Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 13(1): 952, 2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-35177649

RESUMO

Prevalence of Mycobacterium abscessus infections is increasing in patients with respiratory comorbidities. After initial colonisation, M. abscessus smooth colony (S) variants can undergo an irreversible genetic switch into highly inflammatory, rough colony (R) variants, often associated with a decline in pulmonary function. Here, we use an adult zebrafish model of chronic infection with R and S variants to study M. abscessus pathogenesis in the context of fully functioning host immunity. We show that infection with an R variant causes an inflammatory immune response that drives necrotic granuloma formation through host TNF signalling, mediated by the tnfa, tnfr1 and tnfr2 gene products. T cell-dependent immunity is stronger against the R variant early in infection, and regulatory T cells associate with R variant granulomas and limit bacterial growth. In comparison, an S variant proliferates to high burdens but appears to be controlled by TNF-dependent innate immunity early during infection, resulting in delayed granuloma formation. Thus, our work demonstrates the applicability of adult zebrafish to model persistent M. abscessus infection, and illustrates differences in the immunopathogenesis induced by R and S variants during granulomatous infection.


Assuntos
Granuloma/imunologia , Infecções por Mycobacterium não Tuberculosas/imunologia , Mycobacterium abscessus/patogenicidade , Infecção Persistente/imunologia , Animais , Animais Geneticamente Modificados , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Granuloma/microbiologia , Granuloma/patologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Inata , Ativação Linfocitária , Infecções por Mycobacterium não Tuberculosas/microbiologia , Infecções por Mycobacterium não Tuberculosas/patologia , Mycobacterium abscessus/genética , Mycobacterium abscessus/imunologia , Infecção Persistente/microbiologia , Infecção Persistente/patologia , Transdução de Sinais/imunologia , Linfócitos T Reguladores/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
2.
Microbiol Res ; 253: 126887, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34628130

RESUMO

Mycobacterium abscessus complex (MABC) infections cause significant morbidity and mortality among patients with chronic lung disease, like cystic fibrosis. MABC exists in smooth (S) and rough (R) morphotypes, but triggers of morphotype switching and associated pathogenicity or antimicrobial susceptibility are poorly understood. We demonstrate that M. abscessus subspecies abscessus (Mab), massiliense (Mms), and bolletii (Mbl) cultured in Middlebrook (MB) broth exhibit S morphotype, whereas the bacteria grown in Luria Bertani (LB) broth adopt the R morphotype, characterized by low glycopeptidolipid (GPL) expression. The components of broth that mediate this selection are complex, with albumin supplementation promoting growth of S morphotype, but not sufficient for complete selection. Consistent with the findings of other groups, R forms of Mab, Mms and Mbl selected by LB broth were internalized in RAW 264.7 macrophages with higher efficiency than S. Intracellular survival of broth-selected organisms was variable and was higher for S Mab, but lower for S Mms and Mbl. It is proposed that growth in R morphotype is induced during stress conditions, such as nutrient poor environments or during inflammation. One key component of inflammation is release of nitric oxide. We demonstrated that a nitric oxide donor (DETA-NONOate) appears to induce growth in an R morphotype, as indicated by reduced GPL expression of Mab. Mab treated with DETA-NONOate also enhanced susceptibility to azithromycin at sub-MIC concentrations. In conclusion, morphotype and macrophage intracellular bacterial load of MABC subspecies can be manipulated by growing the bacteria in different culture conditions. Nitric oxide may also drive morphotype selection and enhanced azithromycin activity against Mab and macrophage killing.


Assuntos
Mycobacterium abscessus , Óxido Nítrico , Virulência , Azitromicina/farmacologia , Meios de Cultivo Condicionados/farmacologia , Farmacorresistência Bacteriana/efeitos dos fármacos , Humanos , Inflamação , Mycobacterium abscessus/citologia , Mycobacterium abscessus/efeitos dos fármacos , Mycobacterium abscessus/patogenicidade , Óxido Nítrico/farmacologia , Virulência/efeitos dos fármacos
3.
Eur J Med Res ; 26(1): 109, 2021 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-34537085

RESUMO

BACKGROUND: Mycobacterium abscessus subspecies massiliense is a non-tuberculous mycobacteriosis and was subdivided from Mycobacterium abscessus in 2006. This article is the first report on nasopharyngitis caused by Mycobacterium abscessus subspecies massiliense. CASE PRESENTATION: A 45-year-old woman had an 18-month history of recurrent nasopharyngitis and presented with pain in the throat. Mycobacterial tissue culture and polymerase chain reaction testing revealed the presence of Mycobacterium abscessus subspecies massiliense in the nasopharyngeal tissue. This patient underwent surgery, followed by multiple rounds of chemotherapy with oral and intravenous antibiotic agents for 16 weeks. She has had no recurrence during the 56 weeks since treatment. CONCLUSION: It is difficult to detect the presence of Mycobacterium abscessus subspecies massiliense in a culture from the swabbing sample. The tissue culture from a biopsy specimen is mandatory for the identification of the species. Currently, no definite treatment policy is available and only empirical treatment is applied. This case is an important for the diagnosis and treatment of this bacterial infection on nasopharynx.


Assuntos
Infecções por Mycobacterium não Tuberculosas/complicações , Mycobacterium abscessus/patogenicidade , Nasofaringite/microbiologia , Antibacterianos/uso terapêutico , Feminino , Humanos , Pessoa de Meia-Idade , Infecções por Mycobacterium não Tuberculosas/microbiologia , Mycobacterium abscessus/isolamento & purificação , Nasofaringite/tratamento farmacológico , Prognóstico
4.
Nat Microbiol ; 6(10): 1279-1288, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34545208

RESUMO

Mycobacterium abscessus, a multidrug-resistant nontuberculous mycobacterium, has emerged as a major pathogen affecting people with cystic fibrosis (CF). Although originally thought to be acquired independently from the environment, most individuals are infected with one of several dominant circulating clones (DCCs), indicating the presence of global transmission networks of M. abscessus. How and when these clones emerged and spread globally is unclear. Here, we use evolutionary analyses of isolates from individuals both with and without CF to reconstruct the population history, spatiotemporal spread and recent transmission networks of the DCCs. We demonstrate synchronous expansion of six unrelated DCCs in the 1960s, a period associated with major changes in CF care and survival. Each of these clones has spread globally as a result of rare intercontinental transmission events. We show that the DCCs, but not environmentally acquired isolates, exhibit a specific smoking-associated mutational signature and that current transmission networks include individuals both with and without CF. We therefore propose that the DCCs initially emerged in non-CF populations but were then amplified and spread through the CF community. While individuals with CF are probably the most permissive host, non-CF individuals continue to play a key role in transmission networks and may facilitate long-distance transmission.


Assuntos
Infecções por Mycobacterium não Tuberculosas/microbiologia , Infecções por Mycobacterium não Tuberculosas/transmissão , Mycobacterium abscessus/isolamento & purificação , Fibrose Cística/microbiologia , Genoma Bacteriano/genética , Saúde Global , Humanos , Pulmão/microbiologia , Mutação , Mycobacterium abscessus/classificação , Mycobacterium abscessus/genética , Mycobacterium abscessus/patogenicidade , Filogenia , Fumantes
5.
Science ; 372(6541)2021 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-33926925

RESUMO

Although almost all mycobacterial species are saprophytic environmental organisms, a few, such as Mycobacterium tuberculosis, have evolved to cause transmissible human infection. By analyzing the recent emergence and spread of the environmental organism M. abscessus through the global cystic fibrosis population, we have defined key, generalizable steps involved in the pathogenic evolution of mycobacteria. We show that epigenetic modifiers, acquired through horizontal gene transfer, cause saltational increases in the pathogenic potential of specific environmental clones. Allopatric parallel evolution during chronic lung infection then promotes rapid increases in virulence through mutations in a discrete gene network; these mutations enhance growth within macrophages but impair fomite survival. As a consequence, we observe constrained pathogenic evolution while person-to-person transmission remains indirect, but postulate accelerated pathogenic adaptation once direct transmission is possible, as observed for M. tuberculosis Our findings indicate how key interventions, such as early treatment and cross-infection control, might restrict the spread of existing mycobacterial pathogens and prevent new, emergent ones.


Assuntos
Doenças Transmissíveis Emergentes/microbiologia , Evolução Molecular , Aptidão Genética , Pulmão/microbiologia , Infecções por Mycobacterium não Tuberculosas/microbiologia , Mycobacterium abscessus/genética , Mycobacterium abscessus/patogenicidade , Pneumonia Bacteriana/microbiologia , Doenças Transmissíveis Emergentes/transmissão , Conjuntos de Dados como Assunto , Epigênese Genética , Transferência Genética Horizontal , Genoma Bacteriano , Humanos , Mutação , Infecções por Mycobacterium não Tuberculosas/transmissão , Pneumonia Bacteriana/transmissão , Virulência/genética
6.
Exp Mol Med ; 53(1): 136-149, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33473145

RESUMO

Infection with rapidly growing nontuberculous mycobacteria is emerging as a global health issue; however, key host factors remain elusive. Here, we investigated the characteristic immune profiles of peripheral blood mononuclear cells (PBMCs) from patients infected with Mycobacteroides abscessus subsp. abscessus (Mabc) and M. abscessus subsp. massiliense (Mmass). Using an integrated analysis of global mRNA and microRNA expression profiles, we found that several inflammatory cytokines/chemokines [interleukin (IL)-1ß, IL-6, C-X-C motif chemokine ligand 2, and C-C motif chemokine ligand 2] and miR-144-3p were significantly upregulated in PBMCs from patients compared with those from healthy controls (HCs). Notably, there was a strong correlation between the expression levels of miR-144-3p and proinflammatory cytokines/chemokines. Similarly, upregulated expression of miR-144-3p and proinflammatory cytokines/chemokines was found in macrophages and lungs from mice after infection with Mabc and Mmass. We showed that the expression of negative regulators of inflammation (SARM1 and TNIP3) was significantly downregulated in PBMCs from the patients, although they were not putative targets of miR-144-3p. Furthermore, overexpression of miR-144-3p led to a marked increase in proinflammatory cytokines/chemokines and promoted bacterial growth in macrophages. Together, our results highlight the importance of miR-144-3p linking to pathological inflammation during M. abscessus infection.


Assuntos
MicroRNAs , Infecções por Mycobacterium não Tuberculosas , Animais , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Proteínas do Domínio Armadillo/genética , Proteínas do Domínio Armadillo/metabolismo , Células Cultivadas , Quimiocina CXCL2/genética , Quimiocina CXCL2/metabolismo , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos Endogâmicos C57BL , MicroRNAs/genética , MicroRNAs/metabolismo , Mycobacterium abscessus/patogenicidade , Infecções por Mycobacterium não Tuberculosas/genética , Infecções por Mycobacterium não Tuberculosas/metabolismo , Infecções por Mycobacterium não Tuberculosas/microbiologia , Infecções por Mycobacterium não Tuberculosas/patologia
7.
Cells ; 9(11)2020 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-33158165

RESUMO

Peptidoglycan (PG) is made of a polymer of disaccharides organized as a three-dimensional mesh-like network connected together by peptidic cross-links. PG is a dynamic structure that is essential for resistance to environmental stressors. Remodeling of PG occurs throughout the bacterial life cycle, particularly during bacterial division and separation into daughter cells. Numerous autolysins with various substrate specificities participate in PG remodeling. Expression of these enzymes must be tightly regulated, as an excess of hydrolytic activity can be detrimental for the bacteria. In non-tuberculous mycobacteria such as Mycobacterium abscessus, the function of PG-modifying enzymes has been poorly investigated. In this study, we characterized the function of the PG amidase, Ami1 from M. abscessus. An ami1 deletion mutant was generated and the phenotypes of the mutant were evaluated with respect to susceptibility to antibiotics and virulence in human macrophages and zebrafish. The capacity of purified Ami1 to hydrolyze muramyl-dipeptide was demonstrated in vitro. In addition, the screening of a 9200 compounds library led to the selection of three compounds inhibiting Ami1 in vitro. We also report the structural characterization of Ami1 which, combined with in silico docking studies, allows us to propose a mode of action for these inhibitors.


Assuntos
Mycobacterium abscessus/enzimologia , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Animais , Cristalografia por Raios X , Modelos Animais de Doenças , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Deleção de Genes , Humanos , Larva/microbiologia , Macrófagos/microbiologia , Testes de Sensibilidade Microbiana , Simulação de Acoplamento Molecular , Infecções por Mycobacterium não Tuberculosas/microbiologia , Mycobacterium abscessus/patogenicidade , Mycobacterium abscessus/ultraestrutura , N-Acetil-Muramil-L-Alanina Amidase/antagonistas & inibidores , Fenótipo , Homologia Estrutural de Proteína , Células THP-1 , Virulência , Peixe-Zebra
8.
Virulence ; 11(1): 1225-1239, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32835604

RESUMO

The global incidence of Mycobacterium abscessus (Mabc), a rapidly growing nontuberculous mycobacterial strain that causes treatment-refractory pulmonary diseases, is increasing. Despite this, the host factors that allow for protection against infection are largely unknown. In this study, we found that sirtuin 3 (SIRT3), a mitochondrial protein deacetylase, plays a critical role in host defense against Mabc infection. Mabc decreased SIRT3 and upregulated mitochondrial oxidative stress in macrophages. SIRT3 deficiency led to increased bacterial loads, histopathological, and mitochondrial damage, and pathological inflammation during Mabc infection. Administration of scavengers of mitochondrial reactive oxygen species significantly decreased the in vivo Mabc burden and excessive inflammation, and induced SIRT3 expression in infected lungs. Notably, SIRT3 agonist (resveratrol) significantly decreased Mabc growth and attenuated inflammation in mice and zebrafishes, indicating the key role for SIRT3 in metazoan host defense. Collectively, these data strongly suggest that SIRT3 is a host-directed therapeutic target against Mabc infection by controlling mitochondrial homeostasis.


Assuntos
Homeostase , Interações Hospedeiro-Patógeno , Mitocôndrias/fisiologia , Infecções por Mycobacterium não Tuberculosas/prevenção & controle , Sirtuína 3/genética , Animais , Regulação da Expressão Gênica , Macrófagos/microbiologia , Macrófagos/fisiologia , Masculino , Camundongos , Mycobacterium abscessus/crescimento & desenvolvimento , Mycobacterium abscessus/patogenicidade , Estresse Oxidativo , Espécies Reativas de Oxigênio , Sirtuína 3/metabolismo , Peixe-Zebra/microbiologia
9.
Int J Mycobacteriol ; 9(1): 48-52, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32474488

RESUMO

Background: Treatment of Mycobacterium abscessus pulmonary disease (PD) is challenging with frequent side effects and uncertain rates of success. Methods: We performed a retrospective review of all patients at our center with at least one respiratory sample positive for M. abscessus between 2014 and 2019. Electronic health records were reviewed to determine factors associated with M. abscessus infection and clinical outcomes. Results: Thirty-seven patients were identified including 24 with cystic fibrosis (CF), 10 with bronchiectasis, two with chronic obstructive PD (COPD), and one with asthma. American Thoracic Society/Infectious Diseases Society of America criteria for nontuberculous mycobacteria PD were met in 21/37 (56.8%) of cases. Evidence of Aspergillus lung disease was noted in 18 (75.0%) CF patients compared with 3 (23.1%) non-CF patients (P = 0.005). Induction therapy for M. abscessus was given to 22/37 (59.5%) patients (18/24 [75%] with CF and 4/13 [30.8%] without CF). Median duration of induction therapy was 6 weeks (range 3-12). Maintenance antibiotic therapy was prescribed to 17/22 (77.3%) of treated patients. Culture conversion was seen in 15/24 (62.5%) of CF patients compared with 3/13 (23.1%) in the non-CF group (P = 0.034). Culture conversion occurred in 10/22 (45.5%) of treated patients compared with 8/15 (53.3%) untreated patients. Three patients (8.1%) died during follow-up: one with CF and two with COPD. Conclusions: Culture conversion following isolation of M. abscessus from respiratory samples not only is more common in CF than in patients without CF but also frequently occurs spontaneously in both groups. Targeted treatment for M. abscessus did not clearly impact rates of culture conversion.


Assuntos
Antibacterianos/uso terapêutico , Pulmão/microbiologia , Infecções por Mycobacterium não Tuberculosas/microbiologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Antibacterianos/farmacologia , Coinfecção/tratamento farmacológico , Coinfecção/microbiologia , Fibrose Cística/complicações , Fibrose Cística/microbiologia , Registros Eletrônicos de Saúde , Feminino , Humanos , Masculino , Testes de Sensibilidade Microbiana , Pessoa de Meia-Idade , Infecções por Mycobacterium não Tuberculosas/tratamento farmacológico , Infecções por Mycobacterium não Tuberculosas/mortalidade , Mycobacterium abscessus/efeitos dos fármacos , Mycobacterium abscessus/patogenicidade , Estudos Retrospectivos , Escarro/microbiologia , Resultado do Tratamento , Adulto Jovem
10.
PLoS One ; 15(4): e0232161, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32339194

RESUMO

BACKGROUND: We aim to investigate the rate of spontaneous sputum conversion and reversion in patients with Mycobacterium abscessus complex (MABC) lung disease. METHODS: Among 241 patients diagnosed with MABC lung disease between July 2012 and December 2018, 126 patients with persistent sputum positivity for ≥ 6 months without treatment were enrolled at a tertiary referral center in South Korea. Patients were subdivided into two groups, depending on whether or not treatment was initiated within 2 years of diagnosis. The rates of spontaneous sputum culture conversion and reversion was investigated in patients who did not receive treatment within 2 years. RESULTS: The mean age of 126 patients was 62.9 years. During a mean follow-up duration of 3.2 years, 33 (26.2%) patients received treatment within 2 years of diagnosis. Among the remaining 93 patients not receiving treatment within 2 years, spontaneous sputum conversion occurred in 24 (25.8%) patients during a mean follow-up duration of 3.7 years after diagnosis. No significant differences were observed in time to conversion between Mycobacterium abscessus and Mycobacterium massiliense lung diseases. The Cox regression analysis showed that malignancy as a comorbid disease and the lower number of lobes involved were independent predictors of spontaneous sputum conversion. After spontaneous sputum conversion, reversion occurred in 27.8% patients at a median of 18.2 months after conversion. CONCLUSIONS: Among patients with MABC lung disease who did not receive treatment for at least 2 years after diagnosis, approximately one-fourth experienced spontaneous conversion. However, not a few patients experienced reversion after spontaneous conversion.


Assuntos
Pneumopatias/microbiologia , Mycobacterium abscessus/patogenicidade , Escarro/microbiologia , Antibacterianos/uso terapêutico , Feminino , Seguimentos , Humanos , Pneumopatias/tratamento farmacológico , Masculino , Pessoa de Meia-Idade , Infecções por Mycobacterium não Tuberculosas/tratamento farmacológico , Infecções por Mycobacterium não Tuberculosas/microbiologia , Mycobacterium abscessus/efeitos dos fármacos , República da Coreia , Estudos Retrospectivos
12.
Transpl Infect Dis ; 22(3): e13278, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32170822

RESUMO

Nontuberculous mycobacteria are ubiquitous in water and soil, and the subset of rapidly growing mycobacteria species can cause severe infections in immunocompromised patients. Solid organ or hematopoietic stem cell transplantation (HSCT) recipients are known to be susceptible to infection by nontuberculous mycobacteria. The nontuberculous mycobacteria species Mycobacterium massiliense (M massiliense) has been classified as a rapidly growing mycobacteria and recognized as a pathogen causing lung and soft tissue infections in humans. However, there have been only a few reported cases of M massiliense infection after solid organ transplantation and HSCT. We herein report another case of M massiliense infection after allogeneic HSCT, which manifested as soft tissue infection, lung infection, and bacteremia.


Assuntos
Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Infecções por Mycobacterium não Tuberculosas/microbiologia , Síndromes Mielodisplásicas/complicações , Adulto , Antibacterianos/uso terapêutico , Bacteriemia , Feminino , Humanos , Masculino , Testes de Sensibilidade Microbiana , Pessoa de Meia-Idade , Infecções por Mycobacterium não Tuberculosas/diagnóstico por imagem , Infecções por Mycobacterium não Tuberculosas/tratamento farmacológico , Mycobacterium abscessus/efeitos dos fármacos , Mycobacterium abscessus/patogenicidade , Estudos Retrospectivos , Tomografia Computadorizada por Raios X
13.
Transpl Infect Dis ; 22(3): e13274, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32129923

RESUMO

BACKGROUND: Mycobacterium abscessus infection has been associated with variable outcomes following lung transplantation. M abscessus comprises three subspecies (M abscessus subsp abscessus, M abscessus subsp massiliense, and M abscessus subsp bolletii). We investigated whether lung transplantation outcome in cystic fibrosis (CF) patients in a single center was related to the M abscessus subspecies and genetic cluster. METHODS: CF patients with chronic M abscessus infection transplanted at Great Ormond Street Hospital between 2004 and 2017 were retrospectively examined. All M abscessus isolates were identified to subspecies level by polymerase chain reaction and sequencing. Genetic cluster was determined by variable number tandem repeat profiling and whole-genome sequencing (WGS), and sequence type inferred from WGS. RESULTS: Thirteen patients with chronic M abscessus infection underwent heart/lung or lung transplantation. Subspecies identification showed n = 1 with M abscessus bolletii, n = 5 with M abscessus massiliense, and n = 7 with M abscessus abscessus infection. Eight (62%) patients (one with M abscessus massiliense and seven with M abscessus abscessus) died post-lung transplant. The patient with M abscessus bolletii and three patients with M abscessus massiliense did well post-transplant. One patient with M abscessus massiliense is receiving ongoing treatment. CONCLUSIONS: Dramatically worse outcomes are observed in patients infected with M abscessus subspecies abscessus, the majority of whom were infected with ST-1 and ST-26 strains. Patients infected with other M abcsessus strains can have acceptable outcomes.


Assuntos
Fibrose Cística/complicações , Transplante de Pulmão/efeitos adversos , Infecções por Mycobacterium não Tuberculosas/microbiologia , Mycobacterium abscessus/classificação , Adolescente , Criança , Fibrose Cística/microbiologia , Fibrose Cística/cirurgia , DNA Bacteriano/genética , Feminino , Humanos , Masculino , Testes de Sensibilidade Microbiana , Infecções por Mycobacterium não Tuberculosas/fisiopatologia , Mycobacterium abscessus/patogenicidade , Avaliação de Processos e Resultados em Cuidados de Saúde , Filogenia , Estudos Retrospectivos , Análise de Sequência de DNA , Sequenciamento Completo do Genoma
14.
BMC Infect Dis ; 19(1): 1034, 2019 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-31805893

RESUMO

BACKGROUND: The incidence of Taralomyces marneffei infection in HIV-infected individuals has been decreasing, whereas its rate is rising among non-HIV immunodeficient persons, particularly patients with anti-interferon-gamma autoantibodies. T. marneffei usually causes invasive and disseminated infections, including fungemia. T. marneffei oro-pharyngo-laryngitis is an unusual manifestation of talaromycosis. CASE PRESENTATION: A 52-year-old Thai woman had been diagnosed anti-IFNÉ£ autoantibodies for 4 years. She had a sore throat, odynophagia, and hoarseness for 3 weeks. She also had febrile symptoms and lost 5 kg in weight. Physical examination revealed marked swelling and hyperemia of both sides of the tonsils, the uvula and palatal arches including a swelling of the epiglottis, and arytenoid. The right tonsillar biopsy exhibited a few intracellular oval and elongated yeast-like organisms with some central transverse septum seen, which subsequently grew a few colonies of T. marneffei on fungal cultures. The patient received amphotericin B deoxycholate 45 mg/dayfor 1 weeks, followed by oral itraconazole 400 mg/day for several months. Her symptoms completely resolved without complication. CONCLUSION: In patients with anti-IFN-É£ autoantibodies, T. marneffei can rarely cause a local infection involving oropharynx and larynx. Fungal culture and pathological examination are warranted for diagnosis T. marneffei oro-pharyngo-laryngitis. This condition requires a long term antifungal therapy.


Assuntos
Antifúngicos/uso terapêutico , Laringite/tratamento farmacológico , Micoses/tratamento farmacológico , Faringite/tratamento farmacológico , Talaromyces/patogenicidade , Anfotericina B/uso terapêutico , Autoanticorpos/sangue , Ácido Desoxicólico/uso terapêutico , Combinação de Medicamentos , Feminino , Humanos , Interferon gama/imunologia , Itraconazol/uso terapêutico , Laringite/microbiologia , Laringite/patologia , Pessoa de Meia-Idade , Infecções por Mycobacterium não Tuberculosas/tratamento farmacológico , Mycobacterium abscessus/patogenicidade , Micoses/etiologia , Micoses/microbiologia , Faringite/microbiologia , Faringite/patologia , Tailândia
15.
PLoS Pathog ; 15(11): e1008069, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31703112

RESUMO

Free-living amoebae are thought to represent an environmental niche in which amoeba-resistant bacteria may evolve towards pathogenicity. To get more insights into factors playing a role for adaptation to intracellular life, we characterized the transcriptomic activities of the emerging pathogen Mycobacterium abscessus in amoeba and murine macrophages (Mϕ) and compared them with the intra-amoebal transcriptome of the closely related, but less pathogenic Mycobacterium chelonae. Data on up-regulated genes in amoeba point to proteins that allow M. abscessus to resist environmental stress and induce defense mechanisms, as well as showing a switch from carbohydrate carbon sources to fatty acid metabolism. For eleven of the most upregulated genes in amoeba and/or Mϕ, we generated individual gene knock-out M. abscessus mutant strains, from which ten were found to be attenuated in amoeba and/or Mϕ in subsequence virulence analyses. Moreover, transfer of two of these genes into the genome of M. chelonae increased the intra-Mϕ survival of the recombinant strain. One knock-out mutant that had the gene encoding Eis N-acetyl transferase protein (MAB_4532c) deleted, was particularly strongly attenuated in Mϕ. Taken together, M. abscessus intra-amoeba and intra-Mϕ transcriptomes revealed the capacity of M. abscessus to adapt to an intracellular lifestyle, with amoeba largely contributing to the enhancement of M. abscessus intra-Mϕ survival.


Assuntos
Amoeba/genética , Macrófagos/metabolismo , Infecções por Mycobacterium não Tuberculosas/genética , Mycobacterium abscessus/patogenicidade , Transcriptoma , Fatores de Virulência/genética , Virulência/genética , Amoeba/crescimento & desenvolvimento , Amoeba/microbiologia , Animais , Proteínas de Bactérias/genética , Macrófagos/microbiologia , Camundongos , Infecções por Mycobacterium não Tuberculosas/microbiologia , Mycobacterium abscessus/genética , Mycobacterium abscessus/isolamento & purificação
16.
Int J Mol Sci ; 20(23)2019 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-31766758

RESUMO

Nontuberculous mycobacteria (NTM) have recently emerged as important pathogens among cystic fibrosis (CF) patients worldwide. Mycobacterium abscessus is becoming the most worrisome NTM in this cohort of patients and recent findings clarified why this pathogen is so prone to this disease. M. abscessus drug therapy takes up to 2 years and its failure causes an accelerated lung function decline. The M. abscessus colonization of lung alveoli begins with smooth strains producing glycopeptidolipids and biofilm, whilst in the invasive infection, "rough" mutants are responsible for the production of trehalose dimycolate, and consequently, cording formation. Human-to-human M. abscessus transmission was demonstrated among geographically separated CF patients by whole-genome sequencing of clinical isolates worldwide. Using a M. abscessus infected CF zebrafish model, it was demonstrated that CFTR (cystic fibrosis transmembrane conductance regulator) dysfunction seems to have a specific role in the immune control of M. abscessus infections only. This pathogen is also intrinsically resistant to many drugs, thanks to its physiology and to the acquisition of new mechanisms of drug resistance. Few new compounds or drug formulations active against M. abscessus are present in preclinical and clinical development, but recently alternative strategies have been investigated, such as phage therapy and the use of ß-lactamase inhibitors.


Assuntos
Doenças Transmissíveis Emergentes , Fibrose Cística , Farmacorresistência Bacteriana Múltipla/imunologia , Infecções por Mycobacterium não Tuberculosas , Mycobacterium abscessus , Alvéolos Pulmonares , Animais , Doenças Transmissíveis Emergentes/epidemiologia , Doenças Transmissíveis Emergentes/imunologia , Doenças Transmissíveis Emergentes/patologia , Fibrose Cística/epidemiologia , Fibrose Cística/imunologia , Fibrose Cística/patologia , Regulador de Condutância Transmembrana em Fibrose Cística/imunologia , Modelos Animais de Doenças , Humanos , Infecções por Mycobacterium não Tuberculosas/epidemiologia , Infecções por Mycobacterium não Tuberculosas/imunologia , Infecções por Mycobacterium não Tuberculosas/patologia , Mycobacterium abscessus/imunologia , Mycobacterium abscessus/patogenicidade , Alvéolos Pulmonares/imunologia , Alvéolos Pulmonares/microbiologia , Peixe-Zebra
17.
Pathog Dis ; 77(5)2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31583400

RESUMO

Non-pigmented rapidly growing mycobacteria (NPRGM) are widely distributed in water, soil and animals. It has been observed an increasing importance of NPRGM related-infections, particularly due to the high antimicrobial resistance. NPRGM have rough and smooth colony phenotypes, and several studies have showed that rough colony variants are more virulent than smooth ones. However, other studies have failed to validate this observation. In this study, we have performed two models, invitro and in vivo, in order to assess the different pathogenicity of these two phenotypes. We used collection and clinical strains of Mycobacteriumabscessus, Mycobacterium fortuitum and Mycobacteriumchelonae. On the invitro model (macrophages), phagocytosis was higher for M. abscessus and M. fortuitum rough colony variant strains when compared to smooth colony variants. However, we did not find differences with colonial variants of M. chelonae. Survival of Galleriamellonella larvae in the experimental model was lower for M. abscessus and M. fortuitum rough colony variants when compared with larvae infected with smooth colony variants. We did not find differences in larvae infected with M. chelonae.Results of our in vivo study correlated well with the experimental model. This fact could have implications on the interpretation of the clinical significance of the NPRGM isolate colonial variants.


Assuntos
Infecções por Mycobacterium não Tuberculosas/microbiologia , Infecções por Mycobacterium não Tuberculosas/patologia , Mycobacterium abscessus/patogenicidade , Mycobacterium chelonae/patogenicidade , Mycobacterium fortuitum/patogenicidade , Fenótipo , Animais , Modelos Animais de Doenças , Larva , Lepidópteros , Macrófagos/imunologia , Macrófagos/microbiologia , Modelos Teóricos , Mycobacterium abscessus/crescimento & desenvolvimento , Mycobacterium chelonae/crescimento & desenvolvimento , Mycobacterium fortuitum/crescimento & desenvolvimento , Fagocitose , Pigmentos Biológicos/análise , Análise de Sobrevida , Virulência
18.
Sci Rep ; 9(1): 8667, 2019 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-31209261

RESUMO

Mycobacteria share with other actinomycetes the ability to produce large quantities of triacylglycerol (TAG), which accumulate as intracytoplasmic lipid inclusions (ILI) also known as lipid droplets (LD). Mycobacterium tuberculosis (M. tb), the etiologic agent of tuberculosis, acquires fatty acids from the human host which are utilized to synthesize TAG, subsequently stored in the form of ILI to meet the carbon and nutrient requirements of the bacterium during long periods of persistence. However, environmental factors governing mycobacterial ILI formation and degradation remain poorly understood. Herein, we demonstrated that in the absence of host cells, carbon excess and nitrogen starvation promote TAG accumulation in the form of ILI in M. smegmatis and M. abscessus, used as surrogate species of M. tb. Based on these findings, we developed a simple and reversible in vitro model to regulate ILI biosynthesis and hydrolysis in mycobacteria. We also showed that TAG formation is tgs1 dependent and that lipolytic enzymes mediate TAG breakdown. Moreover, we confirmed that the nitrogen-deprived and ILI-rich phenotype was associated with an increased tolerance towards several drugs used for treating mycobacterial infections. Importantly, we showed that the presence of ILI substantially enhanced the bacterial burden and granuloma abundance in zebrafish embryos infected with lipid-rich M. abscessus as compared to embryos infected with lipid-poor M. abscessus, suggesting that ILI are actively contributing to mycobacterial virulence and pathogenesis.


Assuntos
Antituberculosos/farmacologia , Infecções por Mycobacterium não Tuberculosas/metabolismo , Mycobacterium abscessus/efeitos dos fármacos , Mycobacterium smegmatis/efeitos dos fármacos , Nitrogênio/deficiência , Triglicerídeos/biossíntese , Animais , Animais Geneticamente Modificados , Carbono/metabolismo , Tolerância a Medicamentos , Embrião não Mamífero , Ácidos Graxos/metabolismo , Humanos , Isoniazida/farmacologia , Ligases/genética , Ligases/metabolismo , Gotículas Lipídicas/metabolismo , Lipólise , Longevidade/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/microbiologia , Infecções por Mycobacterium não Tuberculosas/tratamento farmacológico , Infecções por Mycobacterium não Tuberculosas/microbiologia , Infecções por Mycobacterium não Tuberculosas/mortalidade , Mycobacterium abscessus/metabolismo , Mycobacterium abscessus/patogenicidade , Mycobacterium smegmatis/metabolismo , Mycobacterium smegmatis/patogenicidade , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/patogenicidade , Rifampina/farmacologia , Virulência , Peixe-Zebra
19.
Front Immunol ; 10: 125, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30766538

RESUMO

Mycobacterium abscessus complex (MAB) is a rapidly growing mycobacterium(RGM) whose clinical significance as an emerging human pathogen has been increasing worldwide. It has two types of colony morphology, a smooth (S) type, producing high glycopeptidolipid (GPL) content, and a rough (R) type, which produces low levels of GPLs and is associated with increased virulence. However, the mechanism responsible for their difference in virulence is poorly known. By ultrastructural examination of murine macrophages infected, we found that MAB-R strains could replicate more actively in the macrophage phagosome than the S variants and that they could escape into cytosol via phagosomal rupture. The cytosolic access of MAB-R strains via phagosomal rupture led to enhanced Type I interferon (IFN) production and cell death, which resulted in their cell-to-cell spreading. This behavior can provide an additional niche for the survival of MAB-R strains. In addition, we found that their enhancement of cell death mediated cell spreading are dependent on Type I IFN signaling via comparison of wild-type and IFNAR1 knockout mice. In conclusion, our data indicated that a transition of MAB-S strains into MAB-R variants increased their virulence via enhanced Type I IFN production, which led to enhanced survival in infected macrophage via cell death mediated cell-to-cell spreading. This result provides not only a novel insight into the difference in virulence between MAB-R and -S variants but also hints to their treatment strategy.


Assuntos
Interferon Tipo I/metabolismo , Macrófagos/imunologia , Infecções por Mycobacterium não Tuberculosas/imunologia , Mycobacterium abscessus/fisiologia , Fagossomos/ultraestrutura , Animais , Morte Celular , Linhagem Celular , Humanos , Evasão da Resposta Imune , Macrófagos/microbiologia , Macrófagos/ultraestrutura , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Mycobacterium não Tuberculosas/transmissão , Mycobacterium abscessus/patogenicidade , Fagossomos/microbiologia , Receptor de Interferon alfa e beta/genética , Transdução de Sinais , Especificidade da Espécie , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA