Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Mol Oncol ; 17(10): 1981-1999, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-36912768

RESUMO

Human epidermal growth factor receptor 2 (ErbB2/HER2) is a tyrosine kinase receptor that is overexpressed in 25% of primary human breast cancers, as well as in multiple other cancers. HER2-targeted therapies improved progression-free and overall survival in patients with HER2+ breast cancers. However, associated resistance mechanisms and toxicity highlight the need for new therapeutic approaches for these cancers. We recently established that, in normal cells, HER2 is stabilized in a catalytically repressed state by direct interaction with members of the ezrin/radixin/moesin (ERM) family. In HER2-overexpressing tumors, the low expression of moesin contributes to the aberrant activation of HER2. Through a screen designed to find moesin-mimicking compounds, we identified ebselen oxide. We show that ebselen oxide, and some derivatives, conferred an efficient allosteric inhibition of overexpressed HER2, as well as mutated and truncated oncogenic forms of HER2, which are resistant to current therapies. Ebselen oxide selectively inhibited anchorage-dependent and -independent proliferation of HER2+ cancer cells and showed a significant benefit in combination with current anti-HER2 therapeutic agents. Finally, ebselen oxide significantly blocked HER2+ breast tumor progression in vivo. Collectively, these data provide evidence that ebselen oxide is a newly identified allosteric inhibitor of HER2 to be considered for therapeutic intervention on HER2+ cancers.


Assuntos
Antineoplásicos , Neoplasias da Mama , Humanos , Feminino , Lapatinib/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia , Receptor ErbB-2/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral
2.
Fluids Barriers CNS ; 19(1): 41, 2022 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-35658915

RESUMO

Endothelial cells (ECs) are constantly submitted in vivo to hemodynamical forces derived from the blood circulation, including shear stress (SS). ECs are able to detect SS and consequently adapt their phenotype, thus affecting many endothelial functions. If a plethora of shear stress-regulated molecular networks have been described in peripheral ECs, less is known about the molecular responses of microvascular brain ECs which constitute the blood-brain barrier (BBB). In this work, we investigated the response of human cerebral microvascular ECs to laminar physiological shear stress using the well characterized hCMEC/D3 cell line. Interestingly, we showed that hCMEC/D3 cells responded to shear stress by aligning perpendicularly to the flow direction, contrary to peripheral endothelial cells which aligned in the flow direction. Whole proteomic profiles were compared between hCMEC/D3 cells cultured either in static condition or under 5 or 10 dyn.cm-2 SS for 3 days. 3592 proteins were identified and expression levels were significantly affected for 3% of them upon both SS conditions. Pathway analyses were performed which revealed that most proteins overexpressed by SS refer to the antioxidant defense, probably mediated by activation of the NRF2 transcriptional factor. Regarding down-regulated proteins, most of them participate to the pro-inflammatory response, cell motility and proliferation. These findings confirm the induction of EC quiescence by laminar physiological SS and reveal a strong protective effect of SS on hCMEC/D3 cells, suggesting a similar effect on the BBB. Our results also showed that SS did not significantly increase expression levels nor did it affect the localization of junctional proteins and did not afect either the functional activity of several ABC transporters (P-glycoprotein and MRPs). This work provides new insights on the response of microvascular brain ECs to SS and on the importance of SS for optimizing in vitro BBB models.


Assuntos
Células Endoteliais , Proteômica , Barreira Hematoencefálica/metabolismo , Encéfalo/irrigação sanguínea , Células Cultivadas , Células Endoteliais/metabolismo , Humanos , Estresse Mecânico
3.
J Neurochem ; 161(6): 492-505, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34822163

RESUMO

COVID-19 is associated with encephalitis in critically ill patients and endothelial dysfunction seems to contribute to this life-threatening complication. Our objective was to determine the hallmark of endothelial activation in COVID-19-related encephalitis. In an observational study in intensive care unit (ICU), we compared vascular biomarkers of critically ill COVID-19 patients with or without encephalitis. To be classified in the encephalitis group, patients had to have new onset of central neurologic symptom, and pathological findings on either brain magnetic resonance imaging (MRI) and/or electroencephalogram (EEG). Among the 32 critically ill COVID-19 consecutive patients, 21 were categorized in the control group and 11 in the encephalitis group. Encephalitis patients had a longer ICU stay than control patients (median length [25th-75th percentile] of 52 [16-79] vs. 20.5 [11-44] days, respectively, p = 0.04). Nine-month overall follow-up mortality reached 21% (7/32 patients), with mortality rates in the encephalitis group and the control group of 27% and 19%, respectively. Encephalitis was associated with significant higher release of soluble endothelial activation markers (sE-selectin, tumor necrosis factor-α (TNF-α), interleukin 6, placental growth factor, and thrombomodulin), but these increases were correlated with TNF-α plasmatic levels. The hypoxia-inducible protein angiopoietin-like 4 (ANGPTL4) was at significantly higher levels in encephalitis patients compared to control patients (p = 0.0099), and in contrary to the other increased factors, was not correlated with TNF-α levels (r = 0.2832, p = 0.1163). Our findings suggest that COVID-19-related encephalitis is a cytokine-associated acute brain dysfunction. ANGPTL4 was the only elevated marker found in encephalitis patients, which was not correlated with systemic inflammation, suggesting that ANGPTL4 might be a relevant factor to predict encephalitis in critically ill COVID-19 patients.


Assuntos
COVID-19 , Encefalite , Proteína 4 Semelhante a Angiopoietina/metabolismo , Biomarcadores , COVID-19/complicações , Estado Terminal , Encefalite/virologia , Células Endoteliais , Humanos , Unidades de Terapia Intensiva , Fator de Necrose Tumoral alfa
4.
Proc Natl Acad Sci U S A ; 118(45)2021 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-34725157

RESUMO

Neisseria meningitidis utilizes type IV pili (T4P) to adhere to and colonize host endothelial cells, a process at the heart of meningococcal invasive diseases leading to meningitis and sepsis. T4P are polymers of an antigenically variable major pilin building block, PilE, plus several core minor pilins that initiate pilus assembly and are thought to be located at the pilus tip. Adhesion of N. meningitidis to human endothelial cells requires both PilE and a conserved noncore minor pilin PilV, but the localization of PilV and its precise role in this process remains to be clarified. Here, we show that both PilE and PilV promote adhesion to endothelial vessels in vivo. The substantial adhesion defect observed for pilV mutants suggests it is the main adhesin. Consistent with this observation, superresolution microscopy showed the abundant distribution of PilV throughout the pilus. We determined the crystal structure of PilV and modeled it within the pilus filament. The small size of PilV causes it to be recessed relative to adjacent PilE subunits, which are dominated by a prominent hypervariable loop. Nonetheless, we identified a conserved surface-exposed adhesive loop on PilV by alanine scanning mutagenesis. Critically, antibodies directed against PilV inhibit N. meningitidis colonization of human skin grafts. These findings explain how N. meningitidis T4P undergo antigenic variation to evade the humoral immune response while maintaining their adhesive function and establish the potential of this highly conserved minor pilin as a vaccine and therapeutic target for the prevention and treatment of N. meningitidis infections.


Assuntos
Aderência Bacteriana , Proteínas de Bactérias/fisiologia , Fímbrias Bacterianas/fisiologia , Neisseria meningitidis/fisiologia , Animais , Anticorpos/uso terapêutico , Proteínas de Bactérias/química , Proteínas de Bactérias/ultraestrutura , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos , Feminino , Fímbrias Bacterianas/química , Fímbrias Bacterianas/ultraestrutura , Humanos , Infecções Meningocócicas/tratamento farmacológico , Camundongos SCID
5.
Cancer Res ; 81(21): 5464-5476, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34493594

RESUMO

Therapies targeting the tyrosine kinase receptor HER2 have significantly improved survival of patients with HER2+ cancer. However, both de novo and acquired resistance remain a challenge, particularly in the brain metastatic setting. Here we report that, unlike other HER tyrosine kinase receptors, HER2 possesses a binding motif in its cytosolic juxtamembrane region that allows interaction with members of the Ezrin/Radixin/Moesin (ERM) family. Under physiologic conditions, this interaction controls the localization of HER2 in ERM-enriched domains and stabilizes HER2 in a catalytically repressed state. In HER2+ breast cancers, low expression of Moesin correlated with increased HER2 expression. Restoring expression of ERM proteins in HER2+ breast cancer cells was sufficient to revert HER2 activation and inhibit HER2-dependent proliferation. A high-throughput assay recapitulating the HER2-ERM interaction allowed for screening of about 1,500 approved drugs. From this screen, we found Zuclopenthixol, an antipsychotic drug that behaved as a Moesin-mimicking compound, because it directly binds the juxtamembrane region of HER2 and specifically inhibits HER2 activation in HER2+ cancers, as well as activation of oncogenic mutated and truncated forms of HER2. Zuclopenthixol efficiently inhibited HER2+ breast tumor progression in vitro and in vivo and, more importantly, showed significant activity on HER2+ brain tumor progression. Collectively, these data reveal a novel class of allosteric HER2 inhibitors, increasing the number of approaches to consider for intervention on HER2+ breast cancers and brain metastases. SIGNIFICANCE: This study demonstrates the functional role of Moesin in maintaining HER2 in a catalytically repressed state and provides novel therapeutic approaches targeting HER2+ breast cancers and brain metastasis using Moesin-mimicking compounds.


Assuntos
Biomimética/métodos , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias da Mama/tratamento farmacológico , Clopentixol/farmacologia , Regulação Neoplásica da Expressão Gênica , Proteínas dos Microfilamentos/metabolismo , Receptor ErbB-2/antagonistas & inibidores , Regulação Alostérica , Animais , Apoptose , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/secundário , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proliferação de Células , Antagonistas de Dopamina/farmacologia , Feminino , Humanos , Camundongos , Camundongos Nus , Proteínas dos Microfilamentos/genética , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
6.
C R Biol ; 344(2): 127-143, 2021 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-34213851

RESUMO

Neisseria meningitidis (meningococcus) is a Gram-negative bacterium responsible for two devastating forms of invasive diseases: purpura fulminans and meningitis. Since the first description of the epidemic nature of the illness at the dawn of the nineteenth century, the scientific knowledge of meningococcal infection has increased greatly. Major advances have been made in the management of the disease with the advent of antimicrobial therapy and the implementation of meningococcal vaccines. More recently, an extensive knowledge has been accumulated on meningococcal interaction with its human host, revealing key processes involved in disease progression and new promising therapeutic approaches.


Neisseria meningitidis (méningocoque) est une bactérie à Gram négatif responsable de deux formes gravissimes de maladies invasives : le purpura fulminans et la méningite. Depuis la première description du caractère épidémique de la maladie à l'aube du 19e siècle, les connaissances scientifiques sur les infections méningococciques ont considérablement augmenté. Des progrès majeurs ont été réalisés dans la gestion de la maladie avec l'avènement des agents antimicrobiens et le développement de vaccins contre le méningocoque. De nombreuses connaissances ont récemment été accumulées sur son interaction avec l'être humain, son unique hôte, révélant les processus clés impliqués dans la progression de la maladie et de nouvelles approches thérapeutiques prometteuses.


Assuntos
Infecções Meningocócicas , Neisseria meningitidis , Púrpura Fulminante , Antibacterianos , Humanos , Infecções Meningocócicas/tratamento farmacológico , Púrpura Fulminante/tratamento farmacológico
7.
PLoS Pathog ; 17(2): e1009299, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33592056

RESUMO

Neisseria meningitidis (the meningococcus) remains a major cause of bacterial meningitis and fatal sepsis. This commensal bacterium of the human nasopharynx can cause invasive diseases when it leaves its niche and reaches the bloodstream. Blood-borne meningococci have the ability to adhere to human endothelial cells and rapidly colonize microvessels. This crucial step enables dissemination into tissues and promotes deregulated inflammation and coagulation, leading to extensive necrotic purpura in the most severe cases. Adhesion to blood vessels relies on type IV pili (TFP). These long filamentous structures are highly dynamic as they can rapidly elongate and retract by the antagonistic action of two ATPases, PilF and PilT. However, the consequences of TFP dynamics on the pathophysiology and the outcome of meningococcal sepsis in vivo have been poorly studied. Here, we show that human graft microvessels are replicative niches for meningococci, that seed the bloodstream and promote sustained bacteremia and lethality in a humanized mouse model. Intriguingly, although pilus-retraction deficient N. meningitidis strain (ΔpilT) efficiently colonizes human graft tissue, this mutant did not promote sustained bacteremia nor induce mouse lethality. This effect was not due to a decreased inflammatory response, nor defects in bacterial clearance by the innate immune system. Rather, TFP-retraction was necessary to promote the release of TFP-dependent contacts between bacteria and, in turn, the detachment from colonized microvessels. The resulting sustained bacteremia was directly correlated with lethality. Altogether, these results demonstrate that pilus retraction plays a key role in the occurrence and outcome of meningococcal sepsis by supporting sustained bacteremia. These findings open new perspectives on the role of circulating bacteria in the pathological alterations leading to lethal sepsis.


Assuntos
Bacteriemia/microbiologia , Modelos Animais de Doenças , Proteínas de Fímbrias/metabolismo , Fímbrias Bacterianas/fisiologia , Infecções Meningocócicas/microbiologia , Neisseria meningitidis/patogenicidade , Sepse/microbiologia , Animais , Bacteriemia/metabolismo , Bacteriemia/patologia , Aderência Bacteriana , Células Endoteliais , Feminino , Proteínas de Fímbrias/genética , Humanos , Infecções Meningocócicas/metabolismo , Infecções Meningocócicas/patologia , Camundongos , Camundongos SCID , Sepse/metabolismo , Sepse/patologia , Transplante de Pele
8.
Cell Microbiol ; 22(4): e13185, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32185901

RESUMO

Neisseria meningitidis (meningococcus) is a Gram-negative bacterium responsible for two devastating forms of invasive diseases: purpura fulminans and meningitis. Interaction with both peripheral and cerebral microvascular endothelial cells is at the heart of meningococcal pathogenesis. During the last two decades, an essential role for meningococcal type IV pili in vascular colonisation and disease progression has been unravelled. This review summarises 20 years of research on meningococcal type IV pilus-dependent virulence mechanisms, up to the identification of promising anti-virulence compounds that target type IV pili.


Assuntos
Aderência Bacteriana , Fímbrias Bacterianas/classificação , Fímbrias Bacterianas/metabolismo , Infecções Meningocócicas/microbiologia , Neisseria meningitidis/patogenicidade , Animais , Células Endoteliais/microbiologia , Humanos , Camundongos , Virulência
9.
Proc Natl Acad Sci U S A ; 117(5): 2606-2612, 2020 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-31964828

RESUMO

Bacterial infections are frequently based on the binding of lectin-like adhesins to specific glycan determinants exposed on host cell receptors. These interactions confer species-specific recognition and tropism for particular host tissues and represent attractive antibacterial targets. However, the wide structural diversity of carbohydrates hampers the characterization of specific glycan determinants. Here, we characterized the receptor recognition of type IV pili (Tfp), a key adhesive factor present in numerous bacterial pathogens, using Neisseria meningitidis as a model organism. We found that meningococcal Tfp specifically recognize a triantennary sialylated poly-N-acetyllactosamine-containing N-glycan exposed on the human receptor CD147/Basigin, while fucosylated derivatives of this N-glycan impaired bacterial adhesion. Corroborating the inhibitory role of fucosylation on receptor recognition, adhesion of the meningococcus on nonhuman cells expressing human CD147 required prior defucosylation. These findings reveal the molecular basis of the selective receptor recognition by meningococcal Tfp and thereby, identify a potential antibacterial target.


Assuntos
Adesinas Bacterianas/metabolismo , Proteínas de Fímbrias/metabolismo , Infecções Meningocócicas/metabolismo , Neisseria meningitidis/metabolismo , Receptores de Superfície Celular/metabolismo , Adesinas Bacterianas/genética , Proteínas de Fímbrias/genética , Fímbrias Bacterianas/genética , Fímbrias Bacterianas/metabolismo , Glicosilação , Humanos , Infecções Meningocócicas/genética , Infecções Meningocócicas/microbiologia , Neisseria meningitidis/genética , Polissacarídeos/metabolismo , Receptores de Superfície Celular/genética
10.
Cell Microbiol ; 22(1): e13132, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31658405

RESUMO

The skull, spine, meninges, and cellular barriers at the blood-brain and the blood-cerebrospinal fluid interfaces well protect the brain and meningeal spaces against microbial invasion. However, once in the bloodstream, a range of pathogenic bacteria is able to reach the brain and cause meningitis. Despite advances in antibacterial therapy, bacterial meningitis remains one of the most important infectious diseases worldwide. The most common causative bacteria in children and adults are Streptococcus pneumoniae and Neisseria meningitidis associated with high morbidity and mortality, while among neonates, most cases of bacterial meningitis are due to group B Streptococcus and Escherichia coli. Here we summarise our current knowledge on the strategies used by these bacterial pathogens to survive in the bloodstream, to colonise the brain vasculature and to cross the blood-brain barrier.


Assuntos
Bactérias/patogenicidade , Barreira Hematoencefálica/microbiologia , Animais , Transporte Biológico , Encéfalo/microbiologia , Células Endoteliais/microbiologia , Humanos , Inflamação , Neisseria meningitidis/patogenicidade , Neisseria meningitidis/fisiologia , Streptococcus pneumoniae/patogenicidade , Streptococcus pneumoniae/fisiologia , Fatores de Virulência
11.
J Nanobiotechnology ; 17(1): 115, 2019 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-31711496
12.
Nat Commun ; 10(1): 4430, 2019 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-31562326

RESUMO

Zika virus (ZIKV) invades and persists in the central nervous system (CNS), causing severe neurological diseases. However the virus journey, from the bloodstream to tissues through a mature endothelium, remains unclear. Here, we show that ZIKV-infected monocytes represent suitable carriers for viral dissemination to the CNS using human primary monocytes, cerebral organoids derived from embryonic stem cells, organotypic mouse cerebellar slices, a xenotypic human-zebrafish model, and human fetus brain samples. We find that ZIKV-exposed monocytes exhibit higher expression of adhesion molecules, and higher abilities to attach onto the vessel wall and transmigrate across endothelia. This phenotype is associated to enhanced monocyte-mediated ZIKV dissemination to neural cells. Together, our data show that ZIKV manipulates the monocyte adhesive properties and enhances monocyte transmigration and viral dissemination to neural cells. Monocyte transmigration may represent an important mechanism required for viral tissue invasion and persistence that could be specifically targeted for therapeutic intervention.


Assuntos
Moléculas de Adesão Celular/metabolismo , Monócitos/metabolismo , Monócitos/virologia , Neurônios/metabolismo , Migração Transendotelial e Transepitelial/fisiologia , Infecção por Zika virus/metabolismo , Zika virus/fisiologia , Zika virus/patogenicidade , Animais , Adesão Celular/fisiologia , Sobrevivência Celular , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Sistema Nervoso Central/virologia , Cerebelo/patologia , Cerebelo/virologia , Modelos Animais de Doenças , Células-Tronco Embrionárias , Endotélio/virologia , Feminino , Humanos , Monócitos/patologia , Neurônios/patologia , Neurônios/virologia , Organoides/metabolismo , Organoides/patologia , Peixe-Zebra , Infecção por Zika virus/patologia , Infecção por Zika virus/virologia
13.
Cell Microbiol ; 21(11): e13063, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31167044

RESUMO

Neisseria meningitidis is a Gram-negative bacterium that asymptomatically colonises the nasopharynx of humans. For an unknown reason, N. meningitidis can cross the nasopharyngeal barrier and invade the bloodstream where it becomes one of the most harmful extracellular bacterial pathogen. This infectious cycle involves the colonisation of two different environments. (a) In the nasopharynx, N. meningitidis grow on the top of mucus-producing epithelial cells surrounded by a complex microbiota. To survive and grow in this challenging environment, the meningococcus expresses specific virulence factors such as polymorphic toxins and MDAΦ. (b) Meningococci have the ability to survive in the extra cellular fluids including blood and cerebrospinal fluid. The interaction of N. meningitidis with human endothelial cells leads to the formation of typical microcolonies that extend overtime and promote vascular injury, disseminated intravascular coagulation, and acute inflammation. In this review, we will focus on the interplay between N. meningitidis and these two different niches at the cellular and molecular level and discuss the use of inhibitors of piliation as a potent therapeutic approach.


Assuntos
Infecções Meningocócicas/microbiologia , Nasofaringe/microbiologia , Neisseria meningitidis/patogenicidade , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Vasos Sanguíneos/microbiologia , Células Endoteliais/patologia , Células Epiteliais/patologia , Interações entre Hospedeiro e Microrganismos , Humanos , Inovirus/crescimento & desenvolvimento , Inovirus/patogenicidade , Infecções Meningocócicas/sangue , Infecções Meningocócicas/líquido cefalorraquidiano , Neisseria meningitidis/metabolismo , Fatores de Virulência
14.
Nat Microbiol ; 4(6): 972-984, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30911127

RESUMO

Bacterial virulence factors are attractive targets for the development of therapeutics. Type IV pili, which are associated with a remarkable array of properties including motility, the interaction between bacteria and attachment to biotic and abiotic surfaces, represent particularly appealing virulence factor targets. Type IV pili are present in numerous bacterial species and are critical for their pathogenesis. In this study, we report that trifluoperazine and related phenothiazines block functions associated with Type IV pili in different bacterial pathogens, by affecting piliation within minutes. Using Neisseria meningitidis as a paradigm of Gram-negative bacterial pathogens that require Type IV pili for pathogenesis, we show that piliation is sensitive to altered activity of the Na+ pumping NADH-ubiquinone oxidoreductase (Na+-NQR) complex and that these compounds probably altered the establishment of the sodium gradient. In vivo, these compounds exert a strong protective effect. They reduce meningococcal colonization of the human vessels and prevent subsequent vascular dysfunctions, intravascular coagulation and overwhelming inflammation, the hallmarks of invasive meningococcal infections. Finally, they reduce lethality. This work provides a proof of concept that compounds with activity against bacterial Type IV pili could beneficially participate in the treatment of infections caused by Type IV pilus-expressing bacteria.


Assuntos
Fímbrias Bacterianas/efeitos dos fármacos , Fímbrias Bacterianas/fisiologia , Infecções Meningocócicas/prevenção & controle , Neisseria meningitidis/efeitos dos fármacos , Fatores de Virulência , Animais , Antibacterianos/farmacologia , Vasos Sanguíneos/lesões , Vasos Sanguíneos/microbiologia , Vasos Sanguíneos/patologia , Combinação de Medicamentos , Complexo I de Transporte de Elétrons , Feminino , Fímbrias Bacterianas/genética , Perfilação da Expressão Gênica , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Bactérias Gram-Negativas , Humanos , Camundongos , Neisseria meningitidis/genética , Neisseria meningitidis/crescimento & desenvolvimento , Fenotiazinas/farmacologia , Pele/patologia , Transplante de Pele , ATPase Trocadora de Sódio-Potássio , Trifluoperazina/farmacologia
15.
Mol Pharm ; 16(3): 1312-1326, 2019 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-30721081

RESUMO

The effect of cannabidiol (CBD), a high-affinity agonist of the transient receptor potential vanilloid-2 (TRPV2) channel, has been poorly investigated in human brain microvessel endothelial cells (BMEC) forming the blood-brain barrier (BBB). TRPV2 expression and its role on Ca2+ cellular dynamics, trans-endothelial electrical resistance (TEER), cell viability and growth, migration, and tubulogenesis were evaluated in human primary cultures of BMEC (hPBMEC) or in the human cerebral microvessel endothelial hCMEC/D3 cell line. Abundant TRPV2 expression was measured in hCMEC/D3 and hPBMEC by qRT-PCR, Western blotting, nontargeted proteomics, and cellular immunofluorescence studies. Intracellular Ca2+ levels were increased by heat and CBD and blocked by the nonspecific TRP antagonist ruthenium red (RR) and the selective TRPV2 inhibitor tranilast (TNL) or by silencing cells with TRPV2 siRNA. CBD dose-dependently induced the hCMEC/D3 cell number (EC50 0.3 ± 0.1 µM), and this effect was fully abolished by TNL or TRPV2 siRNA. A wound healing assay showed that CBD induced cell migration, which was also inhibited by TNL or TRPV2 siRNA. Tubulogenesis of hCMEC/D3 cells in 3D matrigel cultures was significantly increased by 41 and 73% after a 7 or 24 h CBD treatment, respectively, and abolished by TNL. CBD also increased the TEER of hPBMEC monolayers cultured in transwell, and this was blocked by TNL. Our results show that CBD, at extracellular concentrations close to those observed in plasma of patients treated by CBD, induces proliferation, migration, tubulogenesis, and TEER increase in human brain endothelial cells, suggesting CBD might be a potent target for modulating the human BBB.


Assuntos
Neoplasias Encefálicas/irrigação sanguínea , Canabidiol/farmacologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Endoteliais/metabolismo , Microvasos/patologia , Canais de Cátion TRPV/metabolismo , Barreira Hematoencefálica/metabolismo , Cálcio/metabolismo , Cannabis/química , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Impedância Elétrica , Temperatura Alta , Humanos , Extratos Vegetais/farmacologia , Rutênio Vermelho/farmacologia , Canais de Cátion TRPV/antagonistas & inibidores , ortoaminobenzoatos/farmacologia
16.
PLoS Pathog ; 14(4): e1006981, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29630665

RESUMO

Purpura fulminans is a deadly complication of Neisseria meningitidis infections due to extensive thrombosis of microvessels. Although a Disseminated Intra-vascular Coagulation syndrome (DIC) is frequently observed during Gram negative sepsis, it is rarely associated with extensive thrombosis like those observed during meningococcemia, suggesting that the meningococcus induces a specific dysregulation of coagulation. Another specific feature of N. meningitidis pathogenesis is its ability to colonize microvessels endothelial cells via type IV pili. Importantly, endothelial cells are key in controlling the coagulation cascade through the activation of the potent anticoagulant Protein C (PC) thanks to two endothelial cell receptors among which the Endothelial Protein C Receptor (EPCR). Considering that congenital or acquired deficiencies of PC are associated with purpura fulminans, we hypothesized that a defect in the activation of PC following meningococcal adhesion to microvessels is responsible for the thrombotic events observed during meningococcemia. Here we showed that the adhesion of N. meningitidis on endothelial cells results in a rapid and intense decrease of EPCR expression by inducing its cleavage in a process know as shedding. Using siRNA experiments and CRISPR/Cas9 genome edition we identified ADAM10 (A Disintegrin And Metalloproteinase-10) as the protease responsible for this shedding. Surprisingly, ADAM17, the only EPCR sheddase described so far, was not involved in this process. Finally, we showed that this ADAM10-mediated shedding of EPCR induced by the meningococcal interaction with endothelial cells was responsible for an impaired activation of Protein C. This work unveils for the first time a direct link between meningococcal adhesion to endothelial cells and a severe dysregulation of coagulation, and potentially identifies new therapeutic targets for meningococcal purpura fulminans.


Assuntos
Proteína ADAM10/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Receptor de Proteína C Endotelial/metabolismo , Endotélio Vascular/patologia , Proteínas de Membrana/metabolismo , Infecções Meningocócicas/complicações , Microvasos/patologia , Proteína C/metabolismo , Púrpura Fulminante/etiologia , Proteína ADAM10/genética , Secretases da Proteína Precursora do Amiloide/genética , Aderência Bacteriana , Coagulação Sanguínea/fisiologia , Células Cultivadas , Receptor de Proteína C Endotelial/genética , Endotélio Vascular/metabolismo , Endotélio Vascular/microbiologia , Humanos , Proteínas de Membrana/genética , Infecções Meningocócicas/microbiologia , Microvasos/metabolismo , Microvasos/microbiologia , Neisseria meningitidis/fisiologia , Proteína C/genética , Púrpura Fulminante/metabolismo , Púrpura Fulminante/patologia
18.
Virulence ; 8(8): 1808-1819, 2017 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-29099305

RESUMO

Neisseria meningitidis is the causative agent of cerebrospinal meningitis and that of a rapidly progressing fatal septic shock known as purpura fulminans. Meningococcemia is characterized by bacterial adhesion to human endothelial cells of the microvessels. Host specificity has hampered studies on the role of blood vessels colonization in N. meningitidis associated pathogenesis. In this work, using a humanized model of SCID mice allowing the study of bacterial adhesion to human cells in an in vivo context we demonstrate that meningococcal colonization of human blood vessels is a prerequisite to the establishment of sepsis and lethality. To identify the molecular pathways involved in bacterial virulence, we performed transposon insertion site sequencing (Tn-seq) in vivo. Our results demonstrate that 36% of the genes that are important for growth in the blood of mice are dispensable when bacteria colonize human blood vessels, suggesting that human endothelial cells lining the blood vessels are feeding niches for N. meningitidis in vivo. Altogether, our work proposes a new paradigm for meningococcal virulence in which colonization of blood vessels is associated with metabolic adaptation and sustained bacteremia responsible for sepsis and subsequent lethality.


Assuntos
Bacteriemia/microbiologia , Infecções Meningocócicas/sangue , Infecções Meningocócicas/microbiologia , Microvasos/microbiologia , Neisseria meningitidis/fisiologia , Animais , Bacteriemia/sangue , Aderência Bacteriana , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Neisseria meningitidis/genética
19.
Nat Commun ; 8: 15764, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28569760

RESUMO

Neisseria meningitidis (meningococcus) is an invasive bacterial pathogen that colonizes human vessels, causing thrombotic lesions and meningitis. Establishment of tight interactions with endothelial cells is crucial for meningococci to resist haemodynamic forces. Two endothelial receptors, CD147 and the ß2-adrenergic receptor (ß2AR), are sequentially engaged by meningococci to adhere and promote signalling events leading to vascular colonization, but their spatiotemporal coordination is unknown. Here we report that CD147 and ß2AR form constitutive hetero-oligomeric complexes. The scaffolding protein α-actinin-4 directly binds to the cytosolic tail of CD147 and governs the assembly of CD147-ß2AR complexes in highly ordered clusters at bacterial adhesion sites. This multimolecular assembly process increases the binding strength of meningococci to endothelial cells under shear stress, and creates molecular platforms for the elongation of membrane protrusions surrounding adherent bacteria. Thus, the specific organization of cellular receptors has major impacts on host-pathogen interaction.


Assuntos
Actinina/metabolismo , Basigina/metabolismo , Interações Hospedeiro-Patógeno/fisiologia , Neisseria meningitidis/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Aderência Bacteriana/fisiologia , Basigina/genética , Células Endoteliais/metabolismo , Células Endoteliais/microbiologia , Humanos , Complexos Multiproteicos/metabolismo , Neisseria meningitidis/patogenicidade , Receptores Adrenérgicos beta 2/genética
20.
Nat Rev Microbiol ; 15(3): 149-159, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28090076

RESUMO

The blood-brain barrier, which is one of the tightest barriers in the body, protects the brain from insults, such as infections. Indeed, only a few of the numerous blood-borne bacteria can cross the blood-brain barrier to cause meningitis. In this Review, we focus on invasive extracellular pathogens, such as Neisseria meningitidis, Streptococcus pneumoniae, group B Streptococcus and Escherichia coli, to review the obstacles that bacteria have to overcome in order to invade the meninges from the bloodstream, and the specific skills they have developed to bypass the blood-brain barrier. The medical importance of understanding how these barriers can be circumvented is underlined by the fact that we need to improve drug delivery into the brain.


Assuntos
Barreira Hematoencefálica/microbiologia , Escherichia coli/patogenicidade , Meningites Bacterianas/patologia , Neisseria meningitidis/patogenicidade , Streptococcus agalactiae/patogenicidade , Streptococcus pneumoniae/patogenicidade , Sistemas de Liberação de Medicamentos , Humanos , Meninges/microbiologia , Meningites Bacterianas/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA