Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
Add more filters

Publication year range
1.
Immunity ; 29(6): 922-33, 2008 Dec 19.
Article in English | MEDLINE | ID: mdl-19013083

ABSTRACT

Mice with mutations in the gene encoding Fas ligand (FasL) develop lymphoproliferation and systemic autoimmune diseases. However, the cellular subset responsible for the prevention of autoimmunity in FasL-deficient mice remains undetermined. Here, we show that mice with FasL loss on either B or T cells had identical life span as littermates, and both genotypes developed signs of autoimmunity. In addition, we show that T cell-dependent death was vital for the elimination of aberrant T cells and for controlling the numbers of B cells and dendritic cells that dampen autoimmune responses. Furthermore, we show that the loss of FasL on T cells affected the follicular dentritic cell network in the germinal centers, leading to an impaired recall response to exogenous antigen. These results disclose the distinct roles of cellular subsets in FasL-dependent control of autoimmunity and provide further insight into the role of FasL in humoral immunity.


Subject(s)
Autoimmune Diseases/immunology , B-Lymphocytes/immunology , Dendritic Cells/immunology , Fas Ligand Protein/immunology , T-Lymphocytes/immunology , Animals , Antigens/immunology , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Dendritic Cells/metabolism , Fas Ligand Protein/genetics , Lymphatic Diseases/genetics , Lymphatic Diseases/immunology , Lymphatic Diseases/pathology , Mice , Mice, Knockout , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , fas Receptor/immunology , fas Receptor/metabolism
2.
Pharm Res ; 33(8): 1913-22, 2016 08.
Article in English | MEDLINE | ID: mdl-27091031

ABSTRACT

PURPOSE: Busulfan-melphalan high-dose chemotherapy followed by autologous stem cell transplantation is an essential consolidation treatment of high-risk neuroblastoma in children. Main treatment limitation is hepatic veno-occlusive disease, the most severe and frequent extra-hematological toxicity. This life threatening toxicity has been related to a drug interaction between busulfan and melphalan which might be increased by prior disturbance of iron homeostasis, i.e. an increased plasma ferritin level. METHODS: We performed an experimental study of busulfan and melphalan pharmacodynamic and pharmacokinetics in iron overloaded mice. RESULTS: Iron excess dramatically increased the toxicity of melphalan or busulfan melphalan combination in mice but it did not modify the clearance of either busulfan or melphalan. We show that prior busulfan treatment impairs the clearance of melphalan. This clearance alteration was exacerbated in iron overloaded mice demonstrating a pharmacokinetic interaction. Additionally, iron overload increased melphalan toxicity without altering its pharmacokinetics, suggesting a pharmacodynamic interaction between iron and melphalan. Based on iron homeostasis disturbance, we postulated that prior induction of ferritin, through Nrf2 activation after oxidative stress, may be associated with the alteration of melphalan metabolism. CONCLUSION: Iron overload increases melphalan and busulfan-melphalan toxicity through a pharmacodynamic interaction and reveals a pharmacokinetic drug interaction between busulfan and melphalan.


Subject(s)
Busulfan/metabolism , Busulfan/toxicity , Iron Overload/metabolism , Melphalan/metabolism , Melphalan/toxicity , Animals , Antineoplastic Agents, Alkylating/metabolism , Antineoplastic Agents, Alkylating/toxicity , Drug Interactions/physiology , Iron Overload/pathology , Male , Mice , Mice, Inbred C57BL
3.
EMBO Mol Med ; 16(8): 1840-1885, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39009887

ABSTRACT

We have recently identified the uncharacterized ZNF555 protein as a component of a productive complex involved in the morbid function of the 4qA locus in facioscapulohumeral dystrophy. Subsequently named DiPRO1 (Death, Differentiation, and PROliferation related PROtein 1), our study provides substantial evidence of its role in the differentiation and proliferation of human myoblasts. DiPRO1 operates through the regulatory binding regions of SIX1, a master regulator of myogenesis. Its relevance extends to mesenchymal tumors, such as rhabdomyosarcoma (RMS) and Ewing sarcoma, where DiPRO1 acts as a repressor via the epigenetic regulators TIF1B and UHRF1, maintaining methylation of cis-regulatory elements and gene promoters. Loss of DiPRO1 mimics the host defense response to virus, awakening retrotransposable repeats and the ZNF/KZFP gene family. This enables the eradication of cancer cells, reprogramming the cellular decision balance towards inflammation and/or apoptosis by controlling TNF-α via NF-kappaB signaling. Finally, our results highlight the vulnerability of mesenchymal cancer tumors to si/shDiPRO1-based nanomedicines, positioning DiPRO1 as a potential therapeutic target.


Subject(s)
Cell Differentiation , Humans , Cell Proliferation , Myoblasts/metabolism
4.
Am J Pathol ; 180(5): 2084-96, 2012 May.
Article in English | MEDLINE | ID: mdl-22425713

ABSTRACT

Because many of the genes used to produce induced pluripotent stem cells (iPSCs) from somatic cells are either outright established oncogenes, such as c-myc and Klf4, or potentially related to tumorigenesis in various cancers, both the safety and the risks of tumorigenesis linked to iPSC generation require evaluation. In this work, we generated, by lentivirus-mediated gene transfer of Oct4, Sox2, Nanog, and Lin28, two types of iPSCs from human mesenchymal stem cells and human amniotic fluid-derived cells: fully reprogrammed iPSCs with silencing of the four transgenes and partially reprogrammed iPSCs that still express one or several transgenes. We assessed the behavior of these cells during both their differentiation and proliferation using in vivo teratoma assays in nonobese diabetic mice with severe combined immunodeficiency. In contrast to fully reprogrammed iPSCs, 43% of partially reprogrammed iPSC cases (6 of 14 teratomas) generated major dysplasia and malignant tumors, with yolk sac tumors and embryonal carcinomas positive for α-fetoprotein, cytokeratin AE1/AE3, and CD30. This correlated with the expression of one or several transgenes used for the reprogramming, down-regulation of CDK 1A mRNA (p21/CDKN1A), and up-regulation of antiapoptotic Bcl-2 mRNA. Therefore, the oncogenicity of therapeutically valuable patient-specific iPSC-derived cells should be scrupulously evaluated before they are used for any clinical applications.


Subject(s)
Cell Transformation, Neoplastic/pathology , Induced Pluripotent Stem Cells/pathology , Ki-1 Antigen/metabolism , Neoplasms, Germ Cell and Embryonal/pathology , Animals , Cell Differentiation/physiology , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cellular Reprogramming/physiology , Embryonic Stem Cells/cytology , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Karyotype , Kruppel-Like Factor 4 , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms, Germ Cell and Embryonal/genetics , Neoplasms, Germ Cell and Embryonal/metabolism , Teratoma/metabolism , Teratoma/pathology , Transgenes/genetics
5.
Blood ; 117(11): 3065-75, 2011 Mar 17.
Article in English | MEDLINE | ID: mdl-21149635

ABSTRACT

The early emergence of macrophages and their large pattern of tissue distribution during development suggest that they may play a critical role in the initial steps of embryogenesis. In the present study, we show that monocytic cells derived from human embryonic stem cells (hESCs) and from fetal liver follow a differentiation pathway different to that of adult cells, leading to specific functions. Embryonic and fetal monocytic cells differentiated from a CD14(low)CD16(-) precursor to form CD14(high)CD16(+) cells without producing the CD14(high)CD16(-) cell population that predominates in adult peripheral blood. Both demonstrated an enhanced expression of genes encoding tissue-degrading enzymes, chemokines, and scavenger receptors, as was previously reported for M2 macrophages. Compared with adult blood monocytes, embryonic and fetal monocytic cells secreted high amounts of proteins acting on tissue remodeling and angiogenesis, and most of them expressed the Tie2 receptor. Furthermore, they promoted vascular remodeling in xenotransplanted human tumors. These findings suggest that the regulation of human fetal and embryonic monocytic cell differentiation leads to the generation of cells endowed mainly with anti-inflammatory and remodeling functions. Trophic and immunosuppressive functions of M2-polarized macrophages link fetus and tumor development, and hESCs offer a valuable experimental model for in vitro studies of mechanisms sustaining these processes.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Fetus/cytology , Homeostasis , Liver/cytology , Liver/embryology , Monocytes/cytology , Adult , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Line , Cytokines/genetics , Cytokines/metabolism , Embryonic Stem Cells/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Humans , Inflammation/pathology , Lipopolysaccharide Receptors/metabolism , Macrophages/cytology , Macrophages/metabolism , Monocytes/metabolism , Organ Specificity/genetics , Phenotype , Receptors, IgG/metabolism
6.
Nat Med ; 12(2): 214-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16444265

ABSTRACT

The interferon (IFN)-gamma-induced TRAIL effector mechanism is a vital component of cancer immunosurveillance by natural killer (NK) cells in mice. Here we show that the main source of IFN-gamma is not the conventional NK cell but a subset of B220(+)Ly6C(-) dendritic cells, which are atypical insofar as they express NK cell-surface molecules. Upon contact with a variety of tumor cells that are poorly recognized by NK cells, B220(+)NK1.1(+) dendritic cells secrete high levels of IFN-gamma and mediate TRAIL-dependent lysis of tumor cells. Adoptive transfer of these IFN-producing killer dendritic cells (IKDCs) into tumor-bearing Rag2(-/-)Il2rg(-/-) mice prevented tumor outgrowth, whereas transfer of conventional NK cells did not. In conclusion, we identified IKDCs as pivotal sensors and effectors of the innate antitumor immune response.


Subject(s)
Dendritic Cells/classification , Dendritic Cells/immunology , Neoplasms, Experimental/immunology , Adoptive Transfer , Animals , Antigen Presentation , Antigens, Ly , Antigens, Surface/metabolism , Apoptosis Regulatory Proteins/immunology , CD11c Antigen/metabolism , Cell Line, Tumor , Cytotoxicity, Immunologic , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Dendritic Cells/ultrastructure , Female , Interferon-gamma/biosynthesis , Interleukin Receptor Common gamma Subunit , Killer Cells, Natural/immunology , Lectins, C-Type/metabolism , Leukocyte Common Antigens/metabolism , Membrane Glycoproteins/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Microscopy, Electron , NK Cell Lectin-Like Receptor Subfamily B , Receptors, Interleukin/deficiency , Receptors, Interleukin/genetics , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/immunology
7.
Blood ; 115(18): 3686-94, 2010 May 06.
Article in English | MEDLINE | ID: mdl-20223923

ABSTRACT

The mitogen-activated protein kinases (MAPKs) extracellular signal-regulated kinase 1 (ERK1) and ERK2 are among the main signal transduction molecules, but little is known about their isoform-specific functions in vivo. We have examined the role of ERK1 in adult hematopoiesis with ERK1(-/-) mice. Loss of ERK1 resulted in an enhanced splenic erythropoiesis, characterized by an accumulation of erythroid progenitors in the spleen, without any effect on the other lineages or on bone marrow erythropoiesis. This result suggests that the ablation of ERK1 induces a splenic stress erythropoiesis phenotype. However, the mice display no anemia. Deletion of ERK1 did not affect erythropoietin (EPO) serum levels or EPO/EPO receptor signaling and was not compensated by ERK2. Splenic stress erythropoiesis response has been shown to require bone morphogenetic protein 4 (BMP4)-dependent signaling in vivo and to rely on the expansion of a resident specialized population of erythroid progenitors, termed stress erythroid burst-forming units (BFU-Es). A great expansion of stress BFU-Es and increased levels of BMP4 mRNA were found in ERK1(-/-) spleens. The ERK1(-/-) phenotype can be transferred by bone marrow cells. These findings show that ERK1 controls a BMP4-dependent step, regulating the steady state of splenic erythropoiesis.


Subject(s)
Anemia/pathology , Erythroid Precursor Cells/physiology , Erythropoiesis/physiology , Mitogen-Activated Protein Kinase 3/physiology , Spleen/metabolism , Anemia/chemically induced , Animals , Apoptosis , Blotting, Western , Bone Marrow Transplantation , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Colony-Forming Units Assay , Erythroid Precursor Cells/cytology , Erythropoietin/metabolism , Flow Cytometry , Mice , Mice, Inbred C57BL , Oxidants/toxicity , Phenylhydrazines/toxicity , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Spleen/cytology
8.
Invest New Drugs ; 30(1): 144-56, 2012 Feb.
Article in English | MEDLINE | ID: mdl-20924644

ABSTRACT

BACKGROUND: Interstitial hypertension is responsible for poor capillary blood flow and hampered drug delivery. The efficacy of combined sorafenib/bevacizumab treatment given according to different administration schedules has been evaluated by measuring both interstitial pressure (IP) and quantitative dynamic contrast-enhanced ultrasonography (DCE-US) parameters in melanoma-bearing mice. MATERIAL AND METHODS: [corrected] Sixty mice were xenografted with B16F10 melanoma. Animals received a daily administration over 4 days (D0 to D3) of either sorafenib at 30 mg/kg, bevacizumab at 2.5 mg/kg alone, or different schedules of combined treatments. Perfusion parameters determined using an Aplio® sonograph (Toshiba) with SonoVue® contrast agent (Bracco) were compared to IP measurements using fiberoptic probes (Samba®) at D0, D2, D4, D8. RESULTS: The mean baseline IP values ranged between 6.55 and 31.29 mmHg in all the groups. A transient IP decrease occurred at D2 in all treated groups, and especially in the concomitant group which exhibited a significant IP reduction compared to D0. A significant decrease in both the peak intensity and the area under the curve was observed at D4 in the group with concomitant administration of both molecules which yielded maximal inhibition of the tumor volume and the number of vessels. No correlation was found between IP values and volume or perfusion parameters, indicating complex relationships between IP and vascularization. No IP gradients were found between the center and the periphery but IP values in these two regions were significantly correlated (R = 0.93). CONCLUSION: The results suggest that IP variations could be predictive of vascular changes and that one single IP measurement is sufficient to fully characterize the whole tumor.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzenesulfonates/administration & dosage , Extracellular Fluid/metabolism , Melanoma, Experimental/drug therapy , Neovascularization, Pathologic/prevention & control , Perfusion Imaging , Pyridines/administration & dosage , Animals , Bevacizumab , Contrast Media , Drug Administration Schedule , Female , Fiber Optic Technology , Melanoma, Experimental/blood supply , Melanoma, Experimental/diagnostic imaging , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Neovascularization, Pathologic/diagnostic imaging , Neovascularization, Pathologic/metabolism , Niacinamide/analogs & derivatives , Perfusion Imaging/methods , Phenylurea Compounds , Phospholipids , Pressure , Regional Blood Flow , Sorafenib , Sulfur Hexafluoride , Time Factors , Transducers, Pressure , Ultrasonography, Doppler
9.
Exp Cell Res ; 317(3): 262-75, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-20974126

ABSTRACT

Deciphering molecular pathways involved in the early steps of prostate oncogenesis requires both in vitro and in vivo models derived from human primary tumors. However the few recognized models of human prostate epithelial cancer originate from metastases. To date, very few models are proposed from primary tumors and immortalizing normal human prostate cells does not recapitulate the natural history of the disease. By culturing human prostate primary tumor cells onto human epithelial extra-cellular matrix, we successfully selected a new prostate cancer cell line, IGR-CaP1, and clonally-derived subclones. IGR-CaP1 cells, that harbor a tetraploid karyotype, high telomerase activity and mutated TP53, rapidly induced subcutaneous xenografts in nude mice. Furthermore, IGR-CaP1 cell lines, all exhibiting negativity for the androgen receptor and PSA, express the specific prostate markers alpha-methylacyl-CoA racemase and a low level of the prostate-specific membrane antigen PSMA, along with the prostate basal epithelial markers CK5 and CK14. More importantly, these clones express high CD44, CD133, and CXCR4 levels associated with high expression of α2ß1-integrin and Oct4 which are reported to be prostate cancer stemness markers. RT-PCR data also revealed high activation of the Sonic Hedgehog signalling pathway in these cells. Additionally, the IGR-CaP1 cells possess a 3D sphere-forming ability and a renewal capacity by maintaining their CSC potential after xenografting in mice. As a result, the hormone-independent IGR-CaP1 cellular clones exhibit the original features of both basal prostate tissue and cancer stemness. Tumorigenic IGR-CaP1 clones constitute invaluable human models for studying prostate cancer progression and drug assessment in vitro as well as in animals specifically for developing new therapeutic approaches targeting prostate cancer stem cells.


Subject(s)
Extracellular Matrix/pathology , Prostatic Neoplasms/pathology , Tumor Cells, Cultured , Animals , Antigens, Surface/analysis , Biomarkers, Tumor/analysis , Clone Cells/pathology , Humans , Immunophenotyping , Male , Mice , Mice, Nude , Neoplasm Transplantation , Transplantation, Heterologous
10.
Proc Natl Acad Sci U S A ; 106(29): 12127-32, 2009 Jul 21.
Article in English | MEDLINE | ID: mdl-19564597

ABSTRACT

In most mammalian species, a key process of placenta development is the fusion of trophoblast cells into a highly specialized, multinucleated syncytiotrophoblast layer, through which most of the maternofetal exchanges take place. Little is known about this process, despite the recent identification of 2 pairs of envelope genes of retroviral origin, independently acquired by the human (syncytin-1 and syncytin-2) and mouse (syncytin-A and syncytin-B) genomes, specifically expressed in the placenta, and with in vitro cell-cell fusion activity. By generating knockout mice, we show here that homozygous syncytin-A null mouse embryos die in utero between 11.5 and 13.5 days of gestation. Refined cellular and subcellular analyses of the syncytin-A-deficient placentae disclose specific disruption of the architecture of the syncytiotrophoblast-containing labyrinth, with the trophoblast cells failing to fuse into an interhemal syncytial layer. Lack of syncytin-A-mediated trophoblast cell fusion is associated with cell overexpansion at the expense of fetal blood vessel spaces and with apoptosis, adding to the observed maternofetal interface structural defects to provoke decreased vascularization, inhibition of placental transport, and fetal growth retardation, ultimately resulting in death of the embryo. These results demonstrate that syncytin-A is essential for trophoblast cell differentiation and syncytiotrophoblast morphogenesis during placenta development, and they provide evidence that genes captured from ancestral retroviruses have been pivotal in the acquisition of new, important functions in mammalian evolution.


Subject(s)
Endogenous Retroviruses/genetics , Placentation/physiology , Pregnancy Proteins/deficiency , Viral Envelope Proteins/genetics , Animals , Crosses, Genetic , Embryo Loss/metabolism , Embryo Loss/pathology , Embryo, Mammalian/abnormalities , Embryo, Mammalian/pathology , Extraembryonic Membranes/abnormalities , Extraembryonic Membranes/pathology , Female , Gene Targeting , Genotype , Male , Mice , Mice, Knockout , Placenta/abnormalities , Placenta/pathology , Placenta/ultrastructure , Pregnancy , Pregnancy Proteins/metabolism , Trophoblasts/pathology
11.
J Immunother Cancer ; 10(3)2022 03.
Article in English | MEDLINE | ID: mdl-35296557

ABSTRACT

BACKGROUND: Prostate cancer (PC) responds to androgen deprivation therapy (ADT) usually in a transient fashion, progressing from hormone-sensitive PC (HSPC) to castration-resistant PC (CRPC). We investigated a mouse model of PC as well as specimens from PC patients to unravel an unsuspected contribution of thymus-derived T lymphocytes and the intestinal microbiota in the efficacy of ADT. METHODS: Preclinical experiments were performed in PC-bearing mice, immunocompetent or immunodeficient. In parallel, we prospectively included 65 HSPC and CRPC patients (Oncobiotic trial) to analyze their feces and blood specimens. RESULTS: In PC-bearing mice, ADT increased thymic cellularity and output. PC implanted in T lymphocyte-depleted or athymic mice responded less efficiently to ADT than in immunocompetent mice. Moreover, depletion of the intestinal microbiota by oral antibiotics reduced the efficacy of ADT. PC reduced the relative abundance of Akkermansia muciniphila in the gut, and this effect was reversed by ADT. Moreover, cohousing of PC-bearing mice with tumor-free mice or oral gavage with Akkermansia improved the efficacy of ADT. This appears to be applicable to PC patients because long-term ADT resulted in an increase of thymic output, as demonstrated by an increase in circulating recent thymic emigrant cells (sjTRECs). Moreover, as compared with HSPC controls, CRPC patients demonstrated a shift in their intestinal microbiota that significantly correlated with sjTRECs. While feces from healthy volunteers restored ADT efficacy, feces from PC patients failed to do so. CONCLUSIONS: These findings suggest the potential clinical utility of reversing intestinal dysbiosis and repairing acquired immune defects in PC patients.


Subject(s)
Gastrointestinal Microbiome , Prostatic Neoplasms, Castration-Resistant , Androgen Antagonists/pharmacology , Androgen Antagonists/therapeutic use , Androgens/therapeutic use , Animals , Humans , Immune System , Male , Mice , Prostatic Neoplasms, Castration-Resistant/drug therapy
12.
Cancer Discov ; 12(4): 1128-1151, 2022 04 01.
Article in English | MEDLINE | ID: mdl-34930787

ABSTRACT

Gut dysbiosis has been associated with intestinal and extraintestinal malignancies, but whether and how carcinogenesis drives compositional shifts of the microbiome to its own benefit remains an open conundrum. Here, we show that malignant processes can cause ileal mucosa atrophy, with villous microvascular constriction associated with dominance of sympathetic over cholinergic signaling. The rapid onset of tumorigenesis induced a burst of REG3γ release by ileal cells, and transient epithelial barrier permeability that culminated in overt and long-lasting dysbiosis dominated by Gram-positive Clostridium species. Pharmacologic blockade of ß-adrenergic receptors or genetic deficiency in Adrb2 gene, vancomycin, or cohousing of tumor bearers with tumor-free littermates prevented cancer-induced ileopathy, eventually slowing tumor growth kinetics. Patients with cancer harbor distinct hallmarks of this stress ileopathy dominated by Clostridium species. Hence, stress ileopathy is a corollary disease of extraintestinal malignancies requiring specific therapies. SIGNIFICANCE: Whether gut dysbiosis promotes tumorigenesis and how it controls tumor progression remain open questions. We show that 50% of transplantable extraintestinal malignancies triggered a ß-adrenergic receptor-dependent ileal mucosa atrophy, associated with increased gut permeability, sustained Clostridium spp.-related dysbiosis, and cancer growth. Vancomycin or propranolol prevented cancer-associated stress ileopathy. This article is highlighted in the In This Issue feature, p. 873.


Subject(s)
Dysbiosis , Receptors, Adrenergic, beta , Carcinogenesis/pathology , Dysbiosis/chemically induced , Dysbiosis/complications , Dysbiosis/pathology , Humans , Intestinal Mucosa/pathology , Signal Transduction
13.
Int J Cancer ; 128(11): 2748-58, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-20715103

ABSTRACT

Tumor angiogenesis in childhood neuroblastoma is an important prognostic factor suggesting a potential role for antiangiogenic agents in the treatment of high-risk disease. Within the KidsCancerKinome project, we evaluated the new oral selective pan-VEGFR tyrosine kinase inhibitor axitinib (AG-013736) against neuroblastoma cell lines and the subcutaneous and orthotopic xenograft model IGR-N91 derived from a primary bone marrow metastasis. Axitinib reduced cell proliferation in a dose-dependent manner with IC(50) doses between 274 and >10,000 nmol/l. Oral treatment with 30 mg/kg BID for 2 weeks in advanced tumors yielded significant tumor growth delay, with a median time to reach five times initial tumor volume of 11.4 days compared to controls (p = 0.0006) and resulted in significant reduction in bioluminescence. Simultaneous inhibition of VEGFR downstream effector mTOR using rapamycin 20 mg/kg q2d×5 did not statistically enhance tumor growth delay compared to single agent activities. Axitinib downregulated VEGFR-2 phosphorylation resulting in significantly decreased microvessel density (MVD) and overall surface fraction of tumor vessels (OSFV) in all xenografts as measured by CD34 immunohistochemical staining (mean MVD ± SD and OSFV at 14 days 21.27 ± 10.03 in treated tumors vs. 48.79 ± 17.27 in controls and 0.56% vs. 1.29%; p = 0.0006, respectively). We further explored the effects of axitinib on circulating mature endothelial cells (CECs) and endothelial progenitor cells (CEPs) measured by flow cytometry. While only transient modification was observed for CECs, CEP counts were significantly reduced during and up to 14 days after end of treatment. Axitinib has potent antiangiogenic properties that may warrant further evaluation in neuroblastoma.


Subject(s)
Bone Marrow Neoplasms/drug therapy , Imidazoles/therapeutic use , Indazoles/therapeutic use , Neuroblastoma/drug therapy , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-3/antagonists & inhibitors , Xenograft Model Antitumor Assays , Animals , Apoptosis , Axitinib , Blotting, Western , Bone Marrow Neoplasms/secondary , Child , Drug Synergism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans , Male , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Neuroblastoma/blood supply , Neuroblastoma/pathology , Phosphorylation , Sirolimus/pharmacology , Stem Cells/drug effects , Tumor Cells, Cultured
14.
J Exp Med ; 218(10)2021 10 04.
Article in English | MEDLINE | ID: mdl-34495298

ABSTRACT

Cholangiocarcinoma (CCA) results from the malignant transformation of cholangiocytes. Primary sclerosing cholangitis (PSC) and primary biliary cholangitis (PBC) are chronic diseases in which cholangiocytes are primarily damaged. Although PSC is an inflammatory condition predisposing to CCA, CCA is almost never found in the autoimmune context of PBC. Here, we hypothesized that PBC might favor CCA immunosurveillance. In preclinical murine models of cholangitis challenged with syngeneic CCA, PBC (but not PSC) reduced the frequency of CCA development and delayed tumor growth kinetics. This PBC-related effect appeared specific to CCA as it was not observed against other cancers, including hepatocellular carcinoma. The protective effect of PBC was relying on type 1 and type 2 T cell responses and, to a lesser extent, on B cells. Single-cell TCR/RNA sequencing revealed the existence of TCR clonotypes shared between the liver and CCA tumor of a PBC host. Altogether, these results evidence a mechanistic overlapping between autoimmunity and cancer immunosurveillance in the biliary tract.


Subject(s)
Autoimmunity , Bile Duct Neoplasms/immunology , Cholangiocarcinoma/immunology , Cholangitis/immunology , Animals , Bile Duct Neoplasms/pathology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cholangiocarcinoma/pathology , Cholangitis/pathology , Cytokines/metabolism , Female , Forkhead Transcription Factors/metabolism , Liver/immunology , Liver/pathology , Mice, Inbred C57BL , Monitoring, Immunologic , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology
15.
Cell Death Differ ; 28(5): 1532-1547, 2021 05.
Article in English | MEDLINE | ID: mdl-33262469

ABSTRACT

Ileal epithelial cell apoptosis and the local microbiota modulate the effects of oxaliplatin against proximal colon cancer by modulating tumor immunosurveillance. Here, we identified an ileal immune profile associated with the prognosis of colon cancer and responses to chemotherapy. The whole immune ileal transcriptome was upregulated in poor-prognosis patients with proximal colon cancer, while the colonic immunity of healthy and neoplastic areas was downregulated (except for the Th17 fingerprint) in such patients. Similar observations were made across experimental models of implanted and spontaneous murine colon cancer, showing a relationship between carcinogenesis and ileal inflammation. Conversely, oxaliplatin-based chemotherapy could restore a favorable, attenuated ileal immune fingerprint in responders. These results suggest that chemotherapy inversely shapes the immune profile of the ileum-tumor axis, influencing clinical outcome.


Subject(s)
Colonic Neoplasms/physiopathology , Ileal Diseases/complications , Ileum/pathology , Animals , Humans , Mice , Prognosis
16.
Nat Med ; 27(8): 1432-1441, 2021 08.
Article in English | MEDLINE | ID: mdl-34239137

ABSTRACT

Treatment with combined immune checkpoint blockade (CICB) targeting CTLA-4 and PD-1 is associated with clinical benefit across tumor types, but also a high rate of immune-related adverse events. Insights into biomarkers and mechanisms of response and toxicity to CICB are needed. To address this, we profiled the blood, tumor and gut microbiome of 77 patients with advanced melanoma treated with CICB, with a high rate of any ≥grade 3 immune-related adverse events (49%) with parallel studies in pre-clinical models. Tumor-associated immune and genomic biomarkers of response to CICB were similar to those identified for ICB monotherapy, and toxicity from CICB was associated with a more diverse peripheral T-cell repertoire. Profiling of gut microbiota demonstrated a significantly higher abundance of Bacteroides intestinalis in patients with toxicity, with upregulation of mucosal IL-1ß in patient samples of colitis and in pre-clinical models. Together, these data offer potential new therapeutic angles for targeting toxicity to CICB.


Subject(s)
CTLA-4 Antigen/immunology , Gastrointestinal Microbiome , Programmed Cell Death 1 Receptor/immunology , Animals , Cell Line, Tumor , Female , Humans , Interleukin-1beta/immunology , Melanoma , Mice , Mice, Inbred C57BL
17.
Int J Cancer ; 127(5): 1038-51, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20013808

ABSTRACT

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


Subject(s)
Brain Neoplasms/pathology , Carrier Proteins/genetics , Cytokines/genetics , Glioblastoma/pathology , Mutation/genetics , Proto-Oncogene Proteins/genetics , Animals , Apoptosis , Blotting, Western , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , CHO Cells , Cell Adhesion , Cell Movement , Cell Proliferation , Collagen/metabolism , Cricetinae , Cricetulus , Drug Combinations , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation, Neoplastic/physiology , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Immunoenzyme Techniques , Laminin/metabolism , Mice , Mice, Nude , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Proteoglycans/metabolism , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
18.
J Clin Invest ; 117(7): 1844-55, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17557121

ABSTRACT

Tumor radioresponsiveness depends on endothelial cell death, which leads in turn to tumor hypoxia. Radiation-induced hypoxia was recently shown to trigger tumor radioresistance by activating angiogenesis through hypoxia-inducible factor 1-regulated (HIF-1-regulated) cytokines. We show here that combining targeted radioiodide therapy with angiogenic inhibitors, such as canstatin, enhances direct tumor cell apoptosis, thereby overcoming radio-induced HIF-1-dependent tumor survival pathways in vitro and in vivo. We found that following dual therapy, HIF-1alpha increases the activity of the canstatin-induced alpha(v)beta(5) signaling tumor apoptotic pathway and concomitantly abrogates mitotic checkpoint and tetraploidy triggered by radiation. Apoptosis in conjunction with mitotic catastrophe leads to lethal tumor damage. We discovered that HIF-1 displays a radiosensitizing activity that is highly dependent on treatment modalities by regulating key apoptotic molecular pathways. Our findings therefore support a crucial role for angiogenesis inhibitors in shifting the fate of radiation-induced HIF-1alpha activity from hypoxia-induced tumor radioresistance to hypoxia-induced tumor apoptosis. This study provides a basis for developing new biology-based clinically relevant strategies to improve the efficacy of radiation oncology, using HIF-1 as an ally for cancer therapy.


Subject(s)
Apoptosis/radiation effects , Collagen Type IV/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Peptide Fragments/metabolism , Adenoviridae/genetics , Animals , Cell Line , Collagen Type IV/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Genetic Therapy , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Integrins/metabolism , Mice , Mice, Transgenic , Mitochondria/metabolism , Neoplasms/blood supply , Neoplasms/therapy , Peptide Fragments/genetics , Signal Transduction , Symporters/genetics , Symporters/metabolism , Xenograft Model Antitumor Assays
19.
Breast Cancer Res Treat ; 122(1): 145-58, 2010 Jul.
Article in English | MEDLINE | ID: mdl-19771505

ABSTRACT

The suppression of oestrogen receptor alpha (ERalpha) functions by silencing RNAs in association with or not with anti-oestrogens (AEs) both in vitro and in breast cancer cell xenografts was assessed. In vitro, a prolonged decrease in ERalpha protein expression and an enhanced AE-induced inhibition of ERalpha-mediated transcription, together with antiproliferative activity, were observed. Incorporation of ERalpha-siRNAs in pegylated nanocapsules (NC) was achieved; and their intravenous injections in MCF-7 xenografts, in contrast to scramble siRNA containing NCs, lead to decrease in ERalpha protein content and Ki67 labelling in tumour cells. The pure AE RU58668 (RU) both free and entrapped in stealth nanospheres (NS) at very low concentration (8 microg/kg/week) had no effect on tumour growth evolution. However, coinjection of the two nanocarriers potentiated the decrease in ERalpha protein, concomitantly with decreasing tumour vasculature and glucose transporter-1. These data support that the targeted delivery of ERalpha-siRNA in breast tumours potentiates the inhibition of E(2)-induced proliferative activity by encapsulated AE through enhanced anti-vascular activity. In the hormone-independent MDA-MB-231 xenograft model, RU-NS at 4 mg/kg/week induce also a strong tumour vascular normalisation. Together, these findings suggest that the anti-oestrogen activity of RU as well as that of targeted ERalpha-siRNA leads to anti-angiogenic activity. Their delivery in stealth nanocarriers may constitute a new anti-cancer therapeutic strategy in solid tumours.


Subject(s)
Adenocarcinoma/pathology , Breast Neoplasms/pathology , Estradiol/analogs & derivatives , Estrogen Receptor Modulators/therapeutic use , Estrogen Receptor alpha/antagonists & inhibitors , Estrogens , Mammary Neoplasms, Experimental/drug therapy , Nanocapsules/administration & dosage , Neoplasm Proteins/antagonists & inhibitors , Neoplasms, Hormone-Dependent/pathology , RNA, Small Interfering/therapeutic use , Animals , Cell Division/drug effects , Cell Line, Tumor/drug effects , Cell Line, Tumor/transplantation , Drug Synergism , Estradiol/pharmacology , Estradiol/therapeutic use , Estrogen Receptor Modulators/pharmacology , Estrogen Receptor alpha/biosynthesis , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Injections, Intravenous , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Nude , Nanocapsules/chemistry , Nanospheres/administration & dosage , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Polyesters , Polyethylene Glycols , RNA Interference , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/pharmacology , Specific Pathogen-Free Organisms , Xenograft Model Antitumor Assays
20.
Cell Death Discov ; 6(1): 129, 2020 Nov 24.
Article in English | MEDLINE | ID: mdl-33298861

ABSTRACT

Salicylate, the active derivative of aspirin (acetylsalicylate), recapitulates the mode of action of caloric restriction inasmuch as it stimulates autophagy through the inhibition of the acetyltransferase activity of EP300. Here, we directly compared the metabolic effects of aspirin medication with those elicited by 48 h fasting in mice, revealing convergent alterations in the plasma and the heart metabolome. Aspirin caused a transient reduction of general protein acetylation in blood leukocytes, accompanied by the induction of autophagy. However, these effects on global protein acetylation could not be attributed to the mere inhibition of EP300, as determined by epistatic experiments and exploration of the acetyl-proteome from salicylate-treated EP300-deficient cells. Aspirin reduced high-fat diet-induced obesity, diabetes, and hepatosteatosis. These aspirin effects were observed in autophagy-competent mice but not in two different models of genetic (Atg4b-/- or Bcln1+/-) autophagy-deficiency. Aspirin also improved tumor control by immunogenic chemotherapeutics, and this effect was lost in T cell-deficient mice, as well as upon knockdown of an essential autophagy gene (Atg5) in cancer cells. Hence, the health-improving effects of aspirin depend on autophagy.

SELECTION OF CITATIONS
SEARCH DETAIL