您好!欢迎来到首都医科大学附属北京胸科医院官方网站!

高分SCI文摘

您所在的位置: 首页 >>胸科图书馆 >>信息推送 >>高分SCI文摘 >>2019年

高分SCI文摘

2019年

No.13

来源:tushuguan 发布时间:2019-10-15 浏览次数:
字号: + - 14

Medical Abstracts

Keyword: tuberculosis

1. Nature. 2019 Jul;571(7763):72-78. doi: 10.1038/s41586-019-1315-z. Epub 2019 Jun 19.

Large-scale chemical-genetics yields new M. tuberculosis inhibitor classes.

Johnson EO(1)(2)(3), LaVerriere E(1)(4), Office E(1), Stanley M(1)(5), Meyer

E(1)(6), Kawate T(1)(2)(3), Gomez JE(1), Audette RE(7)(8), Bandyopadhyay N(1),

Betancourt N(9)(10), Delano K(1), Da Silva I(9), Davis J(1)(11), Gallo C(1)(12),

Gardner M(7), Golas AJ(1), Guinn KM(7), Kennedy S(1), Korn R(1), McConnell JA(9),

Moss CE(13)(14), Murphy KC(13), Nietupski RM(1), Papavinasasundaram KG(13),

Pinkham JT(7), Pino PA(9), Proulx MK(13), Ruecker N(9), Song N(9), Thompson

M(1)(15), Trujillo C(9), Wakabayashi S(7), Wallach JB(9), Watson C(1)(16),

Ioerger TR(17), Lander ES(1), Hubbard BK(1), Serrano-Wu MH(1), Ehrt S(9),

Fitzgerald M(1), Rubin EJ(7), Sassetti CM(13), Schnappinger D(9), Hung

DT(18)(19)(20).

Author information:

(1)Broad Institute of MIT and Harvard, Cambridge, MA, USA.

(2)Department of Molecular Biology and Center for Computational and Integrative

Biology, Massachusetts General Hospital, Boston, MA, USA.

(3)Department of Genetics, Harvard Medical School, Boston, MA, USA.

New antibiotics are needed to combat rising levels of resistance, with new

Mycobacterium tuberculosis (Mtb) drugs having the highest priority. However,

conventional whole-cell and biochemical antibiotic screens have failed. Here we

develop a strategy termed PROSPECT (primary screening of strains to prioritize

expanded chemistry and targets), in which we screen compounds against pools of

strains depleted of essential bacterial targets. We engineered strains that

target 474 essential Mtb genes and screened pools of 100-150 strains against

activity-enriched and unbiased compound libraries, probing more than 8.5 million

chemical-genetic interactions. Primary screens identified over tenfold more hits

than screening wild-type Mtb alone, with chemical-genetic interactions providing

immediate, direct target insights. We identified over 40 compounds that target

DNA gyrase, the cell wall, tryptophan, folate biosynthesis and RNA polymerase, as

well as inhibitors that target EfpA. Chemical optimization yielded EfpA

inhibitors with potent wild-type activity, thus demonstrating the ability of

PROSPECT to yield inhibitors against targets that would have eluded conventional

drug discovery.

DOI: 10.1038/s41586-019-1315-z

PMID: 31217586 [Indexed for MEDLINE]

2. Acc Chem Res. 2019 Aug 20;52(8):2340-2348. doi: 10.1021/acs.accounts.9b00275.

Epub 2019 Jul 30.

Harnessing Biological Insight to Accelerate Tuberculosis Drug Discovery.

de Wet TJ(1)(2), Warner DF(1)(3), Mizrahi V(1)(3).

Author information:

(1)SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit and DST/NRF Centre of

Excellence for Biomedical TB Research, Department of Pathology and Institute of

Infectious Disease and Molecular Medicine , University of Cape Town ,

Observatory, Cape Town 7925 , South Africa.

(2)Department of Integrative Biomedical Sciences , University of Cape Town ,

Observatory, Cape Town 7925 , South Africa.

(3)Wellcome Centre for Infectious Disease Research in Africa , University of Cape

Town , Observatory, Cape Town 7925 , South Africa.

Tuberculosis (TB) is the leading cause of mortality globally resulting from an

infectious disease, killing almost 1.6 million people annually and accounting for

approximately 30% of deaths attributed to antimicrobial resistance (AMR). This

despite the widespread administration of a neonatal vaccine, and the availability

of an effective combination drug therapy against the causative agent,

Mycobacterium tuberculosis (Mtb). Instead, TB prevalence worldwide is

characterized by high-burden regions in which co-epidemics, such as HIV, and

social and economic factors, undermine efforts to control TB. These elements

additionally ensure conditions that favor the emergence of drug-resistant Mtb

strains, which further threaten prospects for future TB control. To address this

challenge, significant resources have been invested in developing a TB drug

pipeline, an initiative given impetus by the recent regulatory approval of two

new anti-TB drugs. However, both drugs have been reserved for drug-resistant

disease, and the seeming inevitability of new resistance plus the recognized need

to shorten the duration of chemotherapy demands continual replenishment of the

pipeline with high-quality "hits" with novel mechanisms of action. This

represents a massive challenge, which has been undermined by key gaps in our

understanding of Mtb physiology and metabolism, especially during host infection.

Whereas drug discovery for other bacterial infections can rely on predictive in

vitro assays and animal models, for Mtb, inherent metabolic flexibility and

uncertainties about the nutrients available to infecting bacilli in different

host (micro)environments instead requires educated predictions or demonstrations

of efficacy in animal models of arguable relevance to human disease. Even

microbiological methods for enumeration of viable mycobacterial cells are fraught

with complication. Our research has focused on elucidating those aspects of

mycobacterial metabolism that contribute to the robustness of the bacillus to

host immunological defenses and applied antibiotics and that, possibly, drive the

emergence of drug resistance. This work has identified a handful of metabolic

pathways that appear vulnerable to antibiotic targeting. Those highlighted, here,

include the inter-related functions of pantothenate and coenzyme A biosynthesis

and recycling and nucleotide metabolism-the last of which reinforces our view

that DNA metabolism constitutes an under-explored area for new TB drug

development. Although nonessential functions have traditionally been

deprioritized for antibiotic development, a common theme emerging from this work

is that these very functions might represent attractive targets because of the

potential to cripple mechanisms critical to bacillary survival under stress (for

example, the RelMtb-dependent stringent response) or to adaptability under

unfavorable, potentially lethal, conditions including antibiotic therapy (for

example, DnaE2-dependent SOS mutagenesis). The bar, however, is high:

demonstrating convincingly the likely efficacy of this strategy will require

innovative models of human TB disease. In the concluding section, we focus on the

need for improved techniques to elucidate mycobacterial metabolism during

infection and its impact on disease outcomes. Here, we argue that developments in

other fields suggest the potential to break through this barrier by harnessing

chemical-biology approaches in tandem with the most advanced technologies. As

researchers based in a high-burden country, we are impelled to continue

participating in this important endeavor.

DOI: 10.1021/acs.accounts.9b00275

PMCID: PMC6704484

PMID: 31361123

3. Nat Med. 2019 Jul;25(7):1175. doi: 10.1038/s41591-019-0519-y.

Publisher Correction: IFN-γ-independent immune markers of Mycobacterium

tuberculosis exposure.

Lu LL(1)(2), Smith MT(3), Yu KKQ(3), Luedemann C(2), Suscovich TJ(2), Grace

PS(2), Cain A(2), Yu WH(2)(4), McKitrick TR(5), Lauffenburger D(4), Cummings

RD(5), Mayanja-Kizza H(6), Hawn TR(3), Boom WH(7), Stein CM(7)(8), Fortune

SM(1)(2), Seshadri C(9), Alter G(10).

Author information:

(1)Department of Immunology and Infectious Diseases, Harvard TH Chan School of

Public Health, Boston, MA, USA.

(2)Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.

(3)Department of Medicine, University of Washington, Seattle, WA, USA.

Erratum for

Nat Med. 2019 Jun;25(6):977-987.

In the version of this article originally published, there was an error in the

abstract. The word disease should not have been included in the sentence "These

individuals were highly exposed to Mtb but tested negative disease by IFN-γ

release assay and tuberculin skin test, 'resisting' development of classic LTBI".

The sentence should have been "These individuals were highly exposed to Mtb but

tested negative by IFN-γ release assay and tuberculin skin test, 'resisting'

development of classic LTBI." The error has been corrected in the HTML and PDF

versions of this article.

DOI: 10.1038/s41591-019-0519-y

PMID: 31222179

4. Proc Natl Acad Sci U S A. 2019 Aug 13;116(33):16326-16331. doi:

10.1073/pnas.1820683116. Epub 2019 Jul 31.

Phase separation and clustering of an ABC transporter in Mycobacterium

tuberculosis.

Heinkel F(1)(2), Abraham L(3), Ko M(4), Chao J(3)(4), Bach H(4), Hui LT(2), Li

H(1)(2), Zhu M(1)(2), Ling YM(2), Rogalski JC(1), Scurll J(5), Bui JM(1)(2),

Mayor T(1)(2), Gold MR(3), Chou KC(6), Av-Gay Y(3)(4), McIntosh LP(7)(2)(6),

Gsponer J(7)(2).

Author information:

(1)Michael Smith Laboratories, University of British Columbia, Vancouver, BC,

Canada V6T 1Z4.

(2)Department of Biochemistry and Molecular Biology, University of British

Columbia, Vancouver, BC, Canada V6T 1Z3.

(3)Department of Microbiology and Immunology, University of British Columbia,

Vancouver, BC, Canada V6T 1Z3.

Phase separation drives numerous cellular processes, ranging from the formation

of membrane-less organelles to the cooperative assembly of signaling proteins.

Features such as multivalency and intrinsic disorder that enable condensate

formation are found not only in cytosolic and nuclear proteins, but also in

membrane-associated proteins. The ABC transporter Rv1747, which is important for

Mycobacterium tuberculosis (Mtb) growth in infected hosts, has a cytoplasmic

regulatory module consisting of 2 phosphothreonine-binding Forkhead-associated

domains joined by an intrinsically disordered linker with multiple

phospho-acceptor threonines. Here we demonstrate that the regulatory modules of

Rv1747 and its homolog in Mycobacterium smegmatis form liquid-like condensates as

a function of concentration and phosphorylation. The serine/threonine kinases and

sole phosphatase of Mtb tune phosphorylation-enhanced phase separation and

differentially colocalize with the resulting condensates. The Rv1747 regulatory

module also phase-separates on supported lipid bilayers and forms dynamic foci

when expressed heterologously in live yeast and M. smegmatis cells. Consistent

with these observations, single-molecule localization microscopy reveals that the

endogenous Mtb transporter forms higher-order clusters within the Mycobacterium

membrane. Collectively, these data suggest a key role for phase separation in the

function of these mycobacterial ABC transporters and their regulation via

intracellular signaling.

DOI: 10.1073/pnas.1820683116

PMCID: PMC6697873 [Available on 2020-01-31]

PMID: 31366629

Conflict of interest statement: The authors declare no conflict of interest.

5. Proc Natl Acad Sci U S A. 2019 Aug 6;116(32):15907-15913. doi:

10.1073/pnas.1906606116. Epub 2019 Jul 18.

An essential bifunctional enzyme in Mycobacterium tuberculosis for itaconate

dissimilation and leucine catabolism.

Wang H(1), Fedorov AA(2), Fedorov EV(2), Hunt DM(1), Rodgers A(1), Douglas HL(1),

Garza-Garcia A(1), Bonanno JB(2), Almo SC(2), de Carvalho LPS(3).

Author information:

(1)Mycobacterial Metabolism and Antibiotic Research Laboratory, The Francis Crick

Institute, London NW1 1AT, United Kingdom.

(2)Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY

10461.

(3)Mycobacterial Metabolism and Antibiotic Research Laboratory, The Francis Crick

Institute, London NW1 1AT, United Kingdom; luiz.carvalho@crick.ac.uk.

Mycobacterium tuberculosis (Mtb) is the etiological agent of tuberculosis.

One-fourth of the global population is estimated to be infected with Mtb,

accounting for ∼1.3 million deaths in 2017. As part of the immune response to Mtb

infection, macrophages produce metabolites with the purpose of inhibiting or

killing the bacterial cell. Itaconate is an abundant host metabolite thought to

be both an antimicrobial agent and a modulator of the host inflammatory response.

However, the exact mode of action of itaconate remains unclear. Here, we show

that Mtb has an itaconate dissimilation pathway and that the last enzyme in this

pathway, Rv2498c, also participates in l-leucine catabolism. Our results from

phylogenetic analysis, in vitro enzymatic assays, X-ray crystallography, and in

vivo Mtb experiments, identified Mtb Rv2498c as a bifunctional β-hydroxyacyl-CoA

lyase and that deletion of the rv2498c gene from the Mtb genome resulted in

attenuation in a mouse infection model. Altogether, this report describes an

itaconate resistance mechanism in Mtb and an l-leucine catabolic pathway that

proceeds via an unprecedented (R)-3-hydroxy-3-methylglutaryl-CoA (HMG-CoA)

stereospecific route in nature.

Copyright © 2019 the Author(s). Published by PNAS.

DOI: 10.1073/pnas.1906606116

PMCID: PMC6689899

PMID: 31320588

Conflict of interest statement: The authors declare no conflict of interest.

6. Free Radic Biol Med. 2019 Jul 15;143:232-239. doi:

10.1016/j.freeradbiomed.2019.07.012. [Epub ahead of print]

Molecular mechanism for the activation of the anti-tuberculosis drug isoniazid by

Mn(III): First detection and unequivocal identification of the critical

N-centered isoniazidyl radical and its exact location.

Qin L(1), Huang CH(1), Xu D(1), Xie LN(2), Shao J(1), Mao L(1), Kalyanaraman

B(3), Zhu BZ(4).

Author information:

(1)State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research

Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing,

100085, P. R. China; University of Chinese Academy of Sciences, Beijing, 100049,

P. R. China.

(2)State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research

Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing,

100085, P. R. China; National Institute of Environmental Health, Chinese Center

for Disease Control and Prevention, Beijing, 100021, China.

(3)Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, 53226,

USA.

(4)State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research

Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing,

100085, P. R. China; University of Chinese Academy of Sciences, Beijing, 100049,

P. R. China; Linus Pauling Institute, Oregon State University, Corvallis, OR,

97331, USA. Electronic address: bzhu@rcees.ac.cn.

Isoniazid (INH), the most-widely used anti-tuberculosis drug, has been shown to

be activated by Mn(III) to produce the reactive carbon-centered isonicotinic acyl

radical, which was considered to be responsible for its anti-tuberculosis

activity. However, it is still not clear whether the previously-proposed

N-centered isoniazidyl radical intermediate can be initially produced or not; and

if so, what is its exact location on the hydrazine group, distal- or

proximal-nitrogen? Through complementary applications of ESR spin-trapping and

HPLC/MS methods, here we show that the characteristic and transient N-centered

isoniazidyl radical intermediate can be detected and identified from INH

activation uniquely by Mn(III)Acetate not by Mn(III) pyrophosphate. The exact

location of the radical was found to be at the distal-nitrogen of the hydrazine

group by 15N-isotope-labeling techniques via using 15N-labeled INH.

Diisonicotinyl hydrazine was identified as a new reaction product from

INH/Mn(III). Analogous results were observed with other hydrazides. This study

represents the first detection and unequivocal identification of the initial

N-centered isoniazidyl radical and its exact location. These findings should

provide a new perspective on the molecular mechanism of INH activation, which may

have broad biomedical and toxicological significance for future research for more

efficient hydrazide anti-tuberculosis drugs.

Copyright © 2019. Published by Elsevier Inc.

DOI: 10.1016/j.freeradbiomed.2019.07.012

PMID: 31319159

7. J Antimicrob Chemother. 2019 Jul 1;74(7):1795-1798. doi: 10.1093/jac/dkz150.

Characterization of linezolid-resistance-associated mutations in Mycobacterium

tuberculosis through WGS.

Pi R(1)(2), Liu Q(1)(2), Jiang Q(1)(2), Gao Q(1)(2).

Author information:

(1)Key Laboratory of Medical Molecular Virology of Ministries of Education and

Health, School of Basic Medical Sciences and Shanghai Public Health Clinical

Center, Fudan University, Shanghai, China.

(2)Shenzhen Center for Chronic Disease Control, Shenzhen, China.

OBJECTIVES: Linezolid is becoming an important antibiotic for treating MDR/XDR

TB, but the mutations conferring resistance to linezolid remain inadequately

characterized. Herein, we investigated the linezolid-resistance-associated

mutations on a whole-genome scale through parallel selections of resistant

isolates in vitro.

METHODS: Ten parallel Mycobacterium tuberculosis H37Rv cultures were subjected to

spontaneous mutant selection on 7H11 agar plates containing 2.5mg/L linezolid.

The linezolid resistance of resulting colonies was confirmed by growth on a

second linezolid plate. WGS was then performed to identify mutations associated

with linezolid resistance.

RESULTS: Of 181 colonies appearing on the initial linezolid plates, 154 were

confirmed to be linezolid resistant. WGS showed that 88.3% (136/154) of these

isolates had a T460C mutation in rplC, resulting in a C154R substitution. The

other 18 isolates harboured a single mutation in the rrl gene, with G2814T and

G2270T mutations accounting for 7.8% (12/154) and 3.9% (6/154), respectively.

CONCLUSIONS: No mutations in novel genes were associated with linezolid

resistance in a whole-genome investigation of 154 linezolid-resistant isolates

selected in vitro. These results emphasize that rrl and rplC genes should be the

major targets for molecular detection of linezolid resistance.

© The Author(s) 2019. Published by Oxford University Press on behalf of the

British Society for Antimicrobial Chemotherapy. All rights reserved. For

permissions, please email: journals.permissions@oup.com.

DOI: 10.1093/jac/dkz150

PMID: 31225608

8. Antioxid Redox Signal. 2019 Jul 24. doi: 10.1089/ars.2018.7708. [Epub ahead of print]

Truncated Hemoglobin O Carries an Autokinase Activity and Facilitates Adaptation

of Mycobacterium tuberculosis Under Hypoxia.

Hade MD(1), Sethi D(1), Datta H(1), Singh S(1), Thakur N(1), Chhaya A(2), Dikshit

KL(1)(2).

Author information:

(1)1CSIR-Institute of Microbial Technology, Chandigarh, India.

(2)2Department of Biotechnology, Panjab University, Chandigarh, India.

Aims: Although the human pathogen, Mycobacterium tuberculosis (Mtb), is strictly

aerobic and requires efficient supply of oxygen, it can survive long stretches of

severe hypoxia. The mechanism responsible for this metabolic flexibility is

unknown. We have investigated a novel mechanism by which hemoglobin O (HbO),

operates and supports its host under oxygen stress. Results: We discovered that

the HbO exists in a phospho-bound state in Mtb and remains associated with the

cell membrane under hypoxia. Deoxy-HbO carries an autokinase activity that

disrupts its dimeric assembly into monomer and facilitates its association with

the cell membrane, supporting survival and adaptation of Mtb under low oxygen

conditions. Consistent with these observations, deletion of the glbO gene in

Mycobacterium bovis bacillus Calmette-Guerin, which is identical to the glbO gene

of Mtb, attenuated its survival under hypoxia and complementation of the glbO

gene of Mtb rescued this inhibition, but phosphorylation-deficient mutant did

not. These results demonstrated that autokinase activity of the HbO modulates its

physiological function and plays a vital role in supporting the survival of its

host under hypoxia. Innovation and Conclusion: Our study demonstrates that the

redox-dependent autokinase activity regulates oligomeric state and membrane

association of HbO that generates a reservoir of oxygen in the proximity of

respiratory membranes to sustain viability of Mtb under hypoxia. These results

thus provide a novel insight into the physiological function of the HbO and

demonstrate its pivotal role in supporting the survival and adaptation of Mtb

under hypoxia.

DOI: 10.1089/ars.2018.7708

PMID: 31218881

9. Proc Natl Acad Sci U S A. 2019 Jul 2;116(27):13573-13581. doi:

10.1073/pnas.1900176116. Epub 2019 Jun 19.

CarD contributes to diverse gene expression outcomes throughout the genome of

Mycobacterium tuberculosis.

Zhu DX(1), Garner AL(1), Galburt EA(2), Stallings CL(3).

Author information:

(1)Department of Molecular Microbiology, Washington University School of

Medicine, St. Louis, MO 63110.

(2)Department of Biochemistry and Molecular Biophysics, Washington University

School of Medicine, St. Louis, MO 63110.

(3)Department of Molecular Microbiology, Washington University School of

Medicine, St. Louis, MO 63110; stallings@wustl.edu.

The ability to regulate gene expression through transcription initiation

underlies the adaptability and survival of all bacteria. Recent work has revealed

that the transcription machinery in many bacteria diverges from the paradigm that

has been established in Escherichia coli Mycobacterium tuberculosis (Mtb) encodes

the RNA polymerase (RNAP)-binding protein CarD, which is absent in E. coli but is

required to form stable RNAP-promoter open complexes (RPo) and is essential for

viability in Mtb The stabilization of RPo by CarD has been proposed to result in

activation of gene expression; however, CarD has only been examined on limited

promoters that do not represent the typical promoter structure in Mtb In this

study, we investigate the outcome of CarD activity on gene expression from Mtb

promoters genome-wide by performing RNA sequencing on a panel of mutants that

differentially affect CarD's ability to stabilize RPo In all CarD mutants, the

majority of Mtb protein encoding transcripts were differentially expressed,

demonstrating that CarD had a global effect on gene expression. Contrary to the

expected role of CarD as a transcriptional activator, mutation of CarD led to

both up- and down-regulation of gene expression, suggesting that CarD can also

act as a transcriptional repressor. Furthermore, we present evidence that

stabilization of RPo by CarD could lead to transcriptional repression by

inhibiting promoter escape, and the outcome of CarD activity is dependent on the

intrinsic kinetic properties of a given promoter region. Collectively, our data

support CarD's genome-wide role of regulating diverse transcription outcomes.

DOI: 10.1073/pnas.1900176116

PMCID: PMC6613185 [Available on 2019-12-19]

PMID: 31217290

Conflict of interest statement: The authors declare no conflict of interest.

10. Nucleic Acids Res. 2019 Jul 26;47(13):6685-6698. doi: 10.1093/nar/gkz449.

CarD and RbpA modify the kinetics of initial transcription and slow promoter

escape of the Mycobacterium tuberculosis RNA polymerase.

Jensen D(1), Manzano AR(1), Rammohan J(1), Stallings CL(2), Galburt EA(1).

Author information:

(1)Department of Biochemistry and Molecular Biophysics, Washington University

School of Medicine, St. Louis, MO 63110, USA.

(2)Department of Molecular Microbiology, Washington University School of

Medicine, St. Louis, MO 63110, USA.

The pathogen Mycobacterium tuberculosis (Mtb), the causative agent of

tuberculosis, enacts unique transcriptional regulatory mechanisms when subjected

to host-derived stresses. Initiation of transcription by the Mycobacterial RNA

polymerase (RNAP) has previously been shown to exhibit different open complex

kinetics and stabilities relative to Escherichia coli (Eco) RNAP. However,

transcription initiation rates also depend on the kinetics following open complex

formation such as initial nucleotide incorporation and subsequent promoter

escape. Here, using a real-time fluorescence assay, we present the first in-depth

kinetic analysis of initial transcription and promoter escape for the Mtb RNAP.

We show that in relation to Eco RNAP, Mtb displays slower initial nucleotide

incorporation but faster overall promoter escape kinetics on the Mtb rrnAP3

promoter. Furthermore, in the context of the essential transcription factors CarD

and RbpA, Mtb promoter escape is slowed via differential effects on initially

transcribing complexes. Finally, based on their ability to increase the rate of

open complex formation and decrease the rate of promoter escape, we suggest that

CarD and RbpA are capable of activation or repression depending on the

rate-limiting step of a given promoter's basal initiation kinetics.

© The Author(s) 2019. Published by Oxford University Press on behalf of Nucleic

Acids Research.

DOI: 10.1093/nar/gkz449

PMCID: PMC6648326

PMID: 31127308

11. Eur Respir J. 2019 Jul 11;54(1). pii: 1800353. doi: 10.1183/13993003.00353-2018.

Print 2019 Jul.

Analysis of loss to follow-up in 4099 multidrug-resistant pulmonary tuberculosis

patients.

Walker IF(1), Shi O(2)(3), Hicks JP(4), Elsey H(4), Wei X(2), Menzies D(5), Lan

Z(5), Falzon D(6), Migliori GB(7), Pérez-Guzmán C(8)(9), Vargas MH(9)(10),

García-García L(11), Sifuentes Osornio J(12), Ponce-De-León A(13), van der Walt

M(14), Newell JN(4).

Author information:

(1)Nuffield Centre for International Health and Development, University of Leeds,

Leeds, UK i.walker@leeds.ac.uk.

(2)Dalla Lana School of Public Health, University of Toronto, Toronto, ON,

Canada.

(3)Shenzhen Second People's Hospital, Shenzhen University, Shenzhen, China.

Loss to follow-up (LFU) of ≥2 consecutive months contributes to the poor levels

of treatment success in multidrug-resistant tuberculosis (MDR-TB) reported by TB

programmes. We explored the timing of when LFU occurs by month of MDR-TB

treatment and identified patient-level risk factors associated with LFU.We

analysed a dataset of individual MDR-TB patient data (4099 patients from 22

countries). We used Kaplan-Meier survival curves to plot time to LFU and a Cox

proportional hazards model to explore the association of potential risk factors

with LFU.Around one-sixth (n=702) of patients were recorded as LFU. Median

(interquartile range) time to LFU was 7 (3-11)?months. The majority of LFU

occurred in the initial phase of treatment (75% in the first 11?months). Major

risk factors associated with LFU were: age 36-50?years (HR 1.3, 95% CI 1.0-1.6;

p=0.04) compared with age 0-25?years, being HIV positive (HR 1.8, 95% CI 1.2-2.7;

p<0.01) compared with HIV negative, on an individualised treatment regimen (HR

0.7, 95% CI 0.6-1.0; p=0.03) compared with a standardised regimen and a recorded

serious adverse event (HR 0.5, 95% CI 0.4-0.6; p<0.01) compared with no serious

adverse event.Both patient- and regimen-related factors were associated with LFU,

which may guide interventions to improve treatment adherence, particularly in the

first 11?months.

Copyright ©ERS 2019.

DOI: 10.1183/13993003.00353-2018

PMID: 31073080

12. Thorax. 2019 Jul;74(7):675-683. doi: 10.1136/thoraxjnl-2018-212529. Epub 2019 Apr29.

Urban airborne particle exposure impairs human lung and blood Mycobacterium

tuberculosis immunity.

Torres M(1), Carranza C(1), Sarkar S(2), Gonzalez Y(1), Osornio Vargas A(3),

Black K(4), Meng Q(2), Quintana-Belmares R(5), Hernandez M(6), Angeles Garcia

JJF(6), Páramo-Figueroa VH(6), Iñiguez-Garcia MA(7), Flores JL(8), Zhang JJ(9),

Gardner CR(10), Ohman-Strickland P(11), Schwander S(12).

Author information:

(1)Department of Microbiology, Instituto Nacional de Enfermedades Respiratorias,

Mexico City, Mexico.

(2)Environmental and Occupational Health, Rutgers School of Public Health New

Brunswick Campus, Piscataway, New Jersey, USA.

(3)Paediatrics, University of Alberta, Edmonton, Alberta, Canada.

RATIONALE: Associations between urban (outdoor) airborne particulate matter (PM)

exposure and TB and potential biological mechanisms are poorly explored.

OBJECTIVES: To examine whether in vivo exposure to urban outdoor PM in Mexico

City and in vitro exposure to urban outdoor PM2.5 (< 2.5µm median aerodynamic

diameter) alters human host immune cell responses to Mycobacterium tuberculosis.

METHODS: Cellular toxicity (flow cytometry, proliferation assay (MTS assay)), M.

tuberculosis and PM2.5 phagocytosis (microscopy), cytokine-producing cells

(Enzyme-linked immune absorbent spot (ELISPOT)), and signalling pathway markers

(western blot) were examined in bronchoalveolar cells (BAC) and peripheral blood

mononuclear cells (PBMC) from healthy, non-smoking, residents of Mexico City

(n=35; 13 female, 22 male). In vivo-acquired PM burden in alveolar macrophages

(AM) was measured by digital image analysis.

MEASUREMENTS AND MAIN RESULTS: In vitro exposure of AM to PM2.5 did not affect M.

tuberculosis phagocytosis. High in vivo-acquired AM PM burden reduced

constitutive, M. tuberculosis and PM-induced interleukin-1β production in freshly

isolated BAC but not in autologous PBMC while it reduced constitutive production

of tumour necrosis factor-alpha in both BAC and PBMC. Further, PM burden was

positively correlated with constitutive, PM, M. tuberculosis and purified protein

derivative (PPD)-induced interferon gamma (IFN-γ) in BAC, and negatively

correlated with PPD-induced IFN-γ in PBMC.

CONCLUSIONS: Inhalation exposure to urban air pollution PM impairs important

components of the protective human lung and systemic immune response against M.

tuberculosis. PM load in AM is correlated with altered M. tuberculosis-induced

cytokine production in the lung and systemic compartments. Chronic PM exposure

with high constitutive expression of proinflammatory cytokines results in

relative cellular unresponsiveness.

© Author(s) (or their employer(s)) 2019. No commercial re-use. See rights and

permissions. Published by BMJ.

DOI: 10.1136/thoraxjnl-2018-212529

PMID: 31036772

Conflict of interest statement: Competing interests: None declared.

13. Chemistry. 2019 Jul 2;25(37):8894-8902. doi: 10.1002/chem.201901640. Epub 2019

Jun 6.

Synthesis of New Cyclomarin Derivatives and Their Biological Evaluation towards

Mycobacterium Tuberculosis and Plasmodium Falciparum.

Kiefer A(1), Bader CD(2), Held J(3), Esser A(4), Rybniker J(5), Empting M(6),

Müller R(2)(7), Kazmaier U(1).

Author information:

(1)Organic Chemistry, Saarland University, Campus C4.2, 66123, Saarbrücken,

Germany.

(2)Department Microbial Natural Products (MINS), Helmholtz-Institute for

Pharmaceutical Research Saarland (HIPS)-Helmholtz Centre for Infection Research

(HZI), Campus E8.1, 66123, Saarbrücken, Germany.

(3)Department of Tropical Medicine, University of Tübingen, Wilhelmstraße 27,

72074, Tübingen, Germany.

Cyclomarins are highly potent antimycobacterial and antiplasmodial cyclopeptides

isolated from a marine bacterium (Streptomyces sp.). Previous studies have

identified the target proteins and elucidated a novel mode of action, however

there are currently only a few studies examining the structure-activity

relationship (SAR) for both pathogens. Herein, we report the synthesis and

biological evaluation of 17 novel desoxycyclomarin-inspired analogues.

Optimization via side chain modifications of the non-canonical amino acids led to

potent lead structures for each pathogen.

© 2019 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim.

DOI: 10.1002/chem.201901640

PMID: 31012978 [Indexed for MEDLINE]

14. Bioinformatics. 2019 Jul 1;35(13):2276-2282. doi: 10.1093/ bioinformatics/ bty949.

Application of machine learning techniques to tuberculosis drug resistance

analysis.

Kouchaki S(1), Yang Y(1), Walker TM(2)(3), Sarah Walker A(2)(3)(4), Wilson DJ(5),

Peto TEA(2)(3), Crook DW(2)(3)(6); CRyPTIC Consortium, Clifton DA(1).

Author information:

(1)Department of Engineering Science, Institute of Biomedical Engineering.

(2)Nuffield Department of Medicine, University of Oxford.

(3)National Institute of Health Research Oxford Biomedical Research Centre, John

Radcliffe Hospital, Oxford, UK.

(4)Medical Research Council Clinical Trials Unit, University College London, UK.

(5)Nuffield Department of Population Health, Big Data Institute, University of

Oxford, Li Ka Shing Centre for Health Information and Discovery, Oxford, UK.

(6)National Infection Service, Public Health England, Colindale, London, UK.

MOTIVATION: Timely identification of Mycobacterium tuberculosis (MTB) resistance

to existing drugs is vital to decrease mortality and prevent the amplification of

existing antibiotic resistance. Machine learning methods have been widely applied

for timely predicting resistance of MTB given a specific drug and identifying

resistance markers. However, they have been not validated on a large cohort of

MTB samples from multi-centers across the world in terms of resistance prediction

and resistance marker identification. Several machine learning classifiers and

linear dimension reduction techniques were developed and compared for a cohort of

13 402 isolates collected from 16 countries across 6 continents and tested 11

drugs.

RESULTS: Compared to conventional molecular diagnostic test, area under curve of

the best machine learning classifier increased for all drugs especially by

23.11%, 15.22% and 10.14% for pyrazinamide, ciprofloxacin and ofloxacin,

respectively (P<0.01). Logistic regression and gradient tree boosting found to

perform better than other techniques. Moreover, logistic regression/gradient tree

boosting with a sparse principal component analysis/non-negative matrix

factorization step compared with the classifier alone enhanced the best

performance in terms of F1-score by 12.54%, 4.61%, 7.45% and 9.58% for amikacin,

moxifloxacin, ofloxacin and capreomycin, respectively, as well increasing area

under curve for amikacin and capreomycin. Results provided a comprehensive

comparison of various techniques and confirmed the application of machine

learning for better prediction of the large diverse tuberculosis data.

Furthermore, mutation ranking showed the possibility of finding new

resistance/susceptible markers.

AVAILABILITY AND IMPLEMENTATION: The source code can be found at

http://www.robots.ox.ac.uk/ davidc/code.php.

SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics

online.

© The Author(s) 2018. Published by Oxford University Press.

DOI: 10.1093/bioinformatics/bty949

PMCID: PMC6596891

PMID: 30462147

15. Clin Infect Dis. 2019 Jul 2;69(2):295-305. doi: 10.1093/cid/ciy823.

Plasma Biomarkers to Detect Prevalent or Predict Progressive Tuberculosis

Associated With Human Immunodeficiency Virus-1.

Lesosky M(1)(2), Rangaka MX(2)(3)(4), Pienaar C(1), Coussens AK(2)(5), Goliath

R(2), Mathee S(6), Mwansa-Kambafwile J(2), Maartens G(3), Wilkinson

RJ(2)(3)(7)(8), Wilkinson KA(2)(3)(8).

Author information:

(1)Division of Epidemiology & Biostatistics, School of Public Health and Family

Medicine.

(2)Wellcome Centre for Infectious Diseases Research in Africa, Institute of

Infectious Diseases and Molecular Medicine, Observatory, South Africa.

(3)Department of Medicine, Faculty of Health Sciences, University of Cape Town,

Observatory, South Africa.

BACKGROUND: The risk of individuals infected with human immunodeficiency virus

(HIV)-1 developing tuberculosis (TB) is high, while both prognostic and

diagnostic tools remain insensitive. The potential for plasma biomarkers to

predict which HIV-1-infected individuals are likely to progress to active disease

is unknown.

METHODS: Thirteen analytes were measured from QuantiFERON Gold in-tube (QFT)

plasma samples in 421 HIV-1-infected persons recruited within the screening and

enrollment phases of a randomized, controlled trial of isoniazid preventive

therapy. Blood for QFT was obtained pre-randomization. Individuals were

classified into prevalent TB, incident TB, and control groups. Comparisons

between groups, supervised learning methods, and weighted correlation network

analyses were applied utilizing the unstimulated and background-corrected plasma

analyte concentrations.

RESULTS: Unstimulated samples showed higher analyte concentrations in the

prevalent and incident TB groups compared to the control group. The largest

differences were seen for C-X-C motif chemokine 10 (CXCL10), interleukin-2

(IL-2), IL-1α, transforming growth factor-α (TGF-α). A predictive model analysis

using unstimulated analytes discriminated best between the control and prevalent

TB groups (area under the curve [AUC] = 0.9), reasonably well between the

incident and prevalent TB groups (AUC > 0.8), and poorly between the control and

incident TB groups. Unstimulated IL-2 and IFN-γ were ranked at or near the top

for all comparisons, except the comparison between the control vs incident TB

groups. Models using background-adjusted values performed poorly.

CONCLUSIONS: Single plasma biomarkers are unlikely to distinguish between disease

states in HIV-1 co-infected individuals, and combinations of biomarkers are

required. The ability to detect prevalent TB is potentially important, as no

blood test hitherto has been suggested as having the utility to detect prevalent

TB amongst HIV-1 co-infected persons.

© The Author(s) 2018. Published by Oxford University Press for the Infectious

Diseases Society of America.

DOI: 10.1093/cid/ciy823

PMCID: PMC6603269

PMID: 30256919

16. Lancet Infect Dis. 2019 Oct;19(10):1129-1137. doi: 10.1016/ S1473- 3099(19)30309 -3. Epub 2019 Jul 16.

Long-term all-cause mortality in people treated for tuberculosis: a systematic

review and meta-analysis.

Romanowski K(1), Baumann B(2), Basham CA(3), Ahmad Khan F(4), Fox GJ(5), Johnston

JC(6).

Author information:

(1)Provincial TB Services, British Columbia Centre for Disease Control,

Vancouver, BC, Canada.

(2)Department of Medicine, University of British Columbia, Vancouver, BC, Canada.

(3)Provincial TB Services, British Columbia Centre for Disease Control,

Vancouver, BC, Canada; School of Population and Public Health, University of

British Columbia, Vancouver, BC, Canada.

BACKGROUND: Accurate estimates of long-term mortality following tuberculosis

treatment are scarce. This systematic review and meta-analysis aimed to estimate

the post-treatment mortality among tuberculosis survivors, and examine

differences in mortality risk by demographic and clinical characteristics.

METHODS: We systematically searched Embase, MEDLINE, and the Cochrane Database of

Systematic Reviews for cohort studies published in English between Jan 1, 1997,

and May 31, 2018. We included research papers that used a cohort study design,

included bacteriological or clinical confirmation of tuberculosis disease for all

participants, and reported, or provided enough data to calculate, mortality

estimates for people with tuberculosis and a valid control group representative

of the general population. We excluded studies that reported duplicate data, had

a study population of fewer than 50 people overall, had a follow-up period

shorter than 12 months after treatment completion, or had a loss to follow-up of

more than 30%. From eligible studies, we extracted standardised mortality ratios

(SMRs), or calculated them when the data were sufficient, by dividing the sum of

the observed deaths by the sum of the expected deaths. For studies that did not

report SMR as their mortality estimate, either mortality hazard ratios or

mortality rate ratios were extracted and pooled with SMRs. Random-effects

meta-analysis was used to obtain pooled SMRs. Between-study heterogeneity was

estimated with I2. This study was prospectively registered in PROSPERO

(CRD42018092592).

FINDINGS: Of the 7283 unique studies identified, data from ten studies, reporting

on 40?781 individuals and 6922 deaths, were included. The pooled SMR for

all-cause mortality among people with tuberculosis, compared with the control

group, was 2·91 (95% CI 2·21-3·84; I2=99%, pheterogeneity<0·0001). When

restricted to people with confirmed treatment completion or cure, the pooled SMR

was 3·76 (95% CI 3·04-4·66; I2=95%). Effect estimates were similar when

stratified by tuberculosis type, sex, age, and country income category. Causes of

mortality were extracted for 4226 deaths that occurred post-treatment, with most

deaths attributable to cardiovascular disease (20% [95% CI 15-26]; I2=92%).

INTERPRETATION: People treated for tuberculosis have significantly increased

mortality following treatment compared with the general population or matched

controls. These findings support the need for further research to understand and

address the biomedical and social factors that affect the long-term prognosis of

this population.

FUNDING: None.

Copyright © 2019 Elsevier Ltd. All rights reserved.

DOI: 10.1016/S1473-3099(19)30309-3

PMID: 31324519

上一篇: No.14

下一篇: No.12