Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Methods ; 223: 118-126, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38246229

ABSTRACT

Quantitative Systems Pharmacology (QSP) models are increasingly being applied for target discovery and dose selection in immuno-oncology (IO). Typical application involves virtual trial, a simulation of a virtual population of hundreds of model instances with model inputs reflecting individual variability. While the structure of the model and initial parameterisation are based on literature describing the underlying biology, calibration of the virtual population by existing clinical data is frequently required to create tumour and patient population specific model instances. Since comparison of a virtual trial with clinical output requires hundreds of large-scale, non-linear model evaluations, the inference of a virtual population is computationally expensive, frequently becoming a bottleneck. Here, we present novel approach to virtual population inference in IO using emulation of the QSP model and an objective function based on Kolmogorov-Smirnov statistics to maximise congruence of simulated and observed clinical tumour size distributions. We sample the parameter space of a QSP IO model to collect a set of tumour growth time profiles. We evaluate performance of several machine learning approaches in interpolating these time profiles and create a surrogate model, which computes tumor growth profiles faster than the original model and allows examination of tens of millions of virtual patients. We use the surrogate model to infer a virtual population maximising congruence with the waterfall plot of a pembrolizumab clinical trial. We believe that our approach is applicable not only in QSP IO, but also in other applications where virtual populations need to be inferred for computationally expensive mechanistic models.


Subject(s)
Neoplasms , Network Pharmacology , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Medical Oncology , Computer Simulation , Calibration
2.
Int J Mol Sci ; 24(19)2023 Sep 29.
Article in English | MEDLINE | ID: mdl-37834211

ABSTRACT

RNA polymerase III (RNAP III) holoenzyme activity and the processing of its products have been linked to several metabolic dysfunctions in lower and higher eukaryotes. Alterations in the activity of RNAP III-driven synthesis of non-coding RNA cause extensive changes in glucose metabolism. Increased RNAP III activity in the S. cerevisiae maf1Δ strain is lethal when grown on a non-fermentable carbon source. This lethal phenotype is suppressed by reducing tRNA synthesis. Neither the cause of the lack of growth nor the underlying molecular mechanism have been deciphered, and this area has been awaiting scientific explanation for a decade. Our previous proteomics data suggested mitochondrial dysfunction in the strain. Using model mutant strains maf1Δ (with increased tRNA abundance) and rpc128-1007 (with reduced tRNA abundance), we collected data showing major changes in the TCA cycle metabolism of the mutants that explain the phenotypic observations. Based on 13C flux data and analysis of TCA enzyme activities, the present study identifies the flux constraints in the mitochondrial metabolic network. The lack of growth is associated with a decrease in TCA cycle activity and downregulation of the flux towards glutamate, aspartate and phosphoenolpyruvate (PEP), the metabolic intermediate feeding the gluconeogenic pathway. rpc128-1007, the strain that is unable to increase tRNA synthesis due to a mutation in the C128 subunit, has increased TCA cycle activity under non-fermentable conditions. To summarize, cells with non-optimal activity of RNAP III undergo substantial adaptation to a new metabolic state, which makes them vulnerable under specific growth conditions. Our results strongly suggest that balanced, non-coding RNA synthesis that is coupled to glucose signaling is a fundamental requirement to sustain a cell's intracellular homeostasis and flexibility under changing growth conditions. The presented results provide insight into the possible role of RNAP III in the mitochondrial metabolism of other cell types.


Subject(s)
Mitochondria , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Mitochondria/metabolism , Homeostasis , RNA, Transfer/genetics , RNA, Transfer/metabolism , RNA, Untranslated/metabolism
3.
CPT Pharmacometrics Syst Pharmacol ; 12(7): 889-903, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37452454

ABSTRACT

Typical Quantitative Systems Pharmacology (QSP) workflows involve discussion of biology, supported by graphical diagrams, followed by construction of large Ordinary Differential Equation models. QSP Designer facilitates this process by providing enhanced graphical notation, which enables hierarchical presentation with modules and handling of combinatorial complexity with diagram node arrays. Whereas the software includes a simulation engine, a major feature is full model code generation in MATLAB, R, C, and Julia to support multiple modeling communities.


Subject(s)
Network Pharmacology , Pharmacology , Humans , Models, Biological , Software , Computer Simulation , Language
4.
CPT Pharmacometrics Syst Pharmacol ; 12(2): 139-143, 2023 02.
Article in English | MEDLINE | ID: mdl-36418887

ABSTRACT

Immunogenicity against therapeutic proteins frequently causes attrition owing to its potential impact on pharmacokinetics, pharmacodynamics, efficacy, and safety. Predicting immunogenicity is complex because of its multifactorial drivers, including compound properties, subject characteristics, and treatment parameters. To integrate these, the Immunogenicity Simulator was developed using published, predominantly late-stage trial data from 15 therapeutic proteins. This single-blinded evaluation with subject-level data from 10 further monoclonals assesses the Immunogenicity Simulator's credibility for application during the drug development process.


Subject(s)
Drug Development , Network Pharmacology , Humans , Proteins/immunology , Proteins/therapeutic use
5.
Int J Antimicrob Agents ; 60(1): 106606, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35588969

ABSTRACT

The COVID-19 pandemic has severely impacted health systems and economies worldwide. Significant global efforts are therefore ongoing to improve vaccine efficacies, optimize vaccine deployment, and develop new antiviral therapies to combat the pandemic. Mechanistic viral dynamics and quantitative systems pharmacology models of SARS-CoV-2 infection, vaccines, immunomodulatory agents, and antiviral therapeutics have played a key role in advancing our understanding of SARS-CoV-2 pathogenesis and transmission, the interplay between innate and adaptive immunity to influence the outcomes of infection, effectiveness of treatments, mechanisms and performance of COVID-19 vaccines, and the impact of emerging SARS-CoV-2 variants. Here, we review some of the critical insights provided by these models and discuss the challenges ahead.


Subject(s)
COVID-19 , Models, Biological , Antiviral Agents/therapeutic use , COVID-19/epidemiology , COVID-19/pathology , COVID-19/prevention & control , COVID-19/virology , COVID-19 Vaccines , Disease Progression , Humans , Pandemics/prevention & control
6.
CPT Pharmacometrics Syst Pharmacol ; 10(10): 1130-1133, 2021 10.
Article in English | MEDLINE | ID: mdl-34331834

ABSTRACT

Optimal use and distribution of coronavirus disease 2019 (COVID-19) vaccines involves adjustments of dosing. Due to the rapidly evolving pandemic, such adjustments often need to be introduced before full efficacy data are available. As demonstrated in other areas of drug development, quantitative systems pharmacology (QSP) is well placed to guide such extrapolation in a rational and timely manner. Here, we propose for the first time how QSP can be applied in the context of COVID-19 vaccine development.


Subject(s)
COVID-19 Vaccines/administration & dosage , Systems Biology/methods , COVID-19/prevention & control , Drug Dosage Calculations , Humans
7.
Clin Pharmacol Ther ; 109(3): 605-618, 2021 03.
Article in English | MEDLINE | ID: mdl-32686076

ABSTRACT

Drug development in oncology commonly exploits the tools of molecular biology to gain therapeutic benefit through reprograming of cellular responses. In immuno-oncology (IO) the aim is to direct the patient's own immune system to fight cancer. After remarkable successes of antibodies targeting PD1/PD-L1 and CTLA4 receptors in targeted patient populations, the focus of further development has shifted toward combination therapies. However, the current drug-development approach of exploiting a vast number of possible combination targets and dosing regimens has proven to be challenging and is arguably inefficient. In particular, the unprecedented number of clinical trials testing different combinations may no longer be sustainable by the population of available patients. Further development in IO requires a step change in selection and validation of candidate therapies to decrease development attrition rate and limit the number of clinical trials. Quantitative systems pharmacology (QSP) proposes to tackle this challenge through mechanistic modeling and simulation. Compounds' pharmacokinetics, target binding, and mechanisms of action as well as existing knowledge on the underlying tumor and immune system biology are described by quantitative, dynamic models aiming to predict clinical results for novel combinations. Here, we review the current QSP approaches, the legacy of mathematical models available to quantitative clinical pharmacologists describing interaction between tumor and immune system, and the recent development of IO QSP platform models. We argue that QSP and virtual patients can be integrated as a new tool in existing IO drug development approaches to increase the efficiency and effectiveness of the search for novel combination therapies.


Subject(s)
Allergy and Immunology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Development , Immune Checkpoint Inhibitors/therapeutic use , Medical Oncology , Molecular Dynamics Simulation , Neoplasms/drug therapy , Systems Biology , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Computer Simulation , Humans , Immune Checkpoint Inhibitors/adverse effects , Immune Checkpoint Inhibitors/pharmacokinetics , Models, Immunological , Molecular Targeted Therapy , Neoplasms/immunology , Neoplasms/metabolism , Tumor Microenvironment
8.
Clin Pharmacol Ther ; 107(4): 858-870, 2020 04.
Article in English | MEDLINE | ID: mdl-31955413

ABSTRACT

Application of contemporary molecular biology techniques to clinical samples in oncology resulted in the accumulation of unprecedented experimental data. These "omics" data are mined for discovery of therapeutic target combinations and diagnostic biomarkers. It is less appreciated that omics resources could also revolutionize development of the mechanistic models informing clinical pharmacology quantitative decisions about dose amount, timing, and sequence. We discuss the integration of omics data to inform mechanistic models supporting drug development in immuno-oncology. To illustrate our arguments, we present a minimal clinical model of the Cancer Immunity Cycle (CIC), calibrated for non-small cell lung carcinoma using tumor microenvironment composition inferred from transcriptomics of clinical samples. We review omics data resources, which can be integrated to parameterize mechanistic models of the CIC. We propose that virtual trial simulations with clinical Quantitative Systems Pharmacology platforms informed by omics data will be making increasing impact in the development of cancer immunotherapies.


Subject(s)
Carcinoma, Non-Small-Cell Lung/therapy , Data Collection/methods , Immunotherapy/methods , Lung Neoplasms/therapy , Medical Oncology/methods , Pharmacology, Clinical/methods , Carcinoma, Non-Small-Cell Lung/immunology , Data Collection/statistics & numerical data , Drug Development/methods , Drug Development/statistics & numerical data , Humans , Immunity, Cellular/drug effects , Immunity, Cellular/immunology , Immunotherapy/statistics & numerical data , Lung Neoplasms/immunology , Medical Oncology/statistics & numerical data , Pharmacology, Clinical/statistics & numerical data
11.
NPJ Syst Biol Appl ; 4: 33, 2018.
Article in English | MEDLINE | ID: mdl-30131870

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is a serious public health issue associated with high fat, high sugar diets. However, the molecular mechanisms mediating NAFLD pathogenesis are only partially understood. Here we adopt an iterative multi-scale, systems biology approach coupled to in vitro experimentation to investigate the roles of sugar and fat metabolism in NAFLD pathogenesis. The use of fructose as a sweetening agent is controversial; to explore this, we developed a predictive model of human monosaccharide transport, signalling and metabolism. The resulting quantitative model comprising a kinetic model describing monosaccharide transport and insulin signalling integrated with a hepatocyte-specific genome-scale metabolic network (GSMN). Differential kinetics for the utilisation of glucose and fructose were predicted, but the resultant triacylglycerol production was predicted to be similar for monosaccharides; these predictions were verified by in vitro data. The role of physiological adaptation to lipid overload was explored through the comprehensive reconstruction of the peroxisome proliferator activated receptor alpha (PPARα) regulome integrated with a hepatocyte-specific GSMN. The resulting qualitative model reproduced metabolic responses to increased fatty acid levels and mimicked lipid loading in vitro. The model predicted that activation of PPARα by lipids produces a biphasic response, which initially exacerbates steatosis. Our data support the evidence that it is the quantity of sugar rather than the type that is critical in driving the steatotic response. Furthermore, we predict PPARα-mediated adaptations to hepatic lipid overload, shedding light on potential challenges for the use of PPARα agonists to treat NAFLD.

12.
CPT Pharmacometrics Syst Pharmacol ; 6(11): 732-746, 2017 11.
Article in English | MEDLINE | ID: mdl-28782239

ABSTRACT

The scope of physiologically based pharmacokinetic (PBPK) modeling can be expanded by assimilation of the mechanistic models of intracellular processes from systems biology field. The genome scale metabolic networks (GSMNs) represent a whole set of metabolic enzymes expressed in human tissues. Dynamic models of the gene regulation of key drug metabolism enzymes are available. Here, we introduce GSMNs and review ongoing work on integration of PBPK, GSMNs, and metabolic gene regulation. We demonstrate example models.


Subject(s)
Gene Expression Regulation , Metabolic Networks and Pathways , Algorithms , Computer Simulation , Genome, Human , Humans , Metabolic Clearance Rate , Models, Biological , Pharmacokinetics
13.
Int J Cancer ; 139(7): 1608-17, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27225067

ABSTRACT

HOX genes are vital for all aspects of mammalian growth and differentiation, and their dysregulated expression is related to ovarian carcinogenesis. The aim of the current study was to establish the prognostic value of HOX dysregulation as well as its role in platinum resistance. The potential to target HOX proteins through the HOX/PBX interaction was also explored in the context of platinum resistance. HOX gene expression was determined in ovarian cancer cell lines and primary EOCs by QPCR, and compared to expression in normal ovarian epithelium and fallopian tube tissue samples. Statistical analysis included one-way ANOVA and t-tests, using statistical software R and GraphPad. The analysis identified 36 of the 39 HOX genes as being overexpressed in high grade serous EOC compared to normal tissue. We detected a molecular HOX gene-signature that predicted poor outcome. Overexpression of HOXB4 and HOXB9 was identified in high grade serous cell lines after platinum resistance developed. Targeting the HOX/PBX dimer with the HXR9 peptide enhanced the cytotoxicity of cisplatin in platinum-resistant ovarian cancer. In conclusion, this study has shown the HOX genes are highly dysregulated in ovarian cancer with high expression of HOXA13, B6, C13, D1 and D13 being predictive of poor clinical outcome. Targeting the HOX/PBX dimer in platinum-resistant cancer represents a potentially new therapeutic option that should be further developed and tested in clinical trials.


Subject(s)
Adenocarcinoma/genetics , Genes, Homeobox , Ovarian Neoplasms/genetics , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Animals , Apoptosis/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Organoplatinum Compounds/pharmacology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Prognosis
14.
Sci Rep ; 6: 19771, 2016 Jan 27.
Article in English | MEDLINE | ID: mdl-26813959

ABSTRACT

A major roadblock in the effective treatment of cancers is their heterogeneity, whereby multiple molecular landscapes are classified as a single disease. To explore the contribution of cellular metabolism to cancer heterogeneity, we analyse the Metabric dataset, a landmark genomic and transcriptomic study of 2,000 individual breast tumours, in the context of the human genome-scale metabolic network. We create personalized metabolic landscapes for each tumour by exploring sets of active reactions that satisfy constraints derived from human biochemistry and maximize congruency with the Metabric transcriptome data. Classification of the personalized landscapes derived from 997 tumour samples within the Metabric discovery dataset reveals a novel poor prognosis cluster, reproducible in the 995-sample validation dataset. We experimentally follow mechanistic hypotheses resulting from the computational study and establish that active serotonin production is a major metabolic feature of the poor prognosis group. These data support the reconsideration of concomitant serotonin-specific uptake inhibitors treatment during breast cancer chemotherapy.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Metabolome , Metabolomics , Serotonin/biosynthesis , Biomarkers, Tumor , Breast Neoplasms/genetics , Cell Line, Tumor , Cluster Analysis , Computational Biology/methods , Extracellular Matrix , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Metabolomics/methods , Models, Biological , Prognosis , Transcriptome
15.
NPJ Syst Biol Appl ; 2: 16032, 2016.
Article in English | MEDLINE | ID: mdl-28725480

ABSTRACT

Systems Biology has established numerous approaches for mechanistic modeling of molecular networks in the cell and a legacy of models. The current frontier is the integration of models expressed in different formalisms to address the multi-scale biological system organization challenge. We present MUFINS (MUlti-Formalism Interaction Network Simulator) software, implementing a unique set of approaches for multi-formalism simulation of interaction networks. We extend the constraint-based modeling (CBM) framework by incorporation of linear inhibition constraints, enabling for the first time linear modeling of networks simultaneously describing gene regulation, signaling and whole-cell metabolism at steady state. We present a use case where a logical hypergraph model of a regulatory network is expressed by linear constraints and integrated with a Genome-Scale Metabolic Network (GSMN) of mouse macrophage. We experimentally validate predictions, demonstrating application of our software in an iterative cycle of hypothesis generation, validation and model refinement. MUFINS incorporates an extended version of our Quasi-Steady State Petri Net approach to integrate dynamic models with CBM, which we demonstrate through a dynamic model of cortisol signaling integrated with the human Recon2 GSMN and a model of nutrient dynamics in physiological compartments. Finally, we implement a number of methods for deriving metabolic states from ~omics data, including our new variant of the iMAT congruency approach. We compare our approach with iMAT through the analysis of 262 individual tumor transcriptomes, recovering features of metabolic reprogramming in cancer. The software provides graphics user interface with network visualization, which facilitates use by researchers who are not experienced in coding and mathematical modeling environments.

16.
PLoS One ; 10(10): e0139507, 2015.
Article in English | MEDLINE | ID: mdl-26469081

ABSTRACT

An understanding of the dynamics of the metabolic profile of a bacterial cell is sought from a dynamical systems analysis of kinetic models. This modelling formalism relies on a deterministic mathematical description of enzyme kinetics and their metabolite regulation. However, it is severely impeded by the lack of available kinetic information, limiting the size of the system that can be modelled. Furthermore, the subsystem of the metabolic network whose dynamics can be modelled is faced with three problems: how to parameterize the model with mostly incomplete steady state data, how to close what is now an inherently open system, and how to account for the impact on growth. In this study we address these challenges of kinetic modelling by capitalizing on multi-'omics' steady state data and a genome-scale metabolic network model. We use these to generate parameters that integrate knowledge embedded in the genome-scale metabolic network model, into the most comprehensive kinetic model of the central carbon metabolism of E. coli realized to date. As an application, we performed a dynamical systems analysis of the resulting enriched model. This revealed bistability of the central carbon metabolism and thus its potential to express two distinct metabolic states. Furthermore, since our model-informing technique ensures both stable states are constrained by the same thermodynamically feasible steady state growth rate, the ensuing bistability represents a temporal coexistence of the two states, and by extension, reveals the emergence of a phenotypically heterogeneous population.


Subject(s)
Escherichia coli/genetics , Escherichia coli/metabolism , Genomics , Metabolic Flux Analysis , Models, Biological , Carbon/metabolism , Genome, Bacterial/genetics , Kinetics
17.
BMC Genomics ; 16: 372, 2015 May 09.
Article in English | MEDLINE | ID: mdl-25956932

ABSTRACT

BACKGROUND: Mycobacterium tuberculosis continues to kill more people than any other bacterium. Although its archetypal host cell is the macrophage, it also enters, and survives within, dendritic cells (DCs). By modulating the behaviour of the DC, M. tuberculosis is able to manipulate the host's immune response and establish an infection. To identify the M. tuberculosis genes required for survival within DCs we infected primary human DCs with an M. tuberculosis transposon library and identified mutations with a reduced ability to survive. RESULTS: Parallel sequencing of the transposon inserts of the surviving mutants identified a large number of genes as being required for optimal intracellular fitness in DCs. Loci whose mutation attenuated intracellular survival included those involved in synthesising cell wall lipids, not only the well-established virulence factors, pDIM and cord factor, but also sulfolipids and PGL, which have not previously been identified as having a direct virulence role in cells. Other attenuated loci included the secretion systems ESX-1, ESX-2 and ESX-4, alongside many PPE genes, implicating a role for ESX-5. In contrast the canonical ESAT-6 family of ESX substrates did not have intra-DC fitness costs suggesting an alternative ESX-1 associated virulence mechanism. With the aid of a gene-nutrient interaction model, metabolic processes such as cholesterol side chain catabolism, nitrate reductase and cysteine-methionine metabolism were also identified as important for survival in DCs. CONCLUSION: We conclude that many of the virulence factors required for survival in DC are shared with macrophages, but that survival in DCs also requires several additional functions, such as cysteine-methionine metabolism, PGLs, sulfolipids, ESX systems and PPE genes.


Subject(s)
Dendritic Cells/microbiology , Genomics , Lipid Metabolism/genetics , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/pathogenicity , Type VII Secretion Systems/metabolism , Cell Wall/metabolism , Cholesterol/metabolism , DNA Transposable Elements/genetics , Genome, Bacterial/genetics , Humans , Macrophages/microbiology , Mutation , Mycobacterium tuberculosis/cytology , Mycobacterium tuberculosis/metabolism , Oxidative Stress/genetics , Phagosomes/microbiology , Reactive Nitrogen Species/metabolism , Virulence
18.
World J Gastroenterol ; 20(41): 15070-8, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25386055

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is a progressive disease of increasing public health concern. In western populations the disease has an estimated prevalence of 20%-40%, rising to 70%-90% in obese and type II diabetic individuals. Simplistically, NAFLD is the macroscopic accumulation of lipid in the liver, and is viewed as the hepatic manifestation of the metabolic syndrome. However, the molecular mechanisms mediating both the initial development of steatosis and its progression through non-alcoholic steatohepatitis to debilitating and potentially fatal fibrosis and cirrhosis are only partially understood. Despite increased research in this field, the development of non-invasive clinical diagnostic tools and the discovery of novel therapeutic targets has been frustratingly slow. We note that, to date, NAFLD research has been dominated by in vivo experiments in animal models and human clinical studies. Systems biology tools and novel computational simulation techniques allow the study of large-scale metabolic networks and the impact of their dysregulation on health. Here we review current systems biology tools and discuss the benefits to their application to the study of NAFLD. We propose that a systems approach utilising novel in silico modelling and simulation techniques is key to a more comprehensive, better targeted NAFLD research strategy. Such an approach will accelerate the progress of research and vital translation into clinic.


Subject(s)
Lipid Metabolism , Liver/metabolism , Models, Biological , Non-alcoholic Fatty Liver Disease/etiology , Systems Biology , Animals , Computer Simulation , Genetic Predisposition to Disease , Humans , Liver/pathology , Non-alcoholic Fatty Liver Disease/diagnosis , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/therapy , Phenotype , Prognosis , Risk Factors
19.
BMC Genomics ; 15: 270, 2014 Apr 08.
Article in English | MEDLINE | ID: mdl-24708363

ABSTRACT

BACKGROUND: Leprosy has afflicted humankind throughout history leaving evidence in both early texts and the archaeological record. In Britain, leprosy was widespread throughout the Middle Ages until its gradual and unexplained decline between the 14th and 16th centuries. The nature of this ancient endemic leprosy and its relationship to modern strains is only partly understood. Modern leprosy strains are currently divided into 5 phylogenetic groups, types 0 to 4, each with strong geographical links. Until recently, European strains, both ancient and modern, were thought to be exclusively type 3 strains. However, evidence for type 2 strains, a group normally associated with Central Asia and the Middle East, has recently been found in archaeological samples in Scandinavia and from two skeletons from the medieval leprosy hospital (or leprosarium) of St Mary Magdalen, near Winchester, England. RESULTS: Here we report the genotypic analysis and whole genome sequencing of two further ancient M. leprae genomes extracted from the remains of two individuals, Sk14 and Sk27, that were excavated from 10th-12th century burials at the leprosarium of St Mary Magdalen. DNA was extracted from the surfaces of bones showing osteological signs of leprosy. Known M. leprae polymorphisms were PCR amplified and Sanger sequenced, while draft genomes were generated by enriching for M. leprae DNA, and Illumina sequencing. SNP-typing and phylogenetic analysis of the draft genomes placed both of these ancient strains in the conserved type 2 group, with very few novel SNPs compared to other ancient or modern strains. CONCLUSIONS: The genomes of the two newly sequenced M. leprae strains group firmly with other type 2F strains. Moreover, the M. leprae strain most closely related to one of the strains, Sk14, in the worldwide phylogeny is a contemporaneous ancient St Magdalen skeleton, vividly illustrating the epidemic and clonal nature of leprosy at this site. The prevalence of these type 2 strains indicates that type 2F strains, in contrast to later European and associated North American type 3 isolates, may have been the co-dominant or even the predominant genotype at this location during the 11th century.


Subject(s)
Genome, Bacterial , Leprosy/microbiology , Mycobacterium leprae/genetics , Archaeology , Bone and Bones/microbiology , Epidemics , Evolution, Molecular , Genotype , History, 15th Century , History, 16th Century , History, Medieval , Humans , Leprosy/epidemiology , Leprosy/history , Mycobacterium leprae/classification , Mycobacterium leprae/isolation & purification , Osteology , Phylogeny , Polymorphism, Single Nucleotide , Sequence Analysis, DNA , Skeleton , United Kingdom/epidemiology
20.
PLoS One ; 8(9): e75913, 2013.
Article in English | MEDLINE | ID: mdl-24098743

ABSTRACT

The Mycobacterium tuberculosis complex includes bovine and human strains of the tuberculosis bacillus, including Mycobacterium tuberculosis, Mycobacterium bovis and the Mycobacterium bovis BCG vaccine strain. M. bovis has evolved from a M. tuberculosis-like ancestor and is the ancestor of the BCG vaccine. The pathogens demonstrate distinct differences in virulence, host range and metabolism, but the role of metabolic differences in pathogenicity is poorly understood. Systems biology approaches have been used to investigate the metabolism of M. tuberculosis, but not to probe differences between tuberculosis strains. In this study genome scale metabolic networks of M. bovis and M. bovis BCG were constructed and interrogated, along with a M. tuberculosis network, to predict substrate utilisation, gene essentiality and growth rates. The models correctly predicted 87-88% of high-throughput phenotype data, 75-76% of gene essentiality data and in silico-predicted growth rates matched measured rates. However, analysis of the metabolic networks identified discrepancies between in silico predictions and in vitro data, highlighting areas of incomplete metabolic knowledge. Additional experimental studies carried out to probe these inconsistencies revealed novel insights into the metabolism of these strains. For instance, that the reduction in metabolic capability observed in bovine tuberculosis strains, as compared to M. tuberculosis, is not reflected by current genetic or enzymatic knowledge. Hence, the in silico networks not only successfully simulate many aspects of the growth and physiology of these mycobacteria, but also provide an invaluable tool for future metabolic studies.


Subject(s)
Metabolic Networks and Pathways/genetics , Models, Biological , Mycobacterium bovis/metabolism , Mycobacterium tuberculosis/metabolism , Phenotype , Systems Biology/methods , Glucose/pharmacokinetics , Metabolic Networks and Pathways/physiology , Mycobacterium bovis/growth & development , Mycobacterium tuberculosis/growth & development , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL