Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 234
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 141(1): 178-90, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20371353

ABSTRACT

Our findings that PlGF is a cancer target and anti-PlGF is useful for anticancer treatment have been challenged by Bais et al. Here we take advantage of carcinogen-induced and transgenic tumor models as well as ocular neovascularization to report further evidence in support of our original findings of PlGF as a promising target for anticancer therapies. We present evidence for the efficacy of additional anti-PlGF antibodies and their ability to phenocopy genetic deficiency or silencing of PlGF in cancer and ocular disease but also show that not all anti-PlGF antibodies are effective. We also provide additional evidence for the specificity of our anti-PlGF antibody and experiments to suggest that anti-PlGF treatment will not be effective for all tumors and why. Further, we show that PlGF blockage inhibits vessel abnormalization rather than density in certain tumors while enhancing VEGF-targeted inhibition in ocular disease. Our findings warrant further testing of anti-PlGF therapies.


Subject(s)
Neovascularization, Physiologic/drug effects , Pregnancy Proteins/antagonists & inhibitors , Pregnancy Proteins/metabolism , Angiogenesis Inhibitors/therapeutic use , Animals , Antibodies, Monoclonal/therapeutic use , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/prevention & control , Choroid/blood supply , Disease Models, Animal , Eye Diseases/pathology , Humans , Liver Neoplasms, Experimental/blood supply , Liver Neoplasms, Experimental/prevention & control , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Papilloma/blood supply , Papilloma/chemically induced , Papilloma/prevention & control , Placenta Growth Factor , Skin Neoplasms/blood supply , Skin Neoplasms/chemically induced , Skin Neoplasms/prevention & control
2.
Nature ; 542(7639): 49-54, 2017 02 02.
Article in English | MEDLINE | ID: mdl-28024299

ABSTRACT

Lymphatic vessels are lined by lymphatic endothelial cells (LECs), and are critical for health. However, the role of metabolism in lymphatic development has not yet been elucidated. Here we report that in transgenic mouse models, LEC-specific loss of CPT1A, a rate-controlling enzyme in fatty acid ß-oxidation, impairs lymphatic development. LECs use fatty acid ß-oxidation to proliferate and for epigenetic regulation of lymphatic marker expression during LEC differentiation. Mechanistically, the transcription factor PROX1 upregulates CPT1A expression, which increases acetyl coenzyme A production dependent on fatty acid ß-oxidation. Acetyl coenzyme A is used by the histone acetyltransferase p300 to acetylate histones at lymphangiogenic genes. PROX1-p300 interaction facilitates preferential histone acetylation at PROX1-target genes. Through this metabolism-dependent mechanism, PROX1 mediates epigenetic changes that promote lymphangiogenesis. Notably, blockade of CPT1 enzymes inhibits injury-induced lymphangiogenesis, and replenishing acetyl coenzyme A by supplementing acetate rescues this process in vivo.


Subject(s)
Fatty Acids/chemistry , Fatty Acids/metabolism , Lymphangiogenesis , Lymphatic Vessels/cytology , Lymphatic Vessels/metabolism , Acetates/pharmacology , Acetyl Coenzyme A/metabolism , Acetylation/drug effects , Animals , Carnitine O-Palmitoyltransferase/antagonists & inhibitors , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Epigenesis, Genetic , Female , Histones/metabolism , Homeodomain Proteins/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Lymphangiogenesis/drug effects , Lymphangiogenesis/genetics , Lymphatic Vessels/drug effects , Mice , Mice, Inbred C57BL , Oxidation-Reduction/drug effects , Protein Biosynthesis , Transcription, Genetic , Tumor Suppressor Proteins/metabolism , Umbilical Arteries/cytology , Up-Regulation
3.
Cell Mol Life Sci ; 79(5): 255, 2022 Apr 22.
Article in English | MEDLINE | ID: mdl-35460056

ABSTRACT

The urokinase plasminogen activator receptor-associated protein (uPARAP/Endo180) is already known to be a key collagen receptor involved in collagen internalization and degradation in mesenchymal cells and some macrophages. It is one of the four members of the mannose receptor family along with a macrophage mannose receptor (MMR), a phospholipase lipase receptor (PLA2R), and a dendritic receptor (DEC-205). As a clathrin-dependent endocytic receptor for collagen or large collagen fragments as well as through its association with urokinase (uPA) and its receptor (uPAR), uPARAP/Endo180 takes part in extracellular matrix (ECM) remodeling, cell chemotaxis and migration under physiological (tissue homeostasis and repair) and pathological (fibrosis, cancer) conditions. Recent advances that have shown an expanded contribution of this multifunctional protein across a broader range of biological processes, including vascular biology and innate immunity, are summarized in this paper. It has previously been demonstrated that uPARAP/Endo180 assists in lymphangiogenesis through its capacity to regulate the heterodimerization of vascular endothelial growth factor receptors (VEGFR-2 and VEGFR-3). Moreover, recent findings have demonstrated that it is also involved in the clearance of collectins and the regulation of the immune system, something which is currently being studied as a biomarker and a therapeutic target in a number of cancers.


Subject(s)
Mannose-Binding Lectins , Vascular Endothelial Growth Factor A , Carrier Proteins , Collagen/metabolism , Mannose-Binding Lectins/metabolism , Membrane Glycoproteins/metabolism , Receptors, Mitogen/metabolism , Vascular Endothelial Growth Factor A/metabolism
4.
Cell Mol Life Sci ; 79(6): 295, 2022 May 14.
Article in English | MEDLINE | ID: mdl-35567669

ABSTRACT

Although lymph node (LN) metastasis is an important prognostic parameter in cervical cancer, the tissue remodeling at a pre-metastatic state is poorly documented in LNs. We here identified periostin (POSTN) as a component of non-metastatic LNs by applying proteomic analyses and computerized image quantifications on LNs of patients with cervical cancer. We provide evidence for remarkable modifications of POSTN and lymphatic vessel distributions and densities in non-metastatic sentinel and metastatic human LNs, when compared to distant non-metastatic LNs. POSTN deposition at a pre-metastatic stage was demonstrated in a pre-clinical murine model (the ear sponge assay). Its expression by fibroblastic LN cells was assessed by in situ hybridization and in vitro cultures. In vitro, POSTN promoted lymphatic endothelial cell functions and tumor cell proliferation. Accordingly, the in vivo injection of recombinant POSTN together with VEGF-C boosted the lymphangiogenic response, while the metastatic potential of tumor cells was drastically reduced using a POSTN blocking antibody. This translational study also supports the existence of an unprecedented dialog "in cascade", between the primary tumor and the first pelvic nodal relay in early cervical cancer, and subsequently from pelvic LN to para-aortic LNs in locally advanced cervical cancers. Collectively, this work highlights the association of POSTN deposition with lymphangiogenesis in LNs, and provides evidence for a key contribution of POSTN in promoting VEGF-C driven lymphangiogenesis and the seeding of metastatic cells.


Subject(s)
Cell Adhesion Molecules/metabolism , Lymph Nodes , Uterine Cervical Neoplasms , Animals , Endothelial Cells/metabolism , Female , Humans , Lymph Nodes/metabolism , Lymphatic Metastasis/pathology , Mice , Proteomics , Uterine Cervical Neoplasms/metabolism , Vascular Endothelial Growth Factor C/metabolism
5.
Int J Mol Sci ; 24(12)2023 Jun 09.
Article in English | MEDLINE | ID: mdl-37373092

ABSTRACT

MT4-MMP (or MMP-17) belongs to the membrane-type matrix metalloproteinases (MT-MMPs), a distinct subset of the MMP family that is anchored to the cell surface, in this case by a glycosylphosphatidylinositol (GPI) motif. Its expression in a variety of cancers is well documented. However, the molecular mechanisms by which MT4-MMP contributes to tumor development need further investigation. In this review, we aim to summarize the contribution of MT4-MMP in tumorigenesis, focusing on the molecular mechanisms triggered by the enzyme in tumor cell migration, invasiveness, and proliferation, in the tumor vasculature and microenvironment, as well as during metastasis. In particular, we highlight the putative substrates processed and signaling cascades activated by MT4-MMP that may underlie these malignancy processes and compare this with what is known about its role during embryonic development. Finally, MT4-MMP is a relevant biomarker of malignancy that can be used for monitoring cancer progression in patients as well as a potential target for future therapeutic drug development.


Subject(s)
Matrix Metalloproteinase 17 , Neoplasms , Humans , Matrix Metalloproteinase 17/metabolism , Neoplasms/genetics , Matrix Metalloproteinases, Membrane-Associated/metabolism , Tumor Microenvironment
6.
Int J Mol Sci ; 24(11)2023 Jun 03.
Article in English | MEDLINE | ID: mdl-37298669

ABSTRACT

Estetrol (E4) is a natural estrogen with promising therapeutic applications in humans. The European Medicines Agency and the Food and Drug Administration have approved the use of 15 mg E4/3 mg drospirenone for contraceptive indication. Phase III clinical trials with 15-20 mg E4 for the relief of climacteric complaints are currently running. Relevant data from preclinical animal models are needed to characterize the molecular mechanisms and the pharmacological effects of E4 and possibly to reveal new therapeutic applications and to anticipate potential adverse effects. Therefore, it is important to design experimental procedures in rodents that closely mimic or anticipate human E4 exposure. In this study, we compared the effects of E4 exposure after acute or chronic administration in women and mice. Women who received chronic E4 treatment per os at a dose of 15 mg once daily reached a steady state within 6 to 8 days, with a mean plasma concentration of 3.20 ng/mL. Importantly, with subcutaneous, intraperitoneal or oral administration of E4 in mice, a stable concentration over time that would mimic human pharmacokinetics could not be achieved. The use of osmotic minipumps continuously releasing E4 for several weeks provided an exposure profile mimicking chronic oral administration in women. Measurements of the circulating concentration of E4 in mice revealed that the mouse equivalent dose necessary to mimic human treatment does not fit with the allometric prediction. In conclusion, this study highlights the importance of precise definition of the most appropriate dose and route of administration to utilize when developing predictive preclinical animal models to mimic or anticipate specific human treatment.


Subject(s)
Estetrol , United States , Humans , Female , Mice , Animals , Estetrol/adverse effects , Estrogens
7.
Am J Physiol Renal Physiol ; 323(2): F198-F211, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35796462

ABSTRACT

Renal ischemia-reperfusion (I/R) causes acute kidney injury (AKI). Ischemic preconditioning (IPC) attenuates I/R-associated AKI. Whole body irradiation induces renal IPC in mice. Still, the mechanisms remain largely unknown. Furthermore, the impact of kidney-centered irradiation on renal resistance against I/R has not been studied. Renal irradiation (8.5 Gy) was done in male 8- to 12-wk-old C57bl/6 mice using a small animal radiation therapy device. Left renal I/R was performed by clamping the renal pedicles for 30 min, with simultaneous right nephrectomy, at 7, 14, and 28 days postirradiation. The renal reperfusion lasted 48 h. Following I/R, blood urea nitrogen (BUN) and serum creatinine (SCr) levels were lower in preirradiated mice compared with controls; so was the histological Jablonski score of AKI. The metabolomics signature of renal I/R was attenuated in preirradiated mice. The numbers of proliferating cell nuclear antigen (PCNA)-, cluster of differentiation molecule 11b (CD11b)-, and cell surface glycoprotein F4/80-positive cells in the renal parenchyma post-I/R were reduced in preirradiated versus control groups. Such IPC was significantly observed as early as day 14 postirradiation. RNA sequencing showed an upregulation of angiogenesis- and stress response-related signaling pathways in irradiated nonischemic kidneys on day 28. Qualitative RT-PCR confirmed the increased expression of vascular endothelial growth factor (VEGF), activin receptor-like kinase 5 (ALK5), heme oxygenase-1 (HO1), platelet endothelial cell adhesion molecule-1 (PECAM1), NADPH oxidase 2 (NOX2), and heat shock proteins 70 and 27 (HSP70 and HSP27, respectively) in irradiated kidneys compared with controls. In addition, irradiated kidneys showed an increased CD31-positive vascular area compared with controls. A 14-day gavage of irradiated mice with the antiangiogenic drug sunitinib before I/R abrogated the irradiation-induced IPC at both functional and structural levels. Our observations suggest that kidney-centered irradiation activates proangiogenic pathways and induces IPC, with preserved renal function and attenuated inflammation post-I/R.NEW & NOTEWORTHY This study based on a mouse model of renal ischemia-reperfusion (I/R) aimed to 1) test whether and how irradiation strictly centered on the kidney protects against the I/R injury and 2) determine the shortest efficient delay of kidney irradiation to achieve such nephroprotection. Kidney irradiation increased the vascular surface in the renal parenchyma and conferred resistance against renal I/R damage, which highlights novel putative strategies in the field of ischemic acute kidney injury.


Subject(s)
Acute Kidney Injury , Ischemic Preconditioning , Reperfusion Injury , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Acute Kidney Injury/prevention & control , Animals , Ischemia/metabolism , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Reperfusion Injury/pathology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
8.
Reprod Biol Endocrinol ; 20(1): 35, 2022 Feb 19.
Article in English | MEDLINE | ID: mdl-35183206

ABSTRACT

BACKGROUND: Ovarian tissue cryopreservation and transplantation (OTCTP) is currently the main option available to preserve fertility in prepubertal patients undergoing aggressive cancer therapy treatments. However, a major limitation of OTCTP is follicle loss after transplantation. The mouse is a model of choice for studying ovarian function and follicle development after ovarian tissue grafting in vivo. In these mouse models, ovarian tissue or ovaries can be transplanted to different sites. Our aim was to evaluate a new alternative to heterotopic transplantation models that could be useful to test pharmaceutical improvement for ovarian grafts after OTCTP. METHODS: Slow frozen murine whole ovaries were transplanted into the mouse ears (between the external ear skin layer and the cartilage). Ovarian transplants were recovered after 3, 14 or 21 days. Grafts were analyzed by immunohistochemistry and follicle density analyses were performed. RESULTS: An increase of ovarian vascularization (CD31 and Dextran-FITC positive staining), as well as cellular proliferation (Ki67 staining) were observed 3 weeks after transplantation in comparison to 3 days. Fibrosis density, evaluated after Van Gieson staining, decreased 3 weeks after transplantation. Furthermore, transplantation of cryopreserved ovaries into ovariectomized mice favored follicle activation compared to transplantation into non-ovariectomized mice. CONCLUSION: The present study indicates that surgical tissue insertion in the highly vascularized murine ear is an effective model for ovarian grafting. This model could be helpful in research to test pharmaceutical strategies to improve the function and survival of cryopreserved and transplanted ovarian tissue.


Subject(s)
Drug Evaluation, Preclinical/methods , Fertility Agents, Female/therapeutic use , Fertility Preservation/methods , Ovary/transplantation , Transplantation, Heterotopic/methods , Animals , Cell Proliferation/drug effects , Combined Modality Therapy , Female , Fertility Agents, Female/pharmacology , Graft Survival/drug effects , Hormone Replacement Therapy/methods , Mice , Mice, Inbred BALB C , Mice, SCID , Models, Biological
9.
Cell Mol Life Sci ; 78(16): 5987-6002, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34241649

ABSTRACT

Lymph node metastasis is a crucial prognostic parameter in many different types of cancers and a gateway for further dissemination to distant organs. Prior to metastatic dissemination, the primary tumor prepares for the remodeling of the draining (sentinel) lymph node by secreting soluble factors or releasing extracellular vesicles that are transported by lymphatic vessels. These important changes occur before the appearance of the first metastatic cell and create what is known as a pre-metastatic niche giving rise to the subsequent survival and growth of metastatic cells. In this review, the lymph node structure, matrix composition and the emerging heterogeneity of cells forming it are described. Current knowledge of the major cellular and molecular processes associated with nodal pre-metastatic niche formation, including lymphangiogenesis, extracellular matrix remodeling, and immunosuppressive cell enlisting in lymph nodes are additionally summarized. Finally, future directions that research could possibly take and the clinical impact are discussed.


Subject(s)
Lymph Nodes/pathology , Lymphatic Metastasis/pathology , Animals , Extracellular Vesicles/pathology , Humans , Lymphangiogenesis/physiology , Lymphatic Vessels/pathology , Prognosis
10.
Semin Cancer Biol ; 62: 166-181, 2020 05.
Article in English | MEDLINE | ID: mdl-31415910

ABSTRACT

The tumor microenvironment (TME) is a complex meshwork of extracellular matrix (ECM) macromolecules filled with a collection of cells including cancer-associated fibroblasts (CAFs), blood vessel associated smooth muscle cells, pericytes, endothelial cells, mesenchymal stem cells and a variety of immune cells. In tumors the homeostasis governing ECM synthesis and turnover is disturbed resulting in abnormal blood vessel formation and excessive fibrillar collagen accumulations of varying stiffness and organization. The disturbed ECM homeostasis opens up for new types of paracrine, cell-cell and cell-ECM interactions with large consequences for tumor growth, angiogenesis, metastasis, immune suppression and resistance to treatments. As a main producer of ECM and paracrine signals the CAF is a central cell type in these events. Whereas the paracrine signaling has been extensively studied in the context of tumor-stroma interactions, the nature of the numerous integrin-mediated cell-ECM interactions occurring in the TME remains understudied. In this review we will discuss and dissect the role of known and potential CAF interactions in the TME, during both tumorigenesis and chemoresistance-induced events, with a special focus on the "interaction landscape" in desmoplastic breast, lung and pancreatic cancers. As an example of the multifaceted mode of action of the stromal collagen receptor integrin α11ß1, we will summarize our current understanding on the role of this CAF-expressed integrin in these three tumor types.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Cell Transformation, Neoplastic/metabolism , Integrins/metabolism , Neoplasms, Connective Tissue/etiology , Neoplasms, Connective Tissue/metabolism , Animals , Biomarkers, Tumor , Cancer-Associated Fibroblasts/pathology , Cell Transformation, Neoplastic/genetics , Disease Susceptibility , Fibroblasts , Humans , Neoplasms, Connective Tissue/pathology , Organ Specificity , Tumor Microenvironment/genetics
11.
Thorax ; 74(8): 768-779, 2019 08.
Article in English | MEDLINE | ID: mdl-31142617

ABSTRACT

BACKGROUND: Air pollution, including particulates and gazes such as ozone (O3), is detrimental for patient's health and has repeatedly been correlated to increased morbidity and mortality in industrialised countries. Although studies have described a link between ambient particulate matter and increased lung cancer morbidity, no direct relation has yet been established between O3 exposure and metastatic dissemination to lungs. OBJECTIVES: To outline the mechanisms through which pulmonary O3 exposure modulates metastasis kinetics in an experimental mouse model of O3 exposure. METHODS: Metastatic responses to pulmonary O3 exposure were assessed using a reliable experimental mouse model of concomitant pulmonary O3 exposure and tumour cell injection. Roles of neutrophils in O3-induced lung metastasis were highlighted using blocking anti-Ly6G antibodies; moreover, the implication of neutrophil extracellular traps (NETs) in metastatic processes was evaluated using MRP8cre-Pad4lox/lox mice or by treating mice with DNase I. RESULTS: Pulmonary O3 exposure strongly facilitates the establishment of lung metastasis by (1) Inducing a pulmonary injury and neutrophilic inflammation, (2) Influencing very early steps of metastasis, (3) Priming neutrophils' phenotype to release NETs that favour tumour cell colonisation in lungs. The ability of O3-primed neutrophils to enhance lung colonisation by tumour cells was proven after their adoptive transfer in Balb/c mice unexposed to O3. CONCLUSIONS: Pulmonary neutrophils induced by O3 promote metastatic dissemination to lungs by producing NETs. These findings open new perspectives to improve treatment and prevention strategies in patients affected by metastatic diseases.


Subject(s)
Breast Neoplasms/pathology , Extracellular Traps , Lung Neoplasms/secondary , Melanoma/pathology , Neoplasm Metastasis , Neutrophils/pathology , Ozone/toxicity , Animals , Antibodies/pharmacology , Antigens, Ly/immunology , Bronchitis/chemically induced , Bronchitis/pathology , Bronchoalveolar Lavage Fluid/cytology , Cell Line, Tumor , Deoxyribonuclease I/pharmacology , Disease Models, Animal , Leukocyte Count , Lung Injury/chemically induced , Lung Injury/pathology , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Neoplasm Metastasis/genetics , Neoplasm Transplantation , Neutrophils/drug effects , Pneumonia/chemically induced , Pneumonia/pathology , Protein-Arginine Deiminase Type 4/genetics
13.
Int J Mol Sci ; 20(2)2019 Jan 16.
Article in English | MEDLINE | ID: mdl-30654475

ABSTRACT

MT4-MMP (or MMP17) belongs to the Membrane-Type Matrix Metalloproteinase (MT-MMP) family. This family of proteases contributes to extracellular matrix remodeling during several physiological processes, including embryogenesis, organogenesis, tissue regeneration, angiogenesis, wound healing, and inflammation. MT4-MMP (MMP17) presents unique characteristics compared to other members of the family in terms of sequence homology, substrate specificity, and internalization mode, suggesting distinct physiological and pathological functions. While the physiological functions of MT4-MMP are poorly understood, it has been involved in different pathological processes such as arthritis, cardiovascular disease, and cancer progression. The mt4-mmp transcript has been detected in a large diversity of cancers. The contribution of MT4-MMP to tumor development has been further investigated in gastric cancer, colon cancer, head and neck cancer, and more deeply in breast cancer. Given its contribution to different pathologies, particularly cancers, MT4-MMP represents an interesting therapeutic target. In this review, we examine its biological and structural properties, and we propose an overview of its physiological and pathological functions.


Subject(s)
Disease , Glycosylphosphatidylinositols/metabolism , Matrix Metalloproteinases, Membrane-Associated/metabolism , Animals , Humans , Matrix Metalloproteinase Inhibitors/pharmacology , Matrix Metalloproteinases, Membrane-Associated/biosynthesis , Models, Biological
14.
FASEB J ; 31(4): 1678-1688, 2017 04.
Article in English | MEDLINE | ID: mdl-28057697

ABSTRACT

Zonula occludens-1 (ZO-1) is a submembrane scaffolding protein that may display proinvasive functions when it relocates from tight junctions into the cytonuclear compartment. This article examines the functional involvement of ZO-1 in CXCL8/IL-8 chemokine expression in lung and breast tumor cells. ZO-1 small interfering RNA and cDNA transfection experiments emphasized regulation of CXCL8/IL-8 expression via a cytonuclear pool of ZO-1. Luciferase reporter assays highlighted a 173-bp region of CXCL8/IL-8 promoter that responded to ZO-1. Moreover, by using mutated promoter constructs, we identified a NF-κB site as critical in this activation. Furthermore, NF-κB pathway signaling analysis revealed both IκBα and p65 phosphorylation in ZO-1-overexpressing cells, and subsequent p65 silencing validated its requirement for CXCL8/IL-8 induction. Investigation of the functional implication of this regulatory axis next showed the proangiogenic activity of ZO-1 in both ex vivo and in vivo angiogenesis assays. Finally, we found that non-small-cell lung carcinoma that presented a cytonuclear ZO-1 pattern was significantly more angiogenic that that without detectable cytonuclear ZO-1 expression. Taken together, our results demonstrate that ZO-1 regulates CXCL8/IL-8 expression via the NF-κB signaling pathway and its p65 subunit, which subsequently modulates the transcription of IL-8. We also provide evidence of a newly identified regulatory pathway that could promote angiogenesis. Thus, our results support the concept that the ZO-1 shuttle from the cell junction to the cytonuclear compartment may affect both the intrinsic invasive properties of tumor cells and the establishment of the protumoral microenvironment.-Lesage, J., Suarez-Carmona, M., Neyrinck-Leglantier, D., Grelet, S., Blacher, S., Hunziker, W., Birembaut, P., Noël, A., Nawrocki-Raby, B., Gilles, C., Polette, M. Zonula occludens-1/NF-κB/CXCL8: a new regulatory axis for tumor angiogenesis.


Subject(s)
Gene Expression Regulation, Neoplastic , Interleukin-8/metabolism , NF-kappa B/metabolism , Neovascularization, Pathologic/genetics , Zonula Occludens-1 Protein/metabolism , Humans , Interleukin-8/genetics , MCF-7 Cells , Neovascularization, Pathologic/metabolism , Promoter Regions, Genetic
16.
J Mammary Gland Biol Neoplasia ; 22(1): 1-11, 2017 03.
Article in English | MEDLINE | ID: mdl-27889857

ABSTRACT

Estrogens are the subject of intensive researches aiming to elucidate their mechanism of action on the various tissues they target and especially on mammary gland and breast cancer. The use of ready-to-use slow releasing devices to administer steroids, especially estrogens, to small experimental animals remains the method of choice in terms of animal well-being and of safety for both the researcher and the animal. In this study, we evaluated and compared, in vitro and in vivo, the release kinetic of estradiol (E2) over sixty days from two different slow-releasing systems: the matrix pellet (MP) and the reservoir implant (RI). We compared the impact of these systems in three E2-sensitive mouse models : mammary gland development, human MCF7 adenocarcinoma xenograft and mouse melanoma progression. The real amount of E2 that is released from both types of devices could differ from manufacturer specifications due to inadequate release for MP and initial burst effect for RI. Compared to MP, the interindividual variability was reduced with RI thanks to a superior control of the E2 release. Depending on the dose-dependent sensitivity of the physiological or pathological readout studied, this could lead to an improvement of the statistical power of in vivo experiments and thus to a reduction of the required animal number. Altogether, our data draw attention on the importance to adequately select the slow-releasing device that is the most appropriated to a specific experiment to better fulfill the 3Rs rule (Replacement, Reduction, Refinement) related to animal welfare and protection.


Subject(s)
Breast Neoplasms/drug therapy , Estradiol/administration & dosage , Mammary Neoplasms, Experimental/drug therapy , Animals , Cell Line, Tumor , Estrogens/administration & dosage , Female , Humans , MCF-7 Cells , Mice , Mice, Inbred C57BL , Mice, Nude , Rats, Sprague-Dawley , Reproducibility of Results
17.
Lab Invest ; 97(2): 194-206, 2017 02.
Article in English | MEDLINE | ID: mdl-27918556

ABSTRACT

Fatty acid synthase (FASN) is responsible for the endogenous production of fatty acids from acetyl-CoA and malonyl-CoA. Its overexpression is associated with poor prognosis in human cancers including melanomas. Our group has previously shown that the inhibition of FASN with orlistat reduces spontaneous lymphatic metastasis in experimental B16-F10 melanomas, which is a consequence, at least in part, of the reduction of proliferation and induction of apoptosis. Here, we sought to investigate the effects of pharmacological FASN inhibition on lymphatic vessels by using cell culture and mouse models. The effects of FASN inhibitors cerulenin and orlistat on the proliferation, apoptosis, and migration of human lymphatic endothelial cells (HDLEC) were evaluated with in vitro models. The lymphatic outgrowth was evaluated by using a murine ex vivo assay. B16-F10 melanomas and surgical wounds were produced in the ears of C57Bl/6 and Balb-C mice, respectively, and their peripheral lymphatic vessels evaluated by fluorescent microlymphangiography. The secretion of vascular endothelial growth factor C and D (VEGF-C and -D) by melanoma cells was evaluated by ELISA and conditioned media used to study in vitro lymphangiogenesis. Here, we show that cerulenin and orlistat decrease the viability, proliferation, and migration of HDLEC cells. The volume of lymph node metastases from B16-F10 experimental melanomas was reduced by 39% in orlistat-treated animals as well as the expression of VEGF-C in these tissues. In addition, lymphatic vessels from orlistat-treated mice drained more efficiently the injected FITC-dextran. Orlistat and cerulenin reduced VEGF-C secretion and, increase production of VEGF-D by B16-F10 and SK-Mel-25 melanoma cells. Finally, reduced lymphatic cell extensions, were observed following the treatment with conditioned medium from cerulenin- and orlistat-treated B16-F10 cells. Altogether, our results show that FASN inhibitors have anti-metastatic effects by acting on lymphatic endothelium and melanoma cells regardless the increase of lymphatic permeability promoted by orlistat.


Subject(s)
Cerulenin/pharmacology , Fatty Acid Synthases/antagonists & inhibitors , Lactones/pharmacology , Lymphatic Vessels/drug effects , Melanoma, Experimental/prevention & control , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Fatty Acid Synthases/genetics , Fatty Acid Synthases/metabolism , Fatty Acid Synthesis Inhibitors/pharmacology , Humans , Lymphangiogenesis/drug effects , Lymphatic Metastasis , Lymphatic Vessels/metabolism , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Orlistat , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor D/metabolism
18.
Br J Cancer ; 116(6): 742-751, 2017 Mar 14.
Article in English | MEDLINE | ID: mdl-28196064

ABSTRACT

BACKGROUND: Triple-negative breast cancers (TNBC) are heterogeneous cancers with poor prognosis. We aimed to determine the clinical relevance of membrane type-4 matrix metalloproteinase (MT4-MMP), a membrane type matrix metalloproteinase that interacts with epidermal growth factor receptor (EGFR) overexpressed in >50% of TNBC. METHODS: We conducted a retrospective immunohistochemical analysis on human TNBC samples (n=81) and validated our findings in in vitro and in vivo assays. RESULTS: Membrane type-4 matrix metalloproteinase and EGFR are produced in 72.5% of TNBC samples, whereas those proteins are faintly produced by healthy tissues. Unexpectedly, tumour relapse after chemotherapy was reduced in samples highly positive for MT4-MMP. Mechanistically, this is ascribed to a higher sensitivity of MT4-MMP-producing cells to alkylating or intercalating chemotherapeutic agents, as assessed in vitro. In sharp contrast, MT4-MMP expression did not affect tumour cell sensitivity to paclitaxel that interferes with protease trafficking. Importantly, MT4-MMP expression sensitised cancer cells to erlotinib, a tyrosine kinase EGFR inhibitor. In a pre-clinical model, the growth of MT4-MMP overexpressing xenografts, but not of control ones, was reduced by epirubicin or erlotinib. The combination of suboptimal drug doses blocked drastically the growth of MT4-MMP-producing tumours. CONCLUSIONS: We demonstrate that MT4-MMP defines a sub-population of TNBC sensitive to a combination of DNA-targeting chemotherapeutic agents and anti-EGFR drugs.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/metabolism , ErbB Receptors/metabolism , Matrix Metalloproteinases, Membrane-Associated/metabolism , Neoplasm Recurrence, Local/pathology , Triple Negative Breast Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Animals , Epirubicin/administration & dosage , Erlotinib Hydrochloride/administration & dosage , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , Lymphatic Metastasis , Mice , Mice, Nude , Middle Aged , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/metabolism , Neoplasm Staging , Prognosis , Retrospective Studies , Survival Rate , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Xenograft Model Antitumor Assays
19.
Angiogenesis ; 19(1): 53-65, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26446156

ABSTRACT

The only documented activity of a subclass of ADAMTS proteases comprising ADAMTS2, 3 and 14 is the cleavage of the aminopropeptide of fibrillar procollagens. A limited number of in vitro studies suggested that ADAMTS3 is mainly responsible for procollagen II processing in cartilage. Here, we created an ADAMTS3 knockout mouse (Adamts3(-/-)) model to determine in vivo the actual functions of ADAMTS3. Heterozygous Adamts3(+/-) mice were viable and fertile, but their intercrosses demonstrated lethality of Adamts3(-/-) embryos after 15 days of gestation. Procollagens I, II and III processing was unaffected in these embryos. However, a massive lymphedema caused by the lack of lymphatics development, an abnormal blood vessel structure in the placenta and a progressive liver destruction were observed. These phenotypes are most probably linked to dysregulation of the VEGF-C pathways. This study is the first demonstration that an aminoprocollagen peptidase is crucial for developmental processes independently of its primary role in collagen biology and has physiological functions potentially involved in several human diseases related to angiogenesis and lymphangiogenesis.


Subject(s)
ADAM Proteins/metabolism , Embryo, Mammalian/metabolism , Lymphangiogenesis , Neovascularization, Physiologic , Placenta/blood supply , ADAM Proteins/deficiency , Animals , Blood Vessels/pathology , Cartilage/pathology , Collagen/metabolism , Edema/pathology , Embryo Loss/metabolism , Embryo Loss/pathology , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Homozygote , Immunohistochemistry , Liver/embryology , Liver/pathology , Mice , Mutation/genetics , Oligonucleotide Array Sequence Analysis , Placenta/pathology , Pregnancy , Protein Processing, Post-Translational , Skin/pathology , Vascular Endothelial Growth Factor C/metabolism
20.
J Pathol ; 236(4): 491-504, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25880038

ABSTRACT

Epithelial-mesenchymal transition (EMT) programmes provide cancer cells with invasive and survival capacities that might favour metastatic dissemination. Whilst signalling cascades triggering EMT have been extensively studied, the impact of EMT on the crosstalk between tumour cells and the tumour microenvironment remains elusive. We aimed to identify EMT-regulated soluble factors that facilitate the recruitment of host cells in the tumour. Our findings indicate that EMT phenotypes relate to the induction of a panel of secreted mediators, namely IL-8, IL-6, sICAM-1, PAI-1 and GM-CSF, and implicate the EMT-transcription factor Snail as a regulator of this process. We further show that EMT-derived soluble factors are pro-angiogenic in vivo (in the mouse ear sponge assay), ex vivo (in the rat aortic ring assay) and in vitro (in a chemotaxis assay). Additionally, conditioned medium from EMT-positive cells stimulates the recruitment of myeloid cells. In a bank of 40 triple-negative breast cancers, tumours presenting features of EMT were significantly more angiogenic and infiltrated by a higher quantity of myeloid cells compared to tumours with little or no EMT. Taken together, our results show that EMT programmes trigger the expression of soluble mediators in cancer cells that stimulate angiogenesis and recruit myeloid cells in vivo, which might in turn favour cancer spread.


Subject(s)
Angiogenic Proteins/metabolism , Chemotaxis , Cytokines/metabolism , Epithelial-Mesenchymal Transition , Myeloid Cells/metabolism , Neovascularization, Pathologic , Paracrine Communication , Triple Negative Breast Neoplasms/blood supply , Triple Negative Breast Neoplasms/metabolism , Tumor Microenvironment , Angiogenic Proteins/genetics , Animals , Cell Line, Tumor , Culture Media, Conditioned/metabolism , Cytokines/genetics , Female , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice, Inbred C57BL , Mice, SCID , Myeloid Cells/pathology , Phenotype , RNA Interference , Rats , Signal Transduction , Transfection , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL