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











Base de dados
Intervalo de ano de publicação
1.
Commun Biol ; 7(1): 634, 2024 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-38796621

RESUMO

Ebola virus (EBOV) matrix protein VP40 can assemble and bud as virus-like particles (VLPs) when expressed alone in mammalian cells. Nucleoprotein (NP) could be recruited to VLPs as inclusion body (IB) when co-expressed, and increase VLP production. However, the mechanism behind it remains unclear. Here, we use a computational approach to study NP-VP40 interactions. Our simulations indicate that NP may enhance VLP production through stabilizing VP40 filaments and accelerating the VLP budding step. Further, both the relative timing and amount of NP expression compared to VP40 are important for the effective production of IB-containing VLPs. We predict that relative NP/VP40 expression ratio and time are important for efficient production of IB-containing VLPs. We conclude that disrupting the expression timing and amount of NP and VP40 could provide new avenues to treat EBOV infection. This work provides quantitative insights into EBOV proteins interactions and how virion generation and drug efficacy could be influenced.


Assuntos
Ebolavirus , Proteínas do Core Viral , Ebolavirus/metabolismo , Proteínas do Core Viral/metabolismo , Proteínas do Core Viral/genética , Humanos , Vírion/metabolismo , Vírion/genética , Nucleoproteínas/metabolismo , Nucleoproteínas/genética , Proteínas da Matriz Viral/metabolismo , Proteínas da Matriz Viral/genética , Doença pelo Vírus Ebola/virologia , Doença pelo Vírus Ebola/metabolismo
2.
PLoS Comput Biol ; 19(8): e1011425, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37616311

RESUMO

Immunotherapeutic cytokines can activate immune cells against cancers and chronic infections. N-803 is an IL-15 superagonist that expands CD8+ T cells and increases their cytotoxicity. N-803 also temporarily reduced viral load in a limited subset of non-human primates infected with simian immunodeficiency virus (SIV), a model of HIV. However, viral suppression has not been observed in all SIV cohorts and may depend on pre-treatment viral load and the corresponding effects on CD8+ T cells. Starting from an existing mechanistic mathematical model of N-803 immunotherapy of SIV, we develop a model that includes activation of SIV-specific and non-SIV-specific CD8+ T cells by antigen, inflammation, and N-803. Also included is a regulatory counter-response that inhibits CD8+ T cell proliferation and function, representing the effects of immune checkpoint molecules and immunosuppressive cells. We simultaneously calibrate the model to two separate SIV cohorts. The first cohort had low viral loads prior to treatment (≈3-4 log viral RNA copy equivalents (CEQ)/mL), and N-803 treatment transiently suppressed viral load. The second had higher pre-treatment viral loads (≈5-7 log CEQ/mL) and saw no consistent virus suppression with N-803. The mathematical model can replicate the viral and CD8+ T cell dynamics of both cohorts based on different pre-treatment viral loads and different levels of regulatory inhibition of CD8+ T cells due to those viral loads (i.e. initial conditions of model). Our predictions are validated by additional data from these and other SIV cohorts. While both cohorts had high numbers of activated SIV-specific CD8+ T cells in simulations, viral suppression was precluded in the high viral load cohort due to elevated inhibition of cytotoxicity. Thus, we mathematically demonstrate how the pre-treatment viral load can influence immunotherapeutic efficacy, highlighting the in vivo conditions and combination therapies that could maximize efficacy and improve treatment outcomes.


Assuntos
Vírus da Imunodeficiência Símia , Animais , Interleucina-15 , Carga Viral , Imunoterapia , Linfócitos T CD8-Positivos
3.
Tuberculosis (Edinb) ; 139: 102304, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36682272

RESUMO

Non-tuberculous mycobacterial (NTM) infections, and Mycobacterium avium Complex (MAC) in particular, affect women at nearly twice the rate of men, and post-menopausal patients are at higher risk than pre-menopausal patients. The reasons for the disproportionate number of cases in women and post-menopausal patients remain unclear. One possibility is that menopause-associated immunological changes contribute to higher MAC prevalence post-menopause compared to pre-menopause. Menopause-associated immune disruption includes increased cytokine and chemokine production, and reduced cytotoxicity and phagocytosis in macrophages. Here we use an agent-based model of bacterial and host immune interactions in the airway to translate the combined impact of menopause-associated cellular immune disruptions to tissue scale outcomes. Our simulations indicate that menopause-associated immune disruptions can result in increased macrophage recruitment. However, this increase in macrophage number is unable to overcome functional deficits in macrophage phagocytosis and killing, since the post-menopausal simulations also show increased bacterial loads. Post-menopausal conditions are also associated with a lower number of cleared infections, and more simulations that have predominantly extracellular bacteria. Taken together, our work quantifies the potential impact of menopause-associated disruptions of innate immune functions on early MAC infection progression. Our findings will support the development of new therapies targeted to this high-risk group of patients.


Assuntos
Infecção por Mycobacterium avium-intracellulare , Mycobacterium tuberculosis , Masculino , Humanos , Feminino , Pós-Menopausa , Macrófagos/microbiologia , Complexo Mycobacterium avium
4.
Tuberculosis (Edinb) ; 138: 102300, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36621288

RESUMO

Incidence and prevalence of MAC infections are increasing globally, and reinfection is common. Thus, MAC infections present a significant public health challenge. We quantify the impact of MAC biofilms and repeated exposure on infection progression using a computational model of MAC infection in lung airways. MAC biofilms aid epithelial cell invasion, cause premature macrophage apoptosis, and limit antibiotic efficacy. In this computational work we develop an agent-based model that incorporates the interactions between bacteria, biofilm, and immune cells. In this computational model, we perform virtual knockouts to quantify the effects of the biofilm sources (deposited with bacteria vs. formed in the airway), and their impacts on macrophages (inducing apoptosis and slowing phagocytosis). We also quantify the effects of repeated bacterial exposures to assess their impact on infection progression. Our simulations show that chemoattractants released by biofilm-induced apoptosis bias macrophage chemotaxis towards pockets of infected and apoptosed macrophages. This bias results in fewer macrophages finding extracellular bacteria, allowing the extracellular planktonic bacteria to replicate freely. These spatial macrophage trends are further exacerbated with repeated deposition of bacteria. Our model indicates that interventions to abrogate macrophages' apoptotic responses to bacterial biofilms and/or reduce frequency of patient exposure to bacteria will lower bacterial load, and likely overall risk of infection.


Assuntos
Mycobacterium avium , Mycobacterium tuberculosis , Humanos , Carga Bacteriana , Macrófagos/microbiologia , Biofilmes , Pulmão , Complexo Mycobacterium avium
5.
Front Immunol ; 13: 1014515, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36405707

RESUMO

The risk of active tuberculosis disease is 15-21 times higher in those coinfected with human immunodeficiency virus-1 (HIV) compared to tuberculosis alone, and tuberculosis is the leading cause of death in HIV+ individuals. Mechanisms driving synergy between Mycobacterium tuberculosis (Mtb) and HIV during coinfection include: disruption of cytokine balances, impairment of innate and adaptive immune cell functionality, and Mtb-induced increase in HIV viral loads. Tuberculosis granulomas are the interface of host-pathogen interactions. Thus, granuloma-based research elucidating the role and relative impact of coinfection mechanisms within Mtb granulomas could inform cohesive treatments that target both pathogens simultaneously. We review known interactions between Mtb and HIV, and discuss how the structure, function and development of the granuloma microenvironment create a positive feedback loop favoring pathogen expansion and interaction. We also identify key outstanding questions and highlight how coupling computational modeling with in vitro and in vivo efforts could accelerate Mtb-HIV coinfection discoveries.


Assuntos
Coinfecção , Infecções por HIV , HIV-1 , Tuberculose , Humanos , Biologia de Sistemas , Granuloma , Infecções por HIV/complicações
6.
J Infect Dis ; 219(12): 1858-1866, 2019 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-30929010

RESUMO

Despite intensive research efforts, several fundamental disease processes for tuberculosis (TB) remain poorly understood. A central enigma is that host immunity is necessary to control disease yet promotes transmission by causing lung immunopathology. Our inability to distinguish these processes makes it challenging to design rational novel interventions. Elucidating basic immune mechanisms likely requires both in vivo and in vitro analyses, since Mycobacterium tuberculosis is a highly specialized human pathogen. The classic immune response is the TB granuloma organized in three dimensions within extracellular matrix. Several groups are developing cell culture granuloma models. In January 2018, NIAID convened a workshop, entitled "3-D Human in vitro TB Granuloma Model" to advance the field. Here, we summarize the arguments for developing advanced TB cell culture models and critically review those currently available. We discuss how integrating complementary approaches, specifically organoids and mathematical modeling, can maximize progress, and conclude by discussing future challenges and opportunities.


Assuntos
Granuloma/imunologia , Tuberculose/imunologia , Animais , Granuloma/microbiologia , Humanos , Modelos Teóricos , Mycobacterium tuberculosis/imunologia , Organoides/imunologia , Organoides/microbiologia , Tuberculose/microbiologia
7.
PLoS One ; 13(5): e0196322, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29746491

RESUMO

Drug resistant tuberculosis is increasing world-wide. Resistance against isoniazid (INH), rifampicin (RIF), or both (multi-drug resistant TB, MDR-TB) is of particular concern, since INH and RIF form part of the standard regimen for TB disease. While it is known that suboptimal treatment can lead to resistance, it remains unclear how host immune responses and antibiotic dynamics within granulomas (sites of infection) affect emergence and selection of drug-resistant bacteria. We take a systems pharmacology approach to explore resistance dynamics within granulomas. We integrate spatio-temporal host immunity, INH and RIF dynamics, and bacterial dynamics (including fitness costs and compensatory mutations) in a computational framework. We simulate resistance emergence in the absence of treatment, as well as resistance selection during INH and/or RIF treatment. There are four main findings. First, in the absence of treatment, the percentage of granulomas containing resistant bacteria mirrors the non-monotonic bacterial dynamics within granulomas. Second, drug-resistant bacteria are less frequently found in non-replicating states in caseum, compared to drug-sensitive bacteria. Third, due to a steeper dose response curve and faster plasma clearance of INH compared to RIF, INH-resistant bacteria have a stronger influence on treatment outcomes than RIF-resistant bacteria. Finally, under combination therapy with INH and RIF, few MDR bacteria are able to significantly affect treatment outcomes. Overall, our approach allows drug-specific prediction of drug resistance emergence and selection in the complex granuloma context. Since our predictions are based on pre-clinical data, our approach can be implemented relatively early in the treatment development process, thereby enabling pro-active rather than reactive responses to emerging drug resistance for new drugs. Furthermore, this quantitative and drug-specific approach can help identify drug-specific properties that influence resistance and use this information to design treatment regimens that minimize resistance selection and expand the useful life-span of new antibiotics.


Assuntos
Farmacorresistência Bacteriana/fisiologia , Tuberculose Resistente a Múltiplos Medicamentos/tratamento farmacológico , Antituberculosos/uso terapêutico , Simulação por Computador , Farmacorresistência Bacteriana/efeitos dos fármacos , Resistência a Múltiplos Medicamentos , Granuloma/tratamento farmacológico , Isoniazida/farmacologia , Testes de Sensibilidade Microbiana , Mycobacterium tuberculosis/genética , Rifampina/farmacologia , Tuberculose/tratamento farmacológico , Tuberculose/microbiologia , Tuberculose Resistente a Múltiplos Medicamentos/microbiologia
8.
Cell Mol Bioeng ; 10(6): 523-535, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29276546

RESUMO

INTRODUCTION: Tuberculosis (TB), one of the most common infectious diseases, requires treatment with multiple antibiotics taken over at least 6 months. This long treatment often results in poor patient-adherence, which can lead to the emergence of multi-drug resistant TB. New antibiotic treatment strategies are sorely needed. New antibiotics are being developed or repurposed to treat TB, but as there are numerous potential antibiotics, dosing sizes and potential schedules, the regimen design space for new treatments is too large to search exhaustively. Here we propose a method that combines an agent-based multi-scale model capturing TB granuloma formation with algorithms for mathematical optimization to identify optimal TB treatment regimens. METHODS: We define two different single-antibiotic treatments to compare the efficiency and accuracy in predicting optimal treatment regimens of two optimization algorithms: genetic algorithms (GA) and surrogate-assisted optimization through radial basis function (RBF) networks. We also illustrate the use of RBF networks to optimize double-antibiotic treatments. RESULTS: We found that while GAs can locate optimal treatment regimens more accurately, RBF networks provide a more practical strategy to TB treatment optimization with fewer simulations, and successfully estimated optimal double-antibiotic treatment regimens. CONCLUSIONS: Our results indicate surrogate-assisted optimization can locate optimal TB treatment regimens from a larger set of antibiotics, doses and schedules, and could be applied to solve optimization problems in other areas of research using systems biology approaches. Our findings have important implications for the treatment of diseases like TB that have lengthy protocols or for any disease that requires multiple drugs.

9.
PLoS Comput Biol ; 13(8): e1005650, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28817561

RESUMO

Granulomas are complex lung lesions that are the hallmark of tuberculosis (TB). Understanding antibiotic dynamics within lung granulomas will be vital to improving and shortening the long course of TB treatment. Three fluoroquinolones (FQs) are commonly prescribed as part of multi-drug resistant TB therapy: moxifloxacin (MXF), levofloxacin (LVX) or gatifloxacin (GFX). To date, insufficient data are available to support selection of one FQ over another, or to show that these drugs are clinically equivalent. To predict the efficacy of MXF, LVX and GFX at a single granuloma level, we integrate computational modeling with experimental datasets into a single mechanistic framework, GranSim. GranSim is a hybrid agent-based computational model that simulates granuloma formation and function, FQ plasma and tissue pharmacokinetics and pharmacodynamics and is based on extensive in vitro and in vivo data. We treat in silico granulomas with recommended daily doses of each FQ and compare efficacy by multiple metrics: bacterial load, sterilization rates, early bactericidal activity and efficacy under non-compliance and treatment interruption. GranSim reproduces in vivo plasma pharmacokinetics, spatial and temporal tissue pharmacokinetics and in vitro pharmacodynamics of these FQs. We predict that MXF kills intracellular bacteria more quickly than LVX and GFX due in part to a higher cellular accumulation ratio. We also show that all three FQs struggle to sterilize non-replicating bacteria residing in caseum. This is due to modest drug concentrations inside caseum and high inhibitory concentrations for this bacterial subpopulation. MXF and LVX have higher granuloma sterilization rates compared to GFX; and MXF performs better in a simulated non-compliance or treatment interruption scenario. We conclude that MXF has a small but potentially clinically significant advantage over LVX, as well as LVX over GFX. We illustrate how a systems pharmacology approach combining experimental and computational methods can guide antibiotic selection for TB.


Assuntos
Antituberculosos , Biologia Computacional/métodos , Simulação por Computador , Fluoroquinolonas , Granuloma , Mycobacterium tuberculosis , Tuberculose , Animais , Antituberculosos/administração & dosagem , Antituberculosos/farmacocinética , Antituberculosos/farmacologia , Antituberculosos/uso terapêutico , Feminino , Fluoroquinolonas/administração & dosagem , Fluoroquinolonas/farmacocinética , Fluoroquinolonas/farmacologia , Fluoroquinolonas/uso terapêutico , Granuloma/tratamento farmacológico , Granuloma/microbiologia , Humanos , Testes de Sensibilidade Microbiana , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/patogenicidade , Coelhos , Tuberculose/tratamento farmacológico , Tuberculose/microbiologia
10.
Curr Opin Syst Biol ; 3: 170-185, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30714019

RESUMO

Tuberculosis (TB) is an ancient and deadly disease characterized by complex host-pathogen dynamics playing out over multiple time and length scales and physiological compartments. Computational modeling can be used to integrate various types of experimental data and suggest new hypotheses, mechanisms, and therapeutic approaches to TB. Here, we offer a first-time comprehensive review of work on within-host TB models that describe the immune response of the host to infection, including the formation of lung granulomas. The models include systems of ordinary and partial differential equations and agent-based models as well as hybrid and multi-scale models that are combinations of these. Many aspects of M. tuberculosis infection, including host dynamics in the lung (typical site of infection for TB), granuloma formation, roles of cytokine and chemokine dynamics, and bacterial nutrient availability have been explored. Finally, we survey applications of these within-host models to TB therapy and prevention and suggest future directions to impact this global disease.

11.
Front Immunol ; 8: 1843, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29326718

RESUMO

Mycobacterium tuberculosis is the pathogenic bacterium that causes tuberculosis (TB), one of the most lethal infectious diseases in the world. The only vaccine against TB is minimally protective, and multi-drug resistant TB necessitates new therapeutics to treat infection. Developing new therapies requires a better understanding of the complex host immune response to infection, including dissecting the processes leading to formation of granulomas, the dense cellular lesions associated with TB. In this work, we pair experimental and computational modeling studies to explore cytokine regulation in the context of TB. We use our next-generation hybrid multi-scale model of granuloma formation (GranSim) to capture molecular, cellular, and tissue scale dynamics of granuloma formation. We identify TGF-ß1 as a major inhibitor of cytotoxic T-cell effector function in granulomas. Deletion of TGF-ß1 from the system results in improved bacterial clearance and lesion sterilization. We also identify a novel dichotomous regulation of cytotoxic T cells and macrophages by TGF-ß1 and IL-10, respectively. These findings suggest that increasing cytotoxic T-cell effector functions may increase bacterial clearance in granulomas and highlight potential new therapeutic targets for treating TB.

12.
Infect Immun ; 84(5): 1650-1669, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26975995

RESUMO

Granulomas are a hallmark of tuberculosis. Inside granulomas, the pathogen Mycobacterium tuberculosis may enter a metabolically inactive state that is less susceptible to antibiotics. Understanding M. tuberculosis metabolism within granulomas could contribute to reducing the lengthy treatment required for tuberculosis and provide additional targets for new drugs. Two key adaptations of M. tuberculosis are a nonreplicating phenotype and accumulation of lipid inclusions in response to hypoxic conditions. To explore how these adaptations influence granuloma-scale outcomes in vivo, we present a multiscale in silico model of granuloma formation in tuberculosis. The model comprises host immunity, M. tuberculosis metabolism, M. tuberculosis growth adaptation to hypoxia, and nutrient diffusion. We calibrated our model to in vivo data from nonhuman primates and rabbits and apply the model to predict M. tuberculosis population dynamics and heterogeneity within granulomas. We found that bacterial populations are highly dynamic throughout infection in response to changing oxygen levels and host immunity pressures. Our results indicate that a nonreplicating phenotype, but not lipid inclusion formation, is important for long-term M. tuberculosis survival in granulomas. We used virtual M. tuberculosis knockouts to predict the impact of both metabolic enzyme inhibitors and metabolic pathways exploited to overcome inhibition. Results indicate that knockouts whose growth rates are below ∼66% of the wild-type growth rate in a culture medium featuring lipid as the only carbon source are unable to sustain infections in granulomas. By mapping metabolite- and gene-scale perturbations to granuloma-scale outcomes and predicting mechanisms of sterilization, our method provides a powerful tool for hypothesis testing and guiding experimental searches for novel antituberculosis interventions.


Assuntos
Adaptação Fisiológica , Simulação por Computador , Granuloma/microbiologia , Granuloma/patologia , Mycobacterium tuberculosis/imunologia , Mycobacterium tuberculosis/fisiologia , Tuberculose/patologia , Animais , Carbono/metabolismo , Modelos Animais de Doenças , Metabolismo dos Lipídeos , Redes e Vias Metabólicas/genética , Viabilidade Microbiana , Mycobacterium tuberculosis/crescimento & desenvolvimento , Mycobacterium tuberculosis/metabolismo , Primatas , Coelhos , Tuberculose/microbiologia
13.
BMC Syst Biol ; 9: 79, 2015 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-26578235

RESUMO

BACKGROUND: Improvement in tuberculosis treatment regimens requires selection of antibiotics and dosing schedules from a large design space of possibilities. Incomplete knowledge of antibiotic and host immune dynamics in tuberculosis granulomas impacts clinical trial design and success, and variations among clinical trials hamper side-by-side comparison of regimens. Our objective is to systematically evaluate the efficacy of isoniazid and rifampin regimens, and identify modifications to these antibiotics that improve treatment outcomes. RESULTS: We pair a spatio-temporal computational model of host immunity with pharmacokinetic and pharmacodynamic data on isoniazid and rifampin. The model is calibrated to plasma pharmacokinetic and granuloma bacterial load data from non-human primate models of tuberculosis and to tissue and granuloma measurements of isoniazid and rifampin in rabbit granulomas. We predict the efficacy of regimens containing different doses and frequencies of isoniazid and rifampin. We predict impacts of pharmacokinetic/pharmacodynamic modifications on antibiotic efficacy. We demonstrate that suboptimal antibiotic concentrations within granulomas lead to poor performance of intermittent regimens compared to daily regimens. Improvements from dose and frequency changes are limited by inherent antibiotic properties, and we propose that changes in intracellular accumulation ratios and antimicrobial activity would lead to the most significant improvements in treatment outcomes. Results suggest that an increased risk of drug resistance in fully intermittent as compared to daily regimens arises from higher bacterial population levels early during treatment. CONCLUSIONS: Our systems pharmacology approach complements efforts to accelerate tuberculosis therapeutic development.


Assuntos
Antituberculosos/farmacocinética , Simulação por Computador , Isoniazida/farmacocinética , Rifampina/farmacocinética , Tuberculose/tratamento farmacológico , Animais , Antituberculosos/administração & dosagem , Antituberculosos/uso terapêutico , Farmacorresistência Bacteriana , Granuloma/tratamento farmacológico , Granuloma/microbiologia , Isoniazida/administração & dosagem , Isoniazida/uso terapêutico , Mycobacterium tuberculosis/crescimento & desenvolvimento , Rifampina/administração & dosagem , Rifampina/uso terapêutico , Resultado do Tratamento
14.
Integr Biol (Camb) ; 7(5): 591-609, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25924949

RESUMO

Approximately one third of the world's population is infected with Mycobacterium tuberculosis. Limited information about how the immune system fights M. tuberculosis and what constitutes protection from the bacteria impact our ability to develop effective therapies for tuberculosis. We present an in vivo systems biology approach that integrates data from multiple model systems and over multiple length and time scales into a comprehensive multi-scale and multi-compartment view of the in vivo immune response to M. tuberculosis. We describe computational models that can be used to study (a) immunomodulation with the cytokines tumor necrosis factor and interleukin 10, (b) oral and inhaled antibiotics, and


Assuntos
Tuberculose/terapia , Administração por Inalação , Administração Oral , Animais , Antibacterianos/administração & dosagem , Biomarcadores , Biologia Computacional , Simulação por Computador , Citocinas/metabolismo , Sistemas de Liberação de Medicamentos , Desenho de Fármacos , Granuloma/tratamento farmacológico , Humanos , Sistema Imunitário , Interleucina-10/metabolismo , Macrófagos/efeitos dos fármacos , Mycobacterium tuberculosis , Linguagens de Programação , Biologia de Sistemas , Fator de Necrose Tumoral alfa
15.
J Theor Biol ; 367: 166-179, 2015 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-25497475

RESUMO

While active tuberculosis (TB) is a treatable disease, many complex factors prevent its global elimination. Part of the difficulty in developing optimal therapies is the large design space of antibiotic doses, regimens and combinations. Computational models that capture the spatial and temporal dynamics of antibiotics at the site of infection can aid in reducing the design space of costly and time-consuming animal pre-clinical and human clinical trials. The site of infection in TB is the granuloma, a collection of immune cells and bacteria that form in the lung, and new data suggest that penetration of drugs throughout granulomas is problematic. Here we integrate our computational model of granuloma formation and function with models for plasma pharmacokinetics, lung tissue pharmacokinetics and pharmacodynamics for two first line anti-TB antibiotics. The integrated model is calibrated to animal data. We make four predictions. First, antibiotics are frequently below effective concentrations inside granulomas, leading to bacterial growth between doses and contributing to the long treatment periods required for TB. Second, antibiotic concentration gradients form within granulomas, with lower concentrations toward their centers. Third, during antibiotic treatment, bacterial subpopulations are similar for INH and RIF treatment: mostly intracellular with extracellular bacteria located in areas non-permissive for replication (hypoxic areas), presenting a slowly increasing target population over time. Finally, we find that on an individual granuloma basis, pre-treatment infection severity (including bacterial burden, host cell activation and host cell death) is predictive of treatment outcome.


Assuntos
Antibacterianos/uso terapêutico , Simulação por Computador , Imunidade/efeitos dos fármacos , Tuberculose/tratamento farmacológico , Tuberculose/imunologia , Animais , Antibacterianos/farmacocinética , Antibacterianos/farmacologia , Antituberculosos/farmacocinética , Antituberculosos/farmacologia , Antituberculosos/uso terapêutico , Calibragem , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Granuloma/imunologia , Granuloma/patologia , Humanos , Isoniazida/farmacocinética , Isoniazida/uso terapêutico , Camundongos , Modelos Biológicos , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/crescimento & desenvolvimento , Primatas , Rifampina/farmacocinética , Rifampina/uso terapêutico , Fatores de Tempo , Resultado do Tratamento , Tuberculose/microbiologia , Tuberculose/patologia
16.
PLoS One ; 9(11): e112426, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25386849

RESUMO

The standard treatment of tuberculosis (TB) takes six to nine months to complete and this lengthy therapy contributes to the emergence of drug-resistant TB. TB is caused by Mycobacterium tuberculosis (Mtb) and the ability of this bacterium to switch to a dormant phenotype has been suggested to be responsible for the slow clearance during treatment. A recent study showed that the replication rate of a non-virulent mycobacterium, Mycobacterium smegmatis, did not correlate with antibiotic susceptibility. However, the question whether this observation also holds true for Mtb remains unanswered. Here, in order to mimic physiological conditions of TB infection, we established a protocol based on long-term infection of primary human macrophages, featuring Mtb replicating at different rates inside the cells. During conditions that restricted Mtb replication, the bacterial phenotype was associated with reduced acid-fastness. However, these phenotypically altered bacteria were as sensitive to isoniazid, pyrazinamide and ethambutol as intracellularly replicating Mtb. In support of the recent findings with M. smegmatis, we conclude that replication rates of Mtb do not correlate with antibiotic tolerance.


Assuntos
Antituberculosos/farmacologia , Carga Bacteriana , Farmacorresistência Bacteriana , Macrófagos/microbiologia , Mycobacterium tuberculosis/crescimento & desenvolvimento , Antituberculosos/uso terapêutico , Proliferação de Células , Replicação do DNA , Humanos , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/patogenicidade , Fenótipo , Fatores de Tempo , Tuberculose/tratamento farmacológico , Tuberculose/microbiologia
17.
J Ethnopharmacol ; 157: 134-9, 2014 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-25261689

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: The emergence of multidrug-resistant strains of Mycobacterium tuberculosis underscores the need for continuous development of new and efficient methods to determine the susceptibility of isolates of Mycobacterium tuberculosis in the search for novel antimycobacterial agents. Natural products constitute an important source of new drugs, and design and implementation of antimycobacterial susceptibility testing methods are necessary to evaluate the different extracts and compounds. In this study we have explored the antimycobacterial properties of 50 ethanolic extracts from different parts of 46 selected medicinal plants traditionally used in Sudan to treat infectious diseases. MATERIALS AND METHODS: Plants were harvested and ethanolic extracts were prepared. For selected extracts, fractionation with hydrophilic and hydrophobic solvents was undertaken. A luminometry-based assay was used for determination of mycobacterial growth in broth cultures and inside primary human macrophages in the presence or absence of plant extracts and fractions of extracts. Cytotoxicity was also assessed for active fractions of plant extracts. RESULTS: Of the tested extracts, three exhibited a significant inhibitory effect on an avirulent strain of Mycobacterium tubercluosis (H37Ra) at the initial screening doses (125 and 6.25µg/ml). These were bark and leaf extracts of Khaya senegalensis and the leaf extract of Rosmarinus officinalis L. Further fractions of these plant extracts were prepared with n-hexane, chloroform, ethyl acetate, n-butanol, ethanol and water, and the activity of these extracts was retained in hydrophobic fractions. Cytotoxicity assays revealed that the chloroform fraction of Khaya senegalensis bark was non-toxic to human monocyte-derived macrophages and other cell types at the concentrations used and hence, further analysis, including assessment of IC50 and intracellular activity was done with this fraction. CONCLUSION: These results encourage further investigations to identify the active compound(s) within the chloroform fraction of Khaya senegalensis bark.


Assuntos
Antituberculosos/farmacologia , Mycobacterium tuberculosis/efeitos dos fármacos , Extratos Vegetais/farmacologia , Plantas Medicinais/química , Antituberculosos/administração & dosagem , Antituberculosos/isolamento & purificação , Relação Dose-Resposta a Droga , Farmacorresistência Bacteriana Múltipla , Humanos , Concentração Inibidora 50 , Macrófagos/microbiologia , Medicinas Tradicionais Africanas , Testes de Sensibilidade Microbiana , Extratos Vegetais/administração & dosagem , Sudão
18.
J Theor Biol ; 342: 23-32, 2014 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-24112967

RESUMO

There is a large body of literature describing molecular level interactions between Mycobacterium tuberculosis (Mtb) and macrophages. Macrophages initiate a range of anti-bacterial mechanisms in response to infection, and Mtb is capable of surviving and circumventing many of these responses. We apply a computational approach to ask: what are the effects on the cellular level of these opposing interactions? The model considers the interplay between bacterial killing and the pathogen's interference with macrophage function. The results reveal an oscillating balance between host and pathogen, but the balance is transient and varies in length, indicating that stochasticity in the bacterial population or host response could contribute to the diverse incubation periods observed in exposed individuals. The model captures host and strain variation and gives new insight into host-pathogen compatibility and co-evolution.


Assuntos
Interações Hospedeiro-Patógeno , Macrófagos/microbiologia , Macrófagos/patologia , Modelos Biológicos , Mycobacterium tuberculosis/fisiologia , Contagem de Colônia Microbiana , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade , Macrófagos/imunologia , Viabilidade Microbiana , Mycobacterium tuberculosis/crescimento & desenvolvimento , Mycobacterium tuberculosis/patogenicidade , Tuberculose/microbiologia , Tuberculose/patologia , Virulência
19.
J Theor Biol ; 251(4): 616-27, 2008 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-18237749

RESUMO

The tri-frame model gives mathematical expression to the transcription and translation processes, and considers all three reading frames (RFs). RNA polymerases transcribe DNA in single nucleotide increments, but ribosomes translate mRNA in pairings of three (triplets or codons). The set of triplets in the mRNA, starting with the initiation codon (usually AUG) defines the open reading frame (ORF). Since ribosomes do not always translocate three nucleotide positions, two additional RFs are accessible. The -1 RF and the +1 RF are triplet pairings of the mRNA, which are accessed by shifting one nucleotide position in the 5' and 3' directions, respectively. Transcription is modeled as a linear operator that maps the initial codons in all three frames into other codon sets to account for possible transcriptional errors. Translational errors (missense errors) originate from misacylation of tRNAs and misreading of aa-tRNAs by the ribosome. Translation is modeled as a linear mapping from codons into aa-tRNA species, which includes misreading errors. A final transformation from aa-tRNA species into amino acids provides the probability distributions of possible amino acids into which the codons in all three frames could be translated. An important element of the tri-frame model is the ribosomal occupancy probability. It is a vector in R(3) that gives the probability to find the ribosome in the ORF, -1 or +1 RF at each codon position. The sequence of vectors, from the first to the final codon position, gives a history of ribosome frameshifting. The model is powerful: it provides explicit expressions for (1) yield of error-free protein, (2) fraction of prematurely terminated polypeptides, (3) number of transcription errors, (4) number of translation errors and (5) mutations due to frameshifting. The theory is demonstrated for the three genes rpsU, dnaG and rpoD of Escherichia coli, which lie on the same operon, as well as for the prfB gene.


Assuntos
Modelos Genéticos , Fases de Leitura Aberta , Aminoácidos/genética , Animais , Mutação da Fase de Leitura , Humanos , Biossíntese de Proteínas , Transcrição Gênica
20.
Comput Biol Chem ; 31(5-6): 335-46, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17897886

RESUMO

It is generally accepted that the translation rate depends on the availability of cognate aa-tRNAs. In this study it is shown that the key factor that determines translation rate is the competition between near-cognate and cognate aa-tRNAs. The transport mechanism in the cytoplasm is diffusion, thus the competition between cognate, near-cognate and non-cognate aa-tRNAs to bind to the ribosome is a stochastic process. Two competition measures are introduced; C(i) and R(i) (i=1, 64) are quotients of the arrival frequencies of near-cognates vs. cognates and non-cognates vs. cognates, respectively. Furthermore, the reaction rates of bound cognates differ from those of bound near-cognates. If a near-cognate aa-tRNA binds to the A site of the ribosome, it may be rejected at the anti-codon recognition step or proofreading step or it may be accepted. Regardless of its fate, the near-cognates and non-cognates have caused delays of varying duration to the observed rate of translation. Rate constants have been measured at a temperature of 20 degrees C by (Gromadski, K.B., Rodnina, M.V., 2004. Kinetic determinants of high-fidelity tRNA discrimination on the ribosome. Mol. Cell 13, 191-200). These rate constants have been re-evaluated at 37 degrees C, using experimental data at 24.5 degrees C and 37 degrees C (Varenne, S., et al., 1984. Translation in a non-uniform process: effect of tRNA availability on the rate of elongation of nascent polypeptide chains. J. Mol. Biol. 180, 549-576). The key results of the study are: (i) the average time (at 37 degrees C) to add an amino acid, as defined by the ith codon, to the nascent peptide chain is: tau(i)=9.06+1.445x[10.48C(i)+0.5R(i)] (in ms); (ii) the misreading frequency is directly proportional to the near-cognate competition, E(i)=0.0009C(i); (iii) the competition from near-cognates, and not the availability of cognate aa-tRNAs, is the most important factor that determines the translation rate - the four codons with highest near-cognate competition (in the case of E. coli) are [GCC]>[CGG]>[AGG]>[GGA], which overlap only partially with the rarest codons: [AGG]<[CCA]<[GCC]<[CAC]; (iv) based on the kinetic rates at 37 degrees C, the average time to insert a cognate amino acid is 9.06ms and the average delay to process a near-cognate aa-tRNA is 10.45ms and (vii) the model also provides estimates of the vacancy times of the A site of the ribosome - an important factor in frameshifting.


Assuntos
Modelos Biológicos , Biossíntese de Proteínas , RNA de Transferência Aminoácido-Específico/metabolismo , Ribossomos/metabolismo , Algoritmos , Códon/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Cinética , Elongação Traducional da Cadeia Peptídica , Temperatura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA