Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Language
Publication year range
1.
Personal Ment Health ; 2023 Nov 29.
Article in English | MEDLINE | ID: mdl-38031321

ABSTRACT

Since the research on contemporary personality models-and psychopathology-mainly originate from the Western world, we aimed to test the factorial structure of two trait systems assessed with the Personality Inventory for DSM-5 (PID-5) in a non-Western sample and to compare the extracted models' relative associations with binge eating disorder (BED) and bipolar spectrum disorder (BSD) symptoms. A community sample (N = 516; 72% female) was administered the PID-5, which can operationalize both the DSM-5 and ICD-11 systems. The factor structures of both systems were tested using exploratory structural equation modeling (ESEM). The congruence coefficients of all factor loadings with international studies were calculated. The Binge Eating Scale (BES), Bipolar Spectrum Diagnostic Scale (BSDS), and Hypomania Checklist-32-Revised (HCL-32) were used to measure the criterion variables. Linear regression models were used for comparing the DSM-5 and ICD-11 systems in predicting the BED and BSD. The findings supported five-factor solutions for both trait systems. Both systems significantly predicted dimensional measures of both BED and BSD (all p < 0.001). The present findings support an acceptable five-factor structure for both personality systems in the non-Western sample. Different algorithms of maladaptive domains on both systems are related to binge eating and bipolar spectrum psychopathology.

2.
J Transl Med ; 13: 27, 2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25623554

ABSTRACT

BACKGROUND: Endothelial cells (ECs) are responsible for creating a tumor vascular niche as well as producing angiocrine factors. ECs demonstrate functional and phenotypic heterogeneity when located under different microenvironments. Here, we describe a tumor-stimulated mesenchymal phenotype in ECs and investigate its impact on tumor growth, stemness, and invasiveness. METHODS: Xenograft tumor assay in NOD/SCID mice and confocal imaging were conducted to show the acquisition of mesenchymal phenotype in tumor-associated ECs in vivo. Immunocytochemistry, qPCR and flow cytometry techniques showed the appearance of mesenchymal traits in ECs after contact with breast tumor cell lines MDA-MB231 or MCF-7. Cell proliferation, cell migration, and sphere formation assays were applied to display the functional advantages of mesenchymal ECs in tumor growth, invasiveness, and enrichment of tumor initiating cells. qPCR and western blotting were used to investigate the mechanisms underlying EC mesenchymal transition. RESULTS: Our results showed that co-injection of ECs and tumor cells in NOD/SCID mice significantly enhanced tumor growth in vivo with tumor-associated ECs expressing mesenchymal markers while maintaining their intrinsic endothelial trait. We also showed that a mesenchymal phenotype is possibly detectable in human neoplastic breast biopsies as well as ECs pre-exposed to tumor cells (ECs(Mes)) in vitro. The ECs(Mes) acquired prolonged survival, increased migratory behavior and enhanced angiogenic properties. In return, ECs(Mes) were capable of enhancing tumor survival and invasiveness. The mesenchymal phenotypes in ECs(Mes) were the result of a contact-dependent transient phenomenon and reversed upon removal of the neoplastic contexture. We showed a synergistic role for TGFß and notch pathways in this phenotypic change, as simultaneous inhibition of notch and TGFß down-regulated Smad1/5 phosphorylation and Jag1(KD) tumor cells were unable to initiate the process. CONCLUSIONS: Overall, our data proposed a crosstalk mechanism between tumor and microenvironment where tumor-stimulated mesenchymal modulation of ECs enhanced the constitution of a transient mesenchymal/endothelial niche leading to significant increase in tumor proliferation, stemness, and invasiveness. The possible involvement of notch and TGFß pathways in the initiation of mesenchymal phenotype may propose new stromal targets.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Human Umbilical Vein Endothelial Cells/pathology , Mesoderm/pathology , Receptors, Notch/metabolism , Tumor Microenvironment , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mesoderm/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Phenotype , Signal Transduction/genetics , Transcriptome/genetics , Transforming Growth Factor beta/metabolism , Xenograft Model Antitumor Assays
3.
PLoS One ; 9(11): e112424, 2014.
Article in English | MEDLINE | ID: mdl-25380486

ABSTRACT

Treating metastasis has been challenging due to tumors complexity and heterogeneity. This complexity is partly related to the crosstalk between tumor and its microenvironment. Endothelial cells -the building blocks of tumor vasculature- have been shown to have additional roles in cancer progression than angiogenesis and supplying oxygen and nutrients. Here, we show an alternative role for endothelial cells in supporting breast cancer growth and spreading independent of their vascular functions. Using endothelial cells and breast cancer cell lines MDA-MB231 and MCF-7, we developed co-culture systems to study the influence of tumor endothelium on breast tumor development by both in vitro and in vivo approaches. Our results demonstrated that endothelial cells conferred survival advantage to tumor cells under complete starvation and enriched the CD44HighCD24Low/- stem cell population in tumor cells. Moreover, endothelial cells enhanced the pro-metastatic potential of breast cancer cells. The in vitro and in vivo results concordantly confirmed a role for endothelial Jagged1 to promote breast tumor through notch activation. Here, we propose a role for endothelial cells in enhancing breast cancer progression, stemness, and pro-metastatic traits through a perfusion-independent manner. Our findings may be beneficial in developing novel therapeutic approaches.


Subject(s)
Breast Neoplasms/metabolism , Cellular Microenvironment , Endothelial Cells/metabolism , Neoplastic Stem Cells/metabolism , Receptors, Notch/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Breast Neoplasms/pathology , CD24 Antigen/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Line, Tumor , Cell Survival , Cells, Cultured , Coculture Techniques , Endothelial Cells/cytology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Hyaluronan Receptors/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , MCF-7 Cells , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Microscopy, Confocal , Neoplasm Metastasis , Neoplastic Stem Cells/pathology , Reverse Transcriptase Polymerase Chain Reaction , Serrate-Jagged Proteins , Transcription Factor HES-1 , Transplantation, Heterologous
4.
Mol Cancer Ther ; 13(12): 3123-36, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25319392

ABSTRACT

Ovarian cancer is the second leading cause of cancer-related death in women worldwide. Despite optimal cytoreduction and adequate adjuvant therapies, initial tumor response is often followed by relapse suggesting the existence of a tumor niche. Targeted therapies have been evaluated in ovarian cancer to overcome resistant disease. Among them, antiangiogenic therapies inhibit new blood vessel growth, induce endothelial cell apoptosis, and block the incorporation of hematopoietic and endothelial progenitor cells into new blood vessels. Despite in vitro and in vivo successes, antivascular therapy with bevacizumab targeting VEGF-A has limited efficacy in ovarian cancer. The precise molecular mechanisms underlying clinical resistance to anti-VEGF therapies are not yet well understood. Among them, tumor and stromal heterogeneity might determine the treatment outcomes. The present study investigates whether abnormalities in the tumor endothelium may contribute to treatment resistance to bevacizumab and promote a residual microscopic disease. Here, we showed that ovarian cancer cells activate Akt phosphorylation in endothelial cells inducing resistance to bevacizumab leading to an autocrine loop based on FGF2 secretion. Altogether, our results point out the role of an activated endothelium in the resistance to bevacizumab and in the constitution of a niche for a residual disease.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Drug Resistance, Neoplasm , Endothelium/metabolism , Endothelium/pathology , Neoplasm, Residual , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Bevacizumab , Cell Communication , Cell Line , Cell Survival/drug effects , Enzyme Activation , Female , Fibroblast Growth Factor 2/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Models, Biological , Ovarian Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects
5.
Cancer Microenviron ; 7(1-2): 41-59, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24424657

ABSTRACT

The tumor stroma plays an essential role in tumor growth, resistance to therapy and occurrence of metastatic phenotype. Tumor vessels have been considered as passive conducts for nutrients but several studies have demonstrated secretion of pro-tumoral factors by endothelial cells. The failure of anti-angiogenic therapies to meet expectations raised by pre-clinical studies prompt us to better study the cross-talk between endothelial and cancer cells. Here, we hypothesized that tumor cells and the endothelium secrete bio-active microparticles (MPs) participating to a functional cross-talk. We characterized the cancer cells MPs, using breast and ovarian cancer cell lines (MCF7, MDA-MB231, SKOV3, OVCAR3 and a primary cell lines, APOCC). Our data show that MPs from mesenchymal-like cell lines (MDA-MB231, SKOV3 and APOCC) were able to promote an activation of endothelial cells through Akt phosphorylation, compared to MPs from epithelial-like cell lines (OVCAR3 and MCF7). The MPs from mesenchymal-like cells contained increased angiogenic molecules including PDGF, IL8 and angiogenin. The endothelial activation was associated to increased Arf6 expression and MPs secretion. Endothelial activation functionalized an MP dependent pro-tumoral vascular niche promoting cancer cells proliferation, invasiveness, stem cell phenotype and chemoresistance. MPs from cancer and endothelial cells displayed phenotypic heterogeneity, and participated to a functional cross-talk where endothelial activation by cancer MPs resulted in increased secretion of EC-MPs sustaining tumor cells. Such cross-talk may play a role in perfusion independent role of the endothelium.

6.
J Transl Med ; 11: 94, 2013 Apr 10.
Article in English | MEDLINE | ID: mdl-23574623

ABSTRACT

Our vision of cancer has changed during the past decades. Indeed tumors are now perceived as complex entities where tumoral and stromal components interact closely. Among the different elements of tumor stroma the cellular component play a primordial role. Bone Marrow derived mesenchymal cells (MSCs) are attracted to tumor sites and support tumor growth. Endothelial cells (ECs) play a major role in angiogenesis. While the literature documents many aspects of the cross talk between stromal and cancer cells, the role of direct hetero-cellular contact is not clearly established. Recently, Tunneling nanotubes (TnTs) have been shown to support cell-to-cell transfers of plasma membrane components, cytosolic molecules and organelles within cell lines. Herein, we have investigated the formation of heterocellular TnTs between stromal (MSCs and ECs) and cancer cells. We demonstrate that TnTs occur between different cancer cells, stromal cells and cancer-stromal cell lines. We showed that TnTs-like structure occurred in 3D anchorage independent spheroids and also in tumor explant cultures. In our culture condition, TnTs formation occurred after large membrane adhesion. We showed that intercellular transfers of cytoplasmic content occurred similarly between cancer cells and MSCs or ECs, but we highlighted that the exchange of mitochondria occurred preferentially between endothelial cells and cancer cells. We illustrated that the cancer cells acquiring mitochondria displayed chemoresistance. Our results illustrate the perfusion-independent role of the endothelium by showing a direct endothelial to cancer cell mitochondrial exchange associated to phenotypic modulation. This supports another role of the endothelium in the constitution of the metastatic niche.


Subject(s)
Bone Marrow Cells/cytology , Drug Resistance, Neoplasm , Mesenchymal Stem Cells/cytology , Mitochondria/metabolism , Ovarian Neoplasms/drug therapy , Cell Adhesion , Cell Line, Tumor , Cell Membrane/metabolism , Cell Survival , Coculture Techniques/methods , Female , Green Fluorescent Proteins/metabolism , Humans , MCF-7 Cells , Nanotubes/chemistry , Neoplasms/drug therapy , Neoplasms/metabolism , Neovascularization, Pathologic , Ovarian Neoplasms/metabolism
7.
Int J Cancer ; 128(3): 715-25, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-20725999

ABSTRACT

Hyperthermic intraperitoneal chemotherapy (HIPEC) has shown promise in treatment of ovarian carcinosis. Despite its efficiency for the treatment of peritoneal carcinosis from digestive tract neoplasia, it has failed to demonstrate significant benefit in ovarian cancers. It is therefore essential to understand the mechanism underlying resistance to HIPEC in ovarian cancers. Mesenchymal stem cells (MSC) play an important role in the development of ovarian cancer metastasis and resistance to treatments. A recent study suggests that MSCs may be cytotoxic for cancer cells upon heat shock. In contrast, we describe the protective role of MSC against hyperthermia. Using cytokine arrays we determined that the tumor associated MSC (TAMC) secrete pro-tumoral cytokines. We studied the effect of hyperthermia in co-culture setting of TAMC or BM-MCS associated with ovarian cancer cell lines (SKOV3 and CaOV3) with polyvariate flow cytometry. We demonstrate that hyperthermia does not challenge survival of TAMC or bone marrow derived MSC (BM-MSC). Both TAMC and BM-MSC displayed strong protective effect inducing thermotolerance in ovarian cancer cells (OCC). Transwell experiments demonstrated the role of secreted factors. We showed that CXCL12 was inducing thermotolerance and that inhibition of CXCL12/CXCR4 interaction restored cytotoxicity of hyperthermia in co-culture experiments. Contrary to the previous published study we demonstrated that TAMC and BM-MSC co-cultured with OCC induced thermotolerance in a CXCL12 dependant manner. Targeting the interaction between stromal and cancer cells through CXCL12 inhibition might restore hyperthermia sensitivity in ovarian cancers, and thus improve HIPEC efficiency.


Subject(s)
Mesenchymal Stem Cells/physiology , Ovarian Neoplasms/pathology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Survival , Chemokine CXCL12/antagonists & inhibitors , Coculture Techniques , Female , Flow Cytometry , Genes, Reporter , Green Fluorescent Proteins/genetics , Hot Temperature , Humans , Hyperthermia, Induced , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/mortality , Receptors, CXCR4/antagonists & inhibitors , Survival Rate
8.
Scand J Infect Dis ; 38(9): 794-9, 2006.
Article in English | MEDLINE | ID: mdl-16938734

ABSTRACT

In the absence of egg excretion, laboratory diagnosis of recently acquired schistosomiasis is dependent on serology. 42 of 83 Swedish adventure tourists to sub-Saharan Africa had serum anti-schistosome antibodies indicating recent infection. There is little doubt regarding the specificity and sensitivity of serology for the demonstration of infection, but there is a need for alternative serological methods which could be more widely used than the standard immunofluorescence assay (IFA) for antibodies against gut-derived antigens (anti-GAA). We present results suggesting that 40/42 anti-GAA positive cases also react with keyhole limpet haemocyanin (KLH), a readily available commercial antigen. High anti-GAA titres were seen for more than 2 y despite treatment with praziquantel. Thus we are faced with several questions. How likely is it that positive serology means treatment failure? What is the risk involved in chronic infection? What is the prospect for monitoring treatment outcome by serology? We conclude that there is a need for better information on the risk of becoming infected, for improved methods for testing and for monitoring the therapeutic effects in adventure tourists.


Subject(s)
Antibodies, Helminth/blood , Schistosoma/immunology , Schistosomiasis/diagnosis , Travel , Adolescent , Adult , Africa South of the Sahara , Aged , Animals , Drug Monitoring , Female , Fluorescent Antibody Technique, Indirect , Hemocyanins , Humans , Male , Middle Aged , Praziquantel/therapeutic use , Schistosomiasis/drug therapy , Schistosomiasis/immunology , Serologic Tests , Sweden , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL