Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
EMBO J ; 42(7): e111961, 2023 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-36574362

RESUMO

Cytosolic DNA promotes inflammatory responses upon detection by the cyclic GMP-AMP (cGAMP) synthase (cGAS). It has been suggested that cGAS downregulation is an immune escape strategy harnessed by tumor cells. Here, we used glioblastoma cells that show undetectable cGAS levels to address if alternative DNA detection pathways can promote pro-inflammatory signaling. We show that the DNA-PK DNA repair complex (i) drives cGAS-independent IRF3-mediated type I Interferon responses and (ii) that its catalytic activity is required for cGAS-dependent cGAMP production and optimal downstream signaling. We further show that the cooperation between DNA-PK and cGAS favors the expression of chemokines that promote macrophage recruitment in the tumor microenvironment in a glioblastoma model, a process that impairs early tumorigenesis but correlates with poor outcome in glioblastoma patients. Thus, our study supports that cGAS-dependent signaling is acquired during tumorigenesis and that cGAS and DNA-PK activities should be analyzed concertedly to predict the impact of strategies aiming to boost tumor immunogenicity.


Assuntos
Proteína Quinase Ativada por DNA , Glioblastoma , Nucleotidiltransferases , Humanos , Carcinogênese , DNA/metabolismo , Dano ao DNA , Reparo do DNA , Glioblastoma/genética , Imunidade Inata , Inflamação , Nucleotidiltransferases/metabolismo , Microambiente Tumoral , Proteína Quinase Ativada por DNA/metabolismo
2.
EMBO J ; 39(1): e102030, 2020 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-31774199

RESUMO

Glioblastoma is one of the most lethal forms of adult cancer with a median survival of around 15 months. A potential treatment strategy involves targeting glioblastoma stem-like cells (GSC), which constitute a cell autonomous reservoir of aberrant cells able to initiate, maintain, and repopulate the tumor mass. Here, we report that the expression of the paracaspase mucosa-associated lymphoid tissue l (MALT1), a protease previously linked to antigen receptor-mediated NF-κB activation and B-cell lymphoma survival, inversely correlates with patient probability of survival. The knockdown of MALT1 largely impaired the expansion of patient-derived stem-like cells in vitro, and this could be recapitulated with pharmacological inhibitors, in vitro and in vivo. Blocking MALT1 protease activity increases the endo-lysosome abundance, impairs autophagic flux, and culminates in lysosomal-mediated cell death, concomitantly with mTOR inactivation and dispersion from endo-lysosomes. These findings place MALT1 as a new druggable target involved in glioblastoma and unveil ways to modulate the homeostasis of endo-lysosomes.


Assuntos
Biomarcadores Tumorais/metabolismo , Endossomos/patologia , Glioma/patologia , Homeostase , Lisossomos/patologia , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa/metabolismo , Células-Tronco Neoplásicas/patologia , Idoso , Animais , Apoptose , Biomarcadores Tumorais/genética , Proliferação de Células , Endossomos/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Glioma/genética , Glioma/metabolismo , Humanos , Ativação Linfocitária , Lisossomos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa/genética , Células-Tronco Neoplásicas/metabolismo , Proteólise , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
3.
J Autoimmun ; 129: 102826, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35378380

RESUMO

The critical role of neutrophils in pathological inflammation, notably in various autoimmune disorders, is currently the focus of renewed interest. Here, we demonstrate for the first time that activation of neutrophils with various inflammatory stimuli induces the release of extracellular vesicles (EVs) that are internalized by endothelial cells (ECs), thus leading to the transfer of miR-223, miR-142-3p and miR-451 and subsequent endothelial damage. Indeed, while miR-223 has little effect on EC responses, we show that the induced expression of miR-142-3p and miR-451 in ECs results in profound cell damage, especially in inflammatory conditions, characterized by a dramatic increase in cell apoptosis, impaired angiogenic repair responses, and the induction of IL-6, IL-8, CXCL10 and CXCL11 expression. We show that the strong deleterious effect of miR-142-3p may be due in part to its ability to block the activation of ERK1/2 and eNOS-mediated signals in ECs. miR-142-3p also inhibits the expression of RAC1, ROCK2 and CLIC4, three genes that are critical for EC migration and angiogenic responses. Importantly, miR-223, miR-142-3p and miR-451 are markedly increased in kidney biopsies from patients with active ANCA-associated vasculitis, a severe autoimmune disease that is prototypical of a neutrophil-induced microvascular damage. Taken together, our results suggest that miR-142-3p and miR-451 released in EVs by activated neutrophils can target EC to trigger an inflammatory cascade and induce direct vascular damage, and that therapeutic strategies based on the inhibition of these miRNAs in ECs will have implications for neutrophil-mediated inflammatory diseases.


Assuntos
Vesículas Extracelulares , MicroRNAs , Canais de Cloreto/metabolismo , Células Endoteliais/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , Inflamação/metabolismo , MicroRNAs/genética , Neutrófilos/metabolismo
4.
EMBO Rep ; 20(10): e47840, 2019 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-31410978

RESUMO

The activation of mixed lineage kinase-like (MLKL) by receptor-interacting protein kinase-3 (RIPK3) controls the execution of necroptosis, a regulated form of necrosis that occurs in apoptosis-deficient conditions. Active oligomerized MLKL triggers the exposure of phosphatidylserine residues on the cell surface and disrupts the plasma membrane integrity by forming lytic pores. MLKL also governs endosomal trafficking and biogenesis of small extracellular vesicles as well as the production of proinflammatory cytokines during the early steps of necroptosis; however, the molecular basis continues to be elucidated. Here, we find that MLKL oligomers activate Pannexin-1 (PANX1) channels, concomitantly to the loss of phosphatidylserine asymmetry. This plasma membrane "leakiness" requires the small GTPase RAB27A and RAB27B isoforms, which regulate intracellular vesicle trafficking, docking, and fusion with the plasma membrane. Although cells in which PANX1 is silenced or inhibited normally undergo necroptotic death, they display enhanced production of cytokines such as interleukin-8, indicating that PANX1 may tamper with inflammation. These data identify a novel signaling nexus between MLKL, RAB27, and PANX1 and propose ways to interfere with inflammation associated with necroptosis.


Assuntos
Conexinas/metabolismo , Citocinas/metabolismo , Mediadores da Inflamação/metabolismo , Necroptose , Proteínas do Tecido Nervoso/metabolismo , Membrana Celular/metabolismo , Permeabilidade da Membrana Celular , Sobrevivência Celular , Inativação Gênica , Células HT29 , Humanos , Proteínas Quinases/metabolismo , Multimerização Proteica , Vesículas Transportadoras/metabolismo
5.
Int J Mol Sci ; 22(12)2021 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-34204510

RESUMO

Glioblastoma, a subset of aggressive brain tumors, deploy several means to increase blood vessel supply dedicated to the tumor mass. This includes typical program borrowed from embryonic development, such as vasculogenesis and sprouting angiogenesis, as well as unconventional processes, including co-option, vascular mimicry, and transdifferentiation, in which tumor cells are pro-actively engaged. However, these neo-generated vascular networks are morphologically and functionally abnormal, suggesting that the vascularization processes are rather inefficient in the tumor ecosystem. In this review, we reiterate the specificities of each neovascularization modality in glioblastoma, and, how they can be hampered mechanistically in the perspective of anti-cancer therapies.


Assuntos
Glioblastoma/metabolismo , Glioblastoma/patologia , Neovascularização Patológica/metabolismo , Animais , Biomarcadores , Comunicação Celular , Diferenciação Celular , Suscetibilidade a Doenças , Resistencia a Medicamentos Antineoplásicos , Regulação Neoplásica da Expressão Gênica , Glioblastoma/etiologia , Glioblastoma/terapia , Humanos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Tolerância a Radiação , Transdução de Sinais , Microambiente Tumoral
6.
Int J Mol Sci ; 22(7)2021 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-33918324

RESUMO

Epithelial-mesenchymal transition (EMT) is important for the initial steps of metastasis. Although it is well accepted that the nucleoside diphosphate kinase NME1 is a metastasis suppressor, its effect on EMT remains poorly documented, as does that of its closely related isoform, NME2. Here, by using gene silencing, inactivation and overexpression strategies in a variety of cellular models of cancer, we show that NME1 is a powerful inhibitor of EMT. Genetic manipulation of NME2, by contrast, had no effect on the EMT phenotype of cancer cells, indicating a specific function of NME1 in EMT regulation. Loss of NME1 in epithelial cancer cells resulted in a hybrid phenotype intermediate between epithelial and mesenchymal cells, which is known to be associated with cells with a highly metastatic character. Conversely, overexpression of NME1 in mesenchymal cancer cells resulted in a more epithelial phenotype. We found that NME1 expression was negatively associated with EMT markers in many human cancers and was reduced in human breast tumor cell lines with the aggressive 'triple-negative' phenotype when compared to human breast tumor cell lines positive for estrogen receptor. We show that NME1, but not NME2, is an inhibitor of essential concerted intracellular signaling pathways involved in inducing EMT, including the AKT and MAPK (ERK, p38, and JNK) pathways. Additionally, NME1 depletion considerably altered the distribution of E-cadherin, a gatekeeper of the epithelial phenotype, shifting it from the plasma membrane to the cytosol and resulting in less E-cadherin on the cell surface than in control cells. Functional aggregation and dispersion assays demonstrated that inactivation of NME1 decreases E-cadherin-mediated cell-cell adhesion. We conclude that NME1, but not NME2, acts specifically to inhibit EMT and prevent the earliest stages of metastasis.


Assuntos
Transição Epitelial-Mesenquimal , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Animais , Caderinas/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Feminino , Edição de Genes , Humanos , Sistema de Sinalização das MAP Quinases , Camundongos Nus , Neoplasias de Mama Triplo Negativas/metabolismo
7.
Cell Immunol ; 353: 104115, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32388054

RESUMO

The CARMA1-BCL10-MALT1 (CBM) complex couples antigen receptors to the activation of Nuclear Factor κB (NF-κB) transcription factors in T/B lymphocytes. Within this signalosome, the MALT1 paracaspase serves dual roles: it is a crucial adaptor for signal transduction to NF-κB signaling, and a protease that shapes NF-κB activity and lymphocyte activation. Although a subtle choreography of ubiquitination and phosphorylation orchestrate the CBM, how precisely this complex and MALT1 enzyme are regulated continue to be elucidated. Here, we report that the chemical inhibition or the siRNA-based silencing of transforming growth factor beta-activated kinase 1 (TAK1), a known partner of the CBM complex required for NF-κB activation, enhanced the processing of MALT1 substrates. We further show that the assembly of the CBM as well as the ubiquitination of MALT1 was augmented when TAK1 was inhibited. Thus, TAK1 may initiate a negative feedback loop to finely tune the CBM complex activity.


Assuntos
MAP Quinase Quinase Quinases/metabolismo , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Linfócitos B/imunologia , Humanos , Células Jurkat , Ativação Linfocitária , Complexos Multiproteicos/metabolismo , NF-kappa B/metabolismo , Proteínas de Neoplasias/genética , Fosforilação , Transdução de Sinais , Fatores de Transcrição/metabolismo
8.
Cell Immunol ; 353: 104133, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32450431

RESUMO

The natural bioactive glycerophospholipid lysophosphatidic acid (LPA) binds to its cognate G protein-coupled receptors (GPCRs) on the cell surface to promote the activation of several transcription factors, including NF-κB. LPA-mediated activation of NF-κB relies on the formation of a signalosome that contains the scaffold CARMA3, the adaptor BCL10 and the paracaspase MALT1 (CBM complex). The CBM complex has been extensively studied in lymphocytes, where it links antigen receptors to NF-κB activation via the recruitment of the linear ubiquitin assembly complex (LUBAC), a tripartite complex of HOIP, HOIL1 and SHARPIN. Moreover, MALT1 cleaves the LUBAC subunit HOIL1 to further enhance NF-κB activation. However, the contribution of the LUBAC downstream of GPCRs has not been investigated. By using murine embryonic fibroblasts from mice deficient for HOIP, HOIL1 and SHARPIN, we report that the LUBAC is crucial for the activation of NF-κB in response to LPA. Further echoing the situation in lymphocytes, LPA unbridles the protease activity of MALT1, which cleaves HOIL1 at the Arginine 165. The expression of a MALT1-insensitive version of HOIL1 reveals that this processing is involved in the optimal production of the NF-κB target cytokine interleukin-6. Lastly, we provide evidence that the guanine exchange factor GEF-H1 favors MALT1-mediated cleavage of HOIL1 and NF-κB signaling in this context. Together, our results unveil a critical role for the LUBAC as a positive regulator of NF-κB signaling downstream of LPA receptors.


Assuntos
Lisofosfolipídeos/farmacologia , Complexos Multiproteicos/metabolismo , NF-kappa B/metabolismo , Animais , Proteína 10 de Linfoma CCL de Células B/metabolismo , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Técnicas de Cultura de Células , Fibroblastos/metabolismo , Glicerofosfolipídeos/metabolismo , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lisofosfolipídeos/metabolismo , Camundongos , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
9.
Mol Carcinog ; 58(1): 161-168, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30230038

RESUMO

Head and neck squamous cell carcinoma (HNSCC) represent aggressive classes of tumors with a high mortality rate. The mammalian target of rapamycin (mTOR) pathway is instrumental in their initiation and expansion. Although results from pre-clinical models promise mTOR targeting as a potent novel therapeutic approach, its impact on the tumor microenvironment, such as endothelial cells is only scarcely investigated. Here, we first confirmed the effects of mTOR pharmacological inhibition on cell viability, clonogenicity, and proliferation in HNSCC human cell lines, HN26, and HN30. While Everolimus and Torin1 potently blunted mTOR-based proliferation of HN26 and HN30 lines, endothelial cells were left intact. To further explore the possibility of a paracrine bystander action of HNSCC-treated cells on endothelial cells, conditioned medium from Everolimus- and Torin1-challenged HN26 and HN30 cells were collected and applied to naive human endothelial cells. Although endothelial cell viability was again not modified, morphology and mobility were changed. Indeed, spreading of endothelial cells was altered upon challenge with mTOR-pretreated tumor conditioned-media, as measured via cell impedance and imagery. Interestingly, this was associated with an augmentation of focal adhesion kinase (FAK) active phosphorylation and enhanced migratory behavior. From a molecular standpoint, the production of vascular endothelial growth factor was elevated in treated HNSCC cells and might contribute to FAK phosphorylation. Although mTOR inhibition in tumor cells did hinder their growth, it also favors the release of factors that subsequently enable endothelial cell migration. Further studies will address how this paracrine action may affect tumor-driven angiogenesis upon pharmacological treatments.


Assuntos
Carcinoma de Células Escamosas/patologia , Meios de Cultivo Condicionados/farmacologia , Endotélio Vascular/patologia , Neoplasias de Cabeça e Pescoço/patologia , Comunicação Parácrina , Serina-Treonina Quinases TOR/antagonistas & inibidores , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/metabolismo , Proliferação de Células , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/metabolismo , Humanos , Serina-Treonina Quinases TOR/metabolismo , Células Tumorais Cultivadas
10.
J Cell Sci ; 129(9): 1775-80, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-27006117

RESUMO

Antigen-receptor-mediated activation of lymphocytes relies on a signalosome comprising CARMA1 (also known as CARD11), BCL10 and MALT1 (the CBM complex). The CBM activates nuclear factor κB (NF-κB) transcription factors by recruiting the 'linear ubiquitin assembly complex' (LUBAC), and unleashes MALT1 paracaspase activity. Although MALT1 enzyme shapes NF-κB signaling, lymphocyte activation and contributes to lymphoma growth, the identity of its substrates continues to be elucidated. Here, we report that the LUBAC subunit HOIL1 (also known as RBCK1) is cleaved by MALT1 following antigen receptor engagement. HOIL1 is also constitutively processed in the 'activated B-cell-like' (ABC) subtype of diffuse large B-cell lymphoma (DLBCL), which exhibits aberrant MALT1 activity. We further show that the overexpression of MALT1-insensitive HOIL1 mitigates T-cell-receptor-mediated NF-κB activation and subsequent cytokine production in lymphocytes. Thus, our results unveil HOIL1 as a negative regulator of lymphocyte activation cleaved by MALT1. This cleavage could therefore constitute an appealing therapeutic target for modulating immune responses.


Assuntos
Caspases/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Caspases/genética , Células HEK293 , Humanos , Células Jurkat , Linfoma Difuso de Grandes Células B/genética , Linfoma Difuso de Grandes Células B/metabolismo , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa , NF-kappa B/genética , NF-kappa B/metabolismo , Proteínas de Neoplasias/genética , Receptores de Antígenos de Linfócitos T/genética , Fatores de Transcrição/genética , Ubiquitina-Proteína Ligases/genética
11.
Brain ; 140(11): 2939-2954, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-29053791

RESUMO

Glioblastoma are highly aggressive brain tumours that are associated with an extremely poor prognosis. Within these tumours exists a subpopulation of highly plastic self-renewing cancer cells that retain the ability to expand ex vivo as tumourspheres, induce tumour growth in mice, and have been implicated in radio- and chemo-resistance. Although their identity and fate are regulated by external cues emanating from endothelial cells, the nature of such signals remains unknown. Here, we used a mass spectrometry proteomic approach to characterize the factors released by brain endothelial cells. We report the identification of the vasoactive peptide apelin as a central regulator for endothelial-mediated maintenance of glioblastoma patient-derived cells with stem-like properties. Genetic and pharmacological targeting of apelin cognate receptor abrogates apelin- and endothelial-mediated expansion of glioblastoma patient-derived cells with stem-like properties in vitro and suppresses tumour growth in vivo. Functionally, selective competitive antagonists of apelin receptor were shown to be safe and effective in reducing tumour expansion and lengthening the survival of intracranially xenografted mice. Therefore, the apelin/apelin receptor signalling nexus may operate as a paracrine signal that sustains tumour cell expansion and progression, suggesting that apelin is a druggable factor in glioblastoma.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Animais , Apelina , Receptores de Apelina , Neoplasias Encefálicas/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Endoteliais , Glioblastoma/tratamento farmacológico , Células HEK293 , Humanos , Técnicas In Vitro , Espectrometria de Massas , Camundongos , Terapia de Alvo Molecular , Proteômica , RNA Interferente Pequeno , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Blood ; 123(14): 2199-203, 2014 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-24497531

RESUMO

Antigen receptor-mediated nuclear factor κB (NF-κB) activation relies on the formation of a large multi-protein complex that contains CARMA1, BCL10, and MALT1 (CBM complex). This signalosome is pirated in the activated B-cell-like subgroup of diffuse large B-cell lymphoma (ABC DLBCL) to drive aberrant NF-κB activation, thereby promoting cell survival and propagation. Using an unbiased proteomic approach, we screened for additional components of the CBM in lymphocytes. We found that the linear ubiquitin chain assembly complex (LUBAC), which was previously linked to cytokine-mediated NF-κB activation, dynamically integrates the CBM and marshals NF-κB optimal activation following antigen receptor ligation independently of its catalytic activity. The LUBAC also participates in preassembled CBM complex in cells derived from ABC DLBCL. Silencing the LUBAC reduced NF-κB activation and was toxic in ABC DLBCL cell lines. Thus, our findings reveal a role for the LUBAC during lymphocyte activation and in B-cell malignancy.


Assuntos
Linfoma/metabolismo , NF-kappa B/química , NF-kappa B/metabolismo , Domínios e Motivos de Interação entre Proteínas/fisiologia , Receptores de Antígenos de Linfócitos T/metabolismo , Ubiquitina/metabolismo , Catálise , Linhagem Celular Tumoral , Humanos , Células Jurkat , Ativação Linfocitária/fisiologia , Linfoma/patologia , Ligação Proteica , Ubiquitinação/fisiologia
13.
Nutr Cancer ; 68(5): 838-47, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27266882

RESUMO

Oral squamous cell carcinoma (OSCC) exhibited high chemoresistance to current treatments. Here we aimed at identifying and repositioning approved drugs that could be selectively toxic toward OSCC cells. Through a cell-based drug screening of 1,280 chemical molecules, we selected compounds lethal to oral cancer SCC-25 cells, while sparing normal keratinocyte HaCaT cells. Within the chemical library, the natural flavonoid luteolin was identified as a potent cytotoxic agent against oral cancer cells in vitro, along with metixene hydrochloride and nitazoxanide. Of note, they exhibit low toxicity and high efficiency compared to the standard-of-care, such as cisplatin and the epidermal growth factor receptor inhibitor tyrphostin. From a molecular standpoint, luteolin causes phosphorylation of ataxia telangiectasia mutated (ATM) and H2AX in a DNA repair pathway and can be efficiently combined with a checkpoint kinase (CHK) pharmacological inhibitor. Thus, luteolin emerges as a potent cytotoxic and/or adjuvant therapy in oral cancer, as it is a natural compound presenting better effects in vitro compared to conventional chemotherapeutic agents. Future in vivo exploration is next required to provide the proof-of-concept that luteolin could be an efficient anticancer molecule.


Assuntos
Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/patologia , Dano ao DNA/efeitos dos fármacos , Luteolina/farmacologia , Neoplasias Bucais/patologia , Carcinoma de Células Escamosas/tratamento farmacológico , Linhagem Celular Tumoral , Cisplatino/farmacologia , Relação Dose-Resposta a Droga , Quimioterapia Combinada , Humanos , Neoplasias Bucais/tratamento farmacológico , Nitrocompostos , Tiazóis/farmacologia , Tioxantenos/farmacologia , Tirfostinas/farmacologia
14.
Cell Commun Signal ; 13: 11, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25889342

RESUMO

BACKGROUND: The viral G protein-coupled receptor (vGPCR) is proposed to act as one of the predominant mediators of Kaposi's sarcoma (KS), a human herpes virus 8 (HHV8)-elicited disease. The actions of vGPCR manifest pathogenesis, in part, through increased permeability of endothelial cells. Endothelial cell-cell junctions have indeed emerged as an instrumental target involved in the vasculature defects observed within the tumor microenvironment. The pathway leading to adherens junction destabilization has been shown to involve the activation of the small GTPase Rac, in the context of either latent infection or the sole expression of vGPCR. However, the precise molecular mechanisms governed by vGPCR in vascular leakage require further elucidation. FINDINGS: Guanine exchange factors (GEFs) function as critical molecular switches that control the activation of small GTPases. We therefore screened the effects of 80 siRNAs targeting GEFs on vGPCR-driven endothelial permeability and identified switch-associated protein 70 (SWAP70) as necessary for its elevating effects. Pull-down experiments further showed that Rac activation by vGPCR was dependent on SWAP70. Examination of tissues and cells from HHV8-positive patients revealed that SWAP70 was ubiquitously expressed. Furthermore, SWAP70 was found to be crucial for vGPCR-driven endothelial tube formation and endothelial sprouting in vitro. CONCLUSIONS: SWAP70 appears to act as a molecular intermediate between vGPCR and endothelial activation. Because of the important role of vGPCR-mediated endothelial plasticity in KS pathogenesis, inhibition of SWAP70 function could be of interest for blocking vGPCR-driven activities in HHV8-defined diseases.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Herpesvirus Humano 8/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Proteínas Nucleares/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Virais/metabolismo , Proteínas de Ligação a DNA/genética , Fatores de Troca do Nucleotídeo Guanina/genética , Herpesvirus Humano 8/genética , Humanos , Antígenos de Histocompatibilidade Menor , Proteínas Nucleares/genética , Receptores Acoplados a Proteínas G/genética , Proteínas Virais/genética
15.
BMC Cell Biol ; 15: 38, 2014 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-25339290

RESUMO

BACKGROUND: The interleukin-8 chemokine (IL-8) G-protein coupled receptor CXCR2 governs pro-inflammatory and pro-angiogenic responses in leukocytes and endothelial cells. At a molecular standpoint, CXCR2 is widely reported to operate through calcium flux, phosphoinoisitide 3 kinase (PI3K) and mitogen-activated protein kinase (MAPK). While CXCR2 trafficking is suspected to be intertwined with its signaling, the exact mechanism is not fully elucidated. RESULTS: Here, we identified the lysine 327 within the CXCR2 C-terminal tail as a key residue for ubiquitination, internalization, and signaling. First, the substitution to an arginine of K327 mutation was associated with a reduction in CXCR2 poly-ubiquitination. While WT CXCR2 was rapidly internalized following IL-8 administration, K327R mutant remained at the plasma membrane. Finally, K327R mutant failed to promote the recruitment of ß-arrestin2, as estimated by imagery and bioluminescence resonance transfer. As a consequence, the activation of intracellular signaling, including both early events such as ERK phosphorylation and the increase in calcium flux, and the latter activation of the AP1 and NF-κB transcription factors, was blunted. CONCLUSIONS: Overall, our results demonstrate that CXCR2 ubiquitination on K327 residue modulates agonist-activated CXCR2 cell sorting and intracellular signaling. Thus, the inhibition of K327 ubiquitination might emerge as an effective mean to curb exacerbated CXCR2 signaling in several pathological conditions, such as inflammatory diseases and cancer.


Assuntos
Lisina/análise , Receptores de Interleucina-8B/metabolismo , Ubiquitinação , Sequência de Aminoácidos , Animais , Células HEK293 , Humanos , Interleucina-8/metabolismo , Lisina/metabolismo , Dados de Sequência Molecular , Transporte Proteico , Receptores de Interleucina-8B/química , Alinhamento de Sequência , Transdução de Sinais
16.
J Cell Sci ; 125(Pt 17): 4137-46, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22685328

RESUMO

VE-cadherin-mediated cell-cell junction weakening increases paracellular permeability in response to both angiogenic and inflammatory stimuli. Although Semaphorin 3A has emerged as one of the few known anti-angiogenic factors to exhibit pro-permeability activity, little is known about how it triggers vascular leakage. Here we report that Semaphorin 3A induced VE-cadherin serine phosphorylation and internalisation, cell-cell junction destabilisation, and loss of barrier integrity in brain endothelial cells. In addition, high-grade glioma-isolated tumour-initiating cells were found to secrete Semaphorin 3A, which promoted brain endothelial monolayer permeability. From a mechanistic standpoint, Semaphorin 3A impinged upon the basal activity of the serine phosphatase PP2A and disrupted PP2A interaction with VE-cadherin, leading to cell-cell junction disorganization and increased permeability. Accordingly, both pharmacological inhibition and siRNA-based knockdown of PP2A mimicked Semaphorin 3A effects on VE-cadherin. Hence, local Semaphorin 3A production impacts on the PP2A/VE-cadherin equilibrium and contributes to elevated vascular permeability.


Assuntos
Permeabilidade da Membrana Celular , Células Endoteliais/enzimologia , Células Endoteliais/patologia , Proteína Fosfatase 2/metabolismo , Semaforina-3A/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Ativação Enzimática , Glioma/enzimologia , Glioma/metabolismo , Glioma/patologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neoplásicas/metabolismo , Proteína Fosfatase 2/antagonistas & inibidores , Quinases da Família src/metabolismo
17.
Am J Pathol ; 183(6): 1993-2006, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24120378

RESUMO

Preeclampsia is a pregnancy disorder characterized by hypertension and proteinuria. In preeclampsia, the placenta releases factors into the maternal circulation that cause a systemic endothelial dysfunction. Herein, we investigated the effects of plasma from women with preeclamptic and normal pregnancies on the transcriptome of an immortalized human umbilical vein endothelial cell line. The cells were exposed for 24 hours to preeclamptic or normal pregnancy plasma and their transcriptome was analyzed using Agilent microarrays. A total of 116 genes were found differentially expressed: 71 were up-regulated and 45 were down-regulated. In silico analysis revealed significant consistency and identified four functional categories of genes: mitosis and cell cycle progression, anti-apoptotic, fatty acid biosynthesis, and endoplasmic reticulum stress effectors. Moreover, several genes involved in vasoregulation and endothelial homeostasis showed modified expression, including EDN1, APLN, NOX4, and CBS. Promoter analysis detected, among the up-regulated genes, a significant overrepresentation of genes containing activation protein-1 regulatory sites. This correlated with down-regulation of JDP2, a gene encoding a repressor of activation protein-1. The role of JDP2 in the regulation of a subset of genes in the human umbilical vein endothelial cells was confirmed by siRNA inhibition. We characterized transcriptional changes induced by preeclamptic plasma on human umbilical vein endothelial cells, and identified, for the first time to our knowledge, JDP2 as a regulator of a subset of genes modified by preeclamptic plasma.


Assuntos
Células Endoteliais , Regulação da Expressão Gênica , Plasma/metabolismo , Pré-Eclâmpsia , Proteínas Repressoras/metabolismo , Fator de Transcrição AP-1/metabolismo , Adulto , Linhagem Celular Transformada , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Feminino , Humanos , Pré-Eclâmpsia/sangue , Pré-Eclâmpsia/patologia , Gravidez , Transcriptoma
18.
Blood ; 120(13): 2745-56, 2012 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-22898603

RESUMO

DEP-1/CD148 is a receptor-like protein tyrosine phosphatase with antiproliferative and tumor-suppressive functions. Interestingly, it also positively regulates Src family kinases in hematopoietic and endothelial cells, where we showed it promotes VE-cadherin-associated Src activation and endothelial cell survival upon VEGF stimulation. However, the molecular mechanism involved and its biologic functions in endothelial cells remain ill-defined. We demonstrate here that DEP-1 is phosphorylated in a Src- and Fyn-dependent manner on Y1311 and Y1320, which bind the Src SH2 domain. This allows DEP-1-catalyzed dephosphorylation of Src inhibitory Y529 and favors the VEGF-induced phosphorylation of Src substrates VE-cadherin and Cortactin. Accordingly, RNA interference (RNAi)-mediated knockdown of DEP-1 or expression of DEP-1 Y1311F/Y1320F impairs Src-dependent biologic responses mediated by VEGF including permeability, invasion, and branching capillary formation. In addition, our work further reveals that above a threshold expression level, DEP-1 can also dephosphorylate Src Y418 and attenuate downstream signaling and biologic responses, consistent with the quiescent behavior of confluent endothelial cells that express the highest levels of endogenous DEP-1. Collectively, our findings identify the VEGF-dependent phosphorylation of DEP-1 as a novel mechanism controlling Src activation, and show this is essential for the proper regulation of permeability and the promotion of the angiogenic response.


Assuntos
Capilares/metabolismo , Permeabilidade da Membrana Celular , Endotélio Vascular/citologia , Neovascularização Patológica , Tirosina/metabolismo , Quinases da Família src/metabolismo , Antígenos CD/metabolismo , Western Blotting , Caderinas/metabolismo , Adesão Celular , Movimento Celular , Proliferação de Células , Células Cultivadas , Cortactina/metabolismo , Endotélio Vascular/metabolismo , Imunofluorescência , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Imunoprecipitação , Mutação/genética , Invasividade Neoplásica , Fosforilação , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/genética , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/metabolismo , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo
19.
Med Sci (Paris) ; 30(4): 408-14, 2014 Apr.
Artigo em Francês | MEDLINE | ID: mdl-24801036

RESUMO

Tumor initiation and progression is a complex process in which cells acquire aberrant proliferation, survival and migration properties, and the ability to form a dedicated blood vessel network. Recent studies highlighted the existence of an active crosstalk between endothelial cells and the tumor mass. Indeed, cancer stem-like cells have been identified and found in the vicinity of blood vessels, and the latter has been proposed to act as a feeding bed for tumors, especially governing the fate of cancer stem-like cells. Here, we present an overview of the direct interplay between endothelial and cancer cells. We will first introduce the mechanisms involved in tumor angiogenesis. How the microenvironment impacts on endothelial plasticity will next be detailed, focusing on the tumor main secreted factors. Finally, the role of the vascular niche and its angiocrine factors in tumorigenesis will be addressed.


Assuntos
Neoplasias/irrigação sanguínea , Neovascularização Patológica , Humanos , Neoplasias/patologia , Neovascularização Patológica/patologia
20.
STAR Protoc ; 5(2): 103121, 2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38850538

RESUMO

Lysosomes are critical for the sustenance of glioblastoma stem-like cells (GSCs) properties. We present a protocol to enrich and purify lysosomes from patient-derived GSCs in culture. We describe the steps required to stably express a tagged lysosomal protein in GSCs, mechanically lyse cells, magnetically immunopurify lysosomes, and qualitatively assess these organelles. We then detail the procedure for retrieving intact and purified lysosomes from GSCs. We also specify cell culture conditions, storage procedures, and sample preparation for immunoblotting. For complete details on the use and execution of this protocol, please refer to Maghe et al.1.


Assuntos
Glioblastoma , Imunoprecipitação , Lisossomos , Células-Tronco Neoplásicas , Humanos , Glioblastoma/patologia , Glioblastoma/metabolismo , Lisossomos/metabolismo , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Imunoprecipitação/métodos , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA