Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-38645070

RESUMO

Time-lapse microscopy has emerged as a crucial tool in cell biology, facilitating a deeper understanding of dynamic cellular processes. While existing tracking tools have proven effective in detecting and monitoring objects over time, the quantification of signals within these tracked objects often faces implementation constraints. In the context of infectious diseases, the quantification of signals at localized compartments within the cell and around intracellular pathogens can provide even deeper insight into the interactions between the pathogen and host cell organelles. Existing quantitative analysis at a single-phagosome level remains limited and dependent on manual tracking methods. We developed a near-fully automated workflow that performs with limited bias, high-throughput cell segmentation and quantitative tracking of both single cell and single bacterium/phagosome within multi-channel, z-stack, time-lapse confocal microscopy videos. We took advantage of the PyImageJ library to bring Fiji functionality into a Python environment and combined deep-learning-based segmentation from Cellpose with tracking algorithms from Trackmate. Our workflow provides a versatile toolkit of functions for measuring relevant signal parameters at the single-cell level (such as velocity or bacterial burden) and at the single-phagosome level (i.e. assessment of phagosome maturation over time). It's capabilities in both single-cell and single-phagosome quantification, its flexibility and open-source nature should assist studies that aim to decipher for example the pathogenicity of bacteria and the mechanism of virulence factors that could pave the way for the development of innovative therapeutic approaches.

2.
bioRxiv ; 2024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38496427

RESUMO

Type I Interferons (IFNs) generally have a protective role during viral infections, but their function during bacterial infections is dependent on the bacterial species. Legionella pneumophila, Shigella sonnei and Mycobacterium tuberculosis can inhibit type I IFN signaling. Here we examined the role of type I IFN, specifically IFNß, in the context of Salmonella enterica serovar Typhimurium (STm) macrophage infections and the capacity of STm to inhibit type I IFN signaling. We demonstrate that IFNß has no effect on the intracellular growth of STm in infected bone marrow derived macrophages (BMDMs) derived from C57BL/6 mice. STm infection inhibits IFNß signaling but not IFNγ signaling in a murine macrophage cell line. We show that this inhibition is independent of the type III and type VI secretion systems expressed by STm and is also independent of bacterial phagocytosis. The inhibition is Toll-like receptor 4 (TLR4)-dependent as the TLR4 ligand, lipopolysaccharide (LPS), alone is sufficient to inhibit IFNß-mediated signaling and STm-infected, TLR4-deficient BMDMs do not exhibit inhibited IFNß signaling. In summary, we show that macrophages exposed to STm have reduced IFNß signaling via crosstalk with TLR4 signaling, and that IFNß signaling does not affect cell autonomous host defense against STm.

3.
Biol Open ; 13(1)2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-38156988

RESUMO

Accurate quantification of bacterial burden within macrophages, termed bacterial burden quantification (BBQ), is crucial for understanding host-pathogen interactions. Various methods have been employed, each with strengths and weaknesses. This article addresses limitations in existing techniques and introduces two novel, automated methods for BBQ within macrophages based on confocal microscopy data analysis. The first method refines total fluorescence quantification by incorporating filtering steps to exclude uninfected cells, while the second method calculates total bacterial volume per cell to mitigate potential biases in fluorescence-based readouts. These workflows utilize PyImageJ and Cellpose software, providing reliable, unbiased, and rapid quantification of bacterial load. The proposed workflows were validated using Salmonella enterica serovar Typhimurium and Mycobacterium tuberculosis models, demonstrating their effectiveness in accurately assessing bacterial burden. These automated workflows offer valuable tools for studying bacterial interactions within host cells and provide insights for various research applications.


Assuntos
Macrófagos , Salmonella typhimurium , Fluxo de Trabalho , Interações Hospedeiro-Patógeno
4.
bioRxiv ; 2023 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-37873092

RESUMO

Accurate quantification of bacterial burden within macrophages, termed Bacterial Burden Quantification (BBQ), is crucial for understanding host-pathogen interactions. Various methods have been employed, each with strengths and weaknesses. This article addresses limitations in existing techniques and introduces two novel automated methods for BBQ within macrophages based on confocal microscopy data analysis. The first method refines total fluorescence quantification by incorporating filtering steps to exclude uninfected cells, while the second method calculates total bacterial volume per cell to mitigate potential biases in fluorescence-based readouts. These workflows utilize PyImageJ and Cellpose software, providing reliable, unbiased, and rapid quantification of bacterial load. The proposed workflows were validated using Salmonella enterica serovar Typhimurium and Mycobacterium tuberculosis models, demonstrating their effectiveness in accurately assessing bacterial burden. These automated workflows offer valuable tools for studying bacterial interactions within host cells and provide insights for various research applications.

5.
Infect Immun ; 91(11): e0036523, 2023 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-37843413

RESUMO

The host type I interferon (IFN) response protects against Legionella pneumophila infections. Other bacterial pathogens inhibit type I IFN-mediated cell signaling; however, the interaction between this signaling pathway and L. pneumophila has not been well described. Here, we demonstrate that L. pneumophila inhibits the IFN-ß signaling pathway but does not inhibit IFN-γ-mediated cell signaling. The addition of IFN-ß to L. pneumophila-infected macrophages limited bacterial growth independently of NOS2 and reactive nitrogen species. The type IV secretion system of L. pneumophila is required to inhibit IFN-ß-mediated cell signaling. Finally, we show that the inhibition of the IFN-ß signaling pathway occurs downstream of STAT1 and STAT2 phosphorylation. In conclusion, our findings describe a novel host cell signaling pathway inhibited by L. pneumophila via its type IV secretion system.


Assuntos
Interferon Tipo I , Legionella pneumophila , Doença dos Legionários , Humanos , Legionella pneumophila/fisiologia , Sistemas de Secreção Tipo IV , Interferon gama/metabolismo , Transdução de Sinais
6.
Nucleic Acids Res ; 51(18): 9905-9919, 2023 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-37670559

RESUMO

Translational fidelity is critical for microbial fitness, survival and stress responses. Much remains unknown about the genetic and environmental control of translational fidelity and its single-cell heterogeneity. In this study, we used a high-throughput fluorescence-based assay to screen a knock-out library of Escherichia coli and identified over 20 genes critical for stop-codon readthrough. Most of these identified genes were not previously known to affect translational fidelity. Intriguingly, we show that several genes controlling metabolism, including cyaA and crp, enhance stop-codon readthrough. CyaA catalyzes the synthesis of cyclic adenosine monophosphate (cAMP). Combining RNA sequencing, metabolomics and biochemical analyses, we show that deleting cyaA impairs amino acid catabolism and production of ATP, thus repressing the transcription of rRNAs and tRNAs to decrease readthrough. Single-cell analyses further show that cAMP is a major driver of heterogeneity in stop-codon readthrough and rRNA expression. Our results highlight that carbon metabolism is tightly coupled with stop-codon readthrough.


Assuntos
Códon de Terminação , AMP Cíclico , Escherichia coli , Sequência de Bases , Códon de Terminação/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Biossíntese de Proteínas , RNA de Transferência/genética , RNA de Transferência/metabolismo
7.
Chem ; 9(4): 881-900, 2023 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-37346394

RESUMO

Pillar[6]MaxQ (P6AS) functions as an in vivo sequestration agent for methamphetamine and fentanyl. We use 1H NMR, isothermal titration calorimetry, and molecular modelling to deduce the geometry and strength of the P6AS•drug complexes. P6AS forms tight complexes with fentanyl (Kd=9.8 nM), PCP (17.1 nM), MDMA (25.5 nM), mephedrone (52.4 nM), and methamphetamine (101 nM). P6AS has good in vitro biocompatibility according to MTS metabolic, Adenylate Kinase cell death, and hERG ion channel inhibition assays, and the Ames fluctuation test. The no observed adverse effect level for P6AS is 45 mg/kg. The hyperlocomotion of mice treated with methamphetamine (0.5 mg/kg) can be ameliorated by treatment with P6AS (35.7 mg/kg) 5-minutes later, whereas the hyperlocomotion of mice treated with fentanyl (0.1 mg/kg) can be controlled by treatment with P6AS (5 mg/kg) up to 15-minutes later. P6AS has significant potential for development as a broad spectrum in vivo sequestration agent.

8.
Cell Host Microbe ; 30(12): 1650-1652, 2022 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-36521438

RESUMO

A recent study in Science found Mycobacterium tuberculosis inhibits pyroptosis of the host cell by secreting a phosphatase (PtpB). PtpB targets the plasma membrane to dephosphorylate PI4P and PI(4,5)P2, inhibiting recruitment of the pore-forming gasdermin D N-terminal fragment. Pyroptosis inhibition contributes to virulence, as ptpB-deficient Mtb is attenuated in mice.


Assuntos
Mycobacterium tuberculosis , Piroptose , Camundongos , Animais , Proteínas de Ligação a Fosfato/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Membrana Celular/metabolismo , Mycobacterium tuberculosis/metabolismo
9.
Front Immunol ; 13: 791136, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35237260

RESUMO

The inflammasome complex is important for host defense against intracellular bacterial infections. Mycobacterium tuberculosis (Mtb) is a facultative intracellular bacterium which is able to survive in infected macrophages. Here we discuss how the host cell inflammasomes sense Mtb and other related mycobacterial species. Furthermore, we describe the molecular mechanisms of NLRP3 inflammasome sensing of Mtb which involve the type VII secretion system ESX-1, cell surface lipids (TDM/TDB), secreted effector proteins (LpqH, PPE13, EST12, EsxA) and double-stranded RNA acting on the priming and/or activation steps of inflammasome activation. In contrast, Mtb also mediates inhibition of the NLRP3 inflammasome by limiting exposure of cell surface ligands via its hydrolase, Hip1, by inhibiting the host cell cathepsin G protease via the secreted Mtb effector Rv3364c and finally, by limiting intracellular triggers (K+ and Cl- efflux and cytosolic reactive oxygen species production) via its serine/threonine kinase PknF. In addition, Mtb inhibits the AIM2 inflammasome activation via an unknown mechanism. Overall, there is good evidence for a tug-of-war between Mtb trying to limit inflammasome activation and the host cell trying to sense Mtb and activate the inflammasome. The detailed molecular mechanisms and the importance of inflammasome activation for virulence of Mtb or host susceptibility have not been fully investigated.


Assuntos
Infecções por Mycobacterium , Mycobacterium tuberculosis , Humanos , Inflamassomos/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Infecções por Mycobacterium/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo
10.
ChemMedChem ; 17(10): e202200046, 2022 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-35238177

RESUMO

We report studies of the interaction of six acyclic CB[n]-type receptors toward a panel of drugs of abuse by a combination of isothermal titration calorimetry and 1 H NMR spectroscopy. Anthracene walled acyclic CB[n] host (M3) displays highest binding affinity toward methamphetamine (Kd =15 nM) and fentanyl (Kd =4 nM). Host M3 is well tolerated by Hep G2 and HEK 293 cells up to 100 µM according to MTS metabolic and adenylate kinase release assays. An in vivo maximum tolerated dose study with Swiss Webster mice showed no adverse effects at the highest dose studied (44.7 mg kg-1 ). Host M3 is not mutagenic based on the Ames fluctuation test and does not inhibit the hERG ion channel. In vivo efficacy studies showed that pretreatment of mice with M3 significantly reduces the hyperlocomotion after treatment with methamphetamine, but M3 does not function similarly when administered 30 seconds after methamphetamine.


Assuntos
Metanfetamina , Animais , Antracenos/farmacologia , Células HEK293 , Humanos , Dose Máxima Tolerável , Metanfetamina/farmacologia , Camundongos
11.
Chemistry ; 27(69): 17476-17486, 2021 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-34613641

RESUMO

We report the synthesis of two new acyclic sulfated acyclic CB[n]-type receptors (TriM0 and Me4 TetM0) and investigations of their binding properties toward a panel of drugs of abuse (1-13) by a combination of 1 H NMR spectroscopy and isothermal titration calorimetry. TetM0 is the most potent receptor with Ka ≥106  M-1 toward methamphetamine, fentanyl, MDMA and mephedrone. TetM0 is not cytotoxic toward HepG2 and HEK 293 cells below 100 µM according to MTS metabolic and adenylate kinase release assays and is well tolerated in vivo when dosed at 46 mg kg-1 . TetM0 does not inhibit the hERG ion channel and is not mutagenic based on the Ames fluctuation test. Finally, in vivo efficacy studies show that the hyperlocomotion of mice treated with methamphetamine can be greatly reduced by treatment with TetM0 up to 5 minutes later. TetM0 has potential as a broad spectrum in vivo sequestrant for drugs of abuse.


Assuntos
Metanfetamina , Sulfatos , Animais , Células HEK293 , Humanos , Metanfetamina/toxicidade , Camundongos
12.
PLoS Pathog ; 17(7): e1009712, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34324582

RESUMO

Mycobacterium tuberculosis (Mtb) has evolved to evade host innate immunity by interfering with macrophage functions. Interleukin-1ß (IL-1ß) is secreted by macrophages after the activation of the inflammasome complex and is crucial for host defense against Mtb infections. We have previously shown that Mtb is able to inhibit activation of the AIM2 inflammasome and subsequent pyroptosis. Here we show that Mtb is also able to inhibit host cell NLRP3 inflammasome activation and pyroptosis. We identified the serine/threonine kinase PknF as one protein of Mtb involved in the NLRP3 inflammasome inhibition, since the pknF deletion mutant of Mtb induces increased production of IL-1ß in bone marrow-derived macrophages (BMDMs). The increased production of IL-1ß was dependent on NLRP3, the adaptor protein ASC and the protease caspase-1, as revealed by studies performed in gene-deficient BMDMs. Additionally, infection of BMDMs with the pknF deletion mutant resulted in increased pyroptosis, while the IL-6 production remained unchanged compared to Mtb-infected cells, suggesting that the mutant did not affect the priming step of inflammasome activation. In contrast, the activation step was affected since potassium efflux, chloride efflux and the generation of reactive oxygen species played a significant role in inflammasome activation and subsequent pyroptosis mediated by the Mtb pknF mutant strain. In conclusion, we reveal here that the serine/threonine kinase PknF of Mtb plays an important role in innate immune evasion through inhibition of the NLRP3 inflammasome.


Assuntos
Evasão da Resposta Imune/imunologia , Inflamassomos/imunologia , Mycobacterium tuberculosis/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Proteínas Serina-Treonina Quinases/imunologia , Tuberculose/imunologia , Animais , Interações Hospedeiro-Patógeno/imunologia , Camundongos , Mycobacterium tuberculosis/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Tuberculose/metabolismo
13.
Chemistry ; 27(9): 3098-3105, 2021 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-33206421

RESUMO

We report investigations of the use of cucurbit[8]uril (CB[8]) macrocycles as an antidote to counteract the in vivo biological effects of phencyclidine. We investigate the binding of CB[8] and its derivative Me4 CB[8] toward ten drugs of abuse (3-9, 12-14) by a combination of 1 H NMR spectroscopy and isothermal titration calorimetry in phosphate buffered water. We find that the cavity of CB[8] and Me4 CB[8] are able to encapsulate the 1-amino-1-aryl-cyclohexane ring system of phencyclidine (PCP) and ketamine as well as the morphinan skeleton of morphine and hydromorphone with Kd values ≤50 nm. In vitro cytotoxicity (MTS metabolic and adenylate kinase cell death assays in HEK293 and HEPG2 cells) and in vivo maximum tolerated dose studies (Swiss Webster mice) which were performed for Me4 CB[8] indicated good tolerability. The tightest host⋅guest pair (Me4 CB[8]⋅PCP; Kd =2 nm) was advanced to in vivo efficacy studies. The results of open field tests demonstrate that pretreatment of mice with Me4 CB[8] prevents subsequent hyperlocomotion induction by PCP and also that treatment of animals previously dosed with PCP with Me4 CB[8] significantly reduces the locomotion levels.


Assuntos
Hidrocarbonetos Aromáticos com Pontes/química , Imidazóis/química , Fenciclidina/análise , Fenciclidina/química , Animais , Hidrocarbonetos Aromáticos com Pontes/administração & dosagem , Hidrocarbonetos Aromáticos com Pontes/farmacologia , Células HEK293 , Células Hep G2 , Humanos , Imidazóis/administração & dosagem , Imidazóis/farmacologia , Locomoção/efeitos dos fármacos , Camundongos , Fenciclidina/administração & dosagem , Fenciclidina/farmacologia
14.
Front Cell Infect Microbiol ; 10: 595029, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33194845

RESUMO

Mycobacterium tuberculosis (Mtb) is a very successful pathogen, strictly adapted to humans and the cause of tuberculosis. Its success is associated with its ability to inhibit host cell intrinsic immune responses by using an arsenal of virulence factors of different nature. It has evolved to synthesize a series of complex lipids which form an outer membrane and may also be released to enter host cell membranes. In addition, secreted protein effectors of Mtb are entering the host cell cytosol to interact with host cell proteins. We briefly discuss the current model, involving the ESX-1 type seven secretion system and the Mtb lipid phthiocerol dimycoserosate (PDIM), of how Mtb creates pores in the phagosomal membrane to allow Mtb proteins to access to the host cell cytosol. We provide an exhaustive list of Mtb secreted proteins that have effector functions. They modify (mostly inhibit but sometimes activate) host cell pathways such as: phagosome maturation, cell death, cytokine response, xenophagy, reactive oxygen species (ROS) response via NADPH oxidase 2 (NOX2), nitric oxide (NO) response via NO Synthase 2 (NOS2) and antigen presentation via MHC class I and class II molecules. We discuss the host cell targets for each lipid and protein effector and the importance of the Mtb effector for virulence of the bacterium.


Assuntos
Mycobacterium tuberculosis , Proteínas de Bactérias , Humanos , Lipídeos , Macrófagos , Fagossomos
15.
Curr Opin Immunol ; 60: 103-110, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31228759

RESUMO

Regulated cell death via apoptosis not only is important for organismal homeostasis but also serves as an innate defense mechanism. The engulfment of apoptotic infected cells, a process known as efferocytosis, is a common pathway for the destruction of many intracellular bacteria. Some pathogens take advantage of efferocytosis to prevent activation of macrophages and thereby facilitate their dissemination. Conversely, many obligate intracellular bacterial pathogens and some facultative-intracellular bacteria inhibit apoptosis, preventing efferocytosis, and evading innate host defenses. The molecular mechanism of bacterial effectors includes secreted proteins that bind to and inhibit apoptosis cell signaling pathways. We provide an overview of the known bacterial effectors, their host cell targets and their importance for the virulence of human pathogens.


Assuntos
Apoptose , Infecções Bacterianas/imunologia , Infecções Bacterianas/microbiologia , Fenômenos Fisiológicos Bacterianos , Interações Hospedeiro-Patógeno/imunologia , Imunidade , Animais , Apoptose/imunologia , Infecções Bacterianas/metabolismo , Biomarcadores , Humanos , Imunidade Inata , Espaço Intracelular/imunologia , Espaço Intracelular/metabolismo , Espaço Intracelular/microbiologia , Fagocitose/imunologia , Transdução de Sinais
16.
J Immunol ; 202(8): 2348-2359, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30833347

RESUMO

The type I IFNs (IFN-α and -ß) are important for host defense against viral infections. In contrast, their role in defense against nonviral pathogens is more ambiguous. In this article, we report that IFN-ß signaling in murine bone marrow-derived macrophages has a cell-intrinsic protective capacity against Mycobacterium tuberculosis via the increased production of NO. The antimycobacterial effects of type I IFNs were mediated by direct signaling through the IFN-α/ß-receptor (IFNAR), as Ab-mediated blocking of IFNAR1 prevented the production of NO. Furthermore, M. tuberculosis is able to inhibit IFNAR-mediated cell signaling and the subsequent transcription of 309 IFN-ß-stimulated genes in a dose-dependent way. The molecular mechanism of inhibition by M. tuberculosis involves reduced phosphorylation of the IFNAR-associated protein kinases JAK1 and TYK2, leading to reduced phosphorylation of the downstream targets STAT1 and STAT2. Transwell experiments demonstrated that the M. tuberculosis-mediated inhibition of type I IFN signaling was restricted to infected cells. Overall, our study supports the novel concept that M. tuberculosis evolved to inhibit autocrine type I IFN signaling to evade host defense mechanisms.


Assuntos
Comunicação Autócrina/imunologia , Interferon Tipo I/imunologia , Viabilidade Microbiana/imunologia , Mycobacterium tuberculosis/imunologia , Transdução de Sinais/imunologia , Animais , Comunicação Autócrina/genética , Interferon Tipo I/genética , Janus Quinase 1/genética , Janus Quinase 1/imunologia , Camundongos , Camundongos Knockout , Viabilidade Microbiana/genética , Óxido Nítrico/genética , Óxido Nítrico/imunologia , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/imunologia , Transdução de Sinais/genética , TYK2 Quinase/genética , TYK2 Quinase/imunologia
17.
Helv Chim Acta ; 101(6)2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31231137

RESUMO

Mixed self-assembly of ligands 1 and 2, PXDA (3), and Pd(NO3)2 afforded metal organic polyhedra (MOP 1 - MOP 3) which bear 24 covalently attached CB[7] and cyclooctyne moieties. Post assembly modification (PAM) of MOP 3 by covalent strain promoted alkyne azide click reaction provided MOP 4 R bearing covalently attached functionality (PEG, sulfonate, biotin, c-RGD, fluorescein and cyanine). Orthogonal CB[7] guest mediated non-covalent PAM of MOP 4 R with Ad-FITC afforded MOP 5 RGD Ad-FITC and MOP 5 biotin 0020Ad-FITC. Flow cytometry analysis of the uptake of MOP 5 RGD Ad-FITC toward U87 cells demonstrated improved uptake relative to control MOP lacking c-RGD ligands. These results suggest a broad applicability of orthogonally functionalizable (covalent and non-covalent) MOPs in targeted drug delivery and imaging applications.

18.
J Am Chem Soc ; 139(26): 9066-9074, 2017 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-28621947

RESUMO

Mixed self-assembly of ligands 1, 2, 1,6-hexanediamine (HDA), and Pd(NO3)2 afforded Fujita-type metal organic polyhedron MOP1 (diameter ≈ 8.2 nm), which is covalently functionalized with an average of 18 cucurbit[7]uril (CB[7]) units, as evidenced by 1H NMR, diffusion-ordered spectroscopy NMR, and transmission electron microscopy measurements. By virtue of the host-guest properties of CB[7], the inner cavity of MOP can be rendered hydrophobic by using octadecyl HDA (3) as guest during the self-assembly process. The hydrophobic cavity was successfully utilized to trap the hydrophobic dye Nile Red (NR) and the anticancer drug doxorubicin (DOX). The stimuli-responsive release of encapsulated NR or DOX occurs (1) upon addition of a competitive binder (e.g., adamantane ammonium (ADA)) for CB[7], (2) by a dual pH-chemical stimulus involving the protonation state change of adamantane carboxylate at pH 5.8, and (3) by a dual pH-photochemical stimulus involving photoisomerization of trans-6 to cis-6 at pH 5.8. NR is released from NR@MOP2 within HeLa cancer cells. This body of work suggests that the covalent attachment of cucurbit[n]uril to metal organic polyhedra constitutes a promising vehicle for the development of both diagnostic and therapeutic nanoparticles.


Assuntos
Hidrocarbonetos Aromáticos com Pontes/química , Imidazóis/química , Estruturas Metalorgânicas/química , Interações Hidrofóbicas e Hidrofílicas , Ligantes , Estrutura Molecular , Fotoquímica
19.
mBio ; 8(2)2017 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-28270579

RESUMO

The cell wall of Mycobacterium tuberculosis is composed of unique lipids that are important for pathogenesis. Indeed, the first-ever genetic screen in M. tuberculosis identified genes involved in the biosynthesis and transport of the cell wall lipid PDIM (phthiocerol dimycocerosates) as crucial for the survival of M. tuberculosis in mice. Here we show evidence for a novel molecular mechanism of the PDIM-mediated virulence in M. tuberculosis We characterized the DNA interaction and the regulon of Rv3167c, a transcriptional repressor that is involved in virulence regulation of M. tuberculosis, and discovered that it controls the PDIM operon. A loss-of-function genetic approach showed that PDIM levels directly correlate with the capacity of M. tuberculosis to escape the phagosome and induce host cell necrosis and macroautophagy. In conclusion, our study attributes a novel role of the cell wall lipid PDIM in intracellular host cell modulation, which is important for host cell exit and dissemination of M. tuberculosisIMPORTANCEMycobacterium tuberculosis is a major human pathogen that has coevolved with its host for thousands of years. The complex and unique cell wall of M. tuberculosis contains the lipid PDIM (phthiocerol dimycocerosates), which is crucial for virulence of the bacterium, but its function is not well understood. Here we show that PDIM expression by M. tuberculosis is negatively regulated by a novel transcriptional repressor, Rv3167c. In addition, we discovered that the escape of M. tuberculosis from its intracellular vacuole was greatly augmented by the presence of PDIM. The increased release of M. tuberculosis into the cytosol led to increased host cell necrosis. The discovery of a link between the cell wall lipid PDIM and a major pathogenesis pathway of M. tuberculosis provides important insights into the molecular mechanisms of host cell manipulation by M. tuberculosis.


Assuntos
Parede Celular/química , Exocitose , Glicolipídeos/metabolismo , Interações Hospedeiro-Patógeno , Mycobacterium tuberculosis/fisiologia , Fagossomos/microbiologia , Fatores de Virulência/metabolismo , Autofagia , Morte Celular , Linhagem Celular , Deleção de Genes , Humanos , Macrófagos/microbiologia , Macrófagos/fisiologia , Óperon , Proteínas Repressoras/genética , Transcrição Gênica , Virulência
20.
J Am Chem Soc ; 138(43): 14488-14496, 2016 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-27723965

RESUMO

Self-assembly of ligand 1 and Pd(NO3)2 delivers Fujita-type metal-organic polyhedron (MOP) 3 which bears 24 covalently attached methyl viologen units on its external surface, as evidenced by 1H NMR, diffusion-ordered spectroscopy NMR, electrospray mass spectrometry, transmission electron microscopy, and atomic force microscopy measurements. MOP 3 undergoes noncovalent complexation with cucurbit[n]urils to yield MOPs 4-6 with diameter ≈5-6 nm. MOP 5 can be fully loaded with doxorubicin (DOX) prodrug 2 via hetero-ternary complex formation to yield 7. The MOPs exhibit excellent stability toward neutral to slightly acidic pH in 10 mM sodium phosphate buffer, mitigating the concern of disassembly during circulation. The results of MTS assays show that MOP 7 is 10-fold more cytotoxic toward HeLa cells than equimolar quantities of DOX prodrug 2. The enhanced cytotoxicity can be traced to a combination of enhanced cellular uptake of 7 and DOX release as demonstrated by flow cytometry and confocal fluorescence microscopy. The confluence of properties imparted by the polycationic MOP architecture and plug-and-play CB[n] complexation provides a potent new platform for drug delivery application.


Assuntos
Hidrocarbonetos Aromáticos com Pontes/química , Doxorrubicina/química , Portadores de Fármacos/química , Imidazóis/química , Compostos Organometálicos/química , Doxorrubicina/metabolismo , Doxorrubicina/farmacologia , Células HeLa , Humanos , Concentração de Íons de Hidrogênio , Modelos Moleculares , Conformação Molecular , Pró-Fármacos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...