Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Mol Cell ; 62(6): 890-902, 2016 06 16.
Article in English | MEDLINE | ID: mdl-27264869

ABSTRACT

The mouse double minute 2 (MDM2) oncoprotein is recognized as a major negative regulator of the p53 tumor suppressor, but growing evidence indicates that its oncogenic activities extend beyond p53. Here, we show that MDM2 is recruited to chromatin independently of p53 to regulate a transcriptional program implicated in amino acid metabolism and redox homeostasis. Identification of MDM2 target genes at the whole-genome level highlights an important role for ATF3/4 transcription factors in tethering MDM2 to chromatin. MDM2 recruitment to chromatin is a tightly regulated process that occurs during oxidative stress and serine/glycine deprivation and is modulated by the pyruvate kinase M2 (PKM2) metabolic enzyme. Depletion of endogenous MDM2 in p53-deficient cells impairs serine/glycine metabolism, the NAD(+)/NADH ratio, and glutathione (GSH) recycling, impacting their redox state and tumorigenic potential. Collectively, our data illustrate a previously unsuspected function of chromatin-bound MDM2 in cancer cell metabolism.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Chromatin Assembly and Disassembly , Chromatin/metabolism , Colonic Neoplasms/metabolism , Lung Neoplasms/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Serine/metabolism , Tumor Suppressor Protein p53/metabolism , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Proliferation , Chromatin/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Glycine/metabolism , HCT116 Cells , Homeostasis , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Nude , Mutation , Oxidation-Reduction , Oxidative Stress , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-mdm2/genetics , RNA Interference , Thyroid Hormones/genetics , Thyroid Hormones/metabolism , Time Factors , Transcription, Genetic , Transfection , Tumor Burden , Tumor Suppressor Protein p53/genetics , Thyroid Hormone-Binding Proteins
2.
Bioconjug Chem ; 34(11): 2014-2021, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37556437

ABSTRACT

The neuropeptide-Y (NPY) family acts through four G protein-coupled receptor subtypes in humans, namely, Y1, Y2, Y4, and Y5. A growing body of evidence suggest the involvement of the NPY system in several cancers, notably the Y5 subtype, thus acting as a relevant target for the development of radiopharmaceuticals for imaging or targeted radionuclide therapy (TRT). Here, the [cPP(1-7),NPY(19-23),Ala31,Aib32,Gln34]hPP scaffold, further referred to as sY5ago, was modified with a DOTA chelator and radiolabeled with 68Ga and 111In and investigated in vitro and in vivo using the MCF-7 model. For in vivo studies, MCF-7 cells were orthotopically implanted in female nude mice and imaging with small animal positron emission tomography/computed tomography (µPET/CT) was performed. At the end of imaging, the mice were sacrificed. A scrambled version of sY5ago, which was also modified with a DOTA chelator, served as a negative control (DOTA-[Nle]sY5ago_scrambled). sY5ago and DOTA-sY5ago showed subnanomolar affinity toward the Y5 (0.9 ± 0.1 and 0.8 ± 0.1 nM, respectively) and a single binding site at the Y5 was identified. [68Ga]Ga-DOTA-sY5ago and [111In]In-DOTA-sY5ago were hydrophilic and showed high specific internalization (1.61 ± 0.75%/106 cells at 1 h) and moderate efflux (55% of total binding externalized at 45 min). On µPET/CT images, most of the signal was depicted in the kidneys and the liver. MCF-7 tumors were clearly visualized. On biodistribution studies, [68Ga]Ga-DOTA-sY5ago was eliminated by the kidneys (∼60 %ID/g). The kidney uptake is Y5-mediated. A specific uptake was also noted in the liver (5.09 ± 1.15 %ID/g vs 1.13 ± 0.21 %ID/g for [68Ga]Ga-DOTA-[Nle]sY5ago_scrambled, p < 0.05), the lungs (1.03 ± 0.34 %ID/g vs 0.20 %ID/g, p < 0.05), and the spleen (0.85 ± 0.09%ID/g vs 0.16 ± 0.16%ID/g, p < 0.05). In MCF-7 tumors, [68Ga]Ga-DOTA-sY5ago showed 12-fold higher uptake than [68Ga]Ga-DOTA-[Nle]sY5ago_scrambled (3.43 ± 2.32 vs 0.27 ± 0.15 %ID/g, respectively, p = 0.0008) at 1 h post-injection. Finally, a proof-of-principle tissular micro-imaging study on a human primary cancer sample showed weak binding of [111In]In-DOTA-sY5ago in prostatic intra-neoplasia and high binding in the ISUP1 lesion while normal prostate was free of signal.


Subject(s)
Prostatic Neoplasms , Receptors, Neuropeptide Y , Male , Mice , Humans , Animals , Receptors, Neuropeptide Y/metabolism , Radiopharmaceuticals , Gallium Radioisotopes , Mice, Nude , Tissue Distribution , Chelating Agents , Positron-Emission Tomography/methods
3.
Biomater Adv ; 161: 213870, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38701686

ABSTRACT

Wound healing following skin tumour surgery still remains a major challenge. To address this issue, polysaccharide-loaded nanofibrous mats have been engineered as skin patches on the wound site to improve wound healing while simultaneously eliminating residual cancer cells which may cause cancer relapse. The marine derived polysaccharides kappa-carrageenan (KCG) and fucoidan (FUC) were blended with polydioxanone (PDX) nanofibers due to their inherent anti-cancer activity conferred by the sulphate groups as well as their immunomodulatory properties which can reduce inflammation resulting in accelerated wound healing. KCG and FUC were released sustainably from the blend nanofibers via the Korsmeyer-Peppas kinetics. MTT assays, live/dead staining and SEM images demonstrated the toxicity of KCG and FUC towards skin cancer MP 41 cells. In addition, MP 41 cells showed reduced metastatic potential when grown on KCG or FUC containing mats. Both KCG and FUC were non- cytotoxic to healthy L 929 fibroblast cells. In vivo studies on healthy Wistar rats confirmed the non-toxicity of the nanofibrous patches as well as their improved and scarless wound healing potential. In vivo studies on tumour xenograft model further showed a reduction of 7.15 % in tumour volume in only 4 days following application of the transdermal patch.


Subject(s)
Melanoma , Nanofibers , Polysaccharides , Rats, Wistar , Skin Neoplasms , Tissue Scaffolds , Wound Healing , Animals , Wound Healing/drug effects , Nanofibers/chemistry , Rats , Skin Neoplasms/pathology , Melanoma/pathology , Tissue Scaffolds/chemistry , Polysaccharides/pharmacology , Polysaccharides/administration & dosage , Mice , Cell Line, Tumor , Carrageenan/pharmacology , Humans , Polydioxanone/pharmacology , Polydioxanone/chemistry , Neoplasm Recurrence, Local/prevention & control , Neoplasm Recurrence, Local/pathology
4.
ACS Omega ; 8(7): 6994-7004, 2023 Feb 21.
Article in English | MEDLINE | ID: mdl-36844603

ABSTRACT

Neurotensin receptor 2 (NTS2) is a well-known mediator of central opioid-independent analgesia. Seminal studies have highlighted NTS2 overexpression in a variety of tumors including prostate cancer, pancreas adenocarcinoma, and breast cancer. Herein, we describe the first radiometalated neurotensin analogue targeting NTS2. JMV 7488 (DOTA-(ßAla)2-Lys-Lys-Pro-(D)Trp-Ile-TMSAla-OH) was prepared using solid-phase peptide synthesis, then purified, radiolabeled with 68Ga and 111In, and investigated in vitro on HT-29 cells and MCF-7 cells, respectively, and in vivo on HT-29 xenografts. [68Ga]Ga-JMV 7488 and [111In]In-JMV 7488 were quite hydrophilic (logD7.4 = -3.1 ± 0.2 and -2.7 ± 0.2, respectively, p < 0.0001). Saturation binding studies showed good affinity toward NTS2 (K D = 38 ± 17 nM for [68Ga]Ga-JMV 7488 on HT-29 and 36 ± 10 nM on MCF-7 cells; K D = 36 ± 4 nM for [111In]In-JMV 7488 on HT-29 and 46 ± 1 nM on MCF-7 cells) and good selectivity (no NTS1 binding up to 500 nM). On cell-based evaluation, [68Ga]Ga-JMV 7488 and [111In]In-JMV 7488 showed high and fast NTS2-mediated internalization of 24 ± 5 and 25 ± 11% at 1 h for [111In]In-JMV 7488, respectively, along with low NTS2-membrane binding (<8%). Efflux was as high as 66 ± 9% at 45 min for [68Ga]Ga-JMV 7488 on HT-29 and increased for [111In]In-JMV 7488 up to 73 ± 16% on HT-29 and 78 ± 9% on MCF-7 cells at 2 h. Maximum intracellular calcium mobilization of JMV 7488 was 91 ± 11% to that of levocabastine, a known NTS2 agonist on HT-29 cells demonstrating the agonist behavior of JMV 7488. In nude mice bearing HT-29 xenograft, [68Ga]Ga-JMV 7488 showed a moderate but promising significant tumor uptake in biodistribution studies that competes well with other nonmetalated radiotracers targeting NTS2. Significant uptake was also depicted in lungs. Interestingly, mice prostate also demonstrated [68Ga]Ga-JMV 7488 uptake although the mechanism was not NTS2-mediated.

5.
Breast Cancer Res ; 13(1): R17, 2011 Feb 04.
Article in English | MEDLINE | ID: mdl-21294885

ABSTRACT

INTRODUCTION: Novel adjuvant therapies are needed to prevent metastatic relapses in HER2-expressing breast cancer. Here, we tested whether trastuzumab-selected single-chain Fv (scFv) could be used to develop an anti-idiotype-based vaccine to inhibit growth of HER2-positive tumor cells in vitro and in vivo through induction of long-lasting HER-specific immunity. METHODS: BALB/c mice were immunized with anti-trastuzumab anti-idiotype (anti-Id) scFv (scFv40 and scFv69), which mimic human HER2. Their sera were assessed for the presence of HER2-specific Ab1' antibodies and for their ability to reduce viability of SK-OV-3 cells, a HER2-positive cancer cell line, in nude mice. MMTV.f.huHER2(Fo5) transgenic mice were immunized with scFv40 and scFv69 and, then, growth inhibition of spontaneous HER2-positive mammary tumors, humoral response, antibody isotype as well as splenocyte secretion of IL2 and IFN-γ were evaluated. RESULTS: Adoptively-transferred sera from BALB/c mice immunized with scFv40 and scFv69 contain anti-HER2 Ab1' antibodies that can efficiently inhibit growth of SK-OV-3 cell tumors in nude mice. Similarly, prophylactic vaccination with anti-Id scFv69 fully protects virgin or primiparous FVB-MMTV.f.huHER2(Fo5) females from developing spontaneous mammary tumors. Moreover, such vaccination elicits an anti-HER2 Ab1' immune response together with a scFv69-specific Th1 response with IL2 and IFN-γ cytokine secretion. CONCLUSIONS: Anti-trastuzumab anti-Id scFv69, used as a therapeutic or prophylactic vaccine, protects mice from developing HER2-positive mammary tumors by inducing both anti-HER2 Ab1' antibody production and an anti-HER2 Th2-dependent immune response. These results suggest that scFv69 could be used as an anti-Id-based vaccine for adjuvant therapy of patients with HER2-positive tumors to reverse immunological tolerance to HER2.


Subject(s)
Antibodies, Anti-Idiotypic/immunology , Antibodies, Monoclonal, Humanized/immunology , Cancer Vaccines/immunology , Immune Tolerance , Ovarian Neoplasms/immunology , Receptor, ErbB-2/immunology , Single-Chain Antibodies/immunology , Animals , Antibodies, Anti-Idiotypic/blood , CHO Cells , Cell Line, Tumor , Cell Survival/immunology , Cricetinae , Female , HEK293 Cells , Humans , Immunity, Humoral , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, Transgenic , Ovarian Neoplasms/therapy , Th1 Cells/immunology , Th2 Cells/immunology , Trastuzumab
6.
Biomater Sci ; 9(15): 5259-5274, 2021 Jul 27.
Article in English | MEDLINE | ID: mdl-34164641

ABSTRACT

The use of non-invasive scaffold materials which can mimic the innate piezoelectric properties of biological tissues is a promising strategy to promote native tissue regeneration. Piezoelectric and cell instructive electrospun core-shell PDX/PHBV mats have been engineered to promote native tissue and skin regeneration. In depth physicochemical characterisation, in vitro and in vivo studies of a rat model showed that the 20/80 PDX/PHBV composition possessed the right balance of physicochemical and piezoelectric properties leading to enhanced fibroblast stimulation, proliferation and migration, reduced fibroblast-mediated contraction and macrophage-induced inflammation, improved keratinocyte proliferation, proper balance between endothelial cell phenotypes, decreased in vivo fibrosis and accelerated in vivo scarless wound regeneration. Overall, this study highlights the importance of exploiting cell-material interactions to match tissue biological needs to sustain the wound healing cascade.


Subject(s)
Tissue Engineering , Tissue Scaffolds , Animals , Fibroblasts , Polyesters , Rats , Wound Healing
7.
Biomedicines ; 9(4)2021 Mar 30.
Article in English | MEDLINE | ID: mdl-33808509

ABSTRACT

The renal fibrotic process is characterized by a chronic inflammatory state and oxidative stress. Antirhea borbonica (A. borbonica) is a French medicinal plant found in Reunion Island and known for its antioxidant and anti-inflammatory activities mostly related to its high polyphenols content. We investigated whether oral administration of polyphenol-rich extract from A. borbonica could exert in vivo a curative anti-renal fibrosis effect. To this aim, three days after unilateral ureteral obstruction (UUO), mice were daily orally treated either with a non-toxic dose of polyphenol-rich extract from A. borbonica or with caffeic acid (CA) for 5 days. The polyphenol-rich extract from A. borbonica, as well as CA, the predominant phenolic acid of this medicinal plant, exerted a nephroprotective effect through the reduction in the three phases of the fibrotic process: (i) macrophage infiltration, (ii) myofibroblast appearance and (iii) extracellular matrix accumulation. These effects were associated with the mRNA down-regulation of Tgf-ß, Tnf-α, Mcp1 and NfkB, as well as the upregulation of Nrf2. Importantly, we observed an increased antioxidant enzyme activity for GPX and Cu/ZnSOD. Last but not least, desorption electrospray ionization-high resolution/mass spectrometry (DESI-HR/MS) imaging allowed us to visualize, for the first time, CA in the kidney tissue. The present study demonstrates that polyphenol-rich extract from A. borbonica significantly improves, in a curative way, renal tubulointerstitial fibrosis progression in the UUO mouse model.

8.
Carbohydr Polym ; 251: 117025, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33142585

ABSTRACT

Cellulose has been extracted from a wide range of land resources, whereas it has been scarcely exploited from marine resources. Cellulose from green seaweeds can be extracted together with smaller molecules called ulvans. We have successfully extracted and characterized cellulose from Ulva sp. Solid state 13C NMR indicated the presence of ulvans in the cellulose extracts. The extracted cellulose was blended with polylactide and polydioxanone and electrospun into nanofibrous mats with a range of physico-chemical properties. These cellulose-based scaffolds were assessed in vitro using fibroblast cells and showed accelerated cell growth. In vivo biocompatibility studies using a Wistar rat model indicated the absence of foreign body response and enhanced angiogenesis.


Subject(s)
Biocompatible Materials , Cellulose , Nanofibers/therapeutic use , Polysaccharides , Tissue Engineering , Tissue Scaffolds , Animals , Biocompatible Materials/chemistry , Cell Line , Cell Proliferation , Cellulose/chemistry , Female , Fibroblasts , Male , Mice , Neovascularization, Physiologic , Polysaccharides/chemistry , Rats , Rats, Wistar , Skin/metabolism , Ulva/chemistry
9.
ACS Med Chem Lett ; 10(5): 743-748, 2019 May 09.
Article in English | MEDLINE | ID: mdl-31097993

ABSTRACT

Atherosclerosis and its associated clinical complications are major health issues in industrialized countries. Lipoprotein-associated phospholipase A2 (Lp-PLA2) was demonstrated to play an important role in atherogenesis and to be a potential risk prediction factor of plaque rupture. Darapladib is one of the most potent Lp-PLA2 inhibitors with an IC50 of 0.25 nM. Using its affinity for Lp-PLA2, we describe herein the total synthesis of darapladib radiolabeling precursor and the automated radiolabeling process for positron emission tomography (PET) imaging via an arylboronate moiety. The tracer thus obtained was tested in a mouse model of atherosclerosis (ApoE KO) and compared with the widely used [18F]fluorodeoxyglucose ([18F]FDG) PET tracer, known to label metabolically active cells. [18F]Darapladib showed a significant accumulation within mice aortic atheromatous plaques dissected out ex vivo compared to [18F]FDG. Incubation of the radiotracer with human carotid samples showed a strong accumulation within the atherosclerotic plaques and supports its potential for use in PET imaging.

10.
Eur J Med Chem ; 176: 129-134, 2019 Aug 15.
Article in English | MEDLINE | ID: mdl-31102933

ABSTRACT

Cardiovascular disease is the leading cause of mortality and morbidity worldwide. Atherosclerosis accounts for 50% of deaths in western countries. This multifactorial pathology is characterized by the accumulation of lipids and inflammatory cells within the vascular wall, leading to plaque formation. We describe herein the synthesis of a PCTA-based 68Ga3+ chelator coupled to a phospholipid biovector 1,2-distearoyl-sn-glycero-3-phosphoethanolamine (DSPE), which is the main constituent of the phospholipid moiety of High-Density Lipoprotein (HDL) phospholipid moiety. The resulting 68Ga-PCTA-DSPE inserted into HDL particles was compared to 18F-FDG as a PET agent to visualize atherosclerotic plaques. Our agent markedly accumulated within mouse atheromatous aortas and more interestingly in human endarterectomy carotid samples. These results support the potential use of 68Ga-PCTA-DSPE-HDL for atherosclerosis PET imaging.


Subject(s)
Atherosclerosis/diagnostic imaging , Chelating Agents/chemistry , Gallium Radioisotopes/chemistry , Heterocyclic Compounds, 2-Ring/chemistry , Phosphatidylethanolamines/chemistry , Radiopharmaceuticals/chemistry , Animals , Aorta/metabolism , Aorta/pathology , Apolipoproteins E/genetics , Carotid Arteries/metabolism , Carotid Arteries/pathology , Chelating Agents/chemical synthesis , Drug Carriers/chemistry , Drug Development , Heterocyclic Compounds, 2-Ring/chemical synthesis , Humans , Lipoproteins, HDL/chemistry , Liver/metabolism , Mice, Knockout , Myocardium/metabolism , Myocardium/pathology , Phosphatidylethanolamines/chemical synthesis , Positron-Emission Tomography/methods , Radiopharmaceuticals/chemical synthesis
11.
J Comp Neurol ; 526(6): 1006-1016, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29277912

ABSTRACT

Clinical benefit for mechanical thrombectomy (MT) in stroke was recently demonstrated in multiple large prospective studies. Acute hyperglycemia (HG) is an important risk factor of poor outcome in stroke patients, including those that underwent MT. The aim of this therapy is to achieve a complete reperfusion in a short time, given that reperfusion damage is dependent on the duration of ischemia. Here, we investigated the effects of acute HG in a mouse model of ischemic stroke induced by middle cerebral artery occlusion (MCAO). Hyperglycemic (intraperitoneal [ip] injection of glucose) and control (ip saline injection) 10-week male C57BL6 mice were subjected to MCAO (30, 90, and 180 min) followed by reperfusion obtained by withdrawal of the monofilament. Infarct volume, hemorrhagic transformation (HT), neutrophil infiltration, and neurological scores were assessed at 24 hr by performing vital staining, ELISA immunofluorescence, and behavioral test, respectively. Glucose injection led to transient HG (blood glucose = 250-390 mg/dL) that significantly increased infarct volume, HT, and worsened neurological outcome. In addition, we report that HG promoted blood-brain barrier disruption as shown by hemoglobin accumulation in the brain parenchyma and tended to increase neutrophil extravasation within the infarcted area. Acute HG increased neurovascular damage for all MCAO durations tested. HTs were observed as early as 90 min after ischemia under hyperglycemic conditions. This model mimics MT ischemia/reperfusion and allows the exploration of brain injury in hyperglycemic conditions.


Subject(s)
Glucose/therapeutic use , Hyperglycemia , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/therapy , Intracranial Hemorrhages/etiology , Animals , Blood Glucose/metabolism , Disease Models, Animal , Hemoglobins/metabolism , Male , Mice , Mice, Inbred C57BL , Nervous System Diseases/etiology , Neutrophil Infiltration , Neutrophils/metabolism , Time Factors
12.
J Vis Exp ; (124)2017 06 26.
Article in English | MEDLINE | ID: mdl-28671660

ABSTRACT

Hyperglycemia is a major health issue that leads to cardiovascular and cerebral dysfunction. For instance, it is associated with increased neurological problems after stroke and is shown to impair neurogenic processes. Interestingly, the adult zebrafish has recently emerged as a relevant and useful model to mimic hyperglycemia/diabetes and to investigate constitutive and regenerative neurogenesis. This work provides methods to develop zebrafish models of hyperglycemia to explore the impact of hyperglycemia on brain cell proliferation under homeostatic and brain repair conditions. Acute hyperglycemia is established using the intraperitoneal injection of D-glucose (2.5 g/kg bodyweight) into adult zebrafish. Chronic hyperglycemia is induced by immersing adult zebrafish in D-glucose (111 mM) containing water for 14 days. Blood-glucose-level measurements are described for these different approaches. Methods to investigate the impact of hyperglycemia on constitutive and regenerative neurogenesis, by describing the mechanical injury of the telencephalon, dissecting the brain, paraffin embedding and sectioning with a microtome, and performing immunohistochemistry procedures, are demonstrated. Finally, the method of using zebrafish as a relevant model for studying the biodistribution of radiolabeled molecules (here,[18F]-FDG) using PET/CT is also described.


Subject(s)
Hyperglycemia/diagnosis , Neurogenesis/physiology , Positron Emission Tomography Computed Tomography/methods , Acute Disease , Animals , Chronic Disease , Disease Models, Animal , Male , Tissue Distribution , Zebrafish , Zebrafish Proteins/metabolism
13.
Clin Cancer Res ; 23(11): 2806-2816, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-27923843

ABSTRACT

Purpose: AXL receptor tyrosine kinase has been described as a relevant molecular marker and a key player in invasiveness, especially in triple-negative breast cancer (TNBC).Experimental Design: We evaluate the antitumor efficacy of the anti-AXL monoclonal antibody 20G7-D9 in several TNBC cell xenografts or patient-derived xenograft (PDX) models and decipher the underlying mechanisms. In a dataset of 254 basal-like breast cancer samples, genes correlated with AXL expression are enriched in EMT, migration, and invasion signaling pathways.Results: Treatment with 20G7-D9 inhibited tumor growth and bone metastasis formation in AXL-positive TNBC cell xenografts or PDX, but not in AXL-negative PDX, highlighting AXL role in cancer growth and invasion. In vitro stimulation of AXL-positive cancer cells by its ligand GAS6 induced the expression of several EMT-associated genes (SNAIL, SLUG, and VIM) through an intracellular signaling implicating the transcription factor FRA-1, important in cell invasion and plasticity, and increased their migration/invasion capacity. 20G7-D9 induced AXL degradation and inhibited all AXL/GAS6-dependent cell signaling implicated in EMT and in cell migration/invasion.Conclusions: The anti-AXL antibody 20G7-D9 represents a promising therapeutic strategy in TNBC with mesenchymal features by inhibiting AXL-dependent EMT, tumor growth, and metastasis formation. Clin Cancer Res; 23(11); 2806-16. ©2016 AACR.


Subject(s)
Antibodies, Anti-Idiotypic/administration & dosage , Cell Proliferation/drug effects , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/immunology , Triple Negative Breast Neoplasms/therapy , Animals , Antibodies, Anti-Idiotypic/immunology , Cell Movement/drug effects , Cell Movement/immunology , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/immunology , Female , Gene Expression Regulation, Neoplastic/drug effects , Heterografts , Humans , Mice , Neoplasm Metastasis , Proto-Oncogene Proteins/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays , Axl Receptor Tyrosine Kinase
14.
Cancer Res ; 76(12): 3618-28, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27197176

ABSTRACT

Subpopulations of cancer stem-like cells (CSC) are thought to drive tumor progression and posttreatment recurrence in multiple solid tumors. However, the mechanisms that maintain stable proportions of self-renewing CSC within heterogeneous tumors under homeostatic conditions remain poorly understood. Progastrin is a secreted peptide that exhibits tumor-forming potential in colorectal cancer, where it regulates pathways known to modulate colon CSC behaviors. In this study, we investigated the role of progastrin in regulating CSC phenotype in advanced colorectal cancer. Progastrin expression and secretion were highly enriched in colon CSC isolated from human colorectal cancer cell lines and colon tumor biopsies. Progastrin expression promoted CSC self-renewal and survival, whereas its depletion by RNA interference-mediated or antibody-mediated strategies altered the homeostatic proportions of CSC cells within heterogeneous colorectal cancer tumors. Progastrin downregulation also decreased the frequency of ALDH(high) cells, impairing their tumor-initiating potential, and inhibited the high glycolytic activity of ALDH(high) CSC to limit their self-renewal capability. Taken together, our results show how colorectal CSC maintain their tumor-initiating and self-renewal capabilities by secreting progastrin, thereby contributing to the tumor microenvironment to support malignancy. Cancer Res; 76(12); 3618-28. ©2016 AACR.


Subject(s)
Colonic Neoplasms/pathology , Gastrins/physiology , Neoplastic Stem Cells/physiology , Protein Precursors/physiology , Aldehyde Dehydrogenase/metabolism , Animals , Cell Line, Tumor , Cell Survival , Humans , Mice , Tumor Microenvironment
15.
J Clin Invest ; 124(5): 1899-913, 2014 May.
Article in English | MEDLINE | ID: mdl-24667635

ABSTRACT

Deregulation of the Wnt/APC/ß-catenin signaling pathway is an important consequence of tumor suppressor APC dysfunction. Genetic and molecular data have established that disruption of this pathway contributes to the development of colorectal cancer. Here, we demonstrate that the transcriptional coregulator RIP140 regulates intestinal homeostasis and tumorigenesis. Using Rip140-null mice and mice overexpressing human RIP140, we found that RIP140 inhibited intestinal epithelial cell proliferation and apoptosis. Interestingly, following whole-body irradiation, mice lacking RIP140 exhibited improved regenerative capacity in the intestine, while mice overexpressing RIP140 displayed reduced recovery. Enhanced RIP140 expression strongly repressed human colon cancer cell proliferation in vitro and after grafting onto nude mice. Moreover, in murine tissues and human cancer cells, RIP140 stimulated APC transcription and inhibited ß-catenin activation and target gene expression. Finally, RIP140 mRNA and RIP140 protein levels were decreased in human colon cancers compared with those in normal mucosal tissue, and low levels of RIP140 expression in adenocarcinomas from patients correlated with poor prognosis. Together, these results support a tumor suppressor role for RIP140 in colon cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenomatous Polyposis Coli Protein/biosynthesis , Cell Transformation, Neoplastic/metabolism , Colonic Neoplasms/metabolism , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Homeostasis , Intestinal Mucosa/metabolism , Nuclear Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adenomatous Polyposis Coli Protein/genetics , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Colonic Neoplasms/pathology , Epithelial Cells/pathology , Female , Heterografts , Humans , Intestinal Mucosa/pathology , Male , Mice , Mice, Knockout , Mice, Nude , Neoplasm Transplantation , Nuclear Proteins/genetics , Nuclear Receptor Interacting Protein 1
16.
Cancer Res ; 71(3): 1041-9, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21159664

ABSTRACT

Despite recent advances in the treatment of colon cancer, tumor resistance is a frequent cause of chemotherapy failure. To better elucidate the molecular mechanisms involved in resistance to irinotecan (and its active metabolite SN38), we established SN38-resistant clones derived from HCT-116 and SW48 cell lines. These clones show various levels (6- to 60-fold) of resistance to SN-38 and display enhanced levels of activated MAPK p38 as compared with the corresponding parental cells. Because four different isoforms of p38 have been described, we then studied the effect of p38 overexpression or downregulation of each isoform on cell sensivity to SN38 and found that both α and ß isoforms are involved in the development of resistance to SN38. In this line, we show that cell treatment with SB202190, which inhibits p38α and p38ß, enhanced the cytotoxic activity of SN38. Moreover, p38 inhibition sensitized tumor cells derived from both SN38-sensitive and -resistant HCT116 cells to irinotecan treatment in xenograft models. Finally, we detected less phosphorylated p38 in primary colon cancer of patients sensitive to irinotecan-based treatment, compared with nonresponder patients. This indicates that enhanced level of phosphorylated p38 could predict the absence of clinical response to irinotecan. Altogether, our results show that the p38 MAPK pathway is involved in irinotecan sensitivity and suggest that phosphorylated p38 expression level could be used as a marker of clinical resistance to irinotecan. They further suggest that targeting the p38 pathway may be a potential strategy to overcome resistance to irinotecan-based chemotherapies in colorectal cancer.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/enzymology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Camptothecin/analogs & derivatives , Colonic Neoplasms/drug therapy , Colonic Neoplasms/enzymology , Protein Kinase Inhibitors/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Camptothecin/administration & dosage , Camptothecin/pharmacology , Drug Resistance, Neoplasm , Drug Synergism , Female , Fluorouracil/administration & dosage , HCT116 Cells , Humans , Imidazoles/administration & dosage , Imidazoles/pharmacology , Immunohistochemistry , Irinotecan , Isoenzymes , Leucovorin/administration & dosage , MAP Kinase Signaling System , Mice , Mice, Nude , Phosphorylation , Pyridines/administration & dosage , Pyridines/pharmacology , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL