Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
J Cell Mol Med ; 23(6): 4408-4421, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30982221

RESUMO

The intercellular communication mediated by extracellular vesicles (EVs) has gained international interest during the last decade. Interfering with the mechanisms regulating this cellular process might find application particularly in oncology where cancer cell-derived EVs play a role in tumour microenvironment transformation. Although several mechanisms were ascribed to explain the internalization of EVs, little is our knowledge about the fate of their cargos, which are crucial to mediate their function. We recently demonstrated a new intracellular pathway in which a fraction of endocytosed EV-associated proteins is transported into the nucleoplasm of the host cell via a subpopulation of late endosomes penetrating into the nucleoplasmic reticulum. Silencing tetraspanin CD9 both in EVs and recipient cells strongly decreased the endocytosis of EVs and abolished the nuclear transfer of their cargos. Here, we investigated whether monovalent Fab fragments derived from 5H9 anti-CD9 monoclonal antibody (referred hereafter as CD9 Fab) interfered with these cellular processes. To monitor the intracellular transport of proteins, we used fluorescent EVs containing CD9-green fluorescent protein fusion protein and various melanoma cell lines and bone marrow-derived mesenchymal stromal cells as recipient cells. Interestingly, CD9 Fab considerably reduced EV uptake and the nuclear transfer of their proteins in all examined cells. In contrast, the divalent CD9 antibody stimulated both events. By impeding intercellular communication in the tumour microenvironment, CD9 Fab-mediated inhibition of EV uptake, combined with direct targeting of cancerous cells could lead to the development of novel anti-melanoma therapeutic strategies.


Assuntos
Transporte Ativo do Núcleo Celular , Vesículas Extracelulares/efeitos dos fármacos , Fragmentos Fab das Imunoglobulinas/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Melanoma/tratamento farmacológico , Proteínas de Neoplasias/metabolismo , Tetraspanina 29/imunologia , Comunicação Celular , Células Cultivadas , Endocitose/efeitos dos fármacos , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/patologia , Humanos , Fragmentos Fab das Imunoglobulinas/imunologia , Melanoma/imunologia , Melanoma/metabolismo , Melanoma/patologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia
2.
J Biol Chem ; 293(36): 13834-13848, 2018 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-30018135

RESUMO

The endocytic pathway plays an instrumental role in recycling internalized molecules back to the plasma membrane or in directing them to lysosomes for degradation. We recently reported a new role of endosomes-the delivery of components from extracellular vesicles (EVs) to the nucleoplasm of recipient cells. Using indirect immunofluorescence, FRET, immunoisolation techniques, and RNAi, we report here a tripartite protein complex (referred to as the VOR complex) that is essential for the nuclear transfer of EV-derived components by orchestrating the specific localization of late endosomes into nucleoplasmic reticulum. We found that the VOR complex contains the endoplasmic reticulum-localized vesicle-associated membrane protein (VAMP)-associated protein A (VAP-A), the cytoplasmic oxysterol-binding protein-related protein 3 (ORP3), and late endosome-associated small GTPase Rab7. The silencing of VAP-A or ORP3 abrogated the association of Rab7-positive late endosomes with nuclear envelope invaginations and, hence, the transport of endocytosed EV-derived components to the nucleoplasm of recipient cells. We conclude that the VOR complex can be targeted to inhibit EV-mediated intercellular communication, which can have therapeutic potential for managing cancer in which the release of EVs is dysregulated.


Assuntos
Proteínas de Transporte/fisiologia , Retículo Endoplasmático/metabolismo , Endossomos/metabolismo , Complexos Multiproteicos/química , Membrana Nuclear/metabolismo , Proteínas de Transporte Vesicular/fisiologia , Comunicação Celular , Células Cultivadas , Endocitose , Proteínas de Ligação a Ácido Graxo , Humanos , Complexos Multiproteicos/fisiologia , Proteínas R-SNARE , Receptores de Esteroides , Proteínas rab de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7
3.
Int J Mol Sci ; 20(5)2019 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-30841521

RESUMO

The diagnostic approach to thyroid cancer is one of the most challenging issues in oncology of the endocrine system because of its high incidence (3.8% of all new cancer cases in the US) and the difficulty to distinguish benign from malignant non-functional thyroid nodules and establish the cervical lymph node involvement during staging. Routine diagnosis of thyroid nodules usually relies on a fine-needle aspirate biopsy, which is invasive and often inaccurate. Therefore, there is an urgent need to identify novel, accurate, and non-invasive diagnostic procedures. Liquid biopsy, as a non-invasive approach for the detection of diagnostic biomarkers for early tumor diagnosis, prognosis, and disease monitoring, may be of particular benefit in this context. Extracellular vesicles (EVs) are a consistent source of tumor-derived RNA due to their prevalence in circulating bodily fluids, the well-established isolation protocols, and the fact that RNA in phospholipid bilayer-enclosed vesicles is protected from blood-borne RNases. Recent results in other types of cancer, including our recent study on plasma EVs from glioblastoma patients suggest that information derived from analysis of EVs from peripheral blood plasma can be integrated in the routine diagnostic tumor approach. In this review, we will examine the diagnostic and prognostic potential of liquid biopsy to detect tumor-derived nucleic acids in circulating EVs from patients with thyroid carcinoma.


Assuntos
Biomarcadores Tumorais/metabolismo , Vesículas Extracelulares/patologia , Neoplasias da Glândula Tireoide/patologia , Vesículas Extracelulares/metabolismo , Humanos , Neoplasias da Glândula Tireoide/metabolismo
4.
J Cell Biochem ; 118(12): 4414-4424, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28452069

RESUMO

Tumor-derived extracellular vesicles (EVs) are emerging as an important mode of intercellular communication, capable of transferring biologically active molecules that facilitate the malignant growth and metastatic process. CD133 (Prominin-1), a stem cell marker implicated in tumor initiation, differentiation and resistance to anti-cancer therapy, is reportedly associated with EVs in various types of cancer. However, little is known about the factors that regulate the release of these CD133+ EVs. Here, we report that the HDAC6 inhibitor tubacin promoted the extracellular release of CD133+ EVs from human FEMX-I metastatic melanoma and Caco-2 colorectal carcinoma cells, with a concomitant downregulation of intracellular CD133. This effect was specific for tubacin, as inhibition of HDAC6 deacetylase activity by another selective HDAC6 inhibitor, ACY-1215 or the pan-HDAC inhibitor trichostatin A (TSA), and knockdown of HDAC6 did not enhance the release of CD133+ EVs. The tubacin-induced EV release was associated with changes in cellular lipid composition, loss of clonogenic capacity and decrease in the ability to form multicellular aggregates. These findings indicate a novel potential anti-tumor mechanism for tubacin in CD133-expressing malignancies. J. Cell. Biochem. 118: 4414-4424, 2017. © 2017 Wiley Periodicals, Inc.


Assuntos
Antígeno AC133/metabolismo , Anilidas/farmacologia , Micropartículas Derivadas de Células/metabolismo , Desacetilase 6 de Histona/antagonistas & inibidores , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Linhagem Celular Tumoral , Desacetilase 6 de Histona/metabolismo , Humanos
5.
Exp Cell Res ; 319(6): 810-9, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23318676

RESUMO

Prominin-1 (CD133) is the first identified gene of a novel class of pentaspan membrane glycoproteins. It is expressed by various epithelial and non-epithelial cells, and notably by stem and cancer stem cells. In non-cancerous cells such as neuro-epithelial and hematopoietic stem cells, prominin-1 is selectively concentrated in plasma membrane protrusions, and released into the extracellular milieu in association with small vesicles. Previously, we demonstrated that prominin-1 contributes to melanoma cells pro-metastatic properties and suggested that it may constitute a molecular target to prevent prominin-1-expressing melanomas from colonizing and growing in lymph nodes and distant organs. Here, we report that three distinct pools of prominin-1 co-exist in cultures of human FEMX-I metastatic melanoma. Morphologically, in addition to the plasma membrane localization, prominin-1 is found within the intracellular compartments, (e.g., Golgi apparatus) and in association with extracellular membrane vesicles. The latter prominin-1-positive structures appeared in three sizes (small, ≤40 nm; intermediates ~40-80 nm, and large, >80 nm). Functionally, the down-regulation of prominin-1 in FEMX-I cells resulted in a significant reduction of number of lipid droplets as observed by coherent anti-Stokes Raman scattering image analysis and Oil red O staining, and surprisingly in a decrease in the nuclear localization of beta-catenin, a surrogate marker of Wnt activation. Moreover, the T-cell factor/lymphoid enhancer factor (TCF/LEF) promoter activity was 2 to 4 times higher in parental than in prominin-1-knockdown cells. Collectively, our results point to Wnt signaling and/or release of prominin-1-containing membrane vesicles as mediators of the pro-metastatic activity of prominin-1 in FEMX-I melanoma.


Assuntos
Antígenos CD/metabolismo , Glicoproteínas/metabolismo , Melanoma/metabolismo , Peptídeos/metabolismo , Mapeamento de Interação de Proteínas , Via de Sinalização Wnt , beta Catenina/metabolismo , Antígeno AC133 , Antígenos CD/genética , Compostos Azo/metabolismo , Biomarcadores Tumorais/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Membrana Celular/patologia , Movimento Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Glicoproteínas/genética , Complexo de Golgi/metabolismo , Humanos , Imuno-Histoquímica , Lipídeos/análise , Melanoma/patologia , Invasividade Neoplásica/patologia , Peptídeos/genética , Regiões Promotoras Genéticas , Análise Espectral Raman , Fatores de Transcrição TCF/genética , Fatores de Transcrição TCF/metabolismo , Transcrição Gênica , Transfecção , beta Catenina/genética
6.
Mol Cancer ; 12: 62, 2013 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-23767874

RESUMO

Exosomes can be viewed as complex "messages" packaged to survive trips to other cells in the local microenvironment and, through body fluids, to distant sites. A large body of evidence indicates a pro-metastatic role for certain types of cancer exosomes. We previously reported that prominin-1 had a pro-metastatic role in melanoma cells and that microvesicles released from metastatic melanoma cells expressed high levels of prominin-1. With the goal to explore the mechanisms that govern proteo-lipidic-microRNA sorting in cancer exosomes and their potential contribution(s) to the metastatic phenotype, we here employed prominin-1-based immunomagnetic separation in combination with filtration and ultracentrifugation to purify prominin-1-expressing exosomes (prom1-exo) from melanoma and colon carcinoma cells. Prom1-exo contained 154 proteins, including all of the 14 proteins most frequently expressed in exosomes, and multiple pro-metastatic proteins, including CD44, MAPK4K, GTP-binding proteins, ADAM10 and Annexin A2. Their lipid composition resembled that of raft microdomains, with a great enrichment in lyso-phosphatidylcholine, lyso-phosphatidyl-ethanolamine and sphingomyelin. The abundance of tetraspanins and of tetraspanin-associated proteins, together with the high levels of sphingomyelin, suggests that proteolipidic assemblies, probably tetraspanin webs, might be the essential structural determinant in the release process of prominin-1 of stem and cancer stem cells. Micro-RNA profiling revealed 49 species of micro-RNA present at higher concentrations in prom1-exo than in parental cells, including 20 with cancer-related function. Extensive accumulation of prom1-exo was observed 3 h after their addition to cultures of melanoma and bone marrow-derived stromal cells (MSC). Short-term co-culture of melanoma cells and MSC resulted in heterologous prominin-1 transfer. Exposure of MSC to prom1-exo increased their invasiveness. Our study supports the concept that specific populations of cancer exosomes contain multiple determinants of the metastatic potential of the cells from which they are derived.


Assuntos
Antígenos CD/metabolismo , Exossomos/química , Exossomos/metabolismo , Glicoproteínas/metabolismo , Peptídeos/metabolismo , Antígeno AC133 , Células da Medula Óssea/química , Células da Medula Óssea/metabolismo , Linhagem Celular Tumoral , Técnicas de Cocultura , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Humanos , Integrina beta1/metabolismo , Lipídeos/química , Melanoma/química , Melanoma/metabolismo , Microdomínios da Membrana , MicroRNAs/química , MicroRNAs/metabolismo , Ligação Proteica , Proteoma , Células Estromais/química , Células Estromais/metabolismo
7.
Am J Pathol ; 180(6): 2504-15, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22542847

RESUMO

Breast cancer progression involves cancer cell heterogeneity, with generation of invasive/metastatic breast cancer cells within populations of nonmetastatic cells of the primary tumor. Sequential genetic mutations, epithelial-to-mesenchymal transition, interaction with local stroma, and formation of hybrids between cancer cells and normal bone marrow-derived cells have been advocated as tumor progression mechanisms. We report herein the spontaneous in vitro formation of heterotypic hybrids between human bone marrow-derived multipotent stromal cells (MSCs) and two different breast carcinoma cell lines, MDA-MB-231 (MDA) and MA11. Hybrids showed predominantly mesenchymal morphological characteristics, mixed gene expression profiles, and increased DNA ploidy. Both MA11 and MDA hybrids were tumorigenic in immunodeficient mice, and some MDA hybrids had an increased metastatic capacity. Both in culture and as xenografts, hybrids underwent DNA ploidy reduction and morphological reversal to breast carcinoma-like morphological characteristics, while maintaining a mixed breast cancer-mesenchymal expression profile. Analysis of coding single-nucleotide polymorphisms by RNA sequencing revealed genetic contributions from both parental partners to hybrid tumors and metastasis. Because MSCs migrate and localize to breast carcinoma, our findings indicate that formation of MSC-breast cancer cell hybrids is a potential mechanism of the generation of invasive/metastatic breast cancer cells. Our findings reconcile the fusion theory of cancer progression with the common observation that breast cancer metastases are generally aneuploid, but not tetraploid, and are histopathologically similar to the primary neoplasm.


Assuntos
Neoplasias da Mama/patologia , Heterogeneidade Genética , Células-Tronco Multipotentes/patologia , Células-Tronco Neoplásicas/patologia , Células Estromais/patologia , Animais , Neoplasias da Mama/genética , Fusão Celular , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Técnicas de Cocultura , Feminino , Perfilação da Expressão Gênica/métodos , Humanos , Células Híbridas/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Microscopia de Fluorescência , Transplante de Neoplasias , Ploidias , Polimorfismo de Nucleotídeo Único , Transplante Heterólogo , Células Tumorais Cultivadas
8.
Adv Exp Med Biol ; 777: 197-211, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23161084

RESUMO

Innovative approaches to specifically target the melanoma subpopulation responsible for local invasion and metastatic dissemination are needed. Prominin-1 (CD133) expression has been observed in many melanoma cell lines, as well as in primary and metastatic melanomas from patients. Although its function(s) in melanoma is presently unknown, prominin-1 may represent a molecular target, due to its association with melanoma stem cells and with the metastatic phenotype.


Assuntos
Melanoma , Humanos
9.
Nat Commun ; 14(1): 4588, 2023 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-37563144

RESUMO

The mechanism of human immunodeficiency virus 1 (HIV-1) nuclear entry, required for productive infection, is not fully understood. Here, we report that in HeLa cells and activated CD4+ T cells infected with HIV-1 pseudotyped with VSV-G and native Env protein, respectively, Rab7+ late endosomes containing endocytosed HIV-1 promote the formation of nuclear envelope invaginations (NEIs) by a molecular mechanism involving the VOR complex, composed of the outer nuclear membrane protein VAP-A, hyperphosphorylated ORP3 and Rab7. Silencing VAP-A or ORP3 and drug-mediated impairment of Rab7 binding to ORP3-VAP-A inhibited the nuclear transfer of the HIV-1 components and productive infection. In HIV-1-resistant quiescent CD4+ T cells, ORP3 was not hyperphosphorylated and neither VOR complex nor NEIs were formed. This new cellular pathway and its molecular players are potential therapeutic targets, perhaps shared by other viruses that require nuclear entry to complete their life cycle.


Assuntos
Infecções por HIV , HIV-1 , Humanos , HIV-1/metabolismo , Células HeLa , Linfócitos T CD4-Positivos/metabolismo , Produtos do Gene env/metabolismo , Proteínas de Membrana/metabolismo
10.
Int J Cancer ; 131(2): 334-43, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21858806

RESUMO

We have previously reported in vitro heterotypic fusion of glioma cells with neural progenitor cells, producing tetraploid cells expressing genetic complements of each partner. Herein, we investigated the fusogenicity of glioma cells. In 1:1 cocultures of single-labeled cells, U87MG cells presented high frequency of homotypic fusogenic events, producing cells that coexpressed both fluorescent proteins. Six percent of the total cells had 8n DNA content, consistent with the finding that the double-labeled cells were actively proliferating. In coculture with fibroblasts, glioma cell fusogenicity resulted in viable reprogrammed cells, thus emerging as a plausible source of tumor cell heterogeneity. As for heterotypic fusion to happen, glioma cells have to establish direct contact with other cells, the effect of stroma on glioma cells was analyzed. Proliferation assays and array analysis of cancer-related pathways established a promalignant effect of stroma. This effect was mediated by fibronectin and was nearly completely abolished by inhibitors of the epidermal-growth-factor receptor. That stroma elicited transduction signaling through the mitogen-activated-protein kinase/extracellular-signal-regulated kinase pathway, which is linked to increased tumor cell migration through extracellular matrix, suggested that glioma cells may actively approach nontumor cells in stromal niches. According to these results, the fusogenicity of glioma cells emerged as an inherent factor for phenotypic changes leading to glioma progression.


Assuntos
Fusão Celular , Transformação Celular Neoplásica , Glioma/patologia , Glioma/fisiopatologia , Células Híbridas/metabolismo , Microambiente Tumoral , Animais , Linhagem Celular Tumoral , Proliferação de Células , Técnicas de Cocultura , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Matriz Extracelular/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibroblastos/fisiologia , Fibronectinas/metabolismo , Regulação Neoplásica da Expressão Gênica , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Transdução de Sinais
11.
Cells ; 11(16)2022 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-36010551

RESUMO

Intercellular communication between cancer cells themselves or with healthy cells in the tumor microenvironment and/or pre-metastatic sites plays an important role in cancer progression and metastasis. In addition to ligand-receptor signaling complexes, extracellular vesicles (EVs) are emerging as novel mediators of intercellular communication both in tissue homeostasis and in diseases such as cancer. EV-mediated transfer of molecular activities impacting morphological features and cell motility from highly metastatic SW620 cells to non-metastatic SW480 cells is a good in vitro example to illustrate the increased malignancy of colorectal cancer leading to its transformation and aggressive behavior. In an attempt to intercept the intercellular communication promoted by EVs, we recently developed a monovalent Fab fragment antibody directed against human CD9 tetraspanin and showed its effectiveness in blocking the internalization of melanoma cell-derived EVs and the nuclear transfer of their cargo proteins into recipient cells. Here, we employed the SW480/SW620 model to investigate the anti-cancer potential of the anti-CD9 Fab antibody. We first demonstrated that most EVs derived from SW620 cells contain CD9, making them potential targets. We then found that the anti-CD9 Fab antibody, but not the corresponding divalent antibody, prevented internalization of EVs from SW620 cells into SW480 cells, thereby inhibiting their phenotypic transformation, i.e., the change from a mesenchymal-like morphology to a rounded amoeboid-like shape with membrane blebbing, and thus preventing increased cell migration. Intercepting EV-mediated intercellular communication in the tumor niche with an anti-CD9 Fab antibody, combined with direct targeting of cancer cells, could lead to the development of new anti-cancer therapeutic strategies.


Assuntos
Neoplasias do Colo , Vesículas Extracelulares , Comunicação Celular , Neoplasias do Colo/patologia , Vesículas Extracelulares/metabolismo , Humanos , Fragmentos Fab das Imunoglobulinas/metabolismo , Tetraspanina 29/metabolismo , Microambiente Tumoral
13.
Exp Cell Res ; 316(9): 1576-86, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20074564

RESUMO

The phenotypic diversity of breast carcinoma may be explained by the existence of a sub-population of breast cancer cells, endowed with stem cell-like properties and gene expression profiles, able to differentiate along different pathways. A stem cell-like population of CD44(+)CD24(-/low) breast cancer cells was originally identified using cells from metastatic pleural effusions of breast carcinoma patients. We have previously reported that upon in vitro culture as mammospheres under stem cell-like conditions, human MA-11 breast carcinoma cells acquired increased tumorigenicity and lost CD24 expression compared with the parental cell line. We now report that upon passage of MA-11 mammospheres into serum-supplemented cultures, CD24 expression was restored; the rapid increase in CD24 expression was consistent with up-regulation of the antigen, and not with in vitro selection of CD24(+) cells. In tumors derived from subcutaneous injection of MA-11 mammospheres in athymic nude mice, 76.1+/-9.7% of cells expressed CD24, vs. 0.5+/-1% in MA-11 cells dissociated from mammospheres before injection. The tumorigenicity of sorted CD44(+)CD24(-) and CD44(+)CD24(high) MA-11 cells was equal. Single cell-sorted CD24(-) and CD24(high) MA-11 gave rise in vitro to cell populations with heterogeneous CD24 expression. Also, subcutaneous tumors derived from sorted CD24(-) sub-populations and single-cell clones had levels of CD24 expression similar to the unsorted cells. To investigate whether the high expression of CD24 contributed to the tumorigenic potential of MA-11 cells, we silenced CD24 by shRNA. CD24 silencing (95%) resulted in no difference in tumorigenicity upon s.c. injection in athymic nude mice compared with mock-transduced MA-11 cells. Since CD24 silencing was maintained in vivo, our data suggest that the level of expression of CD24 is associated with but does not contribute to tumorigenicity. We then compared the molecular profile of the mammospheres with the adherent cell fraction. Gene expression profiling revealed that the increased tumorigenicity of MA-11 mammospheres was associated with changes in 10 signal transduction pathways, including MAP kinase, Notch and Wnt, and increased expression of aldehyde dehydrogenase, a cancer-initiating cell-associated marker. Our data demonstrate that (i) the level of CD24 expression is neither a stable feature of mammosphere-forming cells nor confers tumorigenic potential to MA-11 cells; (ii) cancer-initiating cell-enriched MA-11 mammospheres have activated specific signal transduction pathways, potential targets for anti-breast cancer therapy.


Assuntos
Neoplasias da Mama/patologia , Antígeno CD24/metabolismo , Transdução de Sinais , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Mama/citologia , Mama/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Antígeno CD24/química , Antígeno CD24/genética , Linhagem Celular Tumoral , Citometria de Fluxo , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , RNA Interferente Pequeno/farmacologia
14.
J Extracell Vesicles ; 10(10): e12132, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34429859

RESUMO

Extracellular vesicles (EVs) are mediators of intercellular communication under both healthy and pathological conditions, including the induction of pro-metastatic traits, but it is not yet known how and where functional cargoes of EVs are delivered to their targets in host cell compartments. We have described that after endocytosis, EVs reach Rab7+ late endosomes and a fraction of these enter the nucleoplasmic reticulum and transport EV biomaterials to the host cell nucleoplasm. Their entry therein and docking to outer nuclear membrane occur through a tripartite complex formed by the proteins VAP-A, ORP3 and Rab7 (VOR complex). Here, we report that the antifungal compound itraconazole (ICZ), but not its main metabolite hydroxy-ICZ or ketoconazole, disrupts the binding of Rab7 to ORP3-VAP-A complexes, leading to inhibition of EV-mediated pro-metastatic morphological changes including cell migration behaviour of colon cancer cells. With novel, smaller chemical drugs, inhibition of the VOR complex was maintained, although the ICZ moieties responsible for antifungal activity and interference with intracellular cholesterol distribution were removed. Knowing that cancer cells hijack their microenvironment and that EVs derived from them determine the pre-metastatic niche, small-sized inhibitors of nuclear transfer of EV cargo into host cells could find cancer therapeutic applications, particularly in combination with direct targeting of cancer cells.


Assuntos
Vesículas Extracelulares/efeitos dos fármacos , Vesículas Extracelulares/metabolismo , Proteínas de Ligação a Ácido Graxo/metabolismo , Itraconazol/farmacologia , Membrana Nuclear/metabolismo , Proteínas de Transporte Vesicular/metabolismo , proteínas de unión al GTP Rab7/metabolismo , Transporte Ativo do Núcleo Celular , Antifúngicos/farmacologia , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Colestenonas/farmacologia , Endocitose , Endossomos/metabolismo , Proteínas de Ligação a Ácido Graxo/química , Humanos , Cetoconazol/farmacologia , Modelos Moleculares , Saponinas/farmacologia , Proteínas de Transporte Vesicular/química , proteínas de unión al GTP Rab7/química
15.
Int J Cancer ; 126(5): 1206-15, 2010 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19653275

RESUMO

The prognosis of patients with glioblastoma multiforme (GBM) is generally poor after surgical tumor resection. With the aim of developing new adjuvant therapeutic strategies, we have investigated primary neural stem/progenitor cells (NSPC) in co-cultures with glioma cells, and in a model of gene therapy on aggressively growing malignant glioma. NSPC exhibited tropism towards medium conditioned by glioma cells, and in adherent low-cell density co-culture, were attracted to, and fused with, tumor cells. Similarly, within 24-48 hr of co-culture in suspension, NSPC-tumor hybrids were observed, representing 2-3% of the total cell population. NSPC were then coinjected into mouse brain with GBM cells, employing NSPC expressing cyclophosphamide (CPA)-activating enzyme cytochrome p450 2B6 (CYP2B6), which catalyzes CPA prodrug transformation into membrane diffusible DNA-alkylating metabolites. Upon CPA administration, NSPC containing CYP2B6 elicited substantial impairment of tumor growth. When implanted intracerebrally at a distant site from the tumor, gene-engineered NSPC specifically targeted GBM grafts, after traveling through brain parenchyma, and hindered tumor growth through local activation of CPA. Directed migration of primary NSPC corresponded closely with intracerebral and tumoral pattern of expression of vascular endothelial growth factor, which is a motility factor for NSPC. Overall, these findings indicate that therapeutic gene delivery mediated by primary NSPC is a potentially valid strategy for treatment of high-grade gliomas.


Assuntos
Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Glioma/terapia , Células Híbridas/transplante , Neurônios/citologia , Células-Tronco/citologia , Animais , Antineoplásicos Alquilantes/farmacologia , Hidrocarboneto de Aril Hidroxilases/genética , Diferenciação Celular , Movimento Celular , Técnicas de Cocultura , Ciclofosfamida/farmacologia , Citocromo P-450 CYP2B6 , Engenharia Genética , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Oxirredutases N-Desmetilantes/genética , Transdução Genética
16.
Cells ; 9(9)2020 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-32825578

RESUMO

Extracellular membrane vesicles (EVs) are emerging as new vehicles in intercellular communication, but how the biological information contained in EVs is shared between cells remains elusive. Several mechanisms have been described to explain their release from donor cells and the initial step of their uptake by recipient cells, which triggers a cellular response. Yet, the intracellular routes and subcellular fate of EV content upon internalization remain poorly characterized. This is particularly true for EV-associated proteins and nucleic acids that shuttle to the nucleus of host cells. In this review, we will describe and discuss the release of EVs from donor cells, their uptake by recipient cells, and the fate of their cargoes, focusing on a novel intracellular route wherein small GTPase Rab7+ late endosomes containing endocytosed EVs enter into nuclear envelope invaginations and deliver their cargo components to the nucleoplasm of recipient cells. A tripartite protein complex composed of (VAMP)-associated protein A (VAP-A), oxysterol-binding protein (OSBP)-related protein-3 (ORP3), and Rab7 is essential for the transfer of EV-derived components to the nuclear compartment by orchestrating the particular localization of late endosomes in the nucleoplasmic reticulum.


Assuntos
Transporte Biológico/fisiologia , Comunicação Celular/fisiologia , Endossomos/metabolismo , Vesículas Extracelulares/metabolismo , Humanos
17.
Stem Cells ; 26(12): 3008-17, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18802032

RESUMO

CD133 (Prominin-1) is considered the most important cancer stem cell (CSC)-associated marker identified so far, with increased expression in the CSC fraction of a large variety of human malignancies, including melanoma. Here we investigated the effects of CD133 downregulation in vitro and in vivo in human metastatic melanoma. The average number of CD133 molecules on the cell surface of FEMX-I melanoma cells was decreased by 8.7-fold and 1.8-fold using two different short hairpin RNAs. Downregulation of CD133, confirmed by immunocytochemistry, Western blotting, microarray analysis, and reverse transcription-polymerase chain reaction, resulted in slower cell growth, reduced cell motility, and decreased capacity to form spheroids under stem cell-like growth conditions. Clonal analysis revealed that the reduction in growth rate was proportional to the extent of CD133 downregulation. Monoclonal antibodies directed against two different epitopes of the CD133 protein induced a specific, dose-dependent cytotoxic effect in FEMX-I cells. The downregulation of CD133 severely reduced the capacity of the cells to metastasize, particularly to the spinal cord. In the CD133 downregulated cells, microarray analysis revealed expression changes for only 143 annotated genes (76 up- and 67 downregulated). Ten of the 76 upregulated genes coded for Wnt inhibitors, suggesting an interaction between CD133 and the canonical Wnt pathway. We conclude that CD133, in addition to its role as a CSC marker, is an important therapeutic target for metastatic melanoma and, potentially, for other CD133-expressing cancer types.


Assuntos
Antígenos CD/biossíntese , Glicoproteínas/biossíntese , Melanoma/metabolismo , Células-Tronco Neoplásicas/metabolismo , Antígeno AC133 , Linhagem Celular Tumoral , Movimento Celular , Separação Celular , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica/métodos , Melanoma/tratamento farmacológico , Metástase Neoplásica , Análise de Sequência com Séries de Oligonucleotídeos , Peptídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Esferoides Celulares/metabolismo , Células Tumorais Cultivadas , Proteínas Wnt/metabolismo
18.
Exp Cell Res ; 314(10): 2110-22, 2008 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-18423605

RESUMO

Malignant tumors comprise a small proportion of cancer-initiating cells (CIC), capable of sustaining tumor formation and growth. CIC are the main potential target for anticancer therapy. However, the identification of molecular therapeutic targets in CIC isolated from primary tumors is an extremely difficult task. Here, we show that after years of passaging under differentiating conditions, glioblastoma, mammary carcinoma, and melanoma cell lines contained a fraction of cells capable of forming spheroids upon in vitro growth under stem cell-like conditions. We found an increased expression of surface markers associated with the stem cell phenotype and of oncogenes in cell lines and clones cultured as spheroids vs. adherent cultures. Also, spheroid-forming cells displayed increased tumorigenicity and an altered pattern of chemosensitivity. Interestingly, also from single retrovirally marked clones, it was possible to isolate cells able to grow as spheroids and associated with increased tumorigenicity. Our findings indicate that short-term selection and propagation of CIC as spheroid cultures from established cancer cell lines, coupled with gene expression profiling, represents a suitable tool to study and therapeutically target CIC: the notion of which genes have been down-regulated during growth under differentiating conditions will help find CIC-associated therapeutic targets.


Assuntos
Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Neoplasias/terapia , Esferoides Celulares , Células-Tronco/fisiologia , Animais , Biomarcadores Tumorais/metabolismo , Técnicas de Cultura de Células , Transformação Celular Neoplásica , Feminino , Perfilação da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Neoplasias/patologia , Análise de Sequência com Séries de Oligonucleotídeos
19.
Clin Cancer Res ; 25(1): 266-276, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30287549

RESUMO

PURPOSE: Glioblastoma (GBM) is the most common primary brain tumor. The identification of blood biomarkers reflecting the tumor status represents a major unmet need for optimal clinical management of patients with GBM. Their high number in body fluids, their stability, and the presence of many tumor-associated proteins and RNAs make extracellular vesicles potentially optimal biomarkers. Here, we investigated the potential role of plasma extracellular vesicles from patients with GBM for diagnosis and follow-up after treatment and as a prognostic tool. EXPERIMENTAL DESIGN: Plasma from healthy controls (n = 33), patients with GBM (n = 43), and patients with different central nervous system malignancies (n = 25) were collected. Extracellular vesicles were isolated by ultracentrifugation and characterized in terms of morphology by transmission electron microscopy, concentration, and size by nanoparticle tracking analysis, and protein composition by mass spectrometry. An orthotopic mouse model of human GBM confirmed human plasma extracellular vesicle quantifications. Associations between plasma extracellular vesicle concentration and clinicopathologic features of patients with GBM were analyzed. All statistical tests were two-sided. RESULTS: GBM releases heterogeneous extracellular vesicles detectable in plasma. Plasma extracellular vesicle concentration was higher in GBM compared with healthy controls (P < 0.001), brain metastases (P < 0.001), and extra-axial brain tumors (P < 0.001). After surgery, a significant drop in plasma extracellular vesicle concentration was measured (P < 0.001). Plasma extracellular vesicle concentration was also increased in GBM-bearing mice (P < 0.001). Proteomic profiling revealed a GBM-distinctive signature. CONCLUSIONS: Higher extracellular vesicle plasma levels may assist in GBM clinical diagnosis: their reduction after GBM resection, their rise at recurrence, and their protein cargo might provide indications about tumor, therapy response, and monitoring.


Assuntos
Glioblastoma/sangue , Recidiva Local de Neoplasia/sangue , Recidiva Local de Neoplasia/genética , Prognóstico , Animais , Biomarcadores Tumorais/sangue , Linhagem Celular Tumoral , Vesículas Extracelulares/genética , Vesículas Extracelulares/patologia , Vesículas Extracelulares/ultraestrutura , Feminino , Glioblastoma/genética , Glioblastoma/patologia , Xenoenxertos , Humanos , Masculino , Camundongos , Microscopia Eletrônica , Recidiva Local de Neoplasia/patologia , Células Neoplásicas Circulantes/metabolismo , Células Neoplásicas Circulantes/patologia , Proteoma/genética
20.
Oncotarget ; 8(9): 14443-14461, 2017 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-28129640

RESUMO

Extracellular membrane vesicles (EVs) function as vehicles of intercellular communication, but how the biomaterials they carry reach the target site in recipient cells is an open question. We report that subdomains of Rab7+ late endosomes and nuclear envelope invaginations come together to create a sub-nuclear compartment, where biomaterials associated with CD9+ EVs are delivered. EV-derived biomaterials were also found in the nuclei of host cells. The inhibition of nuclear import and export pathways abrogated the nuclear localization of EV-derived biomaterials or led to their accumulation therein, respectively, suggesting that their translocation is dependent on nuclear pores. Nuclear envelope invagination-associated late endosomes were observed in ex vivo biopsies in both breast carcinoma and associated stromal cells. The transcriptome of stromal cells exposed to cancer cell-derived CD9+ EVs revealed that the regulation of eleven genes, notably those involved in inflammation, relies on the nuclear translocation of EV-derived biomaterials. Our findings uncover a new cellular pathway used by EVs to reach nuclear compartment.


Assuntos
Materiais Biocompatíveis/metabolismo , Neoplasias da Mama/metabolismo , Endossomos/metabolismo , Vesículas Extracelulares/metabolismo , Mediadores da Inflamação/metabolismo , Células-Tronco Mesenquimais/metabolismo , Membrana Nuclear/metabolismo , Transporte Ativo do Núcleo Celular , Adulto , Neoplasias da Mama/patologia , Comunicação Celular , Células Cultivadas , Exossomos/metabolismo , Feminino , Humanos , Células-Tronco Mesenquimais/citologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa