Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Mol Cancer Res ; 19(4): 612-622, 2021 04.
Article in English | MEDLINE | ID: mdl-33472949

ABSTRACT

The mechanisms whereby the Hippo pathway effector YAP regulates cancer cell stemness, plasticity, and chemoresistance are not fully understood. We previously showed that in 5-fluorouracil (5-FU)-resistant colorectal cancer cells, the transcriptional coactivator YAP is differentially regulated at critical transitions connected with reversible quiescence/dormancy to promote metastasis. Here, we found that experimental YAP activation in 5-FU-sensitive and 5-FU-resistant HT29 colorectal cancer cells enhanced nuclear YAP localization and the transcript levels of the retinoic acid (RA) receptors RARα/γ and RAR target genes CYP26A1, ALDH1A3, and LGR5 through RA Response Elements (RARE). In these two cell models, constitutive YAP activation reinforced the expression of the stemness biomarkers and regulators ALDH1A3, LGR5, and OCT4. Conversely, YAP silencing, RAR/RXR inhibition by the pan-RAR antagonist BMS493, and vitamin A depletion downregulated stemness traits and self-renewal. Regarding the mechanisms engaged, proximity-dependent labeling, nuclear YAP pulldown coupled with mass spectrometry, and chromatin immunoprecipitation (ChIP)/re-ChIP experiments revealed: (i) the nuclear colocalization/interaction of YAP with RARγ and RXRs; and (ii) combined genomic co-occupancy of YAP, RARα/γ, and RXRα interactomes at proximal RAREs of LGR5 and ALDH1A3 promoters. Moreover, activation of the YAP/RAR-RXR cross-talk in colorectal cancer cells promoted RAR self-activation loops via vitamin A metabolism, RA, and active RAR ligands generated by ALDH1A3. Together, our data identify YAP as a bona fide RAR-RXR transcriptional coactivator that acts through RARE-activated stemness genes. IMPLICATIONS: Targeting the newly identified YAP/RAR-RXR cross-talk implicated in cancer cell stemness maintenance may lead to multitarget combination therapies for patients with colorectal cancer.


Subject(s)
Cell Cycle Proteins/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Fluorouracil/pharmacology , Neoplastic Stem Cells/metabolism , Receptors, Retinoic Acid/metabolism , Retinoid X Receptors/metabolism , Transcription Factors/metabolism , Cell Line, Tumor , Cell Self Renewal/physiology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm , HT29 Cells , Humans , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Receptor Cross-Talk , Up-Regulation
4.
Mol Carcinog ; 58(11): 1985-1997, 2019 11.
Article in English | MEDLINE | ID: mdl-31373074

ABSTRACT

Growing body of evidence suggests that epithelial-mesenchymal transition (EMT) is a critical process in tumor progression and chemoresistance in pancreatic cancer (PC). The aim of this study was to analyze the role of EMT-like changes in acquisition of resistance to gemcitabine in pancreatic cells of the mesenchymal or epithelial phenotype. Therefore, chemoresistant BxPC-3, Capan-2, Panc-1, and MiaPaca-2 cells were selected by chronic exposure to increasing concentrations of gemcitabine. We show that gemcitabine-resistant Panc-1 and MiaPaca-2 cells of mesenchymal-like phenotype undergo further EMT-like molecular changes mediated by ERK-ZEB-1 pathway, and that inhibition of ERK1/2 phosphorylation or ZEB-1 expression resulted in a decrease in chemoresistance. Conversely, gemcitabine-resistant BxPC-3 and Capan-2 cells of epithelial-like phenotype did not show such typical EMT-like molecular changes although the expression of the tight junction marker occludin could be found decreased. In pancreatic cancer patients, high ZEB-1 expression was associated with tumor invasion and tumor budding. In addition, tumor budding was essentially observed in patients treated with neoadjuvant chemotherapy. These findings support the notion that gemcitabine treatment induces EMT-like changes that sustain invasion and chemoresistance in PC cells.


Subject(s)
Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/drug effects , Pancreatic Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/adverse effects , Deoxycytidine/pharmacology , Humans , MAP Kinase Signaling System/drug effects , Pancreas/drug effects , Pancreas/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phenotype , Zinc Finger E-box-Binding Homeobox 1/genetics , Gemcitabine
5.
Cancers (Basel) ; 10(5)2018 May 08.
Article in English | MEDLINE | ID: mdl-29738494

ABSTRACT

Intrinsically disordered protein YAP (yes-associated protein) interacts with TEADs transcriptional factors family (transcriptional enhancer associated domain) creating three interfaces. Interface 3, between the Ω-loop of YAP and a shallow pocket of TEAD was identified as the most important TEAD zone for YAP-TEAD interaction. Using the first X-ray structure of the hYAP50⁻71-hTEAD1209⁻426 complex (PDB 3KYS) published in 2010, a protein-protein interaction inhibitors-enriched library (175,000 chemical compounds) was screened against this hydrophobic pocket of TEAD. Four different chemical families have been identified and evaluated using biophysical techniques (thermal shift assay, microscale thermophoresis) and in cellulo assays (luciferase activity in transfected HEK293 cells, RTqPCR in MDA-MB231 cells). A first promising hit with micromolar inhibition in the luciferase gene reporter assay was discovered. This hit also decreased mRNA levels of TEAD target genes.

6.
Mol Genet Metab ; 123(4): 463-471, 2018 04.
Article in English | MEDLINE | ID: mdl-29478817

ABSTRACT

Creatine transporter is currently the focus of renewed interest with emerging roles in brain neurotransmission and physiology, and the bioenergetics of cancer metastases. We here report on amendments of a standard creatine uptake assay which might help clinical chemistry laboratories to extend their current range of measurements of creatine and metabolites in body fluids to functional enzyme explorations. In this respect, short incubation times and the use of a stable-isotope-labeled substrate (D3-creatine) preceded by a creatine wash-out step from cultured fibroblast cells by removal of fetal bovine serum (rich in creatine) from the incubation medium are recommended. Together, these measures decreased, by a first order of magnitude, creatine concentrations in the incubation medium at the start of creatine-uptake studies and allowed to functionally discriminate between 4 hemizygous male and 4 heterozygous female patients with X-linked SLC6A8 deficiency, and between this cohort of eight patients and controls. The functional assay corroborated genetic diagnosis of SLC6A8 deficiency. Gene anomalies in our small cohort included splicing site (c.912G > A [p.Ile260_Gln304del], c.778-2A > G and c.1495 + 2 T > G), substitution (c.407C > T) [p.Ala136Val] and deletion (c.635_636delAG [p.Glu212Valfs*84] and c.1324delC [p.Gln442Lysfs*21]) variants with reduced creatine transporter function validating their pathogenicity, including that of a previously unreported c.1324delC variant. The present assay adaptations provide an easy, reliable and discriminative manner for exploring creatine transporter activity and disease variations. It might apply to drug testing or other evaluations in the genetic and metabolic horizons covered by the emerging functions of creatine and its transporter, in a way, however, requiring and completed by additional studies on female patients and blood-brain barrier permeability properties of selected compounds. As a whole, the proposed assay of creatine transporter positively adds to currently existing measurements of this transporter activity, and determining on a large scale the extent of its exact suitability to detect female patients should condition in the future its transfer in clinical practice.


Subject(s)
Brain Diseases, Metabolic, Inborn/metabolism , Creatine/deficiency , Fibroblasts/metabolism , Mental Retardation, X-Linked/metabolism , Mutation , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Plasma Membrane Neurotransmitter Transport Proteins/deficiency , Adolescent , Brain Diseases, Metabolic, Inborn/genetics , Brain Diseases, Metabolic, Inborn/pathology , Case-Control Studies , Child , Child, Preschool , Cohort Studies , Creatine/genetics , Creatine/metabolism , Female , Fibroblasts/pathology , Follow-Up Studies , Humans , Infant , Male , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/pathology , Plasma Membrane Neurotransmitter Transport Proteins/genetics , Plasma Membrane Neurotransmitter Transport Proteins/metabolism , Prognosis
7.
ChemMedChem ; 12(12): 954-961, 2017 06 21.
Article in English | MEDLINE | ID: mdl-28334506

ABSTRACT

Porphyrin derivatives, in particular verteporfin (VP), a photosensitizer initially designed for cancer therapy, have been identified as inhibitors of the YAP-TEAD interaction and transcriptional activity. Herein we report the efficient convergent synthesis of the dipyrrin half of protoporphyrin IX dimethyl ester (PPIX-DME), in which the sensitive vinyl group was created at the final stage by a dehydroiodination reaction. Two other dipyrrin derivatives were synthesized, including dipyrrin 19 [(Z)-2-((3,5-dimethyl-4-vinyl-2H-pyrrol-2-ylidene)methyl)-3,5-dimethyl-4-vinyl-1H-pyrrole], containing two vinyl groups. We found that VP and dipyrrin 19 showed significant inhibitory effects on TEAD transcriptional activity in MDA-MB-231 human breast cancer cells, whereas other compounds did not show significant changes. In addition, we observed a marked decrease in both YAP and TAZ levels following VP treatment, whereas dipyrrin 19 treatment primarily decreased the levels of YAP and receptor kinase AXL, a downstream target of YAP. Together, our data suggest that, due to their chemical structures, porphyrin- and dipyrrin-related derivatives can directly target YAP and/or TAZ proteins and inhibit TEAD transcriptional activity.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Phosphoproteins/antagonists & inhibitors , Photosensitizing Agents/pharmacology , Porphyrins/chemical synthesis , Porphyrins/pharmacology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Transcription Factors/antagonists & inhibitors , Acyltransferases , Cell Line, Tumor , Hippo Signaling Pathway , Humans , Molecular Structure , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/chemistry , Porphyrins/chemistry , Structure-Activity Relationship , Verteporfin , YAP-Signaling Proteins
8.
Oncotarget ; 7(35): 56699-56712, 2016 Aug 30.
Article in English | MEDLINE | ID: mdl-27527859

ABSTRACT

Our aim was to decipher the role and clinical relevance of the YAP/TAZ transcriptional coactivators in the regulation of the proliferation/quiescence balance in human colon cancer cells (CCC) and survival after 5FU-based chemotherapy. The prognostic value of YAP/TAZ on tumor relapse and overall survival was assessed in a five-year follow-up study using specimens of liver metastases (n = 70) from colon cancer patients. In 5FU-chemoresistant HT29-5F31 and -chemosensitive HCT116 and RKO CCC, a reversible G0 quiescent state mediated by Cyclin E1 down-regulation was induced by 5FU in 5F31 cells and recapitulated in CCC by either YAP/TAZ or Cyclin E1 siRNAs or the YAP inhibitor Verteporfin. Conversely, the constitutive active YAPdc-S127A mutant restricted cellular quiescence in 5FU-treated 5F31 cells and sustained high Cyclin E1 levels through CREB Ser-133 phosphorylation and activation. In colon cancer patients, high YAP/TAZ level in residual liver metastases correlated with the proliferation marker Ki-67 (p < 0.0001), high level of the YAP target CTGF (p = 0.01), shorter disease-free and overall survival (p = 0.008 and 0.04, respectively). By multivariate analysis and Cox regression model, the YAP/TAZ level was an independent factor of overall (Hazard ratio [CI 95%] 2.06 (1.02-4.16) p = 0.045) and disease-free survival (Hazard ratio [CI 95%] 1.98 (1.01-3.86) p = 0.045). Thus, YAP/ TAZ pathways contribute to the proliferation/quiescence switch during 5FU treatment according to the concerted regulation of Cyclin E1 and CREB. These findings provide a rationale for therapeutic interventions targeting these transcriptional regulators in patients with residual chemoresistant liver metastases expressing high YAP/TAZ levels.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Colonic Neoplasms/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclin E/metabolism , Drug Resistance, Neoplasm , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasm Recurrence, Local , Oncogene Proteins/metabolism , Phosphoproteins/metabolism , Aged , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Disease-Free Survival , Fluorouracil/chemistry , Follow-Up Studies , HCT116 Cells , HT29 Cells , Humans , Ki-67 Antigen/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Middle Aged , Mutation , Neoplasm Metastasis , Porphyrins/pharmacology , Prognosis , Proportional Hazards Models , Trans-Activators , Transcription Factors/metabolism , Transcriptional Activation , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Verteporfin , YAP-Signaling Proteins
9.
Curr Med Chem ; 23(11): 1171-84, 2016.
Article in English | MEDLINE | ID: mdl-26980565

ABSTRACT

BACKGROUND: Verteporfin is a porphyrinic photosensitizer clinically used for the photodynamic treatment of age-related macular degeneration. It has been identified almost simultaneously as a YAP/TEAD and an autophagosome inhibitor. Over the last few years, YAP (TAZ), the downstream effectors of the Hippo pathway, have emerged as promising anticancer targets, as shown by several experimental lines of evidence, showing the overproduction of YAP in several cancers. However, YAP was also found to be closely connected to autophagy, mitochondria and reactive oxygen/nitrogen species. We herein, review the recent studies where VP was used without photoactivation as a YAP/TEAD inhibitor or protein oligomerization promoter, focusing on its effects on the YAP/TEAD gene targets and other biomarkers related to autophagy. RESULTS: Since the identification of VP as YAP/TEAD inhibitor, several in vitro and in vivo studies have revealed the new potential of this molecule in different cancers, where YAP is overexpressed. However, detailed structural information about its interaction with YAP is still lacking. Concomitantly, VP was identified as autophagosome inhibitor by promoting oligomerization of p62. Moreover, VP proves to be tumor-selective proteotoxic (by oligomerization of p62, STAT3) in colorectal cancer. Knowledge on the biological properties of the only YAP inhibitor available to date is vital for its pharmacological use on cellular and animal models. CONCLUSION: VP is a multi-target drug interacting with several proteins implicated in major cellular processes. Although this does not impact its clinical use, VP does not seem to be the ideal drug for pharmacological inhibitions of YAP/TEAD.


Subject(s)
Macular Degeneration/drug therapy , Photosensitizing Agents/therapeutic use , Porphyrins/therapeutic use , Animals , Humans , Verteporfin
10.
Cancer Sci ; 106(3): 262-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25580640

ABSTRACT

Our aim was to analyze the potential role of chemokine receptors CXCR2 and CXCR4 signalling pathways in liver metastatic colorectal cancer (CRC) relapse. CXCR2, CXCR4, and their chemokine ligands were evaluated in liver metastases of colorectal cancer in order to study their correlation with overall and disease-free survival of patients having received, or not received, a neoadjuvant chemotherapy regimen. Quantitative RT-PCR and CXCR2 immunohistochemical staining were carried out using CRC liver metastasis samples. Expression levels of CXCR2, CXCR4, and their ligands were statistically analyzed according to treatment with neoadjuvant chemotherapy and patients' outcome. CXCR2 and CXCL7 overexpression are correlated to shorter overall and disease-free survival. By multivariate analysis, CXCR2 and CXCL7 expressions are independent factors of overall and disease-free survival. Neoadjuvant chemotherapy increases significantly the expression of CXCR2: treated group 1.89 (0.02-50.92) vs 0.55 (0.07-3.22), P = 0.016. CXCL7 was overexpressed close to significance, 0.40 (0.00-7.85) vs 0.15 (0.01-7.88), P = 0.12. We show the involvement of CXCL7/CXCR2 signalling pathways as a predictive factor of poor outcome in metastatic CRC. 5-Fluorouracil-based chemotherapy regimens increase the expression of these genes in liver metastasis, providing one explanation for aggressiveness of relapsed drug-resistant tumors. Selective blockage of CXCR2/CXCL7 signalling pathways could provide new potential therapeutic opportunities.


Subject(s)
Colonic Neoplasms/mortality , Colonic Neoplasms/pathology , Liver Neoplasms/pathology , Receptors, Interleukin-8B/biosynthesis , beta-Thromboglobulin/biosynthesis , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Capecitabine , Colonic Neoplasms/drug therapy , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Disease-Free Survival , Female , Fluorouracil/analogs & derivatives , Fluorouracil/therapeutic use , Humans , Leucovorin/therapeutic use , Liver Neoplasms/mortality , Liver Neoplasms/secondary , Male , Middle Aged , Neoadjuvant Therapy , Neoplasm Recurrence, Local , Organoplatinum Compounds/therapeutic use , Receptors, CXCR4/biosynthesis , Receptors, Interleukin-8B/antagonists & inhibitors , Signal Transduction/genetics , beta-Thromboglobulin/antagonists & inhibitors
11.
HPB (Oxford) ; 17(2): 176-84, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25041611

ABSTRACT

BACKGROUND: The aim of the authors was to reassess the impact of a positive surgical margin (R1) after a liver resection for colorectal liver metastases (CLMs) on survival in the era of modern chemotherapy, through their own experience and a literature review. METHODS: Inclusion criteria were: R1 or R0 resection with no local treatment modalities, extra-hepatic metastases or other cancer. RESULTS: Among 337 patients operated between 2000 and 2010, 273 patients were eligible (214 R0/59 R1). The mean follow-up was 43 ± 29 months. Compared with a R0 resection, a R1 resection offered a lower 5-year overall (39.1% versus 54.2%, P = 0.010), disease-free (15.2% versus 31.1%, P = 0.021) and progression-free (i.e. time to the first non-curable recurrence; 33.1% versus 47.3%, P = 0.033) survival rates. Metastases in the R1 group were more numerous, larger and more frequently synchronous. Independent factors of poor survival were: number, size and short-time interval of CLM occurrence, N status, rectal primary, absence of adjuvant chemotherapy, but not a R1 resection. With the more-systematic administration of chemotherapy since 2005, the intergroup difference in progression-free survival disappeared (P = 0.264). CONCLUSION: A R1 resection had no prognostic value per se but reflected a more severe disease. The recent change in the prognostic value of a R1 resection may be linked to the beneficial effect of chemotherapy.


Subject(s)
Liver Neoplasms/mortality , Liver Neoplasms/pathology , Aged , Antineoplastic Agents/therapeutic use , Chemotherapy, Adjuvant , Colonic Neoplasms/pathology , Hepatectomy , Humans , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Middle Aged , Neoplasm Invasiveness , Neoplasm Recurrence, Local/epidemiology , Prognosis , Propensity Score , Rectal Neoplasms/pathology , Survival Analysis
12.
Clin Cancer Res ; 20(4): 837-46, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24323901

ABSTRACT

PURPOSE: Metastasis and drug resistance are the major limitations in the survival and management of patients with cancer. This study aimed to identify the mechanisms underlying HT29 colon cancer cell chemoresistance acquired after sequential exposure to 5-fluorouracil (5FU), a classical anticancer drug for treatment of epithelial solid tumors. We examined its clinical relevance in a cohort of patients with colon cancer with liver metastases after 5FU-based neoadjuvant chemotherapy and surgery. RESULTS: We show that a clonal 5F31 cell population, resistant to 1 µmol/L 5FU, express a typical cancer stem cell-like phenotype and enter into a reversible quiescent G0 state upon reexposure to higher 5FU concentrations. These quiescent cells overexpressed the tyrosine kinase c-Yes that became activated and membrane-associated upon 5FU exposure. This enhanced signaling pathway induced the dissociation of the Yes/YAP (Yes-associated protein) molecular complex and depleted nuclear YAP levels. Consistently, YES1 silencing decreased nuclear YAP accumulation and induced cellular quiescence in 5F31 cells cultured in 5FU-free medium. Importantly, YES1 and YAP transcript levels were higher in liver metastases of patients with colon cancer after 5FU-based neoadjuvant chemotherapy. Moreover, the YES1 and YAP transcript levels positively correlated with colon cancer relapse and shorter patient survival (P < 0.05 and P < 0.025, respectively). CONCLUSIONS: We identified c-Yes and YAP as potential molecular targets to eradicate quiescent cancer cells and dormant micrometastases during 5FU chemotherapy and resistance and as predictive survival markers for colon cancer.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Colonic Neoplasms/metabolism , Fluorouracil/pharmacology , Liver Neoplasms/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-yes/metabolism , Transcription Factors/metabolism , Antimetabolites, Antineoplastic/therapeutic use , Biomarkers, Tumor/metabolism , Cell Cycle Checkpoints , Cell Cycle Proteins , Cell Nucleus/metabolism , Cell Proliferation , Checkpoint Kinase 2/metabolism , Chemotherapy, Adjuvant , Colonic Neoplasms/drug therapy , Colonic Neoplasms/mortality , Colonic Neoplasms/pathology , Disease-Free Survival , Drug Resistance, Neoplasm , Fluorouracil/therapeutic use , Gene Expression , HT29 Cells , Humans , Kaplan-Meier Estimate , Liver Neoplasms/drug therapy , Liver Neoplasms/mortality , Liver Neoplasms/secondary , Neoadjuvant Therapy , Neoplasm Micrometastasis/prevention & control , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Nuclear Proteins/genetics , Proportional Hazards Models , Protein Transport , Proto-Oncogene Proteins c-yes/genetics , Transcription Factors/genetics
13.
Ann Surg ; 256(5): 755-61; discussion 761-2, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23095619

ABSTRACT

BACKGROUND: The objective was to determine the liver regeneration capacity and morbidity and mortality rates after major hepatectomy for colorectal metastases in patients having undergone bevacizumab-based chemotherapy (bev+). PATIENTS AND METHODS: Between 2006 and 2011, 41 patients underwent major hepatectomy within 3 months of bevacizumab and were matched with 41 patients operated on following systemic chemotherapy without bevacizumab (bev-). The matching criteria were the following: number of courses of chemotherapy, chemotherapy-free interval, age, and type of hepatectomy. After measurements of remnant liver volume (RLV) preoperatively and at 1 month (RLV1M), volumetric gain was calculated as absolute (RLV1M-RLV) or relative regeneration [(RLV1M-RLV/RLV)]. Ninety-day morbidity was rated according to the Clavien-Dindo classification. RESULTS: There were 21 right, 9 extended right, and 11 left hepatectomies in each group. Groups were comparable in terms of matching criteria, body mass index, American Society of Anesthesiologists score, and RLV. No mortalities were observed. There were no intergroup differences in overall morbidity (56% in bev+ vs 34.1%; P = 0.075) or postoperative liver failure. A severe complication occurred in 5 bev+ (4 eviscerations) and 4 bev- (bile leakages) (P = 0.95). The median hospital stay was similar in both groups as were the degrees of absolute and relative liver regeneration (143% in bev+ vs 114%; P = 0.20). Liver regeneration was not influenced by the type of hepatectomy, the number of courses of chemotherapy, or age more than 65 years. CONCLUSIONS: In a methodologically robust trial in the largest cohort reported up to date, bevacizumab did not impair liver regeneration after major hepatectomy-even in elderly patients or those with high exposure to chemotherapy.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Colorectal Neoplasms/pathology , Hepatectomy/methods , Liver Neoplasms/drug therapy , Liver Neoplasms/secondary , Liver Neoplasms/surgery , Liver Regeneration , Adult , Age Factors , Aged , Aged, 80 and over , Analysis of Variance , Bevacizumab , Case-Control Studies , Chi-Square Distribution , Combined Modality Therapy , Female , Humans , Length of Stay/statistics & numerical data , Male , Middle Aged , Postoperative Complications , Retrospective Studies , Statistics, Nonparametric , Survival Rate , Tomography, X-Ray Computed , Treatment Outcome
14.
Ann N Y Acad Sci ; 1253: 206-21, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22524425

ABSTRACT

Glycoprotein glycan chains, by virtue of structure, topology of presentation and connection to signal-inducing units, are functional galectin counterreceptors. As example, cross-linking of the α(5)ß(1) integrin by galectin-1 on carcinoma cells leads to G(1) arrest or anoikis. Contact-dependent switching from proliferation to differentiation in cultured neuroblastoma cells (SK-N-MC) also utilizes galectin-1. Activity enhancement of a cell surface sialidase underlies the shift in glycan display to ganglioside GM1. Its pentasaccharide within microdomains becomes the target. Similarly, this recognition pair is upregulated upon T cell activation. Cross-linking of GM1 along with associated α(4)/α(5)ß(1) integrins elicits Ca(2+)-influx via TRPC5 channels as the relevant response for T effector cell (T(eff)) suppression. Unlike T(eff) cells from wild-type mice, those from genetically altered mice lacking GM1 are not suppressed by galectin-1 or regulatory T cells. Similarly, in the context of GM1 deficiency in NOD mice, T(eff) cells are associated with resistance to regulatory T cell suppression, which is reversed by applied GM1. The broad array of glycosphingolipid structures suggests the possible existence of several novel counterreceptors targeted to endogenous lectins, with sulfatide-galectin-4 interplay within apical delivery serving as recent example.


Subject(s)
G(M1) Ganglioside/immunology , Galectins/immunology , Neoplasms/immunology , Neoplasms/therapy , Animals , Anoikis/immunology , Cell Communication/immunology , G(M1) Ganglioside/chemistry , Galectins/chemistry , Glycoproteins/chemistry , Glycoproteins/immunology , Humans , Indazoles , Mice , Models, Immunological , Morpholines , Neoplasms/pathology , Propionates , Signal Transduction/immunology , T-Lymphocytes/immunology
15.
HPB Surg ; 2011: 574378, 2011.
Article in English | MEDLINE | ID: mdl-21436987

ABSTRACT

Serous cystadenoma is a common benign neoplasm that can be managed without surgery in asymptomatic patients provided that the diagnosis is certain. We describe a patient, whose pancreatic cyst exhibited a radiological appearance distinct from that of typical serous cystadenoma, resulting in diagnostic difficulties. CT and MRI showed a 10 cm-polycystic tumor with upstream dilatation of the main pancreatic duct (MPD), suggestive of intraductal papillary mucinous tumor (IPMT). Ultrasonographic aspect and EUS-guided fine-needle aspiration gave arguments for serous cystadenoma. ERCP showed a communication between cysts and the dilated MPD, compatible with IPMT. The patient underwent left pancreatectomy with splenectomy. Pathological examination concluded in a serous cystadenoma, with only a ductal obstruction causing proximal dilatation.

16.
Cancer Res ; 70(11): 4644-54, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20460542

ABSTRACT

Metastasis and drug resistance are major problems in cancer chemotherapy. The purpose of this work was to analyze the molecular mechanisms underlying the invasive potential of drug-resistant colon carcinoma cells. Cellular models included the parental HT-29 cell line and its drug-resistant derivatives selected after chronic treatment with either 5-fluorouracil, methotrexate, doxorubicin, or oxaliplatin. Drug-resistant invasive cells were compared with noninvasive cells using cDNA microarray, quantitative reverse transcription-PCR, flow cytometry, immunoblots, and ELISA. Functional and cellular signaling analyses were undertaken using pharmacologic inhibitors, function-blocking antibodies, and silencing by retrovirus-mediated RNA interference. 5-Fluorouracil- and methotrexate-resistant HT-29 cells expressing an invasive phenotype in collagen type I and a metastatic behavior in immunodeficient mice exhibited high expression of the chemokine receptor CXCR4. Macrophage migration-inhibitory factor (MIF) was identified as the critical autocrine CXCR4 ligand promoting invasion in drug-resistant colon carcinoma HT-29 cells. Silencing of CXCR4 and impairing the MIF-CXCR4 signaling pathways by ISO-1, pAb FL-115, AMD-3100, monoclonal antibody 12G5, and BIM-46187 abolished this aggressive phenotype. Induction of CXCR4 was associated with the upregulation of two genes encoding transcription factors previously shown to control CXCR4 expression (HIF-2alpha and ASCL2) and maintenance of intestinal stem cells (ASCL2). Enhanced CXCR4 expression was detected in liver metastases resected from patients with colon cancer treated by the standard FOLFOX regimen. Combination therapies targeting the CXCR4-MIF axis could potentially counteract the emergence of the invasive metastatic behavior in clonal derivatives of drug-resistant colon cancer cells.


Subject(s)
Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Receptors, CXCR4/biosynthesis , Animals , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Fluorouracil/pharmacology , Gene Expression Profiling , Gene Silencing , HT29 Cells , Humans , Methotrexate/pharmacology , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Organoplatinum Compounds/pharmacology , Oxaliplatin , Phenotype , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Signal Transduction , Up-Regulation
17.
Int J Oncol ; 36(3): 689-98, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20126989

ABSTRACT

REG4, the latest member of the regenerating gene family, is overexpressed in inflammatory bowel diseases and gastrointestinal carcinomas. To date, its pathophysiologic role has not been well established. Using HT-29 models, we previously identified REG4 as being overexpressed in colorectal tumor cells displaying a drug-resistance phenotype; some also displayed invasive properties. Thus, we investigated the potential functions of REG4 in biological processes involved in colorectal tumor progression such as cell proliferation, migration and invasion. Colon cancer cells secreting REG4 (HT29-5M21, HT29-5F7 and HT29/REG4-8) or not (HT-29, HT29/CT1 and Caco-2/TC7) were used to analyze the autocrine and paracrine effects of REG4. REG4 was continuously secreted into the culture medium of colon cancer cells. REG4 stimulated cell growth in a paracrine manner after 24 h of treatment. Notably, REG4 promoted migration and invasion of tumor cells in both an autocrine and paracrine manner, and these effects were significantly decreased by concomitant treatment with an anti-REG4 antibody. Using pharmacological inhibitors, we showed that PI3K/Akt, PKAs, PKCs and Rho-like GTPases, but not MAPK, are involved in REG4 invasion signals. In addition, REG4 expression was found to be increased in tissues harboring proliferation and migration properties such as the developing intestine and tissues from inflammatory bowel disease, hyperplastic polyps, adenoma and colorectal cancers. In various situations, REG4 expression was not confined to proliferating cells, regenerating cells or cells of the invasive front of metastatic tumors, suggesting that extracellular REG4 may act on epithelial cells in a paracrine manner. Altogether, our results indicate that REG4 is a multifunctional secreted protein which acts on colorectal cancer cells in an autocrine and paracrine manner. According to its biological functions and tissue expression, REG4 may play an important role in the development and progression of colorectal cancer, as well as in intestinal morphogenesis and epithelium restitution.


Subject(s)
Colonic Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Lectins, C-Type/biosynthesis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Collagen/chemistry , Colon/embryology , Colon/pathology , Disease Progression , Epithelium/embryology , Epithelium/pathology , Gene Expression Regulation, Developmental , Humans , Mitogens , Neoplasm Invasiveness , Pancreatitis-Associated Proteins , Wound Healing
18.
Biol Chem ; 390(7): 529-44, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19426135

ABSTRACT

We have previously reported that galectin-4, a tandem repeat-type galectin, regulates the raft-dependent delivery of glycoproteins to the apical brush border membrane of enterocyte-like HT-29 cells. N-Acetyllactos-amine-containing glycans, known as galectin ligands, were found enriched in detergent-resistant membranes. Here, we analyzed the potential contribution of N- and/or O-glycans in this mechanism. Structural studies were carried out on the brush border membrane-enriched fraction using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS) and nano-ESI-QTOF-MS/MS. The pattern of N-glycans was very heterogeneous, with the presence of high mannose- and hybrid-type glycans as well as a multitude of complex-type glycans. In contrast, the pattern of O-glycans was very simple with the presence of two major core type 1 O-glycans, sialylated and bisialylated T-antigen structures [Neu5Acalpha2-3Galbeta1-3GalNAc-ol and Neu5Acalpha2- 3Galbeta1-3(Neu5Acalpha2-6)GalNAc-ol]. Thus, N-glycans rather than O-glycans contain the N-acetyllactosamine recognition signals for the lipid raft-based galectin-4-dependent apical delivery. In the presence of 1-deoxymannojirimycin, a drug which inhibits the generation of hybrid-type or complex type N-glycans, the extensively O-glycosylated mucin-like MUC1 glycoprotein was not delivered to the apical brush border but accumulated inside the cells. Altogether, our data demonstrate the crucial role of complex N-glycans in the galectin-4-dependent delivery of glycoproteins to the apical brush border membrane of enterocytic HT-29 cells.


Subject(s)
Enterocytes/cytology , Membrane Glycoproteins/metabolism , Microvilli/metabolism , Polysaccharides/metabolism , Carbohydrate Sequence , Epitopes/metabolism , Glycoside Hydrolases/metabolism , Glycosylation , HT29 Cells , Humans , Hydrofluoric Acid/metabolism , Mass Spectrometry , Membrane Glycoproteins/chemistry , Microscopy, Confocal , Molecular Sequence Data , Mucin-1/metabolism , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/metabolism , Polysaccharides/chemistry , Protein Transport
19.
Traffic ; 10(4): 438-50, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19192249

ABSTRACT

We have previously reported that silencing of galectin-4 expression in polarized HT-29 cells perturbed apical biosynthetic trafficking and resulted in a phenotype similar to the inhibitor of glycosylation, 1-benzyl-2-acetamido-2-deoxy-beta-d-galactopyranoside (GalNAcalpha-O-bn). We now present evidence of a lipid raft-based galectin-4-dependent mechanism of apical delivery of glycoproteins in these cells. First, galectin-4 recruits the apical glycoproteins in detergent-resistant membranes (DRMs) because these glycoproteins were depleted in DRMs isolated from galectin-4-knockdown (KD) HT-29 5M12 cells. DRM-associated glycoproteins were identified as ligands for galectin-4. Structural analysis showed that DRMs were markedly enriched in a series of complex N-glycans in comparison to detergent-soluble membranes. Second, in galectin-4-KD cells, the apical glycoproteins still exit the Golgi but accumulated inside the cells, showing that their recruitment within lipid rafts and their apical trafficking required the delivery of galectin-4 at a post-Golgi level. This lectin that is synthesized on free cytoplasmic ribosomes is externalized from HT-29 cells mostly in the apical medium and follows an apical endocytic-recycling pathway that is required for the apical biosynthetic pathway. Together, our data show that the pattern of N-glycosylation of glycoproteins serves as a recognition signal for endocytosed galectin-4, which drives the raft-dependent apical pathway of glycoproteins in enterocyte-like HT-29 cells.


Subject(s)
Cell Membrane/metabolism , Enterocytes/cytology , Galectin 4/metabolism , Glycoproteins/metabolism , Biomarkers/metabolism , Carbohydrate Conformation , Carbohydrate Sequence , Cell Polarity , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/metabolism , Enterocytes/metabolism , Glycoproteins/chemistry , Golgi Apparatus/metabolism , HT29 Cells , Humans , Membrane Microdomains/chemistry , Membrane Microdomains/metabolism , Molecular Sequence Data , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
20.
J Surg Res ; 150(2): 212-8, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18316097

ABSTRACT

BACKGROUND: Although E-cadherin and beta-catenin are key regulators in tumor invasion and proliferation, few studies have been undertaken on the expression of these genes at the messenger ribonucleic acid (mRNA) level in relation to the progression of colon cancer. PATIENTS AND METHODS: In this study, tissue samples from colectomy (n = 37) or hepatectomy (n = 23) were collected in both tumor and adjacent normal tissues. Real-time quantitative reverse transcriptase-polymerase chain reaction was used to quantify E-cadherin and beta-catenin mRNAs in reference to 18S RNA. RESULTS: E-cadherin and beta-catenin levels in colon carcinomas were not statistically different compared with adjacent normal mucosa and were not correlated with tumor, nodes, and metastases (TNM) stage. Conversely, E-cadherin and beta-catenin levels were significantly higher in liver metastases than in adjacent normal tissue. Interestingly, we found that E-cadherin level in liver metastases was correlated to the TNM stage of the related primary tumor: a higher E-cadherin level was found for State I-II TNM. In addition, a high expression of E-cadherin in liver metastases was associated with a lower occurrence of extra-hepatic metastases after resection of liver metastases. CONCLUSION: Taken together, these data show that E-cadherin and beta-catenin expressions are regulated throughout colon cancer progression.


Subject(s)
Cadherins/metabolism , Carcinoma/secondary , Colonic Neoplasms/pathology , Liver Neoplasms/secondary , beta Catenin/metabolism , Adult , Aged , Aged, 80 and over , Carcinoma/metabolism , Colonic Neoplasms/metabolism , Disease Progression , Female , Humans , Liver Neoplasms/metabolism , Male , Middle Aged , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...