General Discussion PhD thesis

Back to PhD thesis

In this PhD thesis the development, validation and application of the Particle Profiler computational model is described. This model was developed with the aim of analyzing measured detailed lipoprotein profiles, to obtain information about human lipoprotein metabolism that can be used for diagnostic purposes. This general discussion includes the main findings, methodological considerations related to Particle Profiler, a comparison of Particle Profiler with other modeling studies, future research recommendations and a general conclusion.

Main findings

This section includes the main findings presented in this PhD thesis with regard to Particle Profiler. The first section summarizes the knowledge gained during model development and validation, while the second section summarizes the use Particle Profiler had in various applications.

Model Development and Validation

The second chapter of this PhD thesis presents a first version of the Particle Profiler model. Particle Profiler models how the rate of lipoprotein metabolic processes depends on the size of the lipoprotein particle. The model was applied to existing preanalyzed stable isotope tracer data presented by Packard et al. (78). This gave four main findings (125). The model implementation was able to reproduce the original metabolic fluxes calculated by Packard et al., requiring only six parameters to describe all modeled lipolysis and uptake processes. The Particle Profiler results in chapter [2] were able to predict the LDL size shift that was measured in the original study, only using reported flux data measured in four density classes by Packard et al. Therefore, the model predicts a change that is actually measured, which gives confidence in the model’s realism. Furthermore, Particle Profiler was able to indicate relevant differences in physiology between the groups, such as a difference in higher hepatic lipase activity that is known to cause smaller LDL particles. This finding indicates a possible mechanism for the shift in LDL peak size. Finally, the potential for modeling the effects of genetic variants was demonstrated by simulating reductions in ApoB-related uptake affinity and lipolysis affinity. These simulations gave results that correspond to the observed lipoprotein profiles in subjects with such deficiencies. These four findings encouraged further investigation.
         In the third chapter, we further developed and calibrated Particle Profiler model parameters using literature data on metabolic fluxes and lipoprotein profiles from subjects with specific genetic conditions. The development step consisted in replacing the function for particle attachment for the liver with a new function that better matched the observed hepatic lipoprotein uptake in lipoprotein flux studies. In this stage, we also further reduced the number of parameters to five. The calibration step consisted in estimating the model ‘constants’ (those parameters that remain the same for all subjects) using subjects with different genetic backgrounds: LPL deficiency, the apoE 3/3 genotype, and the apoE 2/2 genotype. In these subjects specific processes stand out clearly, which makes it possible to estimate the model constants. We subsequently performed technical model validation using data on lipoprotein concentrations and metabolic fluxes: the model could fit a range of normolipidemic and dyslipidemic subjects from fifteen out of sixteen studies equally well, with an average 9%±5% fit error; only one study showed a larger fit error. This result constitutes a corroboration of the model, which is the best possible technical model validation available.

Model Application

The first model application was an initial clinical validation also in chapter three. There, we showed that a new diagnostic marker based on VLDL metabolic ratios calculated by the Particle Profiler model, better distinguished dyslipidemic from normolipidemic subjects than established markers like triglycerides, HDL cholesterol, and LDL cholesterol. This diagnostic marker is called VLDL performance, and is the average of two metabolic ratios: lipolysis/production in VLDL and uptake/production in VLDL. Two additional model-based markers clearly distinguished subjects with genetic variants apoE 2/2, LPL -/-, homozygous familial hypercholesterolemia, and familial defective apoB from other normolipidemic and dyslipidemic subjects. (149) This indicates that Particle Profiler-derived diagnostics reflect specific traits of dyslipidemia and genetic deficiencies.
         The second application in chapter 4 concerned a nutritional study, which investigated the metabolic effects of dietary medium chain fatty acids (MCFA) versus long chain fatty acids, especially linoleic acid. This application showed that combining Particle Profiler-derived flux ratios with stable isotope-determined VLDL apoB production rates can be used to determine absolute VLDL production, lipolysis and uptake rates. The study showed no significant difference in VLDL production between treatments. However, it did show a difference in lipolysis and uptake, which was attributed to PPAR activation by linoleic acid.
         The third application in chapter 5 was to the GOLDN study, in which we identified three subgroups based on clustering of the lipoprotein profiles. Analysis of the cluster centroid lipoprotein profiles using Particle Profile showed differences in the two metabolic ratios that constitute VLDL performance.  The ‘VLDL performance’ marker was high in the first ‘healthy’ subgroup, lower in the second ‘intermediate’ subgroup, and lowest in the third ‘unhealthy’ subgroup. The difference between the second and third versus the first and second subgroups showed a  larger influence of extrahepatic lipolysis dysfunction versus liver uptake dysfunction. The reported dyslipidemia, therefore, seems to involve two stadia in which extrahepatic lipolysis dysfunction becomes progressively more important compared to liver uptake dysfunction. (139) The subgroups also show difference in lipid response to fibrate therapy, where both the ‘intermediate’ and ‘unhealthy’ subgroup show improved lipid values.
         The final application  in chapter 6 was to lipoprotein profiles measured using NMR in the Framingham Heart study. It is possible to construct many different Particle Profiler-based diagnostics by taking ratios of all processes modeled in  Particle Profiler, and evaluating these ratios for various particle size ranges. In this study, we used the following three size ranges: VLDL to LDL, VLDL only, IDL and LDL.  From all possible Particle Profiler-based diagnostics thus derived, we selected those that consistently contributed to cardiovascular disease risk prediction, using a nonlinear SVM-type statistical approach. We found that two lipoprotein metabolic ratios significantly improved the area under the curve of the ROC curve for general cardiovascular disease, and therefore significantly improve the risk prediction for cardiovascular disease. The first ratio we found is the ‘extrahepatic lipolysis ratio’. This ratio is especially important for predicting risk in people that take blood pressure medication: if people take this medication and extrahepatic lipolysis does not function well, the risk of cardiovascular disease dramatically increases. The second ratio is the ‘hepatic turnover ratio’. This ratio is especially important when a person has a LDL particle number that indicates the borderline between low and medium risk categories. If the liver works well in such a person, risk is low; if the liver does not work well, the person belongs to the medium risk category, which often entails therapeutic intervention. Therefore, we have found two lipoprotein metabolic ratios that significantly improve the existing diagnostic power for general cardiovascular disease, a finding that can really make a difference for people’s lives. For clinical application, further validation is required.

Methodological Considerations Related to Particle Profiler

This section treats two issues that came up during the project and that still require further thought and investigation. The first issue is about how to quantify single particle lipolysis. This issue was dealt with briefly in chapter [2], but a lengthier discussion is useful for examining in detail what can be done about it. The second issue concerns the precise size-dependency of lipoprotein metabolic process rates. This issue comes up as a consequence of the work done in this PhD thesis, and discussing it is useful to trace a path for future investigation.

Quantifying Single Particle Lipolysis

The first methodological issue is that of quantifying single particle lipolysis. In the process of designing a model for interpreting detailed lipoprotein profile measurements, several assumptions need to be made to base the mathematical equations on. Making these assumption explicit is one of the key parts of the learning processes involved in the mathematical modeling of biology. The assumptions make clear what the current state of our knowledge about the system is, and often point to new scientific questions. The Particle Profiler model is based on thinking in terms of processes affecting single lipoprotein particles, and explicitly takes into account the size of the particles. One of the questions that appeared was how the particle lipolysis process, during which the particle loses fat to other tissues, relates to the size of the lipoprotein particle. In this process, there are two steps, as described in chapter [2]. The first step describes how fast the lipolysis process occurs for a particle of a given size. The second step describes how the size of the particle changes after lipolysis. I would like to discuss the assumptions we made for the second step.
         In an earlier single particle-based model by Adiels (20), the particle size decrease by  lipolysis has been treated as a stochastic variable. For our model, stochasticity was not an option, because of the computational overhead it entails. Therefore, we sought for experimental data to underpin the size change through lipolysis, but we found none. Since the LPL protein lipolyzes the triglycerides in the core of the lipoprotein particle (107), we related the size change due to lipolysis to the triglyceride content of the particle. This approach poses two problems, the first being the relation between particle size and triglyceride content, and the second being the manner in which triglycerides are taken out. Because of the scarcity of the data, we had to solve the first problem by assuming that a particle of a given size always has the same triglyceride content, where the relation between triglyceride content and particle size is based on an earlier published model that is extensively linked to experimental data (101). For the second problem, we considered two manners in which triglycerides might be taken out, as a constant number of triglycerides per lipolysis step or as a constant fraction of triglycerides per lipolysis step. In both these cases, the model would be parameterizable with a single parameter, which is the maximal number we could hope to estimate with the available data. We were able to see that taking out a constant number of triglycerides per lipolysis step led to inconsistencies with the experimental data, whereas taking out a fixed fraction of triglycerides was compatible with the experimental data. Therefore, for this problem we decided to use a fixed fraction loss of triglycerides per lipolysis step.
         As the reader will note, these last arguments are largely inspired by practical considerations and the solution is limited by scarcity of experimental data. Therefore, in this area the model leads to an experimental challenge: quantifying how the size and composition of a single lipoprotein particle changes during a lipolysis event. The most feasible approach to this problem would be a combination of experiments and modeling. The experiments would have to be stable isotope labeling studies, in which apoB is labeled, for instance through a labeled leucine infusion. Then at several timepoints after the infusion, plasma samples should be taken and lipoprotein profiles run on these samples. The different lipoprotein subfractions (how many subfractions there are depends on the measurement method used)should be analyzed for their biochemical constituents: apoB, free cholesterol, cholesterol ester, phospholipids, triglycerides and total protein. This analysis will allow Particle Profiler to optimally take into account the lipoprotein composition.  In addition, the apoB stable isotope enrichment should be measured for determining the particle production, lipolysis and uptake fluxes. The methodology for such a study is comparable to that of earlier stable isotope studies (50). These data form the ideal dataset for  applying a version of Particle Profiler to that can take into account lipoprotein tracer data. This version of the model is yet to be developed. This methodology will  lead to improved quantitative understanding of lipoprotein lipolysis.

Precise Size-Dependency of Lipoprotein Metabolic Process Rates

A second issue relates to the new concept that the Particle Profiler model introduces: the relation between the rate of lipoprotein metabolic processes and particle size. It is possible to further refine our understanding of how lipoprotein metabolic process rates (production, lipolysis, and uptake) depend on the size of the lipoprotein particle. In this PhD thesis we have been able to work with apoB kinetics data from many dyslipidemic subjects, mostly published by Packard et al. The only drawback these data have from the perspective of the Particle Profiler model, is the fact that they do not include more than four lipoprotein subfractions. The studies described above, in which apoB kinetics are measured in multiple subfractions, are also necessary to refine our understanding of how lipoprotein metabolism rates depend on particle size. Such studies will need to be carried out in subjects with different genetic backgrounds, similar to the study presented in chapter [3], where we used subjects with LPL deficiency, apoE 3/3 and apoE 2/2 backgrounds. Such studies will allow us to refine our understanding of how lipoprotein metabolic process rates depend on particle size.
         One example where refinement is possible, is in the process of LDL uptake through the LDL receptor. It is known that apoB changes its configuration when the lipoprotein particle changes its size, and that this configuration change affects its interaction with the LDL receptor.(114) Put differently, the affinity of the LDL receptor for LDL particles changes with the size of the particle. However, the currently available data do not contain enough information to include this effect in the model, which is why it is currently not included. Future studies of the type described above would help to refine our understanding of how the apoB-dependent LDL uptake varies with the size of the particle. Next to furthering our understanding, this knowledge would also help to further improve the model-based diagnostics.

Model Application

Particle Profiler can in principle be applied to lipoprotein profiles including VLDL through LDL lipoproteins measured by any method. Several such methods are available, as discussed in the Introduction chapter of this thesis under: “What methods are available for measuring lipoproteins and to what data types do these methods lead?” As Figure 1.2 illustrates, these methods are hard to compare directly, because they subdivide the lipoprotein size spectrum into different subclasses. However, Particle Profiler can analyze all these data types and always give the same output parameters. Therefore, it is possible to compare the Particle Profiler output based on different measurement methods.
         Such a comparison is interesting, but not trivial, since things can go wrong at several levels. First of all, the measurement methods can give mutually inconsistent results. But secondly, the measurement methods can measure the same particle and report different sizes. This method-dependent bias in size measure needs to be corrected for. The bias can be detected by comparing the lipoprotein compositions in each size fraction. Therefore, this size bias-related error can be minimized by measuring the lipoprotein composition in each subfraction (phospholipids, free cholesterol, cholesterol ester, triglycerides and total protein). Clearly, the different lipoprotein profiling measures would have to be carried out on the same samples. It would be very interesting to perform this study from a technical point of view.
         Further applications of Particle Profiler are only limited by the domains in which lipoproteins perform an important role. Chapter [6] of this thesis demonstrates that Particle Profiler has produced new diagnostic markers that improve cardiovascular disease prediction. This area could be carried further by looking at specific cardiovascular endpoints, or by further validating the model in other cohort studies. It would also be interesting to relate Particle Profiler parameters to genetic variants, especially those parameters that are important as diagnostic markers. Other possible domains of application include diabetes type II prediction and obesity research.

Particle Profiler Compared to Other Modeling Studies

Modeling can be used to investigate lipoprotein metabolism in various ways. Here, we will discuss several modeling methodologies and their relation with Particle Profiler.

Modeling in the determination of subclass kinetics

Since lipoprotein subclasses can be physically separated e.g. by ultracentrifugation or FPLC, they are amenable for kinetic studies employing tracers. Isotopically labeled precursors of lipoprotein constituents (fatty acids, cholesterol, lipids, or amino acids) can be supplied by means of intravenous infusion, and the kinetics of accumulation of the isotope in different lipoprotein fractions in humans can be followed in samples taken at different time points (46,91). The kinetic constants that characterize the different processes in which lipoproteins take part, then have to be derived by mathematical modeling of these processes and non-linear least squares fitting of predicted vs. measured isotopic enrichments. Typically, the models used are multicompartment models that include pools of precursors in plasma and various body compartments, pools of assembled (nascent) lipoproteins e.g. in the liver, and pools of subclasses of VLDL, IDL, and LDL in plasma (50,51). The kinetic processes typically include synthesis of pool constituents, exchange reactions between the various pools, and irreversible loss of pool constituents. In most cases, first-order kinetics of reactions is assumed. Estimated parameters include pool sizes and first-order rate constants that can be combined to calculate fractional synthetic rates, fractional catabolic rates and/or fractional transfer rates. These parameters can then be related to environmental factors (e.g. interventions) and disease, providing possible mechanistic clues to observed phenotypic responses. The use and applications of these models particularly in studies on the nutrition/metabolic syndrome has recently been reviewed in (228). Tracer studies combined with kinetic modeling have thus yielded important insights in the kinetics of production and interconversion of lipoproteins in vivo and how these vary in diseased states. Such changes will be reflected, albeit in an indirect and complex manner, in lipoprotein profiles. Thus, modeling directed at extracting flux-related information from lipoprotein profiles may help to reveal and diagnose disease phenotypes.
         The relation between Particle Profiler and lipoprotein kinetics models is two-fold. First of all, Particle Profiler has been developed and validated using results from this type of models. Secondly, Particle Profiler can itself be seen as a kinetic model, but in the current implementation it only models particle subfraction concentrations at steady state. However, in principle the Particle Profiler framework could be extended to include the analysis of stable isotope kinetic data. With such an extension, the model would be able to analyze data from experiments where stable isotope enrichments are measured in many subfractions. This would in turn give information for further refinement of the model, as discussed above under ‘Methodological Considerations Related to Particle Profiler’. So Particle Profiler at once builds forward on the work of earlier kinetic models, and could also become a kinetic model itself if extended further.

Modeling to understand and predict lipoprotein metabolism

Knowing from kinetic as well as from molecular studies which processes are involved in the metabolism of lipoproteins, new, mechanistically driven approaches are emerging in cholesterol and lipoprotein modeling. Such approaches hope to predict what lipoprotein profile results from a given set of parameters characterizing the different biological processes in which lipoproteins are involved. Once the prediction is correct, the model can be subjected to various analyses. For example, sensitivity analysis can be used to predict which interventions should be made to bring about a desired change in the lipoprotein profile, such as an optimization of the LDL/HDL cholesterol ratio. One could also predict the effect of genetic mutations on the lipoprotein profile. The mechanistic models to understand and predict lipoprotein metabolism may further be linked to computational mechanistic models of a specific disease process (for example atherosclerosis, (229)) to generate predictions on how the lipoprotein profile will change depending on disease parameters, provided that the lipoprotein metabolic processes are represented in sufficient detail in the integrated model. This offers the possibility to effectively predict candidate lipoprotein-derived causal disease biomarkers from in silico studies.
         Below we describe several predictive models that have recently been published.
         Plasma LDL-cholesterol levels are in part determined by the rate at which LDL particles are removed from the bloodstream by hepatic uptake. The uptake of LDL by mammalian liver cells occurs mainly via receptor-mediated endocytosis, a process which entails the binding of these particles to specific receptors in specialized areas of the cell surface, the subsequent internalization of the receptor–lipoprotein complex, and ultimately the degradation and release of the ingested lipoproteins’ constituent parts. Receptors are then partly recycled to the cell surface, and there is de novo cholesterol dependent receptor formation by the cell. These processes are key determinants for the principal mechanism of action of cholesterol-lowering statins. These drugs decrease intracellular hepatic cholesterol, leading to an increase of LDL receptor formation that subsequently causes increased extraction of cholesterol from LDL and VLDL lipoproteins, lowering plasma cholesterol as a consequence (230). Three deterministic models for lipoprotein endocytosis and associated events have been published and their properties analyzed. August et al (231)   focused on the effect of model parameters on plasma cholesterol levels, predicting distinct high- and low cholesterol states. Tindall et al (232)  predicted the effect of variation in binding, unbinding and internalization rates, the fraction of receptors recycled and the rate at which the cholesterol dependent free receptors are created by the cell on the overall uptake dynamics of either VLDL or LDL particles and subsequent intracellular cholesterol concentration. Pearson et al (233)  focused particularly on how the competition between the LDL and VLDL particles for binding to the pits on the cell surface affects the intracellular cholesterol concentration.
         Lipoproteins are influenced in an indirect way by many more processes than those studied in the three mechanistic models mentioned above. Considering this fact, Van de Pas et al(234,235) have developed a physiologically-based kinetic (PBK) model for plasma cholesterol that includes all processes in the mouse that are key determinants of plasma cholesterol. The choice for processes to include or not was made based on plasma cholesterol values reported for knockout mouse strains included in the Mouse Genome Database[1] (236). The authors identified no less than 36 key genes for plasma cholesterol, and used 12 of these to build a conceptual model of 20 metabolic and transport processes that directly involve cholesterol  (234). Since kinetic parameters for all these processes are hardly available, the authors took the approach of building an ensemble of 64k submodels each having a unique combination of zero-order and first order apparent kinetics for 16 of the 20 processes included, calibrate each with in vivo data on pool sizes and fluxes, and selecting the best predictive models to generate an average prediction (235).  This model is in the process of being translated to a human model.
         In addition to the conclusions on the behavior of cholesterol metabolism as a whole, the dynamics of LDL and VLDL uptake and binding competition, and the effects thereof on intracellular cholesterol, the described models also show that mechanistic characteristics of lipoprotein metabolism are reflected indirectly in the lipoprotein profile. While the submodels lack detail in the plasma lipoprotein compartments (only 2 are included), especially the PBK approach has created the unique possibility to link processes and genes affecting cholesterol transport in the body as a whole, to changes in the plasma lipoprotein profile. The latter may be subject to analysis using more refined models. All in all, these models either provide a larger whole-body context than Particle Profiler, or zoom in on specific details of processes that are relevant for Particle Profiler. They therefore form a valuable addition to the quantative analysis of lipoprotein metabolism.

Modeling to quantify physiological parameters that underlie different subclass distribution phenotypes

Using the knowledge gained from molecular and kinetic studies, a reverse procedure may be followed: from a measured lipoprotein profile, derive the parameter values of the underlying biological processes. This is the approach Particle Profiler has followed. We have shown that the results of such an analysis are especially useful for diagnostic purposes in chapter [6]. It is of interest that computational modeling for diagnostic purposes poses specific demands for the design, construction and evaluation phases of model development and can benefit from techniques for efficient model formulation, model calculation, workflow structuring and quality control as described in the chapter [7](126).
         Next to particle profiler, only  Hübner et al (61)  have published a model with the intention of analyzing lipoprotein subclass profiles.
Hübner et al (61) have developed a particle-based model solely of plasma lipoproteins that considers the entire protein and lipid composition spectrum of individual lipoprotein complexes, of both ApoB-100- and ApoA-I- containing lipoproteins. Subsequently, their distribution over density (which equals the lipoprotein profile) was calculated and it was shown that cholesterol plasma levels can be simulated as resulting from a steady state of a particle distribution. Their model was able to (i) successfully reproduce clinically measured lipoprotein profiles of healthy subjects; (ii) assign lipoproteins to narrow density classes, named high-resolution density sub-fractions (hrDS), revealing heterogeneous lipoprotein distributions within the major lipoprotein classes; and (iii) generate predictions of changes in the lipoprotein distribution elicited by disorders in underlying molecular processes. Formulated as a stochastic particle population model, a large number of individual lipoprotein particles must be modeled for a simulation, and considerable computational power is required. A potential use of the model for diagnostic purposes was indicated but not demonstrated. The estimation of the 20 parameters of the model will require the measurement of a very highly resolved lipoprotein profile indeed, along with compositional analysis of each subfraction.
         In constructing Particle Profiler, we have taken much care to avoid overparametrization of our model, essentially by limiting ourselves to ApoB-100 containing lipoproteins and by implementing particle size-dependent mass action kinetics as opposed to the non size-dependent  kinetics used in Hübner et al (61). This left only 5 parameters to be estimated from a lipoprotein subclass profile in the final model (chapter [3]). By defining and simulating a large number of lipoprotein compartments that contain lipoproteins in a small size range (as small as 0.1 nm has been used), the computational efficiency was greatly improved compared to a stochastic particle population model. It was shown (237) that the variables calculated by the model, can be combined to calculate (sub)class specific lipoprotein flux ratios that reflect disease physiology of liver and extrahepatic tissue. These ratios can be used as novel lipoprotein profile-based diagnostic markers (Chapter [6]. Hübner et al. have not yet published  any clinical application of their model.

Statistical modeling for the construction of biomarkers that have diagnostic potential

The availability of detailed lipoprotein subclass profiles may lead to new biomarkers that spur the development of new disease concepts (see e.g. (238)). However, rather than affecting specific individual (sub)classes, disease may be associated with more complex patterns of distortion in the lipoprotein profile.  Thus, there is a role for yet another type of modeling used in the analysis of lipoproteins, i.e. multivariate statistics. Of course multivariate statistics approaches are not specific to lipoproteins but we nevertheless mention a few interesting applications. Musunuru et al (57) used principal component analysis (PCA) of ion-mobility detected lipoprotein subfraction concentrations to identify 3 major orthogonal (i.e., zero correlation) components of CVD risk, one representing LDL-associated risk, a second representing HDL-associated protection, and the third (the “atherogenic lipoprotein phenotype”) representing a pattern of decreased large HDL, increased small/medium LDL, and increased triglycerides. The patterns appear to be associated with SNPs in specific genes, suggesting independent mechanistic pathways for development of CVD.
         Self-organizing maps (SOMs) is a data visualization technique that reduces the dimensions of data through the use of self-organizing neural networks. The way SOMs go about reducing dimensions is by producing a map of usually 1 or 2 dimensions which plot the similarities of the data by grouping similar data items together.  SOMs were used by Suna et al (239) and Kumpula et al. (240) to cluster lipoprotein profile phenotypes, leading to the emergence of complex lipoprotein associations. In Kumpula et al. (240) a metabolic subgrouping of the associations between plasma LDL cholesterol concentrations and the structural subtypes of LDL particles was discovered . This type of clustering can for example provide clues as to the correlation of compositional variations within the lipoprotein particles with metabolic and clinical parameters.
         Clustering in general is a helpful method to assign patients to specific subgroups based on lipoprotein profiles. Correlating characteristics of the new subgroups with treatment outcomes may identify larger groups of patients that would benefit from the treatment than current clinical practice allows, as we (241) made clear in the case of fibrate therapy . 
         Particle Profiler can contribute to statistical analyses in two ways. First, as we showed in chapter [5], it can be used to interpret the outcome of lipoprotein clustering. But more importantly, to relate the parameters that result from Particle Profiler analysis to a disease endpoint, as we did in chapter [6], multivariate statistics is needed. In this last case, by generating new variables with specific metabolic information, Particle Profiler has improved the capacity of statistical analyses to relate relevant information from the lipoprotein profile to the disease endpoint.

Future Research Recommendations

In the above discussion, several recommendations for future research have emerged. Briefly, on the fundamental side these include further kinetics studies with stable isotope tracers, in which isotope enrichment is followed in a detailed lipoprotein profile. The Particle Profiler model framework needs to be extended to include kinetics data for these analyses.
         Another possible study would be a comparison of the Particle Profiler outcomes using different lipoprotein profile analysis methods on the same samples. For best results, the methods should measure as many lipoprotein constituents in each of the subfractions as possible.
         The area of cardiovascular disease prediction could be carried further by looking at specific endpoints, or by further validating the model for other populations. It would also be interesting to relate the relevant Particle Profiler parameters to genetic variants. Other possible domains of application include diabetes type II prediction and obesity research.

Concluding Remarks

In this PhD thesis, the Particle Profiler model has been developed, calibrated, corroborated, validated and applied. Particle Profiler has shown its usefulness for indicating different specific types of dyslipidemia, for understanding population-wide dyslipidemia, and for examining the effects of a nutritional intervention. Particle Profiler has also led to new risk biomarkers that are able to improve the diagnosis of cardiovascular disease, which have been identified using data from the Framingham cohort.



[1] available via http://www.informatics.jax.org

References

1.    Wilson, P. W. F., W. P. Castelli, and W. B. Kannel. 1987. Coronary risk prediction in adults (the Framingham Heart Study). Am J Cardiol 59:91G-94G.
2.    Conroy, R. M., K. Pyörälä, A. P. Fitzgerald, S. Sans, A. Menotti, G. De Backer, D. De Bacquer, P. Ducimetière, P. Jousilahti, U. Keil, I. Njolstad, R. G. Oganov, T. Thomsen, H. Tunstall-Pedoe, A. Tverdal, H. Wedel, P. Whincup, L. Witheimsen, and I. M. Graham. 2003. Estimation of ten-year risk of fatal cardiovascular disease in Europe: The SCORE project. Eur Heart J 24:987-1003.
3.    Assmann, G., P. Cullen, and H. Schulte. 2002. Simple Scoring Scheme for Calculating the Risk of Acute Coronary Events Based on the 10-Year Follow-Up of the Prospective Cardiovascular Munster (PROCAM) Study. Circulation 105:310-315.
4.    Cromwell, W. C., J. D. Otvos, M. J. Keyes, M. J. Pencina, L. Sullivan, R. S. Vasan, P. W. F. Wilson, and R. B. D'Agostino. 2007. LDL particle number and risk of future cardiovascular disease in the Framingham Offspring Study-Implications for LDL management. J Clin Lipidol 1:583-592.
5.    Otvos, J. D., E. J. Jeyarajah, D. W. Bennett, and R. M. Krauss. 1992. Development of a proton nuclear magnetic resonance spectroscopic method for determining plasma lipoprotein concentrations and subspecies distributions from a single, rapid measurement. Clin Chem 38:1632-1638.
6.    Usui, S., Y. Hara, S. Hosaki, and M. Okazaki. 2002. A new on-line dual enzymatic method for simultaneous quantification of cholesterol and triglycerides in lipoproteins by HPLC. J Lipid Res 43:805-814.
7.    Caulfield, M. P., S. Li, G. Lee, P. J. Blanche, W. A. Salameh, W. H. Benner, R. E. Reitz, and R. M. Krauss. 2008. Direct Determination of Lipoprotein Particle Sizes and Concentrations by Ion Mobility Analysis. Clin Chem 54:1307-1316.
8.    Caulfield, M. P., S. Li, G. Lee, P. A. Blanche, W. A. Salameh, W. H. Benner, R. E. Reitz, and R. M. Krauss. 2008. In Reply. Clin Chem 54:2088-2089.
9.    Rambaldi, D. C., A. Zattoni, S. Casolari, P. Reschiglian, D. Roessner, and C. Johann. 2007. An Analytical Method for Size and Shape Characterization of Blood Lipoproteins. Clin Chem 53:2026-2029.
10.   Beswick, A., and P. Brindle. 2006. Risk scoring in the assessment of cardiovascular risk. Curr Opin Lipidol 17:375-386.
11.   Walley, T., P. Folino-Gallo, U. Schwabe, and E. van Ganse. 2004. Variations and increase in use of statins across Europe: data from administrative databases. BMJ 328:385-386.
12.   Mills, E. J., B. Rachlis, P. Wu, P. J. Devereaux, P. Arora, and D. Perri. 2008. Primary Prevention of Cardiovascular Mortality and Events With Statin Treatments: A Network Meta-Analysis Involving More Than 65,000 Patients. J Am Coll Cardiol 52:1769-1781.
13.   Brugts, J. J., T. Yetgin, S. E. Hoeks, A. M. Gotto, J. Shepherd, R. G. J. Westendorp, A. J. M. de Craen, R. H. Knopp, H. Nakamura, P. Ridker, R. van Domburg, and J. W. Deckers. 2009. The benefits of statins in people without established cardiovascular disease but with cardiovascular risk factors: meta-analysis of randomised controlled trials. BMJ 338:b2376-b2383.
14.   Austin, M. A., M. C. King, K. M. Vranizan, and R. M. Krauss. 1990. Atherogenic lipoprotein phenotype. A proposed genetic marker for coronary heart disease risk. Circulation 82:495-506.
15.   Noto, H. M. 2009. Latest Insights Into High-Density Lipoprotein Functions. Endocrinologist 19:179-186.
16.   Beck C. 2008. Assembly and Secretion of Atherogenic Lipoproteins. PhD thesis. University of Gothenburg, Göteborg, Sweden
17.   Demant, T., A. Gaw, G. F. Watts, P. Durrington, B. Buckley, C. W. Imrie, C. Wilson, C. J. Packard, and J. Shepherd. 1993. Metabolism of apoB-100-containing lipoproteins in familial hyperchylomicronemia. J Lipid Res 34:147-156.
18.   Young, S. G., B. S. Davies, L. G. Fong, P. Gin, M. M. Weinstein, A. Bensadoun, and A. P. Beigneux. 2007. GPIHBP1: an endothelial cell molecule important for the lipolytic processing of chylomicrons. Curr Opin Lipidol 18:389-396.
19.   de Beer, F., W. L. Hendriks, L. C. van Vark, S. W. A. Kamerling, K. W. van Dijk, M. H. Hofker, A. H. M. Smelt, and L. M. Havekes. 1999. Binding of beta-VLDL to Heparan Sulfate Proteoglycans Requires Lipoprotein Lipase, Whereas ApoE Only Modulates Binding Affinity. Arterioscler Thromb Vasc Biol 19:633-637.
20.   Adiels M. 2004. Compartmental Models of Lipoprotein Kinetics. PhD Thesis, Chalmers University of Technology, Göteborg, Sweden.
21.   Voshol, P. J., P. C. Rensen, K. W. Van Dijk, J. A. Romijn, and L. M. Havekes. 2009. Effect of plasma triglyceride metabolism on lipid storage in adipose tissue: Studies using genetically engineered mouse models. Biochim Biophys Acta-doi:10.1016/j.bbalip.2008.12.015.
22.   Demant, T., L. A. Carlson, L. Holmquist, F. Karpe, P. Nilsson-Ehle, C. J. Packard, and J. Shepherd. 1988. Lipoprotein metabolism in hepatic lipase deficiency: studies on the turnover of apolipoprotein B and on the effect of hepatic lipase on high density lipoprotein. J Lipid Res 29:1603-1611.
23.   Jaye, M., K. J. Lynch, J. Krawiec, D. Marchadier, C. Maugeais, K. Doan, V. South, D. Amin, M. Perrone, and D. J. Rader. 1999. A novel endothelial-derived lipase that modulates HDL metabolism. Nat Genet 21:424-428.
24.   Hirata, K., H. n. L. Dichek, J. A. Cioffi, S. Y. Choi, N. J. Leeper, L. Quintana, G. S. Kronmal, A. D. Cooper, and T. Quertermous. 1999. Cloning of a Unique Lipase from Endothelial Cells Extends the Lipase Gene Family. J Biol Chem 274:14170-14175.
25.   Lamarche, B., and M. E. Paradis. 2007. Endothelial lipase and the metabolic syndrome. Curr Opin Lipidol 18:298-303.
26.   Morton, R. E., and D. B. Zilversmit. 1983. Inter-relationship of lipids transferred by the lipid-transfer protein isolated from human lipoprotein-deficient plasma. J Biol Chem 258:11751-11757.
27.   Yen, F. T., R. J. Deckelbaum, C. J. Mann, Y. L. Marcel, R. W. Milne, and A. R. Tall. 1989. Inhibition of cholesteryl ester transfer protein activity by monoclonal antibody. Effects on cholesteryl ester formation and neutral lipid mass transfer in human plasma. J Clin Invest 83:2018-2024.
28.   Millar, J. S., M. E. Brousseau, M. R. Diffenderfer, P. H. Barrett, F. K. Welty, A. Faruqi, M. L. Wolfe, C. Nartsupha, A. G. Digenio, J. P. Mancuso, G. G. Dolnikowski, E. J. Schaefer, and D. J. Rader. 2006. Effects of the cholesteryl ester transfer protein inhibitor torcetrapib on apolipoprotein B100 metabolism in humans. Arterioscler Thromb Vasc Biol 26:1350-1356.
29.   Krieger, M. 1994. Structures and Functions of Multiligand Lipoprotein Receptors: Macrophage Scavenger Receptors and LDL Receptor-Related Protein (LRP). Annu Rev Biochem 63:601-637.
30.   MacArthur, J. M., J. R. Bishop, K. I. Stanford, L. C. Wang, A. Bensadoun, J. L. Witztum, and J. D. Esko. 2007. Liver heparan sulfate proteoglycans mediate clearance of triglyceride-rich lipoproteins independently of LDL receptor family members. J Clin Invest 117:153-164.
31.   Van Eck, M., M. Hoekstra, R. Out, I. S. Bos, J. K. Kruijt, R. B. Hildebrand, and T. J. van Berkel. 2008. Scavenger receptor BI facilitates the metabolism of VLDL lipoproteins in vivo. J Lipid Res. 49:136-146.
32.   Gofman, J. W., and F. LINDGREN. 1950. The role of lipids and lipoproteins in atherosclerosis. Science 111:166-171.
33.   Lindgren, F. T., H. A. ELLIOTT, and J. W. Gofman. 1951. The ultracentrifugal characterization and isolation of human blood lipids and lipoproteins, with applications to the study of atherosclerosis. J Phys. Colloid Chem 55:80-93.
34.   Gofman, J. W., F. T. Lindgren, and H. ELLIOTT. 1949. Ultracentrifugal studies of lipoproteins of human serum. J Biol Chem 179:973-979.
35.   Havel, R. J., H. A. Eder, and J. H. BRAGDON. 1955. The distribution and chemical composition of ultracentrifugally separated lipoproteins in human serum. J. Clin. Invest 34:1345-1353.
36.   GOFMAN, J. W., M. A. R. T. HANIG, H. B. JONES, M. A. LAUFFER, E. Y. LAWRY, L. A. LEWIS, G. V. MANN, F. E. MOORE, F. R. E. D. OLMSTED, J. F. YEAGER, E. C. ANDRUS, J. H. BARACH, J. W. BEAMS, J. W. FERTIG, J. W. GOFMAN, M. A. LAUFFER, I. H. PAGE, J. A. SHANNON, F. J. STARE, and P. D. WHITE. 1956. Evaluation of Serum Lipoprotein and Cholesterol Measurements as Predictors of Clinical Complications of Atherosclerosis: Report of a Cooperative Study of Lipoproteins and Atherosclerosis. Circulation 14:689-741.
37.   BURSTEIN, M., and J. SAMAILLE. 1958. Determination of serum beta-lipoproteins after selective precipitation by heparin. Presse Med 66:974-975.
38.   Friedewald, W. T., R. I. Levy, and D. S. Fredrickson. 1972. Estimation of the Concentration of Low-Density Lipoprotein Cholesterol in Plasma, Without Use of the Preparative Ultracentrifuge. Clin Chem 18:499-502.
39.   Alaupovic, P. 1991. Apolipoprotein composition as the basis for classifying plasma lipoproteins: Characterization of ApoA- and ApoB-containing lipoprotein families. Progr Lipid Res 30:105-138.
40.   Alaupovic, P. 1996. Significance of apolipoproteins for structure, function, and classification of plasma lipoproteins. Method Enzymol 263:32-60.
41.   Walldius, G., A. H. Aastveit, and I. Jungner. 2006. Stroke mortality and the apoB/apoA-I ratio: Results of the AMORIS prospective study. J Intern Med 259:259-266.
42.   Kaess, B., M. Fischer, A. Baessler, K. Stark, F. Huber, W. Kremer, H. R. Kalbitzer, H. Schunkert, G. Riegger, and C. Hengstenberg. 2008. The lipoprotein subfraction profile: heritability and identification of quantitative trait loci. J Lipid Res 49:715-723.
43.   Kulkarni, K. R. 2006. Cholesterol Profile Measurement by Vertical Auto Profile Method. Clin Lab Med 26:787-802.
44.   Grundy, S. M. 2004. Richard Havel, Howard Eder, and the evolution of lipoprotein analysis. J Clin Invest 114:1034-1037.
45.   Barrett, P. H. R., D. C. Chan, and G. F. Watts. 2006. Design and analysis of lipoprotein tracer kinetics studies in humans. J Lipid Res 47:1607-1619.
46.   Parhofer, K. G., and P. H. Barrett. 2006. What we have learned about VLDL and LDL metabolism from human kinetic studies. J Lipid Res 47:1620-1630.
47.   Phair, R. D., M. G. Hammond, J. A. Bowden, M. Fried, W. R. Fisher, and M. Berman. 1975. Preliminary model for human lipoprotein metabolism in hyperlipoproteinemia. Fed Proc 34:2263-2270.
48.   Beltz, W. F., Y. A. Kesaniemi, B. V. Howard, and S. M. Grundy. 1985. Development of an integrated model for analysis of the kinetics of apolipoprotein B in plasma very low density lipoproteins, intermediate density lipoproteins, and low density lipoproteins. J Clin Invest 76:575-585.
49.   Barrett, P. H., N. Baker, and P. J. Nestel. 1991. Model development to describe the heterogeneous kinetics of apolipoprotein B and triglyceride in hypertriglyceridemic subjects. J Lipid Res 32:743-762.
50.   Packard, C. J., A. Gaw, T. Demant, and J. Shepherd. 1995. Development and application of a multicompartmental model to study very low density lipoprotein subfraction metabolism. J Lipid Res 36:172-187.
51.   Adiels, M., C. Packard, M. J. Caslake, P. Stewart, A. Soro, J. Westerbacka, B. Wennberg, S. O. Olofsson, M. R. Taskinen, and J. Borén. 2005. A new combined multicompartmental model for apolipoprotein B-100 and triglyceride metabolism in VLDL subfractions. J Lipid Res 46:58-67.
52.   Gitlin, D., D. G. Cornwell, D. Nakasato, J. L. Oncley, W. L. Hughes, and C. A. Janeway. 1958. Studies on the metabolism of plasma lipoproteins in the necrotic syndrome. II. The lipoproteins. J Clin Invest 37:720-729.
53.   Bilheimer, D. W., S. Eisenberg, and R. I. Levy. 1972. The metabolism of very low density lipoprotein proteins I. Preliminary in vitro and in vivo observations. BBA: Lipids & Lipid Metab 260:212-221.
54.   Cryer, D. R., T. Matsushima, and J. B. Marsh. 1986. Direct measurement of apolipoprotein B synthesis in human very low density lipoprotein using stable isotopes and mass spectrometry. J Lipid Res 27:508-516.
55.   Lichtenstein, A. H., J. S. Cohn, D. L. Hachey, J. S. Millar, J. M. Ordovas, and E. J. Schaefer. 1990. Comparison of deuterated leucine, valine, and lysine in the measurement of human apolipoprotein A-I and B-100 kinetics. J Lipid Res 31:1693-1701.
56.   Fisher, W. R., V. Venkatakrishnan, E. S. Fisher, P. W. Stacpoole, and L. A. Zech. 1997. The 3H-leucine tracer: Its use in kinetic studies of plasma lipoproteins. Metab Clin Exp 46:333-342.
57.   Musunuru, K., M. Orho-Melander, M. P. Caulfield, S. Li, W. A. Salameh, R. E. Reitz, G. Berglund, B. Hedblad, G. Engstrom, P. T. Williams, S. Kathiresan, O. Melander, and R. M. Krauss. 2009. Ion mobility analysis of lipoprotein subfractions identifies three independent axes of cardiovascular risk. Arterioscler Thromb Vasc Biol 29:1975-1980.
58.   Akobeng, A. K. 2007. Understanding diagnostic tests 1: Sensitivity, specificity and predictive values. Acta Paediatr 96:338-341.
59.   Panteleev, A., N. M. Ananyeva, F. I. Ataullakhanov, and E. L. Saenko. 2007. Mathematical models of blood coagulation and platelet adhesion: Clinical applications. Curr Pharm Des 13:1457-1467.
60.   Mari, A., G. Pacini, E. Murphy, B. Ludvik, and J. J. Nolan. 2001. A Model-Based Method for Assessing Insulin Sensitivity From the Oral Glucose Tolerance Test. Diab Care 24:539-548.
61.   Hubner, K., T. Schwager, K. Winkler, J. G. Reich, and H. G. Holzhutter. 2008. Computational lipidology: predicting lipoprotein density profiles in human blood plasma. PLoS Comput Biol 4:e1000079-doi:10.1371/journal.pcbi.1000079.
62.   Rizzo, M., and K. Berneis. 2006. Should we measure routinely the LDL peak particle size? Int J Cardiol 107:166-170.
63.   Forster, L. F., G. Stewart, D. Bedford, J. P. Stewart, E. Rogers, J. Shepherd, C. J. Packard, and M. J. Caslake. 2002. Influence of atorvastatin and simvastatin on apolipoprotein B metabolism in moderate combined hyperlipidemic subjects with low VLDL and LDL fractional clearance rates. Atherosclerosis 164:129-145.
64.   Issemann, I., and S. Green. 1990. Activation of a member of the steroid hormone receptor superfamily by peroxisome proliferators. Nature 347:645-650.
65.   Schoonjans, K., B. Staels, and J. Auwerx. 1996. The peroxisome proliferator activated receptors (PPARs) and their effects on lipid metabolism and adipocyte differentiation. BBA 1302:93-109.
66.   Staels, B., J. Dallongeville, J. Auwerx, K. Schoonjans, E. Leitersdorf, and J. C. Fruchart. 1998. Mechanism of Action of Fibrates on Lipid and Lipoprotein Metabolism. Circulation 98:2088-2093.
67.   Al-Shayji, I. A. R., J. M. R. Gill, J. Cooney, S. Siddiqui, and M. J. Caslake. 2007. Development of a novel method to determine very low density lipoprotein kinetics. J Lipid Res 48:2086-2095.
68.   Bilz, S., S. Wagner, M. Schmitz, A. Bedynek, U. Keller, and T. Demant. 2004. Effects of atorvastatin versus fenofibrate on apoB-100 and apoA-I kinetics in mixed hyperlipidemia. J Lipid Res 45:174-185.
69.   Demant, T., D. Bedford, C. J. Packard, and J. Shepherd. 1991. Influence of apolipoprotein E polymorphism on apolipoprotein B-100 metabolism in normolipemic subjects. J Clin Invest 88:1490-1501.
70.   Demant, T., C. J. Packard, H. Demmelmair, P. Stewart, A. Bedynek, D. Bedford, D. Seidel, and J. Shepherd. 1996. Sensitive methods to study human apolipoprotein B metabolism using stable isotope-labeled amino acids. Am J Physiol 270:E1022-E1036.
71.   Demant, T., C. Mathes, K. Gutlich, A. Bedynek, H. B. Steinhauer, T. Bosch, C. J. Packard, and G. L. Warwick. 1998. A simultaneous study of the metabolism of apolipoprotein B and albumin in nephrotic patients. Kidney Int 54:2064-2080.
72.   Gaffney, D., L. Forster, M. J. Caslake, D. Bedford, J. P. Stewart, G. Stewart, G. Wieringa, M. Dominiczak, J. P. Miller, and C. J. Packard. 2002. Comparison of apolipoprotein B metabolism in familial defective apolipoprotein B and heterogeneous familial hypercholesterolemia. Atherosclerosis 162:33-43.
73.   Gaw, A., C. J. Packard, E. F. Murray, G. M. Lindsay, B. A. Griffin, M. J. Caslake, B. D. Vallance, A. R. Lorimer, and J. Shepherd. 1993. Effects of simvastatin on apoB metabolism and LDL subfraction distribution. Arterioscler Thromb 13:170-189.
74.   Gaw, A., C. J. Packard, G. M. Lindsay, E. F. Murray, B. A. Griffin, M. J. Caslake, I. Colquhoun, D. J. Wheatley, A. R. Lorimer, and J. Shepherd. 1996. Effects of colestipol alone and in combination with simvastatin on apolipoprotein B metabolism. Arterioscler Thromb Vasc Biol 16:236-249.
75.   Lundahl, B., C. Skoglund-Andersson, M. Caslake, D. Bedford, P. Stewart, A. Hamsten, C. J. Packard, and F. Karpe. 2006. Microsomal triglyceride transfer protein -493T variant reduces IDL plus LDL apoB production and the plasma concentration of large LDL particles. Am J Physiol Endocrinol Metab 290:E739-E745.
76.   Nierman, M. C., B. H. C. M. Prinsen, J. Rip, R. J. Veldman, J. A. Kuivenhoven, J. J. P. Kastelein, M. de Sain-van der Velden, and E. S. G. Stroes. 2005. Enhanced Conversion of Triglyceride-Rich Lipoproteins and Increased Low-Density Lipoprotein Removal in LPLS447X Carriers. Arterioscler Thromb Vasc Biol 25:2410-2415.
77.   Packard, C. J., J. Shepherd, G. M. Lindsay, A. Gaw, and M. R. Taskinen. 1993. Thyroid replacement therapy and its influence on postheparin plasma lipases and apolipoprotein-B metabolism in hypothyroidism. J Clin Endocrinol Metab 76:1209-1216.
78.   Packard, C. J., T. Demant, J. P. Stewart, D. Bedford, M. J. Caslake, G. Schwertfeger, A. Bedynek, J. Shepherd, and D. Seidel. 2000. Apolipoprotein B metabolism and the distribution of VLDL and LDL subfractions. J Lipid Res 41:305-318.
79.   Schmitz, M., G. M. Michl, R. Walli, J. Bogner, A. Bedynek, D. Seidel, F. D. Goebel, and T. Demant. 2001. Alterations of apolipoprotein B metabolism in HIV-infected patients with antiretroviral combination therapy. J Acquir Immune Defic Syndr 26:225-235.
80.   Kannel, W. B., W. P. Castelli, T. Gordon, and P. M. McNamara. 1971. Serum cholesterol, lipoproteins, and the risk of coronary heart disease. The Framingham study. Ann Int Med 74:1-12.
81.   Freedman, D. S., J. D. Otvos, E. J. Jeyarajah, I. Shalaurova, A. Cupples, H. Parise, R. B. D'Agostino, P. W. F. Wilson, and E. J. Schaefer. 2004. Sex and age differences in lipoprotein subclasses measured by nuclear magnetic resonance spectroscopy: The Framingham study. Clin Chem 50:1189-1200.
82.   Metges, C. C., and G. Wolfram. 1991. Medium and Long Chain Triglycerides Labeled with 13C: A Comparison of Oxidation after Oral or Parenteral Administration in Humans. J Nutr 121:31-36.
83.   Pasman WJ, de Graaf AA. 2009. The effect of body fat distribution on the physiological response to a dietary fat intervention. Rep. 7261, TNO, Zeist, the Netherlands.
84.   Berman, M., M. Hall, III, R. I. Levy, S. Eisenberg, D. W. Bilheimer, R. D. Phair, and R. H. Goebel. 1978. Metabolism of apoB and apoC lipoproteins in man: kinetic studies in normal and hyperlipoproteinemic subjects. J. Lipid Res. 19:38-56.
85.   Fisher, W. R., L. A. Zech, P. Bardalaye, G. Warmke, and M. Berman. 1980. The metabolism of apolipoprotein B in subjects with hypertriglyceridemia and polydisperse LDL. J. Lipid Res. 21:760-774.
86.   Fisher, W. R., L. A. Zech, L. L. Kilgore, and P. W. Stacpoole. 1991. Metabolic pathways of apolipoprotein B in heterozygous familial hypercholesterolemia: studies with a [3H]leucine tracer. J. Lipid Res. 32:1823-1836.
87.   Packard, C. J., A. Munro, A. R. Lorimer, A. M. Gotto, and J. Shepherd. 1984. Metabolism of apolipoprotein B in large triglyceride-rich very low density lipoproteins of normal and hypertriglyceridemic subjects. J Clin Invest 74:2178-2192.
88.   Ginsberg, H. N., N. A. Le, and J. C. Gibson. 1985. Regulation of the production and catabolism of plasma low density lipoproteins in hypertriglyceridemic subjects. Effect of weight loss. J. Clin. Invest 75:614-623.
89.   Krul, E. S., K. G. Parhofer, P. H. Barrett, R. D. Wagner, and G. Schonfeld. 1992. ApoB-75, a truncation of apolipoprotein B associated with familial hypobetalipoproteinemia: genetic and kinetic studies. J. Lipid Res. 33:1037-1050.
90.   Malmstrom, R., C. J. Packard, T. D. Watson, S. Rannikko, M. Caslake, D. Bedford, P. Stewart, H. Yki-Jarvinen, J. Shepherd, and M. R. Taskinen. 1997. Metabolic basis of hypotriglyceridemic effects of insulin in normal men. Arterioscler. Thromb. Vasc. Biol. 17:1454-1464.
91.   Patterson, B. W., B. Mittendorfer, N. Elias, R. Satyanarayana, and S. Klein. 2002. Use of stable isotopically labeled tracers to measure very low density lipoprotein-triglyceride turnover. J. Lipid Res. 43:223-233.
92.   Zech, L. A., S. M. Grundy, D. Steinberg, and M. Berman. 1979. Kinetic model for production and metabolism of very low density lipoprotein triglycerides. Evidence for a slow production pathway and results for normolipidemic subjects. J. Clin. Invest 63:1262-1273.
93.   Melish, J., N. A. Le, H. Ginsberg, D. Steinberg, and W. V. Brown. 1980. Dissociation of apoprotein B and triglyceride production in very-low-density lipoproteins. Am. J. Physiol 239:E354-E362.
94.   Harris, W. S., W. E. Connor, D. R. Illingworth, D. W. Rothrock, and D. M. Foster. 1990. Effects of fish oil on VLDL triglyceride kinetics in humans. J. Lipid Res. 31:1549-1558.
95.   Campos, H., D. Perlov, C. Khoo, and F. M. Sacks. 2001. Distinct patterns of lipoproteins with apoB defined by presence of apoE or apoC-III in hypercholesterolemia and hypertriglyceridemia. Journal of Lipid Research 42:1239-1249.
96.   Tomiyasu, K., B. W. Walsh, K. Ikewaki, H. Judge, and F. M. Sacks. 2001. Differential metabolism of human VLDL according to content of apoE and apoC-III. Arterioscler Thromb Vasc Biol 21:1494-1500.
97.   Zheng, C., C. Khoo, K. Ikewaki, and F. M. Sacks. 2007. Rapid turnover of apolipoprotein C-III-containing triglyceride-rich lipoproteins contributing to the formation of LDL subfractions. J. Lipid Res. 48:1190-1203.
98.   Chan, D. C., G. F. Watts, M. N. Nguyen, and P. H. Barrett. 2006. Apolipoproteins C-III and A-V as predictors of very-low-density lipoprotein triglyceride and apolipoprotein B-100 kinetics. Arterioscler. Thromb. Vasc. Biol. 26:590-596.
99.   Cohn, J. S., B. W. Patterson, K. D. Uffelman, J. Davignon, and G. Steiner. 2004. Rate of production of plasma and very-low-density lipoprotein (VLDL) apolipoprotein C-III is strongly related to the concentration and level of production of VLDL triglyceride in male subjects with different body weights and levels of insulin sensitivity. J. Clin. Endocrinol. Metab 89:3949-3955.
100.   Okazaki, M., S. Usui, M. Ishigami, N. Sakai, T. Nakamura, Y. Matsuzawa, and S. Yamashita. 2005. Identification of Unique Lipoprotein Subclasses for Visceral Obesity by Component Analysis of Cholesterol Profile in High-Performance Liquid Chromatography. Arterioscler Thromb Vasc Biol 25:578-584.
101.   Tuzikov, F. V., N. A. Tuzikova, R. V. Galimov, L. E. Panin, and G. A. Nevinsky. 2002. General model to describe the structure and dynamic balance between different human serum lipoproteins and its practical application. Med. Sci. Monitor 8:MT79-88.
102.   Blasiole, D. A., R. A. Davis, and A. D. Attie. 2007. The physiological and molecular regulation of lipoprotein assembly and secretion. Mol. Biosyst. 3:608-619.
103.   Stillemark-Billton, P., C. Beck, J. Borén, and S. O. Olofsson. 2005. Relation of the size and intracellular sorting of apoB to the formation of VLDL 1 and VLDL 2. J Lipid Res. 46:104-114.
104.   Olofsson, S. O., and J. Borén. 2005. Apolipoprotein B: a clinically important apolipoprotein which assembles atherogenic lipoproteins and promotes the development of atherosclerosis. Journal of Internal Medicine 258:395-410.
105.   Olofsson, S. O., P. Bostrom, L. Andersson, M. Rutberg, J. Perman, and J. Borén. 2008. Lipid droplets as dynamic organelles connecting storage and efflux of lipids. Biochim. Biophys. Acta-doi:10.1016/j.bbalip.2008.08.001.
106.   Lichtenstein, L., J. F. Berbee, S. J. van Dijk, K. W. Van Dijk, A. Bensadoun, I. P. Kema, P. J. Voshol, M. Muller, P. C. Rensen, and S. Kersten. 2007. Angptl4 upregulates cholesterol synthesis in liver via inhibition of LPL- and HL-dependent hepatic cholesterol uptake. Arterioscler Thromb Vasc Biol 27:2420-2427.
107.   Stein, Y., and O. Stein. 2003. Lipoprotein lipase and atherosclerosis. Atherosclerosis 170:1-9.
108.   Zambon, A., S. S. Deeb, P. Pauletto, G. Crepaldi, and J. D. Brunzell. 2003. Hepatic lipase: A marker for cardiovascular disease risk and response to therapy. Curr. Opin. Lipidol. 14:179-189.
109.   Connelly, P. W., G. F. Maguire, C. Vezina, R. A. Hegele, and A. Kuksis. 1994. Kinetics of lipolysis of very low density lipoproteins by lipoprotein lipase. Importance of particle number and noncompetitive inhibition by particles with low triglyceride content. J. Biol. Chem. 269:20554-20560.
110.   Schreier, L., G. Berg, V. Zago, A. I. Gonzalez, and R. Wikinski. 2002. Kinetics of in vitro lipolysis of human very low-density lipoprotein by lipoprotein lipase. Nutrition, Metabolism and Cardiovascular Diseases 12:13-18.
111.   Guerin, M., P. J. Dolphin, C. Talussot, J. Gardette, F. Berthezene, and M. J. Chapman. 1995. Pravastatin Modulates Cholesteryl Ester Transfer From HDL to ApoB-Containing Lipoproteins and Lipoprotein Subspecies Profile in Familial Hypercholesterolemia. Arterioscler Thromb Vasc Biol 15:1359-1368.
112.   Campos, H., D. M. Dreon, and R. M. Krauss. 1995. Associations of hepatic and lipoprotein lipase activities with changes in dietary composition and low density lipoprotein subclasses. J. Lipid Res. 36:462-472.
113.   Tan, C. E., L. Forster, M. J. Caslake, D. Bedford, T. D. G. Watson, M. McConnell, C. J. Packard, and J. Shepherd. 1995. Relations between plasma lipids and postheparin plasma lipases and VLDL and LDL subfraction patterns in normolipemic men and women. Arterioscler Thromb Vasc Biol 15:1839-1848.
114.   Chen, G. C., W. Liu, P. Duchateau, J. Allaart, R. L. Hamilton, C. M. Mendel, K. Lau, D. A. Hardman, P. H. Frost, M. J. Malloy, and . 1994. Conformational differences in human apolipoprotein B-100 among subspecies of low density lipoproteins (LDL). Association of altered proteolytic accessibility with decreased receptor binding of LDL subspecies from hypertriglyceridemic subjects. J Biol. Chem 269:29121-29128.
115.   Nigon, F., P. Lesnik, M. Rouis, and M. J. Chapman. 1991. Discrete subspecies of human low density lipoproteins are heterogeneous in their interaction with the cellular LDL receptor. J. Lipid Res. 32:1741-1753.
116.   Campos, H., K. S. Arnold, M. E. Balestra, T. L. Innerarity, and R. M. Krauss. 1996. Differences in Receptor Binding of LDL Subfractions. Arterioscler Thromb Vasc Biol 16:794-801.
117.   Thompson, A., E. Di Angelantonio, N. Sarwar, S. Erqou, D. Saleheen, R. P. F. Dullaart, B. Keavney, Z. Ye, and J. Danesh. 2008. Association of Cholesteryl Ester Transfer Protein Genotypes With CETP Mass and Activity, Lipid Levels, and Coronary Risk. JAMA 299:2777-2788.
118.   Magkos, F., and L. S. Sidossis. 2004. Measuring very low density lipoprotein-triglyceride kinetics in man in vivo: how different the various methods really are. Curr. Opin. Clin. Nutr. Metab Care 7:547-555.
119.   Adiels, M., M. R. Taskinen, C. Packard, M. J. Caslake, A. Soro-Paavonen, J. Westerbacka, S. Vehkavaara, A. Häkkinen, S. O. Olofsson, H. Yki-Järvinen, and J. Borén. 2006. Overproduction of large VLDL particles is driven by increased liver fat content in man. Diabetologia 49:755-765.
120.   Cromwell, W. C., and J. D. Otvos. 2004. Low-density lipoprotein particle number and risk for cardiovascular disease. Curr. Atheroscl. Rep. 6:381-387.
121.   Grundy, S. M., J. I. Cleeman, C. N. Merz, H. B. Brewer, Jr., L. T. Clark, D. B. Hunninghake, R. C. Pasternak, S. C. Smith, Jr., and N. J. Stone. 2004. Implications of recent clinical trials for the National Cholesterol Education Program Adult Treatment Panel III guidelines. Circulation 110:227-239.
122.   Olivecrona, T., M. Hultin, M. Bergö, and G. Olivecrona. 1997. Lipoprotein lipase: regulation and role in lipoprotein metabolism. Proceedings of the Nutrition Society 56:723-729.
123.   Olivecrona, T., and G. Bengtsson-Olivecrona. 1993. Lipoprotein lipase and hepatic lipase. Current Opinion in Lipidology 4:187-196.
124.   Frayn, K. N. 2003. Metabolic Regulation, A Human Perspective, Blackwell Science Ltd., Oxford. 264.
125.   van Schalkwijk, D. B., A. A. de Graaf, B. van Ommen, K. van Bochove, P. C. N. Rensen, L. M. Havekes, N. C. A. van de Pas, H. C. J. Hoefsloot, J. van der Greef, and A. P. Freidig. 2009. Improved cholesterol phenotype analysis by a model relating lipoprotein life cycle processes to particle size. J Lipid Res 50:2398-2411.
126.   van Schalkwijk, D. B., K. van Bochove, B. van Ommen, A. P. Freidig, E. P. van Someren, J. van der Greef, and A. A. de Graaf. 2010. Developing computational model-based diagnostics to analyse clinical chemistry data. Brief. Bioinform. 11:403-416.
127.   James, R. W., B. Martin, D. Pometta, J. C. Fruchart, P. Duriez, P. Puchois, J. P. Farriaux, A. Tacquet, T. Demant, R. J. Clegg, and . 1989. Apolipoprotein B metabolism in homozygous familial hypercholesterolemia. J Lipid Res. 30:159-169.
128.   Obuchowski, N. A. 2005. ROC analysis. Am J Roentgenol. 184:364-372.
129.   Ruel, I. L., P. Couture, J. S. Cohn, and B. Lamarche. 2005. Plasma metabolism of apoB-containing lipoproteins in patients with hepatic lipase deficiency. Atherosclerosis 180:355-366.
130.   Walker, C., and B. V. Reamy. 2009. Diets for cardiovascular disease prevention: What is the evidence? Am Fam Phys 79:571-578.
131.   St-Onge, M. P., and A. Bosarge. 2008. Weight-loss diet that includes consumption of medium-chain triacylglycerol oil leads to a greater rate of weight and fat mass loss than does olive oil. Am J Clin Nutr 87:621-626.
132.   Marten, B., M. Pfeuffer, and J. Schrezenmeir. 2006. Medium-chain triglycerides. Int Dairy J 16:1374-1382.
133.   Horrobin, D. F., and Y. S. Huang. 1987. The role of linoleic acid and its metabolites in the lowering of plasma cholesterol and the prevention of cardiovascular disease. Int J Cardiol 17:241-255.
134.   Bach, A. C., and V. K. Babayan. 1982. Medium-chain triglycerides: An update. Am J Clin Nutr 36:950-962.
135.   Shen, B. W., A. M. Scanu, and F. J. Kezdy. 1977. Structure of human serum lipoproteins inferred from compositional analysis. Proc. Natl. Acad. Sci. U. S. A. 74:837-841.
136.   Forman, B. M., J. Chen, and R. M. Evans. 1997. Hypolipidemic drugs, polyunsaturated fatty acids, and eicosanoids are ligands for peroxisome proliferator-activated receptors alpha and delta. Proc Natl. Acad. Sci U. S. A 94:4312-4317.
137.   Krey, G., O. Braissant, F. L'Horset, E. Kalkhoven, M. Perroud, M. G. Parker, and W. Wahli. 1997. Fatty Acids, Eicosanoids, and Hypolipidemic Agents Identified as Ligands of Peroxisome Proliferator-Activated Receptors by Coactivator-Dependent Receptor Ligand Assay. Mol Endocrinol 11:779-791.
138.   Kliewer, S. A., S. S. Sundseth, S. A. Jones, P. J. Brown, G. B. Wisely, C. S. Koble, P. Devchand, W. Wahli, T. M. Willson, J. M. Lenhard, and J. M. Lehmann. 1997. Fatty acids and eicosanoids regulate gene expression through direct interactions with peroxisome proliferator-activated receptors <alpha> and <gamma>. Proc Natl Acad Sci USA 94:4318-4323.
139.   van Bochove, C., D. B. van Schalkwijk, L. D. Parnell, C. Q. Lai, J. M. Ordovas, A. A. de Graaf, B. van Ommen, and D. K. Arnett. 2012. Clustering by plasma lipoprotein profile reveals two distinct subgroups with positive lipid response to fenofibrate therapy. PLoS One 7:e38072.
140.   Ginsberg, H. N., D. E. Bonds, L. C. Lovato, J. R. Crouse, M. B. Elam, P. E. Linz, P. J. O'Connor, L. A. Leiter, D. Weiss, E. Lipkin, and J. L. Fleg. 2007. Evolution of the lipid trial protocol of the Action to Control Cardiovascular Risk in Diabetes (ACCORD) trial. Am J Cardiol. 99:56i-67i.
141.   Lear, S. A., K. H. Humphries, S. Kohli, J. J. Frohlich, C. L. Birmingham, and G. B. J. Mancini. 2007. Visceral adipose tissue, a potential risk factor for carotid atherosclerosis: Results of the Multicultural Community Health Assessment Trial (M-CHAT). Stroke 38:2422-2429.
142.   Huxley, R., S. Mendis, E. Zheleznyakov, S. Reddy, and J. Chan. 2010. Body mass index, waist circumference and waist:hip ratio as predictors of cardiovascular riska review of the literature. Eur J Clin Nutr 64:16-22.
143.   Biolo, G., G. Toigo, and G. Guarnieri. 2001. Slower activation of insulin action in upper body obesity. Metabolism 50:19-23.
144.   McCloy, U., M. A. Ryan, P. B. Pencharz, R. J. Ross, and S. C. Cunnane. 2004. A comparison of the metabolism of eighteen-carbon 13C-unsaturated fatty acids in healthy women. J Lipid Res 45:474-485.
145.   van Eijk, H. M. H., and N. E. P. Deutz. 2003. Plasma Protein Synthesis Measurements Using a Proteomics Strategy. J Nutr 133:2084S-2089S.
146.   ISO 5508:1990 Animal and vegetable fats and oils - Analysis by gas chromatography of methyl esters of fatty acids.  1990.
Ref Type: Statute
147.   ISO 5509:2000 Animal and vegetable fats and oils - Preparation of methyl esters of fatty acids.  2000.
Ref Type: Statute
148.   Verschuren, L., R. Kleemann, E. H. Offerman, A. J. Szalai, S. J. Emeis, H. M. Princen, and T. Kooistra. 2005. Effect of low dose atorvastatin versus diet-induced cholesterol lowering on atherosclerotic lesion progression and inflammation in apolipoprotein E*3-Leiden transgenic mice. Arterioscler. Thromb. Vasc. Biol. 25:161-167.
149.   van Schalkwijk, D. B., B. van Ommen, A. P. Freidig, J. van der Greef, and A. A. de Graaf. 2011. Diagnostic Markers based on a Computational Model of Lipoprotein Metabolism. J Clin Bioinform. 1:29.
150.   Sato, K., Y. Cho, S. Tachibana, T. Chiba, W. J. Schneider, and Y. Akiba. 2005. Impairment of VLDL secretion by medium-chain fatty acids in chicken primary hepatocytes is affected by the chain length. J. Nutr. 135:1636-1641.
151.   Tachibana, S., K. Sato, Y. Cho, T. Chiba, W. J. Schneider, and Y. Akiba. 2005. Octanoate reduces very low-density lipoprotein secretion by decreasing the synthesis of apolipoprotein B in primary cultures of chicken hepatocytes. BBA-mol cell biol l 1737:36-43.
152.   Asakura, L., A. M. P. Lottenberg, M. Q. T. S. Neves, V. S. Nunes, J. C. Rocha, M. Passarelli, E. R. Nakandakare, and E. C. R. Quint+úo. 2000. Dietary medium-chain triacylglycerol prevents the postprandial rise of plasma triacylglycerols but induces hypercholesterolemia in primary hypertriglyceridemic subjects. Am J Clin Nutr 71:701-705.
153.   Hill, J. O., J. C. Peters, L. I. Swift, D. Yang, T. Sharp, N. Abumrad, and H. L. Greene. 1990. Changes in blood lipids during six days of overfeeding with medium or long chain triglycerides. J Lipid Res 31:407-416.
154.   Swift, L. L., J. O. Hill, J. C. Peters, and H. L. Greene. 1992. Plasma lipids and lipoproteins during 6 d of maintenance feeding with long-chain, medium-chain, and mixed-chain triglycerides. Am J Clin Nutr 56:881-886.
155.   Seidell, J. C., L. Perusse, J. P. Despres, and C. Bouchard. 2001. Waist and hip circumferences have independent and opposite effects on cardiovascular disease risk factors: the Quebec Family Study. Am J Clin Nutr 74:315-321.
156.   St-Onge, M. P., and P. J. H. Jones. 2003. Greater rise in fat oxidation with medium-chain triglyceride consumption relative to long-chain triglyceride is associated with lower initial body weight and greater loss of subcutaneous adipose tissue. Int J Obesity 27:1565-1571.
157.   St-Onge, M. P., R. Ross, W. D. Parsons, and P. J. H. Jones. 2003. Medium-chain triglycerides increase energy expenditure and decrease adiposity in overweight men. Obesity Res 11:395-402.
158.   Turkenkopf, I. J., C. A. Maggio, and M. R. C. Greenwood. 1982. Effect of high fat weanling diets containing either medium-chain triglycerides or long-chain triglycerides on the development of obesity in the Zucker rat. J. Nutr. 112:1254-1263.
159.   Thomas, T. R., K. E. Horner, M. M. Langdon, J. Q. Zhang, E. S. Krul, G. Y. Sun, and R. H. Cox. 2001. Effect of exercise and medium-chain fatty acids on postprandial lipemia. J Appl Physiol 90:1239-1246.
160.   Wierzbicki, A. S. 2010. Fibrates: No ACCORD on their use in the treatment of dyslipidaemia. Curr Opin Lipidol 21:352-358.
161.   Elam, M., L. C. Lovato, and H. Ginsberg. 2011. Role of fibrates in cardiovascular disease prevention, the ACCORD-Lipid perspective. Curr Opin Lipidol 22:55-61.
162.   Bruckert, E., J. Labreuche, D. Deplanque, P. J. Touboul, and P. Amarenco. 2011. Fibrates effect on cardiovascular risk is greater in patients with high triglyceride levels or atherogenic dyslipidemia profile: A systematic review and meta-analysis. J Cardiovasc Pharmacol 57:267-272.
163.   Committee of Principal Investigators. 1978. A co-operative trial in the primary prevention of ischaemic heart disease using clofibrate. Report from the Committee of Principal Investigators. Br. Heart J 40:1069-1118.
164.   The Coronary Drug Project Research Group. 1975. Clofibrate and Niacin in Coronary Heart Disease. JAMA 231:360-381.
165.   Frick, M. H., O. Elo, K. Haapa, O. P. Heinonen, P. Heinsalmi, P. Helo, J. K. Huttunen, P. Kaitaniemi, P. Koskinen, and V. Manninen. 1987. Helsinki Heart Study: primary-prevention trial with gemfibrozil in middle-aged men with dyslipidemia. Safety of treatment, changes in risk factors, and incidence of coronary heart disease. N Engl J Med 317:1237-1245.
166.   Rubins, H. B., S. J. Robins, D. Collins, C. L. Fye, J. W. Anderson, M. B. Elam, F. H. Faas, E. Linares, E. J. Schaefer, G. Schectman, T. J. Wilt, J. Wittes, and The Veterans Affairs High-Density Lipoprotein Cholesterol Intervention Trial Study Group. 1999. Gemfibrozil for the Secondary Prevention of Coronary Heart Disease in Men with Low Levels of High-Density Lipoprotein Cholesterol. N Engl J Med 341:410-418.
167.   The BIP study group. 2000. Secondary Prevention by Raising HDL Cholesterol and Reducing Triglycerides in Patients With Coronary Artery Disease : The Bezafibrate Infarction Prevention (BIP) Study. Circulation 102:21-27.
168.   Meade, T., R. Zuhrie, C. Cook, and J. Cooper. 2002. Bezafibrate in men with lower extremity arterial disease: randomised controlled trial. BMJ 325:1139.
169.   DAIS investigators. 2001. Effect of fenofibrate on progression of coronary-artery disease in type 2 diabetes: the Diabetes Atherosclerosis Intervention Study, a randomised study. Lancet 357:905-910.
170.   The FIELD study investigators. 2005. Effects of long-term fenofibrate therapy on cardiovascular events in 9795 people with type 2 diabetes mellitus (the FIELD study): randomised controlled trial. Lancet 366:1849-1861.
171.   Scott, R., M. d'Emden, J. Best, P. Drury, C. Ehnholm, A. Kesaniemi, C. Pardy, D. Tse, P. Barter, M. R. Taskinen, S. Copt, A. Keech, and on behalf of the FIELD Investigators. 2007. Abstract 3691: Features Of Metabolic Syndrome Identify Individuals With Type 2 Diabetes Mellitus At High Risk For Cardiovascular Events And Greater Absolute Benefits Of Fenofibrate. Circulation 116:II-838.
172.   Manninen, V., L. Tenkanen, P. Koskinen, J. K. Huttunen, M. Manttari, O. P. Heinonen, and M. H. Frick. 1992. Joint effects of serum triglyceride and LDL cholesterol and HDL cholesterol concentrations on coronary heart disease risk in the Helsinki Heart Study. Implications for treatment. Circulation 85:37-45.
173.   Robins, S. J., H. B. Rubins, F. H. Faas, E. J. Schaefer, M. B. Elam, J. W. Anderson, and D. Collins. 2003. Insulin Resistance and Cardiovascular Events With Low HDL Cholesterol. Diabetes Care 26:1513-1517.
174.   Wierzbicki, A. S. 2006. FIELDS of dreams, fields of tears: a perspective on the fibrate trials. Int. J Clin Pract. 60:442-449.
175.   Lai, C. Q., D. K. Arnett, D. Corella, R. J. Straka, M. Y. Tsai, J. M. Peacock, X. Adiconis, L. D. Parnell, J. E. Hixson, M. A. Province, and J. M. Ordovas. 2007. Fenofibrate Effect on Triglyceride and Postprandial Response of Apolipoprotein A5 Variants: The GOLDN Study. Arterioscler Thromb Vasc Biol 27:1417-1425.
176.   Matthews, D. R., J. P. Hosker, A. S. Rudenski, B. A. Naylor, D. F. Treacher, and R. C. Turner. 1985. Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia 28:412-419.
177.   Otvos, J. D. 2002. Measurement of lipoprotein subclass profiles by nuclear magnetic resonance spectroscopy. Clin Lab 48:171-180.
178.   Otvos, J. D., E. J. Jeyarajah, and W. C. Cromwell. 2002. Measurement issues related to lipoprotein heterogeneity. Am J Cardiol 90:22i-29i.
179.   Jeyarajah, E. J., W. C. Cromwell, and J. D. Otvos. 2006. Lipoprotein particle analysis by nuclear magnetic resonance spectroscopy. Clin. Lab Med. 26:847-870.
180.   Expert Panel on Detection, E. a. T. o. H. B. C. i. A. 2001. Executive Summary of the Third Report of the National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III). JAMA 285:2486-2497.
181.   Kleemann, R., E. M. van, L. Verschuren, A. M. van den Hoek, M. Koek, P. Y. Wielinga, A. Jie, L. Pellis, I. Bobeldijk-Pastorova, T. Kelder, K. Toet, S. Wopereis, N. Cnubben, C. Evelo, O. B. van, and T. Kooistra. 2010. Time-resolved and tissue-specific systems analysis of the pathogenesis of insulin resistance. PLoS One. 5:e8817.
182.   El Harchaoui, K., W. A. van der Steeg, E. S. G. Stroes, J. A. Kuivenhoven, J. D. Otvos, N. J. Wareham, B. A. Hutten, J. J. P. Kastelein, K. T. Khaw, and S. M. Boekholdt. 2007. Value of Low-Density Lipoprotein Particle Number and Size as Predictors of Coronary Artery Disease in Apparently Healthy Men and Women: The EPIC-Norfolk Prospective Population Study. J. Am. Coll. Cardiol. 49:547-553.
183.   Després, J. P., I. Lemieux, G. R. Dagenais, B. Cantin, and B. Lamarche. 2000. HDL-cholesterol as a marker of coronary heart disease risk: the Québec cardiovascular study. Atherosclerosis 153:263-272.
184.   Zeljkovic, A., V. Spasojevic-Kalimanovska, J. Vekic, Z. Jelic-Ivanovic, A. Topic, N. Bogavac-Stanojevic, S. Spasic, A. Vujovic, and D. Kalimanovska-Ostric. 2008. Does simultaneous determination of LDL and HDL particle size improve prediction of coronary artery disease risk? Clin. Exp. Med. 8:109-116.
185.   The ACCORD Study Group. 2010. Effects of Combination Lipid Therapy in Type 2 Diabetes Mellitus. N Engl J Med. 362:1563-1574.
186.   D'Agostino, R. B., Sr., R. S. Vasan, M. J. Pencina, P. A. Wolf, M. Cobain, J. M. Massaro, and W. B. Kannel. 2008. General Cardiovascular Risk Profile for Use in Primary Care: The Framingham Heart Study. Circulation 117:743-753.
187.   Ala-Korpela, M., A. Korhonen, J. Keisala, S. H+¦rkk+¦, P. Korpi, L. P. Ingman, J. Jokisaari, M. J. Savolainen, and Y. A. Kes+ñniemi. 1994. 1H NMR-based absolute quantitation of human lipoproteins and their lipid contents directly from plasma. J Lipid Res 35:2292-2304.
188.   Rambaldi, D. C., P. Reschiglian, A. Zattoni, and C. Johann. 2009. Enzymatic determination of cholesterol and triglycerides in serum lipoprotein profiles by asymmetrical flow field-flow fractionation with on-line, dual detection. Anal. Chim. Acta 654:64-70.
189.   Kulkarni, K. R., D. W. Garber, S. M. Marcovina, and J. P. Segrest. 1994. Quantification of cholesterol in all lipoprotein classes by the VAP-II method. J Lipid Res 35:159-168.
190.   Hastie, T., R. Tibshirani, and J. H. Friedman. 2009. The elements of statistical learning: data mining, inference, and prediction, Springer-Verlag, New York. 553.
191.   Tsivtsivadze, E., T. Pahikkala, J. Boberg, T. Salakoski, and T. Heskes. 2010. Co-Regularized Least-Squares for Label Ranking. In Preference Learning. 107-123.
192.   Guyon, I., S. Gunn, M. Nikravesh, and L. A. Zadeh. 2006. Feature Extraction: Foundations and Applications (Studies in Fuzziness and Soft Computing), Springer-Verlag, New York.
193.   Lin, H.-T., C.-J. Lin, and R. C. Weng. 2007. A note on Platt's probabilistic outputs for support vector machines. Machine Learning 68:267-276.
194.   Platt, J. C. 1999. Probabilistic Outputs for Support Vector Machines and Comparisons to Regularized Likelihood Methods. In Advances in large margin classifiers. MIT Press 61-74.
195.   Pencina, M. J., R. B. D'Agostino, Sr., R. B. D'Agostino, Jr., and R. S. Vasan. 2008. Evaluating the added predictive ability of a new marker: from area under the ROC curve to reclassification and beyond. Stat. Med 27:157-172.
196.   Ben-Zvi, A., S. D. Vernon, and G. Broderick. 2009. Model-based therapeutic correction of hypothalamic-pituitary-adrenal axis dysfunction. PLoS Computational Biology 5:-e1000273. doi:10.1371/journal.pcbi.1000273.
197.   Clark, T. J., P. M. Barton, A. Coomarasamy, J. K. Gupta, and K. S. Khan. 2006. Investigating postmenopausal bleeding for endometrial cancer: Cost-effectiveness of initial diagnostic strategies. BJOG: An International Journal of Obstetrics and Gynaecology 113:502-510.
198.   Danhof, M., E. C. M. de Lange, O. E. la Pasqua, B. A. Ploeger, and R. A. Voskuyl. 2008. Mechanism-based pharmacokinetic-pharmacodynamic (PK-PD) modeling in translational drug research. Trends in Pharmacological Sciences 29:186-191.
199.   Rajman, I. 2008. PK/PD modelling and simulations: utility in drug development. Drug Discovery Today 13:341-346.
200.   Steinman, D. A. 2004. Image-based Computational Fluid Dynamics: A New Paradigm for Monitoring Hemodynamics and Atherosclerosis. Current Drug Targets - Cardiovascular & Haematological Disorders 4:183-197.
201.   Oresic, M., A. Vidal-Puig, and V. Hännninen. 2006. Metabolomic approaches to phenotype characterization and applications to complex diseases. Expert Review of Molecular Diagnostics 6:575-585.
202.   Scalbert, A., L. Brennan, O. Fiehn, T. Hankemeier, B. Kristal, B. van Ommen, E. Pujos-Guillot, E. Verheij, D. Wishart, and S. Wopereis. 2009. Mass-spectrometry-based metabolomics: limitations and recommendations for future progress with particular focus on nutrition research. Metabolomics 5:435-458.
203.   Picardi, A., and P. Pozzilli. 2003. Dynamic tests in the clinical management of diabetes. J Endocrinol. Invest 26:99-106.
204.   Lutz, A. M., J. K. Willmann, F. V. Cochran, P. Ray, and S. S. Gambhir. 2008. Cancer Screening: A Mathematical Model Relating Secreted Blood Biomarker Levels to Tumor Sizes. PLoS medicine 5:e170.
205.   Iakovidis, I., P. Wilson, and J. C. Healy. 2004. E-health: current situation and examples of implemented and beneficial e-health applications, IOS Press, Fairfax, V.A., U.S.A.
206.   Martin-Martin, L. S., A. Latini, A. Pagano, A. Ragno, R. Stasi, A. Coppè, G. Davoli, A. Crescenzi, A. Alimonti, and A. Migliore. 2003. A new mathematical model based on clinical and laboratory variables for the diagnosis of Sjögren's syndrome. Clinical Rheumatology 22:123-126.
207.   Wei, J. T., Z. Zhang, S. D. Barnhill, K. R. Madyastha, H. Zhang, and J. E. Oesterling. 1998. Understanding artificial neural networks and exploring their potential applications for the practicing urologist. Urology 52:161-172.
208.   Schneider, J., G. Peltri, N. Bitterlich, K. Neu, H. G. Velcovsky, H. Morr, N. Katz, and E. Eigenbrodt. 2003. Fuzzy logic-based tumor marker profiles including a new marker tumor M2-PK improved sensitivity to the detection of progression in lung cancer patients. Anticancer Res 23:899-906.
209.   Kiran, M., S. Coakley, N. Walkinshaw, P. McMinn, and M. Holcombe. 2007. Validation and discovery from computational biology models. Biosystems 93:141-150.
210.   Sun, T., P. McMinn, S. Coakley, M. Holcombe, R. Smallwood, and S. MacNeil. 2007. An integrated systems biology approach to understanding the rules of keratinocyte colony formation. J R Soc Interface 4:1077-1092.
211.   Hoops, S., S. Sahle, R. Gauges, C. Lee, J. Pahle, N. Simus, M. Singhal, L. Xu, P. Mendes, and U. Kummer. 2006. COPASI--a COmplex PAthway SImulator. Bioinformatics 22:3067-3074.
212.   Needham, C. J., J. R. Bradford, A. J. Bulpitt, and D. R. Westhead. 2007. A Primer on Learning in Bayesian Networks for Computational Biology. PLoS Comput Biol 3:e129-doi:10.1371/journal.pcbi.0030129.
213.   Friedman, N., M. Linial, I. Nachman, and D. Pe'er. 2004. Using Bayesian Networks to Analyze Expression Data. Journal of Computational Biology 7:601-620.
214.   Hecker, M., S. Lambeck, S. Toepfer, E. van Someren, and R. Guthke. 2009. Gene regulatory network inference: Data integration in dynamic models--A review. Biosystems 96:86-103.
215.   Doucet, P., and P. B. Sloep. 1992. Mathematical Modeling in the Life Sciences, Ellis Horwood Limited, Chichester, Great Britain.
216.   Nissen, S. E., J. C. Tardif, S. J. Nicholls, J. H. Revkin, C. L. Shear, W. T. Duggan, W. Ruzyllo, W. B. Bachinsky, G. P. Lasala, and E. M. Tuzcu. 2007. Effect of torcetrapib on the progression of coronary atherosclerosis. N Engl J Med 356:1304-1316.
217.   Barter, P. J., M. Caulfield, M. Eriksson, S. M. Grundy, J. J. P. Kastelein, M. Komajda, J. Lopez-Sendon, L. Mosca, J. C. Tardif, D. D. Waters, C. L. Shear, J. H. Revkin, K. A. Buhr, M. R. Fisher, A. R. Tall, and B. Brewer. 2007. Effects of torcetrapib in patients at high risk for coronary events. N Engl J Med 357:2109-2122.
218.   Otvos, J. D., E. J. Jeyarajah, and D. W. Bennett. 1991. Quantification of plasma lipoproteins by proton nuclear magnetic resonance spectroscopy. Clin Chem 37:377-386.
219.   Park, I., K. J. Paeng, Y. Yoon, J. H. Song, and M. H. Moon. 2002. Separation and selective detection of lipoprotein particles of patients with coronary artery disease by frit-inlet asymmetrical flow field-flow fractionation. J Chromatogr. B Analyt. Technol. Biomed. Life Sci. 780:415-422.
220.   Chung, B. H., J. P. Segrest, J. T. Cone, J. Pfau, J. C. Geer, and L. A. Duncan. 1981. High resolution plasma lipoprotein cholesterol profiles by a rapid, high volume semi-automated method. J Lipid Res 22:1003-1014.
221.   Mora, S., J. D. Otvos, N. Rifai, R. S. Rosenson, J. E. Buring, and P. M. Ridker. 2009. Lipoprotein Particle Profiles by Nuclear Magnetic Resonance Compared With Standard Lipids and Apolipoproteins in Predicting Incident Cardiovascular Disease in Women. Circulation 119:931-939.
222.   Ashyraliyev, M., Y. Fomekong-Nanfack, J. A. Kaandorp, and J. G. Blom. 2009. Systems biology: Parameter estimation for biochemical models. FEBS Journal 276:886-902.
223.   Press, W. H., S. A. Teukolsky, W. T. Vetterling, and B. P. Flannery. 2007. Numerical Recipies 3rd Ediation: The Art of Scientific Computing, Cambridge University Press, Cambridge, Great Britain.
224.   Nocedal, J., and S. J. Wright. 2000. Numerical Optimization, Springer, New York, N.Y., U.S.A.
225.   Marquardt, D. W. 1963. An Algorithm for Least-Squares Estimation of Nonlinear Parameters. SIAM J Appl Math 11:431-441.
226.   Beck, K. 2002. Test Driven Development: By Example, Addison-Wesley Professional
227.   Lanzén, A., and T. Oinn. 2008. The Taverna Interaction Service: Enabling manual interaction in workflows. Bioinformatics 24:1118-1120.
228.   de Graaf, A. A., A. P. Freidig, B. De Roos, N. Jamsheedi, M. Heinemann, J. A. C. Rullmann, K. Hall, M. Adiels, and B. van Ommen. 2009. Nutritional systems biology modeling: from molecular mechanisms to physiology. PLoS Comput Biol 5:e1000554.
229.   Pappalardo, F., S. Musumeci, and S. Motta. 2008. Modeling immune system control of atherogenesis. Bioinformatics 24:1715-1721.
230.   Davidson, M. H., and P. P. Toth. 2004. Comparative effects of lipid-lowering therapies. Progr Cardiovasc Dis 47:73-104.
231.   August, E., K. H. Parker, and M. Barahona. 2007. A dynamical model of lipoprotein metabolism. Bull. Math. Biol 69:1233-1254.
232.   Tindall, M. J., J. A. Wattis, B. J. O'Malley, L. Pickersgill, and K. G. Jackson. 2009. A continuum receptor model of hepatic lipoprotein metabolism. J Theor. Biol 257:371-384.
233.   Pearson, T., J. A. Wattis, B. O'Malley, L. Pickersgill, H. Blackburn, K. G. Jackson, and H. M. Byrne. 2009. Mathematical modelling of competitive LDL/VLDL binding and uptake by hepatocytes. J Math. Biol 58:845-880.
234.   van de Pas, N. C., A. E. Soffers, A. P. Freidig, O. B. van, R. A. Woutersen, I. M. Rietjens, and A. A. de Graaf. 2010. Systematic construction of a conceptual minimal model of plasma cholesterol levels based on knockout mouse phenotypes. Biochim Biophys Acta 1801:646-654.
235.   van de Pas, N. C. A., R. A. Woutersen, B. van Ommen, I. M. C. M. Rietjens, and A. A. de Graaf. 2010. A physiologically-based kinetic model for the prediction of plasma cholesterol concentrations in the mouse. BBA-mol cell biol l 1811:333-342.
236.   Eppig, J. T., J. A. Blake, C. J. Bult, J. A. Kadin, and J. E. Richardson. 2007. The mouse genome database (MGD): new features facilitating a model system. Nucleic Acids Research 35:D630-D637.
237.   van Schalkwijk, D. B., B. van Ommen, A. A. Freidig, J. van der Greef, and A. A. de Graaf. 2010. MS547 Introducing the VLDL performance diagnostic. Atheroscl suppl 11:220.
238.   Bloomgarden, Z. T. 2010. 2009 World Congress on the Insulin Resistance Syndrome: Cardiovascular Disease Concepts. Diab Care 33:e85-e89.
239.   Suna, T., A. Salminen, P. Soininen, R. Laatikainen, P. Ingman, S. Makela, M. J. Savolainen, M. L. Hannuksela, M. Jauhiainen, M. R. Taskinen, K. Kaski, and M. la-Korpela. 2007. 1H NMR metabonomics of plasma lipoprotein subclasses: elucidation of metabolic clustering by self-organising maps. NMR Biomed. 20:658-672.
240.   Kumpula, L. S., S. M. Makela, V. P. Makinen, A. Karjalainen, J. M. Liinamaa, K. Kaski, M. J. Savolainen, M. L. Hannuksela, and M. la-Korpela. 2010. Characterization of metabolic interrelationships and in silico phenotyping of lipoprotein particles using self-organizing maps. J Lipid Res 51:431-439.
241.   van Schalkwijk, D. B., K. van Bochove, L. D. Parnell, C. Q. Lai, J. M. Ordovas, A. A. de Graaf, B. van Ommen, and D. K. Arnett. 2010. MS327 Clustering by plasma lipoprotein profile reveals large fenofibrate responder subgroup. Atheroscl suppl 11:175.
242.   Stone, N. J. 2006. Successful control of dyslipidemia in patients with metabolic syndrome: focus on lifestyle changes. Clinical Cornerstone 8:S15-20.
243.   Bakker, S. 2006. Bereik en behoud je ideale gewicht!, De Zonnestraal
244.   Covey, S. R. 1989. The Seven Habits of Highly Effective People, Franklin Covey
245.   MacIntyre, A. 2009. Dependent Rational Animals, Gerald Duckworth & Co Ltd
246.   Aquinas, T. 2008. Summa Theologica, New Advent
247.   Aristoteles. 2005. Ethica Nicomachea, Project Gutenberg
248.   Aquinas, T. 1999. Questiones Disputatae de Virtutibus. R. McInerny, and J. Kenny, editors. St. Augustine's Press, South Bend, Indiana.
249.   Havard, A. 2007. Virtuous Leadership, Scepter
250.   MacIntyre, A. 2007. After Virtue, University of Notre Dame Press
251.   Kinneging, A. 2006. Geografie van goed en kwaad, Spectrum

Back to PhD thesis

No comments: