Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Antimicrob Agents Chemother ; 65(7): e0031621, 2021 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-33941518

RESUMO

Chronic pulmonary methicillin-resistant Staphylococcus aureus (MRSA) disease in cystic fibrosis (CF) has a high probability of recurrence following treatment with standard-of-care antibiotics and represents an area of unmet need associated with reduced life expectancy. We developed a lipoglycopeptide therapy customized for pulmonary delivery that not only demonstrates potent activity against planktonic MRSA, but also against protected colonies of MRSA in biofilms and within cells, the latter of which have been linked to clinical antibiotic failure. A library of next-generation potent lipoglycopeptides was synthesized with an emphasis on attaining superior pharmacokinetics (PK) and pharmacodynamics to similar compounds of their class. Our strategy focused on hydrophobic modification of vancomycin, where ester and amide functionality were included with carbonyl configuration and alkyl length as key variables. Candidates representative of each carbonyl attachment chemistry demonstrated potent activity in vitro, with several compounds being 30 to 60 times more potent than vancomycin. Selected compounds were advanced into in vivo nose-only inhalation PK evaluations in rats, where RV94, a potent lipoglycopeptide that utilizes an inverted amide linker to attach a 10-carbon chain to vancomycin, demonstrated the most favorable lung residence time after inhalation. Further in vitro evaluation of RV94 showed superior activity to vancomycin against an expanded panel of Gram-positive organisms, cellular accumulation and efficacy against intracellular MRSA, and MRSA biofilm killing. Moreover, in vivo efficacy of inhaled nebulized RV94 in a 48 h acute model of pulmonary MRSA (USA300) infection in neutropenic rats demonstrated statistically significant antibacterial activity that was superior to inhaled vancomycin.


Assuntos
Staphylococcus aureus Resistente à Meticilina , Infecções Estafilocócicas , Animais , Antibacterianos/uso terapêutico , Lipoglicopeptídeos , Pulmão , Testes de Sensibilidade Microbiana , Ratos , Infecções Estafilocócicas/tratamento farmacológico , Vancomicina
3.
BMC Microbiol ; 16(1): 270, 2016 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-27829372

RESUMO

BACKGROUND: Mycobacterium avium subsp. hominissuis is a common intracellular pathogen that infects patients with HIV/AIDS and cause lung infection in patients with underlying lung pathology. M.avium preferably infects macrophages and uses diverse mechanisms to alter phagosome maturation. Once in the macrophage, the pathogen can alter the host cellular defenses by secreting proteins into the cytosol of host cells, but despite considerable research, only a few secreted effector proteins have been identified. We hypothesized that the environmental cues inside the phagosome can trigger bacterial protein secretion. To identify M. avium secretome within the phagosome, we utilized a previously established in vitro system that mimics the metal ion concentrations and pH of the M. avium phagosome. RESULTS: M. avium was exposed to phagosome metal concentrations for different time points and exported proteins were profiled and analyzed against bacterial proteins secreted in the culture medium. Mass spectrometric analysis of the secreted proteome identified several proteins, of which 46 were unique to bacteria incubated in the metal mixture. Ten of potential effectors were selected and secretion of these proteins was monitored within M. avium infected mononuclear phagocytic cells using the beta-lactamase FRET-based reporter system. In addition, pull-down assay was performed for secreted calmodulin-like protein MAV_1356 protein to evaluate for eukaryotic target. All examined M. avium proteins were secreted into the macrophage cytosol, and gene expression analysis suggested that the metal environment likely stimulates secretion of pre-made proteins. Further investigation of bacterial secreted MAV_1356 protein, lead to the observation that the MAV_1356 interacts with the host proteins Annexin A1 and Protein S100-A8. CONCLUSIONS: We established an in vitro system for the study if proteins secreted intracellularly, and revealed that the metal mixture mimicking the concentration of metals in the phagosome environment, triggers protein secretion.


Assuntos
Proteínas de Bactérias/metabolismo , Mycobacterium avium/genética , Mycobacterium avium/metabolismo , Fagossomos/metabolismo , Proteínas de Bactérias/genética , Calmodulina/metabolismo , Cátions/metabolismo , Linhagem Celular , Citosol/metabolismo , DNA Bacteriano/genética , Escherichia coli/genética , Interações Hospedeiro-Patógeno , Humanos , Macrófagos/microbiologia , Metais/metabolismo , Metais/farmacologia , Monócitos/microbiologia , Mycobacterium avium/isolamento & purificação , Proteoma/metabolismo , RNA Bacteriano/genética , beta-Lactamases/metabolismo
4.
Infect Immun ; 83(2): 625-36, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25422262

RESUMO

"Mycobacterium avium subsp. hominissuis" is an opportunistic environmental pathogen that causes respiratory illness in immunocompromised patients, such as those with cystic fibrosis as well as other chronic respiratory diseases. Currently, there is no efficient approach to prevent or treat M. avium subsp. hominissuis infection in the lungs. During initial colonization of the airways, M. avium subsp. hominissuis forms microaggregates composed of 3 to 20 bacteria on human respiratory epithelial cells, which provides an environment for phenotypic changes leading to efficient mucosal invasion in vitro and in vivo. DNA microarray analysis was employed to identify genes associated with the microaggregate phenotype. The gene encoding microaggregate-binding protein 1 (MBP-1) (MAV_3013) is highly expressed during microaggregate formation. When expressed in noninvasive Mycobacterium smegmatis, MBP-1 increased the ability of the bacteria to bind to HEp-2 epithelial cells. Using anti-MBP-1 immune serum, microaggregate binding to HEp-2 cells was significantly reduced. By far-Western blotting, and verified by coimmunoprecipitation, we observed that MBP-1 interacts with the host cytoskeletal protein vimentin. As visualized by confocal microscopy, microaggregates, as well as MBP-1, induced vimentin polymerization at the site of bacterium-host cell contact. Binding of microaggregates to HEp-2 cells was inhibited by treatment with an antivimentin antibody, suggesting that MBP-1 expression is important for M. avium subsp. hominissuis adherence to the host cell. MBP-1 immune serum significantly inhibited M. avium subsp. hominissuis infection throughout the respiratory tracts of mice. This study characterizes a pathogenic mechanism utilized by M. avium subsp. hominissuis to bind and invade the host respiratory epithelium, suggesting new potential targets for the development of antivirulence therapy.


Assuntos
Aderência Bacteriana/genética , Proteínas de Bactérias/genética , Células Epiteliais/microbiologia , Mycobacterium avium/patogenicidade , Mucosa Respiratória/microbiologia , Animais , Aderência Bacteriana/imunologia , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/metabolismo , Linhagem Celular Tumoral , Células Epiteliais/citologia , Feminino , Humanos , Soros Imunes/imunologia , Soros Imunes/farmacologia , Hospedeiro Imunocomprometido , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Mycobacterium/microbiologia , Infecções por Mycobacterium/patologia , Mycobacterium avium/genética , Mycobacterium avium/imunologia , Mycobacterium smegmatis/genética , Mycobacterium smegmatis/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Mucosa Respiratória/citologia , Infecções Respiratórias/microbiologia , Infecções Respiratórias/patologia , Vimentina/antagonistas & inibidores , Vimentina/imunologia , Vimentina/metabolismo
5.
Infect Immun ; 82(1): 405-12, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24191301

RESUMO

Mycobacterium avium subsp. hominissuis is an opportunistic human pathogen that has been shown to form biofilm in vitro and in vivo. Biofilm formation in vivo appears to be associated with infections in the respiratory tract of the host. The reasoning behind how M. avium subsp. hominissuis biofilm is allowed to establish and persist without being cleared by the innate immune system is currently unknown. To identify the mechanism responsible for this, we developed an in vitro model using THP-1 human mononuclear phagocytes cocultured with established M. avium subsp. hominissuis biofilm and surveyed various aspects of the interaction, including phagocyte stimulation and response, bacterial killing, and apoptosis. M. avium subsp. hominissuis biofilm triggered robust tumor necrosis factor alpha (TNF-α) release from THP-1 cells as well as superoxide and nitric oxide production. Surprisingly, the hyperstimulated phagocytes did not effectively eliminate the cells of the biofilm, even when prestimulated with gamma interferon (IFN-γ) or TNF-α or cocultured with natural killer cells (which have been shown to induce anti-M. avium subsp. hominissuis activity when added to THP-1 cells infected with planktonic M. avium subsp. hominissuis). Time-lapse microscopy and the TUNEL (terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling) assay determined that contact with the M. avium subsp. hominissuis biofilm led to early, widespread onset of apoptosis, which is not seen until much later in planktonic M. avium subsp. hominissuis infection. Blocking TNF-α or TNF-R1 during interaction with the biofilm significantly reduced THP-1 apoptosis but did not lead to elimination of M. avium subsp. hominissuis. Our data collectively indicate that M. avium subsp. hominissuis biofilm induces TNF-α-driven hyperstimulation and apoptosis of surveilling phagocytes, which prevents clearance of the biofilm by cells of the innate immune system and allows the biofilm-associated infection to persist.


Assuntos
Apoptose/fisiologia , Biofilmes/crescimento & desenvolvimento , Complexo Mycobacterium avium/fisiologia , Infecção por Mycobacterium avium-intracellulare/fisiopatologia , Fagócitos/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Células Cultivadas , Técnicas de Cocultura , Humanos , Imunidade Celular , Imunidade Inata , Células Matadoras Naturais/fisiologia , Complexo Mycobacterium avium/imunologia , Óxido Nítrico/metabolismo , Superóxidos/metabolismo
6.
Pharmaceutics ; 15(9)2023 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-37765219

RESUMO

The increased prevalence of pulmonary methicillin-resistant Staphylococcus aureus (MRSA) infection in patients living with cystic fibrosis (CF) is concerning due to a correlation with reduced life expectancy and lack of available treatment options. RV94 is a next generation lipoglycopeptide designed for pulmonary delivery that preclinically demonstrated high potency against MRSA in planktonic and protected colonies and improved pulmonary clearance relative to same class molecules. Here, RV94 was formulated into a dry powder for inhalation (DPI) to investigate the localized treatment of pulmonary MRSA presented in a potentially more convenient dosage form. RV94 DPI was generated using a spray-drying process with 12.5 wt% trileucine and demonstrated aerosol characteristics (2.0 µm MMAD and 73% FPF) predictive of efficient pulmonary deposition. In vivo PK from a single dose of RV94 DPI delivered by inhalation to rats yielded lung levels (127 µg/g) much greater than the MRSA minimum inhibitory concentration (0.063 µg/mL), low systemic levels (0.1 µg/mL), and a lung t1/2 equal to 3.5 days. In a rat acute pulmonary MRSA model, a single dose of RV94 DPI delivered by inhalation either up to seven days prior to or 24 h after infection resulted in a statistically significant reduction in lung MRSA titer.

7.
PLoS One ; 17(8): e0272575, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36044421

RESUMO

PURPOSE: Neutrophil serine proteases (NSPs) are implicated in numerous inflammatory diseases. Thus, a robust methodology to monitor and quantify NSPs is important to study disease progression and evaluate the effect of pharmacological interventions. A comparison of the various methods used to extract NSPs from neutrophil granulocytes has not been published, providing the impetus to conduct this method optimization and comparison study. METHODS: Two NSP recovery methodologies were evaluated on samples from five human donors: zymosan stimulation and cell pellet extraction. For the zymosan stimulation method, 1 mL donor blood was added to zymosan and samples were incubated at 37°C for 30 min while shaking. Samples were then centrifuged, and the plasma was collected for quantitation of NSP activity. For the cell pellet extraction procedure, 2 mL whole blood samples were centrifuged into white blood cell pellets following red blood cell lysis. To each pellet, three sequential lysis steps were performed using either 0.05% Nonidet P-40 Substitute (NP40) or 0.02% Triton X-100 lysis buffers under agitation followed by centrifugation. NSP activities were quantified using an exogenous peptide substrate specific to each of the three NSPs being analyzed: neutrophil elastase, cathepsin G, and proteinase 3. RESULTS AND DISCUSSION: The zymosan stimulation method resulted in lower recovery of active NSPs and was unable to stimulate significant release of active cathepsin G. In contrast, the NP40 pellet extraction method showed consistent inter-donor NSP release with greater recoveries of active NSPs than the Triton method or the zymosan stimulation method. Overall, the pellet extraction procedure provided 13.3-fold greater recovery of active neutrophil elastase, 283-fold greater recovery of active cathepsin G, and 2.9-fold greater recovery of active proteinase 3 than the zymosan method. CONCLUSION: The NP40 cell pellet extraction method resulted in greater extraction of active NSPs compared to the other methods investigated here, which may allow for a more accurate and complete biomarker profile when evaluating human clinical samples.


Assuntos
Métodos Analíticos de Preparação de Amostras , Serina Proteases , Células Sanguíneas/química , Células Sanguíneas/enzimologia , Catepsina G/química , Catepsina G/metabolismo , Humanos , Elastase de Leucócito/química , Elastase de Leucócito/metabolismo , Mieloblastina , Neutrófilos/química , Neutrófilos/metabolismo , Serina Proteases/química , Serina Proteases/metabolismo , Zimosan/farmacologia
8.
Int J Microbiol ; 2019: 9167271, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31281365

RESUMO

Bovine tuberculosis (bTB) is a highly transmissible infection and remains of great concern as a zoonosis. The worldwide incidence of bTB is in rise, creating potential reservoir and increased infection risk for humans and animals. In attempts to identify novel surface antigens of Mycobacterium bovis as a proof-of-concept for potential inducers of protective immunity, we investigated surface proteome of M. bovis BCG strain that was cultured under the granuloma-like condition. We also demonstrated that the pathogen exposed to the biologically relevant environment has greater binding and invasion abilities to host cells than those of bacteria incubated under regular laboratory conditions. A total of 957 surface-exposed proteins were identified for BCG cultured under laboratory condition, whereas 1,097 proteins were expressed under the granuloma-like condition. The overexpression of Mb1524, Mb01_03198, Mb1595_p3681 (PhoU1 same as phoY1_1), and Mb1595_p0530 (HbhA) surface proteins in Mycobacterium smegmatis leads to increased binding and invasion to mucosal cells. We also examined the immunogenicity of purified recombinant proteins and tested M. smegmatis overexpressing these surface antigens for the induction of protective immunity in mice. Significantly high levels of specific IgA and IgG antibodies were observed in recombinant protein immunized groups by both inhalation and intraperitoneal (IP) routes, but only IP delivery induced high total IgA and IgG levels. We did not detect major differences in antibody levels in the M. smegmatis group that overexpressed surface antigens. In addition, the bacterial load was significantly reduced in the lungs of mice immunized with the combination of inhaled recombinant proteins. Our findings suggest that the activation of the mucosal immunity can lead to increased ability to confer protection upon M. bovis BCG infection.

9.
Microorganisms ; 7(5)2019 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-31117286

RESUMO

Mycobacterium avium subspecies hominissuis (MAH) is an opportunistic pathogen that is ubiquitous in the environment and often isolated from faucets and showerheads. MAH mostly infects humans with an underlying disease, such as chronic pulmonary disorder, cystic fibrosis, or individuals that are immunocompromised. In recent years, MAH infections in patients without concurrent disease are increasing in prevalence as well. This pathogen is resistant to many antibiotics due to the impermeability of its envelope and due to the phenotypic resistance established within the host macrophages, making difficult to treat MAH infections. By screening a MAH transposon library for mutants that are susceptible to killing by reactive nitrogen intermediaries, we identified the MAV_4644 (MAV_4644:Tn) gene knockout clone that was also significantly attenuated in growth within the host macrophages. Complementation of the mutant restored the wild-type phenotype. The MAV_4644 gene encodes a dual-function protein with a putative pore-forming function and ADP-ribosyltransferase activity. Protein binding assay suggests that MAV_4644 interacts with the host lysosomal peptidase cathepsin Z (CTSZ), a key regulator of the cell signaling and inflammation. Pathogenic mycobacteria have been shown to suppress the action of many cathepsins to establish their intracellular niche. Our results demonstrate that knocking-down the cathepsin Z in human macrophages rescues the attenuated phenotype of MAV_4644:Tn clone. Although, the purified cathepsin Z by itself does not have any killing effect on MAH, it contributes to bacterial killing in the presence of the nitric oxide (NO). Our data suggest that the cathepsin Z is involved in early macrophage killing of MAH, and the virulence factor MAV_4644 protects the pathogen from this process.

10.
Comp Immunol Microbiol Infect Dis ; 66: 101332, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31437674

RESUMO

Records of all Diagnostic laboratory submissions from 2012 to 2015 were examined and subjected to analysis according to species, location of infection, species of bacteria, and antibiotic resistance/susceptibility. A total of 23.8% of all culture isolates were Staphylococcus sp. Of those Staphylococcus, 43% were isolated from surgical site infections. Staphylococcus pseudintermedius accounted for approximately 28% of all staphylococcus cultures, while methicillin-resistant (MR) S. pseudintermedius accounted for 8% of all staphylococcus cultures. Environmental samples were also collected by swabbing surfaces in the intensive care unit (ICU) and anesthesia prep room at the OSU VTH. Isolated bacterial colonies were subjected to PCR for species identification and for the presence of the mecA gene. Ability of horizontal transfer in vitro of the mecA gene was evaluated by incubating the mecA positive bacterium, with the mecA negative bacterium, and then plated onto agar plates infused with known concentration of oxacillin. Colonies were then subjected to PCR for species and mecA identification. Horizontal transfer of the mecA gene was demonstrated and confirmed via PCR from MR S. epidermidis to MS S. pseudintermedius in an in vitro model that mimicked the veterinary hospital environment. Biofilms were established using four Staphylococcus species isolated from swabbing the Intensive Care Unit (ICU) and anesthesia prep room and were resistant when exposed to the current cleaning agent. Staphylococcus species makeup nearly » of all infections at OSU VDL during the four years of the study, and MS S. pseudintermedius was shown to acquire the mecA gene from an environmental strain.


Assuntos
Farmacorresistência Bacteriana/genética , Hospitais Veterinários , Hospitais de Ensino , Infecções Estafilocócicas/veterinária , Staphylococcus/efeitos dos fármacos , Staphylococcus/isolamento & purificação , Animais , Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Desinfecção/métodos , Testes de Sensibilidade Microbiana , Proteínas de Ligação às Penicilinas/genética , Estudos Retrospectivos , Infecções Estafilocócicas/epidemiologia , Staphylococcus/genética
11.
Artigo em Inglês | MEDLINE | ID: mdl-29740544

RESUMO

Mycobacterium avium subsp. hominissuis (M. avium) is a member of the non-tuberculous mycobacteria (NTM), and is a common cause of lung infection in patients with chronic NTM lung conditions. M. avium is an environmental bacterium believed to be transmitted from environmental sources. In this work we used a recently developed model in Caenorhabditis elegans to ask whether M. avium can be transmitted from host-to-host, and the bacterial genes associated with host colonization. Infection of C. elegans was carried out by placing the nematode in cultured with M. avium. Bacteria eliminated from the intestines of infected C. elegans were used to infect naïve nematodes. In parallel experiments, to identify colonization associated genes, a transposon library of M. avium was screened for the ability to bind to HEp-2 mucosal cells. Thirty clones were identified and five selected clones with impaired adherence to HEp-2 epithelial cells were used to infect C. elegans to determine the degree of colonization. It was determined that M. avium eliminated from infected C. elegans were able to colonize a naïve C. elegans with high efficiency. Thirty of the most adherence-deficient M. avium clones obtained from the HEp-2 cell screening were sequenced to identify the location of the transposon. Many of the genes associated with the bacterial cell wall synthesis were shown to be inactivated in the selected mutants. Five out of the 30 bacterial clones were then used to infect C. elegans. All five mutants had impaired ability to colonize C. elegans compared with the wild type bacteria (decrease of 1.5-2.0 logs, p < 0.05). The limitation of this work is that the model can be used for initial screening, but other more complex systems should be used to confirm the findings. C. elegans can be used as a model to test for M. avium adherence/colonization-associated virulence determinants. All the tested adherence-deficient clones that were examined had impaired ability to colonize the host C. elegans, and some can be potentially used to prevent colonization.


Assuntos
Caenorhabditis elegans/microbiologia , Genes Bacterianos/genética , Infecções por Mycobacterium/microbiologia , Infecções por Mycobacterium/transmissão , Mycobacterium avium/genética , Mycobacterium avium/patogenicidade , Animais , Proteínas de Bactérias/genética , Linhagem Celular , Modelos Animais de Doenças , Células Epiteliais/microbiologia , Regulação Bacteriana da Expressão Gênica , Mucosa Intestinal/microbiologia , Infecções Respiratórias/transmissão , Fatores de Virulência
12.
Virulence ; 9(1): 1287-1300, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30134761

RESUMO

Mycobacterium avium subsp. hominissuis is an opportunistic intracellular pathogen associated with disease in patients either immunosuppression or chronic lung pathology. Once in the host, M. avium preferentially infects and replicates within the phagocytic cells. The host driven macrophage apoptosis appears to be an essential aspect of innate immunity during bacterial infection; however, the existing evidence suggests that M. avium has evolved adaptive approaches to trigger the phagocyte apoptosis, exit apoptotic cells or via ingestion of infected apoptotic bodies subsequently infect neighboring macrophages. By evaluating 4,000 transposon mutants of M. avium in THP-1 cells, we identified clones that can trigger a new form of early host cell apoptosis, which is only observed upon entry into the "secondary-infected" macrophages. Inactivation of MAVA5_06970 gene lead to significant attenuation in intracellular growth within macrophages and mice, and impaired M. avium to induce rapid apoptosis in the "secondary-infected" cells as measured by Annexin V-FITC detection assay. Complementation of MAVA5_06970 gene corrected the attenuation as well as apoptotic phenotypes. The MAVA5_06970 gene encodes for a secreted protein. Using the pull-down assay and then confirmed with the yeast two-hybrid screen, we found that MAVA5_06970 effector interacts with the Secreted Phosphoprotein 1, the cytokine also known as Osteopontin. This interaction enhances the THP-1 cell apoptosis and, consequently, restricts the production of interleukin-12 that likely may limit the activation of the type I immunity pathway in vivo. This work identified a key virulence effector of M. avium that contributes to the cell-to-cell spread of the pathogen.


Assuntos
Apoptose , Proteínas de Bactérias/genética , Macrófagos/microbiologia , Mycobacterium avium/patogenicidade , Osteopontina/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Células Cultivadas , Elementos de DNA Transponíveis , Técnicas de Inativação de Genes , Interações Hospedeiro-Patógeno , Humanos , Interleucina-12/imunologia , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Mycobacterium avium/genética , Células THP-1 , Fator de Necrose Tumoral alfa/imunologia , Virulência
13.
J Med Microbiol ; 66(7): 1063-1075, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28671535

RESUMO

PURPOSE: Mycobacterium avium subsp. hominissuis is a member of the M. avium complex, a heterogeneous group of bacteria that cause lung infection in immunocompetent patients or disseminated infection in patients with immunosuppression. The bacteria belonging to this complex have variable virulence, depending on the strain considered, and therefore a representative of the most common clinical phenotype was analysed. METHODOLOGY: The genomic sequences of four M. avium subsp. hominissuis isolates obtained from clinical specimens were completed. Mav101, Mav100 and MavA5 were isolated from the blood of patients with AIDS. MavA5 was disseminated from the lung, while Mav3388 was isolated from the lungs of a patient with chronic lung disease. The sequences were annotated using the published Mav104 genome as a blueprint. Functional and virulence analyses of the sequences were carried out. Mice studies comparing the virulence of the strains were performed. RESULTS: Findings showed that while Mav101 was very similar to Mav104, there were numerous differences between Mav104 and the remaining strains at nucleotide and predicted protein levels. The presence of genes associated with biofilm formation and several known virulence-related genes were sometimes differentially present among the isolates, suggesting overlapping functions by different genetic determinants. CONCLUSIONS: The sequences provided important information about M. avium heterogenicity and evolution as a pathogen. The limitation is the lack of understanding on possible overlapping functions of genes/proteins.


Assuntos
Variação Genética , Genoma Bacteriano , Mycobacterium avium/genética , Mycobacterium avium/isolamento & purificação , Tuberculose/microbiologia , Fatores de Virulência/genética , Síndrome da Imunodeficiência Adquirida/complicações , Animais , Modelos Animais de Doenças , Humanos , Camundongos Endogâmicos C57BL
14.
mBio ; 7(6)2016 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-27923918

RESUMO

Extracellular DNA (eDNA) is an integral biofilm matrix component of numerous pathogens, including nontuberculous mycobacteria (NTM). Cell lysis is the source of eDNA in certain bacteria, but the source of eDNA remains unidentified for NTM, as well as for other eDNA-containing bacterial species. In this study, conditions affecting eDNA export were examined, and genes involved with the eDNA export mechanism were identified. After a method for monitoring eDNA in real time in undisturbed biofilms was established, different conditions affecting eDNA were investigated. Bicarbonate positively influenced eDNA export in a pH-independent manner in Mycobacterium avium, M. abscessus, and M. chelonae The surface-exposed proteome of M. avium in eDNA-containing biofilms revealed abundant carbonic anhydrases. Chemical inhibition of carbonic anhydrases with ethoxzolamide significantly reduced eDNA export. An unbiased transposon mutant library screen for eDNA export in M. avium identified many severely eDNA-attenuated mutants, including one not expressing a unique FtsK/SpoIIIE-like DNA-transporting pore, two with inactivation of carbonic anhydrases, and nine with inactivation of genes belonging to a unique genomic region, as well as numerous mutants involved in metabolism and energy production. Complementation of nine mutants that included the FtsK/SpoIIIE and carbonic anhydrase significantly restored eDNA export. Interestingly, several attenuated eDNA mutants have mutations in genes encoding proteins that were found with the surface proteomics, and many more mutations are localized in operons potentially encoding surface proteins. Collectively, our data strengthen the evidence of eDNA export being an active mechanism that is activated by the bacterium responding to bicarbonate. IMPORTANCE: Many bacteria contain extracellular DNA (eDNA) in their biofilm matrix, as it has various biological and physical functions. We recently reported that nontuberculous mycobacteria (NTM) can contain significant quantities of eDNA in their biofilms. In some bacteria, eDNA is derived from dead cells, but that does not appear to be the case for all eDNA-containing organisms, including NTM. In this study, we found that eDNA export in NTM is conditionally dependent on the molecules to which the bacteria are exposed and that bicarbonate positively influences eDNA export. We also identified genes and proteins important for eDNA export, which begins to piece together a description of a mechanism for eDNA. Better understanding of eDNA export can give new targets for the development of antivirulence drugs, which are attractive because resistance to classical antibiotics is currently a significant problem.


Assuntos
Bicarbonatos/metabolismo , Biofilmes/crescimento & desenvolvimento , DNA Bacteriano/metabolismo , Micobactérias não Tuberculosas/efeitos dos fármacos , Micobactérias não Tuberculosas/metabolismo , Proteínas de Bactérias/análise , Anidrases Carbônicas/metabolismo , Elementos de DNA Transponíveis , Técnicas de Inativação de Genes , Teste de Complementação Genética , Concentração de Íons de Hidrogênio , Proteínas de Membrana/análise , Proteínas de Membrana Transportadoras/metabolismo , Mutagênese Insercional , Micobactérias não Tuberculosas/genética , Micobactérias não Tuberculosas/fisiologia
15.
Virulence ; 6(7): 694-703, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26252358

RESUMO

The environmental opportunistic pathogen Mycobacterium avium subsp hominissuis (MAH), a member of the nontuberculous mycobacteria (NTM) cluster, causes respiratory as well as disseminated disease in patients such as those with chronic respiratory illnesses or AIDS. Currently, there is no effective method to prevent NTM respiratory infections. The formation of mycobacterial microaggregates comprises of phenotypic changes that lead to efficient adherence and invasion of the respiratory mucosa in vitro and in vivo. Microaggregate adhesion to the respiratory epithelium is mediated in part through the mycobacterial protein, MAV_3013 (MBP-1). Through DNA microarray analysis, the small hypothetical gene MAV_0831 (Microaggregate Invasion Protein-1, MIP-1) was identified as being upregulated during microaggregate formation. When MIP-1 was overexpressed in poorly-invasive Mycobacterium smegmatis, it provided the bacterium the ability to bind and enter epithelial cells. In addition, incubating microaggregates with recombinant MIP-1 protein enhanced the ability of microaggregates to invade HEp-2 cells, and exposure to anti-MIP-1 immune serum reduced the invasion of the host epithelium. Through protein-protein interaction assays, MIP-1 was found to bind to the host protein filamin A, a cytoskeletal actin-binding protein integral to the modulation of host cell shape and migration. As visualized by immunofluorescence, filamin A was able to co-localize with microaggregates and to a lesser extent planktonic bacteria. Invasion of HEp-2 cells by microaggregates and planktonic bacteria was also inhibited by the addition of anti-filamin A antibody suggesting that filamin A plays an important role during infection. In addition, at earlier time points binding and invasion assay results suggest that MBP-1 participates significantly during the first interactions with the host cell while MIP-1 becomes important once the bacteria adhere to the host epithelium. In summary, we have unveiled one more step associated with MAH crossing the respiratory mucosa.


Assuntos
Células Epiteliais/microbiologia , Mycobacterium avium/metabolismo , Mycobacterium avium/patogenicidade , Animais , Aderência Bacteriana , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Linhagem Celular , Feminino , Filaminas/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Camundongos Endogâmicos C57BL , Infecções por Mycobacterium/microbiologia , Infecções por Mycobacterium/patologia , Mycobacterium avium/genética , Mucosa Respiratória/microbiologia , Infecções Respiratórias/microbiologia , Infecções Respiratórias/patologia
16.
PLoS One ; 10(5): e0128772, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26010725

RESUMO

Mycobacterium avium subsp. hominissuis is an opportunistic pathogen that is associated with biofilm-related infections of the respiratory tract and is difficult to treat. In recent years, extracellular DNA (eDNA) has been found to be a major component of bacterial biofilms, including many pathogens involved in biofilm-associated infections. To date, eDNA has not been described as a component of mycobacterial biofilms. In this study, we identified and characterized eDNA in a high biofilm-producing strain of Mycobacterium avium subsp. hominissuis (MAH). In addition, we surveyed for presence of eDNA in various MAH strains and other nontuberculous mycobacteria. Biofilms of MAH A5 (high biofilm-producing strain) and MAH 104 (reference strain) were established at 22°C and 37°C on abiotic surfaces. Acellular biofilm matrix and supernatant from MAH A5 7 day-old biofilms both possess abundant eDNA, however very little eDNA was found in MAH 104 biofilms. A survey of MAH clinical isolates and other clinically relevant nontuberculous mycobacterial species revealed many species and strains that also produce eDNA. RAPD analysis demonstrated that eDNA resembles genomic DNA. Treatment with DNase I reduced the biomass of MAH A5 biofilms when added upon biofilm formation or to an already established biofilm both on abiotic surfaces and on top of human pharyngeal epithelial cells. Furthermore, co-treatment of an established biofilm with DNase 1 and either moxifloxacin or clarithromycin significantly increased the susceptibility of the bacteria within the biofilm to these clinically used antimicrobials. Collectively, our results describe an additional matrix component of mycobacterial biofilms and a potential new target to help treat biofilm-associated nontuberculous mycobacterial infections.


Assuntos
Biofilmes , DNA Bacteriano/fisiologia , Farmacorresistência Bacteriana , Mycobacterium avium/fisiologia , Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Linhagem Celular , Claritromicina/farmacologia , Desoxirribonuclease I/farmacologia , Farmacorresistência Bacteriana/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/microbiologia , Fluoroquinolonas/farmacologia , Humanos , Moxifloxacina , Mycobacterium avium/efeitos dos fármacos
17.
PLoS One ; 9(9): e108703, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25264757

RESUMO

Pulmonary infections caused by nontuberculous mycobacteria (NTM) are an increasing problem in individuals with chronic lung conditions and current therapies are lacking. We investigated the activity of liposomal amikacin for inhalation (LAI) against NTM in vitro as well as in a murine model of respiratory infection. Macrophage monolayers were infected with three strains of Mycobacterium avium, two strains of Mycobacterium abscessus, and exposed to LAI or free amikacin for 4 days before enumerating bacterial survival. Respiratory infection was established in mice by intranasal inoculation with M. avium and allowing three weeks for the infection to progress. Three different regimens of inhaled LAI were compared to inhaled saline and parenterally administered free amikacin over a 28 day period. Bacteria recovered from the mice were analyzed for acquired resistance to amikacin. In vitro, liposomal amikacin for inhalation was more effective than free amikacin in eliminating both intracellular M. avium and M. abscessus. In vivo, inhaled LAI demonstrated similar effectiveness to a ∼25% higher total dose of parenterally administered amikacin at reducing M. avium in the lungs when compared to inhaled saline. Additionally, there was no acquired resistance to amikacin observed after the treatment regimen. The data suggest that LAI has the potential to be an effective therapy against NTM respiratory infections in humans.


Assuntos
Amicacina/administração & dosagem , Amicacina/uso terapêutico , Antituberculosos/uso terapêutico , Sistemas de Liberação de Medicamentos , Infecções por Mycobacterium não Tuberculosas/tratamento farmacológico , Micobactérias não Tuberculosas/efeitos dos fármacos , Infecções Respiratórias/tratamento farmacológico , Administração por Inalação , Aerossóis , Amicacina/farmacologia , Animais , Antituberculosos/administração & dosagem , Antituberculosos/farmacologia , Linhagem Celular , Contagem de Colônia Microbiana , Modelos Animais de Doenças , Farmacorresistência Bacteriana/efeitos dos fármacos , Feminino , Humanos , Espaço Intracelular/microbiologia , Lipossomos , Pulmão/efeitos dos fármacos , Pulmão/microbiologia , Pulmão/patologia , Camundongos Endogâmicos C57BL , Testes de Sensibilidade Microbiana , Infecções por Mycobacterium não Tuberculosas/microbiologia , Infecções por Mycobacterium não Tuberculosas/patologia , Micobactérias não Tuberculosas/isolamento & purificação , Infecções Respiratórias/microbiologia , Infecções Respiratórias/patologia
18.
mBio ; 5(4)2014 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-25139900

RESUMO

Inhibition of apoptotic death of macrophages by Mycobacterium tuberculosis represents an important mechanism of virulence that results in pathogen survival both in vitro and in vivo. To identify M. tuberculosis virulence determinants involved in the modulation of apoptosis, we previously screened a transposon bank of mutants in human macrophages, and an M. tuberculosis clone with a nonfunctional Rv3354 gene was identified as incompetent to suppress apoptosis. Here, we show that the Rv3354 gene encodes a protein kinase that is secreted within mononuclear phagocytic cells and is required for M. tuberculosis virulence. The Rv3354 effector targets the metalloprotease (JAMM) domain within subunit 5 of the COP9 signalosome (CSN5), resulting in suppression of apoptosis and in the destabilization of CSN function and regulatory cullin-RING ubiquitin E3 enzymatic activity. Our observation suggests that alteration of the metalloprotease activity of CSN by Rv3354 possibly prevents the ubiquitin-dependent proteolysis of M. tuberculosis-secreted proteins. IMPORTANCE : Macrophage protein degradation is regulated by a protein complex called a signalosome. One of the signalosomes associated with activation of ubiquitin and protein labeling for degradation was found to interact with a secreted protein from M. tuberculosis, which binds to the complex and inactivates it. The interference with the ability to inactivate bacterial proteins secreted in the phagocyte cytosol may have crucial importance for bacterial survival within the phagocyte.


Assuntos
Proteínas de Bactérias/metabolismo , Macrófagos/microbiologia , Metaloproteases/metabolismo , Complexos Multiproteicos/metabolismo , Mycobacterium tuberculosis/genética , Peptídeo Hidrolases/metabolismo , Proteínas Quinases/metabolismo , Apoptose , Proteínas de Bactérias/genética , Complexo do Signalossomo COP9 , Linhagem Celular , Interações Hospedeiro-Patógeno , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Complexos Multiproteicos/antagonistas & inibidores , Mycobacterium tuberculosis/enzimologia , Mycobacterium tuberculosis/patogenicidade , Proteínas Quinases/genética , Proteólise , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
19.
Artigo em Inglês | MEDLINE | ID: mdl-25072031

RESUMO

Johne's disease is caused by Mycobacterium avium subsp. paratuberculosis (MAP), which results in serious economic losses worldwide in farmed livestock such as cattle, sheep, and goats. To control this disease, an effective vaccine with minimal adverse effects is needed. In order to identify a live vaccine for Johne's disease, we evaluated eight attenuated mutant strains of MAP using a C57BL/6 mouse model. The persistence of the vaccine candidates was measured at 6, 12, and 18 weeks post vaccination. Only strains 320, 321, and 329 colonized both the liver and spleens up until the 12-week time point. The remaining five mutants showed no survival in those tissues, indicating their complete attenuation in the mouse model. The candidate vaccine strains demonstrated different levels of protection based on colonization of the challenge strain in liver and spleen tissues at 12 and 18 weeks post vaccination. Based on total MAP burden in both tissues at both time points, strain 315 (MAP1566::Tn5370) was the most protective whereas strain 318 (intergenic Tn5367 insertion between MAP0282c and MAP0283c) had the most colonization. Mice vaccinated with an undiluted commercial vaccine preparation displayed the highest bacterial burden as well as enlarged spleens indicative of a strong infection. Selected vaccine strains that showed promise in the mouse model were moved forward into a goat challenge model. The results suggest that the mouse trial, as conducted, may have a relatively poor predictive value for protection in a ruminant host such as goats.


Assuntos
Vacinas Bacterianas/imunologia , Mycobacterium avium subsp. paratuberculosis/imunologia , Paratuberculose/prevenção & controle , Vacinas Atenuadas/imunologia , Animais , Vacinas Bacterianas/administração & dosagem , Bovinos , Modelos Animais de Doenças , Feminino , Fígado/imunologia , Fígado/microbiologia , Fígado/patologia , Camundongos , Mutação , Mycobacterium avium subsp. paratuberculosis/genética , Baço/imunologia , Baço/microbiologia , Baço/patologia , Fatores de Tempo , Vacinação , Vacinas Atenuadas/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA