Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 13(6): e1006367, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28570642

RESUMEN

Tuberculosis remains a global pandemic and drives lung matrix destruction to transmit. Whilst pathways driving inflammatory responses in macrophages have been relatively well described, negative regulatory pathways are less well defined. We hypothesised that Mycobacterium tuberculosis (Mtb) specifically targets negative regulatory pathways to augment immunopathology. Inhibition of signalling through the PI3K/AKT/mTORC1 pathway increased matrix metalloproteinase-1 (MMP-1) gene expression and secretion, a collagenase central to TB pathogenesis, and multiple pro-inflammatory cytokines. In patients with confirmed pulmonary TB, PI3Kδ expression was absent within granulomas. Furthermore, Mtb infection suppressed PI3Kδ gene expression in macrophages. Interestingly, inhibition of the MNK pathway, downstream of pro-inflammatory p38 and ERK MAPKs, also increased MMP-1 secretion, whilst suppressing secretion of TH1 cytokines. Cross-talk between the PI3K and MNK pathways was demonstrated at the level of eIF4E phosphorylation. Mtb globally suppressed the MMP-inhibitory pathways in macrophages, reducing levels of mRNAs encoding PI3Kδ, mTORC-1 and MNK-1 via upregulation of miRNAs. Therefore, Mtb disrupts negative regulatory pathways at multiple levels in macrophages to drive a tissue-destructive phenotype that facilitates transmission.


Asunto(s)
Macrófagos/inmunología , Mycobacterium tuberculosis/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Humanos , Macrófagos/microbiología , Metaloproteinasa 1 de la Matriz/genética , Metaloproteinasa 1 de la Matriz/inmunología , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Complejos Multiproteicos/genética , Complejos Multiproteicos/inmunología , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/fisiología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/inmunología , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/inmunología , Tuberculosis Pulmonar/genética , Tuberculosis Pulmonar/microbiología , Tuberculosis Pulmonar/patología
2.
J Infect Dis ; 212(3): 463-73, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-25676469

RESUMEN

A central tenet of tuberculosis pathogenesis is that caseous necrosis leads to extracellular matrix destruction and bacterial transmission. We reconsider the underlying mechanism of tuberculosis pathology and demonstrate that collagen destruction may be a critical initial event, causing caseous necrosis as opposed to resulting from it. In human tuberculosis granulomas, regions of extracellular matrix destruction map to areas of caseous necrosis. In mice, transgenic expression of human matrix metalloproteinase 1 causes caseous necrosis, the pathological hallmark of human tuberculosis. Collagen destruction is the principal pathological difference between humanised mice and wild-type mice with tuberculosis, whereas the release of proinflammatory cytokines does not differ, demonstrating that collagen breakdown may lead to cell death and caseation. To investigate this hypothesis, we developed a 3-dimensional cell culture model of tuberculosis granuloma formation, using bioelectrospray technology. Collagen improved survival of Mycobacterium tuberculosis-infected cells analyzed on the basis of a lactate dehydrogenase release assay, propidium iodide staining, and measurement of the total number of viable cells. Taken together, these findings suggest that collagen destruction is an initial event in tuberculosis immunopathology, leading to caseous necrosis and compromising the immune response, revealing a previously unappreciated role for the extracellular matrix in regulating the host-pathogen interaction.


Asunto(s)
Matriz Extracelular/química , Matriz Extracelular/metabolismo , Granuloma/metabolismo , Granuloma/patología , Tuberculosis/metabolismo , Tuberculosis/patología , Animales , Colágeno/metabolismo , Granuloma/microbiología , Interacciones Huésped-Patógeno , Humanos , Pulmón/química , Pulmón/patología , Neoplasias Pulmonares/patología , Metaloproteinasa 1 de la Matriz/genética , Metaloproteinasa 1 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Necrosis/metabolismo , Necrosis/patología
3.
Infect Immun ; 83(2): 730-42, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25452551

RESUMEN

A recombinant macrophage infectivity potentiator (rMIP) protein of Neisseria meningitidis induces significant serum bactericidal antibody production in mice and is a candidate meningococcal vaccine antigen. However, bioinformatics analysis of MIP showed some amino acid sequence similarity to human FK506-binding proteins (FKBPs) in residues 166 to 252 located in the globular domain of the protein. To circumvent the potential concern over generating antibodies that could recognize human proteins, we immunized mice with recombinant truncated type I rMIP proteins that lacked the globular domain and the signal leader peptide (LP) signal sequence (amino acids 1 to 22) and contained the His purification tag at either the N or C terminus (C-term). The immunogenicity of truncated rMIP proteins was compared to that of full (i.e., full-length) rMIP proteins (containing the globular domain) with either an N- or C-terminal His tag and with or without the LP sequence. By comparing the functional murine antibody responses to these various constructs, we determined that C-term His truncated rMIP (-LP) delivered in liposomes induced high levels of antibodies that bound to the surface of wild-type but not Δmip mutant meningococci and showed bactericidal activity against homologous type I MIP (median titers of 128 to 256) and heterologous type II and III (median titers of 256 to 512) strains, thereby providing at least 82% serogroup B strain coverage. In contrast, in constructs lacking the LP, placement of the His tag at the N terminus appeared to abrogate bactericidal activity. The strategy used in this study would obviate any potential concerns regarding the use of MIP antigens for inclusion in bacterial vaccines.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Proteínas Bacterianas/inmunología , Meningitis Meningocócica/inmunología , Vacunas Meningococicas/inmunología , Neisseria meningitidis/inmunología , Proteínas de Unión a Tacrolimus/inmunología , Secuencia de Aminoácidos , Animales , Proteínas Bacterianas/genética , Clonación Molecular , Reacciones Cruzadas/inmunología , Humanos , Meningitis Meningocócica/prevención & control , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Alineación de Secuencia , Vacunación
4.
Mol Microbiol ; 85(3): 461-77, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22646689

RESUMEN

The transcriptional regulator PrfA controls key virulence determinants of the facultative intracellular pathogen Listeria monocytogenes. PrfA-dependent gene expression is strongly induced within host cells. While the basis of this activation is unknown, the structural homology of PrfA with the cAMP receptor protein (Crp) and the finding of constitutively activated PrfA* mutants suggests it may involve ligand-induced allostery. Here, we report the identification of a solvent-accessible cavity within the PrfA N-terminal domain that may accommodate an activating ligand. The pocket occupies a similar position to the cAMP binding site in Crp but lacks the cyclic nucleotide-anchoring motif and has its entrance on the opposite side of the ß-barrel. Site-directed mutations in this pocket impaired intracellular PrfA-dependent gene activation without causing extensive structural/functional alterations to PrfA. Two substitutions, L48F and Y63W, almost completely abolished intracellular virulence gene induction and thus displayed the expected phenotype for allosteric activation-deficient PrfA mutations. Neither PrfA(allo) substitution affected vacuole escape and initial intracellular growth of L. monocytogenes in epithelial cells and macrophages but caused defective cell-to-cell spread and strong attenuation in mice. Our data support the hypothesis that PrfA is allosterically activated during intracellular infection and identify the probable binding site for the effector ligand. They also indicate that PrfA allosteric activation is not required for early intracellular survival but is essential for full Listeria virulence and colonization of host tissues.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidad , Mutación , Factores de Terminación de Péptidos/química , Factores de Terminación de Péptidos/genética , Activación Transcripcional , Regulación Alostérica/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Sitios de Unión , Línea Celular , AMP Cíclico/metabolismo , Femenino , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Fagosomas/metabolismo , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Alineación de Secuencia , Vacuolas , Virulencia/genética
5.
Cytoskeleton (Hoboken) ; 80(7-8): 266-274, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36855298

RESUMEN

Septins are evolutionarily conserved GTP-binding proteins known for their roles in cell division and host defence against Shigella infection. Although septin group members are viewed to function as hetero-oligomeric complexes, the role of individual septins within these complexes or in isolation is poorly understood. Decades of work using mouse models has shown that some septins (including SEPT7) are essential for animal development, while others (including SEPT6) are dispensable, suggesting that some septins may compensate for the absence of others. The zebrafish genome encodes 19 septin genes, representing the full complement of septin groups described in mice and humans. In this report, we characterise null mutants for zebrafish Sept6 (a member of the SEPT6 group) and Sept15 (a member of the SEPT7 group) and test their role in zebrafish development and host defence against Shigella infection. We show that null mutants for Sept6 and Sept15 are both viable, and that expression of other zebrafish septins are not significantly affected by their mutation. Consistent with previous reports using knockdown of Sept2, Sept7b, and Sept15, we show that Sept6 and Sept15 are required for host defence against Shigella infection. These results highlight Shigella infection of zebrafish as a powerful system to study the role of individual septins in vivo.


Asunto(s)
Disentería Bacilar , Septinas , Animales , Disentería Bacilar/genética , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Septinas/genética , Septinas/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo
6.
Sci Adv ; 9(36): eadf9706, 2023 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-37672585

RESUMEN

Trained immunity is a long-term memory of innate immune cells, generating an improved response upon reinfection. Shigella is an important human pathogen and inflammatory paradigm for which there is no effective vaccine. Using zebrafish larvae, we demonstrate that after Shigella training, neutrophils are more efficient at bacterial clearance. We observe that Shigella-induced protection is nonspecific and has differences with training by BCG and ß-glucan. Analysis of histone ChIP-seq on trained neutrophils revealed that Shigella training deposits the active H3K4me3 mark on promoter regions of 1612 genes, dramatically changing the epigenetic landscape of neutrophils toward enhanced microbial recognition and mitochondrial ROS production. Last, we demonstrate that mitochondrial ROS plays a key role in enhanced antimicrobial activity of trained neutrophils. It is envisioned that signals and mechanisms we discover here can be used in other vertebrates, including humans, to suggest new therapeutic strategies involving neutrophils to control bacterial infection.


Asunto(s)
Infecciones por Enterobacteriaceae , Epigénesis Genética , Mycobacterium bovis , Neutrófilos , Inmunidad Entrenada , beta-Glucanos , Infecciones por Enterobacteriaceae/inmunología , Animales , Pez Cebra , Larva , Neutrófilos/inmunología , Neutrófilos/metabolismo , Shigella flexneri/fisiología , Mycobacterium bovis/inmunología , beta-Glucanos/administración & dosificación , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo
7.
Cytoskeleton (Hoboken) ; 80(7-8): 254-265, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35460543

RESUMEN

Apoptosis is a form of regulated cell death essential for tissue homeostasis and embryonic development. Apoptosis also plays a key role during bacterial infection, yet some intracellular bacterial pathogens (such as Shigella flexneri, whose lipopolysaccharide can block apoptosis) can manipulate cell death programs as an important survival strategy. Septins are a component of the cytoskeleton essential for mitochondrial dynamics and host defense, however, the role of septins in regulated cell death is mostly unknown. Here, we discover that septins promote mitochondrial (i.e., intrinsic) apoptosis in response to treatment with staurosporine (a pan-kinase inhibitor) or etoposide (a DNA topoisomerase inhibitor). Consistent with a role for septins in mitochondrial dynamics, septins promote the release of mitochondrial protein cytochrome c in apoptotic cells and are required for the proteolytic activation of caspase-3, caspase-7, and caspase-9 (core components of the apoptotic machinery). Apoptosis of HeLa cells induced in response to infection by S. flexneri ΔgalU (a lipopolysaccharide mutant unable to block apoptosis) is also septin-dependent. In vivo, zebrafish larvae are significantly more susceptible to infection with S. flexneri ΔgalU (as compared to infection with wildtype S. flexneri), yet septin deficient larvae are equally susceptible to infection with S. flexneri ΔgalU and wildtype S. flexneri. These data provide a new molecular framework to understand the complexity of mitochondrial apoptosis and its ability to combat bacterial infection.

8.
Antibiotics (Basel) ; 11(10)2022 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-36289932

RESUMEN

We evaluated a novel physiological 3-D bioelectrospray model of the tuberculosis (TB) granuloma to test the activity of a known anti-TB drug, clofazimine; three carbapenems with potential activity, including one currently used in therapy; and nitazoxanide, an anti-parasitic compound with possible TB activity (all chosen as conventional drug susceptibility was problematical). PBMCs collected from healthy donors were isolated and infected with M. tuberculosis H37Rv lux (i.e., luciferase). Microspheres were generated with the infected cells; the anti-microbial compounds were added and bacterial luminescence was monitored for at least 21 days. Clavulanate was added to each carbapenem to inhibit beta-lactamases. M. tuberculosis (MTB) killing efficacy was dose dependent. Clofazimine was the most effective drug inhibiting MTB growth at 2 mg/L with good killing activity at both concentrations tested. It was the only drug that killed bacteria at the lowest concentration tested. Carbapenems showed modest initial activity that was lost at around day 10 of incubation and clavulanate did not increase killing activity. Of the carbapenems tested, tebipenem was the most efficient in killing MTB, albeit at a high concentration. Nitazoxanide was effective only at concentrations not achievable with current dosing (although this might partly have been an artefact related to extensive protein binding).

9.
Emerg Infect Dis ; 16(1): 136-8, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20031061

RESUMEN

Two species of Listeria are pathogenic; L. monocytogenes infects humans and animals, and L. ivanovii has been considered to infect ruminants only. We report L. ivanovii-associated gastroenteritis and bacteremia in a man. This isolate was indistinguishable from prototypic ruminant strains. L. ivanovii is thus an enteric opportunistic human pathogen.


Asunto(s)
Listeria/patogenicidad , Listeriosis/microbiología , Animales , Bacteriemia/microbiología , Gastroenteritis/microbiología , Cabras/microbiología , Humanos , Huésped Inmunocomprometido , Listeriosis/epidemiología , Masculino , Persona de Mediana Edad , Infecciones Oportunistas/microbiología , Paris/epidemiología
10.
JCI Insight ; 5(18)2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32780727

RESUMEN

BACKGROUNDTuberculosis (TB) kills more people than any other infection, and new diagnostic tests to identify active cases are required. We aimed to discover and verify novel markers for TB in nondepleted plasma.METHODSWe applied an optimized quantitative proteomics discovery methodology based on multidimensional and orthogonal liquid chromatographic separation combined with high-resolution mass spectrometry to study nondepleted plasma of 11 patients with active TB compared with 10 healthy controls. Prioritized candidates were verified in independent UK (n = 118) and South African cohorts (n = 203).RESULTSWe generated the most comprehensive TB plasma proteome to date, profiling 5022 proteins spanning 11 orders-of-magnitude concentration range with diverse biochemical and molecular properties. We analyzed the predominantly low-molecular weight subproteome, identifying 46 proteins with significantly increased and 90 with decreased abundance (peptide FDR ≤ 1%, q ≤ 0.05). Verification was performed for novel candidate biomarkers (CFHR5, ILF2) in 2 independent cohorts. Receiver operating characteristics analyses using a 5-protein panel (CFHR5, LRG1, CRP, LBP, and SAA1) exhibited discriminatory power in distinguishing TB from other respiratory diseases (AUC = 0.81).CONCLUSIONWe report the most comprehensive TB plasma proteome to date, identifying novel markers with verification in 2 independent cohorts, leading to a 5-protein biosignature with potential to improve TB diagnosis. With further development, these biomarkers have potential as a diagnostic triage test.FUNDINGColciencias, Medical Research Council, Innovate UK, NIHR, Academy of Medical Sciences, Program for Advanced Research Capacities for AIDS, Wellcome Centre for Infectious Diseases Research.


Asunto(s)
Biomarcadores/sangre , Mycobacterium tuberculosis/metabolismo , Proteoma/análisis , Tuberculosis Pulmonar/sangre , Tuberculosis Pulmonar/epidemiología , Estudios de Casos y Controles , Femenino , Estudios de Seguimiento , Redes Reguladoras de Genes , Humanos , Masculino , Perú/epidemiología , Estudios Prospectivos , Proteoma/metabolismo , Curva ROC , Sudáfrica/epidemiología , Tuberculosis Pulmonar/microbiología , Tuberculosis Pulmonar/patología
11.
Elife ; 92020 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-32091388

RESUMEN

Previously, we developed a 3-dimensional cell culture model of human tuberculosis (TB) and demonstrated its potential to interrogate the host-pathogen interaction (Tezera et al., 2017a). Here, we use the model to investigate mechanisms whereby immune checkpoint therapy for cancer paradoxically activates TB infection. In patients, PD-1 is expressed in Mycobacterium tuberculosis (Mtb)-infected lung tissue but is absent in areas of immunopathology. In the microsphere model, PD-1 ligands are up-regulated by infection, and the PD-1/PD-L1 axis is further induced by hypoxia. Inhibition of PD-1 signalling increases Mtb growth, and augments cytokine secretion. TNF-α is responsible for accelerated Mtb growth, and TNF-α neutralisation reverses augmented Mtb growth caused by anti-PD-1 treatment. In human TB, pulmonary TNF-α immunoreactivity is increased and circulating PD-1 expression negatively correlates with sputum TNF-α concentrations. Together, our findings demonstrate that PD-1 regulates the immune response in TB, and inhibition of PD-1 accelerates Mtb growth via excessive TNF-α secretion.


Asunto(s)
Inmunoterapia/métodos , Tuberculosis Latente/patología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Hipoxia de la Célula , Granuloma/metabolismo , Humanos , Tuberculosis Latente/inmunología , Microesferas , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/crecimiento & desarrollo , Mycobacterium tuberculosis/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba
12.
Curr Opin Pharmacol ; 42: 16-21, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29990957

RESUMEN

Mycobacterium tuberculosis (Mtb) kills more humans than any other infection and drug resistant strains are progressively emerging. Whilst the successful development of new agents for multi-drug resistant Mtb represents a major step forward, this progress must be balanced against recent disappointments in treatment-shortening trials. Consequently, there is a pressing need to strengthen the pipeline of drugs to treat tuberculosis (TB) and develop innovative therapeutic regimes. Approaches that bridge diverse disciplines are likely to be required to provide systems that address the limitations of current experimental models. Mtb is an obligate human pathogen that has undergone extensive co-evolution, resulting in a complex interplay between the host and pathogen. This chronic interaction involves multiple micro-environments, which may underlie some of the challenges in developing new drugs. The authors propose that advanced cell culture models of TB are likely to be an important addition to the experimental armamentarium in developing new approaches to TB, and here we review recent progress in this area and discuss the principal challenges.


Asunto(s)
Antituberculosos/farmacología , Antituberculosos/uso terapéutico , Tuberculosis Resistente a Múltiples Medicamentos/tratamiento farmacológico , Tuberculosis/tratamiento farmacológico , Animales , Humanos , Mycobacterium tuberculosis/efectos de los fármacos
13.
Bio Protoc ; 7(14)2017 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-28904991

RESUMEN

Standard cell culture models have been used to investigate disease pathology and to test new therapies for over fifty years. However, these model systems have often failed to mimic the changes occurring within three-dimensional (3-D) space where pathology occurs in vivo. To truthfully represent this, an emerging paradigm in biology is the importance of modelling disease in a physiologically relevant 3-D environment. One of the approaches for 3-D cell culture is bioelectrospray technology. This technique uses an alginate-based 3-D environment as an inert backbone within which mammalian cells and extracellular matrix can be incorporated. These alginate-based matrices produce highly reproducible results and can be mixed with different extracellular matrix components. This protocol describes a 3-D system incorporating mycobacteria, primary human blood mononuclear cells and collagen-alginate matrix to dissect the host-pathogen interaction in tuberculosis.

14.
mBio ; 8(1)2017 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-28174307

RESUMEN

Antimicrobial resistance presents one of the most significant threats to human health, with the emergence of totally drug-resistant organisms. We have combined bioengineering, genetically modified bacteria, longitudinal readouts, and fluidics to develop a transformative platform to address the drug development bottleneck, utilizing Mycobacterium tuberculosis as the model organism. We generated microspheres incorporating virulent reporter bacilli, primary human cells, and an extracellular matrix by using bioelectrospray methodology. Granulomas form within the three-dimensional matrix, and mycobacterial stress genes are upregulated. Pyrazinamide, a vital first-line antibiotic for treating human tuberculosis, kills M. tuberculosis in a three-dimensional culture but not in a standard two-dimensional culture or Middlebrook 7H9 broth, demonstrating that antibiotic sensitivity within microspheres reflects conditions in patients. We then performed pharmacokinetic modeling by combining the microsphere system with a microfluidic plate and demonstrated that we can model the effect of dynamic antibiotic concentrations on mycobacterial killing. The microsphere system is highly tractable, permitting variation of cell content, the extracellular matrix, sphere size, the infectious dose, and the surrounding medium with the potential to address a wide array of human infections and the threat of antimicrobial resistance. IMPORTANCE: Antimicrobial resistance is a major global threat, and an emerging concept is that infection should be studied in the context of host immune cells. Tuberculosis is a chronic infection that kills over a million people every year and is becoming progressively more resistant to antibiotics. Recent major studies of shorter treatment or new vaccination approaches have not been successful, demonstrating that transformative technologies are required to control tuberculosis. We have developed an entirely new system to study the infection of host cells in a three-dimensional matrix by using bioengineering. We showed that antibiotics that work in patients are effective in this microsphere system but not in standard infection systems. We then combined microspheres with microfluidics to model drug concentration changes in patients and demonstrate the effect of increasing antibiotic concentrations on bacterial survival. This system can be widely applied to address the threat of antimicrobial resistance and develop new treatments.


Asunto(s)
Técnicas Bacteriológicas/métodos , Técnicas de Cultivo de Célula/métodos , Farmacorresistencia Bacteriana , Microfluídica/métodos , Microesferas , Modelos Teóricos , Mycobacterium tuberculosis/efectos de los fármacos , Células Cultivadas , Humanos , Mycobacterium tuberculosis/crecimiento & desarrollo
15.
Elife ; 62017 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-28063256

RESUMEN

Cell biology differs between traditional cell culture and 3-dimensional (3-D) systems, and is modulated by the extracellular matrix. Experimentation in 3-D presents challenges, especially with virulent pathogens. Mycobacterium tuberculosis (Mtb) kills more humans than any other infection and is characterised by a spatially organised immune response and extracellular matrix remodelling. We developed a 3-D system incorporating virulent mycobacteria, primary human blood mononuclear cells and collagen-alginate matrix to dissect the host-pathogen interaction. Infection in 3-D led to greater cellular survival and permitted longitudinal analysis over 21 days. Key features of human tuberculosis develop, and extracellular matrix integrity favours the host over the pathogen. We optimised multiparameter readouts to study emerging therapeutic interventions: cytokine supplementation, host-directed therapy and immunoaugmentation. Each intervention modulates the host-pathogen interaction, but has both beneficial and harmful effects. This methodology has wide applicability to investigate infectious, inflammatory and neoplastic diseases and develop novel drug regimes and vaccination approaches.


Asunto(s)
Interacciones Huésped-Patógeno/efectos de los fármacos , Leucocitos Mononucleares/efectos de los fármacos , Modelos Biológicos , Mycobacterium tuberculosis/patogenicidad , Esferoides Celulares/efectos de los fármacos , Alginatos/química , Antígenos Bacterianos/farmacología , Proteínas Bacterianas/farmacología , Quimiocina CCL2/biosíntesis , Quimiocina CCL2/metabolismo , Quimiocina CXCL10/biosíntesis , Quimiocina CXCL10/metabolismo , Técnicas de Cocultivo , Colágeno/química , Dinoprostona/farmacología , Matriz Extracelular/química , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/inmunología , Regulación de la Expresión Génica , Ácido Glucurónico/química , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Ácidos Hexurónicos/química , Interacciones Huésped-Patógeno/inmunología , Humanos , Interleucina-12/biosíntesis , Interleucina-12/metabolismo , Interleucina-1beta/biosíntesis , Interleucina-1beta/metabolismo , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/microbiología , Microesferas , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/fisiología , Esferoides Celulares/inmunología , Esferoides Celulares/microbiología , Virulencia
16.
Expert Rev Vaccines ; 14(12): 1633-49, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26468663

RESUMEN

Peptidyl prolyl cis/trans isomerases (PPIases) are a superfamily of proteins ubiquitously distributed among living organisms, which function primarily to assist the folding and structuring of unfolded and partially folded polypeptide chains and proteins. In this review, we focus specifically on the Macrophage Infectivity Potentiator (MIP)-like PPIases, which are members of the immunophilin family of FK506-binding proteins (FKBP). MIP-like PPIases have accessory roles in virulence and are candidates for inclusion in vaccines protective against both animal and human bacterial pathogens. A structural vaccinology approach obviates any issues over molecular mimicry and potential cross-reactivity with human FKBP proteins and studies with a representative antigen, the Neisseria meningitidis-MIP, support this strategy. Moreover, a dual approach of vaccination and drug targeting could be considered for controlling bacterial infectious diseases of humans and animals.


Asunto(s)
Proteínas Bacterianas/inmunología , Vacunas Bacterianas/inmunología , Macrófagos/inmunología , Isomerasa de Peptidilprolil/inmunología , Proteínas de Unión a Tacrolimus/inmunología , Vacunas de Subunidad/inmunología , Secuencia de Aminoácidos , Animales , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/prevención & control , Diseño de Fármacos , Humanos , Legionella pneumophila/inmunología , Meningitis Meningocócica/inmunología , Meningitis Meningocócica/microbiología , Meningitis Meningocócica/prevención & control , Datos de Secuencia Molecular , Neisseria meningitidis/inmunología , Neisseria meningitidis/patogenicidad
17.
Vaccine ; 31(22): 2584-90, 2013 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-23566947

RESUMEN

Sera from individuals colonized with Neisseria meningitidis and from patients with meningococcal disease contain antibodies specific for the neisserial heat-shock/chaperonin (Chp)60 protein. In this study, immunization of mice with recombinant (r)Chp60 in saline; adsorbed to aluminium hydroxide; in liposomes and detergent micelles, with and without the adjuvant MonoPhosphoryl Lipid A (MPLA), induced high and similar (p>0.05) levels of antibodies that recognized Chp60 in outer membranes (OM). FACS analysis and immuno-fluorescence experiments demonstrated that Chp60 was surface-expressed on meningococci. By western blotting, murine anti-rChp60 sera recognized a protein of Mr 60kDa in meningococcal cell lysates. However, cross-reactivity with human HSP60 protein was also observed. By comparing translated protein sequences of strains, 40 different alleles were found in meningococci in the Bacterial Isolate Genome Sequence database with an additional 5 new alleles found in our selection of 13 other strains from colonized individuals and patients. Comparison of the non-redundant translated amino acid sequences from all the strains revealed ≥97% identity between meningococcal Chp60 proteins, and in our 13 strains the protein was expressed to high and similar levels. Bactericidal antibodies (median reciprocal titres of 32-64) against the homologous strain MC58 were induced by immunization with rChp60 in liposomes, detergent micelles and on Al(OH)3. Bactericidal activity was influenced by the addition of MPLA and the delivery formulation used. Moreover, the biological activity of anti-Chp60 antisera did not extend significantly to heterologous meningococcal strains. Thus, in order to provide broad coverage, vaccines based on Chp60 would require multiple proteins and specific bactericidal epitope identification.


Asunto(s)
Chaperonina 60/inmunología , Infecciones Meningocócicas/inmunología , Vacunas Meningococicas/farmacología , Neisseria meningitidis/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/genética , Anticuerpos Antibacterianos/inmunología , Formación de Anticuerpos , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/farmacología , Western Blotting , Chaperonina 60/genética , Chaperonina 60/farmacología , Reacciones Cruzadas , Técnica del Anticuerpo Fluorescente , Humanos , Infecciones Meningocócicas/microbiología , Infecciones Meningocócicas/prevención & control , Vacunas Meningococicas/genética , Vacunas Meningococicas/inmunología , Ratones , Ratones Endogámicos BALB C , Proteínas Mitocondriales/inmunología , Neisseria meningitidis/genética , Conejos , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/farmacología
18.
Curr Opin Microbiol ; 14(2): 118-27, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21388862

RESUMEN

Listeria monocytogenes is the causative agent of listeriosis, a severe foodborne infection. These bacteria live as soil saprotrophs on decaying plant matter but also as intracellular parasites, using the cell cytosol as a replication niche. PrfA, a regulatory protein, integrates a number of environmental cues that signal the transition between these two contrasting lifestyles, activating a set of key virulence factors during host infection. While a number of details concerning the general mode of action of this virulence master switch have been elucidated, others remain unsolved. Recent work has revealed additional mechanisms that contribute to L. monocytogenes virulence modulation, often via cross-talk with PrfA, or by regulating new genes involved in host colonization.


Asunto(s)
Proteínas Bacterianas/biosíntesis , Regulación Bacteriana de la Expresión Génica , Listeria monocytogenes/patogenicidad , Factores de Terminación de Péptidos/metabolismo , Factores de Virulencia/biosíntesis , Animales , Proteínas Bacterianas/metabolismo , Microbiología Ambiental , Humanos , Listeriosis/microbiología , Listeriosis/veterinaria , Modelos Biológicos , Plantas/microbiología , Transducción de Señal , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA