Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 21
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Int J Mol Sci ; 25(10)2024 May 17.
Article de Anglais | MEDLINE | ID: mdl-38791496

RÉSUMÉ

This Special Issue intends to underscore several topics related to cellular signaling transduction, considering the consensus that nowadays, the best scientific approach for the prevention, diagnosis, and treatment of several diseases is the study of the regulatory networks that determine the response to therapy and the maintenance of homeostasis or its dysregulation [...].


Sujet(s)
Exosomes , Vésicules extracellulaires , Immunothérapie , Tumeurs , Humains , Exosomes/métabolisme , Tumeurs/thérapie , Tumeurs/immunologie , Tumeurs/métabolisme , Immunothérapie/méthodes , Vésicules extracellulaires/métabolisme , Animaux , Transduction du signal
2.
Front Pharmacol ; 14: 1275833, 2023.
Article de Anglais | MEDLINE | ID: mdl-37841928

RÉSUMÉ

Introduction: Long non-coding RNA H19 (lncH19) is highly expressed in colorectal cancer (CRC) and plays critical roles in tumor development, proliferation, metastasis, and drug resistance. Indeed, the expression of lncH19 usually affects the outcomes of chemo-, endocrine, and targeted therapies. ITF2357 (givinostat) is a histone deacetylase inhibitor (HDACi) that revealed a significant anti-tumor action by inducing apoptosis in different tumor models, including leukemia, melanoma, and glioblastoma. However, no data are present in the literature regarding the use of this compound for CRC treatment. Here, we investigate the role of lncH19 in ITF2357-induced apoptosis in CRC cells. Methods: The HCT-116 CRC cell line was stably silenced for H19 to investigate the role of this lncRNA in ITF2357-induced cell death. Cell viability assays and flow cytometric analyses were performed to assess the anti-proliferative and pro-apoptotic effects of ITF2357 in CRC cell lines that are silenced or not for lncH19. RT-PCR and Western blot were used to study the effects of ITF2357 on autophagy and apoptosis markers. Finally, bioinformatics analyses were used to identify miRNAs targeting pro-apoptotic factors that can be sponged by lncH19. Results: ITF2357 increased the expression levels of H19 and reduced HCT-116 cell viability, inducing apoptosis, as demonstrated by the increase in annexin-V positivity, caspase 3 cleavage, and poly (ADP-ribose) polymerase (PARP-1) degradation. Interestingly, the apoptotic effect of ITF2357 was much less evident in lncH19-silenced cells. We showed that lncH19 plays a functional role in the pro-apoptotic activity of the drug by stabilizing TP53 and its transcriptional targets, NOXA and PUMA. ITF2357 also induced autophagy in CRC cells, which was interpreted as a pro-survival response not correlated with lncH19 expression. Furthermore, ITF2357 induced apoptosis in 5-fluorouracil-resistant HCT-116 cells that express high levels of lncH19. Conclusion: This study shows that lncH19 expression contributes to ITF2357-induced apoptosis by stabilizing TP53. Overall, we suggest that lncH19 expression may be exploited to favor HDACi-induced cell death and overcome 5-fluorouracil chemoresistance.

3.
Sci Rep ; 13(1): 17448, 2023 10 14.
Article de Anglais | MEDLINE | ID: mdl-37838795

RÉSUMÉ

Anomalous aortic origin of the coronary artery (AAOCA) is a rare disease associated with sudden cardiac death, usually related to physical effort in young people. Clinical routine tests fail to assess the ischemic risk, calling for novel diagnostic approaches. To this aim, some recent studies propose to assess the coronary blood flow (CBF) in AAOCA by computational simulations but they are limited by the use of data from literature retrieved from normal subjects. To overcome this limitation and obtain a reliable assessment of CBF, we developed a fully patient-specific lumped parameter model based on clinical imaging and in-vivo data retrieved during invasive coronary functional assessment of subjects with AAOCA. In such a way, we can estimate the CBF replicating the two hemodynamic conditions in-vivo analyzed. The model can mimic the effective coronary behavior with high accuracy and could be a valuable tool to quantify CBF in AAOCA. It represents the first step required to move toward a future clinical application with the aim of improving patient care. The study was registered at Clinicaltrial.gov with (ID: NCT05159791, date 2021-12-16).


Sujet(s)
Aorte , Coeur , Humains , Vaisseaux coronaires/imagerie diagnostique , Mort subite cardiaque , Hémodynamique
4.
Int J Mol Sci ; 24(11)2023 May 25.
Article de Anglais | MEDLINE | ID: mdl-37298188

RÉSUMÉ

Flavonoids are polyphenolic phytochemical compounds found in many plants, fruits, vegetables, and leaves. They have a multitude of medicinal applications due to their anti-inflammatory, antioxidative, antiviral, and anticarcinogenic properties. Furthermore, they also have neuroprotective and cardioprotective effects. Their biological properties depend on the chemical structure of flavonoids, their mechanism of action, and their bioavailability. The beneficial effects of flavonoids have been proven for a variety of diseases. In the last few years, it is demonstrated that the effects of flavonoids are mediated by inhibiting the NF-κB (Nuclear Factor-κB) pathway. In this review, we have summarized the effects of some flavonoids on the most common diseases, such as cancer, cardiovascular, and human neurodegenerative diseases. Here, we collected all recent studies describing the protective and prevention role of flavonoids derived from plants by specifically focusing their action on the NF-κB signaling pathway.


Sujet(s)
Tumeurs , Maladies neurodégénératives , Humains , Facteur de transcription NF-kappa B/métabolisme , Flavonoïdes/pharmacologie , Flavonoïdes/usage thérapeutique , Flavonoïdes/métabolisme , Maladies neurodégénératives/traitement médicamenteux , Transduction du signal , Tumeurs/traitement médicamenteux
5.
Cell Biol Int ; 47(3): 634-647, 2023 Mar.
Article de Anglais | MEDLINE | ID: mdl-36378586

RÉSUMÉ

Angiogenesis, a process characterized by the formation of new blood vessels from pre-existing ones, is a crucial step in tumor growth and dissemination. Given the ability of tumors to interfere with multiple or different molecular pathways to promote angiogenesis, there is an increasing need to therapeutically block tumor progression by targeting multiple antiangiogenic pathways. Natural polyphenols present health-protective properties, which are likely attributed to their ability to activate multiple pathways involved in inflammation, carcinogenesis, and angiogenesis. Recently, increased attention has been addressed to the ability of flavonoids, the most abundant polyphenols in the diet, to prevent cancer by suppressing angiogenesis. Here we investigate the mechanisms by which xanthohumol (the major prenylated flavonoid of the hop plant Humulus lupulus L.) and nobiletin (flavonoid from red-orange Citrus sinensis) can modulate the effects of Tumor Necrosis Factor-α (TNF-α) on human umbilical vein endothelial cells (HUVEC). The results reported in this paper show that xanthohumol and nobiletin pretreatment of HUVEC inhibits the effects induced by TNF-α on cell migration, invasion capability, and colon cancer cell adhesion on the endothelial monolayer. Moreover, the pretreatment reduces metalloproteinases and adhesion molecules' expression. Finally, our results highlight that xanthohumol and nobiletin can counteract the effects of TNF-α on angiogenesis and invasiveness, mainly through Vascular Endothelial Growth Factor and NF-κB pathways. Since angiogenesis plays an important pathological role in the progression of several diseases, our findings may provide clues for developing xanthohumol and nobiletin as therapeutic agents against angiogenesis-associated diseases.


Sujet(s)
Facteur de transcription NF-kappa B , Tumeurs , Humains , Facteur de transcription NF-kappa B/métabolisme , Facteur de nécrose tumorale alpha/pharmacologie , Facteur de nécrose tumorale alpha/métabolisme , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Flavonoïdes/pharmacologie , Transduction du signal , Tumeurs/métabolisme , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Polyphénols/métabolisme , Polyphénols/pharmacologie
6.
BMC Cancer ; 22(1): 567, 2022 May 20.
Article de Anglais | MEDLINE | ID: mdl-35596172

RÉSUMÉ

BACKGROUND: The uncontrolled proliferation of cancer cells determines hypoxic conditions within the neoplastic mass with consequent activation of specific molecular pathways that allow cells to survive despite oxygen deprivation. The same molecular pathways are often the cause of chemoresistance. This study aims to investigate the role of the hypoxia-induced miR-675-5p in 5-Fluorouracil (5-FU) resistance on colorectal cancer (CRC) cells. METHODS: CRC cell lines were treated with 5-Fu and incubated in normoxic or hypoxic conditions; cell viability has been evaluated by MTT assay. MiR-675-5p levels were analysed by RT-PCR and loss and gain expression of the miRNA has been obtained by the transfection of miRNA antagomir or miRNA mimic. Total protein expression of different apoptotic markers was analysed through western blot assay. MirWalk 2.0 database search engine was used to investigate the putative targets of the miR-675-5p involved in the apoptotic process. Finally, the luciferase assay was done to confirm Caspase-3 as a direct target of the miR-675-5p. RESULTS: Our data demonstrated that hypoxia-induced miR-675-5p counteracts the apoptotic signal induced by 5-FU, thus taking part in the drug resistance response. We showed that the apoptotic markers, cleaved PARP and cleaved caspase-3, increased combining miR-675-5p inhibition with 5-FU treatment. Moreover, we identified pro-caspase-3 among the targets of the miR-675-5p. CONCLUSION: Our data demonstrate that the inhibition of hypoxia-induced miR-675-5p combined with 5-FU treatment can enhances drug efficacy in both prolonged hypoxia and normoxia, indicating a possible strategy to partially overcome chemoresistance.


Sujet(s)
Tumeurs colorectales , Résistance aux médicaments antinéoplasiques , microARN , Apoptose/génétique , Caspase-3/métabolisme , Lignée cellulaire tumorale , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Résistance aux médicaments antinéoplasiques/génétique , Fluorouracil/pharmacologie , Fluorouracil/usage thérapeutique , Régulation de l'expression des gènes tumoraux , Humains , Hypoxie/génétique , Hypoxie/métabolisme , microARN/génétique , microARN/métabolisme
7.
Cells ; 10(12)2021 11 30.
Article de Anglais | MEDLINE | ID: mdl-34943863

RÉSUMÉ

In the last decade, an increasing number of studies have demonstrated that non-coding RNA (ncRNAs) cooperate in the gene regulatory networks with other biomolecules, including coding RNAs, DNAs and proteins. Among them, microRNAs (miRNAs), long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) are involved in transcriptional and translation regulation at different levels. Intriguingly, ncRNAs can be packed in vesicles, released in the extracellular space, and finally internalized by receiving cells, thus affecting gene expression also at distance. This review focuses on the mechanisms through which the ncRNAs can be selectively packaged into extracellular vesicles (EVs).


Sujet(s)
Vésicules extracellulaires/métabolisme , ARN/métabolisme , Animaux , Humains , Modèles biologiques , Protéines/métabolisme , ARN/génétique , Transport des ARN
8.
Int J Mol Sci ; 21(16)2020 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-32764403

RÉSUMÉ

The correct concentration of oxygen in all tissues is a hallmark of cellular wellness, and the negative regulation of oxygen homeostasis is able to affect the cells and tissues of the whole organism. The cellular response to hypoxia is characterized by the activation of multiple genes involved in many biological processes. Among them, hypoxia-inducible factor (HIF) represents the master regulator of the hypoxia response. The active heterodimeric complex HIF α/ß, binding to hypoxia-responsive elements (HREs), determines the induction of at least 100 target genes to restore tissue homeostasis. A growing body of evidence demonstrates that hypoxia signaling can act by generating contrasting responses in cells and tissues. Here, this dual and controversial role of hypoxia and the HIF signaling pathway is discussed, with particular reference to the effects induced on the complex activities of the immune system and on mechanisms determining cell and tissue responses after an injury in both acute and chronic human diseases related to the heart, lung, liver, and kidney.


Sujet(s)
Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Hypoxie cellulaire/génétique , Facteur-1 induit par l'hypoxie/génétique , Facteurs de transcription/génétique , Protéines de liaison à l'ADN/génétique , Humains , Protéines nucléaires/génétique , Oxygène/métabolisme , ARN messager/génétique , Transduction du signal/génétique
9.
Int J Mol Sci ; 20(4)2019 Feb 13.
Article de Anglais | MEDLINE | ID: mdl-30781795

RÉSUMÉ

The long non-coding RNA H19 (lncH19) is broadly transcribed in the first stage of development and silenced in most cells of an adult organism; it appears again in several tumors where, through different molecular mediators, promotes cell proliferation, motility and metastases. LncH19 has been associated with hypoxia-inducible factor 1-alpha (HIF-1α) activation and, in some tumors, it has proved to be necessary and required to sustain hypoxic responses. Here we propose to investigate a putative role for the lncH19 in hypoxia induced multiple myeloma (MM) progression. Transcriptional analysis of MM cell lines (RPMI and MM1.S) exposed to normoxia or hypoxia (1% O2) was done in order to evaluate lncH19 levels under hypoxic stimulation. Then, to investigate the role of lncH19 in hypoxia mediated MM progression, transcriptional, protein and functional assays have been performed on hypoxia stimulated MM cell lines, silenced or not for lncH19. Our data demonstrated that hypoxic stimulation in MM cell lines induced the overexpression of lncH19, which, in turn, is required for the expression of the hypoxia induced genes involved in MM dissemination, such as C-X-C Motif Chemokine Receptor 4 (CXCR4) and Snail. Moreover, adhesion assays demonstrated that lncH19 silencing abrogates the increased adhesion on stromal cells induced by the hypoxic condition. Finally, Western blot analysis indicated that lncH19 silencing impaired HIF1α nuclear translocation. The LncH19, required for the induction of hypoxic responses in MM cells, could represent a new therapeutic target for MM.


Sujet(s)
Hypoxie/génétique , Myélome multiple/génétique , Myélome multiple/anatomopathologie , ARN long non codant/métabolisme , Adhérence cellulaire , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Humains , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Modèles biologiques , ARN long non codant/génétique , Cellules stromales/métabolisme , Cellules stromales/anatomopathologie
10.
J Cell Mol Med ; 20(10): 1829-39, 2016 10.
Article de Anglais | MEDLINE | ID: mdl-27196940

RÉSUMÉ

Chronic myelogenous leukaemia (CML) is a clonal myeloproliferative disorder. Recent evidence indicates that altered crosstalk between CML and mesenchymal stromal cells may affect leukaemia survival; moreover, vesicles released by both tumour and non-tumour cells into the microenvironment provide a suitable niche for cancer cell growth and survival. We previously demonstrated that leukaemic and stromal cells establish an exosome-mediated bidirectional crosstalk leading to the production of IL8 in stromal cells, thus sustaining the survival of CML cells. Human cell lines used are LAMA84 (CML cells), HS5 (stromal cells) and bone marrow primary stromal cells; gene expression and protein analysis were performed by real-time PCR and Western blot. IL8 and MMP9 secretions were evaluated by ELISA. Exosomes were isolated from CML cells and blood samples of CML patients. Here, we show that LAMA84 and CML patients' exosomes contain amphiregulin (AREG), thus activating epidermal growth factor receptor (EGFR) signalling in stromal cells. EGFR signalling increases the expression of SNAIL and its targets, MMP9 and IL8. We also demonstrated that pre-treatment of HS5 with LAMA84 exosomes increases the expression of annexin A2 that promotes the adhesion of leukaemic cells to the stromal monolayer, finally supporting the growth and invasiveness of leukaemic cells. Leukaemic and stromal cells establish a bidirectional crosstalk: exosomes promote proliferation and survival of leukaemic cells, both in vitro and in vivo, by inducing IL8 secretion from stromal cells. We propose that this mechanism is activated by a ligand-receptor interaction between AREG, found in CML exosomes, and EGFR in bone marrow stromal cells.


Sujet(s)
Cellules de la moelle osseuse/métabolisme , Cellules de la moelle osseuse/anatomopathologie , Microenvironnement cellulaire , Récepteurs ErbB/métabolisme , Exosomes/métabolisme , Leucémie myéloïde chronique BCR-ABL positive/métabolisme , Leucémie myéloïde chronique BCR-ABL positive/anatomopathologie , Amphiréguline/métabolisme , Annexine A2/métabolisme , Adhérence cellulaire , Lignée cellulaire tumorale , Humains , Interleukine-8/métabolisme , Matrix metalloproteinase 9/génétique , Matrix metalloproteinase 9/métabolisme , Phosphorylation , ARN messager/génétique , ARN messager/métabolisme , Petit ARN interférent/métabolisme , Facteurs de transcription de la famille Snail/génétique , Facteurs de transcription de la famille Snail/métabolisme , Cellules stromales/métabolisme
11.
Biomed Res Int ; 2015: 821613, 2015.
Article de Anglais | MEDLINE | ID: mdl-26583135

RÉSUMÉ

In recent years the role of tumor microenvironment in the progression of hematological malignancies has been widely recognized. Recent studies have focused on how cancer cells communicate within the microenvironment. Among several factors (cytokines, growth factors, and ECM molecules), a key role has been attributed to extracellular vesicles (EV), released from different cell types. EV (microvesicles and exosomes) may affect stroma remodeling, host cell functions, and tumor angiogenesis by inducing gene expression modulation in target cells, thus promoting cancer progression and metastasis. Microvesicles and exosomes can be recovered from the blood and other body fluids of cancer patients and contain and deliver genetic and proteomic contents that reflect the cell of origin, thus constituting a source of new predictive biomarkers involved in cancer development and serving as possible targets for therapies. Moreover, due to their specific cell-tropism and bioavailability, EV can be considered natural vehicles suitable for drug delivery. Here we will discuss the recent advances in the field of EV as actors in hematological cancer progression, pointing out the role of these vesicles in the tumor-host interplay and in their use as biomarkers for hematological malignancies.


Sujet(s)
Exosomes/génétique , Vésicules extracellulaires/métabolisme , Tumeurs hématologiques/génétique , Néovascularisation pathologique/génétique , Vésicules extracellulaires/génétique , Régulation de l'expression des gènes tumoraux , Tumeurs hématologiques/traitement médicamenteux , Tumeurs hématologiques/anatomopathologie , Humains , Néovascularisation pathologique/traitement médicamenteux , Néovascularisation pathologique/anatomopathologie , Protéomique , Microenvironnement tumoral/génétique
12.
Leuk Res ; 39(8): 883-96, 2015 Aug.
Article de Anglais | MEDLINE | ID: mdl-26055960

RÉSUMÉ

BACKGROUND: Acute myeloid leukemia (AML) represents a heterogeneous disorder with recurrent chromosomal alterations and molecular abnormalities. Among AML with normal karyotype (NK-AML) FLT3 activating mutation, internal tandem duplication (FLT3-ITD), is present in about 30% of patients, conferring unfavorable outcome. Our previous data demonstrated specific up-regulation of miR-155 in FLT3-ITD+ AML. miR-155 is known to be directly implicated in normal hematopoiesis and in some pathologies such as myeloid hyperplasia and acute lymphoblastic leukemia. METHODS AND RESULTS: To investigate about the potential influence of miR-155 de-regulation in FLT3-mutated AML we generated a transcription factors regulatory network and combined this with data from multiple sources that predict miR-155 interactions. From these analyses, we derived a sub-network, called "miR-155 module" that describes functional relationship among miR-155 and transcription factors in FLT3-mutated AML. We found that "miR-155 module" is characterized by the presence of six transcription factors as central hubs: four miR-155 regulators (JUN, RUNX1, FOSb, JUNB) and two targets of miR-155 (SPI1, CEBPB) all known to be "master" genes of myelopoiesis. We found, in FLT3-mutated AML, a significant down-regulation of miR-155 target genes CEBPB and SPI1 and up-regulation of miR-155 regulator genes JUN and RUNX1. We also showed that PKC412-related FLT3 inhibition, in MV4-11 cell line, causes down-regulation of miR-155 and increased level of mRNA and protein of miR-155 target SPI1. We showed in experiments of miR-155 mimic in K562 cell line, a high increase of miR-155 and an inverse correlation with the mRNA levels of its targets SPI1 and CEBPB. Moreover silencing of miR-155 in primary AMLs causes mRNA up-regulation of its target SPI1 and CEBPB. CONCLUSION: Our results suggest that activating mutation of FLT3 in AML can lead, through the induction of JUN, to an increased expression of miR-155, which then causes down-regulation of SPI1 and CEBPB and consequently may causes block of myeloid differentiation.


Sujet(s)
Réseaux de régulation génique/physiologie , Leucémie aigüe myéloïde/génétique , microARN/physiologie , Mutation , Tyrosine kinase-3 de type fms/génétique , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Femelle , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes dans la leucémie , Humains , Cellules K562 , Mâle , Adulte d'âge moyen , Jeune adulte
13.
Cell Commun Signal ; 13: 8, 2015 Feb 03.
Article de Anglais | MEDLINE | ID: mdl-25644060

RÉSUMÉ

BACKGROUND: Chronic myeloid leukemia (CML) is a clonal hematopoietic stem cell disorder in which leukemic cells display a reciprocal t(9:22) chromosomal translocation that results in the formation of the chimeric BCR-ABL oncoprotein, with a constitutive tyrosine kinase activity. Consequently, BCR-ABL causes increased proliferation, inhibition of apoptosis, and altered adhesion of leukemic blasts to the bone marrow (BM) microenvironment. It has been well documented that cancer cells can generate their own signals in order to sustain their growth and survival, and recent studies have revealed the role of cancer-derived exosomes in activating signal transduction pathways involved in cancer cell proliferation. Exosomes are small vesicles of 40-100 nm in diameter that are initially formed within the endosomal compartment, and are secreted when a multivesicular body (MVB) fuses with the plasma membrane. These vesicles are released by many cell types including cancer cells, and are considered messengers in intercellular communication. We have previously shown that CML cells released exosomes able to affect the tumor microenvironment. RESULTS: CML cells, exposed up to one week, to exosomes showed a dose-dependent increased proliferation compared with controls. Moreover, exosome treatment promotes the formation of LAMA84 colonies in methylcellulose. In a CML xenograft model, treatment of mice with exosomes caused a greater increase in tumor size compared with controls (PBS-treated mice). Real time PCR and Western Blot analysis showed, in both in vitro and in vivo samples, an increase in mRNA and protein levels of anti-apoptotic molecules, such as BCL-w, BCL-xl, and survivin, and a reduction of the pro-apoptotic molecules BAD, BAX and PUMA. We also found that TGF- ß1 was enriched in CML-exosomes. Our investigations showed that exosome-stimulated proliferation of leukemia cells, as well as the exosome-mediated activation of an anti-apoptotic phenotype, can be inhibited by blocking TGF-ß1 signaling. CONCLUSIONS: CML-derived exosomes promote, through an autocrine mechanism, the proliferation and survival of tumor cells, both in vitro and in vivo, by activating anti-apoptotic pathways. We propose that this mechanism is activated by a ligand-receptor interaction between TGF-ß1, found in CML-derived exosomes, and the TGF- ß1 receptor in CML cells.


Sujet(s)
Communication autocrine , Prolifération cellulaire , Exosomes/métabolisme , Leucémie myéloïde chronique BCR-ABL positive/métabolisme , Transduction du signal , Animaux , Protéines régulatrices de l'apoptose/génétique , Protéines régulatrices de l'apoptose/métabolisme , Lignée cellulaire tumorale , Exosomes/génétique , Exosomes/anatomopathologie , Humains , Protéines IAP/génétique , Protéines IAP/métabolisme , Leucémie myéloïde chronique BCR-ABL positive/génétique , Leucémie myéloïde chronique BCR-ABL positive/anatomopathologie , Souris , Souris nude , Souris SCID , Protéines tumorales/génétique , Protéines tumorales/métabolisme , Survivine , Facteur de croissance transformant bêta-1/génétique , Facteur de croissance transformant bêta-1/métabolisme
14.
Cancer Lett ; 348(1-2): 71-6, 2014 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-24657661

RÉSUMÉ

Chronic myelogenous leukemia (CML) is a myeloproliferative disorder characterized by the Bcr-Abl oncoprotein with constitutive tyrosine kinase activity. Exosomes are nanovesicles released by cancer cells that are involved in cell-to-cell communication thus potentially affecting cancer progression. It is well known that bone marrow stromal microenvironment contributes to disease progression through the establishment of a bi-directional crosstalk with cancer cells. Our hypothesis is that exosomes could have a functional role in this crosstalk. Interleukin-8 (IL 8) is a proinflammatory chemokine that activates multiple signalling pathways downstream of two receptors (CXCR1 and CXCR2). We demonstrated that exosomes released from CML cells stimulate bone marrow stromal cells to produce IL 8 that, in turn, is able to modulate both in vitro and in vivo the leukemia cell malignant phenotype.


Sujet(s)
Exosomes/métabolisme , Interleukine-8/métabolisme , Leucémie myéloïde chronique BCR-ABL positive/métabolisme , Cellules souches mésenchymateuses/métabolisme , Communication paracrine , Animaux , Adhérence cellulaire , Lignée cellulaire tumorale , Mouvement cellulaire , Survie cellulaire , Hétérogreffes , Humains , Interleukine-8/génétique , Leucémie myéloïde chronique BCR-ABL positive/anatomopathologie , Mâle , Souris , Souris de lignée NOD , Souris SCID , Phénotype , Transduction du signal , Niche de cellules souches , Microenvironnement tumoral , Régulation positive
15.
Int J Mol Sci ; 14(3): 5338-66, 2013 Mar 06.
Article de Anglais | MEDLINE | ID: mdl-23466882

RÉSUMÉ

Cell to cell communication is essential for the coordination and proper organization of different cell types in multicellular systems. Cells exchange information through a multitude of mechanisms such as secreted growth factors and chemokines, small molecules (peptides, ions, bioactive lipids and nucleotides), cell-cell contact and the secretion of extracellular matrix components. Over the last few years, however, a considerable amount of experimental evidence has demonstrated the occurrence of a sophisticated method of cell communication based on the release of specialized membranous nano-sized vesicles termed exosomes. Exosome biogenesis involves the endosomal compartment, the multivesicular bodies (MVB), which contain internal vesicles packed with an extraordinary set of molecules including enzymes, cytokines, nucleic acids and different bioactive compounds. In response to stimuli, MVB fuse with the plasma membrane and vesicles are released in the extracellular space where they can interact with neighboring cells and directly induce a signaling pathway or affect the cellular phenotype through the transfer of new receptors or even genetic material. This review will focus on exosomes as intercellular signaling organelles involved in a number of physiological as well as pathological processes and their potential use in clinical diagnostics and therapeutics.

16.
PLoS One ; 7(8): e42310, 2012.
Article de Anglais | MEDLINE | ID: mdl-22879938

RÉSUMÉ

The Bcr/Abl kinase has been targeted for the treatment of chronic myelogenous leukaemia (CML) by imatinib mesylate. While imatinib has been extremely effective for chronic phase CML, blast crisis CML are often resistant. New therapeutic options are therefore needed for this fatal disease. Although more common in solid tumors, increased microvessel density was also reported in chronic myelogenous leukaemia and was associated with a significant increase of angiogenic factors, suggesting that vascularity in hematologic malignancies is a controlled process and may play a role in the leukaemogenic process thus representing an alternative therapeutic target. Carboxyamidotriazole-orotate (CTO) is the orotate salt form of carboxyamidotriazole (CAI), an orally bioavailable signal transduction inhibitor that in vitro has been shown to possess antileukaemic activities. CTO, which has a reduced toxicity, increased oral bioavailability and stronger efficacy when compared to the parental compound, was tested in this study for its ability to affect imatinib-resistant CML tumor growth in a xenograft model. The active cross talk between endothelial cells and leukemic cells in the bone marrow involving exosomes plays an important role in modulating the process of neovascularization in CML. We have thus investigated the effects of CTO on exosome-stimulated angiogenesis. Our results indicate that CTO may be effective in targeting both cancer cell growth and the tumor microenvironment, thus suggesting a potential therapeutic utility for CTO in leukaemia patients.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Exosomes/métabolisme , Leucémie myéloïde chronique BCR-ABL positive/traitement médicamenteux , Leucémie myéloïde chronique BCR-ABL positive/anatomopathologie , Néovascularisation pathologique/traitement médicamenteux , Acide orotique/pharmacologie , Pipérazines/usage thérapeutique , Pyrimidines/usage thérapeutique , Triazoles/usage thérapeutique , Animaux , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Benzamides , Adhérence cellulaire/effets des médicaments et des substances chimiques , Molécules d'adhérence cellulaire/métabolisme , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Exosomes/effets des médicaments et des substances chimiques , Extracellular Signal-Regulated MAP Kinases/métabolisme , Protéines de fusion bcr-abl/métabolisme , Régulation de l'expression des gènes dans la leucémie/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/cytologie , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Humains , Mésilate d'imatinib , Interleukine-8/métabolisme , Leucémie myéloïde chronique BCR-ABL positive/enzymologie , Mâle , Souris , Néovascularisation pathologique/anatomopathologie , Acide orotique/analogues et dérivés , Acide orotique/usage thérapeutique , Phosphorylation/effets des médicaments et des substances chimiques , Phosphotyrosine/métabolisme , Pipérazines/pharmacologie , Protéines proto-oncogènes c-akt/métabolisme , Pyrimidines/pharmacologie , ARN messager/génétique , ARN messager/métabolisme , Triazoles/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe
17.
Cancer Lett ; 300(2): 205-14, 2011 Jan 28.
Article de Anglais | MEDLINE | ID: mdl-21041018

RÉSUMÉ

Mutation of the Bcr-Abl oncoprotein is one of most frequent mechanisms by which chronic myelogenous leukemia (CML) cells become resistant to imatinib. Here, we show that treatment of cell lines harbouring wild type or mutant BCR-ABL with carboxyamidotriazole (CAI), a calcium influx and signal transduction inhibitor, inhibits cell growth, the expression of Bcr-Abl and its downstream signalling, and induces apoptosis. Moreover, we show that CAI acts by increasing intracellular ROS. Clinically significant, CAI has also inhibitory effects on T315I Bcr-Abl mutant, a mutation that causes CML cells to become insensitive to imatinib and second generation abl kinase inhibitors.


Sujet(s)
Antinéoplasiques/pharmacologie , Protéines de fusion bcr-abl/génétique , Leucémie myéloïde chronique BCR-ABL positive/génétique , Oxydoréduction/effets des médicaments et des substances chimiques , Triazoles/pharmacologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Benzamides , Technique de Western , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Humains , Mésilate d'imatinib , Souris , Mutation , Pipérazines/pharmacologie , Pyrimidines/pharmacologie , Espèces réactives de l'oxygène/métabolisme
18.
J Cell Physiol ; 215(1): 111-21, 2008 Apr.
Article de Anglais | MEDLINE | ID: mdl-17924401

RÉSUMÉ

Although imatinib mesylate (IM) has revolutionized the treatment of chronic myeloid leukemia (CML), some patients develop resistance with progression of leukemia. Alternative or additional targeting of signaling pathways deregulated in bcr-abl-driven CML cells may provide a feasible option for improving clinical response and overcoming resistance. In this study, we show that carboxyamidotriazole (CAI), an orally bioavailable calcium influx and signal transduction inhibitor, is equally effective in inhibiting the proliferation and bcr-abl dependent- and independent-signaling pathways in imatinib-resistant CML cells. CAI inhibits phosphorylation of cellular proteins including STAT5 and CrkL at concentrations that induce apoptosis in IM-resistant CML cells. The combination of imatinib and CAI also down-regulated bcr-abl protein levels. Since CAI is already available for clinical use, these results suggest that it may be an effective addition to the armamentarium of drugs for the treatment of CML.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Leucémie myéloïde chronique BCR-ABL positive/anatomopathologie , Pipérazines/pharmacologie , Pyrimidines/pharmacologie , Triazoles/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Benzamides , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Régulation négative/effets des médicaments et des substances chimiques , Activation enzymatique/effets des médicaments et des substances chimiques , Antienzymes/pharmacologie , Protéines de fusion bcr-abl/génétique , Protéines de fusion bcr-abl/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Mésilate d'imatinib , Hybridation fluorescente in situ , Leucémie myéloïde chronique BCR-ABL positive/enzymologie , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Sondes moléculaires/métabolisme , Phosphotyrosine/métabolisme , Protéines G ras/métabolisme
19.
J Proteome Res ; 6(11): 4330-42, 2007 Nov.
Article de Anglais | MEDLINE | ID: mdl-17935311

RÉSUMÉ

The aim of the present study was the molecular profiling of different Ph+ chronic myelogenous leukemia (CML) cell lines (LAMA84, K562, and KCL22) by a proteomic approach. By employing two-dimensional gel electrophoresis combined with mass spectrometry analysis, we have identified 191 protein spots corresponding to 142 different proteins. Among these, 63% were cancer-related proteins and 74% were described for the first time in leukemia cells. Multivariate analysis highlighted significant differences in the global proteomic profile of the three CML cell lines. In particular, the detailed analysis of 35 differentially expressed proteins revealed that LAMA84 cells preferentially expressed proteins associated with an invasive behavior, while K562 and KCL22 cells preferentially expressed proteins involved in drug resistance. These data demonstrate that these CML cell lines, although representing the same pathological phenotype, show characteristics in their protein expression profile that suggest different phenotypic leukemia subclasses. These data contribute a new potential characterization of the CML phenotype and may help to understand interpatient variability in the progression of disease and in the efficacy of a treatment.


Sujet(s)
Analyse de profil d'expression de gènes , Régulation de l'expression des gènes dans la leucémie , Leucémie myéloïde chronique BCR-ABL positive/génétique , Leucémie myéloïde chronique BCR-ABL positive/métabolisme , Protéomique/méthodes , Orange acridine/pharmacologie , Lignée cellulaire tumorale , Mouvement cellulaire , Évolution de la maladie , Résistance aux médicaments antinéoplasiques , Électrophorèse bidimensionnelle sur gel , Éthidium/pharmacologie , Humains , Cellules K562 , Analyse multifactorielle , Invasion tumorale , Phénotype
20.
Int J Cancer ; 121(3): 528-35, 2007 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-17372905

RÉSUMÉ

The extravasation of cancer cells is a key step of the metastatic cascade. Polymorphisms in genes encoding adhesion molecules can facilitate metastasis by increasing the strength of interaction between tumor and endothelial cells as well as impacting other properties of cancer cells. We investigated the Ser128Arg (a561c at the nucleotide level) polymorphism in the E-selectin gene in patients with metastatic colon cancer and its functional significance. Genotyping for a561c polymorphism was performed on 172 cancer patients and on an age-matched control population. The colon cancer group was divided into groups with (M(+)) and without observable metastasis (M(-)). For in vitro functional assays, Huvec transfected cells expressing wild-type (WT) or the S128R variant of E-selectin were established to study in vitro binding ability and signal transduction processes of T84 colon cancer cell line. Our results demonstrated that the Arginine(128) allele was more prevalent in the M(+) group than in the M(-) group or normal controls (p < 0.005; odds ratio, 1.56; 95% confidence interval (CI) 1.16-1.92; p < 0.001, odds ratio = 1.65; CI = 1.24-1.99, respectively). In vitro, S128R E-selectin transfected Huvec cells, supported increased adhesion as well as increased cellular signaling of T84 cancer cells compared to WT E-selectin and mock-transfected Huvec cells. These findings suggest that the E-selectin S128R polymorphism can functionally affect tumor-endothelial interactions as well as motility and signaling properties of neoplastic cells that may modulate the metastatic phenotype.


Sujet(s)
Tumeurs du côlon/génétique , Sélectine E/génétique , Métastase tumorale/génétique , Polymorphisme génétique , Arginine , Adhérence cellulaire , Mouvement cellulaire , Tumeurs du côlon/anatomopathologie , Femelle , Humains , Mâle , Adulte d'âge moyen , Phénotype , Sérine , Transduction du signal/génétique , Transfection , Cellules cancéreuses en culture
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...