Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.948
Filter
1.
Ther Adv Respir Dis ; 18: 17534666241282272, 2024.
Article in English | MEDLINE | ID: mdl-39311055

ABSTRACT

BACKGROUND: It remains unclear whether erythrocyte sedimentation rate (ESR) accurately predicts prognosis during treatment and how ESR changes. OBJECTIVES: We aimed to assess the predictive values of ESR as a prognostic factor of Mycobacterium avium complex pulmonary disease (MAC-PD) while on anti-mycobacterial treatment and its changes according to the treatment responses. DESIGN: This study is a retrospective cohort study. METHODS: This study included patients aged 18 years or older who initiated anti-mycobacterial treatment for MAC-PD at Seoul National University Hospital between January 1, 2009 and March 31, 2022. ESR should be measured at least twice, with a minimum interval of 3 months, during the initial 12 months from the commencement of antibiotic treatment. A mixed linear regression and Cox proportional-hazards models were used to analyze repeated ESR data and the association with patient survival. RESULTS: Of a total of 825 patients who initiated antibiotic treatment for MAC-PD, 369 patients were included in the analysis. Increased levels of ESR during the treatment process were associated with a higher risk of mortality (adjusted hazard ratio 1.03; 95% confidence interval, 1.02-1.03) after adjusting age, sex, comorbidities, presence of cavity, acid-fast bacilli smear positivity, and culture conversion at 12 months. During the treatment, ESR at 12 months of treatment significantly decreased compared to baseline ESR in both the culture-converted and not-converted groups, which was categorized based on whether the culture conversion was achieved within the 12 months after treatment initiation. CONCLUSION: ESR predicted mortality during treatment and decreased over time, regardless of treatment outcomes. Our results underscore the importance of administering anti-mycobacterial treatment even in patients who did not achieve a microbiological cure.


Subject(s)
Biomarkers , Blood Sedimentation , Mycobacterium avium , Tuberculosis , Prognosis , Tuberculosis/diagnosis , Tuberculosis/drug therapy , Tuberculosis/microbiology , Retrospective Studies , Cohort Studies , Humans , Male , Female , Middle Aged , Aged , Anti-Bacterial Agents/therapeutic use , Kaplan-Meier Estimate
2.
J Water Health ; 22(9): 1663-1676, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39340379

ABSTRACT

Outdoor decorative fountains usually attract residents to visit. However, opportunistic pathogens (OPs) can proliferate and grow in the stagnant fountain water, posing potential health risks to visitors due to the inhalation of spaying aerosols. In this study, the abundance of selected OPs and associated microbial communities in three large outdoor decorative fountain waters were investigated using quantitative PCR and 16S rRNA sequencing. The results indicated that Mycobacteria avium and Pseudomonas aeruginosa were consistently detected in all decorative fountain waters throughout the year. Redundancy analysis showed that OPs abundance was negatively correlated with water temperature but positively correlated with nutrient concentrations. The gene copy numbers of M. avium varied between 2.4 and 3.9 log10 (gene copies/mL), which were significantly lower than P. aeruginosa by several orders of magnitude, reaching 6.5-7.1 log10 (gene copies/mL) during winter. The analysis of taxonomic composition and prediction of functional potential also revealed pathogenic microorganisms and infectious disease metabolic pathways associated with microbial communities in different decorative fountain waters. This study provided a deeper understanding of the pathogenic conditions of the outdoor decorative fountain water, and future works should focus on accurately assessing the health risks posed by OPs in aerosols.


Subject(s)
Mycobacterium avium , Pseudomonas aeruginosa , RNA, Ribosomal, 16S , Water Microbiology , Pseudomonas aeruginosa/isolation & purification , Pseudomonas aeruginosa/genetics , RNA, Ribosomal, 16S/analysis , Mycobacterium avium/isolation & purification , Mycobacterium avium/genetics , Microbiota , Environmental Monitoring
3.
PLoS One ; 19(9): e0311167, 2024.
Article in English | MEDLINE | ID: mdl-39348373

ABSTRACT

The treatment of Mycobacterium avium infections is still long, complex, and often poorly tolerated, besides emergence of resistances. New active molecules that are more effective and better tolerated are deeply needed. Mefloquine and its enantiomers ((+) Erythro-mefloquine ((+)-EMQ) and (-)-Erythro-mefloquine ((-)-EMQ)) have shown efficacy in both in vitro and in vivo, in a mouse model of M. avium intraveinous infection. However, no study reports aerosol model of infection or combination with gold standard treatment. That was the aim of our study. In an aerosol model of M. avium infection in BALB/c mice, we used five treatment groups as followed: Clarithromycin-Ethambutol-Rifampicin (CLR-EMB-RIF, standard of care, n = 15), CLR-EMB-MFQ (n = 15), CLR-EMB-(+)-EMQ (n = 15), CLR-EMB-(-)-EMQ (n = 15) and an untreated group (n = 25). To evaluate drug efficacy, we sacrificed each month over 3 months, 5 mice from each group. Lung homogenates were diluted and plated for colony forming unit count (CFU) expressed in Log10. At each time point, we found a significant difference between the untreated group and each of the treatment groups (p<0.005). The (+)-EMQ-CLR-EMB group was the group with the lowest CFU count at each time point but never reached statistical significance. The results of each group 3 months after treatment are: (+)-EMQ-CLR-EMB (4.43 ± 0.26), RIF-CLR-EMB (4.83 ± 0.37), (-)-EMQ-CLR-EMB (4.82 ± 0.18), MFQ-CLR-EMB (4.70 ± 0.21). In conclusion, MFQ and its enantiomers appear to be as effective as rifampicin in combination therapy. Further studies are needed to evaluate the ability of these drugs to prevent selection of clarithromycin resistant strains and potential for lung sterilization.


Subject(s)
Disease Models, Animal , Mefloquine , Mice, Inbred BALB C , Mycobacterium avium , Animals , Mefloquine/pharmacology , Mice , Mycobacterium avium/drug effects , Stereoisomerism , Female , Rifampin/pharmacology , Clarithromycin/pharmacology , Antitubercular Agents/pharmacology , Antitubercular Agents/chemistry , Ethambutol/pharmacology , Drug Therapy, Combination , Tuberculosis/drug therapy , Tuberculosis/microbiology , Lung/microbiology , Lung/drug effects , Lung/pathology
4.
EBioMedicine ; 105: 105198, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38889480

ABSTRACT

BACKGROUND: Disease susceptibility and progression of Mycobacterium avium complex pulmonary disease (MAC-PD) is associated with multiple factors, including low body mass index (BMI). However, the specific impact of low BMI on MAC-PD progression remains poorly understood. This study aims to examine the progression of MAC-PD in the context of low BMI, utilising a disease-resistant mouse model. METHODS: We employed a MAC infection-resistant female A/J mouse model to compare the progression of MAC-PD under two dietary conditions: one group was fed a standard protein diet, representing protein-energy unrestricted conditions, and the other was fed a low protein diet (LPD), representing protein-energy restriction. FINDINGS: Our results reveal that protein-energy restriction significantly exacerbates MAC-PD progression by disrupting lipid metabolism. Mice fed an LPD showed elevated fatty acid levels and related gene expressions in lung tissues, similar to findings of increased fatty acids in the serum of patients who exhibited the MAC-PD progression. These mice also exhibited increased CD36 expression and lipid accumulation in macrophages upon MAC infection. In vitro experiments emphasised the crucial role of CD36-mediated palmitic acid uptake in bacterial proliferation. Importantly, in vivo studies demonstrated that administering anti-CD36 antibody to LPD-fed A/J mice reduced macrophage lipid accumulation and impeded bacterial growth, resulting in remarkable slowing disease progression. INTERPRETATION: Our findings indicate that the metabolic status of host immune cells critically influences MAC-PD progression. This study highlights the potential of adequate nutrient intake in preventing MAC-PD progression, suggesting that targeting CD36-mediated pathways might be a host-directed therapeutic strategy to managing MAC infection. FUNDING: This research was funded by the National Research Foundation of Korea, the Korea Research Institute of Bioscience and Biotechnology, and the Korea National Institute of Health.


Subject(s)
Disease Models, Animal , Disease Progression , Lipid Metabolism , Mycobacterium avium-intracellulare Infection , Animals , Female , Mice , Mycobacterium avium-intracellulare Infection/microbiology , Mycobacterium avium-intracellulare Infection/metabolism , CD36 Antigens/metabolism , CD36 Antigens/genetics , Macrophages/metabolism , Humans , Mycobacterium avium Complex , Lung/metabolism , Lung/microbiology , Lung/pathology , Fatty Acids/metabolism , Mycobacterium avium , Disease Susceptibility
5.
Ann Agric Environ Med ; 31(2): 298-301, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38940116

ABSTRACT

Introduction and Objective. Pets infected with zoonotic pathogens might become a source of infections for their owners, especially those who are immuno-compromised. The aim of this report is to describe a case of chronic, untreatable pneumonia in a domestic ferret. Materials and method. The subject was a 5-year-old female ferret suffering from recurrent pneumonia. Ante-mortally, swabs from the nasal cavity, alveolus and throat were collected from the animal. Post-mortally, lesioned organ fragments were collected. Standard microbiological testing was performed. Additionally, mycobacterial diagnosis including culture and molecular tests was performed. Results. The co-infection of Mycobacterium avium and Klebsiella pneumoniae was microbiologically confirmed. Conclusions. This case demonstrates the need to pay attention to the possibility of zoonotic pathogens in ferrets. Veterinarians diagnosing ferrets are potentially exposed to Mycobacteria spp. infections and other pathogens.


Subject(s)
Coinfection , Ferrets , Klebsiella Infections , Klebsiella pneumoniae , Mycobacterium avium , Animals , Ferrets/microbiology , Female , Klebsiella pneumoniae/isolation & purification , Coinfection/veterinary , Coinfection/microbiology , Klebsiella Infections/veterinary , Klebsiella Infections/microbiology , Klebsiella Infections/diagnosis , Mycobacterium avium/isolation & purification , Tuberculosis/veterinary , Tuberculosis/microbiology , Tuberculosis/diagnosis , Fatal Outcome
6.
Microbiol Spectr ; 12(8): e0016724, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-38916320

ABSTRACT

Mycobacterium tuberculosis (Mtb) as well as nontuberculous mycobacteria are intracellular pathogens whose treatment is extensive and increasingly impaired due to the rise of mycobacterial drug resistance. The loss of antibiotic efficacy has raised interest in the identification of host-directed therapeutics (HDT) to develop novel treatment strategies for mycobacterial infections. In this study, we identified amiodarone as a potential HDT candidate that inhibited both intracellular Mtb and Mycobacterium avium in primary human macrophages without directly impairing bacterial growth, thereby confirming that amiodarone acts in a host-mediated manner. Moreover, amiodarone induced the formation of (auto)phagosomes and enhanced autophagic targeting of mycobacteria in macrophages. The induction of autophagy by amiodarone is likely due to enhanced transcriptional regulation, as the nuclear intensity of the transcription factor EB, the master regulator of autophagy and lysosomal biogenesis, was strongly increased. Furthermore, blocking lysosomal degradation with bafilomycin impaired the host-beneficial effect of amiodarone. Finally, amiodarone induced autophagy and reduced bacterial burden in a zebrafish embryo model of tuberculosis, thereby confirming the HDT activity of amiodarone in vivo. In conclusion, we have identified amiodarone as an autophagy-inducing antimycobacterial HDT that improves host control of mycobacterial infections. IMPORTANCE: Due to the global rise in antibiotic resistance, there is a strong need for alternative treatment strategies against intracellular bacterial infections, including Mycobacterium tuberculosis (Mtb) and non-tuberculous mycobacteria. Stimulating host defense mechanisms by host-directed therapy (HDT) is a promising approach for treating mycobacterial infections. This study identified amiodarone, an antiarrhythmic agent, as a potential HDT candidate that inhibits the survival of Mtb and Mycobacterium avium in primary human macrophages. The antimycobacterial effect of amiodarone was confirmed in an in vivo tuberculosis model based on Mycobacterium marinum infection of zebrafish embryos. Furthermore, amiodarone induced autophagy and inhibition of the autophagic flux effectively impaired the host-protective effect of amiodarone, supporting that activation of the host (auto)phagolysosomal pathway is essential for the mechanism of action of amiodarone. In conclusion, we have identified amiodarone as an autophagy-inducing HDT that improves host control of a wide range of mycobacteria.


Subject(s)
Amiodarone , Autophagy , Macrophages , Mycobacterium tuberculosis , Tuberculosis , Zebrafish , Amiodarone/pharmacology , Autophagy/drug effects , Animals , Zebrafish/microbiology , Humans , Macrophages/microbiology , Macrophages/immunology , Macrophages/drug effects , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/growth & development , Tuberculosis/drug therapy , Tuberculosis/microbiology , Disease Models, Animal , Mycobacterium avium/drug effects , Lysosomes/drug effects , Lysosomes/metabolism , Lysosomes/microbiology
7.
PLoS One ; 19(5): e0303516, 2024.
Article in English | MEDLINE | ID: mdl-38728330

ABSTRACT

Increasingly prevalent, nontuberculous mycobacteria (NTM) infections affect approximately 20% of people with cystic fibrosis (CF). Previous studies of CF sputum identified lower levels of the host metabolite itaconate in those infected with NTM. Itaconate can inhibit the growth of M. tuberculosis (MTB) in vitro via the inhibition of the glyoxylate cycle enzyme (ICL), but its impact on NTM is unclear. To test itaconic acid's (IA) effect on NTM growth, laboratory and CF clinical strains of Mycobacterium abscessus and Mycobacterium avium were cultured in 7H9 minimal media supplemented with 1-10 mM of IA and short-chain fatty acids (SCFA). M. avium and M. abscessus grew when supplemented with SCFAs, whereas the addition of IA (≥ 10 mM) completely inhibited NTM growth. NTM supplemented with acetate or propionate and 5 mM IA displayed slower growth than NTM cultured with SCFA and ≤ 1 mM of IA. However, IA's inhibition of NTM was pH dependent; as similar and higher quantities (100 mM) of pH adjusted IA (pH 7) did not inhibit growth in vitro, while in an acidic minimal media (pH 6.1), 1 to 5 mM of non-pH adjusted IA inhibited growth. None of the examined isolates displayed the ability to utilize IA as a carbon source, and IA added to M. abscessus isocitrate lyase (ICL) decreased enzymatic activity. Lastly, the addition of cell-permeable 4-octyl itaconate (4-OI) to THP-1 cells enhanced NTM clearance, demonstrating a potential role for IA/itaconate in host defense against NTM infections.


Subject(s)
Succinates , Succinates/pharmacology , Succinates/metabolism , Humans , Hydrogen-Ion Concentration , Nontuberculous Mycobacteria/drug effects , Nontuberculous Mycobacteria/growth & development , THP-1 Cells , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium avium/drug effects , Mycobacterium avium/growth & development , Mycobacterium abscessus/drug effects , Mycobacterium abscessus/growth & development , Mycobacterium abscessus/metabolism
8.
R I Med J (2013) ; 107(5): 7-10, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38687260

ABSTRACT

CASE: We report a rare case of mycobacterial periprosthetic joint infection (PJI) after primary total knee arthroplasty 14 years earlier. Progressive knee pain over three years with a negative PJI infectious workup led to revision total knee arthroplasty. A surprising result was isolation of Mycobacterium avium from tissue cultures taken at time of revision surgery. After six months of antibiotic treatment, the patient is alive with well- functioning pain-free TKA at over one-year follow-up. CONCLUSION: Periprosthetic joint infection can present acutely or chronically years following total knee arthroplasty. Depending on the infecting organism, patients can present with sepsis, or a more indolent slower course that mimics aseptic loosening. In the absence of positive pre-operative labs and cultures, and based on the Musculoskeletal Infection Society (MSIS) criteria, aseptic loosening is a diagnosis of exclusion. An atypical infectious organism should be considered a possible cause and may require specialized cultures of operative specimens.


Subject(s)
Arthroplasty, Replacement, Knee , Prosthesis-Related Infections , Reoperation , Aged , Female , Humans , Male , Anti-Bacterial Agents/therapeutic use , Arthroplasty, Replacement, Knee/adverse effects , Mycobacterium avium/isolation & purification , Mycobacterium avium-intracellulare Infection/diagnosis , Prosthesis-Related Infections/microbiology , Prosthesis-Related Infections/diagnosis , Prosthesis-Related Infections/drug therapy
9.
Thorax ; 79(8): 778-787, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38508718

ABSTRACT

INTRODUCTION: Novel therapeutic strategies are urgently needed for Mycobacterium avium complex pulmonary disease (MAC-PD). Human mesenchymal stromal cells (MSCs) can directly inhibit MAC growth, but their effect on intracellular bacilli is unknown. We investigated the ability of human MSCs to reduce bacterial replication and inflammation in MAC-infected macrophages and in a murine model of MAC-PD. METHODS: Human monocyte-derived macrophages (MDMs) were infected with M. avium Chester strain and treated with human bone marrow-derived MSCs. Intracellular and extracellular colony-forming units (CFUs) were counted at 72 hours. Six-week-old female balb/c mice were infected by nebulisation of M. avium Chester. Mice were treated with 1×106 intravenous human MSCs or saline control at 21 and 28 days post-infection. Lungs, liver and spleen were harvested 42 days post-infection for bacterial counts. Cytokines were quantified by ELISA. RESULTS: MSCs reduced intracellular bacteria in MDMs over 72 hours (median 35% reduction, p=0.027). MSC treatment increased extracellular concentrations of prostaglandin E2 (PGE2) (median 10.1-fold rise, p=0.002) and reduced tumour necrosis factor-α (median 28% reduction, p=0.025). Blocking MSC PGE2 production by cyclo-oxygenase-2 (COX-2) inhibition with celecoxib abrogated the antimicrobial effect, while this was restored by adding exogenous PGE2. MSC-treated mice had lower pulmonary CFUs (median 18% reduction, p=0.012), but no significant change in spleen or liver CFUs compared with controls. CONCLUSION: MSCs can modulate inflammation and reduce intracellular M. avium growth in human macrophages via COX-2/PGE2 signalling and inhibit pulmonary bacterial replication in a murine model of chronic MAC-PD.


Subject(s)
Disease Models, Animal , Mesenchymal Stem Cells , Mice, Inbred BALB C , Mycobacterium avium-intracellulare Infection , Animals , Mice , Female , Humans , Mycobacterium avium-intracellulare Infection/microbiology , Mycobacterium avium Complex , Mesenchymal Stem Cell Transplantation/methods , Macrophages/microbiology , Dinoprostone/metabolism , Sulfonamides/pharmacology , Mycobacterium avium
11.
Cell Biochem Biophys ; 82(2): 885-894, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38430410

ABSTRACT

To investigate the effects and mechanisms of Mycobacterium avium MAV-5183 protein on apoptosis in mouse Ana-1 macrophages. A pET-21a-MAV-5183 recombinant plasmid was constructed. The recombinant MAV-5183 protein was cloned, expressed, purified, and identified using an anti-His-tagged antibody. Rabbits were immunized to obtain antiserum, and its potency and immunoreactivity were assessed through WB. Mouse Ana-1 macrophages were incubated with varying concentrations of MAV-5183 protein. Flow cytometry, following ANNEXIN V-FITC/PI double staining, detected apoptosis. Western Blot analysis was conducted to identify apoptosis-related molecules Caspase-9/8/3 and vesicle-related molecules ASC, NLRP3, and Cleaved-casp1. ELISA measured TNF-α and IL-6 levels in the culture supernatant. LDH activity and ROS levels were analyzed separately. RT-qPCR measured mRNA levels of Caspase-9/8/3, ASC, NLRP3, Caspase-1, IL-1ß, Bax, MAPK-p38, Bcl-2, TNF-α, and IL-6. MAV-5183 protein was successfully cloned, purified, and identified. In in vitro studies on Ana-1 macrophages, MAV-5183 protein increased the expression of Caspase-9/8/3, ASC, NLRP3 (P < 0.01), induced ROS secretion (P < 0.05), and promoted inflammatory cytokine secretion (TNF-α, IL-6, P < 0.0001); however, it did not significantly affect LDH (P > 0.05). MAV-5183 also induced apoptosis in Ana-1 macrophages (P < 0.05). RT-qPCR results indicated a significant increase in mRNA expression of Caspase-9/8/3, ASC, NLRP3, TNF-α, IL-6, MAPK-p38, and pro-apoptotic factor Bax (P < 0.01), with no significant effect on Bcl-2 and IL-1ß mRNA (P > 0.05). The data indicate that MAV-5183 induces macrophage apoptosis through a caspase-dependent pathway and promotes inflammatory cytokine secretion via ROS.


Subject(s)
Apoptosis , Macrophages , Mycobacterium avium , Animals , Mice , Macrophages/metabolism , Macrophages/cytology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/metabolism , Rabbits , Interleukin-6/metabolism , Interleukin-6/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Cell Line , Recombinant Proteins/pharmacology
12.
Cell Mol Biol (Noisy-le-grand) ; 70(2): 18-23, 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38430046

ABSTRACT

Mycobacterium avium subspecies paratuberculosis (MAP) is the causative agent of paratuberculosis, which is currently prevalent in many parts of Iran and produces severe economic loss. It is hence necessary to identify and isolate the animals infected with this bacterium, so this research aimed to isolate MAP from milk and fecal samples of ELISA-positive animals and determine the molecular identity of isolates. After performing ELISA on 3,700 bovine blood samples, 115 samples of milk and feces were taken from ELISA-positive cattle and were cultured on Herald's egg yolk medium with and without mycobactin-J and then the acid-fastness of positive samples was determined using Ziehl-Neelsen staining. The 16S rRNA-PCR test was performed after DNA extraction to determine the molecular identity of isolates. Primers IS6110 and IS901 were employed to ensure that the isolates were not related to members of M. tuberculosis complex and  M. avium, respectively. Primer IS900 was also used to determine the molecular identity of MAP isolates. Also, expression levels of MAP-related genes (IS900, ISMAP02, F57, MAP2191, MAP4027) were evaluated via qPCR. Finally, positive samples were confirmed based on the Nested-PCR. Results showed that a total of 9 isolates were obtained from the culture of 90 ELISA-positive samples. The results revealed that all grown samples were positive for acid-fastness. The 16S rRNA-PCR test revealed the 543 bp band, which confirms the presence of Mycobacterium in the samples. The PCR test with Primer IS900 generated the 398 bp fragment in the first step and the 298 bp fragment in the second step, indicating the presence of MAP in samples. Also, relative expression analysis revealed that MAP-related genes were significantly higher in ELIZA-positive samples than in negative ones. Based on the study findings, it can be concluded that MAP-infected animals can be identified by ELISA. In addition, mycobacterium can be isolated by culturing the samples on appropriate media and then its molecular identity can be determined by using nested-PCR.


Subject(s)
Paratuberculosis , Animals , Cattle , Paratuberculosis/diagnosis , RNA, Ribosomal, 16S/genetics , Polymerase Chain Reaction , Coloring Agents , Mycobacterium avium , DNA, Bacterial/genetics
15.
J Infect Chemother ; 30(8): 752-756, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38369123

ABSTRACT

BACKGROUND: Mycobacterium avium is associated with pulmonary disease in otherwise healthy adults. Several clarithromycin-refractory cases have been reported, including some cases caused by clarithromycin-susceptible strains. OBJECTIVES: To characterize the reason for the discrepancy between clinical response and antibiotic susceptibility results. METHODS: We conducted population analysis of clarithromycin-tolerant and heteroresistant subpopulations of M. avium cultured in vitro and in homogenates of infected lungs of mice. The minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) were determined for 28 M. avium and two M. kansasii strains. Mice were intranasally infected with M. avium and treated with or without clarithromycin (100 mg/kg) thrice weekly. They were sacrificed on day 35 and the bacteria in lung homogenates were tested for clarithromycin resistance. Population analysis assays were performed based on colony growth on plates containing two-fold dilutions of clarithromycin. RESULTS: The MBC/MIC ratios were ≥8 in all 28 strains of M. avium tested. In the population analysis assay, several colonies were observed on the plates containing clarithromycin concentrations above the MIC (2-64 mg/L). No growth of M. kansasii colonies was observed on the plates containing clarithromycin concentrations ≥2 mg/L. M. avium in the homogenates of infected lungs showed clearer clarithromycin-resistant subpopulations than in vitro, regardless of clarithromycin exposure. CONCLUSION: M. avium shows intrinsic heterogeneous resistance (heteroresistance) to clarithromycin. This may explain the observed discrepancies between clarithromycin susceptibility testing results and clinical response to clarithromycin treatment. Further studies are needed to confirm a link between heteroresistance and clinical outcomes.


Subject(s)
Clarithromycin , Drug Resistance, Bacterial , Microbial Sensitivity Tests , Mycobacterium avium , Clarithromycin/pharmacology , Clarithromycin/therapeutic use , Animals , Mice , Mycobacterium avium/drug effects , Lung/microbiology , Female , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Humans
16.
Antimicrob Agents Chemother ; 68(3): e0091723, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38334410

ABSTRACT

Mycobacterium avium complex (MAC) is a serious disease that is mainly caused by infection with the non-tuberculous mycobacteria (NTM), Mycobacterium avium and Mycobacterium intracellulare. Seven new compounds, designated mavintramycins A-G (1-7), were isolated along with structurally related compounds, including amicetin (9) and plicacetin (10), from the culture broth of Streptomyces sp. OPMA40551 as anti-MAC compounds that were active against M. avium and M. intracellulare. Among them, mavintramycin A showed the most potent and selective inhibition of M. avium and M. intracellulare. Furthermore, mavintramycin A was active against more than 40 clinically isolated M. avium, including multidrug-resistant strains, and inhibited the growth of M. avium in a persistent infection cell model using THP-1 macrophages. Mavintramycin A also exhibited in vivo efficacy in silkworm and mouse infection assays with NTM. An experiment to elucidate its mechanism of action revealed that mavintramycin A inhibits protein synthesis by binding to 23S ribosomal RNA in NTM. Mavintramycin A, with a different chemical structure from those of clinically used agents, is a promising drug candidate for the treatment of MAC infectious disease.


Subject(s)
Communicable Diseases , Mycobacterium avium-intracellulare Infection , Animals , Mice , Mycobacterium avium Complex , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Mycobacterium avium-intracellulare Infection/drug therapy , Mycobacterium avium-intracellulare Infection/microbiology , Mycobacterium avium
17.
Microbiol Spectr ; 12(4): e0233923, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38363108

ABSTRACT

Macrolide antibiotics such as clarithromycin (CLR) and azithromycin are the key drugs used in multidrug therapy for Mycobacterium avium complex (MAC) diseases. For these antibacterial drugs, drug susceptibility has been correlated with clinical response in MAC diseases. We have previously demonstrated the correlation between drug susceptibility and mutations in the 23S rRNA gene, which confers resistance to macrolides. Herein, we developed a rapid detection method using the amplification refractory mutation system (ARMS)-loop-mediated isothermal amplification (LAMP) technique to identify mutations in the 23S rRNA gene of M. avium. We examined the applicability of the ARMS-LAMP method to genomic DNA extracted from six genotypes of M. avium clinical isolates. The M. avium isolates were classified into 21 CLR-resistant and 9 CLR-susceptible strains based on the results of drug susceptibility tests; the 23S rRNA genes of these strains were sequenced and analyzed using the ARMS-LAMP method. Sequence analysis revealed that the 9 CLR-sensitive strains were wild-type strains, whereas the 21 CLR-resistant strains comprised 20 mutant-type strains and one wild-type strain. Using ARMS-LAMP, no amplification from genomic DNAs of the 10 wild-type strains was observed using the mutant-type mismatch primer sets (MTPSs); however, amplification from the 20 mutant-type strain DNAs was observed using the MTPSs. The rapid detection method developed by us integrates ARMS-LAMP with a real-time turbidimeter, which can help determine drug resistance in a few hours. In conclusion, ARMS-LAMP might be a new clinically beneficial technology for rapid detection of mutations.IMPORTANCEMultidrug therapy for pulmonary Mycobacterium avium complex disease is centered on the macrolide antibiotics clarithromycin and azithromycin, and resistance to macrolides is an important prognosticator for clinical aggravation. Therefore, it is important to develop a quick and easy method for detecting resistance to macrolides. Drug resistance is known to be correlated with mutations in macrolide resistance genes. We developed a rapid detection method using amplification refractory mutation system (ARMS)-loop-mediated isothermal amplification (LAMP) to identify a mutation in the 23S rRNA gene, which is a macrolide resistance gene. Furthermore, we examined the applicability of this method using M. avium clinical isolates. The rapid method developed by us for detection of the macrolide resistance gene by integrating ARMS-LAMP and a real-time turbidimeter can help in detection of drug resistance within a few hours. Since this method does not require expensive equipment or special techniques and shows high analytical speed, it would be very useful in clinical practice.


Subject(s)
Anti-Bacterial Agents , Lung Diseases , Molecular Diagnostic Techniques , Nucleic Acid Amplification Techniques , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Macrolides/pharmacology , Macrolides/therapeutic use , Clarithromycin/pharmacology , Mycobacterium avium , Azithromycin , Drug Therapy, Combination , Drug Resistance, Bacterial/genetics , Leprostatic Agents/therapeutic use , Mutation , Mycobacterium avium Complex , Lung Diseases/drug therapy , Microbial Sensitivity Tests
18.
Front Cell Infect Microbiol ; 14: 1335104, 2024.
Article in English | MEDLINE | ID: mdl-38379773

ABSTRACT

Background: The accurate identification of the Mycobacterium tuberculosis complex (MTBC) and different nontuberculous mycobacteria (NTM) species is crucial for the timely diagnosis of NTM infections and for reducing poor prognoses. Nucleotide matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS) has been extensively used for microbial identification with high accuracy and throughput. However, its efficacy for Mycobacterium species identification has been less studied. The objective of this study was to evaluate the performance of nucleotide MALDI-TOF-MS for Mycobacterium species identification. Methods: A total of 933 clinical Mycobacterium isolates were preliminarily identified as NTM by the MPB64 test. These isolates were identified by nucleotide MALDI-TOF-MS and Sanger sequencing. The performance of nucleotide MALDI-TOF MS for identifying various Mycobacterium species was analyzed based on Sanger sequencing as the gold standard. Results: The total correct detection rate of all 933 clinical Mycobacterium isolates using nucleotide MALDI-TOF-MS was 91.64% (855/933), and mixed infections were detected in 18.65% (174/933) of the samples. The correct detection rates for Mycobacterium intracellulare, Mycobacterium abscessus, Mycobacterium kansasii, Mycobacterium avium, MTBC, Mycobacterium gordonae, and Mycobacterium massiliense were 99.32% (585/589), 100% (86/86), 98.46% (64/65), 94.59% (35/37), 100.00% (34/34), 95.65% (22/23), and 100% (19/19), respectively. For the identification of the MTBC, M. intracellulare, M. abscessus, M. kansasii, M. avium, M. gordonae, and M. massiliense, nucleotide MALDI-TOF-MS and Sanger sequencing results were in good agreement (k > 0.7). Conclusion: In conclusion, nucleotide MALDI-TOF-MS is a promising approach for identifying MTBC and the most common clinical NTM species.


Subject(s)
Mycobacterium Infections, Nontuberculous , Mycobacterium abscessus , Mycobacterium , Humans , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Mycobacterium/genetics , Nontuberculous Mycobacteria/genetics , Mycobacterium Infections, Nontuberculous/diagnosis , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium avium
19.
Antimicrob Agents Chemother ; 68(3): e0115723, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38259101

ABSTRACT

Mycobacterium avium complex pulmonary disease is treated with an azithromycin, ethambutol, and rifampicin regimen, with limited efficacy. The role of rifampicin is controversial due to inactivity, adverse effects, and drug interactions. Here, we evaluated the efficacy of clofazimine as a substitute for rifampicin in an intracellular hollow-fiber infection model. THP-1 cells, which are monocytes isolated from peripheral blood from an acute monocytic leukemia patient, were infected with M. avium ATCC 700898 and exposed to a regimen of azithromycin and ethambutol with either rifampicin or clofazimine. Intrapulmonary pharmacokinetic profiles of azithromycin, ethambutol, and rifampicin were simulated. For clofazimine, a steady-state average concentration was targeted. Drug concentrations and bacterial densities were monitored over 21 days. Exposures to azithromycin and ethambutol were 20%-40% lower than targeted but within clinically observed ranges. Clofazimine exposures were 1.7 times higher than targeted. Until day 7, both regimens were able to maintain stasis. Thereafter, regrowth was observed for the rifampicin-containing regimen, while the clofazimine-containing regimen yielded a 2 Log10 colony forming unit (CFU) per mL decrease in bacterial load. The clofazimine regimen also successfully suppressed the emergence of macrolide tolerance. In summary, substitution of rifampicin with clofazimine in the hollow-fiber model improved the antimycobacterial activity of the regimen. Clofazimine-containing regimens merit investigation in clinical trials.


Subject(s)
Lung Diseases , Mycobacterium avium-intracellulare Infection , Humans , Rifampin/pharmacology , Rifampin/therapeutic use , Clofazimine/pharmacology , Clofazimine/therapeutic use , Ethambutol/pharmacology , Ethambutol/therapeutic use , Azithromycin/pharmacology , Mycobacterium avium , Mycobacterium avium-intracellulare Infection/drug therapy , Drug Therapy, Combination , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Mycobacterium avium Complex , Lung Diseases/microbiology
20.
mSphere ; 9(2): e0051823, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38240581

ABSTRACT

Sudapyridine (WX-081) is a structural analog of bedaquiline (BDQ), which shows anti-tuberculosis and non-tuberculous mycobacteria (NTM) activities but, unlike BDQ, did not prolong QT interval in animal model studies. This study evaluated the antibacterial activity of this novel compound against Mycobacterium avium, Mycobacterium abscessus, and Mycobacterium chelonae in vitro and in vivo. The minimum inhibitory concentration (MIC) of WX-081 against three kinds of non-tuberculous mycobacteria (NTM) clinical strains was determined using microplate-based alamarBlue assay (MABA), and the antibacterial activity of WX-081 against NTM in J774A.1 cells and mice was evaluated. MIC ranges of WX-081 against clinical strains of M. avium and M. abscessus were 0.05-0.94 µg/mL, 0.88-7.22 µg/mL (M. abscessus subsp. abscessus), and 0.22-8.67 µg/mL (M. abscessus subsp. massiliense), respectively, which were slightly higher than those of BDQ. For M. avium, M. abscessus, and M. chelonae, WX-081 can reduce the intracellular bacterial load by 0.13-1.18, 0.18-1.50, and 0.17-1.03 log10 colony forming units (CFU)/mL, respectively, in a concentration-dependent manner. WX-081 has bactericidal activity against three NTM species in mice. WX-081 exhibited anti-NTM activity to the same extent as BDQ both in vivo and in vitro. WX-081 is a promising clinical candidate and should be studied further in clinical trials. IMPORTANCE: Due to the rapidly increased cases globally, non-tuberculous mycobacteria (NTM) disease has become a significant public health problem. NTM accounted for 11.57% of all mycobacterial isolates in China, with a high detection rate of Mycobacterium abscessus, Mycobacterium avium, and Mycobacterium chelonae during 2000-2019. Treatment of NTM infection is often challenging, as natural resistance to most antibiotics is quite common among different NTM species. Hence, identifying highly active anti-NTM agents is a priority for potent regimen establishment. The pursuit of new drugs to treat multidrug-resistant tuberculosis may also identify some agents with strong activity against NTM. Sudapyridine (WX-081) is a structural analog of bedaquiline (BDQ), which was developed to retain the anti-tuberculosis efficacy but eliminates the severe side effects of BDQ. This study initially evaluated the antimicrobial activity of this novel compound against M. avium, M. abscessus, and M. chelonae in vitro, in macrophages and mice, respectively.


Subject(s)
Mycobacterium Infections, Nontuberculous , Mycobacterium abscessus , Mycobacterium chelonae , Pyridines , Tuberculosis , Animals , Mice , Mycobacterium avium , Mycobacterium Infections, Nontuberculous/drug therapy , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL