Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Immunol Rev ; 285(1): 147-167, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30129209

RESUMO

Immune responses to pathogens are complex and not well understood in many diseases, and this is especially true for infections by persistent pathogens. One mechanism that allows for long-term control of infection while also preventing an over-zealous inflammatory response from causing extensive tissue damage is for the immune system to balance pro- and anti-inflammatory cells and signals. This balance is dynamic and the immune system responds to cues from both host and pathogen, maintaining a steady state across multiple scales through continuous feedback. Identifying the signals, cells, cytokines, and other immune response factors that mediate this balance over time has been difficult using traditional research strategies. Computational modeling studies based on data from traditional systems can identify how this balance contributes to immunity. Here we provide evidence from both experimental and mathematical/computational studies to support the concept of a dynamic balance operating during persistent and other infection scenarios. We focus mainly on tuberculosis, currently the leading cause of death due to infectious disease in the world, and also provide evidence for other infections. A better understanding of the dynamically balanced immune response can help shape treatment strategies that utilize both drugs and host-directed therapies.


Assuntos
Biologia Computacional/métodos , Inflamação/imunologia , Pulmão/patologia , Modelos Imunológicos , Mycobacterium tuberculosis/fisiologia , Tuberculose/imunologia , Animais , Antituberculosos/uso terapêutico , Retroalimentação Fisiológica , Humanos , Inflamação/terapia , Pulmão/efeitos dos fármacos , Modelos Teóricos , Transdução de Sinais , Tuberculose/terapia
2.
PLoS Comput Biol ; 16(2): e1007684, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32058996

RESUMO

Identification of differentially expressed genes (DEGs) is well recognized to be variable across independent replications of genome-wide transcriptional studies. These are often employed to characterize disease state early in the process of discovery and prioritize novel targets aimed at addressing unmet medical need. Increasing reproducibility of biological findings from these studies could potentially positively impact the success rate of new clinical interventions. This work demonstrates that statistically sound combination of gene expression data with prior knowledge about biology in the form of large protein interaction networks can yield quantitatively more reproducible observations from studies characterizing human disease. The novel concept of Well-Associated Proteins (WAPs) introduced herein-gene products significantly associated on protein interaction networks with the differences in transcript levels between control and disease-does not require choosing a differential expression threshold and can be computed efficiently enough to enable false discovery rate estimation via permutation. Reproducibility of WAPs is shown to be on average superior to that of DEGs under easily-quantifiable conditions suggesting that they can yield a significantly more robust description of disease. Enhanced reproducibility of WAPs versus DEGs is first demonstrated with four independent data sets focused on systemic sclerosis. This finding is then validated over thousands of pairs of data sets obtained by random partitions of large studies in several other diseases. Conditions that individual data sets must satisfy to yield robust WAP scores are examined. Reproducible identification of WAPs can potentially benefit drug target selection and precision medicine studies.


Assuntos
Biologia Computacional/métodos , Perfilação da Expressão Gênica , Mapas de Interação de Proteínas , Proteínas/química , Área Sob a Curva , Reações Falso-Positivas , Regulação da Expressão Gênica , Humanos , Modelos Lineares , Análise Multivariada , Medicina de Precisão , Probabilidade , Reprodutibilidade dos Testes , Escleroderma Sistêmico/genética
3.
J Chem Inf Model ; 59(2): 673-688, 2019 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-30642173

RESUMO

Chemical synthesis planning is a key aspect in many fields of chemistry, especially drug discovery. Recent implementations of machine learning and artificial intelligence techniques for retrosynthetic analysis have shown great potential to improve computational methods for synthesis planning. Herein, we present a multiscale, data-driven approach for retrosynthetic analysis with deep highway networks (DHN). We automatically extracted reaction rules (i.e., ways in which a molecule is produced) from a data set consisting of chemical reactions derived from U.S. patents. We performed the retrosynthetic reaction prediction task in two steps: first, we built a DHN model to predict which group of reactions (consisting of chemically similar reaction rules) was employed to produce a molecule. Once a reaction group was identified, a DHN trained on the subset of reactions within the identified reaction group, was employed to predict the transformation rule used to produce a molecule. To validate our approach, we predicted the first retrosynthetic reaction step for 40 approved drugs using our multiscale model and compared its predictive performance with a conventional model trained on all machine-extracted reaction rules employed as a control. Our multiscale approach showed a success rate of 82.9% at generating valid reactants from retrosynthetic reaction predictions. Comparatively, the control model trained on all machine-extracted reaction rules yielded a success rate of 58.5% on the validation set of 40 pharmaceutical molecules, indicating a significant statistical improvement with our approach to match known first synthetic reaction of the tested drugs in this study. While our multiscale approach was unable to outperform state-of-the-art rule-based systems curated by expert chemists, multiscale classification represents a marked enhancement in retrosynthetic analysis and can be easily adapted for use in a range of artificial intelligence strategies.


Assuntos
Quimioinformática/métodos , Aprendizado Profundo , Técnicas de Química Sintética , Bases de Dados de Produtos Farmacêuticos , Descoberta de Drogas , Patentes como Assunto , Estados Unidos
4.
J Immunol ; 194(2): 664-77, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25512604

RESUMO

Although almost a third of the world's population is infected with the bacterial pathogen Mycobacterium tuberculosis, our understanding of the functions of many immune factors involved in fighting infection is limited. Determining the role of the immunosuppressive cytokine IL-10 at the level of the granuloma has proven difficult because of lesional heterogeneity and the limitations of animal models. In this study, we take an in silico approach and, through a series of virtual experiments, we predict several novel roles for IL-10 in tuberculosis granulomas: 1) decreased levels of IL-10 lead to increased numbers of sterile lesions, but at the cost of early increased caseation; 2) small increases in early antimicrobial activity cause this increased lesion sterility; 3) IL-10 produced by activated macrophages is a major mediator of early antimicrobial activity and early host-induced caseation; and 4) increasing levels of infected macrophage derived IL-10 promotes bacterial persistence by limiting the early antimicrobial response and preventing lesion sterilization. Our findings, currently only accessible using an in silico approach, suggest that IL-10 at the individual granuloma scale is a critical regulator of lesion outcome. These predictions suggest IL-10-related mechanisms that could be used as adjunctive therapies during tuberculosis.


Assuntos
Interleucina-10/imunologia , Ativação de Macrófagos , Macrófagos/imunologia , Mycobacterium tuberculosis/imunologia , Tuberculose/imunologia , Animais , Granuloma/genética , Granuloma/imunologia , Granuloma/microbiologia , Humanos , Interleucina-10/genética , Tuberculose/genética
5.
Infect Immun ; 83(1): 324-38, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25368116

RESUMO

Mycobacterium tuberculosis, the causative agent of tuberculosis (TB), induces formation of granulomas, structures in which immune cells and bacteria colocalize. Macrophages are among the most abundant cell types in granulomas and have been shown to serve as both critical bactericidal cells and targets for M. tuberculosis infection and proliferation throughout the course of infection. Very little is known about how these processes are regulated, what controls macrophage microenvironment-specific polarization and plasticity, or why some granulomas control bacteria and others permit bacterial dissemination. We take a computational-biology approach to investigate mechanisms that drive macrophage polarization, function, and bacterial control in granulomas. We define a "macrophage polarization ratio" as a metric to understand how cytokine signaling translates into polarization of single macrophages in a granuloma, which in turn modulates cellular functions, including antimicrobial activity and cytokine production. Ultimately, we extend this macrophage ratio to the tissue scale and define a "granuloma polarization ratio" describing mean polarization measures for entire granulomas. Here we coupled experimental data from nonhuman primate TB granulomas to our computational model, and we predict two novel and testable hypotheses regarding macrophage profiles in TB outcomes. First, the temporal dynamics of granuloma polarization ratios are predictive of granuloma outcome. Second, stable necrotic granulomas with low CFU counts and limited inflammation are characterized by short NF-κB signal activation intervals. These results suggest that the dynamics of NF-κB signaling is a viable therapeutic target to promote M1 polarization early during infection and to improve outcome.


Assuntos
Granuloma/imunologia , Granuloma/microbiologia , Macrófagos/imunologia , Macrófagos/microbiologia , Tuberculose/imunologia , Tuberculose/microbiologia , Animais , Simulação por Computador , Modelos Animais de Doenças , Granuloma/patologia , Macaca fascicularis , NF-kappa B/imunologia , Tuberculose/patologia
6.
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
7.
J Clin Invest ; 130(3): 1233-1251, 2020 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-32039915

RESUMO

Smooth muscle cell (SMC) proliferation has been thought to limit the progression of thoracic aortic aneurysm and dissection (TAAD) because loss of medial cells associates with advanced disease. We investigated effects of SMC proliferation in the aortic media by conditional disruption of Tsc1, which hyperactivates mTOR complex 1. Consequent SMC hyperplasia led to progressive medial degeneration and TAAD. In addition to diminished contractile and synthetic functions, fate-mapped SMCs displayed increased proteolysis, endocytosis, phagocytosis, and lysosomal clearance of extracellular matrix and apoptotic cells. SMCs acquired a limited repertoire of macrophage markers and functions via biogenesis of degradative organelles through an mTOR/ß-catenin/MITF-dependent pathway, but were distinguishable from conventional macrophages by an absence of hematopoietic lineage markers and certain immune effectors even in the context of hyperlipidemia. Similar mTOR activation and induction of a degradative SMC phenotype in a model of mild TAAD due to Fbn1 mutation greatly worsened disease with near-uniform lethality. The finding of increased lysosomal markers in medial SMCs from clinical TAAD specimens with hyperplasia and matrix degradation further supports the concept that proliferation of degradative SMCs within the media causes aortic disease, thus identifying mTOR-dependent phenotypic modulation as a therapeutic target for combating TAAD.


Assuntos
Aorta/enzimologia , Aneurisma da Aorta Torácica/enzimologia , Dissecção Aórtica/enzimologia , Miócitos de Músculo Liso/enzimologia , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Dissecção Aórtica/genética , Dissecção Aórtica/patologia , Animais , Aorta/patologia , Aneurisma da Aorta Torácica/genética , Aneurisma da Aorta Torácica/patologia , Modelos Animais de Doenças , Lisossomos/enzimologia , Lisossomos/genética , Lisossomos/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Camundongos Knockout para ApoE , Fator de Transcrição Associado à Microftalmia/genética , Fator de Transcrição Associado à Microftalmia/metabolismo , Miócitos de Músculo Liso/patologia , Serina-Treonina Quinases TOR/genética , Proteína 1 do Complexo Esclerose Tuberosa/genética , Proteína 1 do Complexo Esclerose Tuberosa/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
8.
J Exp Med ; 216(8): 1874-1890, 2019 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-31196980

RESUMO

To define the role of ERK1/2 signaling in the quiescent endothelium, we induced endothelial Erk2 knockout in adult Erk1-/- mice. This resulted in a rapid onset of hypertension, a decrease in eNOS expression, and an increase in endothelin-1 plasma levels, with all mice dying within 5 wk. Immunostaining and endothelial fate mapping showed a robust increase in TGFß signaling leading to widespread endothelial-to-mesenchymal transition (EndMT). Fibrosis affecting the cardiac conduction system was responsible for the universal lethality in these mice. Other findings included renal endotheliosis, loss of fenestrated endothelia in endocrine organs, and hemorrhages. An ensemble computational intelligence strategy, comprising deep learning and probabilistic programing of RNA-seq data, causally linked the loss of ERK1/2 in HUVECs in vitro to activation of TGFß signaling, EndMT, suppression of eNOS, and induction of endothelin-1 expression. All in silico predictions were verified in vitro and in vivo. In summary, these data establish the key role played by ERK1/2 signaling in the maintenance of vascular normalcy.


Assuntos
Endotélio/metabolismo , Hipertensão/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Animais , Aprendizado Profundo , Modelos Animais de Doenças , Endotelina-1/metabolismo , Transição Epitelial-Mesenquimal/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/genética , Óxido Nítrico Sintase Tipo III/metabolismo , RNA-Seq , Transfecção , Fator de Crescimento Transformador beta/metabolismo
9.
Clin Pharmacol Ther ; 105(4): 1031-1039, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30402880

RESUMO

M281 is a fully human, anti-neonatal Fc receptor (FcRn) antibody that inhibits FcRn-mediated immunoglobulin G (IgG) recycling to decrease pathogenic IgG while preserving IgG production. A randomized, double-blind, placebo-controlled, first-in-human study with 50 normal healthy volunteers was designed to probe safety and the physiological maximum for reduction of IgG. Intravenous infusion of single ascending doses up to 60 mg/kg induced dose-dependent serum IgG reductions, which were similar across all IgG subclasses. Multiple weekly doses of 15 or 30 mg/kg achieved mean IgG reductions of ≈85% from baseline and maintained IgG reductions ≥75% from baseline for up to 24 days. M281 was well tolerated, with no serious or severe adverse events (AEs), few moderate AEs, and a low incidence of infection-related AEs similar to placebo treatment. The tolerability and consistency of M281 pharmacokinetics and pharmacodynamics support further evaluation of M281 in diseases mediated by pathogenic IgG.


Assuntos
Anticorpos/metabolismo , Anticorpos/uso terapêutico , Antígenos de Histocompatibilidade Classe I/metabolismo , Imunoglobulina G/metabolismo , Receptores Fc/metabolismo , Adulto , Anticorpos/efeitos adversos , Método Duplo-Cego , Feminino , Voluntários Saudáveis , Humanos , Infusões Intravenosas/métodos , Masculino , Adulto Jovem
10.
Arthritis Res Ther ; 21(1): 216, 2019 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-31647025

RESUMO

BACKGROUND: The goal of this study is to use comprehensive molecular profiling to characterize clinical response to anti-TNF therapy in a real-world setting and identify reproducible markers differentiating good responders and non-responders in rheumatoid arthritis (RA). METHODS: Whole-blood mRNA, plasma proteins, and glycopeptides were measured in two cohorts of biologic-naïve RA patients (n = 40 and n = 36) from the Corrona CERTAIN (Comparative Effectiveness Registry to study Therapies for Arthritis and Inflammatory coNditions) registry at baseline and after 3 months of anti-TNF treatment. Response to treatment was categorized by EULAR criteria. A cell type-specific data analysis was conducted to evaluate the involvement of the most common immune cell sub-populations. Findings concordant between the two cohorts were further assessed for reproducibility using selected NCBI-GEO datasets and clinical laboratory measurements available in the CERTAIN database. RESULTS: A treatment-related signature suggesting a reduction in neutrophils, independent of the status of response, was indicated by a high level of correlation (ρ = 0.62; p < 0.01) between the two cohorts. A baseline, response signature of increased innate cell types in responders compared to increased adaptive cell types in non-responders was identified in both cohorts. This result was further assessed by applying the cell type-specific analysis to five other publicly available RA datasets. Evaluation of the neutrophil-to-lymphocyte ratio at baseline in the remaining patients (n = 1962) from the CERTAIN database confirmed the observation (odds ratio of good/moderate response = 1.20 [95% CI = 1.03-1.41, p = 0.02]). CONCLUSION: Differences in innate/adaptive immune cell type composition at baseline may be a major contributor to response to anti-TNF treatment within the first 3 months of therapy.


Assuntos
Imunidade Adaptativa/fisiologia , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/genética , Perfilação da Expressão Gênica/métodos , Imunidade Inata/fisiologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Imunidade Adaptativa/efeitos dos fármacos , Adulto , Idoso , Antirreumáticos/farmacologia , Antirreumáticos/uso terapêutico , Artrite Reumatoide/imunologia , Estudos de Coortes , Feminino , Humanos , Imunidade Inata/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Resultado do Tratamento , Fator de Necrose Tumoral alfa/imunologia
11.
Nat Metab ; 1(9): 912-926, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31572976

RESUMO

Atherosclerosis is a progressive vascular disease triggered by interplay between abnormal shear stress and endothelial lipid retention. A combination of these and, potentially, other factors leads to a chronic inflammatory response in the vessel wall, which is thought to be responsible for disease progression characterized by a buildup of atherosclerotic plaques. Yet molecular events responsible for maintenance of plaque inflammation and plaque growth have not been fully defined. Here we show that endothelial TGFß signaling is one of the primary drivers of atherosclerosis-associated vascular inflammation. Inhibition of endothelial TGFß signaling in hyperlipidemic mice reduces vessel wall inflammation and vascular permeability and leads to arrest of disease progression and regression of established lesions. These pro-inflammatory effects of endothelial TGFß signaling are in stark contrast with its effects in other cell types and identify it as an important driver of atherosclerotic plaque growth and show the potential of cell-type specific therapeutic intervention aimed at control of this disease.


Assuntos
Aterosclerose/metabolismo , Endotélio Vascular/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Vasculite/metabolismo , Animais , Permeabilidade Capilar , Linhagem Celular , Progressão da Doença , Endotélio Vascular/patologia , Humanos , Camundongos , Camundongos Knockout , Fator de Crescimento Transformador beta/genética
12.
Obesity (Silver Spring) ; 24(1): 102-12, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26576827

RESUMO

OBJECTIVE: T cell inflammation plays pivotal roles in obesity-associated type 2 diabetes (T2DM). The identification of dominant sources of T cell inflammation in humans remains a significant gap in understanding disease pathogenesis. It was hypothesized that cytokine profiles from circulating T cells identify T cell subsets and T cell cytokines that define T2DM-associated inflammation. METHODS: Multiplex analyses were used to quantify T cell-associated cytokines in αCD3/αCD28-stimulated PBMCs, or B cell-depleted PBMCs, from subjects with T2DM or BMI-matched controls. Cytokine measurements were subjected to multivariate (principal component and partial least squares) analyses. Flow cytometry detected intracellular TNFα in multiple immune cell subsets in the presence/absence of antibodies that neutralize T cell cytokines. RESULTS: T cell cytokines were generally higher in T2DM samples, but Th17 cytokines are specifically important for classifying individuals correctly as T2DM. Multivariate analyses indicated that B cells support Th17 inflammation in T2DM but not control samples, while monocytes supported Th17 inflammation regardless of T2DM status. Partial least squares regression analysis indicated that both Th17 and Th1 cytokines impact %HbA1c. CONCLUSIONS: Among various T cell subsets, Th17 cells are major contributors to inflammation and hyperglycemia and are uniquely supported by B cells in obesity-associated T2DM.


Assuntos
Citocinas/imunologia , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/imunologia , Obesidade/complicações , Obesidade/imunologia , Células Th17/imunologia , Fator de Necrose Tumoral alfa/imunologia , Adulto , Idoso , Linfócitos B/imunologia , Células Cultivadas , Feminino , Humanos , Inflamação/complicações , Inflamação/imunologia , Leucócitos Mononucleares/imunologia , Masculino , Pessoa de Meia-Idade , Monócitos/imunologia , Adulto Jovem
13.
Cell Mol Bioeng ; 8(1): 119-136, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26366228

RESUMO

Biologically related processes operate across multiple spatiotemporal scales. For computational modeling methodologies to mimic this biological complexity, individual scale models must be linked in ways that allow for dynamic exchange of information across scales. A powerful methodology is to combine a discrete modeling approach, agent-based models (ABMs), with continuum models to form hybrid models. Hybrid multi-scale ABMs have been used to simulate emergent responses of biological systems. Here, we review two aspects of hybrid multi-scale ABMs: linking individual scale models and efficiently solving the resulting model. We discuss the computational choices associated with aspects of linking individual scale models while simultaneously maintaining model tractability. We demonstrate implementations of existing numerical methods in the context of hybrid multi-scale ABMs. Using an example model describing Mycobacterium tuberculosis infection, we show relative computational speeds of various combinations of numerical methods. Efficient linking and solution of hybrid multi-scale ABMs is key to model portability, modularity, and their use in understanding biological phenomena at a systems level.

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.
PLoS One ; 8(7): e68680, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23869227

RESUMO

Interleukin-10 (IL-10) and tumor necrosis factor-α (TNF-α) are key anti- and pro-inflammatory mediators elicited during the host immune response to Mycobacterium tuberculosis (Mtb). Understanding the opposing effects of these mediators is difficult due to the complexity of processes acting across different spatial (molecular, cellular, and tissue) and temporal (seconds to years) scales. We take an in silico approach and use multi-scale agent based modeling of the immune response to Mtb, including molecular scale details for both TNF-α and IL-10. Our model predicts that IL-10 is necessary to modulate macrophage activation levels and to prevent host-induced tissue damage in a granuloma, an aggregate of cells that forms in response to Mtb. We show that TNF-α and IL-10 parameters related to synthesis, signaling, and spatial distribution processes control concentrations of TNF-α and IL-10 in a granuloma and determine infection outcome in the long-term. We devise an overall measure of granuloma function based on three metrics - total bacterial load, macrophage activation levels, and apoptosis of resting macrophages - and use this metric to demonstrate a balance of TNF-α and IL-10 concentrations is essential to Mtb infection control, within a single granuloma, with minimal host-induced tissue damage. Our findings suggest that a balance of TNF-α and IL-10 defines a granuloma environment that may be beneficial for both host and pathogen, but perturbing the balance could be used as a novel therapeutic strategy to modulate infection outcomes.


Assuntos
Granuloma/patologia , Interleucina-10/fisiologia , Mycobacterium tuberculosis/imunologia , Tuberculose/patologia , Fator de Necrose Tumoral alfa/fisiologia , Biologia Computacional , Granuloma/metabolismo , Humanos , Interleucina-10/metabolismo , Macrófagos/imunologia , Modelos Teóricos , Transdução de Sinais , Tuberculose/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA