Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.020
Filter
Add more filters

Publication year range
1.
Cell ; 185(11): 1860-1874.e12, 2022 05 26.
Article in English | MEDLINE | ID: mdl-35568033

ABSTRACT

Two mycobacteriophages were administered intravenously to a male with treatment-refractory Mycobacterium abscessus pulmonary infection and severe cystic fibrosis lung disease. The phages were engineered to enhance their capacity to lyse M. abscessus and were selected specifically as the most effective against the subject's bacterial isolate. In the setting of compassionate use, the evidence of phage-induced lysis was observed using molecular and metabolic assays combined with clinical assessments. M. abscessus isolates pre and post-phage treatment demonstrated genetic stability, with a general decline in diversity and no increased resistance to phage or antibiotics. The anti-phage neutralizing antibody titers to one phage increased with time but did not prevent clinical improvement throughout the course of treatment. The subject received lung transplantation on day 379, and systematic culturing of the explanted lung did not detect M. abscessus. This study describes the course and associated markers of a successful phage treatment of M. abscessus in advanced lung disease.


Subject(s)
Bacteriophages , Cystic Fibrosis , Mycobacterium Infections, Nontuberculous , Mycobacterium abscessus , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacteriophages/genetics , Cystic Fibrosis/drug therapy , Humans , Lung , Male , Mycobacterium Infections, Nontuberculous/therapy , Mycobacterium abscessus/physiology
2.
Mol Cell ; 82(17): 3166-3177.e5, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35905736

ABSTRACT

Rifampicin (RIF), the frontline drug against M. tuberculosis, is completely ineffective against M. abscessus, partially due to the presence of an ADP-ribosyltransferase (Arr) that inactivates RIF. Using RNA-seq, we show that exposure of M. abscessus to sublethal doses of RIF and Rifabutin (RBT), a close analog of RIF, results in an ∼25-fold upregulation of Mab_helR in laboratory and clinical isolates. An isogenic deletion in Mab_helR results in RIF/RBT hypersensitivity, and overexpression of Mab_helR confers RIF tolerance in M. tuberculosis. We demonstrate an increased HelR-RNAP association in RIF-exposed bacteria and a MabHelR-mediated dissociation of RNAP from stalled initiation complexes in vitro. Finally, we show that the tip of the PCh-loop of Mab_helR, present in proximity to RIF, is critical for conferring RIF resistance but dispensable for dissociation of stalled RNAP complexes, suggesting that HelR-mediated RIF resistance requires a step in addition to displacement of RIF-stalled RNAP.


Subject(s)
Mycobacterium abscessus , Mycobacterium tuberculosis , Rifamycins , Tuberculosis , DNA-Directed RNA Polymerases/genetics , Drug Resistance, Bacterial/genetics , Humans , Microbial Sensitivity Tests , Mycobacterium tuberculosis/genetics , Rifabutin/pharmacology , Rifampin/pharmacology , Rifamycins/pharmacology , Tuberculosis/microbiology
3.
Proc Natl Acad Sci U S A ; 121(17): e2403206121, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38630725

ABSTRACT

Mycobacterium abscessus is increasingly recognized as the causative agent of chronic pulmonary infections in humans. One of the genes found to be under strong evolutionary pressure during adaptation of M. abscessus to the human lung is embC which encodes an arabinosyltransferase required for the biosynthesis of the cell envelope lipoglycan, lipoarabinomannan (LAM). To assess the impact of patient-derived embC mutations on the physiology and virulence of M. abscessus, mutations were introduced in the isogenic background of M. abscessus ATCC 19977 and the resulting strains probed for phenotypic changes in a variety of in vitro and host cell-based assays relevant to infection. We show that patient-derived mutational variations in EmbC result in an unexpectedly large number of changes in the physiology of M. abscessus, and its interactions with innate immune cells. Not only did the mutants produce previously unknown forms of LAM with a truncated arabinan domain and 3-linked oligomannoside chains, they also displayed significantly altered cording, sliding motility, and biofilm-forming capacities. The mutants further differed from wild-type M. abscessus in their ability to replicate and induce inflammatory responses in human monocyte-derived macrophages and epithelial cells. The fact that different embC mutations were associated with distinct physiologic and pathogenic outcomes indicates that structural alterations in LAM caused by nonsynonymous nucleotide polymorphisms in embC may be a rapid, one-step, way for M. abscessus to generate broad-spectrum diversity beneficial to survival within the heterogeneous and constantly evolving environment of the infected human airway.


Subject(s)
Mycobacterium abscessus , Humans , Bacterial Proteins/genetics , Lipopolysaccharides/chemistry , Mutation
4.
Eur J Immunol ; 54(7): e2350610, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38576227

ABSTRACT

Mycobacterium abscessus is an emerging pathogen that causes chronic pulmonary infection. Treatment is challenging owing in part to our incomplete understanding of M. abscessus virulence mechanisms that enable pathogen persistence, such as the differing pathogenicity of M. abscessus smooth (S) and rough (R) colony morphotype. While R M. abscessus is associated with chronic infection and worse patient outcomes, it is unknown how immune responses to S and R M. abscessus differ in an acute pulmonary infection setting. In this study, immunological outcomes of M. abscessus infection with S and R morphotypes were examined in an immune-competent C3HeB/FeJ murine model. R M. abscessus infection was associated with the rapid production of inflammatory chemokines and recruitment of activated, MHC-II+ Ly6C+ macrophages to lungs and mediastinal LN (mLN). While both S and R M. abscessus increased T helper 1 (Th1) phenotype T cells in the lung, this was markedly delayed in mice infected with S M. abscessus. However, histopathological involvement and bacterial clearance were similar regardless of colony morphotype. These results demonstrate the importance of M. abscessus colony morphotype in shaping the development of pulmonary immune responses to M. abscessus, which further informs our understanding of M. abscessus host-pathogen interactions.


Subject(s)
Adaptive Immunity , Immunity, Innate , Lung , Mycobacterium Infections, Nontuberculous , Mycobacterium abscessus , Animals , Mycobacterium abscessus/immunology , Mice , Mycobacterium Infections, Nontuberculous/immunology , Lung/immunology , Lung/pathology , Lung/microbiology , Disease Models, Animal , Macrophages/immunology , Th1 Cells/immunology , Mice, Inbred C3H , Female
5.
Immunol Rev ; 301(1): 48-61, 2021 05.
Article in English | MEDLINE | ID: mdl-33713043

ABSTRACT

Immunity against different Mycobacteria species targeting the lung requires distinctly different pulmonary immune responses for bacterial clearance. Many parameters of acquired and regulatory immune responses differ quantitatively and qualitatively from immunity during infection with Mycobacteria species. Nontuberculosis Mycobacteria species (NTM) Mycobacterium avium- (M avium), Mycobacterium abscessus-(M abscessus), and the Mycobacteria species Mycobacterium tuberculosis-(Mtb). Herein, we discuss the potential implications of acquired and regulatory immune responses in the context of animal and human studies, as well as future directions for efforts to treat Mycobacteria diseases.


Subject(s)
Mycobacterium abscessus , Mycobacterium tuberculosis , Tuberculosis , Animals , Humans , Mycobacterium avium
6.
J Infect Dis ; 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39189818

ABSTRACT

BACKGROUND: Mycobacterium abscessus complex (MABC), an opportunistic nontuberculous mycobacteria (NTM), can lead to poor clinical outcomes in pulmonary infections. Conflicting data exist on person-to-person transmission of MABC within and across healthcare facilities. To investigate further, a comprehensive retrospective study across five healthcare institutions on the Island of Montréal was undertaken. METHODS: We analyzed the genomes of 221 MABC isolates obtained from 115 individuals (2010-2018) to identify possible links. Genetic similarity, defined as ≤25 single-nucleotide polymorphisms (SNPs), was investigated through a blinded epidemiological inquiry. RESULTS: Bioinformatics analyses identified 28 sequence types (STs), including globally observed dominant circulating clones (DCCs). Further analysis revealed 210 isolate pairs within the SNP threshold. Among these pairs, there was one possible lab contamination where isolates from different patients processed in the same lab differed by only 2 SNPs. There were 37 isolate pairs from patients who had provided specimens from the same hospital; however, epidemiological analysis found no evidence of healthcare-associated person-to-person transmission between these patients. Additionally, pan-genome analysis showed higher discriminatory power than core genome analysis for examining genomic similarity. CONCLUSIONS: Genomics alone is insufficient to establish MABC transmission, particularly considering the genetic similarity and wide distribution of DCCs, although pan-genome analysis has the potential to add further insight. Our findings indicate that MABC infections in Montréal are unlikely attributable to healthcare-associated person-to-person transmission.

7.
J Bacteriol ; 206(3): e0033523, 2024 03 21.
Article in English | MEDLINE | ID: mdl-38319218

ABSTRACT

Mycobacterium abscessus is increasingly recognized for causing infections that are notoriously difficult to treat, owing to its large arsenal of intrinsic antibiotic resistance mechanisms. Tools for the genetic manipulation of the pathogen are critical for enabling a better understanding of M. abscessus biology, pathogenesis, and antibiotic resistance mechanisms. However, existing methods are largely recombination-based, which are relatively inefficient. Meanwhile, CRISPR/Cas9 has revolutionized the field of genome editing including its recent adaptation for use in mycobacteria. In this study, we report a streamlined and efficient method for rapid genetic disruptions in M. abscessus. Harnessing the CRISPR1 loci from Streptococcus thermophilus, we have developed a dual-plasmid workflow that introduces Cas9 and sgRNA cassettes in separate steps but requires no other additional factors to engineer mutations in single genes or multiple genes simultaneously or sequentially using multiple targeting sgRNAs. Importantly, the efficiency of mutant generation is several orders of magnitude higher than reported for homologous recombination-based methods. This work, thus, reports the first application of CRISPR/Cas9 for gene editing in M. abscessus and is an important tool in the arsenal for the genetic manipulation of this human pathogen. IMPORTANCE: Mycobacterium abscessus is an opportunistic pathogen of increasing clinical importance due to its poor clinical outcomes and limited treatment options. Drug discovery and development in this highly antibiotic-resistant species will require further understanding of M. abscessus biology, pathogenesis, and antibiotic resistance mechanisms. However, existing methods for facile genetic engineering are relatively inefficient. This study reports on the first application of CRISPR/Cas9 for gene editing in M. abscessus using a dual-plasmid workflow. We establish that our method is easily programmable, efficient, and versatile for genetic disruptions in M. abscessus. This is a critical advancement to facilitating targeted gene function studies in this emerging pathogen.


Subject(s)
CRISPR-Cas Systems , Mycobacterium abscessus , Humans , RNA, Guide, CRISPR-Cas Systems , Mycobacterium abscessus/genetics , Gene Editing/methods , Plasmids/genetics
8.
Proteomics ; 24(10): e2300332, 2024 May.
Article in English | MEDLINE | ID: mdl-38238893

ABSTRACT

Nontuberculous Mycobacteria (NTM) are a group of emerging bacterial pathogens that have been identified in cystic fibrosis (CF) patients with microbial lung infections. The treatment of NTM infection in CF patients is challenging due to the natural resistance of NTM species to many antibiotics. Mycobacterium abscessus is one of the most common NTM species found in the airways of CF patients. In this study, we characterized the extracellular vesicles (EVs) released by drug-sensitive M. abscessus untreated or treated with clarithromycin (CLR), one of the frontline anti-NTM drugs. Our data show that exposure to CLR increases mycobacterial protein trafficking into EVs as well as the secretion of EVs in culture. Additionally, EVs released by CLR-treated M. abscessus increase M. abscessus resistance to CLR when compared to EVs from untreated M. abscessus. Proteomic analysis further indicates that EVs released by CLR-treated M. abscessus carry an increased level of 50S ribosomal subunits, the target of CLR. Taken together, our results suggest that EVs play an important role in M. abscessus resistance to CLR treatment.


Subject(s)
Anti-Bacterial Agents , Clarithromycin , Drug Resistance, Bacterial , Extracellular Vesicles , Mycobacterium Infections, Nontuberculous , Mycobacterium abscessus , Extracellular Vesicles/metabolism , Extracellular Vesicles/drug effects , Mycobacterium abscessus/drug effects , Mycobacterium abscessus/metabolism , Clarithromycin/pharmacology , Drug Resistance, Bacterial/drug effects , Humans , Anti-Bacterial Agents/pharmacology , Mycobacterium Infections, Nontuberculous/microbiology , Proteomics/methods , Bacterial Proteins/metabolism
9.
J Biol Chem ; 299(8): 104979, 2023 08.
Article in English | MEDLINE | ID: mdl-37390990

ABSTRACT

Mycobacterium abscessus causes severe lung infections. Clinical isolates can have either smooth (S) or rough (R) colony morphotypes; of these, S but not R variants have abundant cell wall glycopeptidolipids (GPL) consisting of a peptidolipid core substituted by a 6-deoxy-α-L-talose (6-dTal) and rhamnose residues. Deletion of gtf1, encoding the 6-dTal transferase, results in the S-to-R transition, mycobacterial cord formation, and increased virulence, underscoring the importance of 6-dTal in infection outcomes. However, since 6-dTal is di-O-acetylated, it is unclear whether the gtf1 mutant phenotypes are related to the loss of the 6-dTal or the result of the absence of acetylation. Here, we addressed whether M. abscessus atf1 and atf2, encoding two putative O-acetyltransferases located within the gpl biosynthetic locus, transfer acetyl groups to 6-dTal. We found deletion of atf1 and/or atf2 did not drastically alter the GPL acetylation profile, suggesting there are additional enzymes with redundant functions. We subsequently identified two paralogs of atf1 and atf2, MAB_1725c and MAB_3448. While deletion of MAB_1725c and MAB_3448 had no effect on GPL acetylation, the triple atf1-atf2-MAB_1725c mutant did not synthetize fully acetylated GPL, and the quadruple mutant was totally devoid of acetylated GPL. Moreover, both triple and quadruple mutants accumulated hyper-methylated GPL. Finally, we show deletion of atf genes resulted in subtle changes in colony morphology but had no effect on M. abscessus internalization by macrophages. Overall, these findings reveal the existence of functionally redundant O-acetyltransferases and suggest that O-acetylation influences the glycan moiety of GPL by deflecting biosynthetic flux in M. abscessus.


Subject(s)
Acetyltransferases , Macrophages , Mycobacterium Infections, Nontuberculous , Mycobacterium abscessus , Humans , Acetyltransferases/genetics , Acetyltransferases/metabolism , Macrophages/microbiology , Mycobacterium abscessus/enzymology , Mycobacterium abscessus/genetics , Mycobacterium Infections, Nontuberculous/microbiology
10.
Emerg Infect Dis ; 30(3): 548-554, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38407146

ABSTRACT

Because epidemiologic and environmental risk factors for nontuberculous mycobacteria (NTM) have been reported only infrequently, little information exists about those factors. The state of Virginia, USA, requires certain ecologic features to be included in reports to the Virginia Department of Health, presenting a unique opportunity to study those variables. We analyzed laboratory reports of Mycobacterium avium complex (MAC) and M. abscessus infections in Virginia during 2021-2023. MAC/M. abscessus was isolated from 6.19/100,000 persons, and 2.37/100,000 persons had MAC/M. abscessus lung disease. M. abscessus accounted for 17.4% and MAC for 82.6% of cases. Saturated vapor pressure was associated with MAC/M. abscessus prevalence (prevalence ratio 1.414, 95% CI 1.011-1.980; p = 0.043). Self-supplied water use was a protective factor (incidence rate ratio 0.304, 95% CI 0.098-0.950; p = 0.041). Our findings suggest that a better understanding of geographic clustering and environmental water exposures could help develop future targeted prevention and control efforts.


Subject(s)
Carbamates , Mycobacterium abscessus , Nontuberculous Mycobacteria , Pyrazines , Pyridines , Virginia/epidemiology , Mycobacterium avium Complex , Water
11.
Antimicrob Agents Chemother ; 68(8): e0064824, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39016592

ABSTRACT

Mycobacteroides abscessus (Mab or Mycobacterium abscessus) is a fast-growing mycobacterium that is ubiquitous in the environment and can cause opportunistic disease in people with lung comorbidity and immunodeficiency. There are no Food and Drug Administration-approved drugs for this disease, and repurposed antibiotics have a poor microbiological response. To address the need for effective new antibiotics, we determined the efficacy of epetraborole (EBO) against three Mab clinical isolates in a mouse model of lung Mab infection. Reduction in lung Mab burden over 4 weeks of treatment was the study end point. EBO was administered orally once daily at doses of 25 and 50 mg/kg, which achieved exposures approximating the once-daily dosing of 250 mg and 500 mg, respectively, in humans. EBO administration led to a gradual reduction in the lung Mab burden. After 4 weeks of treatment, the efficacies of 25 and 50 mg/kg EBO against isolates ATCC 19977 and M9501 were comparable. However, against isolate M9530, 50 mg/kg EBO was more efficacious than 25 mg/kg and comparable with parenteral imipenem, one of the most efficacious antibiotics against Mab. We also undertook a dose-ranging study by evaluating the efficacies of once-daily oral administration of 0.5, 5, 10, 25, and 100 mg/kg EBO against M9501 over 4 weeks. Once-daily oral 100 mg/kg EBO was as effective as twice-daily 100 mg/kg imipenem injection. Our study suggests that EBO could address the unmet need for effective oral treatment options for Mab lung disease, given the high rates of Mab drug resistance and limited tolerable intravenous options.


Subject(s)
Anti-Bacterial Agents , Disease Models, Animal , Mycobacterium Infections, Nontuberculous , Mycobacterium abscessus , Animals , Mice , Mycobacterium abscessus/drug effects , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium Infections, Nontuberculous/microbiology , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Lung/microbiology , Lung/drug effects , Female , Microbial Sensitivity Tests
12.
Antimicrob Agents Chemother ; 68(8): e0152023, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-38990014

ABSTRACT

Mycobacterium abscessus pulmonary infections are increasingly problematic, especially for immunocompromised individuals and those with underlying lung conditions. Currently, there is no reliable standardized treatment, underscoring the need for improved preclinical drug testing. We present a simplified immunosuppressed mouse model using only four injections of cyclophosphamide, which allows for sustained M. abscessus lung burden for up to 16 days. This model proved effective for antibiotic efficacy evaluation, as demonstrated with imipenem or amikacin.


Subject(s)
Amikacin , Anti-Bacterial Agents , Cyclophosphamide , Disease Models, Animal , Mycobacterium Infections, Nontuberculous , Mycobacterium abscessus , Animals , Cyclophosphamide/pharmacology , Mycobacterium abscessus/drug effects , Mice , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium Infections, Nontuberculous/microbiology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Amikacin/pharmacology , Amikacin/therapeutic use , Imipenem/pharmacology , Imipenem/therapeutic use , Lung/microbiology , Lung/drug effects , Immunocompromised Host , Female
13.
Antimicrob Agents Chemother ; 68(8): e0002924, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-38990015

ABSTRACT

Mycobacterium abscessus infections are emerging in cystic fibrosis patients, and treatment success rate in these patients is only 33% due to extreme antibiotic resistance. Thus, new treatment options are essential. An interesting target could be Lsr2, a nucleoid-associated protein involved in mycobacterial virulence. Zafirlukast is a Food and Drug Administration (FDA)-approved drug against asthma that was shown to bind Lsr2. In this study, zafirlukast treatment is shown to reduce M. abscessus growth, with a minimal inhibitory concentration of 16 µM and a bactericidal concentration of 64 µM in replicating bacteria only. As an initial response, DNA condensation, a known stress response of mycobacteria, occurs after 1 h of treatment with zafirlukast. During continued zafirlukast treatment, the morphology of the bacteria alters and the structural integrity of the bacteria is lost. After 4 days of treatment, reduced viability is measured in different culture media, and growth of M. abscessus is reduced in a dose-dependent manner. Using transmission electron microscopy, we demonstrated that the hydrophobic multilayered cell wall and periplasm are disorganized and ribosomes are reduced in size and relocalized. In summary, our data demonstrate that zafirlukast alters the morphology of M. abscessus and is bactericidal at 64 µM. The bactericidal concentration of zafirlukast is relatively high, and it is only effective on replicating bacteria but as zafirlukast is an FDA-approved drug, and currently used as an anti-asthma treatment, it could be an interesting drug to further study in in vivo experiments to determine whether it could be used as an antibiotic for M. abscessus infections.


Subject(s)
Anti-Bacterial Agents , Microbial Sensitivity Tests , Mycobacterium abscessus , Phenylcarbamates , Sulfonamides , Tosyl Compounds , Mycobacterium abscessus/drug effects , Mycobacterium abscessus/genetics , Anti-Bacterial Agents/pharmacology , Tosyl Compounds/pharmacology , Sulfonamides/pharmacology , Phenylcarbamates/pharmacology , Indoles/pharmacology , Humans , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , DNA, Bacterial/genetics
14.
Antimicrob Agents Chemother ; 68(7): e0058524, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38837394

ABSTRACT

Individuals with compromised lung function and immunity are susceptible to developing chronic Mycobacterium abscessus infection. Current treatment recommendations typically involve using one ß-lactam antibiotic in combination with non-ß-lactam antibiotics. However, a recent case study (B. Becken, K. M. Dousa, J. L. Johnson, S. M. Holland, and R. A. Bonomo, Antimicrob Agents Chemother 68:e00319-24, 2024, https://doi.org/10.1128/aac.00319-24) demonstrated successful treatment of chronic M. abscessus lung disease in a child using two ß-lactam antibiotics simultaneously. This commentary reviews the emerging evidence and outstanding questions regarding dual ß-lactam therapy for M. abscessus infections.


Subject(s)
Anti-Bacterial Agents , Lung Diseases , Mycobacterium Infections, Nontuberculous , Mycobacterium abscessus , beta-Lactams , Mycobacterium abscessus/drug effects , Humans , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium Infections, Nontuberculous/microbiology , Anti-Bacterial Agents/therapeutic use , beta-Lactams/therapeutic use , Lung Diseases/drug therapy , Lung Diseases/microbiology , Drug Therapy, Combination , Child
15.
Antimicrob Agents Chemother ; 68(7): e0031924, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38757973

ABSTRACT

Treatment of Mycobacterium abscessus infection presents significant challenges, exacerbated by the emergence of macrolide-resistant strains that necessitate the use of multiple antimicrobials in combination and carry the potential for significant toxic effects. Select dual beta-lactam combinations, with or without beta-lactamase inhibitors, have been shown to be highly active in vitro. Herein, we describe a 6-year-old child with underlying mild bilateral lower lobe cylindrical bronchiectatic lung disease who developed pulmonary Mycobacterium abscessus infection and was treated with a multi-drug regimen including two ß-lactam antibiotics, achieving both early clinical and microbiological cure. This case highlights the potential benefit of dual ß-lactam therapy for the treatment of drug-resistant Mycobacterium abscessus infection.


Subject(s)
Anti-Bacterial Agents , Mycobacterium Infections, Nontuberculous , Mycobacterium abscessus , beta-Lactams , Humans , Mycobacterium abscessus/drug effects , Child , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium Infections, Nontuberculous/microbiology , beta-Lactams/therapeutic use , beta-Lactams/pharmacology , Microbial Sensitivity Tests , Male , Drug Therapy, Combination
16.
Antimicrob Agents Chemother ; 68(5): e0134823, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38572960

ABSTRACT

Mycobacterium abscessus (M. abscessus) inherently displays resistance to most antibiotics, with the underlying drug resistance mechanisms remaining largely unexplored. Efflux pump is believed to play an important role in mediating drug resistance. The current study examined the potential of efflux pump inhibitors to reverse levofloxacin (LFX) resistance in M. abscessus. The reference strain of M. abscessus (ATCC19977) and 60 clinical isolates, including 41 M. abscessus subsp. abscessus and 19 M. abscessus subsp. massilense, were investigated. The drug sensitivity of M. abscessus against LFX alone or in conjunction with efflux pump inhibitors, including verapamil (VP), reserpine (RSP), carbonyl cyanide 3-chlorophenylhydrazone (CCCP), or dicyclohexylcarbodiimide (DCC), were determined by AlarmarBlue microplate assay. Drug-resistant regions of the gyrA and gyrB genes from the drug-resistant strains were sequenced. The transcription level of the efflux pump genes was monitored using qRT-PCR. All the tested strains were resistant to LFX. The drug-resistant regions from the gyrA and gyrB genes showed no mutation associated with LFX resistance. CCCP, DCC, VP, and RSP increased the susceptibility of 93.3% (56/60), 91.7% (55/60), 85% (51/60), and 83.3% (50/60) isolates to LFX by 2 to 32-fold, respectively. Elevated transcription of seven efflux pump genes was observed in isolates with a high reduction in LFX MIC values in the presence of efflux pump inhibitors. Efflux pump inhibitors can improve the antibacterial activity of LFX against M. abscessus in vitro. The overexpression of efflux-related genes in LFX-resistant isolates suggests that efflux pumps are associated with the development of LFX resistance in M. abscessus.


Subject(s)
Anti-Bacterial Agents , Levofloxacin , Microbial Sensitivity Tests , Mycobacterium abscessus , Reserpine , Levofloxacin/pharmacology , Anti-Bacterial Agents/pharmacology , Mycobacterium abscessus/drug effects , Mycobacterium abscessus/genetics , Reserpine/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , DNA Gyrase/genetics , DNA Gyrase/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Drug Resistance, Bacterial/genetics , Humans , Verapamil/pharmacology
17.
Antimicrob Agents Chemother ; 68(2): e0132723, 2024 Feb 07.
Article in English | MEDLINE | ID: mdl-38206003

ABSTRACT

Non-tuberculosis mycobacteria (NTM) can cause severe respiratory infection in patients with underlying pulmonary conditions, and these infections are extremely difficult to treat. In this report, we evaluate a nitric oxide (NO)-releasing prodrug [methyl tris diazeniumdiolate (MD3)] against a panel of NTM clinical isolates and as a treatment for acute and chronic NTM infections in vivo. Its efficacy in inhibiting growth or killing mycobacteria was explored in vitro alongside evaluation of the impact to primary human airway epithelial tissue. Airway epithelial tissues remained viable after exposure at concentrations of MD3 needed to kill mycobacteria, with no inherent toxic effect from drug scaffold after NO liberation. Resistance studies conducted via serial passage with representative Mycobacterium abscessus isolates demonstrated no resistance to MD3. When administered directly into the lung via intra-tracheal administration in mice, MD3 demonstrated significant reduction in M. abscessus bacterial load in both acute and chronic models of M. abscessus lung infection. In summary, MD3 is a promising treatment for complex NTM pulmonary infection, specifically those caused by M. abscessus, and warrants further exploration as a therapeutic.


Subject(s)
Mycobacterium Infections, Nontuberculous , Mycobacterium , Prodrugs , Humans , Animals , Mice , Nitric Oxide , Anti-Bacterial Agents/pharmacology , Prodrugs/pharmacology , Mycobacterium Infections, Nontuberculous/drug therapy , Nontuberculous Mycobacteria , Microbial Sensitivity Tests
18.
Antimicrob Agents Chemother ; 68(5): e0017424, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38557171

ABSTRACT

Mycobacterium abscessus (MAB) infections pose a growing public health threat. Here, we assessed the in vitro activity of the boronic acid-based ß-lactamase inhibitor, vaborbactam, with different ß-lactams against 100 clinical MAB isolates. Enhanced activity was observed with meropenem and ceftaroline with vaborbactam (1- and >4-fold MIC50/90 reduction). CRISPRi-mediated blaMAB gene knockdown showed a fourfold MIC reduction to ceftaroline but not the other ß-lactams. Our findings demonstrate vaborbactam's potential in combination therapy against MAB infections.


Subject(s)
Anti-Bacterial Agents , Boronic Acids , Cefoxitin , Ceftaroline , Cephalosporins , Imipenem , Meropenem , Microbial Sensitivity Tests , Mycobacterium abscessus , Mycobacterium abscessus/drug effects , Meropenem/pharmacology , Boronic Acids/pharmacology , Anti-Bacterial Agents/pharmacology , Cephalosporins/pharmacology , Imipenem/pharmacology , Cefoxitin/pharmacology , Humans , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium Infections, Nontuberculous/microbiology , beta-Lactamase Inhibitors/pharmacology
19.
Antimicrob Agents Chemother ; : e0145623, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38651855

ABSTRACT

Mycobacterium abscessus is an emerging opportunistic pathogen responsible for chronic lung diseases, especially in patients with cystic fibrosis. Treatment failure of M. abscessus infections is primarily associated with intrinsic or acquired antibiotic resistance. However, there is growing evidence that antibiotic tolerance, i.e., the ability of bacteria to transiently survive exposure to bactericidal antibiotics through physiological adaptations, contributes to the relapse of chronic infections and the emergence of acquired drug resistance. Yet, our understanding of the molecular mechanisms that underlie antibiotic tolerance in M. abscessus remains limited. In the present work, a mutant with increased cross-tolerance to the first- and second-line antibiotics cefoxitin and moxifloxacin, respectively, has been isolated by experimental evolution. This mutant harbors a mutation in serB2, a gene involved in L-serine biosynthesis. Metabolic changes caused by this mutation alter the intracellular redox balance to a more reduced state that induces overexpression of the transcriptional regulator WhiB7 during the stationary phase, promoting tolerance through activation of a WhiB7-dependant adaptive stress response. These findings suggest that alteration of amino acid metabolism and, more generally, conditions that trigger whiB7 overexpression, makes M. abscessus more tolerant to antibiotic treatment.

20.
Biochem Biophys Res Commun ; 690: 149249, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38000294

ABSTRACT

The anti-tuberculosis therapeutic bedaquiline (BDQ) is used against Mycobacterium abscessus. In M. abscessus BDQ is only bacteriostatic and less potent compared to M. tuberculosis or M. smegmatis. Here we demonstrate its reduced ATP synthesis inhibition against M. abscessus inside-out vesicles, including the F1FO-ATP synthase. Molecular dynamics simulations and binding free energy calculations highlight the differences in drug-binding of the M. abscessus and M. smegmatis FO-domain at the lagging site, where the drug deploys its mechanistic action, inhibiting ATP synthesis. These data pave the way for improved anti-M. abscessus BDQ analogs.


Subject(s)
Mycobacterium abscessus , Mycobacterium tuberculosis , Antitubercular Agents/pharmacology , Diarylquinolines/pharmacology , Diarylquinolines/metabolism , Mycobacterium tuberculosis/metabolism , Nitric Oxide Synthase/metabolism , Adenosine Triphosphate/metabolism , Microbial Sensitivity Tests
SELECTION OF CITATIONS
SEARCH DETAIL