Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Microorganisms ; 11(12)2023 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-38138118

RESUMO

Influenza A virus (IAV) infections represent a substantial global health challenge and are often accompanied by coinfections involving secondary viruses or bacteria, resulting in increased morbidity and mortality. The clinical impact of coinfections remains poorly understood, with conflicting findings regarding fatality. Isolating the impact of each pathogen and mechanisms of pathogen synergy during coinfections is challenging and further complicated by host and pathogen variability and experimental conditions. Factors such as cytokine dysregulation, immune cell function alterations, mucociliary dysfunction, and changes to the respiratory tract epithelium have been identified as contributors to increased lethality. The relative significance of these factors depends on variables such as pathogen types, infection timing, sequence, and inoculum size. Mathematical biological modeling can play a pivotal role in shedding light on the mechanisms of coinfections. Mathematical modeling enables the quantification of aspects of the intra-host immune response that are difficult to assess experimentally. In this narrative review, we highlight important mechanisms of IAV coinfection with bacterial and viral pathogens and survey mathematical models of coinfection and the insights gained from them. We discuss current challenges and limitations facing coinfection modeling, as well as current trends and future directions toward a complete understanding of coinfection using mathematical modeling and computer simulation.

2.
Bull Math Biol ; 84(8): 88, 2022 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-35829841

RESUMO

Age-induced dysregulation of the immune response is a major contributor to the morbidity and mortality related to influenza a virus infections. Experimental data have shown substantial changes to the activation and maintenance of the immune response will occur with age, but it remains unclear which of these many interrelated changes are most critical to controlling the survival of the host during infection. To ascertain which mechanisms are predominantly responsible for the increased morbidity in elderly hosts, we developed an ordinary differential equation model to simulate the immune response to pandemic H1N1 infection. We fit this model to experimental data measured in young and old macaques. We determined that the severity of the infection in the elderly hosts is caused by a dysregulation in the innate immune response. We also simulated CD8+ T cell exhaustion, a common consequence of chronic and extensive infections. Our simulations indicate that while T cell exhaustion is possible in both age groups, its effects are more severe in the elderly population, as their dysregulated immune response cannot easily compensate for the exhausted T cells. Finally, we explore a therapeutic approach to reversing T cell exhaustion through an inflammatory stimulus. A controlled increase in inflammatory signals can lead to a higher chance of surviving the infection, but excess inflammation will likely lead to septic death. These results indicate that our model captures distinctions in the predominant mechanisms controlling the immune response in younger and older hosts and allows for simulations of clinically relevant therapeutic strategies post-infection.


Assuntos
Vírus da Influenza A Subtipo H1N1 , Influenza Humana , Idoso , Linfócitos T CD8-Positivos , Humanos , Vírus da Influenza A Subtipo H1N1/fisiologia , Influenza Humana/epidemiologia , Conceitos Matemáticos , Modelos Biológicos
3.
J Theor Biol ; 532: 110918, 2022 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-34592264

RESUMO

Respiratory viral infections pose a serious public health concern, from mild seasonal influenza to pandemics like those of SARS-CoV-2. Spatiotemporal dynamics of viral infection impact nearly all aspects of the progression of a viral infection, like the dependence of viral replication rates on the type of cell and pathogen, the strength of the immune response and localization of infection. Mathematical modeling is often used to describe respiratory viral infections and the immune response to them using ordinary differential equation (ODE) models. However, ODE models neglect spatially-resolved biophysical mechanisms like lesion shape and the details of viral transport, and so cannot model spatial effects of a viral infection and immune response. In this work, we develop a multiscale, multicellular spatiotemporal model of influenza infection and immune response by combining non-spatial ODE modeling and spatial, cell-based modeling. We employ cellularization, a recently developed method for generating spatial, cell-based, stochastic models from non-spatial ODE models, to generate much of our model from a calibrated ODE model that describes infection, death and recovery of susceptible cells and innate and adaptive responses during influenza infection, and develop models of cell migration and other mechanisms not explicitly described by the ODE model. We determine new model parameters to generate agreement between the spatial and original ODE models under certain conditions, where simulation replicas using our model serve as microconfigurations of the ODE model, and compare results between the models to investigate the nature of viral exposure and impact of heterogeneous infection on the time-evolution of the viral infection. We found that using spatially homogeneous initial exposure conditions consistently with those employed during calibration of the ODE model generates far less severe infection, and that local exposure to virus must be multiple orders of magnitude greater than a uniformly applied exposure to all available susceptible cells. This strongly suggests a prominent role of localization of exposure in influenza A infection. We propose that the particularities of the microenvironment to which a virus is introduced plays a dominant role in disease onset and progression, and that spatially resolved models like ours may be important to better understand and more reliably predict future health states based on susceptibility of potential lesion sites using spatially resolved patient data of the state of an infection. We can readily integrate the immune response components of our model into other modeling and simulation frameworks of viral infection dynamics that do detailed modeling of other mechanisms like viral internalization and intracellular viral replication dynamics, which are not explicitly represented in the ODE model. We can also combine our model with available experimental data and modeling of exposure scenarios and spatiotemporal aspects of mechanisms like mucociliary clearance that are only implicitly described by the ODE model, which would significantly improve the ability of our model to present spatially resolved predictions about the progression of influenza infection and immune response.


Assuntos
COVID-19 , Influenza Humana , Viroses , Humanos , Imunidade Inata , SARS-CoV-2
4.
Bull Math Biol ; 83(7): 79, 2021 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-34037874

RESUMO

The pandemic outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has quickly spread worldwide, creating a serious health crisis. The virus is primarily associated with flu-like symptoms but can also lead to severe pathologies and death. We here present an ordinary differential equation model of the intrahost immune response to SARS-CoV-2 infection, fitted to experimental data gleaned from rhesus macaques. The model is calibrated to data from a nonlethal infection, but the model can replicate behavior from various lethal scenarios as well. We evaluate the sensitivity of the model to biologically relevant parameters governing the strength and efficacy of the immune response. We also simulate the effect of both anti-inflammatory and antiviral drugs on the host immune response and demonstrate the ability of the model to lessen the severity of a formerly lethal infection with the addition of the appropriately calibrated drug. Our model emphasizes the importance of tight control of the innate immune response for host survival and viral clearance.


Assuntos
COVID-19/imunologia , Imunidade Inata , Macaca mulatta/imunologia , Modelos Imunológicos , SARS-CoV-2 , Imunidade Adaptativa , Envelhecimento/imunologia , Animais , Anti-Inflamatórios/administração & dosagem , Anti-Inflamatórios/farmacologia , Antivirais/administração & dosagem , Antivirais/farmacologia , COVID-19/epidemiologia , Simulação por Computador , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Interações entre Hospedeiro e Microrganismos/efeitos dos fármacos , Interações entre Hospedeiro e Microrganismos/imunologia , Humanos , Conceitos Matemáticos , Pandemias , Sistema Respiratório/imunologia , Sistema Respiratório/virologia , SARS-CoV-2/imunologia , Carga Viral/imunologia , Tratamento Farmacológico da COVID-19
5.
Biophys J ; 119(11): 2290-2298, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33129831

RESUMO

Over 50% of drugs fail in stage 3 clinical trials, many because of a poor understanding of the drug's mechanisms of action (MoA). A better comprehension of drug MoA will significantly improve research and development (R&D). Current proposed algorithms, such as ProTINA and DeMAND, can be overly complex. Additionally, they are unable to predict whether the drug-induced gene expression or the topology of the networks used to model gene regulation primarily impacts accurate drug target inference. In this work, we evaluate how network and gene expression data affect ProTINA's accuracy. We find that network topology predominantly determines the accuracy of ProTINA's predictions. We further show that the size of an interaction network and/or selecting cell-specific networks has a limited effect on accuracy. We then demonstrate that a specific network topology measure, betweenness, can be used to improve drug target prediction. Based on these results, we create a new algorithm, TREAP, that combines betweenness values and adjusted p-values for target inference. TREAP offers an alternative approach to drug target inference and is advantageous because it is not computationally demanding, provides easy-to-interpret results, and is often more accurate at predicting drug targets than current state-of-the-art approaches.


Assuntos
Algoritmos , Preparações Farmacêuticas , Biologia Computacional , Regulação da Expressão Gênica , Redes Reguladoras de Genes
6.
BMC Public Health ; 14: 1019, 2014 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-25266818

RESUMO

BACKGROUND: Agent based models (ABM) are useful to explore population-level scenarios of disease spread and containment, but typically characterize infected individuals using simplified models of infection and symptoms dynamics. Adding more realistic models of individual infections and symptoms may help to create more realistic population level epidemic dynamics. METHODS: Using an equation-based, host-level mathematical model of influenza A virus infection, we develop a function that expresses the dependence of infectivity and symptoms of an infected individual on initial viral load, age, and viral strain phenotype. We incorporate this response function in a population-scale agent-based model of influenza A epidemic to create a hybrid multiscale modeling framework that reflects both population dynamics and individualized host response to infection. RESULTS: At the host level, we estimate parameter ranges using experimental data of H1N1 viral titers and symptoms measured in humans. By linearization of symptoms responses of the host-level model we obtain a map of the parameters of the model that characterizes clinical phenotypes of influenza infection and immune response variability over the population. At the population-level model, we analyze the effect of individualizing viral response in agent-based model by simulating epidemics across Allegheny County, Pennsylvania under both age-specific and age-independent severity assumptions. CONCLUSIONS: We present a framework for multi-scale simulations of influenza epidemics that enables the study of population-level effects of individual differences in infections and symptoms, with minimal additional computational cost compared to the existing population-level simulations.


Assuntos
Epidemias , Vírus da Influenza A Subtipo H1N1/imunologia , Influenza Humana/epidemiologia , Modelos Teóricos , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Humanos , Vírus da Influenza A Subtipo H1N1/isolamento & purificação , Pessoa de Meia-Idade , Pennsylvania/epidemiologia , Adulto Jovem
7.
J Theor Biol ; 353: 44-54, 2014 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-24594373

RESUMO

The seriousness of pneumococcal pneumonia in mouse models has been shown to depend both on bacterial serotype and murine strain. We here present a simple ordinary differential equation model of the intrahost immune response to bacterial pneumonia that is capable of capturing diverse experimentally determined responses of various murine strains. We discuss the main causes of such differences while accounting for the uncertainty in the estimation of model parameters. We model the bacterial population in both the lungs and blood, the cellular death caused by the infection, and the activation and immigration of phagocytes to the infected tissue. The ensemble model suggests that inter-strain differences in response to streptococcus pneumonia inoculation reside in the strength of nonspecific immune response and the rate of extrapulmonary phagocytosis.


Assuntos
Interações Hospedeiro-Patógeno , Modelos Biológicos , Pneumonia Pneumocócica/microbiologia , Streptococcus pneumoniae/fisiologia , Animais , Modelos Animais de Doenças , Pulmão/microbiologia , Pulmão/patologia , Camundongos Endogâmicos , Pneumonia Pneumocócica/patologia , Análise de Componente Principal , Reprodutibilidade dos Testes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...