Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
J Pathol ; 258(4): 408-425, 2022 12.
Article in English | MEDLINE | ID: mdl-36102377

ABSTRACT

Cancer-associated fibroblasts (CAFs) are orchestrators of the pancreatic ductal adenocarcinoma (PDAC) microenvironment. Previously we described four CAF subtypes with specific molecular and functional features. Here, we have refined our CAF subtype signatures using RNAseq and immunostaining with the goal of defining bioinformatically the phenotypic stromal and tumor epithelial states associated with CAF diversity. We used primary CAF cultures grown from patient PDAC tumors, human data sets (in-house and public, including single-cell analyses), genetically engineered mouse PDAC tissues, and patient-derived xenografts (PDX) grown in mice. We found that CAF subtype RNAseq signatures correlated with immunostaining. Tumors rich in periostin-positive CAFs were significantly associated with shorter overall survival of patients. Periostin-positive CAFs were characterized by high proliferation and protein synthesis rates and low α-smooth muscle actin expression and were found in peri-/pre-tumoral areas. They were associated with highly cellular tumors and with macrophage infiltrates. Podoplanin-positive CAFs were associated with immune-related signatures and recruitment of dendritic cells. Importantly, we showed that the combination of periostin-positive CAFs and podoplanin-positive CAFs was associated with specific tumor microenvironment features in terms of stromal abundance and immune cell infiltrates. Podoplanin-positive CAFs identified an inflammatory CAF (iCAF)-like subset, whereas periostin-positive CAFs were not correlated with the published myofibroblastic CAF (myCAF)/iCAF classification. Taken together, these results suggest that a periostin-positive CAF is an early, activated CAF, associated with aggressive tumors, whereas a podoplanin-positive CAF is associated with an immune-related phenotype. These two subpopulations cooperate to define specific tumor microenvironment and patient prognosis and are of putative interest for future therapeutic stratification of patients. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Adenocarcinoma , Cancer-Associated Fibroblasts , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Mice , Animals , Cancer-Associated Fibroblasts/pathology , Pancreatic Neoplasms/pathology , Tumor Microenvironment , Adenocarcinoma/metabolism , Carcinoma, Pancreatic Ductal/genetics , Pancreatic Neoplasms
2.
Int J Mol Sci ; 23(4)2022 Feb 19.
Article in English | MEDLINE | ID: mdl-35216426

ABSTRACT

As is the case with most eucaryotic cells, cancer cells are able to secrete extracellular vesicles (EVs) as a communication means towards their environment and surrounding cells. EVs are represented by microvesicles and smaller vesicles called exosomes, which are known for their involvement in cancer aggressiveness. The release of such EVs requires the intervention of trafficking-associated proteins, mostly represented by the RAB-GTPases family. In particular, RAB27A is known for its role in addressing EVs-to-be secreted towards the the plasma membrane. In this study, shRNAs targeting RAB27A were used in colorectal (CRC) and glioblastoma (GB) cell lines in order to alter EVs secretion. To study and monitor EVs secretion in cell lines' supernatants, nanoparticle tracking analysis (NTA) was used through the NanoSight NS300 device. Since it appeared that NanoSight failed to detect the decrease in the EVs secretion, we performed another approach to drop EVs secretion (RAB27A-siRNA, indomethacin, Nexihnib20). Similar results were obtained i.e., no variation in EVs concentration. Conversely, NTA allowed us to monitor EVs up-secretion following rotenone treatment or hypoxia conditions. Therefore, our data seemed to point out the insufficiency of using only this technique for the assessment of EVs secretion decrease.


Subject(s)
Biotechnology/methods , Extracellular Vesicles/metabolism , Nanoparticles/metabolism , Cell Line, Tumor , Cell-Derived Microparticles/metabolism , Exosomes/metabolism , HCT116 Cells , Humans , Neoplasms/metabolism , Protein Transport/physiology
3.
Cancers (Basel) ; 13(5)2021 Mar 02.
Article in English | MEDLINE | ID: mdl-33801266

ABSTRACT

Autophagy is a homeostatic process involved in the degradation of disabled proteins and organelles using lysosomes. This mechanism requires the recruitment of specialized proteins for vesicle trafficking, that may also be involved in other types of machinery such as the biogenesis and secretion of extracellular vesicles (EVs), and particularly small EVs called exosomes. Among these proteins, Rab-GTPases may operate in both pathways, thus representing an interesting avenue for further study regarding the interaction between autophagy and extracellular vesicle machinery. Both mechanisms are involved in the development of colorectal cancer (CRC), particularly in cancer stem cell (CSC) survival and communication, although they are not specific to CRC or CSCs. This highlights the importance of studying the crosstalk between autophagy and EVs biogenesis and release.

4.
Cell Mol Gastroenterol Hepatol ; 11(5): 1405-1436, 2021.
Article in English | MEDLINE | ID: mdl-33482394

ABSTRACT

BACKGROUND & AIMS: Cancer-associated fibroblasts (CAFs) from pancreatic adenocarcinoma (PDA) present high protein synthesis rates. CAFs express the G-protein-coupled somatostatin receptor sst1. The sst1 agonist SOM230 blocks CAF protumoral features in vitro and in immunocompromised mice. We have explored here the therapeutic potential of SOM230, and underlying mechanisms, in immunocompetent models of murine PDA mimicking the heavy fibrotic and immunosuppressive stroma observed in patient tumors. METHODS: Large-scale mass spectrometry analyses were performed on media conditioned from 9 patient PDA-derived CAF primary cultures. Spontaneous transgenic and experimental (orthotopic co-graft of tumor cells plus CAFs) PDA-bearing mice were longitudinally ultrasound-monitored for tumor and metastatic progression. Histopathology and flow cytometry analyses were performed on primary tumors and metastases. Stromal signatures were functionally validated through bioinformatics using several published, and 1 original, PDA database. RESULTS: Proteomics on the CAF secretome showed that SOM230 controls stromal activities including inflammatory responses. Among the identified secreted proteins, we validated that colony-stimulating factor 1 (CSF-1) (a macrophage growth factor) was reduced by SOM230 in the tumor and plasma of PDA-harboring mice, alongside intratumor stromal normalization (reduced CAF and macrophage activities), and dramatic metastasis reduction. In transgenic mice, these SOM230 benefits alleviate the chemotherapy-induced (gemcitabine) immunosuppressive stroma reshaping. Mechanistically, SOM230 acts in vivo on CAFs through sst1 to disrupt prometastatic CAF production of CSF-1 and cross-talk with macrophages. We found that in patients, stromal CSF-1 was associated with aggressive PDA forms. CONCLUSIONS: We propose SOM230 as an antimetastatic therapy in PDA for its capacity to remodel the fibrotic and immunosuppressive myeloid stroma. This pharmacotherapy should benefit PDA patients treated with chemotherapies.


Subject(s)
Cancer-Associated Fibroblasts/drug effects , Carcinoma, Pancreatic Ductal/drug therapy , Macrophages/drug effects , Pancreatic Neoplasms/drug therapy , Secretome/drug effects , Somatostatin/analogs & derivatives , Aged , Aged, 80 and over , Animals , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/secondary , Female , Hormones/pharmacology , Humans , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Somatostatin/pharmacology
5.
J Cell Mol Med ; 25(1): 47-60, 2021 01.
Article in English | MEDLINE | ID: mdl-33325631

ABSTRACT

Colorectal cancer (CRC) is the third most common cancer worldwide. Even if 5-fluorouracil (5-FU) is used as the first-line chemotherapeutic drug, responsiveness is only 20-30%. Acquired resistance to 5-FU contributes to both poor patient prognosis and relapse, emphasizing the need to identify biomarkers. Sortilin, a vacuolar protein sorting 10 protein (Vps10p), implicated in protein trafficking, is over expressed in CRC cell lines cultured 72 hours in presence of 5-FU. This overexpression was also observed in 5-FU-resistant cells derived from these cell lines as well as in CRC primary cultures (or patients derived cell lines). A significantly higher expression of sortilin was observed in vivo, in 5-FU-treated tumours engrafted in Nude mice, as compared with non-treated tumour. A study of transcriptional regulation allowed identifying a decrease in ATF3 expression, as an explanation of sortilin overexpression following 5-FU treatment. In silico analysis revealed SORT1 expression correlation with poor prognosis. Moreover, sortilin expression was found to be positively correlated with CRC tumour grades. Collectively, our findings identify sortilin as a potential biomarker of 5-FU resistance associated with poor clinical outcomes and aggressiveness in CRC. As a new prognostic factor, sortilin expression could be used to fight against CRC.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm , Fluorouracil/therapeutic use , Adaptor Proteins, Vesicular Transport/genetics , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Disease-Free Survival , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice, Nude , Neoplasm Grading , Prognosis , Protein Transport/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Treatment Outcome , Xenograft Model Antitumor Assays
6.
Cell Death Dis ; 11(12): 1027, 2020 12 02.
Article in English | MEDLINE | ID: mdl-33268796

ABSTRACT

Initially, NEUROTENSIN (NTS) has been shown to play physiological and biological functions as a neuro-transmitter/modulator in the central nervous system and as an endocrine factor in the periphery, through its binding to two kinds of receptors: NTSR1 and 2 (G protein-coupled receptors) and NTSR3/sortilin (a vacuolar protein-sorting 10-domain receptor). NTS also plays oncogenic roles in many types of cancer, including digestive cancers. In tumor tissues, NTS and NTSR1 expression is higher than in healthy ones and is associated with poor prognosis. NTS and NTRS1 promote cancer progression and play key functions in metastatic processes; they modulate several signaling pathways and they contribute to changes in the tumor microenvironment. Conversely, NTRS2 involvement in digestive cancers is poorly understood. Discovered for mediating NTS biological effects, sortilin recently emerged as a promising target as its expression was found to be increased in various types of cancers. Because it can be secreted, a soluble form of sortilin (sSortilin) appears as a new serum biomarker which, on the basis of recent studies, promises to be useful in both the diagnosis and tumor progression monitoring. More precisely, it appears that soluble sortilin can be associated with other receptors like TRKB. These associations occur in exosomes and trigger the aggressiveness of cancers like glioblastoma, leading to the concept of a possible composite theranostic biomarker. This review summarizes the oncogenic roles of the NTS signaling pathways in digestive cancers and discusses their emergence as promising early diagnostic and/or prognostic biomarkers and therapeutic targets.


Subject(s)
Gastrointestinal Neoplasms/metabolism , Neurotensin/metabolism , Signal Transduction , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/pathology , Humans , Models, Biological , Oncogenes , Receptors, Neurotensin/metabolism
7.
EMBO Mol Med ; 12(11): e12010, 2020 11 06.
Article in English | MEDLINE | ID: mdl-33025708

ABSTRACT

Cancer-associated fibroblasts (CAFs) are considered the most abundant type of stromal cells in pancreatic ductal adenocarcinoma (PDAC), playing a critical role in tumour progression and chemoresistance; however, a druggable target on CAFs has not yet been identified. Here we report that focal adhesion kinase (FAK) activity (evaluated based on 397 tyrosine phosphorylation level) in CAFs is highly increased compared to its activity in fibroblasts from healthy pancreas. Fibroblastic FAK activity is an independent prognostic marker for disease-free and overall survival of PDAC patients (cohort of 120 PDAC samples). Genetic inactivation of FAK within fibroblasts (FAK kinase-dead, KD) reduces fibrosis and immunosuppressive cell number within primary tumours and dramatically decreases tumour spread. FAK pharmacologic or genetic inactivation reduces fibroblast migration/invasion, decreases extracellular matrix (ECM) expression and deposition by CAFs, modifies ECM track generation and negatively impacts M2 macrophage polarization and migration. Thus, FAK activity within CAFs appears as an independent PDAC prognostic marker and a druggable driver of tumour cell invasion.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Carcinoma, Pancreatic Ductal/drug therapy , Cell Line, Tumor , Fibroblasts , Humans , Phosphorylation , Prognosis
8.
Cancer Sci ; 111(9): 3142-3154, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32536012

ABSTRACT

Colorectal cancer (CRC) is a public health problem. It is the third most common cancer in the world, with nearly 1.8 million new cases diagnosed in 2018. The only curative treatment is surgery, especially for early tumor stages. When there is locoregional or distant invasion, chemotherapy can be introduced, in particular 5-fluorouracil (5-FU). However, the disease can become tolerant to these pharmaceutical treatments: resistance emerges, leading to early tumor recurrence. Different mechanisms can explain this 5-FU resistance. Some are disease-specific, whereas others, such as drug efflux, are evolutionarily conserved. These mechanisms are numerous and complex and can occur simultaneously in cells exposed to 5-FU. In this review, we construct a global outline of different mechanisms from disruption of 5-FU-metabolic enzymes and classic cellular processes (apoptosis, autophagy, glucose metabolism, oxidative stress, respiration, and cell cycle perturbation) to drug transporters and epithelial-mesenchymal transition induction. Particular interest is directed to tumor microenvironment function as well as epigenetic alterations and miRNA dysregulation, which are the more promising processes that will be the subject of much research in the future.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Colorectal Neoplasms/metabolism , Drug Resistance, Neoplasm , Fluorouracil/pharmacology , Signal Transduction/drug effects , Antimetabolites, Antineoplastic/metabolism , Biomarkers , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Energy Metabolism/drug effects , Epigenesis, Genetic/drug effects , Fluorouracil/metabolism , Gene Expression Regulation, Neoplastic , Humans , Metabolic Networks and Pathways , Oxidative Stress/drug effects , Thymidylate Synthase/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics
9.
Cell Death Dis ; 10(2): 123, 2019 02 11.
Article in English | MEDLINE | ID: mdl-30741921

ABSTRACT

Cancers of the digestive system, including esophageal, gastric, pancreatic, hepatic, and colorectal cancers, have a high incidence and mortality worldwide. Efficient therapies have improved patient care; however, many challenges remain including late diagnosis, disease recurrence, and resistance to therapies. Mechanisms responsible for these aforementioned challenges are numerous. This review focuses on neurotrophins, including NGF, BDNF, and NT3, and their specific tyrosine kinase receptors called tropomyosin receptor kinase (Trk A, B, C, respectively), associated with sortilin and the p75 neurotrophin receptor (p75NTR), and their implication in digestive cancers. Globally, p75NTR is a frequently downregulated tumor suppressor. On the contrary, Trk and their ligands are considered oncogenic factors. New therapies which target NT and/or their receptors, or use them as diagnosis biomarkers could help us to combat digestive cancers.


Subject(s)
Neoplasms/pathology , Nerve Growth Factors/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/metabolism , Nerve Growth Factors/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , Signal Transduction
10.
Oncotarget ; 8(49): 85185-85202, 2017 Oct 17.
Article in English | MEDLINE | ID: mdl-29156712

ABSTRACT

By radiolabelling monomeric (m) and polymeric (p) IgA with technetium 99m (99mTc), this study assessed IgA biodistribution and tumour-targeting potency. IgA directed against carcinoembryonic antigen (CEA), a colorectal cancer marker, was selected to involve IgA mucosal tropism. Ig was radiolabelled with 99mTc-tricarbonyl after derivatisation by 2-iminothiolane. 99mTc-IgA was evaluated by in vitro analysis. The biodistributions of radiolabelled anti-CEA mIgA, pIgA and IgG were compared in normal mice. Anti-CEA pIgA tumour uptake was studied in mice bearing the WiDr caecal orthotopic graft. IgA radiolabelling was obtained with a high yield, was stable in PBS and murine plasma, and did not alter IgA binding functionality (Kd ≈ 25 nM). Biodistribution studies in normal mice confirmed that radiolabelled pIgA - and to a lesser extent, mIgA - showed strong and fast mucosal tropism and a shorter serum half-life than IgG. In caecal tumour model mice, evaluation of the anti-CEA-pIgA biodistribution showed a high uptake in lung metastases, confirmed by histological analysis. However, no radioactivity uptake increase in the tumoural caecum was discerned from normal intestinal tissue, probably due to high IgA caecal natural tropism. In microSPECT/CT imaging, 99mTc-IgA confirmed its diagnostic potency of tumour in mucosal tissue, even if detection threshold by in vivo imaging was higher than post mortem studies. Contribution of the FcαRI receptor, studied with transgenic mouse model (Tsg SCID-CD89), did not appear to be determinant in 99mTc-IgA uptake. Pre-clinical experiments highlighted significant differences between 99mTc-IgA and 99mTc-IgG biodistributions. Furthermore, tumoural model studies suggested potential targeting potency of pIgA in mucosal tissues.

11.
Oncotarget ; 8(49): 85969-85983, 2017 Oct 17.
Article in English | MEDLINE | ID: mdl-29156770

ABSTRACT

Angiogenesis plays a critical role in glioblastoma growth and progression. We therefore aimed at evaluating the anti-angiogenic properties of an oligopeptide originating from SCO-spondin (NX) on a model of human glioblastoma. To this end, we studied the impact of NX treatment on human brain endothelial cells (HBMECs) alone or co-cultured with glioblastoma cells (U87-MG) on apoptosis, proliferation, migration and release of angiogenic factors. We further investigated the anti-angiogenic potential of NX on human glioblastoma cells grown on chorio-allantoic membrane (CAM) or in glioblastoma xenografts. The results of our experiments showed that NX treatment impaired the microvascular network and induced a decrease in cell proliferation, vascularization and tumor growth in the CAM model as well as in xenotransplants. Interestingly, our in vitro experiments showed that NX impairs HBMECs migration but also regulates the release of angiogenic factors from U87-MG. These results are confirmed by the profiling of NX-treated U87-MG grown on CAM that highlighted modifications of several genes involved in angiogenesis. In conclusion, NX inhibits tumorigenesis by impairing the ability of glioblastoma cells to induce angiogenesis and by inhibiting endothelial cell migration. This molecule might therefore be an interesting candidate for future cancer therapies.

12.
Oncol Lett ; 13(6): 4571-4576, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28588719

ABSTRACT

Colorectal cancer (CRC) is a common and lethal disease. It is the third most common type of cancer in the world, behind lung and breast cancer, with almost 1.4 million new cases diagnosed in 2012. The risk of developing CRC is influenced by environmental and genetic factors. Adenocarcinomas comprise the vast majority (98%) of CRCs. A patient's likelihood of survival is associated with the tumor stage at the time of diagnosis. With regular screening, CRC can be identified early, when treatment is the most effective. However, CRC is typically asymptomatic until the advanced stages. The combination of the absence of symptoms and current screening methodology results in a significant number of patients being diagnosed in advanced stages. The purpose of the present review is to discuss and summarize the biomarkers linked to CRC progression, particularly the controversial E-cadherin, which is a calcium-dependent cell-cell adhesion molecule involved in the mesenchymal-epithelial transition.

13.
J Cell Mol Med ; 21(10): 2610-2622, 2017 10.
Article in English | MEDLINE | ID: mdl-28597984

ABSTRACT

Colorectal cancer (CRC) is the most common digestive cancer in the Western world. Despite effective therapies, resistance and/or recurrence frequently occur. The present study investigated the impact of two survival pathways-neurotrophic factors (TrkB/BDNF) and autophagy-on cell fate and tumour evolution. In vitro studies were performed on two CRC cell lines, SW480 (primary tumour) and SW620 (lymph node invasion), which were also used for subcutaneous xenografts on a nude mouse model. In addition, the presence of neurotrophic factors (NTs) and autophagy markers were assessed in tissue samples representative of different stages. On the basis of our previous study (which demonstrated that TrkB overexpression is associated with prosurvival signaling in CRC cells), we pharmacologically inhibited NTs pathways with K252a. As expected, an inactivation of the PI3K/AKT pathway was observed and CRC cells initiated autophagy. Conversely, blocking the autophagic flux with chloroquine or with ATG5-siRNA overactivated TrkB/BDNF signaling. In vitro, dual inhibition improved the effectiveness of single treatment by significantly reducing metabolic activity and enhancing apoptotic cell death. These findings were accentuated in vivo, in which dual inhibition induced a spectacular reduction in tumour volume following long-term treatment (21 days for K252a and 12 days for CQ). Finally, significant amounts of phospho-TrkB and LC3II were found in the patients' tissues, highlighting their relevance in CRC tumour biology. Taken together, our results show that targeting NTs and autophagy pathways potentially constitutes a new therapeutic approach for CRC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Autophagy/drug effects , Brain-Derived Neurotrophic Factor/antagonists & inhibitors , Colorectal Neoplasms/drug therapy , Receptor, trkB/antagonists & inhibitors , Xenograft Model Antitumor Assays , Aged , Aged, 80 and over , Animals , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Carbazoles/administration & dosage , Cell Line, Tumor , Chloroquine/administration & dosage , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Enzyme Inhibitors/administration & dosage , Female , Humans , Indole Alkaloids/administration & dosage , Male , Mice, Inbred Strains , Mice, Nude , RNA Interference , Receptor, trkB/metabolism , Retrospective Studies , Tumor Burden/drug effects
14.
J Cancer Res Clin Oncol ; 143(7): 1177-1190, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28289897

ABSTRACT

PURPOSE: Multiple studies have attempted to demonstrate the interest of the cell adhesion marker, E cadherin, as a diagnostic and prognosis marker in colorectal cancer (CRC). However, it was considered non specific. MATERIALS AND METHODS: Studies were carried out with CRC cell lines and patients' cohort operated for CRC. The expression of E cadherin was studied after 5 fluorouracil (5FU) treatment and correlated to CRC relapse, chemo-resistance and survival. RESULTS: In CRC cell lines derived from high tumor stages, extracellular domain of E cadherin expression decreased after 5FU treatment whereas it increased in supernatants. Interestingly, only specific cleaved forms at 55 kDa of E cadherin were detected in supernatants. In CRC surgical patients, more importantly concerning extracellular E cadherin domain, a decreased expression was observed in tissues in function of CRC stages whereas an increased expression was found in sera. Moreover, there is an increasing trend of survival with weak serum E cadherin secretion, reinforcing the implication of this protein in CRC evolution. DISCUSSION: The extracellular domain can be defined as a 5FU resistance marker and allow CRC monitoring.


Subject(s)
Biomarkers, Tumor/blood , Cadherins/metabolism , Colorectal Neoplasms/pathology , Neoplasm Invasiveness/pathology , Adult , Aged , Antigens, CD , Antineoplastic Agents/therapeutic use , Blotting, Western , Colorectal Neoplasms/blood , Disease-Free Survival , Drug Resistance, Neoplasm/physiology , Female , Fluorouracil/therapeutic use , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/blood , Neoplasm Recurrence, Local/pathology , Protein Domains
15.
Biochim Biophys Acta Gen Subj ; 1861(7): 1676-1690, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28188858

ABSTRACT

BACKGROUND: Photodynamic therapy, using porphyrins as photosensitizers (PS), has been approved in treatment of several solid tumors. However, commonly used PS induce death but also resistance pathways in cancer cells and an alteration of surrounding normal tissues. Because polyamines (PA) are actively accumulated in cancer cells by the Polyamine Transport System (PTS), they may enable PS to specifically target cancer cells. Here, we investigated whether new protoporphyrin IX-polyamine derivatives were effective PS against prostate cancer and whether PA increased PDT specificity after 630nm irradiation. METHODS: CHO and CHO-MG cells (differing in their PTS activity) were used to assess efficacy of polyamine vectorization. MTT assays were performed on human prostate non-malignant (RWPE-1) and malignant (PC-3, DU 145 and LNCaP) cell lines to test PS phototoxicity. ROS generation, DNA fragmentation and cell signalling were assessed by ELISA/EIA, western-blots and gel shift assays. Finally, PS effects were studied on tumor growth in nude mice. RESULTS: Our PS were more effective on cancer cells compared to non-malignant cells and more effective than PpIX alone. PpIX-PA generated ROS production involved in induction of apoptotic intrinsic pathways. Different pathways involved in apoptosis resistance were studied: PS inhibited Bcl-2, Akt, and NF-κB but activated p38/COX-2/PGE2 pathways which were not implicated in apoptosis resistance in our model. In vivo experiments showed PpIX-PA efficacy was greater than results obtained with PpIX. CONCLUSIONS: All together, our results showed that PpIX-PA exerted its maximum effects without activating resistance pathways and appears to be a good candidate for prostate cancer PDT treatment.


Subject(s)
Photochemotherapy , Photosensitizing Agents/therapeutic use , Polyamines/therapeutic use , Prostatic Neoplasms/drug therapy , Protoporphyrins/therapeutic use , Animals , Apoptosis/drug effects , CHO Cells , Cell Line, Tumor , Cricetulus , Humans , Male , Polyamines/pharmacology , Prostatic Neoplasms/pathology , Protoporphyrins/pharmacology , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays
16.
Biomed Rep ; 6(1): 89-94, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28123714

ABSTRACT

Previous studies have reported the association between brain-derived neurotrophic factor (BDNF) and tumor development in numerous cancers. However, the accurate implication of the two specific ligands of tropomyosin kinase B receptor, BDNF and neurotrophic factor 4 (NT4/5), has not been studied in colorectal cancer (CRC) patients. The present study investigated the significance of serum BDNF and the NT4/5 in association with the intake of psychoactive drugs in CRC patients. Soluble BDNF and NT4 in the serum were assessed by ELISA. Although no correlation of BDNF and NT4 with the CRC stage was identified, a positive correlation was found between NT4 and the intake of psychoactive drugs (P=0.0457). For BDNF, a correlation was found in particular with the intake of benzodiazepine (P=0.0221). As BDNF and NT4/5 are implicated in the response of psychoactive treatments applied to manage depression, which frequently occurs in cancer patients, they cannot be used as prognostic or diagnostic markers for CRC in these patients. However, high expression of BDNF and NT4 was significantly associated with better survival. Therefore, these NTs may be used as markers for monitoring depression or predicting survival in CRC patients.

17.
Oncotarget ; 7(27): 41584-41598, 2016 Jul 05.
Article in English | MEDLINE | ID: mdl-27177087

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) shows a rich stroma where cancer-associated fibroblasts (CAFs) represent the major cell type. CAFs are master secretors of proteins with pro-tumor features. CAF targeting remains a promising challenge for PDA, a devastating disease where treatments focusing on cancer cells have failed. We previously introduced a novel pharmacological CAF-targeting approach using the somatostatin analog SOM230 (pasireotide) that inhibits protein synthesis in CAFs, and subsequent chemoprotective features of CAF secretome. Using primary cultures of CAF isolated from human PDA resections, we here report that CAF secretome stimulates in vitro cancer cell survival, migration and invasive features, that are abolished when CAFs are treated with SOM230. Mechanistically, SOM230 inhibitory effect on CAFs depends on the somatostatin receptor subtype sst1 expressed in CAFs but not in non-activated pancreatic fibroblasts, and on protein synthesis shutdown through eiF4E-Binding Protein-1 (4E-BP1) expression decrease. We identify interleukin-6 as a SOM230-inhibited CAF-secreted effector, which stimulates cancer cell features through phosphoinositide 3-kinase activation. In vivo, mice orthotopically co-xenografted with the human pancreatic cancer MiaPaCa-2 cells and CAFs develop pancreatic tumors, on which SOM230 treatment does not inhibit growth but abrogates metastasis. Consistently, CAF secretome stimulates epithelial-to-mesenchymal transition in cancer cells, which is reversed upon CAF treatment with SOM230. Our results highlight a novel promising anti-metastatic potential for SOM230 indirectly targeting pancreatic cancer cell invasion through pharmacological inhibition of stromal CAFs.


Subject(s)
Antineoplastic Agents/therapeutic use , Cancer-Associated Fibroblasts/drug effects , Carcinoma, Pancreatic Ductal/drug therapy , Molecular Targeted Therapy/methods , Pancreatic Neoplasms/drug therapy , Somatostatin/analogs & derivatives , Animals , Antineoplastic Agents/pharmacology , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Cancer-Associated Fibroblasts/physiology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Nude , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Somatostatin/pharmacology , Somatostatin/therapeutic use , Xenograft Model Antitumor Assays
18.
Oncotarget ; 7(23): 34480-97, 2016 Jun 07.
Article in English | MEDLINE | ID: mdl-27120782

ABSTRACT

p75NTR, a member of TNF receptor family, is the low affinity receptor common to several mature neurotrophins and the high affinity receptor for pro-neurotrophins. Brain-Derived Neurotrophic Factor (BDNF), a member of neurotrophin family has been described to play an important role in development and progression of several cancers, through its binding to a high affinity tyrosine kinase receptor B (TrkB) and/or p75NTR. However, the functions of these two receptors in renal cell carcinoma (RCC) have never been investigated. An overexpression of p75NTR, pro-BDNF, and to a lesser extent for TrkB and sortilin, was detected by immunohistochemistry in a cohort of 83 clear cell RCC tumors. p75NTR, mainly expressed in tumor tissues, was significantly associated with higher Fuhrman grade in multivariate analysis. In two derived-RCC lines, 786-O and ACHN cells, we demonstrated that pro-BDNF induced cell survival and migration, through p75NTR as provided by p75NTR RNA silencing or blocking anti-p75NTR antibody. This mechanism is independent of TrkB activation as demonstrated by k252a, a tyrosine kinase inhibitor for Trk neurotrophin receptors. Taken together, these data highlight for the first time an important role for p75NTR in renal cancer and indicate a putative novel target therapy in RCC.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Carcinoma, Renal Cell/pathology , Cell Movement/physiology , Kidney Neoplasms/pathology , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Protein Precursors/metabolism , Receptor, trkB/metabolism , Receptors, Nerve Growth Factor/metabolism , Adaptor Proteins, Vesicular Transport/biosynthesis , Aged , Aged, 80 and over , Antibodies, Blocking/pharmacology , Biopsy , Brain-Derived Neurotrophic Factor/biosynthesis , Carbazoles/pharmacology , Cell Line, Tumor , Cell Survival/physiology , Female , Humans , Indole Alkaloids/pharmacology , Male , Membrane Glycoproteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Protein Binding , Protein Precursors/biosynthesis , RNA Interference , RNA, Small Interfering/genetics , Receptor, trkB/biosynthesis , Receptors, Nerve Growth Factor/biosynthesis , Receptors, Nerve Growth Factor/genetics
19.
World J Gastroenterol ; 22(2): 490-500, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26811602

ABSTRACT

Colorectal cancer (CRC) is the second most common cause of cancer-related death in western countries. Approximately one-quarter of newly diagnosed patients for CRC have metastases, and a further 40%-50% experience disease recurrence or develop metastases after all standard therapies. Therefore, understanding the molecular mechanisms involved in the progression of CRC and subsequently developing novel therapeutic targets is crucial to improve management of CRC and patients' long-term survival. Several tyrosine kinase receptors have been implicated in CRC development, progression and metastasis, including epidermal growth factor receptor (EGFR) and vascular EGFR. Recently, tropomyosin-related kinase B (TrkB), a tyrosine kinase receptor, has been reported in CRC and found to clearly exert several biological and clinical features, such as tumor cell growth and survival in vitro and in vivo, metastasis formation and poor prognosis. Here we review the significance of TrkB and its ligand brain derived-neurotrophic factor in CRC. We focus on their expression in CRC tumor samples, and their functional roles in CRC cell lines and in in vivo models. Finally we discuss therapeutic approaches that can lead to the development of novel therapeutic agents for treating TrkB-expressing CRC tumors.


Subject(s)
Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Membrane Glycoproteins/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Brain-Derived Neurotrophic Factor/metabolism , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Drug Discovery , Humans , Membrane Glycoproteins/metabolism , Molecular Targeted Therapy , Protein-Tyrosine Kinases/metabolism , Receptor, trkB , Signal Transduction/drug effects
20.
Anal Bioanal Chem ; 407(28): 8433-43, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26427501

ABSTRACT

Despite effective treatments, relapse of colorectal cancer (CRC) is frequent, in part caused by the existence of tumor-initiating cells (TICs). Different subtypes of TICs, quiescent and activated, coexist in tumors, defining the tumor aggressiveness and therapeutic response. These subtypes have been sorted by hyperlayer sedimentation field-flow fractionation (SdFFF) from WiDr and HCT116 cell lines. On the basis of a new strategy, including TIC SdFFF sorting, 3D Matrigel amplification, and grafting of corresponding TIC colonies on the chick chorioallantoic membrane (CAM), specific tumor matrices could be obtained. If tumors had similar architectural structure with vascularization by the host system, they had different proliferative indices in agreement with their initial quiescent or activated state. Protein analysis also revealed that tumors obtained from a population enriched for "activated" TICs lost "stemness" properties and became invasive. In contrast, tumors obtained from a population enriched for "quiescent" TICs kept their stemness properties and seemed to be less proliferative and invasive. Then, it was possible to produce different kinds of tumor which could be used as selective supports to study carcinogenesis and therapy sensitivity.


Subject(s)
Biomarkers, Tumor/genetics , Cell Separation/methods , Colorectal Neoplasms/diagnosis , Models, Biological , Neoplastic Stem Cells/classification , Animals , Biomarkers, Tumor/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Cell Line, Tumor , Cell Movement , Cell Separation/instrumentation , Chick Embryo , Chorioallantoic Membrane/blood supply , Chorioallantoic Membrane/pathology , Collagen/chemistry , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Drug Combinations , Fractionation, Field Flow/instrumentation , Fractionation, Field Flow/methods , Gene Expression , HCT116 Cells , Humans , Keratin-20/genetics , Keratin-20/metabolism , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Laminin/chemistry , Neoplasm Invasiveness , Neoplastic Stem Cells/pathology , Neovascularization, Pathologic/pathology , Proteoglycans/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...