Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
ALTEX ; 40(3): 534-540, 2023.
Article in English | MEDLINE | ID: mdl-36888967

ABSTRACT

Progress in developing new tools, assays, and approaches to assess human hazard and health risk provides an opportunity to re-evaluate the necessity of dog studies for the safety evaluation of agrochemicals. A workshop was held where partic­ipants discussed the strengths and limitations of past use of dogs for pesticide evaluations and registrations. Opportunities were identified to support alternative approaches to answer human safety questions without performing the required 90-day dog study. Development of a decision tree for determining when the dog study might not be necessary to inform pesticide safety and risk assessment was proposed. Such a process will require global regulatory authority participation to lead to its acceptance. The identification of unique effects in dogs that are not identified in rodents will need further evaluation and determination of their relevance to humans. The establishment of in vitro and in silico approaches that can provide critical data on relative species sensitivity and human relevance will be an important tool to advance the decision process. Promising novel tools including in vitro comparative metabolism studies, in silico models, and high-throughput assays able to identify metabolites and mechanisms of action leading to development of adverse outcome pathways will need further development. To replace or eliminate the 90-day dog study, a collaborative, multidisciplinary, international effort that transcends organi­zations and regulatory agencies will be needed in order to develop guidance on when the study would not be necessary for human safety and risk assessment.


Subject(s)
Adverse Outcome Pathways , Pesticides , Animals , Dogs , Humans , Agrochemicals/toxicity , Pesticides/toxicity , Risk Assessment , Computer Simulation
2.
PLoS One ; 16(7): e0254208, 2021.
Article in English | MEDLINE | ID: mdl-34292999

ABSTRACT

Nanoparticles have the potential to increase the efficacy of anticancer drugs whilst reducing off-target side effects. However, there remain uncertainties regarding the cellular uptake kinetics of nanoparticles which could have implications for nanoparticle design and delivery. Polymersomes are nanoparticle candidates for cancer therapy which encapsulate chemotherapy drugs. Here we develop a mathematical model to simulate the uptake of polymersomes via endocytosis, a process by which polymersomes bind to the cell surface before becoming internalised by the cell where they then break down, releasing their contents which could include chemotherapy drugs. We focus on two in vitro configurations relevant to the testing and development of cancer therapies: a well-mixed culture model and a tumour spheroid setup. Our mathematical model of the well-mixed culture model comprises a set of coupled ordinary differential equations for the unbound and bound polymersomes and associated binding dynamics. Using a singular perturbation analysis we identify an optimal number of ligands on the polymersome surface which maximises internalised polymersomes and thus intracellular chemotherapy drug concentration. In our mathematical model of the spheroid, a multiphase system of partial differential equations is developed to describe the spatial and temporal distribution of bound and unbound polymersomes via advection and diffusion, alongside oxygen, tumour growth, cell proliferation and viability. Consistent with experimental observations, the model predicts the evolution of oxygen gradients leading to a necrotic core. We investigate the impact of two different internalisation functions on spheroid growth, a constant and a bond dependent function. It was found that the constant function yields faster uptake and therefore chemotherapy delivery. We also show how various parameters, such as spheroid permeability, lead to travelling wave or steady-state solutions.


Subject(s)
Antineoplastic Agents , Drug Carriers , Endocytosis , Models, Biological , Nanoparticles/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Biological Transport , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Humans , Kinetics , Nanoparticles/chemistry
3.
PLoS One ; 16(2): e0244070, 2021.
Article in English | MEDLINE | ID: mdl-33556073

ABSTRACT

A major bottleneck in the study of human liver physiology is the provision of stable liver tissue in sufficient quantity. As a result, current approaches to modelling human drug efficacy and toxicity rely heavily on immortalized human and animal cell lines. These models are informative but do possess significant drawbacks. To address the issues presented by those models, researchers have turned to pluripotent stem cells (PSCs). PSCs can be generated from defined genetic backgrounds, are scalable, and capable of differentiation to all the cell types found in the human body, representing an attractive source of somatic cells for in vitro and in vivo endeavours. Although unlimited numbers of somatic cell types can be generated in vitro, their maturation still remains problematic. In order to develop high fidelity PSC-derived liver tissue, it is necessary to better understand the cell microenvironment in vitro including key elements of liver physiology. In vivo a major driver of zonated liver function is the oxygen gradient that exists from periportal to pericentral regions. In this paper, we demonstrate how cell culture conditions for PSC-derived liver sphere systems can be optimised to recapitulate physiologically relevant oxygen gradients by using mathematical modelling. The mathematical model incorporates some often-understated features and mechanisms of traditional spheroid systems such as cell-specific oxygen uptake, media volume, spheroid size, and well dimensions that can lead to a spatially heterogeneous distribution of oxygen. This mathematical modelling approach allows for the calibration and identification of culture conditions required to generate physiologically realistic function within the microtissue through recapitulation of the in vivo microenvironment.


Subject(s)
Hepatocytes/metabolism , Liver/metabolism , Oxygen/metabolism , Pluripotent Stem Cells/metabolism , Hepatocytes/cytology , Humans , Liver/cytology , Models, Theoretical , Pluripotent Stem Cells/cytology
4.
Interface Focus ; 10(2): 20190041, 2020 Apr 06.
Article in English | MEDLINE | ID: mdl-32194929

ABSTRACT

In early preclinical drug development, potential candidates are tested in the laboratory using isolated cells. These in vitro experiments traditionally involve cells cultured in a two-dimensional monolayer environment. However, cells cultured in three-dimensional spheroid systems have been shown to more closely resemble the functionality and morphology of cells in vivo. While the increasing usage of hepatic spheroid cultures allows for more relevant experimentation in a more realistic biological environment, the underlying physical processes of drug transport, uptake and metabolism contributing to the spatial distribution of drugs in these spheroids remain poorly understood. The development of a multiscale mathematical modelling framework describing the spatio-temporal dynamics of drugs in multicellular environments enables mechanistic insight into the behaviour of these systems. Here, our analysis of cell membrane permeation and porosity throughout the spheroid reveals the impact of these properties on drug penetration, with maximal disparity between zonal metabolism rates occurring for drugs of intermediate lipophilicity. Our research shows how mathematical models can be used to simulate the activity and transport of drugs in hepatic spheroids and in principle any organoid, with the ultimate aim of better informing experimentalists on how to regulate dosing and culture conditions to more effectively optimize drug delivery.

5.
Curr Protoc Toxicol ; 81(1): e87, 2019 09.
Article in English | MEDLINE | ID: mdl-31529797

ABSTRACT

Herein, we describe a protocol for the preparation and analysis of primary isolated rat hepatocytes in a 3D cell culture format described as spheroids. The hepatocyte cells spontaneously self-aggregate into spheroids without the need for synthetic extracellular matrices or hydrogels. Primary rat hepatocytes (PRHs) are a readily available source of primary differentiated liver cells and therefore conserve many of the required liver-specific functional markers, and elicit the natural in vivo phenotype when compared with common hepatic cells lines. We describe the liquid-overlay technique which provides an ultra-low attachment surface on which PRHs can be cultured as spheroids. © 2019 The Authors. Basic Protocol 1: Preparation of agarose-coated plates Basic Protocol 2: Primary rat hepatocyte isolation procedure Basic Protocol 3: Primary rat hepatocyte spheroid culture Basic Protocol 4: Immunofluorescent analysis of PRH spheroids.


Subject(s)
Cell Culture Techniques/methods , Hepatocytes/physiology , Spheroids, Cellular , Animals , Culture Media , Rats
6.
Sci Rep ; 9(1): 6333, 2019 04 19.
Article in English | MEDLINE | ID: mdl-31004119

ABSTRACT

Redox cycling is an understated mechanism of toxicity associated with a plethora of xenobiotics, responsible for preventing the effective treatment of serious conditions such as malaria and cardiomyopathy. Quinone compounds are notorious redox cyclers, present in drugs such as doxorubicin, which is used to treat a host of human cancers. However, the therapeutic index of doxorubicin is undermined by dose-dependent cardiotoxicity, which may be a function of futile redox cycling. In this study, a doxorubicin-specific in silico quinone redox metabolism model is described. Doxorubicin-GSH adduct formation kinetics are thermodynamically estimated from its reduction potential, while the remainder of the model is parameterised using oxygen consumption rate data, indicative of hydroquinone auto-oxidation. The model is then combined with a comprehensive glutathione metabolism model, facilitating the simulation of quinone redox cycling, and adduct-induced GSH depletion. Simulations suggest that glutathione pools are most sensitive to exposure duration at pharmacologically and supra-pharmacologically relevant doxorubicin concentrations. The model provides an alternative method of investigating and quantifying redox cycling induced oxidative stress, circumventing the experimental difficulties of measuring and tracking radical species. This in silico framework provides a platform from which GSH depletion can be explored as a function of a compound's physicochemical properties.


Subject(s)
Benzoquinones/metabolism , Glutathione/metabolism , Models, Biological , Neoplasms/metabolism , Animals , Humans , Neoplasms/drug therapy , Oxidation-Reduction
7.
Toxicol In Vitro ; 55: 160-172, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30578835

ABSTRACT

Many in vitro liver cell models, such as 2D systems, that are used to assess the hepatotoxic potential of xenobiotics suffer major limitations arising from a lack of preservation of physiological phenotype and metabolic competence. To circumvent some of these limitations there has been increased focus on producing more representative 3D models. Here we have used a novel approach to construct a size-controllable 3D hepatic spheroid model using freshly isolated primary rat hepatocytes (PRH) utilising the liquid-overlay technique whereby PRH spontaneously self-assemble in to 3D microtissues. This system produces viable spheroids with a compact in vivo-like structure for up to 21 days with sustained albumin production for the duration of the culture period. F-actin was seen throughout the spheroid body and P-glycoprotein (P-gp) and multidrug resistance-associated protein 2 (MRP2) transporters had polarised expression on the canalicular membrane of hepatocytes within the spheroids upon formation (day 3). The MRP2 transporter was able to functionally transport 5 µM 5-chloromethylfluorescein diacetate (CMFDA) substrates into these canalicular structures. These PRH spheroids display in vivo characteristics including direct cell-cell contacts, cellular polarisation, 3D cellular morphology, and formation of functional secondary structures throughout the spheroid. Such a well-characterised system could be readily exploited for pre-clinical and non-clinical repeat-dose investigations and could make a significant contribution to replace, reduce and refine the use of animals for applied research.


Subject(s)
Hepatocytes , Spheroids, Cellular , Albumins/metabolism , Animals , Cells, Cultured , Drug Evaluation, Preclinical/methods , Fluoresceins/pharmacology , Fluorescent Dyes/pharmacology , Male , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/metabolism , Rats, Wistar , Spheroids, Cellular/metabolism , Spheroids, Cellular/ultrastructure , Toxicity Tests/methods , Urea/metabolism
8.
PLoS One ; 13(11): e0207803, 2018.
Article in English | MEDLINE | ID: mdl-30496306

ABSTRACT

Cancer cells depend on glucose metabolism via glycolysis as a primary energy source, despite the presence of oxygen and fully functioning mitochondria, in order to promote growth, proliferation and longevity. Glycolysis relies upon NAD+ to accept electrons in the glyceraldehyde-3-phosphate dehydrogenase (GAPDH) reaction, linking the redox state of the cytosolic NAD+ pool to glycolytic rate. The free cytosolic NAD+/NADH ratio is involved in over 700 oxidoreductive enzymatic reactions and as such, the NAD+/NADH ratio is regarded as a metabolic readout of overall cellular redox state. Many experimental techniques that monitor or measure total NAD+ and NADH are unable to distinguish between protein-bound and unbound forms. Yet total NAD+/NADH measurements yield little information, since it is the free forms of NAD+ and NADH that determine the kinetic and thermodynamic influence of redox potential on glycolytic rate. Indirect estimations of free NAD+/NADH are based on the lactate/pyruvate (L/P) ratio at chemical equilibrium, but these measurements are often undermined by high lability. To elucidate the sensitivity of the free NAD+/NADH ratio to changes in extracellular substrate, an in silico model of hepatocarcinoma glycolysis was constructed and validated against in vitro data. Model simulations reveal that over experimentally relevant concentrations, changes in extracellular glucose and lactate concentration during routine cancer cell culture can lead to significant deviations in the NAD+/NADH ratio. Based on the principles of chemical equilibrium, the model provides a platform from which experimentally challenging situations may be examined, suggesting that extracellular substrates play an important role in cellular redox and bioenergetic homeostasis.


Subject(s)
Cytosol/metabolism , Extracellular Space/metabolism , Models, Biological , NAD/metabolism , Adenosine Triphosphate/metabolism , Cytoplasm/metabolism , Energy Metabolism , Glycolysis , Lactic Acid/metabolism , Metabolic Flux Analysis
9.
iScience ; 4: 84-96, 2018 Jun 29.
Article in English | MEDLINE | ID: mdl-30240756

ABSTRACT

Many xenobiotics can bind to off-target receptors and cause toxicity via the dysregulation of downstream transcription factors. Identification of subsequent off-target toxicity in these chemicals has often required extensive chemical testing in animal models. An alternative, integrated in vitro/in silico approach for predicting toxic off-target functional responses is presented to refine in vitro receptor identification and reduce the burden on in vivo testing. As part of the methodology, mathematical modeling is used to mechanistically describe processes that regulate transcriptional activity following receptor-ligand binding informed by transcription factor signaling assays. Critical reactions in the signaling cascade are identified to highlight potential perturbation points in the biochemical network that can guide and optimize additional in vitro testing. A physiologically based pharmacokinetic model provides information on the timing and localization of different levels of receptor activation informing whole-body toxic potential resulting from off-target binding.

10.
CPT Pharmacometrics Syst Pharmacol ; 7(6): 394-403, 2018 06.
Article in English | MEDLINE | ID: mdl-29667370

ABSTRACT

Paracetamol (acetaminophen (APAP)) is one of the most commonly used analgesics in the United Kingdom and the United States. However, exceeding the maximum recommended dose can cause serious liver injury and even death. Promising APAP toxicity biomarkers are thought to add value to those used currently and clarification of the functional relationships between these biomarkers and liver injury would aid clinical implementation of an improved APAP toxicity identification framework. The framework currently used to define an APAP overdose is highly dependent upon time since ingestion and initial dose; information that is often highly unpredictable. A pharmacokinetic/pharmacodynamic (PK/PD) APAP model has been built in order to understand the relationships between a panel of biomarkers and APAP dose. Visualization and statistical tools have been used to predict initial APAP dose and time since administration. Additionally, logistic regression analysis has been applied to histology data to provide a prediction of the probability of liver injury.


Subject(s)
Acetaminophen/toxicity , Chemical and Drug Induced Liver Injury/diagnosis , Drug Overdose/complications , Acetaminophen/pharmacokinetics , Animals , Biomarkers , Disease Models, Animal , Drug Overdose/diagnosis , Humans , Logistic Models , Male , Mice , Models, Statistical , Models, Theoretical
11.
Toxicol In Vitro ; 48: 262-275, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29408671

ABSTRACT

Xenobiotic safety assessment is an area that impacts a multitude of different industry sectors such as medicinal drugs, agrochemicals, industrial chemicals, cosmetics and environmental contaminants. As such there are a number of well-developed in vitro, in vivo and in silico approaches to evaluate their properties and potential impact on the environment and to humans. Additionally, there is the continual investment in multidisciplinary scientists to explore non-animal surrogate technologies to predict specific toxicological outcomes and to improve our understanding of the biological processes regarding the toxic potential of xenobiotics. Here we provide a concise, critical evaluation of a number of in vitro systems utilised to assess the hepatotoxic potential of xenobiotics.


Subject(s)
Chemical and Drug Induced Liver Injury/pathology , Hepatocytes/drug effects , Toxicity Tests/methods , Xenobiotics/toxicity , Animal Testing Alternatives , Animals , Cells, Cultured , Culture Techniques , Humans
12.
Geospat Health ; 11(1 Suppl): 387, 2016 Mar 31.
Article in English | MEDLINE | ID: mdl-27063733

ABSTRACT

Outbreaks of Rift Valley fever (RVF), a relatively recently emerged zoonosis endemic to large parts of sub-Saharan Africa that has the potential to spread beyond the continent, have profound health and socio-economic impacts, particularly in communities where resilience is already low. Here output from a new, dynamic disease model [the Liverpool RVF (LRVF) model], driven by downscaled, bias-corrected climate change data from an ensemble of global circulation models from the Inter-Sectoral Impact Model Intercomparison Project run according to two radiative forcing scenarios [representative concentration pathway (RCP)4.5 and RCP8.5], is combined with results of a spatial assessment of social vulnerability to the disease in eastern Africa. The combined approach allowed for analyses of spatial and temporal variations in the risk of RVF to the end of the current century. Results for both scenarios highlight the high-risk of future RVF outbreaks, including in parts of eastern Africa to date unaffected by the disease. The results also highlight the risk of spread from/to countries adjacent to the study area, and possibly farther afield, and the value of considering the geography of future projections of disease risk. Based on the results, there is a clear need to remain vigilant and to invest not only in surveillance and early warning systems, but also in addressing the socio-economic factors that underpin social vulnerability in order to mitigate, effectively, future impacts.


Subject(s)
Climate Change , Models, Theoretical , Rift Valley Fever/epidemiology , Rift Valley Fever/transmission , Africa, Eastern/epidemiology , Animals , Disease Outbreaks , Geography , Humans , Population Surveillance , Risk Factors , Vulnerable Populations
13.
Geospat Health ; 11(1 Suppl): 393, 2016 Mar 31.
Article in English | MEDLINE | ID: mdl-27063736

ABSTRACT

The effect of climate change on the spatiotemporal dynamics of malaria transmission is studied using an unprecedented ensemble of climate projections, employing three diverse bias correction and downscaling techniques, in order to partially account for uncertainty in climate- driven malaria projections. These large climate ensembles drive two dynamical and spatially explicit epidemiological malaria models to provide future hazard projections for the focus region of eastern Africa. While the two malaria models produce very distinct transmission patterns for the recent climate, their response to future climate change is similar in terms of sign and spatial distribution, with malaria transmission moving to higher altitudes in the East African Community (EAC) region, while transmission reduces in lowland, marginal transmission zones such as South Sudan. The climate model ensemble generally projects warmer and wetter conditions over EAC. The simulated malaria response appears to be driven by temperature rather than precipitation effects. This reduces the uncertainty due to the climate models, as precipitation trends in tropical regions are very diverse, projecting both drier and wetter conditions with the current state-of-the-art climate model ensemble. The magnitude of the projected changes differed considerably between the two dynamical malaria models, with one much more sensitive to climate change, highlighting that uncertainty in the malaria projections is also associated with the disease modelling approach.


Subject(s)
Climate Change , Malaria/transmission , Models, Theoretical , Africa, Eastern/epidemiology , Animals , Humans , Malaria/epidemiology , Risk Assessment , Temperature , Uncertainty
14.
Geospat Health ; 11(1 Suppl): 394, 2016 Mar 31.
Article in English | MEDLINE | ID: mdl-27063737

ABSTRACT

Outbreaks of Rift Valley fever (RVF) in eastern Africa have previously occurred following specific rainfall dynamics and flooding events that appear to support the emergence of large numbers of mosquito vectors. As such, transmission of the virus is considered to be sensitive to environmental conditions and therefore changes in climate can impact the spatiotemporal dynamics of epizootic vulnerability. Epidemiological information describing the methods and parameters of RVF transmission and its dependence on climatic factors are used to develop a new spatio-temporal mathematical model that simulates these dynamics and can predict the impact of changes in climate. The Liverpool RVF (LRVF) model is a new dynamic, process-based model driven by climate data that provides a predictive output of geographical changes in RVF outbreak susceptibility as a result of the climate and local livestock immunity. This description of the multi-disciplinary process of model development is accessible to mathematicians, epidemiological modellers and climate scientists, uniting dynamic mathematical modelling, empirical parameterisation and state-of-the-art climate information.


Subject(s)
Climate , Culicidae/growth & development , Rift Valley Fever/epidemiology , Rift Valley Fever/transmission , Africa, Eastern/epidemiology , Animals , Disease Outbreaks , Humans , Insect Vectors , Livestock , Models, Theoretical , Rain
15.
Math Biosci ; 258: 33-43, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25245610

ABSTRACT

HIF (hypoxia inducible factor) is an oxygen-regulated transcription factor that mediates the intracellular response to hypoxia in human cells. There is increasing evidence that cell signaling pathways encode temporal information, and thus cell fate may be determined by the dynamics of protein levels. We have developed a mathematical model to describe the transient dynamics of the HIF-1α protein measured in single cells subjected to hypoxic shock. The essential characteristics of these data are modeled with a system of differential equations describing the feedback inhibition between HIF-1α and prolyl hydroxylases (PHD) oxygen sensors. Heterogeneity in the single-cell data is accounted through parameter variation in the model. We previously identified the PHD2 isoform as the main PHD sensor responsible for controlling the HIF-1α transient response, and make here testable predictions regarding HIF-1α dynamics subject to repetitive hypoxic pulses. The model is further developed to describe the dynamics of HIF-1α in cells cultured as 3D spheroids, with oxygen dynamics parameterized using experimental measurements of oxygen within spheroids. We show that the dynamics of HIF-1α and transcriptional targets of HIF-1α display a non-monotone response to the oxygen dynamics. Specifically we demonstrate that the dynamic transient behavior of HIF-1α results in differential dynamics in transcriptional targets.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/metabolism , Models, Theoretical , Animals , HeLa Cells , Humans
16.
J Biol Chem ; 289(9): 5549-64, 2014 Feb 28.
Article in English | MEDLINE | ID: mdl-24394419

ABSTRACT

Intracellular signaling involving hypoxia-inducible factor (HIF) controls the adaptive responses to hypoxia. There is a growing body of evidence demonstrating that intracellular signals encode temporal information. Thus, the dynamics of protein levels, as well as protein quantity and/or localization, impacts on cell fate. We hypothesized that such temporal encoding has a role in HIF signaling and cell fate decisions triggered by hypoxic conditions. Using live cell imaging in a controlled oxygen environment, we observed transient 3-h pulses of HIF-1α and -2α expression under continuous hypoxia. We postulated that the well described prolyl hydroxylase (PHD) oxygen sensors and HIF negative feedback regulators could be the origin of the pulsatile HIF dynamics. We used iterative mathematical modeling and experimental analysis to scrutinize which parameter of the PHD feedback could control HIF timing and we probed for the functional redundancy between the three main PHD proteins. We identified PHD2 as the main PHD responsible for HIF peak duration. We then demonstrated that this has important consequences, because the transient nature of the HIF pulse prevents cell death by avoiding transcription of p53-dependent pro-apoptotic genes. We have further shown the importance of considering HIF dynamics for coupling mathematical models by using a described HIF-p53 mathematical model. Our results indicate that the tight control of HIF transient dynamics has important functional consequences on the cross-talk with key signaling pathways controlling cell survival, which is likely to impact on HIF targeting strategies for hypoxia-associated diseases such as tumor progression and ischemia.


Subject(s)
Apoptosis/physiology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Signal Transduction/physiology , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Hypoxia/physiology , Cell Survival/physiology , HeLa Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...