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








Base de dados
Intervalo de ano de publicação
1.
NPJ Vaccines ; 9(1): 102, 2024 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-38858404

RESUMO

We recently developed an immuno-oncotherapy against human papillomavirus (HPV)-induced tumors based on a lentiviral vector encoding the Early E6 and E7 oncoproteins of HPV16 and HPV18 genotypes, namely "Lenti-HPV-07". The robust and long-lasting anti-tumor efficacy of Lenti-HPV-07 is dependent on CD8+ T-cell induction and remodeling of the tumor microenvironment. Here, we first established that anti-vector immunity induced by Lenti-HPV-07 prime has no impact on the efficacy of a homologous boost to amplify anti-HPV T-cell immunity. To longitudinally monitor the evolution of the T-cell repertoire generated after the prime, homologous or heterologous boost with Lenti-HPV-07, we tracked T-cell clonotypes by deep sequencing of T-Cell Receptor (TCR) variable ß and α chain mRNA, applied to whole peripheral blood cells (PBL) and a T cell population specific of an immunodominant E7HPV16 epitope. We observed a hyper-expansion of clonotypes post prime, accompanied by increased frequencies of HPV-07-specific T cells. Additionally, there was a notable diversification of clonotypes post boost in whole PBL, but not in the E7HPV16-specific T cells. We then demonstrated that the effector functions of such Lenti-HPV-07-induced T cells synergize with anti-checkpoint inhibitory treatments by systemic administration of anti-TIM3 or anti-NKG2A monoclonal antibodies. While Lenti-HPV-07 is about to enter a Phase I/IIa clinical trial, these results will help better elucidate its mode of action in immunotherapy against established HPV-mediated malignancies.

2.
EMBO Mol Med ; 15(10): e17723, 2023 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-37675835

RESUMO

Human papillomavirus (HPV) infections are the cause of all cervical and numerous oropharyngeal and anogenital cancers. The currently available HPV vaccines, which induce neutralizing antibodies, have no therapeutic effect on established tumors. Here, we developed an immuno-oncotherapy against HPV-induced tumors based on a non-integrative lentiviral vector encoding detoxified forms of the Early E6 and E7 oncoproteins of HPV16 and 18 genotypes, namely, "Lenti-HPV-07". A single intramuscular injection of Lenti-HPV-07 into mice bearing established HPV-induced tumors resulted in complete tumor eradication in 100% of the animals and was also effective against lung metastases. This effect correlated with CD8+ T-cell induction and profound remodeling of the tumor microenvironment. In the intra-tumoral infiltrates of vaccinated mice, the presence of large amounts of activated effector, resident memory, and transcription factor T cell factor-1 (TCF-1)+ "stem-like" CD8+ T cells was associated with full tumor eradication. The Lenti-HPV-07-induced immunity was long-lasting and prevented tumor growth after a late re-challenge, mimicking tumor relapse. Lenti-HPV-07 therapy synergizes with an anti-checkpoint inhibitory treatment and therefore shows promise as an immuno-oncotherapy against established HPV-mediated malignancies.

3.
Microbes Infect ; 25(7): 105142, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37080384

RESUMO

Human Angiotensin-Converting Enzyme 2 (hACE2) is the major receptor enabling host cell invasion by SARS-CoV-2 via interaction with Spike. The murine ACE2 does not interact efficiently with SARS-CoV-2 Spike and therefore the laboratory mouse strains are not permissive to SARS-CoV-2 replication. Here, we generated new hACE2 transgenic mice, which harbor the hACE2 gene under the human keratin 18 promoter, in "HHD-DR1" background. HHD-DR1 mice are fully devoid of murine Major Histocompatibility Complex (MHC) molecules of class-I and -II and express only MHC molecules from Human Leukocyte Antigen (HLA) HLA 02.01, DRA01.01, DRB1.01.01 alleles, widely expressed in human populations. We selected three transgenic strains, with various hACE2 mRNA expression levels and distinctive profiles of lung and/or brain permissiveness to SARS-CoV-2 replication. These new hACE2 transgenic strains display high permissiveness to the replication of SARS-CoV-2 Omicron sub-variants, while the previously available B6.K18-ACE22Prlmn/JAX mice have been reported to be poorly susceptible to infection with Omicron. As a first application, one of these MHC- and ACE2-humanized strains was successfully used to show the efficacy of a lentiviral-based COVID-19 vaccine.


Assuntos
Enzima de Conversão de Angiotensina 2 , COVID-19 , Animais , Camundongos , Humanos , Enzima de Conversão de Angiotensina 2/genética , SARS-CoV-2/genética , Vacinas contra COVID-19 , Permissividade , Complexo Principal de Histocompatibilidade , Camundongos Transgênicos
4.
Mol Ther ; 30(9): 2984-2997, 2022 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-35484842

RESUMO

As the coronavirus disease 2019 (COVID-19) pandemic continues and new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern emerge, the adaptive immunity initially induced by the first-generation COVID-19 vaccines starts waning and needs to be strengthened and broadened in specificity. Vaccination by the nasal route induces mucosal, humoral, and cellular immunity at the entry point of SARS-CoV-2 into the host organism and has been shown to be the most effective for reducing viral transmission. The lentiviral vaccination vector (LV) is particularly suitable for this route of immunization owing to its non-cytopathic, non-replicative, and scarcely inflammatory properties. Here, to set up an optimized cross-protective intranasal booster against COVID-19, we generated an LV encoding stabilized spike of SARS-CoV-2 Beta variant (LV::SBeta-2P). mRNA vaccine-primed and -boosted mice, with waning primary humoral immunity at 4 months after vaccination, were boosted intranasally with LV::SBeta-2P. A strong boost effect was detected on cross-sero-neutralizing activity and systemic T cell immunity. In addition, mucosal anti-spike IgG and IgA, lung-resident B cells, and effector memory and resident T cells were efficiently induced, correlating with complete pulmonary protection against the SARS-CoV-2 Delta variant, demonstrating the suitability of the LV::SBeta-2P vaccine candidate as an intranasal booster against COVID-19. LV::SBeta-2P vaccination was also fully protective against Omicron infection of the lungs and central nervous system, in the highly susceptible B6.K18-hACE2IP-THV transgenic mice.


Assuntos
COVID-19 , Vacinas Virais , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais , COVID-19/prevenção & controle , Vacinas contra COVID-19 , Humanos , Pulmão , Camundongos , Mucosa , SARS-CoV-2/genética , Vacinação , Vacinas Sintéticas , Vacinas de mRNA
5.
Vaccines (Basel) ; 11(1)2022 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-36679857

RESUMO

Following the breakthrough of numerous severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants in recent months and the incomplete efficiency of the currently available vaccines, development of more effective vaccines is desirable. Non-integrative, non-cytopathic and non-inflammatory lentiviral vectors elicit sterilizing prophylaxis against SARS-CoV-2 in preclinical animal models and are particularly suitable for mucosal vaccination, which is acknowledged as the most effective in reducing viral transmission. Here, we demonstrate that a single intranasal administration of a vaccinal lentiviral vector encoding a stabilized form of the original SARS-CoV-2 Spike glycoprotein induces full-lung protection of respiratory tracts and strongly reduces pulmonary inflammation in the susceptible Syrian golden hamster model against the prototype SARS-CoV-2. In addition, we show that a lentiviral vector encoding stabilized Spike of SARS-CoV-2 Beta variant (LV::SBeta-2P) prevents pathology and reduces infectious viral loads in lungs and nasal turbinates following inoculation with the SARS-CoV-2 Omicron variant. Importantly, an intranasal boost with LV::SBeta-2P improves cross-seroneutralization much better in LV::SBeta-2P-primed hamsters than in their counterparts primed with an LV-encoding Spike from the ancestral SARS-CoV-2. These results strongly suggest that an immune imprint with the original Spike sequence has a negative impact on cross-protection against new variants. Our results tackle the issue of vaccine effectiveness in people who have already been vaccinated and have vanished immunity and indicate the efficiency of LV-based intranasal vaccination, either as a single dose or as booster.

6.
Microb Cell Fact ; 19(1): 178, 2020 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-32894164

RESUMO

BACKGROUND: Cyclodipeptide oxidases (CDOs) are enzymes involved in the biosynthesis of 2,5-diketopiperazines, a class of naturally occurring compounds with a large range of pharmaceutical activities. CDOs belong to cyclodipeptide synthase (CDPS)-dependent pathways, in which they play an early role in the chemical diversification of cyclodipeptides by introducing Cα-Cß dehydrogenations. Although the activities of more than 100 CDPSs have been determined, the activities of only a few CDOs have been characterized. Furthermore, the assessment of the CDO activities on chemically-synthesized cyclodipeptides has shown these enzymes to be relatively promiscuous, making them interesting tools for cyclodipeptide chemical diversification. The purpose of this study is to provide the first completely microbial toolkit for the efficient bioproduction of a variety of dehydrogenated 2,5-diketopiperazines. RESULTS: We mined genomes for CDOs encoded in biosynthetic gene clusters of CDPS-dependent pathways and selected several for characterization. We co-expressed each with their associated CDPS in the pathway using Escherichia coli as a chassis and showed that the cyclodipeptides and the dehydrogenated derivatives were produced in the culture supernatants. We determined the biological activities of the six novel CDOs by solving the chemical structures of the biologically produced dehydrogenated cyclodipeptides. Then, we assessed the six novel CDOs plus two previously characterized CDOs in combinatorial engineering experiments in E. coli. We co-expressed each of the eight CDOs with each of 18 CDPSs selected for the diversity of cyclodipeptides they synthesize. We detected more than 50 dehydrogenated cyclodipeptides and determined the best CDPS/CDO combinations to optimize the production of 23. CONCLUSIONS: Our study establishes the usefulness of CDPS and CDO for the bioproduction of dehydrogenated cyclodipeptides. It constitutes the first step toward the bioproduction of more complex and diverse 2,5-diketopiperazines.


Assuntos
Biotecnologia/métodos , Dicetopiperazinas/metabolismo , Escherichia coli/enzimologia , Oxirredutases/metabolismo , Peptídeo Sintases/metabolismo , Vias Biossintéticas/genética , Dicetopiperazinas/química , Escherichia coli/genética , Oxirredutases/genética , Peptídeo Sintases/genética , Filogenia
7.
Sci Rep ; 9(1): 15009, 2019 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-31611595

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

8.
Sci Rep ; 9(1): 9208, 2019 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-31239480

RESUMO

Prenylated indole diketopiperazine (DKP) alkaloids are important bioactive molecules or their precursors. In the context of synthetic biology, efficient means for their biological production would increase their chemical diversification and the discovery of novel bioactive compounds. Here, we prove the suitability of the Escherichia coli chassis for the production of prenylated indole DKP alkaloids. We used enzyme combinations not found in nature by co-expressing bacterial cyclodipeptide synthases (CDPSs) that assemble the DKP ring and fungal prenyltransferases (PTs) that transfer the allylic moiety from the dimethylallyl diphosphate (DMAPP) to the indole ring of tryptophanyl-containing cyclodipeptides. Of the 11 tested combinations, seven resulted in the production of eight different prenylated indole DKP alkaloids as determined by LC-MS/MS and NMR characterization. Two were previously undescribed. Engineering E. coli by introducing a hybrid mevalonate pathway for increasing intracellular DMAPP levels improved prenylated indole DKP alkaloid production. Purified product yields of 2-26 mg/L per culture were obtained from culture supernatants. Our study paves the way for the bioproduction of novel prenylated indole DKP alkaloids in a tractable chassis that can exploit the cyclodipeptide diversity achievable with CDPSs and the numerous described PT activities.


Assuntos
Dicetopiperazinas/química , Escherichia coli/genética , Escherichia coli/metabolismo , Engenharia Genética , Alcaloides Indólicos/química , Alcaloides Indólicos/metabolismo , Prenilação
9.
Cell Rep ; 23(4): 1072-1084, 2018 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-29694886

RESUMO

The pathogenic potential of Mycobacterium tuberculosis largely depends on ESX secretion systems exporting members of the multigenic Esx, Esp, and PE/PPE protein families. To study the secretion and regulation patterns of these proteins while circumventing immune cross-reactions due to their extensive sequence homologies, we developed an approach that relies on the recognition of their MHC class II epitopes by highly discriminative T cell receptors (TCRs) of a panel of T cell hybridomas. The latter were engineered so that each expresses a unique fluorescent reporter linked to specific antigen recognition. The resulting polychromatic and multiplexed imaging assay enabled us to measure the secretion of mycobacterial effectors inside infected host cells. We applied this novel technology to a large panel of mutants, clinical isolates, and host-cell types to explore the host-mycobacteria interplay and its impact on the intracellular bacterial secretome, which also revealed the unexpected capacity of phagocytes from lung granuloma to present mycobacterial antigens via MHC class II.


Assuntos
Sistemas de Secreção Bacterianos/imunologia , Epitopos de Linfócito T/imunologia , Granuloma do Sistema Respiratório , Mycobacterium tuberculosis/imunologia , Fagócitos , Tuberculose Pulmonar , Animais , Linhagem Celular Tumoral , Granuloma do Sistema Respiratório/imunologia , Granuloma do Sistema Respiratório/microbiologia , Granuloma do Sistema Respiratório/patologia , Antígenos de Histocompatibilidade Classe II/imunologia , Camundongos , Fagócitos/imunologia , Fagócitos/microbiologia , Fagócitos/patologia , Tuberculose Pulmonar/imunologia , Tuberculose Pulmonar/patologia
10.
Nat Microbiol ; 3(2): 181-188, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29335553

RESUMO

Mycobacterium tuberculosis requires a large number of secreted and exported proteins for its virulence, immune modulation and nutrient uptake. Most of these proteins are transported by the different type VII secretion systems1,2. The most recently evolved type VII secretion system, ESX-5, secretes dozens of substrates belonging to the PE and PPE families, which are named for conserved proline and glutamic acid residues close to the amino terminus3,4. However, the role of these proteins remains largely elusive 1 . Here, we show that mutations of ppe38 completely block the secretion of two large subsets of ESX-5 substrates, that is, PPE-MPTR and PE_PGRS, together comprising >80 proteins. Importantly, hypervirulent clinical M. tuberculosis strains of the Beijing lineage have such a mutation and a concomitant loss of secretion 5 . Restoration of PPE38-dependent secretion partially reverted the hypervirulence phenotype of a Beijing strain, and deletion of ppe38 in moderately virulent M. tuberculosis increased virulence. This indicates that these ESX-5 substrates have an important role in virulence attenuation. Phylogenetic analysis revealed that deletion of ppe38 occurred at the branching point of the 'modern' Beijing sublineage and is shared by Beijing outbreak strains worldwide, suggesting that this deletion may have contributed to their success and global distribution6,7.


Assuntos
Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/metabolismo , Animais , Antígenos de Bactérias/genética , Pequim , DNA Bacteriano/genética , Modelos Animais de Doenças , Deleção de Genes , Humanos , Camundongos Endogâmicos BALB C , Mutação , Mycobacterium tuberculosis/patogenicidade , Fenótipo , Filogenia , Tuberculose Pulmonar/microbiologia , Sistemas de Secreção Tipo VII , Virulência/genética , Fatores de Virulência
11.
PLoS Pathog ; 13(5): e1006399, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28545104

RESUMO

Sensing and response to changes in nutrient availability are essential for the lifestyle of environmental and pathogenic bacteria. Serine/threonine protein kinase G (PknG) is required for virulence of the human pathogen Mycobacterium tuberculosis, and its putative substrate GarA regulates the tricarboxylic acid cycle in M. tuberculosis and other Actinobacteria by protein-protein binding. We sought to understand the stimuli that lead to phosphorylation of GarA, and the roles of this regulatory system in pathogenic and non-pathogenic bacteria. We discovered that M. tuberculosis lacking garA was severely attenuated in mice and macrophages and furthermore that GarA lacking phosphorylation sites failed to restore the growth of garA deficient M. tuberculosis in macrophages. Additionally we examined the impact of genetic disruption of pknG or garA upon protein phosphorylation, nutrient utilization and the intracellular metabolome. We found that phosphorylation of GarA requires PknG and depends on nutrient availability, with glutamate and aspartate being the main stimuli. Disruption of pknG or garA caused opposing effects on metabolism: a defect in glutamate catabolism or depletion of intracellular glutamate, respectively. Strikingly, disruption of the phosphorylation sites of GarA was sufficient to recapitulate defects caused by pknG deletion. The results suggest that GarA is a cellular target of PknG and the metabolomics data demonstrate that the function of this signaling system is in metabolic regulation. This function in amino acid homeostasis is conserved amongst the Actinobacteria and provides an example of the close relationship between metabolism and virulence.


Assuntos
Aminoácidos/metabolismo , Proteínas de Bactérias/metabolismo , Metabolômica , Mycobacterium tuberculosis , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Ácido Aspártico/metabolismo , Proteínas de Bactérias/genética , Ácido Glutâmico/metabolismo , Homeostase , Macrófagos/microbiologia , Camundongos , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/patogenicidade , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Tuberculose/microbiologia , Virulência
12.
Cell Microbiol ; 19(7)2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28095608

RESUMO

Although phthiocerol dimycocerosates (DIM) are major virulence factors of Mycobacterium tuberculosis (Mtb), the causative agent of human tuberculosis, little is known about their mechanism of action. Localized in the outer membrane of mycobacterial pathogens, DIM are predicted to interact with host cell membranes. Interaction with eukaryotic membranes is a property shared with another virulence factor of Mtb, the early secretory antigenic target EsxA (also known as ESAT-6). This small protein, which is secreted by the type VII secretion system ESX-1 (T7SS/ESX-1), is involved in phagosomal rupture and cell death induced by virulent mycobacteria inside host phagocytes. In this work, by the use of several knock-out or knock-in mutants of Mtb or Mycobacterium bovis BCG strains and different cell biological assays, we present conclusive evidence that ESX-1 and DIM act in concert to induce phagosomal membrane damage and rupture in infected macrophages, ultimately leading to host cell apoptosis. These results identify an as yet unknown function for DIM in the infection process and open up a new research field for the study of the interaction of lipid and protein virulence factors of Mtb.


Assuntos
Antígenos de Bactérias/metabolismo , Apoptose/fisiologia , Proteínas de Bactérias/metabolismo , Lipídeos/fisiologia , Macrófagos/metabolismo , Mycobacterium bovis/patogenicidade , Mycobacterium tuberculosis/patogenicidade , Fagossomos/metabolismo , Linhagem Celular Tumoral , Membrana Celular/patologia , Humanos , Macrófagos/microbiologia , Fagossomos/microbiologia , Células THP-1 , Fatores de Virulência
13.
Nat Microbiol ; 1: 15019, 2016 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-27571976

RESUMO

Mycobacterium tuberculosis is a major, globally spread, aerosol-transmitted human pathogen, thought to have evolved by clonal expansion from a Mycobacterium canettii-like progenitor. In contrast, extant M. canettii strains are rare, genetically diverse, and geographically restricted mycobacteria of only marginal epidemiological importance. Here, we show that the contrasting evolutionary success of these two groups is linked to loss of lipooligosaccharide biosynthesis and subsequent morphotype changes. Spontaneous smooth-to-rough M. canettii variants were found to be mutated in the polyketide-synthase-encoding pks5 locus and deficient in lipooligosaccharide synthesis, a phenotype restored by complementation. Importantly, these rough variants showed an altered host-pathogen interaction and increased virulence in cellular- and animal-infection models. In one variant, lipooligosaccharide deficiency occurred via homologous recombination between two pks5 genes and removal of the intervening acyltransferase-encoding gene. The resulting single pks5 configuration is similar to that fixed in M. tuberculosis, which is known to lack lipooligosaccharides. Our results suggest that pks5-recombination-mediated bacterial surface remodelling increased virulence, driving evolution from putative generalist mycobacteria towards professional pathogens of mammalian hosts.


Assuntos
Vias Biossintéticas , Evolução Molecular , Lipopolissacarídeos/biossíntese , Mycobacterium/genética , Mycobacterium/patogenicidade , Policetídeo Sintases/genética , Policetídeo Sintases/metabolismo , Animais , Modelos Animais de Doenças , Deleção de Genes , Teste de Complementação Genética , Recombinação Homóloga , Interações Hospedeiro-Patógeno , Humanos , Camundongos , Infecções por Mycobacterium/microbiologia , Infecções por Mycobacterium/patologia , Virulência
14.
Sci Rep ; 5: 16918, 2015 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-26603639

RESUMO

Mycobacterium tuberculosis, the agent of human tuberculosis has developed different virulence mechanisms and virulence-associated tools during its evolution to survive and multiply inside the host. Based on previous reports and by analogy with other bacteria, phospholipases C (PLC) of M. tuberculosis were thought to be among these tools. To get deeper insights into the function of PLCs, we investigated their putative involvement in the intracellular lifestyle of M. tuberculosis, with emphasis on phagosomal rupture and virulence, thereby re-visiting a research theme of longstanding interest. Through the construction and use of an M. tuberculosis H37Rv PLC-null mutant (ΔPLC) and control strains, we found that PLCs of M. tuberculosis were not required for induction of phagosomal rupture and only showed marginal, if any, impact on virulence of M. tuberculosis in the cellular and mouse infection models used in this study. In contrast, we found that PLC-encoding genes were strongly upregulated under phosphate starvation and that PLC-proficient M. tuberculosis strains survived better than ΔPLC mutants under conditions where phosphatidylcholine served as sole phosphate source, opening new perspectives for studies on the role of PLCs in the lifecycle of M. tuberculosis.


Assuntos
Proteínas de Bactérias/metabolismo , Mycobacterium tuberculosis/enzimologia , Mycobacterium tuberculosis/patogenicidade , Tuberculose/enzimologia , Fosfolipases Tipo C/metabolismo , Animais , Proteínas de Bactérias/genética , Linhagem Celular , Colorimetria , Feminino , Humanos , Estágios do Ciclo de Vida , Pulmão/microbiologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Óperon/genética , Fagossomos/metabolismo , Fosfatidilcolinas/metabolismo , Baço/microbiologia , Tuberculose/microbiologia , Tuberculose/patologia , Fosfolipases Tipo C/deficiência , Fosfolipases Tipo C/genética , Virulência/genética
15.
Sci Transl Med ; 7(289): 289ra85, 2015 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-26019221

RESUMO

Inflammation adversely affects the health of millions of people worldwide, and there is an unmet medical need for better anti-inflammatory drugs. We evaluated the therapeutic interest of mycolactone, a polyketide-derived macrolide produced by Mycobacterium ulcerans. Bacterial production of mycolactone in human skin causes a combination of ulcerative, analgesic, and anti-inflammatory effects. Whereas ulcer formation is mediated by the proapoptotic activity of mycolactone on skin cells via hyperactivation of Wiskott-Aldrich syndrome proteins, analgesia results from neuronal hyperpolarization via signaling through angiotensin II type 2 receptors. Mycolactone also blunts the capacity of immune cells to produce inflammatory mediators by an independent mechanism of protein synthesis blockade. In an attempt to isolate the structural determinants of mycolactone's immunosuppressive activity, we screened a library of synthetic subunits of mycolactone for inhibition of cytokine production by activated T cells. The minimal structure retaining immunosuppressive activity was a truncated version of mycolactone, missing one of the two core-branched polyketide chains. This compound inhibited the inflammatory cytokine responses of human primary cells at noncytotoxic doses and bound to angiotensin II type 2 receptors comparably to mycolactone in vitro. Notably, it was considerably less toxic than mycolactone in human primary dermal fibroblasts modeling ulcerative activity. In mouse models of human diseases, it conferred systemic protection against chronic skin inflammation and inflammatory pain, with no apparent side effects. In addition to establishing the anti-inflammatory potency of mycolactone in vivo, our study therefore highlights the translational potential of mycolactone core-derived structures as prospective immunosuppressants.


Assuntos
Inflamação/tratamento farmacológico , Macrolídeos/uso terapêutico , Animais , Doença Crônica , Células HeLa , Humanos , Imunomodulação , Inflamação/patologia , Células Jurkat , Macrolídeos/química , Camundongos , Mycobacterium ulcerans/fisiologia , Dor/complicações , Dor/tratamento farmacológico , Substâncias Protetoras/uso terapêutico , Acetato de Tetradecanoilforbol/farmacologia
16.
Infect Immun ; 83(2): 780-91, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25486995

RESUMO

Mycobacterium abscessus is a pathogenic, rapidly growing mycobacterium involved in pulmonary and cutaneo-mucous infections worldwide, to which cystic fibrosis patients are exquisitely susceptible. The analysis of the genome sequence of M. abscessus showed that this bacterium is endowed with the metabolic pathways typically found in environmental microorganisms that come into contact with soil, plants, and aquatic environments, where free-living amoebae are frequently present. M. abscessus also contains several genes that are characteristically found only in pathogenic bacteria. One of them is MAB_0555, encoding a putative phospholipase C (PLC) that is absent from most other rapidly growing mycobacteria, including Mycobacterium chelonae and Mycobacterium smegmatis. Here, we report that purified recombinant M. abscessus PLC is highly cytotoxic to mouse macrophages, presumably due to hydrolysis of membrane phospholipids. We further showed by constructing and using an M. abscessus PLC knockout mutant that loss of PLC activity is deleterious to M. abscessus intracellular survival in amoebae. The importance of PLC is further supported by the fact that M. abscessus PLC was found to be expressed only in amoebae. Aerosol challenge of mice with M. abscessus strains that were precultured in amoebae enhanced M. abscessus lung infectivity relative to M. abscessus grown in broth culture. Our study underlines the importance of PLC for the virulence of M. abscessus. Despite the difficulties of isolating M. abscessus from environmental sources, our findings suggest that M. abscessus has evolved in close contact with environmental protozoa, which supports the argument that amoebae may contribute to the virulence of opportunistic mycobacteria.


Assuntos
Amoeba/fisiologia , Infecções por Mycobacterium não Tuberculosas/imunologia , Mycobacterium/patogenicidade , Fosfolipases Tipo C/fisiologia , Amoeba/microbiologia , Animais , Sequência de Bases , Células Cultivadas , Técnicas de Cocultura , Fibrose Cística/microbiologia , Técnicas de Inativação de Genes , Genoma Bacteriano/genética , Macrófagos/imunologia , Lipídeos de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Mycobacterium/enzimologia , Mycobacterium/genética , Infecções por Mycobacterium não Tuberculosas/microbiologia , Proteínas Recombinantes , Análise de Sequência de DNA , Fosfolipases Tipo C/genética , Fatores de Virulência/genética
17.
Future Microbiol ; 9(8): 969-85, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25302954

RESUMO

Mycobacterium tuberculosis, the etiological agent of human TB, is the most important mycobacterial pathogen in terms of global patient numbers and gravity of disease. The molecular mechanisms by which M. tuberculosis causes disease are complex and the result of host-pathogen coevolution that might have started already in the time of its Mycobacterium canettii-like progenitors. Despite research progress, M. tuberculosis still holds many secrets of its successful strategy for circumventing host defences, persisting in the host and developing resistance, which makes anti-TB treatment regimens extremely long and often inefficient. Here, we discuss what we have learned from recent studies on the evolution of the pathogen and its putative new drug targets that are essential for mycobacterial growth under in vitro or in vivo conditions.


Assuntos
Antituberculosos/farmacologia , Desenho de Fármacos , Evolução Molecular , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/patogenicidade , Tuberculose/microbiologia , Animais , Antituberculosos/classificação , Genes Bacterianos , Genes Essenciais , Humanos , Camundongos , Terapia de Alvo Molecular , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/crescimento & desenvolvimento , Tuberculose/tratamento farmacológico , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
18.
J Med Chem ; 57(17): 7382-95, 2014 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-25158122

RESUMO

Mycolactone is a complex macrolide toxin produced by Mycobacterium ulcerans, the causative agent of skin lesions called Buruli ulcers. Mycolactone-mediated activation of neural (N) Wiskott-Aldrich syndrome proteins (WASP) induces defects in cell adhesion underpinning cytotoxicity and disease pathogenesis. We describe the chemical synthesis of 23 novel mycolactone analogues that differ in structure and modular assembly of the lactone core with its northern and southern polyketide side chains. The lactone core linked to southern chain was the minimal structure binding N-WASP and hematopoietic homolog WASP, where the number and configuration of hydroxyl groups on the acyl side chain impacted the degree of binding. A fluorescent derivative of this compound showed time-dependent accumulation in target cells. Furthermore, a simplified version of mycolactone mimicked the natural toxin for activation of WASP in vitro and induced comparable alterations of epithelial cell adhesion. Therefore, it constitutes a structural and functional surrogate of mycolactone for WASP/N-WASP-dependent effects.


Assuntos
Toxinas Bacterianas/química , Macrolídeos/química , Proteína da Síndrome de Wiskott-Aldrich/química , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/farmacologia , Adesão Celular/efeitos dos fármacos , Células HeLa , Humanos , Cinética , Macrolídeos/metabolismo , Macrolídeos/farmacologia , Modelos Químicos , Estrutura Molecular , Mycobacterium ulcerans/química , Ligação Proteica , Proteína da Síndrome de Wiskott-Aldrich/metabolismo
19.
Mol Microbiol ; 90(3): 612-29, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23998761

RESUMO

Mycobacterium abscessus is an emerging pathogen that is increasingly recognized as a relevant cause of human lung infection in cystic fibrosis patients. This highly antibiotic-resistant mycobacterium is an exception within the rapidly growing mycobacteria, which are mainly saprophytic and non-pathogenic organisms. M. abscessus manifests as either a smooth (S) or a rough (R) colony morphotype, which is of clinical importance as R morphotypes are associated with more severe and persistent infections. To better understand the molecular mechanisms behind the S/R alterations, we analysed S and R variants of three isogenic M. abscessus S/R pairs using an unbiased approach involving genome and transcriptome analyses, transcriptional fusions and integrating constructs. This revealed different small insertions, deletions (indels) or single nucleotide polymorphisms within the non-ribosomal peptide synthase gene cluster mps1-mps2-gap or mmpl4b in the three R variants, consistent with the transcriptional differences identified within this genomic locus that is implicated in the synthesis and transport of Glyco-Peptido-Lipids (GPL). In contrast to previous reports, the identification of clearly defined genetic lesions responsible for the loss of GPL-production or transport makes a frequent switching back-and-forth between smooth and rough morphologies in M. abscessus highly unlikely, which is important for our understanding of persistent M. abscessus infections.


Assuntos
Genes Bacterianos , Lipídeos/biossíntese , Infecções por Mycobacterium não Tuberculosas/microbiologia , Mycobacterium/genética , Peptídeo Sintases/genética , Proteínas de Bactérias/genética , Sequência de Bases , Perfilação da Expressão Gênica , Variação Genética , Genoma Bacteriano , Humanos , Mutação INDEL , Dados de Sequência Molecular , Família Multigênica , Mycobacterium/classificação , Mycobacterium/patogenicidade , Polimorfismo de Nucleotídeo Único
20.
PLoS Negl Trop Dis ; 5(7): e1237, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21811642

RESUMO

BACKGROUND: Mycobacterium ulcerans, the causative agent of Buruli ulcer (BU), is unique among human pathogens in its capacity to produce a polyketide-derived macrolide called mycolactone, making this molecule an attractive candidate target for diagnosis and disease monitoring. Whether mycolactone diffuses from ulcerated lesions in clinically accessible samples and is modulated by antibiotic therapy remained to be established. METHODOLOGY/PRINCIPAL FINDING: Peripheral blood and ulcer exudates were sampled from patients at various stages of antibiotic therapy in Ghana and Ivory Coast. Total lipids were extracted from serum, white cell pellets and ulcer exudates with organic solvents. The presence of mycolactone in these extracts was then analyzed by a recently published, field-friendly method using thin layer chromatography and fluorescence detection. This approach did not allow us to detect mycolactone accurately, because of a high background due to co-extracted human lipids. We thus used a previously established approach based on high performance liquid chromatography coupled to mass spectrometry. By this means, we could identify structurally intact mycolactone in ulcer exudates and serum of patients, and evaluate the impact of antibiotic treatment on the concentration of mycolactone. CONCLUSIONS/SIGNIFICANCE: Our study provides the proof of concept that assays based on mycolactone detection in serum and ulcer exudates can form the basis of BU diagnostic tests. However, the identification of mycolactone required a technology that is not compatible with field conditions and point-of-care assays for mycolactone detection remain to be worked out. Notably, we found mycolactone in ulcer exudates harvested at the end of antibiotic therapy, suggesting that the toxin is eliminated by BU patients at a slow rate. Our results also indicated that mycolactone titres in the serum may reflect a positive response to antibiotics, a possibility that it will be interesting to examine further through longitudinal studies.


Assuntos
Toxinas Bacterianas/sangue , Úlcera de Buruli/sangue , Úlcera de Buruli/microbiologia , Mycobacterium ulcerans/isolamento & purificação , Adolescente , Adulto , Antibacterianos/uso terapêutico , Toxinas Bacterianas/análise , Biomarcadores/análise , Biomarcadores/sangue , Úlcera de Buruli/diagnóstico , Úlcera de Buruli/tratamento farmacológico , Cromatografia Líquida de Alta Pressão , Cromatografia em Camada Fina , Exsudatos e Transudatos/química , Exsudatos e Transudatos/microbiologia , Feminino , Humanos , Leucócitos Mononucleares/química , Leucócitos Mononucleares/microbiologia , Macrolídeos , Masculino , Espectrometria de Massas , Mycobacterium ulcerans/química , Ferimentos e Lesões/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA