Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
mBio ; 14(4): e0034023, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37350592

ABSTRACT

Nicotinamide adenine dinucleotide (NAD) and its phosphorylated derivative (NADP) are essential cofactors that participate in hundreds of biochemical reactions and have emerged as therapeutic targets in cancer, metabolic disorders, neurodegenerative diseases, and infections, including tuberculosis. The biological basis for the essentiality of NAD(P) in most settings, however, remains experimentally unexplained. Here, we report that inactivation of the terminal enzyme of NAD synthesis, NAD synthetase (NadE), elicits markedly different metabolic and microbiologic effects than those of the terminal enzyme of NADP biosynthesis, NAD kinase (PpnK), in Mycobacterium tuberculosis (Mtb). Inactivation of NadE led to parallel reductions of both NAD and NADP pools and Mtb viability, while inactivation of PpnK selectively depleted NADP pools but only arrested growth. Inactivation of each enzyme was accompanied by metabolic changes that were specific for the affected enzyme and associated microbiological phenotype. Bacteriostatic levels of NAD depletion caused a compensatory remodeling of NAD-dependent metabolic pathways in the absence of an impact on NADH/NAD ratios, while bactericidal levels of NAD depletion resulted in a disruption of NADH/NAD ratios and inhibition of oxygen respiration. These findings reveal a previously unrecognized physiologic specificity associated with the essentiality of two evolutionarily ubiquitous cofactors. IMPORTANCE The current course for cure of Mycobacterium tuberculosis (Mtb)-the etiologic agent of tuberculosis (TB)-infections is lengthy and requires multiple antibiotics. The development of shorter, simpler treatment regimens is, therefore, critical to the goal of eradicating TB. NadE, an enzyme required for the synthesis of the ubiquitous cofactor NAD, is essential for survival of Mtb and regarded as a promising drug target. However, the basis of this essentiality was not clear due to its role in the synthesis of both NAD and NADP. Here, we resolve this ambiguity through a combination of gene silencing and metabolomics. We specifically show that NADP deficiency is bacteriostatic, while NAD deficiency is bactericidal due to its role in Mtb's respiratory capacity. These results argue for a prioritization of NAD biosynthesis inhibitors in anti-TB drug development.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Humans , NAD/metabolism , NADP/metabolism , Ligases/metabolism
2.
Nat Commun ; 13(1): 2203, 2022 04 22.
Article in English | MEDLINE | ID: mdl-35459278

ABSTRACT

The ability of Mycobacterium tuberculosis (Mtb) to resist and tolerate antibiotics complicates the development of improved tuberculosis (TB) chemotherapies. Here we define the Mtb protein CinA as a major determinant of drug tolerance and as a potential target to shorten TB chemotherapy. By reducing the fraction of drug-tolerant persisters, genetic inactivation of cinA accelerated killing of Mtb by four antibiotics in clinical use: isoniazid, ethionamide, delamanid and pretomanid. Mtb ΔcinA was killed rapidly in conditions known to impede the efficacy of isoniazid, such as during nutrient starvation, during persistence in a caseum mimetic, in activated macrophages and during chronic mouse infection. Deletion of CinA also increased in vivo killing of Mtb by BPaL, a combination of pretomanid, bedaquiline and linezolid that is used to treat highly drug-resistant TB. Genetic and drug metabolism studies suggest that CinA mediates drug tolerance via cleavage of NAD-drug adducts.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis, Multidrug-Resistant , Animals , Antitubercular Agents/pharmacology , Antitubercular Agents/therapeutic use , Drug Tolerance , Isoniazid/pharmacology , Mice , Mycobacterium tuberculosis/genetics , Tuberculosis, Multidrug-Resistant/drug therapy
3.
Microbiol Spectr ; 9(2): e0092821, 2021 10 31.
Article in English | MEDLINE | ID: mdl-34550010

ABSTRACT

Phosphopantetheinyl hydrolase, PptH (Rv2795c), is a recently discovered enzyme from Mycobacterium tuberculosis that removes 4'-phosphopantetheine (Ppt) from holo-carrier proteins (CPs) and thereby opposes the action of phosphopantetheinyl transferases (PPTases). PptH is the first structurally characterized enzyme of the phosphopantetheinyl hydrolase family. However, conditions for optimal activity of PptH have not been defined, and only one substrate has been identified. Here, we provide biochemical characterization of PptH and demonstrate that the enzyme hydrolyzes Ppt in vitro from more than one M. tuberculosis holo-CP as well as holo-CPs from other organisms. PptH provided the only detectable activity in mycobacterial lysates that dephosphopantetheinylated acyl carrier protein M (AcpM), suggesting that PptH is the main Ppt hydrolase in M. tuberculosis. We could not detect a role for PptH in coenzyme A (CoA) salvage, and PptH was not required for virulence of M. tuberculosis during infection of mice. It remains to be determined why mycobacteria conserve a broadly acting phosphohydrolase that removes the Ppt prosthetic group from essential CPs. We speculate that the enzyme is critical for aspects of the life cycle of M. tuberculosis that are not routinely modeled. IMPORTANCE Tuberculosis (TB), caused by Mycobacterium tuberculosis, was the leading cause of death from an infectious disease before COVID, yet the in vivo essentiality and function of many of the protein-encoding genes expressed by M. tuberculosis are not known. We biochemically characterize M. tuberculosis's phosphopantetheinyl hydrolase, PptH, a protein unique to mycobacteria that removes an essential posttranslational modification on proteins involved in synthesis of lipids important for the bacterium's cell wall and virulence. We demonstrate that the enzyme has broad substrate specificity, but it does not appear to have a role in coenzyme A (CoA) salvage or virulence in a mouse model of TB.


Subject(s)
Bacterial Proteins/metabolism , Mycobacterium tuberculosis/enzymology , Pantetheine/analogs & derivatives , Phosphoric Diester Hydrolases/metabolism , Animals , Cell Wall/metabolism , Female , Humans , Lipids/biosynthesis , Mice , Mice, Inbred C57BL , Pantetheine/metabolism , Protein Processing, Post-Translational , Tuberculosis/pathology , Virulence/physiology
4.
J Bacteriol ; 2020 Jun 01.
Article in English | MEDLINE | ID: mdl-32482725

ABSTRACT

The Mycobacterium tuberculosis H37Rv genome has been sequenced and annotated over 20 years ago, yet roughly half of the protein-coding genes still lack a predicted function. We characterized two genes of unknown function, rv3679 and rv3680, for which inconsistent findings regarding their importance for virulence in mice have been reported. We confirmed that a rv3679-80 deletion mutant (Δrv3679-80) was virulent in mice and discovered that Δrv3679-80 suffered from a glycerol-dependent recovery defect on agar plates following mouse infection. Glycerol also exacerbated killing of Δrv3679-80 by nitric oxide. Rv3679-Rv3680 have previously been shown to form a complex with ATPase activity and we demonstrate that the ability of M. tuberculosis to cope with elevated levels of glycerol and nitric oxide requires intact ATP-binding motifs in both Rv3679 and Rv3680. Inactivation of glycerol kinase or Rv2370c, a protein of unknown function, suppressed glycerol mediated toxicity in Δrv3679-80 Glycerol catabolism led to increased intracellular methylglyoxal pools and Δrv3679-80 was hypersusceptible to extracellular methylglyoxal suggesting that glycerol toxicity in Δrv3679-80 is caused by methylglyoxal. Rv3679 and Rv3680 interacted with Rv1509, and Rv3679 had numerous additional interactors including proteins of the type II fatty acid synthase (FASII) pathway and mycolic acid modifying enzymes linking Rv3679 to fatty acid or lipid synthesis. This work provides experimentally determined roles for Rv3679 and Rv3680 and stimulates future research on these and other proteins of unknown function.Importance A better understanding of the pathogenesis of tuberculosis requires a better understanding of gene function in M. tuberculosis This work provides the first functional insight into the Rv3679/Rv3680 ATPase complex. We demonstrate that M. tuberculosis requires this complex and specifically its ATPase activity to resist glycerol and nitric oxide toxicity. We provide evidence that the glycerol-derived metabolite methylglyoxal causes toxicity in the absence of Rv3679/Rv3680. We further show that glycerol-dependent toxicity is reversed when glycerol kinase (GlpK) is inactivated. Our work uncovered other genes of unknown function that interact with Rv3679 and/or Rv3680 genetically or physically, underscoring the importance of understanding uncharacterized genes.

5.
Tuberculosis (Edinb) ; 121: 101914, 2020 03.
Article in English | MEDLINE | ID: mdl-32279870

ABSTRACT

Mycobacteria are important causes of disease in human and animal hosts. Diseases caused by mycobacteria include leprosy, tuberculosis (TB), nontuberculous mycobacteria (NTM) infections and Buruli Ulcer. To better understand and treat mycobacterial disease, clinicians, veterinarians and scientists use a range of discipline-specific approaches to conduct basic and applied research, including conducting epidemiological surveys, patient studies, wildlife sampling, animal models, genetic studies and computational simulations. To foster the exchange of knowledge and collaboration across disciplines, the Many Hosts of Mycobacteria (MHM) conference series brings together clinical, veterinary and basic scientists who are dedicated to advancing mycobacterial disease research. Started in 2007, the MHM series recently held its 8th conference at the Albert Einstein College of Medicine (Bronx, NY). Here, we review the diseases discussed at MHM8 and summarize the presentations on research advances in leprosy, NTM and Buruli Ulcer, human and animal TB, mycobacterial disease comorbidities, mycobacterial genetics and 'omics, and animal models. A mouse models workshop, which was held immediately after MHM8, is also summarized. In addition to being a resource for those who were unable to attend MHM8, we anticipate this review will provide a benchmark to gauge the progress of future research concerning mycobacteria and their many hosts.


Subject(s)
Bacteriology , Biomedical Research , Infectious Disease Medicine , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium/pathogenicity , Tuberculosis/microbiology , Animals , Congresses as Topic , Diffusion of Innovation , Disease Models, Animal , Host-Pathogen Interactions , Humans , Mycobacterium/genetics , Mycobacterium Infections, Nontuberculous/diagnosis , Mycobacterium Infections, Nontuberculous/epidemiology , Tuberculosis/diagnosis , Tuberculosis/epidemiology
6.
Science ; 363(6433): 1345-1349, 2019 03 22.
Article in English | MEDLINE | ID: mdl-30898933

ABSTRACT

Excessive consumption of beverages sweetened with high-fructose corn syrup (HFCS) is associated with obesity and with an increased risk of colorectal cancer. Whether HFCS contributes directly to tumorigenesis is unclear. We investigated the effects of daily oral administration of HFCS in adenomatous polyposis coli (APC) mutant mice, which are predisposed to develop intestinal tumors. The HFCS-treated mice showed a substantial increase in tumor size and tumor grade in the absence of obesity and metabolic syndrome. HFCS increased the concentrations of fructose and glucose in the intestinal lumen and serum, respectively, and the tumors transported both sugars. Within the tumors, fructose was converted to fructose-1-phosphate, leading to activation of glycolysis and increased synthesis of fatty acids that support tumor growth. These mouse studies support the hypothesis that the combination of dietary glucose and fructose, even at a moderate dose, can enhance tumorigenesis.


Subject(s)
Carcinogenesis/pathology , Diet/adverse effects , High Fructose Corn Syrup/adverse effects , Intestinal Neoplasms/pathology , Tumor Burden , Adenomatous Polyposis Coli Protein/genetics , Animals , High Fructose Corn Syrup/administration & dosage , Mice , Mice, Mutant Strains , Neoplasm Grading
7.
Science ; 363(6426)2019 02 01.
Article in English | MEDLINE | ID: mdl-30705156

ABSTRACT

Mycobacterium tuberculosis (Mtb) is the leading infectious cause of death in humans. Synthesis of lipids critical for Mtb's cell wall and virulence depends on phosphopantetheinyl transferase (PptT), an enzyme that transfers 4'-phosphopantetheine (Ppt) from coenzyme A (CoA) to diverse acyl carrier proteins. We identified a compound that kills Mtb by binding and partially inhibiting PptT. Killing of Mtb by the compound is potentiated by another enzyme encoded in the same operon, Ppt hydrolase (PptH), that undoes the PptT reaction. Thus, loss-of-function mutants of PptH displayed antimicrobial resistance. Our PptT-inhibitor cocrystal structure may aid further development of antimycobacterial agents against this long-sought target. The opposing reactions of PptT and PptH uncover a regulatory pathway in CoA physiology.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Coenzyme A/metabolism , Guanidine/analogs & derivatives , Hydrolases/antagonists & inhibitors , Mycobacterium tuberculosis/enzymology , Transferases (Other Substituted Phosphate Groups)/antagonists & inhibitors , Urea/analogs & derivatives , Acyl Carrier Protein/metabolism , Animals , Catalytic Domain , Drug Resistance, Bacterial/genetics , Female , Guanidine/pharmacology , Hydrolases/genetics , Lipid Metabolism , Loss of Function Mutation , Mice , Mice, Inbred BALB C , Mycobacterium tuberculosis/genetics , Operon , Protein Binding , Protein Structure, Tertiary , Small Molecule Libraries , Urea/pharmacology
8.
Proc Natl Acad Sci U S A ; 114(17): 4495-4500, 2017 04 25.
Article in English | MEDLINE | ID: mdl-28396391

ABSTRACT

Persistence, manifested as drug tolerance, represents a significant obstacle to global tuberculosis control. The bactericidal drugs isoniazid and rifampicin kill greater than 99% of exponentially growing Mycobacterium tuberculosis (Mtb) cells, but the remaining cells are persisters, cells with decreased metabolic rate, refractory to killing by these drugs, and able to generate drug-resistant mutants. We discovered that the combination of cysteine or other small thiols with either isoniazid or rifampicin prevents the formation of drug-tolerant and drug-resistant cells in Mtb cultures. This effect was concentration- and time-dependent, relying on increased oxygen consumption that triggered enhanced production of reactive oxygen species. In infected murine macrophages, the addition of N-acetylcysteine to isoniazid treatment potentiated the killing of Mtb Furthermore, we demonstrate that the addition of small thiols to Mtb drug treatment shifted the menaquinol/menaquinone balance toward a reduced state that stimulates Mtb respiration and converts persister cells to metabolically active cells. This prevention of both persister cell formation and drug resistance leads ultimately to mycobacterial cell death. Strategies to enhance respiration and initiate oxidative damage should improve tuberculosis chemotherapies.


Subject(s)
Antitubercular Agents/pharmacology , Drug Resistance, Bacterial/physiology , Mycobacterium tuberculosis/drug effects , Oxygen Consumption/physiology , Animals , Cell Line , DNA Breaks , Isoniazid , Macrophages/metabolism , Macrophages/microbiology , Mice , Mycobacterium tuberculosis/physiology , Reactive Oxygen Species , Rifampin
9.
Microbiol Spectr ; 5(1)2017 01.
Article in English | MEDLINE | ID: mdl-28155811

ABSTRACT

Accumulating evidence has left little doubt about the importance of persistence or metabolism in the biology and chemotherapy of tuberculosis. However, knowledge of the intersection between these two factors has only recently begun to emerge. Here, we provide a focused review of metabolic characteristics associated with Mycobacterium tuberculosis persistence. We focus on metabolism because it is the biochemical foundation of all physiologic processes and a distinguishing hallmark of M. tuberculosis physiology and pathogenicity. In addition, it serves as the chemical interface between host and pathogen. Existing knowledge, however, derives largely from physiologic contexts in which replication is the primary biochemical objective. The goal of this review is to reframe current knowledge of M. tuberculosis metabolism in the context of persistence, where quiescence is often a key distinguishing characteristic. Such a perspective may help ongoing efforts to develop more efficient cures and inform on novel strategies to break the cycle of transmission sustaining the pandemic.


Subject(s)
Host-Pathogen Interactions , Metabolism , Mycobacterium tuberculosis/pathogenicity , Animals , Humans
10.
ACS Infect Dis ; 3(1): 18-33, 2017 01 13.
Article in English | MEDLINE | ID: mdl-27704782

ABSTRACT

A potent, noncytotoxic indazole sulfonamide was identified by high-throughput screening of >100,000 synthetic compounds for activity against Mycobacterium tuberculosis (Mtb). This noncytotoxic compound did not directly inhibit cell wall biogenesis but triggered a slow lysis of Mtb cells as measured by release of intracellular green fluorescent protein (GFP). Isolation of resistant mutants followed by whole-genome sequencing showed an unusual gene amplification of a 40 gene region spanning from Rv3371 to Rv3411c and in one case a potential promoter mutation upstream of guaB2 (Rv3411c) encoding inosine monophosphate dehydrogenase (IMPDH). Subsequent biochemical validation confirmed direct inhibition of IMPDH by an uncompetitive mode of inhibition, and growth inhibition could be rescued by supplementation with guanine, a bypass mechanism for the IMPDH pathway. Beads containing immobilized indazole sulfonamides specifically interacted with IMPDH in cell lysates. X-ray crystallography of the IMPDH-IMP-inhibitor complex revealed that the primary interactions of these compounds with IMPDH were direct pi-pi interactions with the IMP substrate. Advanced lead compounds in this series with acceptable pharmacokinetic properties failed to show efficacy in acute or chronic murine models of tuberculosis (TB). Time-kill experiments in vitro suggest that sustained exposure to drug concentrations above the minimum inhibitory concentration (MIC) for 24 h were required for a cidal effect, levels that have been difficult to achieve in vivo. Direct measurement of guanine levels in resected lung tissue from tuberculosis-infected animals and patients revealed 0.5-2 mM concentrations in caseum and normal lung tissue. The high lesional levels of guanine and the slow lytic, growth-rate-dependent effect of IMPDH inhibition pose challenges to developing drugs against this target for use in treating TB.


Subject(s)
Antitubercular Agents/pharmacology , IMP Dehydrogenase/antagonists & inhibitors , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Sulfonamides/pharmacology , Animals , Drug Design , Drug Discovery , Drug Resistance, Bacterial , Gene Expression Regulation, Bacterial/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Humans , Mice , Mice, Inbred C57BL , Molecular Structure , Mutation , Protein Conformation , Rabbits , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/pharmacokinetics , Tuberculosis/drug therapy
11.
PLoS Pathog ; 12(12): e1006043, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27936238

ABSTRACT

Trehalose biosynthesis is considered an attractive target for the development of antimicrobials against fungal, helminthic and bacterial pathogens including Mycobacterium tuberculosis. The most common biosynthetic route involves trehalose-6-phosphate (T6P) synthase OtsA and T6P phosphatase OtsB that generate trehalose from ADP/UDP-glucose and glucose-6-phosphate. In order to assess the drug target potential of T6P phosphatase, we generated a conditional mutant of M. tuberculosis allowing the regulated gene silencing of the T6P phosphatase gene otsB2. We found that otsB2 is essential for growth of M. tuberculosis in vitro as well as for the acute infection phase in mice following aerosol infection. By contrast, otsB2 is not essential for the chronic infection phase in mice, highlighting the substantial remodelling of trehalose metabolism during infection by M. tuberculosis. Blocking OtsB2 resulted in the accumulation of its substrate T6P, which appears to be toxic, leading to the self-poisoning of cells. Accordingly, blocking T6P production in a ΔotsA mutant abrogated otsB2 essentiality. T6P accumulation elicited a global upregulation of more than 800 genes, which might result from an increase in RNA stability implied by the enhanced neutralization of toxins exhibiting ribonuclease activity. Surprisingly, overlap with the stress response caused by the accumulation of another toxic sugar phosphate molecule, maltose-1-phosphate, was minimal. A genome-wide screen for synthetic lethal interactions with otsA identified numerous genes, revealing additional potential drug targets synergistic with OtsB2 suitable for combination therapies that would minimize the emergence of resistance to OtsB2 inhibitors.


Subject(s)
Bacterial Proteins/metabolism , Mycobacterium tuberculosis/enzymology , Phosphoric Monoester Hydrolases/metabolism , Sugar Phosphates/metabolism , Trehalose/analogs & derivatives , Tuberculosis/enzymology , Animals , Chromatography, Thin Layer , Disease Models, Animal , Female , Gene Expression Profiling , Gene Knockdown Techniques , Glucosyltransferases/metabolism , In Vitro Techniques , Mice , Mice, Inbred C57BL , Nuclear Magnetic Resonance, Biomolecular , Real-Time Polymerase Chain Reaction , Trehalose/metabolism
12.
J Biol Chem ; 291(13): 7060-9, 2016 Mar 25.
Article in English | MEDLINE | ID: mdl-26858255

ABSTRACT

Mycobacterium tuberculosis (Mtb) displays a high degree of metabolic plasticity to adapt to challenging host environments. Genetic evidence suggests thatMtbrelies mainly on fatty acid catabolism in the host. However,Mtbalso maintains a functional glycolytic pathway and its role in the cellular metabolism ofMtbhas yet to be understood. Pyruvate kinase catalyzes the last and rate-limiting step in glycolysis and theMtbgenome harbors one putative pyruvate kinase (pykA, Rv1617). Here we show thatpykAencodes an active pyruvate kinase that is allosterically activated by glucose 6-phosphate (Glc-6-P) and adenosine monophosphate (AMP). Deletion ofpykApreventsMtbgrowth in the presence of fermentable carbon sources and has a cidal effect in the presence of glucose that correlates with elevated levels of the toxic catabolite methylglyoxal. Growth attenuation was also observed in media containing a combination of short chain fatty acids and glucose and surprisingly, in media containing odd and even chain fatty acids alone. Untargeted high sensitivity metabolomics revealed that inactivation of pyruvate kinase leads to accumulation of phosphoenolpyruvate (P-enolpyruvate), citrate, and aconitate, which was consistent with allosteric inhibition of isocitrate dehydrogenase by P-enolpyruvate. This metabolic block could be relieved by addition of the α-ketoglutarate precursor glutamate. Taken together, our study identifies an essential role of pyruvate kinase in preventing metabolic block during carbon co-catabolism inMtb.


Subject(s)
Bacterial Proteins/metabolism , Carbon/metabolism , Glycolysis/genetics , Mycobacterium tuberculosis/metabolism , Pyruvate Kinase/metabolism , Aconitic Acid/metabolism , Adenosine Monophosphate/metabolism , Adenosine Monophosphate/pharmacology , Allosteric Regulation , Animals , Bacterial Proteins/genetics , Citric Acid/metabolism , Culture Media/chemistry , Enzyme Activation , Fatty Acids, Volatile/pharmacology , Female , Gene Deletion , Gene Expression , Glucose/metabolism , Glucose-6-Phosphate/metabolism , Glucose-6-Phosphate/pharmacology , Glutamic Acid/metabolism , Glutamic Acid/pharmacology , Glycolysis/drug effects , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Ketoglutaric Acids/metabolism , Mice , Mice, SCID , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , Phosphoenolpyruvate/metabolism , Pyruvaldehyde/metabolism , Pyruvate Kinase/genetics , Survival Analysis , Tuberculosis/microbiology , Tuberculosis/mortality
13.
mBio ; 6(6): e01313-15, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26578674

ABSTRACT

UNLABELLED: Mycobacterium haemophilum is an emerging pathogen associated with a variety of clinical syndromes, most commonly skin infections in immunocompromised individuals. M. haemophilum exhibits a unique requirement for iron supplementation to support its growth in culture, but the basis for this property and how it may shape pathogenesis is unclear. Using a combination of Illumina, PacBio, and Sanger sequencing, the complete genome sequence of M. haemophilum was determined. Guided by this sequence, experiments were performed to define the basis for the unique growth requirements of M. haemophilum. We found that M. haemophilum, unlike many other mycobacteria, is unable to synthesize iron-binding siderophores known as mycobactins or to utilize ferri-mycobactins to support growth. These differences correlate with the absence of genes associated with mycobactin synthesis, secretion, and uptake. In agreement with the ability of heme to promote growth, we identified genes encoding heme uptake machinery. Consistent with its propensity to infect the skin, we show at the whole-genome level the genetic closeness of M. haemophilum with Mycobacterium leprae, an organism which cannot be cultivated in vitro, and we identify genes uniquely shared by these organisms. Finally, we identify means to express foreign genes in M. haemophilum. These data explain the unique culture requirements for this important pathogen, provide a foundation upon which the genome sequence can be exploited to improve diagnostics and therapeutics, and suggest use of M. haemophilum as a tool to elucidate functions of genes shared with M. leprae. IMPORTANCE: Mycobacterium haemophilum is an emerging pathogen with an unknown natural reservoir that exhibits unique requirements for iron supplementation to grow in vitro. Understanding the basis for this iron requirement is important because it is fundamental to isolation of the organism from clinical samples and environmental sources. Defining the molecular basis for M. haemophilium's growth requirements will also shed new light on mycobacterial strategies to acquire iron and can be exploited to define how differences in such strategies influence pathogenesis. Here, through a combination of sequencing and experimental approaches, we explain the basis for the iron requirement. We further demonstrate the genetic closeness of M. haemophilum and Mycobacterium leprae, the causative agent of leprosy which cannot be cultured in vitro, and we demonstrate methods to genetically manipulate M. haemophilum. These findings pave the way for the use of M. haemophilum as a model to elucidate functions of genes shared with M. leprae.


Subject(s)
Culture Media/chemistry , Genome, Bacterial , Mycobacterium haemophilum/growth & development , Mycobacterium haemophilum/genetics , Base Sequence , Heme/genetics , Heme/metabolism , Hemoglobins/metabolism , Humans , Iron/metabolism , Mycobacterium leprae/genetics , Oxazoles/metabolism , Phenotype , Sequence Analysis, DNA
14.
J Environ Manage ; 151: 427-36, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25596544

ABSTRACT

We examine sensitivity of estimates of recreation demand using the Travel Cost Method (TCM) to four factors. Three of the four have been routinely and widely discussed in the TCM literature: a) Poisson verses negative binomial regression; b) application of Englin correction to account for endogenous stratification; c) truncation of the data set to eliminate outliers. A fourth issue we address has not been widely modeled: the potential effect on recreation demand of the interaction between income and travel cost. We provide a straightforward comparison of all four factors, analyzing the impact of each on regression parameters and consumer surplus estimates. Truncation has a modest effect on estimates obtained from the Poisson models but a radical effect on the estimates obtained by way of the negative binomial. Inclusion of an income-travel cost interaction term generally produces a more conservative but not a statistically significantly different estimate of consumer surplus in both Poisson and negative binomial models. It also generates broader confidence intervals. Application of truncation, the Englin correction and the income-travel cost interaction produced the most conservative estimates of consumer surplus and eliminated the statistical difference between the Poisson and the negative binomial. Use of the income-travel cost interaction term reveals that for visitors who face relatively low travel costs, the relationship between income and travel demand is negative, while it is positive for those who face high travel costs. This provides an explanation of the ambiguities on the findings regarding the role of income widely observed in the TCM literature. Our results suggest that policies that reduce access to publicly owned resources inordinately impact local low income recreationists and are contrary to environmental justice.


Subject(s)
Recreation/economics , Travel/economics , Environment , Humans , Income , Models, Economic , Public Policy
15.
ACS Infect Dis ; 1(1): 73-5, 2015 Jan 09.
Article in English | MEDLINE | ID: mdl-27620146
16.
PLoS Pathog ; 10(11): e1004510, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25412183

ABSTRACT

In chronic infection, Mycobacterium tuberculosis bacilli are thought to enter a metabolic program that provides sufficient energy for maintenance of the protonmotive force, but is insufficient to meet the demands of cellular growth. We sought to understand this metabolic downshift genetically by targeting succinate dehydrogenase, the enzyme which couples the growth processes controlled by the TCA cycle with the energy production resulting from the electron transport chain. M. tuberculosis contains two operons which are predicted to encode succinate dehydrogenase enzymes (sdh-1 and sdh-2); we found that deletion of Sdh1 contributes to an inability to survive long term stationary phase. Stable isotope labeling and mass spectrometry revealed that Sdh1 functions as a succinate dehydrogenase during aerobic growth, and that Sdh2 is dispensable for this catalysis, but partially overlapping activities ensure that the loss of one enzyme can incompletely compensate for loss of the other. Deletion of Sdh1 disturbs the rate of respiration via the mycobacterial electron transport chain, resulting in an increased proportion of reduced electron carrier (menaquinol) which leads to increased oxygen consumption. The loss of respiratory control leads to an inability to recover from stationary phase. We propose a model in which succinate dehydrogenase is a governor of cellular respiration in the adaptation to low oxygen environments.


Subject(s)
Bacterial Proteins/metabolism , Models, Biological , Mycobacterium tuberculosis/enzymology , Oxygen Consumption/physiology , Succinate Dehydrogenase/metabolism , Animals , Bacterial Proteins/genetics , Mice , Mice, Knockout , Microbial Viability/genetics , Mycobacterium tuberculosis/genetics , Succinate Dehydrogenase/genetics
17.
Microbiol Spectr ; 2(3)2014 Jun.
Article in English | MEDLINE | ID: mdl-25346874

ABSTRACT

Mycobacteria inhabit a wide range of intracellular and extracellular environments. Many of these environments are highly dynamic and therefore mycobacteria are faced with the constant challenge of redirecting their metabolic activity to be commensurate with either replicative growth or a non-replicative quiescence. A fundamental feature in this adaptation is the ability of mycobacteria to respire, regenerate reducing equivalents and generate ATP via oxidative phosphorylation. Mycobacteria harbor multiple primary dehydrogenases to fuel the electron transport chain and two terminal respiratory oxidases, an aa3 -type cytochrome c oxidase and cytochrome bd-type menaquinol oxidase, are present for dioxygen reduction coupled to the generation of a protonmotive force. Hypoxia leads to the downregulation of key respiratory complexes, but the molecular mechanisms regulating this expression are unknown. Despite being obligate aerobes, mycobacteria have the ability to metabolize in the absence of oxygen and a number of reductases are present to facilitate the turnover of reducing equivalents under these conditions (e.g. nitrate reductase, succinate dehydrogenase/fumarate reductase). Hydrogenases and ferredoxins are also present in the genomes of mycobacteria suggesting the ability of these bacteria to adapt to an anaerobic-type of metabolism in the absence of oxygen. ATP synthesis by the membrane-bound F1FO-ATP synthase is essential for growing and non-growing mycobacteria and the enzyme is able to function over a wide range of protonmotive force values (aerobic to hypoxic). The discovery of lead compounds that target respiration and oxidative phosphorylation in Mycobacterium tuberculosis highlights the importance of this area for the generation of new front line drugs to combat tuberculosis.


Subject(s)
Adenosine Triphosphate/metabolism , Energy Metabolism , Mycobacterium tuberculosis/metabolism , Oxidative Phosphorylation , Aerobiosis , Metabolic Networks and Pathways , Oxidoreductases/metabolism
18.
mBio ; 5(4)2014 Aug 12.
Article in English | MEDLINE | ID: mdl-25118234

ABSTRACT

UNLABELLED: Succinate:quinone oxidoreductase (Sdh) is a membrane-bound complex that couples the oxidation of succinate to fumarate in the cytoplasm to the reduction of quinone to quinol in the membrane. Mycobacterial species harbor genes for two putative sdh operons, but the individual roles of these two operons are unknown. In this communication, we show that Mycobacterium smegmatis mc(2)155 expresses two succinate dehydrogenases designated Sdh1 and Sdh2. Sdh1 is encoded by a five-gene operon (MSMEG_0416-MSMEG_0420), and Sdh2 is encoded by a four-gene operon (MSMEG_1672-MSMEG_1669). These two operons are differentially expressed in response to carbon limitation, hypoxia, and fumarate, as monitored by sdh promoter-lacZ fusions. While deletion of the sdh1 operon did not yield any growth phenotypes on succinate or other nonfermentable carbon sources, the sdh2 operon could be deleted only in a merodiploid background, demonstrating that Sdh2 is essential for growth. Sdh activity and succinate-dependent proton pumping were detected in cells grown aerobically, as well as under hypoxia. Fumarate reductase activity was absent under these conditions, indicating that neither Sdh1 nor Sdh2 could catalyze the reverse reaction. Sdh activity was inhibited by the Sdh inhibitor 3-nitroproprionate (3NP), and treatment with 3NP dissipated the membrane potential of wild-type or Δsdh1 mutant cells under hypoxia but not that of cells grown aerobically. These data imply that Sdh2 is the generator of the membrane potential under hypoxia, an essential role for the cell. IMPORTANCE: Complex II or succinate dehydrogenase (Sdh) is a major respiratory enzyme that couples the oxidation of succinate to fumarate in the cytoplasm to the reduction of quinone to quinol in the membrane. Mycobacterial species harbor genes for two putative sdh operons, sdh1 and sdh2, but the individual roles of these two operons are unknown. In this communication, we show that sdh1 and sdh2 are differentially expressed in response to energy limitation, oxygen tension, and alternative electron acceptor availability, suggesting distinct functional cellular roles. Sdh2 was essential for growth and generation of the membrane potential in hypoxic cells. Given the essentiality of succinate dehydrogenase and oxidative phosphorylation in the growth cycle of Mycobacterium tuberculosis, the potential exists to develop new antituberculosis agents against the mycobacterial succinate dehydrogenase. This enzyme has been proposed as a potential target for the development of new chemotherapeutic agents against intracellular parasites and mitochondrion-associated disease.


Subject(s)
Membrane Potentials/physiology , Mycobacterium smegmatis/enzymology , Mycobacterium smegmatis/genetics , Operon , Oxygen , Succinate Dehydrogenase/genetics , Succinate Dehydrogenase/metabolism , Fumarates/metabolism , Gene Expression , Mitochondria/metabolism , Mutation , Mycobacterium smegmatis/growth & development , Mycobacterium smegmatis/metabolism , Oxidation-Reduction , Oxygen Consumption , Phenotype , Sequence Alignment , Succinate Dehydrogenase/chemistry , Succinates/metabolism
19.
mBio ; 5(4): e01275-14, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-25028424

ABSTRACT

The new medicinal compound bedaquiline (BDQ) kills Mycobacterium tuberculosis by inhibiting F1Fo-ATP synthase. BDQ is bacteriostatic for 4 to 7 days and kills relatively slowly compared to other frontline tuberculosis (TB) drugs. Here we show that killing with BDQ can be improved significantly by inhibiting cytochrome bd oxidase, a non-proton-pumping terminal oxidase. BDQ was instantly bactericidal against a cytochrome bd oxidase null mutant of M. tuberculosis, and the rate of killing was increased by more than 50%. We propose that this exclusively bacterial enzyme should be a high-priority target for new drug discovery. Importance: A major drawback of current TB chemotherapy is its long duration. New drug regimens with rapid killing kinetics are desperately needed. Our study demonstrates that inhibition of a nonessential bacterial enzyme greatly improves the efficacy of the latest TB drug bedaquiline and emphasizes that screening for compounds with synergistic killing mechanisms is a promising strategy.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proteins/genetics , Cytochromes/genetics , Diarylquinolines/pharmacology , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , Oxidoreductases/genetics , Mutation
20.
PLoS Pathog ; 10(5): e1004115, 2014 May.
Article in English | MEDLINE | ID: mdl-24809459

ABSTRACT

Mycobacterium tuberculosis bacilli display two signature features: acid-fast staining and the capacity to induce long-term latent infections in humans. However, the mechanisms governing these two important processes remain largely unknown. Ser/Thr phosphorylation has recently emerged as an important regulatory mechanism allowing mycobacteria to adapt their cell wall structure/composition in response to their environment. Herein, we evaluated whether phosphorylation of KasB, a crucial mycolic acid biosynthetic enzyme, could modulate acid-fast staining and virulence. Tandem mass spectrometry and site-directed mutagenesis revealed that phosphorylation of KasB occurred at Thr334 and Thr336 both in vitro and in mycobacteria. Isogenic strains of M. tuberculosis with either a deletion of the kasB gene or a kasB_T334D/T336D allele, mimicking constitutive phosphorylation of KasB, were constructed by specialized linkage transduction. Biochemical and structural analyses comparing these mutants to the parental strain revealed that both mutant strains had mycolic acids that were shortened by 4-6 carbon atoms and lacked trans-cyclopropanation. Together, these results suggested that in M. tuberculosis, phosphorylation profoundly decreases the condensing activity of KasB. Structural/modeling analyses reveal that Thr334 and Thr336 are located in the vicinity of the catalytic triad, which indicates that phosphorylation of these amino acids would result in loss of enzyme activity. Importantly, the kasB_T334D/T336D phosphomimetic and deletion alleles, in contrast to the kasB_T334A/T336A phosphoablative allele, completely lost acid-fast staining. Moreover, assessing the virulence of these strains indicated that the KasB phosphomimetic mutant was attenuated in both immunodeficient and immunocompetent mice following aerosol infection. This attenuation was characterized by the absence of lung pathology. Overall, these results highlight for the first time the role of Ser/Thr kinase-dependent KasB phosphorylation in regulating the later stages of mycolic acid elongation, with important consequences in terms of acid-fast staining and pathogenicity.


Subject(s)
3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , Bacterial Proteins/metabolism , Mycobacterium tuberculosis/metabolism , Mycobacterium tuberculosis/pathogenicity , Mycolic Acids/metabolism , Protein Serine-Threonine Kinases/metabolism , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/genetics , Animals , Bacterial Proteins/genetics , Catalytic Domain/genetics , Cell Wall/metabolism , Lipid Metabolism/genetics , Mice , Mice, Inbred C57BL , Mice, SCID , Microbiological Techniques/methods , Models, Molecular , Mycobacterium tuberculosis/genetics , Mycolic Acids/chemistry , Phosphorylation , Staining and Labeling/methods , Tuberculosis/diagnosis , Tuberculosis/metabolism , Tuberculosis/microbiology , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...