Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Genes Dev ; 28(23): 2621-35, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25452273

RESUMO

Increased PI 3-kinase (PI3K) signaling in pancreatic ductal adenocarcinoma (PDAC) correlates with poor prognosis, but the role of class I PI3K isoforms during its induction remains unclear. Using genetically engineered mice and pharmacological isoform-selective inhibitors, we found that the p110α PI3K isoform is a major signaling enzyme for PDAC development induced by a combination of genetic and nongenetic factors. Inactivation of this single isoform blocked the irreversible transition of exocrine acinar cells into pancreatic preneoplastic ductal lesions by oncogenic Kras and/or pancreatic injury. Hitting the other ubiquitous isoform, p110ß, did not prevent preneoplastic lesion initiation. p110α signaling through small GTPase Rho and actin cytoskeleton controls the reprogramming of acinar cells and regulates cell morphology in vivo and in vitro. Finally, p110α was necessary for pancreatic ductal cancers to arise from Kras-induced preneoplastic lesions by increasing epithelial cell proliferation in the context of mutated p53. Here we identify an in vivo context in which p110α cellular output differs depending on the epithelial transformation stage and demonstrate that the PI3K p110α is required for PDAC induced by oncogenic Kras, the key driver mutation of PDAC. These data are critical for a better understanding of the development of this lethal disease that is currently without efficient treatment.


Assuntos
Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/fisiopatologia , Classe Ia de Fosfatidilinositol 3-Quinase/genética , Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/fisiopatologia , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Animais , Animais Geneticamente Modificados , Proliferação de Células , Células Epiteliais/citologia , Inativação Gênica , Humanos , Camundongos , Mutação , Proteínas Proto-Oncogênicas p21(ras)/genética , Transdução de Sinais
2.
Int J Mol Sci ; 23(19)2022 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-36233054

RESUMO

Bladder cancer is the 10th most common cancer in the world and has a high risk of recurrence and metastasis. In order to sustain high energetic needs, cancer cells undergo complex metabolic adaptations, such as a switch toward aerobic glycolysis, that can be exploited therapeutically. Reactive oxygen species (ROS) act as key regulators of cancer metabolic reprogramming and tumorigenesis, but the sources of ROS remain unidentified. Monoamine oxidases (MAOs) are mitochondrial enzymes that generate H2O2 during the breakdown of catecholamines and serotonin. These enzymes are particularly important in neurological disorders, but recently, a new link between MAOs and cancer has been uncovered, involving their production of ROS. At present, the putative role of MAOs in bladder cancer has never been evaluated. We observed that human urothelial tumor explants and the bladder cancer cell line AY27 expressed both MAO-A and MAO-B isoforms. Selective inhibition of MAO-A or MAO-B limited mitochondrial ROS accumulation, cell cycle progression and proliferation of bladder cancer cells, while only MAO-A inhibition prevented cell motility. To test whether ROS contributed to MAO-induced tumorigenesis, we used a mutated form of MAO-A which was unable to produce H2O2. Adenoviral transduction of the WT MAO-A stimulated the proliferation and migration of AY27 cells while the Lys305Met MAO-A mutant was inactive. This was consistent with the fact that the antioxidant Trolox strongly impaired proliferation and cell cycle progression. Most interestingly, AY27 cells were highly dependent on glucose metabolism to sustain their growth, and MAO inhibitors potently reduced glycolysis and oxidative phosphorylation, due to pyruvate depletion. Accordingly, MAO inhibitors decreased the expression of proteins involved in glucose transport (GLUT1) and transformation (HK2). In conclusion, urothelial cancer cells are characterized by a metabolic shift toward glucose-dependent metabolism, which is important for cell growth and is under the regulation of MAO-dependent oxidative stress.


Assuntos
Carcinoma , Neoplasias da Bexiga Urinária , Antioxidantes/metabolismo , Carcinogênese/metabolismo , Carcinoma/metabolismo , Catecolaminas/metabolismo , Proliferação de Células , Glucose/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Humanos , Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/farmacologia , Monoaminoxidase/genética , Monoaminoxidase/metabolismo , Inibidores da Monoaminoxidase/metabolismo , Inibidores da Monoaminoxidase/farmacologia , Estresse Oxidativo , Piruvatos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Serotonina/metabolismo , Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/metabolismo
3.
Int J Mol Sci ; 21(11)2020 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-32481768

RESUMO

INTRODUCTION: Multiple myeloma (MM) is a B-cell neoplasm characterized by clonal expansion of malignant plasma cells (MM cells) in the bone-marrow (BM) compartment. BM mesenchymal stromal cells (MSC) from newly diagnosed MM patients were shown to be involved in MM pathogenesis and chemoresistance. The patients displayed a distinct transcriptome and were functionally different from healthy donors' (HD) MSC. Our aim was to determine whether MM-MSC also contributed to relapse. METHODS: We obtained and characterized patients' MSC samples at diagnosis, two years after intensive treatment, without relapse and at relapse. RESULTS: Transcriptomic analysis revealed differences in gene expression between HD and MM-MSC, whatever the stage of the disease. An easier differentiation towards adipogenesis at the expense of osteoblatogeneis was observed, even in patients displaying a complete response to treatment. Although their transcriptome was similar, we found that MSC from relapsed patients had an increased immunosuppressive ability, compared to those from patients in remission. CONCLUSION: We demonstrated that imprinting of MSC transcriptome demonstrated at diagnosis of MM, persisted even after the apparent disappearance of MM cells induced by treatment, suggesting the maintenance of a local context favorable to relapse.


Assuntos
Regulação Neoplásica da Expressão Gênica , Impressão Genômica , Células-Tronco Mesenquimais/metabolismo , Mieloma Múltiplo/metabolismo , Transcriptoma , Adipogenia , Idoso , Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Diferenciação Celular , Técnicas de Cocultura , Resistencia a Medicamentos Antineoplásicos , Feminino , Perfilação da Expressão Gênica , Humanos , Imunossupressores/farmacologia , Leucócitos Mononucleares/metabolismo , Masculino , Pessoa de Meia-Idade , Mieloma Múltiplo/terapia , Recidiva Local de Neoplasia , Análise de Componente Principal , Recidiva , Linfócitos T/citologia
4.
Int J Mol Sci ; 20(19)2019 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-31547627

RESUMO

The tumor bulk is composed of a highly heterogeneous population of cancer cells, as well as a large variety of resident and infiltrating host cells, extracellular matrix proteins, and secreted proteins, collectively known as the tumor microenvironment (TME). The TME is essential for driving tumor development by promoting cancer cell survival, migration, metastasis, chemoresistance, and the ability to evade the immune system responses. Therapeutically targeting tumor-associated macrophages (TAMs), cancer-associated fibroblasts (CAFs), regulatory T-cells (T-regs), and mesenchymal stromal/stem cells (MSCs) is likely to have an impact in cancer treatment. In this review, we focus on describing the normal physiological functions of each of these cell types and their behavior in the cancer setting. Relying on the specific surface markers and secreted molecules in this context, we review the potential targeting of these cells inducing their depletion, reprogramming, or differentiation, or inhibiting their pro-tumor functions or recruitment. Different approaches were developed for this targeting, namely, immunotherapies, vaccines, small interfering RNA, or small molecules.


Assuntos
Imunoterapia , Neoplasias , Microambiente Tumoral/imunologia , Fibroblastos Associados a Câncer/imunologia , Fibroblastos Associados a Câncer/patologia , Diferenciação Celular/imunologia , Humanos , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/patologia , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias/terapia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia
5.
Cancer Metastasis Rev ; 33(1): 17-39, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24357056

RESUMO

Ovarian adenocarcinoma is characterized by a late detection, dissemination of cancer cells into the whole peritoneum, and the frequent acquisition of chemoresistance. If these particularities can be explained in part by intrinsic properties of ovarian cancer cells, an increased number of studies show the importance of the tumor microenvironment in tumor progression. Ovarian cancer cells can regulate the composition of their stroma in promoting the formation of ascitic fluid, rich in cytokines and bioactive lipids, and in stimulating the differentiation of stromal cells into a pro-tumoral phenotype. In return, cancer-associated fibroblasts, cancer-associated mesenchymal stem cells, tumor-associated macrophages, or other peritoneal cells, such as adipocytes and mesothelial cells can regulate tumor growth, angiogenesis, dissemination, and chemoresistance. This review focuses on the current knowledge about the roles of stromal cells and the associated secreted factors on tumor progression. We also summarize the different studies showing that targeting the microenvironment represents a great potential for improving the prognosis of patients with ovarian adenocarcinoma.


Assuntos
Adenocarcinoma/patologia , Neoplasias Ovarianas/patologia , Microambiente Tumoral , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/metabolismo , Antineoplásicos/uso terapêutico , Citocinas/metabolismo , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/metabolismo , Células Estromais/metabolismo , Células Estromais/patologia
6.
BMC Cancer ; 15: 569, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26231656

RESUMO

BACKGROUND: The interaction of SDF-1alpha with its receptor CXCR4 plays a role in the occurrence of distant metastasis in many solid tumors. This interaction increases migration from primary sites as well as homing at distant sites. METHODS: Here we investigated how SDF-1α could modulate both migration and adhesion of cancer cells through the modulation of RhoGTPases. RESULTS: We show that different concentrations of SDF-1α modulate the balance of adhesion and migration in cancer cells. Increased migration was obtained at 50 and 100 ng/ml of SDF-1α; however migration was reduced at 200 ng/ml. The adhesion between breast cancer cells and BMHC was significantly increased by SDF-1α treatment at 200 ng/ml and reduced using a blocking monoclonal antibody against CXCR4. We showed that at low SDF-1α concentration, RhoA was activated and overexpressed, while at high concentration Rac1 was promoting SDF-1α mediating-cell adhesion. CONCLUSION: We conclude that SDF-1α concentration modulates migration and adhesion of breast cancer cells, by controlling expression and activation of RhoGTPases.


Assuntos
Neoplasias da Mama/metabolismo , Quimiocina CXCL12/farmacologia , Células Estromais/fisiologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Quimiocina CXCL12/metabolismo , Técnicas de Cocultura , Feminino , Humanos , Células MCF-7 , Células Estromais/citologia
7.
Bull Cancer ; 111(2): 164-175, 2024 Feb.
Artigo em Francês | MEDLINE | ID: mdl-37689530

RESUMO

Clinical research in the area of cancer is of utmost importance in order to improve patient care, both in terms of overall survival and quality of life. The implementation of clinical trials on medicinal products, now falling under EU Regulation 536/2014, is conditioned on prior scientific authorisation from the French National Agency for the Safety of Medicines and Health Products and a favorable ethical opinion from a Research Ethics Committee (REC). OBJECTIVE: The objective of this work is to report on the main problematic issues identified during the evaluation of oncology dossiers by the REC in order to present the expected elements and thus optimise the evaluation procedures. METHODS: The National Conference of the Research Ethics Committees analysed the questions raised by the REC during their evaluation of clinical trials of oncology drugs submitted to the European information system in 2022. RESULTS: Out of a total of fourteen dossiers, nine were subject to ethical questions on the protocol and all dossiers required modifications to the information documents. DISCUSSION: The heterogeneous quality of the dossiers reminds the need to submit well-argued, methodologically robust protocols with supervised research procedures that are safe for the participants. The drafting of information documents needs to be thoroughly reconsidered in order to present clear, concise, loyal and respectful documents for patients' rights.


Assuntos
Neoplasias , Qualidade de Vida , Humanos , Comitês de Ética em Pesquisa , Oncologia , Neoplasias/terapia
8.
Breast Cancer Res ; 15(1): R6, 2013 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-23339407

RESUMO

INTRODUCTION: RhoB has been reported to exert positive and negative effects on cancer pathophysiology but an understanding of its role in breast cancer remains incomplete. Analysis of data from the Oncomine database showed a positive correlation between RhoB expression and positivity for both estrogen receptor alpha (ERα) and progesterone receptor (PR). METHODS: This finding was validated by our analysis of a tissue microarray constructed from a cohort of 113 patients and then investigated in human cell models. RESULTS: We found that RhoB expression in tissue was strongly correlated with ERα and PR expression and inversely correlated with tumor grade, tumor size and count of mitosis. In human breast cancer cell lines, RhoB attenuation was associated with reduced expression of both ERα and PR, whereas elevation of RhoB was found to be associated with ERα overexpression. Mechanistic investigations suggested that RhoB modulates ERα expression, controlling both its protein and mRNA levels, and that RhoB modulates PR expression by accentuating the recruitment of ERα and other major co-regulators to the promoter of PR gene. A major consequence of RhoB modulation was that RhoB differentially regulated the proliferation of breast cancer cell lines. Interestingly, we documented crosstalk between RhoB and ERα, with estrogen treatment leading to RhoB activation. CONCLUSION: Taken together, our findings offer evidence that in human breast cancer RhoB acts as a positive function to promote expression of ERα and PR in a manner correlated with cell proliferation.


Assuntos
Neoplasias da Mama/genética , Receptor alfa de Estrogênio/biossíntese , Receptores de Progesterona/biossíntese , Proteína rhoB de Ligação ao GTP/biossíntese , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , RNA Mensageiro/biossíntese , Análise Serial de Tecidos
9.
Nucleic Acids Res ; 39(22): 9605-19, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21880593

RESUMO

In mammalian cells, the main pathway for DNA double-strand breaks (DSBs) repair is classical non-homologous end joining (C-NHEJ). An alternative or back-up NHEJ (B-NHEJ) pathway has emerged which operates preferentially under C-NHEJ defective conditions. Although B-NHEJ appears particularly relevant to genomic instability associated with cancer, its components and regulation are still largely unknown. To get insights into this pathway, we have knocked-down Ku, the main contributor to C-NHEJ. Thus, models of human cell lines have been engineered in which the expression of Ku70/80 heterodimer can be significantly lowered by the conditional induction of a shRNA against Ku70. On Ku reduction in cells, resulting NHEJ competent protein extracts showed a shift from C- to B-NHEJ that could be reversed by addition of purified Ku protein. Using a cellular fractionation protocol after treatment with a strong DSBs inducer followed by western blotting or immunostaining, we established that, among C-NHEJ factors, Ku is the main counteracting factor against mobilization of PARP1 and the MRN complex to damaged chromatin. In addition, Ku limits PAR synthesis and single-stranded DNA production in response to DSBs. These data support the involvement of PARP1 and the MRN proteins in the B-NHEJ route for the repair of DNA DSBs.


Assuntos
Antígenos Nucleares/metabolismo , Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades , Proteínas de Ligação a DNA/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Antígenos Nucleares/genética , Fracionamento Celular , Linhagem Celular , Cromatina/química , Cromatina/metabolismo , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Técnicas de Silenciamento de Genes , Humanos , Autoantígeno Ku , Poli(ADP-Ribose) Polimerase-1 , Inibidores de Poli(ADP-Ribose) Polimerases
10.
Therapie ; 78(3): 247-257, 2023.
Artigo em Francês | MEDLINE | ID: mdl-36031434

RESUMO

More and more practitioners are being led to propose to their patients an analysis of their genetic characteristics (structural or functional genomics) either to confirm a diagnosis or to adapt a treatment (personalized medicine). Patients treated in hospitals are also offered analyses of their genetic characteristics either because they have not objected to their biological samples taken during care being used for research, or because they are included in a clinical study. The revised bioethics laws for application on 1/1/2021 provide a complete and clear framework for practices concerning genetics. However, most patients or healthy volunteers who have agreed (or not objected) to genetic testing are not aware of the potential consequences for them and their families. This is mostly due to a lack of information (inclusion of a sample without seeking express consent) or partial information due to either a lack of training of the prescriber himself or, in the context of clinical trials, an inadequate information leaflet. In order to help prescribers and evaluators of clinical trial applications to propose fair and clear information and consent letters to patients and thus ensure patient protection, we have produced an exegesis of the legal and ethical elements on which to base a proposal for genetic characteristics analysis. We include examples of requests for analysis of genetic characteristics, specifying the cases in which the research is authorized or not and mentioning the corresponding legal texts. We have generated here a useful pedagogical tool for all prescribers, researchers or simply French citizens.

11.
Bull Cancer ; 110(10): 1002-1014, 2023 Oct.
Artigo em Francês | MEDLINE | ID: mdl-37532643

RESUMO

INTRODUCTION: More and more French cancer patients are offered by their physicians having their genetic characteristics analyzed (diagnosis, adaptation of treatment plans, etc.). In oncology, considering the development of personalized medicine, these analyses are commonplace. Analyses of germline (hereditary) genetic characteristics require information from patients who must sign an informed consent (article 16.10 of the Civil Code and articles L. 1131-3 and L. 1122-1-1 of the Public Health Code). However, prescribing physicians are rarely geneticists and have little training in genetics. Patients report that few are able to answer their questions and often sign a consent that is not truly informed. METHODS: To identify the genetic knowledge and training needs of prescribers, we conducted an online survey of physicians prescribing genetic testing in oncology between January and March 2020. The survey consisted of 17 closed questions and 3 open questions. RESULTS: We obtained 35 usable questionnaires which show that 50% of the prescribing physicians questioned lack knowledge of genetics, but do not express a need for training. They were interested in the provision of a digital teaching aid for patients. DISCUSSION: We have therefore made a film for patients, available in free access, which aims to shed light on the analysis of genetic characteristics. The film helps physicians to explain the offered analyses and their consequences (https://youtu.be/5lWUSsteavs).

12.
J Immunother Cancer ; 11(8)2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37586764

RESUMO

BACKGROUND: Ovarian adenocarcinoma (OVAD) frequently metastasizes to the peritoneal cavity and manifests by the formation of ascites, which constitutes a tumor-promoting microenvironment. In the peritoneal cavity, two developmentally, phenotypically and functionally distinct macrophage subsets, immunocompetent large peritoneal macrophages (LPM) and immunosuppressive small peritoneal macrophages (SPM), coexist. Because peroxisome proliferator-activated receptor γ (PPARγ) is a critical factor participating in macrophage differentiation and cooperates with CCAAT/enhancer binding protein ß (C/EBPß), a transcription factor essential for SPM-to-LPM differentiation, PPARγ could be also involved in the regulation of SPM/LPM balance and could be a promising therapeutic target. METHODS: To evaluate the 15(S)-hydroxyeicosatetraenoic acid (HETE), a PPARγ endogenous ligand, impact on ovarian tumor growth, we intraperitoneally injected 15(S)-HETE into a murine ovarian cancer model. This experimental model consists in the intraperitoneally injection of ID8 cells expressing luciferase into syngeneic C57BL/6 female mice. This ID8 orthotopic mouse model is a well-established experimental model of end-stage epithelial OVAD. Tumor progression was monitored using an in vivo imaging system. Peritoneal immune cells in ascites were analyzed by flow cytometry and cell sorting. To determine whether the impact of 15(S)-HETE in tumor development is mediated through the macrophages, these cells were depleted by injection of liposomal clodronate. To further dissect how 15(S)-HETE mediated its antitumor effect, we assessed the tumor burden in tumor-bearing mice in which the PPARγ gene was selectively disrupted in myeloid-derived cells and in mice deficient of the recombination-activating gene Rag2. Finally, to validate our data in humans, we isolated and treated macrophages from ascites of individuals with OVAD. RESULTS: Here we show, in the murine experimental model of OVAD, that 15(S)-HETE treatment significantly suppresses the tumor growth, which is associated with the differentiation of SPM into LPM and the LPM residency in the peritoneal cavity. We demonstrate that C/EBPß and GATA6 play a central role in SPM-to-LPM differentiation and in LPM peritoneal residence through PPARγ activation during OVAD. Moreover, this SPM-to-LPM switch is associated with the increase of the effector/regulatory T-cell ratio. Finally, we report that 15(S)-HETE attenuates immunosuppressive properties of human ovarian tumor-associated macrophages from ascites. CONCLUSION: Altogether, these results promote PPARγ as a potential therapeutic target to restrain OVAD development and strengthen the use of PPARγ agonists in anticancer therapy.


Assuntos
Adenocarcinoma , Neoplasias Ovarianas , PPAR gama , Animais , Feminino , Humanos , Camundongos , Ascite , Carcinoma Epitelial do Ovário , Terapia de Imunossupressão , Imunossupressores , Macrófagos Peritoneais , Camundongos Endogâmicos C57BL , Neoplasias Ovarianas/tratamento farmacológico , Microambiente Tumoral
13.
Int J Mol Sci ; 13(8): 9545-9571, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22949815

RESUMO

Tumor development principally occurs following the accumulation of genetic and epigenetic alterations in tumor cells. These changes pave the way for the transformation of chemosensitive cells to chemoresistant ones by influencing the uptake, metabolism, or export of drugs at the cellular level. Numerous reports have revealed the complexity of tumors and their microenvironment with tumor cells located within a heterogeneous population of stromal cells. These stromal cells (fibroblasts, endothelial or mesothelial cells, adipocytes or adipose tissue-derived stromal cells, immune cells and bone marrow-derived stem cells) could be involved in the chemoresistance that is acquired by tumor cells via several mechanisms: (i) cell-cell and cell-matrix interactions influencing the cancer cell sensitivity to apoptosis; (ii) local release of soluble factors promoting survival and tumor growth (crosstalk between stromal and tumor cells); (iii) direct cell-cell interactions with tumor cells (crosstalk or oncologic trogocytosis); (iv) generation of specific niches within the tumor microenvironment that facilitate the acquisition of drug resistance; or (v) conversion of the cancer cells to cancer-initiating cells or cancer stem cells. This review will focus on the implication of each member of the heterogeneous population of stromal cells in conferring resistance to cytotoxins and physiological mediators of cell death.


Assuntos
Morte Celular , Resistencia a Medicamentos Antineoplásicos , Neoplasias/patologia , Células Estromais/patologia , Microambiente Tumoral , Animais , Humanos , Neoplasias/tratamento farmacológico , Transdução de Sinais , Células Estromais/efeitos dos fármacos , Células Tumorais Cultivadas
14.
Gynecol Oncol ; 122(1): 155-62, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21463889

RESUMO

OBJECTIVE: Improvement of the management and outcome of ovarian cancers may require intraoperative detection and therapeutic intervention to treat minimal residual disease after complete surgery. The aim of this study was to validate the importance of fluorescence in the peroperative detection of human ovarian adenocarcinoma cells and to determine its efficiency in detecting infra millimetric tumor metastases. METHODS: A fluorescent RAFT-(cRGD)4 tracer molecule (AngioStamp®) was used. The tracer is based on a biomarker, which has a very high affinity for the α(v)ß3 integrin, which is overexpressed in a large ratio of cancer cells and neovessel endothelial cells during angiogenesis. Infrared fluorescence was visualized with Fluobeam®, an open fluorescent imaging system that could potentially be used in peroperative conditions in the future. RESULTS: This novel technique allowed the specific detection of residual tumor deposits and inframillimetric metastases, smaller than 500µm, which were resected under fluorescent guidance. AngioStamp® was able to detect all types of cell lines, derived from human ovarian adenocarcinomas, before or after chemotherapy treatment in animals. The effectiveness of AngioStamp® for the detection of various human ovarian adenocarcinomas was assessed on 10 different fragments of tumor, implanted subcutaneously in nude mice. All implanted tumor fragments were visualized by AngioStamp®. CONCLUSIONS: The high rate of recurrence after apparently complete surgery and/or complete clinical response to chemotherapy implies that most patients have undetected minimal residual disease. Novel techniques such as laparoscopic or laparotomic fluorescence may prove to be crucial in reassessing the definition of primary outcome in ovarian cancer management.


Assuntos
Adenocarcinoma/patologia , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/cirurgia , Neoplasias Peritoneais/patologia , Neoplasias Peritoneais/cirurgia , Adenocarcinoma/cirurgia , Animais , Linhagem Celular Tumoral , Diagnóstico por Imagem , Feminino , Fluorescência , Humanos , Período Intraoperatório , Camundongos , Camundongos Nus , Camundongos Transgênicos , Neoplasia Residual , Transplante Heterólogo
15.
Int J Cancer ; 126(9): 2090-101, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-19739074

RESUMO

The microenvironment is known to play a dominant role in cancer progression. Cells closely associated with tumoral cells, named hospicells, have been recently isolated from the ascites of ovarian cancer patients. Whilst these cells present no specific markers from known cell lineages, they do share some homology with bone marrow-derived or adipose tissue-derived human mesenchymal stem cells (CD9, CD10, CD29, CD146, CD166, HLA-1). We studied the role of hospicells in ovarian carcinoma progression. In vitro, these cells had no effect on the growth of human ovarian carcinoma cell lines OVCAR-3, SKOV-1 and IGROV-1. In vivo, their co-injection with adenocarcinoma cells enhanced tumor growth whatever the tumor model used (subcutaneous and intraperitoneally established xenografts in athymic mice). In addition, their injection increased the development of ascites in tumor-bearing mice. Fluorescent macroscopy revealed an association between hospicells and ovarian adenocarcinoma cells within the tumor mass. Tumors obtained by coinjection of hospicells and human ovarian adenocarcinoma cells presented an increased microvascularization indicating that the hospicells could promote tumorigenicity of ovarian tumor cells in vivovia their action on angiogenesis. This effect on angiogenesis could be attributed to the increased HIF1alpha and VEGF expression associated with the presence of the hospicells. Collectively, these data indicate a role for these ascite-derived stromal cells in promoting tumor growth by increasing angiogenesis.


Assuntos
Ascite/patologia , Neoplasias/etiologia , Neovascularização Patológica/etiologia , Células Estromais/fisiologia , Adenocarcinoma/patologia , Animais , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Camundongos , Transplante de Neoplasias , Neoplasias Ovarianas/patologia , Fenótipo , Transplante Heterólogo , Fator A de Crescimento do Endotélio Vascular/fisiologia
16.
Electrophoresis ; 31(2): 396-402, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20084633

RESUMO

An analytical methodology for quality control analyses of IgG and their impurities is presented using a new UV-LIF (266 nm) detector inside the cassette of a CE instrument and its performance was evaluated. The observed sensitivity was very close to that obtained by silver staining of slab gels (LOD of 25 ng/mL), while the sensitivity of the analysis is 80 times better than with CE/UV absorption (214 nm). Examples of the analysis of pharmaceutical and other commercial IgGs are provided and the kinetics of the reduction of IgG by beta-mercaptoethanol is reported, demonstrating the ease of performing the analysis.


Assuntos
Contaminação de Medicamentos , Eletroforese Capilar/métodos , Preparações Farmacêuticas/química , Espectrometria de Fluorescência/métodos , Animais , Imunoglobulina G/química , Desnaturação Proteica , Coelhos , Ratos , Sensibilidade e Especificidade , Coloração pela Prata
17.
Med Sci (Paris) ; 36(3): 264-270, 2020 Mar.
Artigo em Francês | MEDLINE | ID: mdl-32228846

RESUMO

Star Wars, a "general public" film saga, raises questions about human nature and transhumanism. It features different characters who are neither "real" humans nor robots; there are creatures that can be likened to advanced humans (cyborgs, chimeras or genetically-modified humans). Based on the "Star Wars" movie, we will approach some ways of modifying the human person both in his body and in his consciousness and we will wonder about the man of tomorrow by asking ourselves if George Lucas (director of the first film released) might have not been a visionary of the men of tomorrow.


TITLE: George Lucas : Prophète du transhumanisme ? ABSTRACT: La Guerre des étoiles, saga cinématographique « grand public ¼, permet de se questionner sur la nature humaine et sur le transhumanisme. Elle met en effet en scène différents personnages qui ne sont ni de « vrais ¼ humains, ni des robots, des créatures pouvant être assimilées à des humains évolués (des cyborgs, des chimères ou des humains génétiquement modifiés). À partir de cette approche cinématographique, nous aborderons certains moyens de modification de la personne humaine à la fois dans son corps et dans sa conscience, et nous nous interrogerons sur l'homme de demain, en se demandant si George Lucas (réalisateur du premier film) n'était pas un visionnaire des hommes de demain.


Assuntos
Pessoas Famosas , Obras de Ficção como Assunto , Humanismo , Filmes Cinematográficos , Ciência nas Artes , Animais , California , Meio Ambiente Extraterreno , Melhoramento Genético , História do Século XX , História do Século XXI , Características Humanas , Humanismo/história , Humanos , Camundongos , Filmes Cinematográficos/história , Filosofia , Ciência nas Artes/história , Pesquisa com Células-Tronco/história
18.
J Mol Cell Biol ; 12(3): 202-215, 2020 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-31504643

RESUMO

Factors released by surrounding cells such as cancer-associated mesenchymal stromal cells (CA-MSCs) are involved in tumor progression and chemoresistance. In this study, we characterize the mechanisms by which naïve mesenchymal stromal cells (MSCs) can acquire a CA-MSCs phenotype. Ovarian tumor cells trigger the transformation of MSCs to CA-MSCs by expressing pro-tumoral genes implicated in the chemoresistance of cancer cells, resulting in the secretion of high levels of CXC chemokine receptors 1 and 2 (CXCR1/2) ligands such as chemokine (C-X-C motif) ligand 1 (CXCL1), CXCL2, and interleukin 8 (IL-8). CXCR1/2 ligands can also inhibit the immune response against ovarian tumor cells. Indeed, through their released factors, CA-MSCs promote the differentiation of monocytes towards M2 macrophages, which favors tumor progression. When CXCR1/2 receptors are inhibited, these CA-MSC-activated macrophages lose their M2 properties and acquire an anti-tumoral phenotype. Both ex vivo and in vivo, we used a CXCR1/2 inhibitor to sensitize ovarian tumor cells to carboplatin and circumvent the pro-tumoral effects of CA-MSCs. Since high concentrations of CXCR1/2 ligands in patients' blood are associated with chemoresistance, CXCR1/2 inhibition could be a potential therapeutic strategy to revert carboplatin resistance.


Assuntos
Comunicação Celular , Resistencia a Medicamentos Antineoplásicos , Fatores Imunológicos/biossíntese , Células-Tronco Mesenquimais/metabolismo , Neoplasias/metabolismo , Animais , Antineoplásicos/farmacologia , Biomarcadores , Biópsia , Diferenciação Celular , Linhagem Celular Tumoral , Biologia Computacional , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Perfilação da Expressão Gênica , Humanos , Imunomodulação , Macrófagos/imunologia , Macrófagos/metabolismo , Células-Tronco Mesenquimais/citologia , Camundongos , Modelos Biológicos , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Receptores CXCR/genética , Receptores CXCR/metabolismo , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
19.
J Immunother Cancer ; 8(1)2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32503947

RESUMO

BACKGROUND: Besides the interest of an early detection of ovarian cancer, there is an urgent need for new predictive and prognostic biomarkers of tumor development and cancer treatment. In healthy patients, circulating blood monocytes are typically subdivided into classical (85%), intermediate (5%) and non-classical (10%) populations. Although these circulating monocyte subsets have been suggested as biomarkers in several diseases, few studies have investigate their potential as a predictive signature for tumor immune status,tumor growth and treatment adaptation. METHODS: In this study, we used a homogeneous cohort of 28 chemotherapy-naïve patients with ovarian cancer to evaluate monocyte subsets as biomarkers of the ascites immunological status. We evaluated the correlations between circulating monocyte subsets and immune cells and tumor burden in peritoneal ascites. Moreover, to validate the use of circulating monocyte subsets tofollow tumor progression and treatment response, we characterized blood monocytes from ovarian cancer patients included in a phase 1 clinical trial at baseline and following murlentamab treatment. RESULTS: We demonstrate here a robust expansion of the intermediate blood monocytes (IBMs) in ovarian cancer patients. We establish a significant positive correlation between IBM percentage and tumor-associate macrophages with a CCR2high/CD163high/CD206high/CD86lowprofile. Moreover, IBM expansion is associated with a decreased effector/regulatory T-cell ratio in ascites and with the presence of soluble immunosuppressive mediators. We also establish that IBM proportion positively correlates with the peritoneum tumor burden. Finally, the study of IBMs in patients with ovarian cancer under immunotherapy during the phase clinical trial supports IBMs to follow the evolution of tumor development and the treatment adaptation. CONCLUSIONS: This study, which links IBM level with immunosuppression and tumor burden in peritoneum, identifies IBMs as apotential predictive signature of ascites immune status and as a biomarker ofovarian cancer development and treatment response. TRIAL REGISTRATION NUMBER: EudraCT: 2015-004252-22 NCT02978755.


Assuntos
Ascite/genética , Biomarcadores Tumorais/metabolismo , Imunoterapia/métodos , Receptores de Lipopolissacarídeos/metabolismo , Monócitos/metabolismo , Receptores de IgG/metabolismo , Progressão da Doença , Feminino , Humanos , Masculino , Microambiente Tumoral
20.
Cancer Immunol Res ; 7(2): 321-334, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30610060

RESUMO

Macrophage-mediated cytotoxicity is controlled by surface receptor expression and activation. Despite the numerous studies documenting the role of macrophage C-type lectin receptors (CLR) in pathogen elimination, little is known about their contribution to antitumor responses. Here, we report that IL13 inhibits T-cell lymphoma and ovarian adenocarcinoma development in tumor-bearing mice through the conversion of tumor-supporting macrophages to cytotoxic effectors, characterized by a CLR signature composed of dectin-1 and mannose receptor (MR). We show that dectin-1 and MR are critical for the recognition of tumor cells through sialic acid-specific glycan structure on their surface and for the subsequent activation of macrophage tumoricidal response. Finally, we validated that IL13 antitumor effect mediated by dectin-1 and MR overexpression on macrophages can extend to various types of human tumors. Therefore, these results identify these CLRs as potential targets to promote macrophage antitumor response and represent an attractive approach to elicit tumor-associated macrophage tumoricidal properties.


Assuntos
Interleucina-13/genética , Lectinas Tipo C/genética , Macrófagos/imunologia , Macrófagos/metabolismo , Lectinas de Ligação a Manose/genética , Neoplasias/etiologia , Neoplasias/metabolismo , Receptores de Superfície Celular/genética , Animais , Arginase/metabolismo , Linhagem Celular Tumoral , Expressão Gênica , Humanos , Interleucina-13/metabolismo , Lectinas Tipo C/metabolismo , Receptor de Manose , Lectinas de Ligação a Manose/metabolismo , Camundongos , Camundongos Knockout , Ácido N-Acetilneuramínico/metabolismo , Necrose/genética , Necrose/imunologia , Neoplasias/mortalidade , Neoplasias/patologia , Prognóstico , Espécies Reativas de Oxigênio/metabolismo , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Linfócitos T/imunologia , Linfócitos T/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA