Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters

Publication year range
1.
Cell ; 183(2): 411-428.e16, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32970988

ABSTRACT

The colon is primarily responsible for absorbing fluids. It contains a large number of microorganisms including fungi, which are enriched in its distal segment. The colonic mucosa must therefore tightly regulate fluid influx to control absorption of fungal metabolites, which can be toxic to epithelial cells and lead to barrier dysfunction. How this is achieved remains unknown. Here, we describe a mechanism by which the innate immune system allows rapid quality check of absorbed fluids to avoid intoxication of colonocytes. This mechanism relies on a population of distal colon macrophages that are equipped with "balloon-like" protrusions (BLPs) inserted in the epithelium, which sample absorbed fluids. In the absence of macrophages or BLPs, epithelial cells keep absorbing fluids containing fungal products, leading to their death and subsequent loss of epithelial barrier integrity. These results reveal an unexpected and essential role of macrophages in the maintenance of colon-microbiota interactions in homeostasis. VIDEO ABSTRACT.


Subject(s)
Gastrointestinal Microbiome/physiology , Intestinal Mucosa/metabolism , Macrophages/metabolism , Animals , Colon/metabolism , Epithelial Cells/metabolism , Epithelium , Female , Homeostasis , Immunity, Innate/immunology , Intestinal Mucosa/microbiology , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Microbiota , Signal Transduction
2.
Int J Mol Sci ; 24(17)2023 08 25.
Article in English | MEDLINE | ID: mdl-37686015

ABSTRACT

Gynaecological serous carcinomas (GSCs) constitute a distinctive entity among female tumours characterised by a very poor prognosis. In addition to late-stage diagnosis and a high rate of recurrent disease associated with massive peritoneal carcinomatosis, the systematic acquisition of resistance to first-line chemotherapy based on platinum determines the unfavourable outcome of GSC patients. To explore the molecular mechanisms associated with platinum resistance, we generated patient-derived organoids (PDOs) from liquid biopsies of GSC patients. PDOs are emerging as a relevant preclinical model system to assist in clinical decision making, mainly from tumoural tissue and particularly for personalised therapeutic options. To approach platinum resistance in a GSC context, proficient PDOs were generated from the ascitic fluid of ovarian, primary peritoneal and uterine serous carcinoma patients in platinum-sensitive and platinum-resistant clinical settings from the uterine aspirate of a uterine serous carcinoma patient, and we also induced platinum resistance in vitro in a representative platinum-sensitive PDO. Histological and immunofluorescent characterisation of these ascites-derived organoids showed resemblance to the corresponding original tumours, and assessment of platinum sensitivity in these preclinical models replicated the clinical setting of the corresponding GSC patients. Differential gene expression profiling of a panel of 770 genes representing major canonical cancer pathways, comparing platinum-sensitive and platinum-resistant PDOs, revealed cellular response to DNA damage stimulus as the principal biological process associated with the acquisition of resistance to the first-line therapy for GSC. Additionally, candidate genes involved in regulation of cell adhesion, cell cycles, and transcription emerged from this proof-of-concept study. In conclusion, we describe the generation of PDOs from liquid biopsies in the context of gynaecological serous carcinomas to explore the molecular determinants of platinum resistance.


Subject(s)
Ascites , Cystadenocarcinoma, Serous , Humans , Female , Organoids , Peritoneum , Ascitic Fluid , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/genetics
3.
J Cell Sci ; 132(6)2019 03 15.
Article in English | MEDLINE | ID: mdl-30765467

ABSTRACT

In the early stages of metastasis, cancer cells exit the primary tumor and enter the vasculature. Although most studies have focused on the tumor invasive front, cancer cells from the tumor core can also potentially metastasize. To address cell motility in the tumor core, we imaged tumor explants from spontaneously forming tumors in mice in real time using long-term two-photon microscopy. Cancer cells in the tumor core are remarkably dynamic and exhibit correlated migration patterns, giving rise to local 'currents' and large-scale tissue dynamics. Although cells exhibit stop-and-start migration with intermittent pauses, pausing does not appear to be required during division. Use of pharmacological inhibitors indicates that migration patterns in tumors are actively driven by the actin cytoskeleton. Under these conditions, we also observed a relationship between migration speed and correlation length, suggesting that cells in tumors are near a jamming transition. Our study provides new insight into the dynamics of cancer cells in the tumor core, opening new avenues of research in understanding the migratory properties of cancer cells and later metastasis.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Actin Cytoskeleton/pathology , Cell Movement , Neoplastic Cells, Circulating/pathology , Animals , Carcinogenesis/chemically induced , Mice , Microscopy, Fluorescence, Multiphoton , Neoplasms, Experimental , Primary Cell Culture , Tamoxifen/pharmacology
4.
Int J Cancer ; 145(7): 1852-1859, 2019 10 01.
Article in English | MEDLINE | ID: mdl-30714617

ABSTRACT

Anal squamous cell carcinoma (ASCC) is a rare tumour, but its incidence is increasing. Standard chemoradiotherapy fails to cure 20% of patients and no targeted therapy is currently approved for recurrent ASCC. The PI3K/Akt/mTOR pathway is frequently altered in this poorly characterised carcinoma. IGF2 was identified here as a key factor in ASCC oncogenesis, as IGF2 was shown to play a crucial role in the PI3K pathway with frequent (~60%) and mutually exclusive genomic alterations in IGF2, IGF1R, PTEN and PIK3CA genes. We also demonstrated that IGF2 expression is mainly due to cancer-associated fibroblasts and that IGF2 secreted by cancer-associated fibroblasts from ASCC samples promotes proliferation of a human ASCC cell line via IGF2 paracrine signalling. Altogether, these results highlight the key role of the IGF2/PI3K axis, and the major role of cancer-associated fibroblasts in tumour growth via IGF2 secretion, suggesting a major role of IGF2/IGF1R inhibitors in ASCC therapies.


Subject(s)
Anus Neoplasms/metabolism , Cancer-Associated Fibroblasts/metabolism , Carcinoma, Squamous Cell/metabolism , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Animals , Anus Neoplasms/genetics , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mutation , Neoplasm Transplantation , Paracrine Communication , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction
5.
Int J Mol Sci ; 18(6)2017 Jun 13.
Article in English | MEDLINE | ID: mdl-28608814

ABSTRACT

Colorectal cancer (CRC) is one of the major causes of cancer-related deaths. Early detection of tumor relapse is crucial for determining the most appropriate therapeutic management. In clinical practice, computed tomography (CT) is routinely used, but small tumor changes are difficult to visualize, and reliable blood-based prognostic and monitoring biomarkers are urgently needed. The aim of this study was to prospectively validate a gene expression panel (composed of GAPDH, VIL1, CLU, TIMP1, TLN1, LOXL3 and ZEB2) for detecting circulating tumor cells (CTCs) as prognostic and predictive tool in blood samples from 94 metastatic CRC (mCRC) patients. Patients with higher gene panel expression before treatment had a reduced progression-free survival (PFS) and overall-survival (OS) rates compared with patients with low expression (p = 0.003 and p ≤ 0.001, respectively). Patients with increased expression of CTCs markers during treatment presented PFS and OS times of 8.95 and 11.74 months, respectively, compared with 14.41 and 24.7 for patients presenting decreased expression (PFS; p = 0.020; OS; p ≤ 0.001). Patients classified as non-responders by CTCs with treatment, but classified as responders by CT scan, showed significantly shorter survival times (PFS: 8.53 vs. 11.70; OS: 10.37 vs. 24.13; months). In conclusion, our CTCs detection panel demonstrated efficacy for early treatment response assessment in mCRC patients, and with increased reliability compared to CT scan.


Subject(s)
Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Neoplastic Cells, Circulating/pathology , Biomarkers, Tumor/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Male , Neoplasm Metastasis/diagnosis , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Neoplasm Metastasis/therapy , Neoplastic Cells, Circulating/metabolism , Prognosis , Prospective Studies , Treatment Outcome
6.
Carcinogenesis ; 35(12): 2679-86, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25233929

ABSTRACT

Myometrial infiltration represents a main clinical determinant of endometrial carcinomas (EC) presenting as aggressive high-grade deeply invasive neoplasms, substantially associated with risk of recurrence and death. The up-regulation of ETV5 transcription factor linked to the promotion of epithelial to mesenchymal transition is considered as a basic mechanism underlying the initial steps of EC invasion. In this work, we aimed to investigate the transcription program of tumor invasion regulated by ETV5. We performed a comparative Chip-on-chip analysis at invasive front and superficial area of human EC. ETV5 specific binding to promoter regions of genes related to cellular migration, adhesion and invasion at deep invasion tumor areas highlighted the relevance of neural networks associated with cellular plasticity. Interestingly, brain-derived neurotrophic factor (BDNF) demonstrated a principal role orchestrating ETV5-mediated epithelial-to-mesenchymal transition in endometrial cancer. Impairment of the BDNF/tropomyosin-related kinase B (TrkB)/extracellular signal-regulated kinase axis in endometrial cancer cell lines reversed the aggressive and invasive phenotype promoted by the up-regulation of ETV5 at the invasive front of EC. Likewise, BDNF directly impacted on the efficiency of ETV5 promoted metastasis in a mice model of endometrial distant dissemination. These results translate the recognized role of BDNF/TrkB on neural plasticity into a relevant cancer metastasis event; suggest common mechanisms shared by neural development and tumor invasion; and offer new therapeutic opportunities specifically directed against disseminated disease in endometrial cancer.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cell Movement , DNA-Binding Proteins/genetics , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Transcription Factors/genetics , Animals , Blotting, Western , Brain-Derived Neurotrophic Factor/genetics , Cell Proliferation , Chromatin Immunoprecipitation , DNA-Binding Proteins/metabolism , Endometrial Neoplasms/genetics , Female , Fluorescent Antibody Technique , Humans , Mice , Neoplasm Invasiveness , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/metabolism , Tumor Cells, Cultured
7.
Int J Cancer ; 135(11): 2633-43, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-24752533

ABSTRACT

Circulating tumor cells (CTCs), proposed as major players in cancer dissemination, have demonstrated clinical prognostic significance in several cancer types. However, their predictive value remains unclear. Here we evaluated the clinical utility of six CTC markers (tissue specific and epithelial to mesenchymal transition transcripts) both as prognostic and predictive tools in metastatic colorectal cancer (mCRC) patients. CTCs were immunoisolated from blood in 50 mCRC patients at baseline and at 4 and 16 weeks after treatment onset. Expression levels of GAPDH, VIL1, CLU, TIMP1, LOXL3 and ZEB2 were determined by qualitative polymerase chain reaction and normalized to the unspecific cell isolation marker CD45. At baseline, median progression-free survival (PFS) and overall survival (OS) for patients with high CTC markers were 6.3 and 12.7 months, respectively, versus 12.7 and 24.2 for patients with low CTC markers (PFS; p = 0.0003; OS; p = 0.044). Concerning response to therapy, PFS and OS for patients with increased CTC markers along treatment were, respectively, 6.6 and 13.1 months, compared with 12.7 and 24.3 for patients presenting CTC markers reduction (PFS; p = 0.004; OS; p = 0.007). Of note, CTC markers identified therapy-refractory patients not detected by standard image techniques. Patients with increased CTC markers along treatment, but classified as responders by computed tomography, showed significantly shorter survival times (PFS: 7.8 vs. 13.2; OS: 14.4 vs. 24.4; months). In conclusion, we have generated a CTC marker panel for prognosis evaluation and the identification of patients benefiting or not from therapy in mCRC. Our methodology efficiently classified patients earlier than routine computed tomography and from a minimally invasive liquid biopsy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/analysis , Colorectal Neoplasms/blood , Liver Neoplasms/blood , Neoplastic Cells, Circulating/pathology , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/administration & dosage , Bevacizumab , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Capecitabine , Cetuximab , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Epithelial-Mesenchymal Transition , Female , Fluorouracil/administration & dosage , Fluorouracil/analogs & derivatives , Follow-Up Studies , Humans , Irinotecan , Liver Neoplasms/drug therapy , Liver Neoplasms/mortality , Liver Neoplasms/secondary , Male , Middle Aged , Neoplasm Staging , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Prognosis , Survival Rate
8.
Methods Mol Biol ; 2764: 179-203, 2024.
Article in English | MEDLINE | ID: mdl-38393596

ABSTRACT

Cells within a tumor interact by generating, transmitting, and sensing mechanical forces. Among all the cells of the tumor microenvironment, cancer-associated fibroblasts (CAFs) are a paradigmatic example of mechanical communication. In different steps of tumor progression, CAFs pull and push on cancer cells, regulating cancer cell migration, invasion, compartmentalization, and signaling. There is thus an increasing need to experimentally address mechanical interactions within a tumor. A common technique to measure these interactions is laser ablation. Cutting a tissue region with a high-power laser triggers a sudden tissue displacement whose direction and magnitude reveal the local mechanical stresses. In this chapter, we provide a detailed protocol to perform laser ablations in vitro and ex vivo. First, we describe how to prepare cocultures of primary CAFs and cancer cells and tumor explants. Then, we explain how to perform laser ablations in these two systems and how to analyze the induced tissue displacements using particle image velocimetry (PIV). Overall, we provide a workflow to perform, analyze, and interpret laser ablations to explore tumor mechanical interactions.


Subject(s)
Cancer-Associated Fibroblasts , Laser Therapy , Neoplasms , Humans , Cancer-Associated Fibroblasts/pathology , Fibroblasts/pathology , Neoplasms/pathology , Coculture Techniques , Tumor Microenvironment , Cell Line, Tumor , Cell Movement
9.
Biomed Pharmacother ; 173: 116345, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38442670

ABSTRACT

Antagonists of the A2B adenosine receptor have recently emerged as targeted anticancer agents and immune checkpoint inhibitors within the realm of cancer immunotherapy. This study presents a comprehensive evaluation of novel Biginelli-assembled pyrimidine chemotypes, including mono-, bi-, and tricyclic derivatives, as A2BAR antagonists. We conducted a comprehensive examination of the adenosinergic profile (both binding and functional) of a large compound library consisting of 168 compounds. This approach unveiled original lead compounds and enabled the identification of novel structure-activity relationship (SAR) trends, which were supported by extensive computational studies, including quantum mechanical calculations and free energy perturbation (FEP) analysis. In total, 25 molecules showed attractive affinity (Ki < 100 nM) and outstanding selectivity for A2BAR. From these, five molecules corresponding to the new benzothiazole scaffold were below the Ki < 10 nM threshold, in addition to a novel dual A2A/A2B antagonist. The most potent compounds, and the dual antagonist, showed enantiospecific recognition in the A2BAR. Two A2BAR selective antagonists and the dual A2AAR/A2BAR antagonist reported in this study were assessed for their impact on colorectal cancer cell lines. The results revealed a significant and dose-dependent reduction in cell proliferation. Notably, the A2BAR antagonists exhibited remarkable specificity, as they did not impede the proliferation of non-tumoral cell lines. These findings support the efficacy and potential that A2BAR antagonists as valuable candidates for cancer therapy, but also that they can effectively complement strategies involving A2AAR antagonism in the context of immune checkpoint inhibition.


Subject(s)
Antineoplastic Agents , Colorectal Neoplasms , Humans , Purinergic P1 Receptor Antagonists , Receptor, Adenosine A2B/metabolism , Adenosine A2 Receptor Antagonists/pharmacology , Structure-Activity Relationship , Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy
10.
Sci Adv ; 9(33): eade2120, 2023 08 18.
Article in English | MEDLINE | ID: mdl-37585527

ABSTRACT

Fibroblasts play a fundamental role in tumor development. Among other functions, they regulate cancer cells' migration through rearranging the extracellular matrix, secreting soluble factors, and establishing direct physical contacts with cancer cells. Here, we report that migrating fibroblasts deposit on the substrate a network of tubular structures that serves as a guidance cue for cancer cell migration. Such membranous tubular network, hereafter called tracks, is stably anchored to the substrate in a ß5-integrin-dependent manner. We found that cancer cells specifically adhere to tracks by using clathrin-coated structures that pinch and engulf tracks. Tracks thus represent a spatial memory of fibroblast migration paths that is read and erased by cancer cells directionally migrating along them. We propose that fibroblast tracks represent a topography-based intercellular communication system capable of steering cancer cell migration.


Subject(s)
Cues , Neoplasms , Humans , Cell Movement/physiology , Fibroblasts/physiology , Extracellular Matrix
11.
Nat Commun ; 14(1): 6966, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37907483

ABSTRACT

During tumor progression, cancer-associated fibroblasts (CAFs) accumulate in tumors and produce an excessive extracellular matrix (ECM), forming a capsule that enwraps cancer cells. This capsule acts as a barrier that restricts tumor growth leading to the buildup of intratumoral pressure. Combining genetic and physical manipulations in vivo with microfabrication and force measurements in vitro, we found that the CAFs capsule is not a passive barrier but instead actively compresses cancer cells using actomyosin contractility. Abrogation of CAFs contractility in vivo leads to the dissipation of compressive forces and impairment of capsule formation. By mapping CAF force patterns in 3D, we show that compression is a CAF-intrinsic property independent of cancer cell growth. Supracellular coordination of CAFs is achieved through fibronectin cables that serve as scaffolds allowing force transmission. Cancer cells mechanosense CAF compression, resulting in an altered localization of the transcriptional regulator YAP and a decrease in proliferation. Our study unveils that the contractile capsule actively compresses cancer cells, modulates their mechanical signaling, and reorganizes tumor morphology.


Subject(s)
Cancer-Associated Fibroblasts , Neoplasms , Cancer-Associated Fibroblasts/pathology , Mechanotransduction, Cellular , Cell Line, Tumor , Fibroblasts/pathology , Tumor Microenvironment , Neoplasms/pathology
12.
J Cell Mol Med ; 16(10): 2342-9, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22304365

ABSTRACT

The accuracy in the diagnosis of metastatic colorectal cancer (mCRC) represents one of the challenges in the clinical management of patients. The detection of circulating tumour cells (CTC) is becoming a promising alternative to current detection techniques, as it focuses on one of the players of the metastatic disease and it should provide with more specific and sensitive detection rates. Here, we describe an improved method of detection of CTC from mCRC patients by combining immune-enrichment, optimal purification of RNA from very low cell numbers, and the selection of accurate PCR probes. As a result, we obtained a logistic model that combines GAPDH and VIL1 normalized to CD45 rendering powerful results in the detection of CTC from mCRC patients (AUROC value 0.8599). We further demonstrated the utility of this model at the clinical setting, as a reliable prognosis tool to determine progression-free survival in mCRC patients. Overall, we developed a strategy that ameliorates the specificity and sensitivity in the detection of CTC, resulting in a robust and promising logistic model for the clinical management of metastatic colorectal cancer patients.


Subject(s)
Biomarkers, Tumor/analysis , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/pathology , Neoplastic Cells, Circulating/metabolism , Adult , Aged , Aged, 80 and over , Disease-Free Survival , Epithelial Cells/metabolism , Female , Humans , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/metabolism , Logistic Models , Male , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Middle Aged , Prognosis , Reproducibility of Results , Sensitivity and Specificity
13.
Biomed Pharmacother ; 153: 113408, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36076535

ABSTRACT

G-protein coupled receptors (GPCRs) have been largely targeted in a wide range of diseases, but few therapies have been directed against GPCRs in the field of cancer, partly because of the lack of effective target identification strategies. Here, using colorectal cancer (CRC) as a model, we explored the gene expression of a panel of GPCRs in tumor and stromal cells, identifying specific gene sets defining each cellular compartment. We selected the adenosine receptor 2B (A2BAR), specifically expressed in cancer cell lines compared with stromal cells, to explore the use of fluorescent ligands that can be used for target visualization. Fluorescent probes allowed semi-quantitative receptor mapping in living cells and validated the specific expression of A2BAR in CRC cell lines. As well, fluorescent ligands were effective at monitoring real-time A2BAR receptor labeling using live-imaging modalities, and displayed high efficiency when used to label complex 3D cellular systems such as tumor spheroids. Finally, we validated A2BAR as a potential pharmacological tool in CRC, using selective antagonists, finding a reduction in tumor cell proliferation. This proof-of-concept study suggests the use of fluorescent ligands for GPCR characterization through imaging, and as possible new tools used for target validation in drug screening methodologies.


Subject(s)
Colorectal Neoplasms , Receptors, G-Protein-Coupled , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Fluorescent Dyes , Humans , Ligands , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
14.
EMBO Mol Med ; 14(9): e15670, 2022 09 07.
Article in English | MEDLINE | ID: mdl-36069081

ABSTRACT

Centrosome amplification, the presence of more than two centrosomes in a cell is a common feature of most human cancer cell lines. However, little is known about centrosome numbers in human cancers and whether amplification or other numerical aberrations are frequently present. To address this question, we have analyzed a large cohort of primary human epithelial ovarian cancers (EOCs) from 100 patients. We found that rigorous quantitation of centrosome number in tumor samples was extremely challenging due to tumor heterogeneity and extensive tissue disorganization. Interestingly, even if centrosome clusters could be identified, the incidence of centrosome amplification was not comparable to what has been described in cultured cancer cells. Surprisingly, centrosome loss events where a few or many nuclei were not associated with centrosomes were clearly noticed and overall more frequent than centrosome amplification. Our findings highlight the difficulty of characterizing centrosome numbers in human tumors, while revealing a novel paradigm of centrosome number defects in EOCs.


Subject(s)
Centrosome , Ovarian Neoplasms , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/pathology , Cell Line , Centrosome/metabolism , Centrosome/pathology , Female , Humans , Ovarian Neoplasms/pathology
15.
Nat Commun ; 11(1): 404, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31964880

ABSTRACT

Although fibroblast heterogeneity is recognized in primary tumors, both its characterization in and its impact on metastases remain unknown. Here, combining flow cytometry, immunohistochemistry and RNA-sequencing on breast cancer samples, we identify four Cancer-Associated Fibroblast (CAF) subpopulations in metastatic lymph nodes (LN). Two myofibroblastic subsets, CAF-S1 and CAF-S4, accumulate in LN and correlate with cancer cell invasion. By developing functional assays on primary cultures, we demonstrate that these subsets promote metastasis through distinct functions. While CAF-S1 stimulate cancer cell migration and initiate an epithelial-to-mesenchymal transition through CXCL12 and TGFß pathways, highly contractile CAF-S4 induce cancer cell invasion in 3-dimensions via NOTCH signaling. Patients with high levels of CAFs, particularly CAF-S4, in LN at diagnosis are prone to develop late distant metastases. Our findings suggest that CAF subset accumulation in LN is a prognostic marker, suggesting that CAF subsets could be examined in axillary LN at diagnosis.


Subject(s)
Breast Neoplasms/pathology , Cancer-Associated Fibroblasts/metabolism , Lymphatic Metastasis/pathology , Myofibroblasts/metabolism , Adult , Aged , Aged, 80 and over , Axilla , Breast Neoplasms/mortality , Breast Neoplasms/therapy , Cancer-Associated Fibroblasts/pathology , Cell Proliferation , Cell Separation , Chemokine CXCL12/metabolism , Epithelial-Mesenchymal Transition , Female , Flow Cytometry , Follow-Up Studies , Humans , Kaplan-Meier Estimate , Lymph Nodes/cytology , Lymph Nodes/pathology , Middle Aged , Myofibroblasts/pathology , Neoplasm Invasiveness/pathology , Primary Cell Culture , Prognosis , Progression-Free Survival , Receptors, Notch/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Tumor Cells, Cultured , Tumor Microenvironment
16.
Curr Opin Cell Biol ; 56: 71-79, 2019 02.
Article in English | MEDLINE | ID: mdl-30308331

ABSTRACT

The most abundant cell type in the tumor microenvironment are cancer-associated fibroblasts (CAFs). CAFs play an important role in tumor growth and progression. Besides direct communication with cancer cells via secreted molecules or cell-cell adhesions, CAFs also indirectly affect cancer cell behavior by remodeling the extracellular matrix (ECM). Here, we summarize recent findings on the distinct mechanisms that CAFs use to modify ECM, specifically, their proteolytic versus force-dependent activity. We then review the consequences of CAF force transmission on the physico-chemical properties of the matrix, focusing on the deposition of new matrix components, and the alteration of the organization and stiffness of the ECM. CAFs promote tumor invasion by creating the roads cancer cells use to escape the tumor mass. However, there is also evidence that CAFs can prevent invasion, possibly by forming a physical barrier around the tumor edge. We discuss the controversial role of CAFs in tumor progression.


Subject(s)
Cancer-Associated Fibroblasts/pathology , Tumor Microenvironment , Animals , Cancer-Associated Fibroblasts/metabolism , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Humans , Neoplasms/pathology , Signal Transduction
17.
Sci Rep ; 9(1): 12124, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31431650

ABSTRACT

Previously, Aznar et al., showed that Daple/CCDC88C enables Wnt receptors to transactivate trimeric G-proteins during non-canonical Wnt signaling via a novel G-protein binding and activating (GBA) motif. By doing so, Daple serves two opposing roles; earlier during oncogenesis it suppresses neoplastic transformation and tumor growth, but later it triggers epithelial-to-mesenchymal-transition (EMT). We have identified and characterized two isoforms of the human Daple gene. While both isoforms cooperatively suppress tumor growth via their GBA motif, only the full-length transcript triggers EMT and invasion. Both isoforms are suppressed during colon cancer progression, and their reduced expression carries additive prognostic significance. These findings provide insights into the opposing roles of Daple during cancer progression and define the G-protein regulatory GBA motif as one of the minimal modules essential for Daple's role as a tumor suppressor.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Microfilament Proteins/metabolism , Neoplasms/metabolism , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , COS Cells , Cell Proliferation/physiology , Chlorocebus aethiops , Cohort Studies , Colon/metabolism , Genes, Tumor Suppressor , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Microfilament Proteins/genetics , NIH 3T3 Cells , Neoplasms/genetics , Protein Binding , Protein Isoforms/metabolism , RNA, Messenger/metabolism
18.
Cancers (Basel) ; 11(10)2019 Oct 19.
Article in English | MEDLINE | ID: mdl-31635093

ABSTRACT

Epigenetic modifications of glyco-genes have been documented in different types of cancer and are tightly linked to proliferation, invasiveness, metastasis, and drug resistance. This study aims to investigate the diagnostic, prognostic, and therapy-response predictive value of the glyco-gene B4GALT1 in colorectal cancer (CRC) patients. A Kaplan-Meier analysis was conducted in 1418 CRC patients (GEO and TCGA datasets) to assess the prognostic and therapy-response predictive values of the aberrant expression and methylation status of B4GALT1. Quantitative methylation-specific PCR (QMSP) and droplet digital quantitative methylation-specific PCR (dd-QMSP) were respectively used to detect hypermethylated B4GALT1 in metastasis and plasma in four cohorts of metastatic CRC cases (mCRC). Both the downregulated expression and promoter hypermethylation of B4GALT1 have a negative prognostic impact on CRC. Interestingly a low expression level of B4GALT1 was significantly associated with poor cetuximab response (progression-free survival (PFS) p = 0.01) particularly in wild-type (WT)-KRAS patients (p = 0.03). B4GALT1 promoter was aberrantly methylated in liver and lung metastases. The detection of hypermethylated B4GALT1 in plasma of mCRC patients showed a highly discriminative receiver operating characteristic (ROC) curve profile (area under curve (AUC) value 0.750; 95% CI: 0.592-0.908, p = 0.008), clearly distinguishing mCRC patients from healthy controls. Based on an optimal cut-off value defined by the ROC analysis, B4GALT1 yield a 100% specificity and a 50% sensitivity. These data support the potential value of B4GALT1 as an additional novel biomarker for the prediction of cetuximab response, and as a specific and sensitive diagnostic circulating biomarker that can be detected in CRC.

19.
Methods Mol Biol ; 1749: 163-173, 2018.
Article in English | MEDLINE | ID: mdl-29525997

ABSTRACT

Cell migration is a process that ensures correct cell localization and function in development and homeostasis. In disease such as cancer, cells acquire an upregulated migratory capacity that leads to their dissemination throughout the body. Live imaging of cell migration allows for better understanding of cell behaviors in development, adult tissue homeostasis and disease. We have optimized live imaging procedures to track cell migration in adult murine tissue explants derived from: (1) healthy gut; (2) primary intestinal carcinoma; and (3) the liver, a common metastatic site. To track epithelial cell migration in the gut, we generated an inducible fluorescent reporter mouse, enabling us to visualize and track individual cells in unperturbed gut epithelium. To image intratumoral cancer cells, we use a spontaneous intestinal cancer model based on the activation of Notch1 and deletion of p53 in the mouse intestinal epithelium, which gives rise to aggressive carcinoma. Interaction of cancer cells with a metastatic niche, the mouse liver, is addressed using a liver colonization model. In summary, we describe a method for long-term 3D imaging of tissue explants by two-photon excitation microscopy. Explant culturing and imaging can help understand dynamic behavior of cells in homeostasis and disease, and would be applicable to various tissues.


Subject(s)
Cell Movement/physiology , Optical Imaging/methods , Organ Culture Techniques/methods , Animals , Cells, Cultured , Intestines/cytology , Liver/cytology , Liver Neoplasms/pathology , Mice
20.
Cancer Res ; 77(13): 3431-3441, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28536280

ABSTRACT

The interaction between circulating tumor cells (CTC) and endothelial cells during extravasation is a critical process during metastatic colonization, but its mechanisms remain poorly characterized. Here we report that the luminal side of liver blood vessels contains fibronectin deposits that are enriched in mice bearing primary tumors and are also present in vessels from human livers affected with metastases. Cancer cells attached to endothelial fibronectin deposits via talin1, a major component of focal adhesions. Talin1 depletion impaired cancer cell adhesion to the endothelium and transendothelial migration, resulting in reduced liver metastasis formation in vivo Talin1 expression levels in patient CTC's correlated with prognosis and therapy response. Together, our findings uncover a new mechanism for liver metastasis formation involving an active contribution of hepatic vascular fibronectin and talin1 in cancer cells. Cancer Res; 77(13); 3431-41. ©2017 AACR.


Subject(s)
Fibronectins/metabolism , Liver Neoplasms/blood , Liver Neoplasms/pathology , Neoplastic Cells, Circulating/pathology , Animals , Cell Adhesion/physiology , Cell Line, Tumor , Humans , Mice , Mice, Nude , Neoplasm Metastasis , Transendothelial and Transepithelial Migration
SELECTION OF CITATIONS
SEARCH DETAIL