Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
J Pathol ; 249(3): 343-355, 2019 11.
Article in English | MEDLINE | ID: mdl-31257576

ABSTRACT

The identification of novel regulators of tumor progression is a key challenge to gain knowledge on the biology of small intestinal neuroendocrine tumors (SI-NETs). We recently identified the loss of the axon guidance protein semaphorin 3F as a protumoral event in SI-NETs. Interestingly the expression of its receptor neuropilin-2 (NRP-2) was still maintained. This study aimed at deciphering the potential role of NRP-2 as a contributor to SI-NET progression. The role of NRP-2 in SI-NET progression was addressed using an approach integrating human tissue and serum samples, cell lines and in vivo models. Data obtained from human SI-NET tissues showed that membranous NRP-2 expression is present in a majority of tumors, and is correlated with invasion, metastatic abilities, and neovascularization. In addition, NRP-2 soluble isoform was found elevated in serum samples from metastatic patients. In preclinical mouse models of NET progression, NRP-2 silencing led to a sustained antitumor effect, partly driven by the downregulation of VEGFR2. In contrast, its ectopic expression conferred a gain of aggressiveness, driven by the activation of various oncogenic signaling pathways. Lastly, NRP-2 inhibition led to a decrease of tumor cell viability, and sensitized to therapeutic agents. Overall, our results point out NRP-2 as a potential therapeutic target for SI-NETs, and will foster the development of innovative strategies targeting this receptor. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Carcinoma, Neuroendocrine/metabolism , Intestinal Neoplasms/metabolism , Intestine, Small/metabolism , Neuropilin-2/metabolism , Aged , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Carcinoma, Neuroendocrine/drug therapy , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/secondary , Cell Line, Tumor , Cell Movement , Everolimus/pharmacology , Female , Gene Expression Regulation, Neoplastic , Humans , Intestinal Neoplasms/drug therapy , Intestinal Neoplasms/genetics , Intestinal Neoplasms/pathology , Intestine, Small/pathology , Male , Mice, Nude , Neoplasm Invasiveness , Neovascularization, Pathologic , Neuropilin-2/blood , Neuropilin-2/genetics , Protein Kinase Inhibitors/pharmacology , Signal Transduction , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
2.
Mol Cancer Ther ; 17(1): 60-72, 2018 01.
Article in English | MEDLINE | ID: mdl-29051320

ABSTRACT

Streptozotocin-based chemotherapy is the first-line chemotherapy recommended for advanced pancreatic neuroendocrine tumors (pNETs), whereas targeted therapies, including mTOR inhibitors, are available in second-line treatment. Unfortunately, objective response rates to both treatments are limited. Because mTOR pathway activation, commonly observed in pNETs, has been reported as one of the major mechanisms accounting for chemoresistance, we investigated the potential benefit of mTOR inhibition combined with streptozotocin treatment in a subset of pNETs, namely insulinomas. To evaluate the potential of mTOR inhibition in combination with streptozotocin, we selected four different inhibitors acting at various levels of the pathway (everolimus: inhibition of mTORC1; MK-2206: inhibition of AKT; BKM120: inhibition of PI3K, mTORC1, and mTORC2; and BEZ235: inhibition of mTORC1 and mTORC2). Effects on cell viability and apoptosis were assessed in insulinoma cell lines INS-1E (rat) and MIN6 (mouse) in vitro and were confirmed in vivo by using a mouse model of hepatic tumor dissemination after intrasplenic xenograft. In vitro, all four combinations display synergistic effects. These combinations lead to heterogeneous mTOR pathway inhibition, in agreement with their respective target, and increased apoptosis. In vivo, tumor growth in the liver was significantly inhibited by combining streptozotocin with everolimus (P = 0.0014), BKM120 (P = 0.0092), or BEZ235 (P = 0.008) as compared to each agent alone. These results suggest that targeting the mTOR pathway in combination with streptozotocin could be of potential benefit for insulinomas and pNET patients and thus support further clinical investigations. Mol Cancer Ther; 17(1); 60-72. ©2017 AACR.


Subject(s)
Insulinoma/drug therapy , Streptozocin/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/therapeutic use , Animals , Female , Humans , Insulinoma/pathology , Mice , Mice, Nude , Streptozocin/pharmacology , TOR Serine-Threonine Kinases/pharmacology
3.
Oncotarget ; 8(13): 20974-20987, 2017 Mar 28.
Article in English | MEDLINE | ID: mdl-28423496

ABSTRACT

mTOR and Unfolded Protein Response (UPR) are two signaling pathways frequently activated in cancer cells. The mTOR pathway has been shown to be up-regulated in most gastroenteropancreatic neuroendocrine tumors. In contrast, little is known about the UPR status in neoplastic neuroendocrine cells. However, these hormone-producing cells are likely to present distinctive adaptations of this pathway, as other secretory cells. We therefore analyzed the status of the three axes of UPR and their relation to mTOR pathway in two gastrointestinal neuroendocrine tumors (GI-NET) cell lines STC-1 and GluTag. At baseline, pharmacological inducers activate the three arms of UPR: PERK, ATF6 and IRE1. Although hypoxia stimulates the PERK, ATF6 and IRE-1 pathways in both cell lines, glucose depletion activates UPR only in STC-1 cell line. Strikingly, P-p70S6K1 increases concomitantly to P-PERK and BiP in response to thapsigargin treatment, glucose depletion or hypoxia. We found that different mTOR inhibitors activate the PERK signaling pathway. To confirm that mTOR inhibition modulates PERK activation, we inhibited PERK and showed that it decreased cell viability when associated to mTOR inhibition, indicating that mTOR drives a PERK-dependent survival pathway. In conclusion, in GI-NET cell lines, UPR signaling is functional and PERK arm is induced by mTOR inhibition. These observations open up new perspectives for therapeutic strategies: the crosstalk between mTOR and UPR might contribute to the resistance to mTOR inhibitors and could be targeted by mTOR and PERK inhibitors in combination therapy.


Subject(s)
Cell Proliferation/drug effects , Gastrointestinal Neoplasms/pathology , Heat-Shock Proteins/metabolism , Neuroendocrine Tumors/pathology , TOR Serine-Threonine Kinases/metabolism , Unfolded Protein Response/drug effects , eIF-2 Kinase/metabolism , Apoptosis , Biomarkers, Tumor/metabolism , Cell Hypoxia , Endoplasmic Reticulum Chaperone BiP , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/metabolism , Glucose , Heat-Shock Proteins/antagonists & inhibitors , Humans , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/metabolism , Protein Kinase Inhibitors/pharmacology , Signal Transduction , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tumor Cells, Cultured , eIF-2 Kinase/antagonists & inhibitors
4.
Invest New Drugs ; 34(5): 654-62, 2016 10.
Article in English | MEDLINE | ID: mdl-27230034

ABSTRACT

Objectives Several targeted therapies are available for metastatic neuroendocrine tumours (NETs) but no predictive factor of response to these treatments has been identified yet. Our aim was to identify and evaluate clinical, biological, histological and functional markers of response to everolimus. Methods We retrospectively reviewed 53 patients with NETs treated with everolimus (68 % in clinical trials). Clinical, biological and histological data were analyzed. The functional marker p-p70S6K, a main effector of the mTOR pathway, was studied by immunohistochemistry in 43 cases. Prognostic factors of progression-free survival (PFS) were studied by Kaplan Meier analysis. Results All patients had metastatic and progressive disease before everolimus treatment. Objective response was 9 % and median PFS was 8.1 (4.7-11.5) months. Hypercholesterolemia (HR = 0.13, p < 0.0001) was associated with longer PFS, whereas presence of bone metastases (HR = 3.1, p < 0.001) and overexpression of p-p70S6K by tumor cells (HR = 2.5, p = 0.01) were associated with shorter PFS under everolimus at multivariate analysis. Conclusion Clinical markers are not useful to predict response to everolimus. However, occurrence of hypercholesterolemia under treatment may be an early marker of response. Prospective studies are required to confirm these results and to assess whether p-p70S6K immunostaining is a prognostic or predictive marker of no-response to everolimus.


Subject(s)
Antineoplastic Agents/therapeutic use , Everolimus/therapeutic use , Hypercholesterolemia/chemically induced , Neuroendocrine Tumors/drug therapy , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Everolimus/adverse effects , Everolimus/pharmacology , Female , Humans , Hypercholesterolemia/metabolism , Kaplan-Meier Estimate , Male , Middle Aged , Neuroendocrine Tumors/metabolism , Prognosis , TOR Serine-Threonine Kinases/metabolism , Treatment Outcome
5.
Oncotarget ; 6(34): 36731-45, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26447612

ABSTRACT

Gastro-intestinal neuroendocrine tumors (GI-NETs) are rare neoplasms, frequently metastatic, raising difficult clinical and therapeutic challenges due to a poor knowledge of their biology. As neuroendocrine cells express both epithelial and neural cell markers, we studied the possible involvement in GI-NETs of axon guidance molecules, which have been shown to decrease tumor cell proliferation and metastatic dissemination in several tumor types. We focused on the role of Semaphorin 3F (SEMA3F) in ileal NETs, one of the most frequent subtypes of GI-NETs.SEMA3F expression was detected in normal neuroendocrine cells but was lost in most of human primary tumors and all their metastases. SEMA3F loss of expression was associated with promoter gene methylation. After increasing endogenous SEMA3F levels through stable transfection, enteroendocrine cell lines STC-1 and GluTag showed a reduced proliferation rate in vitro. In two different xenograft mouse models, SEMA3F-overexpressing cells exhibited a reduced ability to form tumors and a hampered liver dissemination potential in vivo. This resulted, at least in part, from the inhibition of mTOR and MAPK signaling pathways.This study demonstrates an anti-tumoral role of SEMA3F in ileal NETs. We thus suggest that SEMA3F and/or its cellular signaling pathway could represent a target for ileal NET therapy.


Subject(s)
Axon Guidance/physiology , Ileal Neoplasms/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neuroendocrine Tumors/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Disease Progression , Female , Heterografts , Humans , Ileal Neoplasms/genetics , Ileal Neoplasms/pathology , Membrane Proteins/genetics , Mice , Nerve Tissue Proteins/genetics , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/pathology , Signal Transduction
6.
Cancer Res ; 75(17): 3519-28, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26122847

ABSTRACT

Collapsin response mediator protein 5 (CRMP5) belongs to a family of five cytosolic proteins that play a major role in nervous system development. This protein was first described in cancer-induced autoimmune processes, causing neurodegenerative disorders (paraneoplastic neurologic syndromes). CRMP5 expression has been reported to serve as a biomarker for high-grade lung neuroendocrine carcinomas; however, its functional roles have not been examined in any setting of cancer pathophysiology. In this study, we report two different CRMP5 expression patterns observed in human glioblastoma (GBM) biopsies that establish connections between CRMP5 expression, Notch receptor signaling, and GBM cell proliferation. We demonstrated that elevated CRMP5 promotes Notch receptor expression and Akt activation in human tumor cell lines, GBM stem cells, and primary tumor biopsies. We have shown that the high CRMP5 and Notch expression in GBM xenograft is related to stem cells. This suggests that high CRMP5 expression pattern in GBM biopsies encompasses a subset of stem cells. Mechanistically, CRMP5 functioned by hijacking Notch receptors from Itch-dependent lysosomal degradation. Our findings suggest that CRMP5 serves as a major mediator of Notch signaling and Akt activation by controlling the degradation of the Notch receptor, with implications for defining a biomarker signature in GBM that correlates with and may predict patient survival.


Subject(s)
Brain Neoplasms/genetics , Glioblastoma/genetics , Nerve Tissue Proteins/biosynthesis , Receptors, Notch/genetics , Adult , Aged , Aged, 80 and over , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Humans , Hydrolases , Male , Mice , Microtubule-Associated Proteins , Middle Aged , Nerve Tissue Proteins/genetics , Receptors, Notch/metabolism , Signal Transduction , Survival Analysis , Xenograft Model Antitumor Assays
7.
J Neural Transm (Vienna) ; 122(7): 975-91, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25976431

ABSTRACT

Brain banks manage and store fully clinically and pathologically characterised brains. The diversity of techniques used in research projects increases. These biological resource centres are made to adapt brain tissue processing. Furthermore, the development of more sensitive techniques to analyse nucleic acids and proteins offers new fields of exploration when combined with laser capture microdissection in order to decipher the physiopathology of diseases at the cell level. In this study, our goal was to evaluate procedures and set a workflow compatible with the constraints of brain banks, from brain sampling to laser capture microdissection and pre-analytical quality assessment. We compared various methods of freezing brain tissue, focused on morphological quality preservation of brain microscopical structures and on the quality of nucleic acid or protein yields. Staining protocols combined with strategies to lower neurones autofluorescence were adapted for the same purpose. Finally, we found that laser capture microdissection is possible in the setting of brain banks. However, the entire process has to be envisioned from the autopsy to the analysis. The impact on protein or nucleic acid quality is a limitation that restricts the amount of samples available for this purpose.


Subject(s)
Brain/pathology , Microdissection , Neurons/pathology , Tissue Banks , Workflow , Adult , Aged , Aged, 80 and over , Brain Diseases/pathology , Europe , Female , Humans , Male , Middle Aged , Neurons/metabolism , Postmortem Changes , Proteins/genetics , Proteins/metabolism , Specimen Handling , Staining and Labeling
8.
Mol Cell Endocrinol ; 399: 154-63, 2015 Jan 05.
Article in English | MEDLINE | ID: mdl-25224486

ABSTRACT

Small-intestinal neuroendocrine tumors (SI-NETs) are defined as locally invasive only after extension to the muscularis propria. To gain further insight into the molecular mechanisms, we applied a proteomic approach to an orthotopic xenograft model to identify candidate proteins evaluable in human SI-NETs. After grafting STC-1 neuroendocrine tumor cells on the caecum of nude mice, comparative proteomic studies were performed between the pre-invasive and the invasive stages, respectively 2 and 8 weeks after grafting. We identified 24 proteins displaying at least a 1.5-fold differential expression between 2 and 8 week-stages. Most were cytoskeleton-associated proteins, among which five showed decreasing expression levels (CRMP2, TCP1ε, TPM2, vimentin, desmin) and two increasing expression levels (14-3-3γ, CK8). Changes for CRMP2, TCP1ε, TPM2 and 14-3-3γ were confirmed in experimental tumors and in a series of 28 human SI-NETs. In conclusion, our results underline the relevance of proteomics to identify novel biomarkers of tissue invasion.


Subject(s)
Cytoskeletal Proteins/biosynthesis , Enteroendocrine Cells/metabolism , Gene Expression Regulation, Neoplastic , Intestinal Neoplasms/metabolism , Neoplasm Proteins/biosynthesis , Proteomics , Animals , Cell Line, Tumor , Enteroendocrine Cells/pathology , Humans , Intestinal Neoplasms/pathology , Mice , Mice, Nude , Mice, Transgenic , Neoplasm Transplantation
9.
Am J Surg Pathol ; 32(11): 1699-708, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18769332

ABSTRACT

The diagnosis of high-grade neuroendocrine tumors has strong clinical relevance because it identifies patients at higher risk of an unfavorable outcome who should receive multimodal treatment. However, these tumors can be mistaken for poorly differentiated nonsmall cell carcinoma or carcinoid lung tumors. In fact, no immunohistochemical marker can currently distinguish between histologic lung subtypes. Because the collapsin response mediator protein (CRMP) family is involved in an autoimmune disease associated with small cell lung carcinoma, we explored the relationship between CRMP5 expression and lung tumor behavior. Using World Health Organization morphologic criteria, 123 lung neuroendocrine tumors and 41 randomly selected non-neuroendocrine tumors were classified. CRMP5 protein expression in tumors, metastases, and healthy lung tissue was assessed using immunostaining method. Strong and extensive CRMP5 expression was seen in 98.6% of high-grade neuroendocrine lung tumors, including small cell lung carcinoma and large cell lung neuroendocrine carcinoma, but not in any of the squamous cell carcinomas or lung adenocarcinomas in our series. In contrast, the majority of low-grade neuroendocrine lung tumors were negative for CRMP5 staining, although weak CRMP5 expression was seen in some, with 2 different staining patterns of either scattered positive cells or small foci of positive cells. Our findings point at CRMP5 as a novel marker for routine pathologic evaluation of lung tumors surgical samples in distinguishing between highly aggressive neuroendocrine carcinoma and the other lung cancers.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Neuroendocrine/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Nerve Tissue Proteins/biosynthesis , Adult , Blotting, Western , Carcinoma, Neuroendocrine/mortality , Disease-Free Survival , Female , Gene Expression , Humans , Hydrolases , Immunohistochemistry , Kaplan-Meier Estimate , Lung Neoplasms/mortality , Male , Microtubule-Associated Proteins , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...