Semianar Write Up For Rachael

You might also like

Download as docx, pdf, or txt
Download as docx, pdf, or txt
You are on page 1of 30

CHAPTER ONE

INTRODUCTION

Precision nutrition is a cutting-edge approach to optimizing health and well-being by tailoring

dietary recommendations to individual needs. It recognizes that each person is unique, with

distinct physiological, genetic, and lifestyle factors that influence their nutritional requirements.

This personalized approach aims to navigate the intricate landscape of individual dietary needs,

acknowledging that a one-size-fits-all approach to nutrition may not be effective for everyone.

Fiamoncini J, Rundle M, (2018)

In the pursuit of precision nutrition, various factors are taken into account, such as genetics,

metabolism, body composition, medical history, and lifestyle choices. Advances in technology,

including genetic testing and other biomarker assessments, play a crucial role in gathering

relevant information to create highly personalized nutrition plans.

The fundamental principle of precision nutrition is to move beyond generic dietary guidelines

and instead focus on tailoring nutritional advice to address the specific needs and goals of each

individual. Lopez-Miranda J, Williams C, Lairon D. (2007 This approach recognizes that what

works for one person may not work for another, even if they share similar demographics.

Precision nutrition involves the collaboration of nutritionists, healthcare professionals, and

individuals themselves to gather comprehensive data and design a personalized roadmap for

optimal nutrition. This may include identifying specific nutrient deficiencies, understanding

individual responses to certain foods, and considering lifestyle factors that impact dietary

choices.
The ultimate goal of precision nutrition is to enhance overall health, support weight

management, optimize athletic performance, and mitigate the risk of chronic diseases. Ordovas

JM, Corella D. (2004) By embracing this individualized approach, individuals can make

informed choices about their nutrition, leading to more sustainable and effective results.

Dietary information is also collected independent of genetics, as a feature in its own right.

Information on diet is particularly important in precision nutrition approaches to bodyweight

management Zeevi D, Korem T, Zmora N, et al. (2015). In some cases, not only dietary features

in the long term but also the content of an individual meal and the timing features of the meal

(e.g., timing of the meal, time elapsed since the previous meal, etc.) are required to be known.

This is the case in research investigating postprandial meal responses, where the composition of

an individual meal in relation to its postprandial effect is relevant to know. Meal timing features

are relevant due to their impact on health. Metabolomics is an increasingly popular field that

quantifies the presence of small molecules in a sample with high accuracy using sophisticated

techniques such as nuclear magnetic resonance and mass spectrometry Rodgers GP, Collins FS.

(2020). As the field of metabolomics develops further and these techniques become more

frequently used, metabolomics will have a role to play in precision nutrition such as by

investigating how different individuals metabolize foods and by establishing phenotypes.

However, the measurement of clinically relevant biochemical parameters measured with

traditional methods (i.e., not assessed from a metabolomics perspective) is currently more

commonly seen and represents features in the group of clinical biochemical parameters. Included

here are common clinical measures such as blood-sugar, hormonal levels, blood counts, and

other parameters deemed to be relevant for a given precision nutrition intervention. other

precision nutrition features are the microbiome, due to its emerging role in health and
relationship with nutritional intake Ordovas JM, Ferguson LR, Tai ES, Mathers JC (2018);

activity parameters (PA amount and intensity, sedentary behavior, and energy expenditure (EE)),

due to their established interaction with health and disease; anthropometric features, such as

height, weight, body mass index (BMI), etc.; and personal features, which includes information

about individuals that can have an impact on model outcome such age, medical information and

disease status, medication use, socioeconomic status, stress, and sleep.

Nutrition is a fundamental pillar of health, and diet is the modifiable factor that exerts the

greatest impact on human health and wellbeing (1). Dietary recommendations have traditionally

been based on a one-size-fits-all approach which assumes that individual nutritional

requirements and responses mimic the average response observed in study populations (2). The

advancement of personalized nutrition or precision nutrition (precision nutrition) strategies has

improved our understanding of how factors such as genetic, microbiome, and metabolic

signatures, may predict whether what we eat supports or harms our health and to what degree

Berry SE, Valdes AM, et al. (2020).

Studies in the field of nutritional genomics have unveiled associations between genetic factors

and metabolic responses to food, nutrient requirements, dietary preferences, and disease

outcomes. Advances in this and other areas of precision nutrition have added new dimensions

that help explain the variability in responses observed in otherwise well-controlled trials of diet

and nutrients. In particular, promising research results support the predictive potential of

assessments of the gut microbiome and metabolome—among other factors—and showcase the

individual but malleable qualities of our biology Perez-Martinez P, Phillips. (2013).

As we bring new perspectives to the multiple dimensions of food and health, we are also

overcoming some of the barriers created by previous reductionistic thinking about nutrition. It is
in this context that precision nutrition is driving the scientific journey toward a more

personalized, predictive, and integrative systems approach to understanding how nature and

nurture interact to shape our health and wellbeing.

Precision nutrition can be considered as occurring at three levels: (1) conventional nutrition

based on general guidelines for population groups by age, gender and social determinants; (2)

individualized nutrition that adds phenotypic information about the person's current nutritional

status (e.g. anthropometry, biochemical and metabolic analysis, physical activity, among others),

and (3) genotype-directed nutrition based on rare or common gene variation Gibney MJ, Walsh

MC (2013). The ultimate goal is to integrate such sources of information to ensure that health-

care professionals, including dietitians, physicians, pharmacists and genetic counselors, know

sufficient concepts about nutrigenetics and nutrigenomics to decide on the most appropriate level

of care to achieve a precision nutrition which integrates phenotypical and genotypical issues as

well as social, environmental and metabolic factors Juma S, Imrham V, Vijayagopal P, Prasad C

(2014).

Precision nutrition is founded upon the concept of biological variability between individuals in

response to nutrition Ordovas JM, Ferguson LR, Tai ES, Mathers JC (2018). Thus, if the

variables responsible for causing this variation and their effect on a desired outcome variable can

be known, the outcome variable can be predicted, and this can be translated into nutrition advice.

What, then, are these variables? The answer to this question depends on the desired outcome

variable. There is no set of fixed variables that will provide any given output. Instead, features

thought to be of importance to predicting the outcome are selected on a per situation basis. In

some cases, this can reach to large numbers of individual features. however, they can be

separated into groups, here referred to as precision nutrition elements. one common precision
nutrition element is genetics. genetics is understood as a reason for many obvious examples of

variation, such as eye color and hair color, and this is extended to response to nutrition. indeed,

in some circumstances such as pku, genetics is an extremely relevant feature for precision

nutrition approaches. there is also some known relationships between genetics and weight

management, lactose (as in the case of lactose intolerance), metabolic syndrome , and more.

however, unlike with eye and hair color, what has become clear is that genetics can rarely

explain nutritional response entirely. in some cases, the genetic contribution is virtually absent,

as berry et al. witnessed when predicting postprandial triglycerides. another relevant factor is not

only genes alone but also their interaction with nutritional intake, termed nutrigenomics. genetic

variation impacts metabolism of dietary components, but also dietary components regulate gene

expression and signaling. failing to account for this interaction will naturally lead to

compromised accuracy of precision nutrition models, meaning dietary information is often

collected in precision nutrition approaches. gene-diet interactions for various chronic conditions

are known and as more continue to be discovered, precision nutrition approaches considering

nutrigenomics can be improved Zeevi D, Korem T, Zmora N, et al. (2015).


CHAPTER TWO

LITERATURE REVIEW

2.1 OVERVIEW OF PRECISION NUTRITION FRAMEWORKS

Suboptimal diet and nutritional imbalance are well established as contributors to the global

burden of non-communicable diseases with a high incidence worldwide (Roth et al., 2018).

Globally, in 2017, an estimated 11 million deaths were attributable to dietary risk factors, with

more than half of these deaths associated with high sodium intake (Afshin et al., 2019). National

dietary guidelines designed to influence consumers’ nutritional behavior and dietary patterns

have had minimal impact (Kalmpourtzidou, Eilander, & Talsma, 2020). Thus, health targets, as

well as environmental targets, highlight the need for integrated population efforts to prioritize

food-based guidelines that shift diets towards high intake of whole grains, fruit and vegetables,

nuts and seeds and legumes (Springmann et al., 2020). However, the “one-size-fits-all” nature of

national and international dietary guidelines does not account for the diverse biological and

sociocultural factors that drive human conduct (Herforth et al., 2019). Human variability is wide-

ranging, and can include genetic, phenotypic, and physiological determinants, medical history,

and lifestyle practices, such as dietary habits and physical activity, as well as sociocultural and

socioeconomic factors, such as the food environment, gastronomy, and educational attainment

(Ordovas, Ferguson, Tai, & Mathers, 2018). This range of potential dietary influences has led to

the emergence of personalized and precision nutrition investigations to improve dietary patterns

(Ordovas et al., 2018), which are now central pillars of many national and international nutrition

research priorities and position statements (Ferguson et al., 2016; Kohlmeier et al., 2016).
The connections between nutrition and wellbeing are numerous and wide-ranging, concerning

both health maintenance as well as disease prevention and management. To operationalize

individualized interventions to improve population and planetary health, the measurement and

scoring of dietary intake is required (Martínez-González et al., 2021). Nutritional status

assessment has been routinely performed to identify and treat undernutrition, however efforts are

now increasingly focused on quantifying the contributing factors to overnutrition and obesity

(Martínez-González et al., 2021). Indeed, the concept of personalized nutrition has been a long-

standing endeavor, which was first alluded to by the ancient Greeks, including Hippocrates

(“your food is the base of your health), and Galen ("personal attitudes and unique responses to

food”), and has since evolved to include nutriomics and the development of global tools to

quantify and categorize individual dietary intakes (Ordovas et al., 2018).

The ultimate goal of personalized and precision nutrition is to preserve or ameliorate health and

wellbeing using dietary interventions, products or services that leveraging human variability

(Ferguson et al., 2016; Ordovas et al., 2018). However, there is no international consensus on the

definition of these terms, and terminology varies depending on the country, health field and

scope of the research question (Bush et al., 2020). For example, the terms precision public health

and precision health have been coined to consider the needs of digital health interventions and

data-driven public health systems to prevent non-communicable diseases (Canfell et al., 2022),

as well as the social determinants of health inequity (Olstad & McIntyre, 2019) when designing

precision nutrition approaches. For the purpose of this review, personalized nutrition will be

defined as an approach in which genetic, metagenomic, physiological, phenotypic, nutritional,

and other relevant information are used to design tailored nutritional advice and support for each

individual (Jinnette et al., 2021). In turn, the overarching term of precision nutrition is defined as
a methodology to integrate genetic, metabolic and environmental information at scale, which can

utilize high-throughput metabolomics, metagenomic and epigenetic approaches (Ordovas et al.,

2018).

2.2 SYSTEMS BIOLOGY APPROACH

Systems biology has been responsible for some of the most important developments in the

science of human health and environmental sustainability. It is a holistic approach to deciphering

the complexity of biological systems that starts from the understanding that the networks that

form the whole of living organisms are more than the sum of their parts. It is collaborative,

integrating many scientific disciplines – biology, computer science, engineering, bioinformatics,

physics and others – to predict how these systems change over time and under varying

conditions, and to develop solutions to the world’s most pressing health and environmental

issues (Betts, Gonzalez, 2016).

This ability to design predictive, multiscale models enables our scientists to discover new

biomarkers for disease, stratify patients based on unique genetic profiles, and target drugs and

other treatments. Systems biology, ultimately, creates the potential for entirely new kinds of

exploration, and drives constant innovation in biology-based technology and computation

(McMahon et a; 2014).

Because systems biology requires constant attention to a very complex, very human social

experiment, ISB fosters the kind of financial, social and psychological environment in which the

world’s best scientists, technologists, engineers and mathematicians can collaborate and do their

best work.
A fundamental tenet of systems biology is that solving challenging biological problems always

requires the development of new technologies in order to explore new dimensions of data space

(Valleé Marcotte, 2016). New data types require novel analytical tools. This virtuous cycle of

biology driving technology driving computation can exist only in a cross-disciplinary

environment where biologists, chemists, computer scientists, engineers, mathematicians,

physicists, physicians and others can come together in teams to tackle grand challenges. This is

ISB. And this describes what we call the “innovation engine” (depicted below) that drives our

ability to develop intellectual property, which we share through open-access platforms or by

spinning out companies.

Fig 2.1: System Biology Approach

2.2.1 CROSS-DISCIPLINARY TEAMS

In describing systems biology and the distinguishing characteristics of ISB’s approach, we

always emphasize how our lab groups are intentionally and necessarily cross-disciplinary. One
of our labs, for example, includes molecular biologists, microbiologists, geneticists, engineers,

oceanographers, and even an astrophysicist (Ouellette, 2014). The complexity of biology in this

age of “big data” requires diverse teams in order to tackle such vast amounts of data and to make

sense of it all. New technologies that crunch data faster and more efficiently also permit

researchers to re-analyze existing datasets, a process which often reveals undiscovered

information. Complementary skills empower any of our groups of researchers to better

understand biological or environmental challenges from different perspectives and to arrive at

shareable insights more quickly. Our interdisciplinary teams have contributed notable advances

to everything from ocean acidification to neurodegenerative diseases and tuberculosis to multiple

cancers.

2.2.2 NETWORK OF NETWORKS

Network of NetworksWith networks, we can organize and integrate information at different

levels. Social networks have transformed communications in the 21st Century, democratizing

our platforms for communications. At ISB we are also concerned with networks (Rudkowska,

2015). One of the tenets of systems biology we often refer to is the “Network of Networks.” On a

biological level, our bodies are made up of many networks that are integrated at and

communicating on multiple scales. From our genome to the molecules and cells that makeup the

organs in our bodies all the way out to ourselves in our world: we are fundamentally a network

of networks. Systems biology looks at these networks across scales to integrate behaviors at

different levels, to formulate hypotheses for biological function and to provide spatial and

temporal insights into dynamical biological changes. It is not enough to understand only one part

of a system when studying the complexity of biology. Therefore the framework of the “Network

of Networks” provides meaningful insight into understanding how systems biology’s approach is
different, more integrated and more capable of analyzing and predicting state transitions in

biological systems.

Fig 2.2: Network of Networks

2.2.3 MULTISCALE MODELING

Whether we explicitly recognize it or not, multiscale phenomena are part of our daily lives. We

organize our time in days, months and years, as a result of the multiscale dynamics of the solar

system. Our society is organized in a hierarchical structure, from towns to states, countries and

continents. The human body is a complex machine, with many little parts that work by

themselves or with other parts to perform specific functions (Tremblay et al, 2015). Organelles

inside each cell in our bodies interact with one another to maintain a healthy functioning cell that

moves, differentiates and dies. These subcellular organelles and their processes govern each

cell’s signalling mechanism to interact with its neighboring cells, and form multi-cellular

systems called tissues (e.g. epithelial tissue, muscular tissue). Two or more types of tissues work
together to form an organ that performs a specific task (e.g. mouth, stomach, liver). Two or more

organs work together to form organ systems, such as the digestive system and the nervous

system, that perform more complex tasks. All these organ systems interact with each other to

enable a healthy functioning organism. Traditional approaches to modeling real world systems

focus on a single scale that imparts a limited understanding of the system. The pace at which

biotechnology has grown has enabled us to collect large volumes of data capturing behavior at

multiple scales of a biological system. Genetic as well as environmental alterations to the DNA,

expression levels of RNAs, expression of genes and synthesis of proteins – all this is measurable

now within a matter of days at a rapidly declining cost. So, it is really up to scientists and data

analysts to make use of this variety of data types and build integrative models that enable a

comprehensive understanding of the system under study. Multiscale models do just that. By

integrating models at different scales and allowing flow of information between them, multiscale

models describe a system in its entirety, and as such, are intrinsic to the principles of systems

biology (Palatini et al, 2009).

2.3 NUTRIGENOMICS AND NUTRIGENETICS

With the completion of human genome sequencing and entering the-Omics area, the new term

“Nutritional Genomics” tends to replace the former “nutrient-gene interactions” (Rasinpera et al,

2004). It has been demonstrated that numerous genetic polymorphisms can influence protein

structure function. The Nutritional genomic area includes two parts: first Nutrigenomics that is

the study of interaction between dietary components and the genome, and the regulating changes

in proteins and other metabolism; second Nutrigenetics that identify the response to dietary

components with regard to genetic differences.


Nutrients are as environmental factors can interact with genetic material. It has been clearly

demonstrated that DNA metabolism and repair depend on a wide range of dietary factors that act

as cofactors or substrates in metabolic pathway, but much less is known about the impact of

cofactors and/or micronutrients deficiency or excess on the fidelity of DNA replication and

repair (3). Although the nutrients can influence the development of a particular phenotype, the

response to a specific nutrient that determined by the individual genotype has also to be

considered (Fig. 2.3).

Fig 2.3: Nutrigenetics and nutrigenomics

2.4 METABOLOMICS AND OTHER OMICS TECHNOLOGIES


Biomedical researchers are increasingly relying on metabolomics and other omics data types to

study and evaluate disease mechanisms and phenotypes. Omics data include, but are not limited

to, measurements of the metabolome, proteome, transcriptome, genome, microbiome, and

exposome. These measurements include the presence (binary), quantification (abundance),

and/or characterization (chemical or biological function) of molecules or entities, such as

metabolites, proteins, microbial taxa, genes, or transcripts. For simplicity, we refer to these

molecules or entities as “analytes” throughout this work. Multi-omics data may also include

descriptors from multiple timepoints in one or more omic modalities, phenotype information

such as case/control labels, and relevant clinical variables such as age and sex. Collectively,

these data provide holistic insights into disease-driven biological pathway dysregulation, which

in turn provides preliminary evidence to drive the identification of new targets or intervention

strategies (Giner et al, 2001). While the utility of assessing multi-omics data is clear, the

integration of metabolomic data with other omic data poses significant computational challenges

that range from the need for developing statistical methods that are appropriately adapted to

multi-omics integration, to the need for providing comprehensive open-source resources that

provide validated relationships between omics types, biological pathways, and diseases. Multi-

omics integration typically follows the general workflow depicted in Figure 2.4.
Fig 2.4: The metabolome in the context of other omics data types and broad approaches

for their integration.

Ongoing efforts to support the integrative analysis of multi-omics data include the development

of statistical methods, computational tools, and pipelines/workflows. Statistical and

computational methods comprise novel metrics or novel applications of metrics that describe the

relationship between multiple omic data. These include univariate and multivariate analyses,

correlation networks, and traditional machine learning and deep learning techniques. A tool is an

implementation of a method with proven utility, many of which are designed to be user-friendly

software. Tools are often downloadable as an executable file or stored in a public code

repository. A series of methods and tools can be combined into workflows to perform an

analysis. Supported analyses could span the conversion of raw data (e.g., direct output from
instruments or a matrix of un-normalized metabolite and gene levels) to interpretable data that

explain a biological system under study. Workflows are particularly useful for conducting

repetitive tasks, and typically provide default parameters that are globally applicable, hence

making them user-friendly. Examples of open-source and user-friendly workflows include

MetaboAnalyst, XCMS, mixomics, miodin, and many others reviewed elsewhere (Goni et al,

2015). These workflows are particularly useful for end users that may not have a strong data

analysis or computational background and are invaluable for outputting reproducible results.

Ideally, methods, tools, and workflows provide up-to-date, publicly available datasets that can be

used as an input for testing and benchmarking, allowing users to readily evaluate the utility of

these workflows for their own purpose.

Many high-level analytical concepts are employed across workflows and data modalities.

Understanding the general steps taken by many workflows is crucial to compare the different

resources available for performing these tasks. Starting with raw data, prior to analysis, the data

quality of each omic data must be carefully assessed to ensure that measurements are

reproducible. This step typically requires a comparison of analyte measurements across technical

replicates using metrics such as standard deviation or the coefficient of variation. Samples should

also be evaluated, making sure that the overall distribution of analyte measurements is consistent

across samples. We note that identification of potential outliers (analytes or samples) is critical,

as some analytical models for multi-omics analyses (e.g., Principal Components Analysis and

Student’s t-test) could be strongly affected by the presence of outliers (Rukh et al, 2013). Other

preprocessing steps may include normalization to account for differences in experimental effects

such as differences in amounts of starting material and batch effects. Data are then typically

transformed so that they follow a Gaussian or “Normal” distribution, which is commonly used
for statistical analyses. Importantly, as some analyses will not work on missing data, missing

values can be imputed. We note that the imputation method used can affect downstream analysis

results, and thus imputation is still an active area of research (Olsen et al, 2016). Finally, noting

that the range of values may differ between omic modalities, appropriate scaling (e.g., to a

standard deviation of 1, z-scores) within and across omic datasets is critical for ensuring that

each omic modality contributes to the analyses and that the effect of one omic modality does not

dominate all analyses performed. Special scaling considerations should also be taken for time-

series data.

After preprocessing, omics data can be integrated in multiple ways. One can analyze or model

each omic modality separately and then integrate results (a posteriori integration) or one can

integrate data for all omic modalities before any statistical or computational modeling (a priori

integration). Depending on which integration approach is utilized, data may be pretreated

differently. For example, scaling analyte measurements appropriately within each omic modality

is particularly critical when applying a priori integration. Additionally, the sample origin of the

multi-omics datasets dictates which integration approach can be used. For example, a priori

integration requires the measurements to be collected in the same biospecimens (tissue, blood,

etc.) or individuals to allow measurements to be matched to the same sample, while a posteriori

integration does not. When the analysis is performed on the same individual but different

biospecimens, e.g., genomic data from blood and metabolomic data from urine, we note that it is

not possible to evaluate direct relationships between genes and metabolites and how they may

relate to phenotype. However, it is feasible to evaluate whether one omic modality (e.g.,

metabolites) could act as a biomarker for what is occurring at another level (e.g., genome), or
one omic modality can be used to corroborate findings (e.g., biological pathways) uncovered in

another omic modality (Qi et al, 2012).

Recognizing that other reviews provide a comprehensive list of available methods, software,

and/or workflows (Brunkwall et al, 2016), we instead focus on providing concepts and

considerations that are useful when choosing a method or tool appropriate for one’s desired data

types and analyses. As such, we discuss existing guidelines in data curation and tool

development and describe the building blocks that are used for developing computational tools

and workflows, namely unsupervised clustering approaches to assess data quality or separation

by sample type, approaches for modeling co-regulations between multiple omic modalities,

approaches for identifying multi-omics factors associated with a phenotype (supervised

methods), and methods that provide a biological, chemical, and/or disease context to multi-omics

data (e.g., pathway analysis). We provide a summary illustration of these approaches in Figure

2.5.
Fig 2.5: Analysis techniques on a dataset with n samples and d analytes in two classes.

Further, the review focuses on open-source resources, and we reference example research

projects that make use of these resources in the context of metabolomic and other omic data,

denoting whether and to what extent the data used is publicly available.

2.5 INDIVIDUALIZED DIETARY ASSESSMENT

Dietary assessment is an evaluation of food and nutrient intake and dietary pattern of an

individual or individuals in the household or population group over time. It is one of the four

approaches in nutrition assessment to evaluating the nutritional status of individuals

comprehensively.

The other three are anthropometrics, biochemical parameters and clinical examination (Gibson,

2005). Dietary assessment methods are usually categorized according to the nature of the method

used as shown in Figure 1. Indirect methods utilize secondary data for assessing diets, while

direct methods collect primary dietary data from individuals. Prospective and retrospective

methods refer to the time food consumption is recorded. Prospective methods involve recording

the diet when the foods are being consumed; retrospective methods are based on a recall of food

intake that have already been consumed.

Furthermore, dietary assessment can be qualitative (types of food consumed) or quantitative

(types and amounts of food consumed). The latter allows estimation of an individual’s food,

energy and nutrient intakes. Selection of an appropriate method for dietary assessment depends

on the purposes of the study, which may be to measure food consumption, nutrient intake or

eating habits.

2.5.1 INDIRECT METHODS


Indirect methods use secondary information (e.g. food supply, agricultural statistics, food

expenditure) to estimate food available for consumption at the national and household levels.

Firstly, we will look at the Food Balance Sheet (FBS) 4, which provides food consumption

information at national level: food consumption is estimated from the point of view of food

supply.

Secondly, we will discuss about the Household Consumption and Expenditure Survey (HCES),

which provide food consumption information at household level: food consumption is estimated

from the point of view of food demand. Neither of the two methods directly obtain primary

dietary data from individuals to evaluate dietary intake or food consumption on an individual

basis. Indirect methods are useful for identifying trends in food availability and consumption

across different geographical regions and time (Chen et al, 2016).

2.5.2 DIRECT METHODS

Direct methods using individual-based dietary assessment can be classified into two groups, as

described in Figure 2.6. Retrospective methods measure food intake from the past. These

methods include 24-hour recall, food frequency questionnaires (FFQ), and dietary history.

Prospective methods assess current food intake.

These methods include food records and the duplicate meal method. Estimates obtained from

direct methods are used to identify trends in food consumption, food and nutrient intakes, eating

patterns, and to evaluate diet–disease associations. Information provided by such methods can

also be used to calculate relevant food-based indicators for monitoring and evaluation purposes.

This section will describe dietary assessment methods used in individualbased surveys, and
analyse their application, focusing mainly on low resource settings, and will also cover the issues

of precision and accuracy

Fig 2.6: Overview of dietary assessment methods to estimate food and nutrient

consumption at national, household and individual level


CHAPTER THREE

CONCLUSION AND RECOMMENDATION

CONCLUSION

In conclusion, precision nutrition represents a groundbreaking approach to navigating the

complex landscape of individual dietary needs. By acknowledging and embracing the unique

biological, genetic, and lifestyle factors that influence how individuals respond to food, precision

nutrition offers a more tailored and effective way to optimize health outcomes. The emphasis on

personalized assessments, data-driven insights, and behavioral considerations underscores the

importance of treating each person as a distinct entity with specific nutritional requirements.

This approach is not only about addressing existing health concerns but also preventing future

issues and promoting overall well-being. The adaptability and flexibility inherent in precision

nutrition recognize that dietary needs are dynamic and may change over time, requiring

continuous assessment and adjustment.

The collaborative nature of precision nutrition, involving healthcare professionals and

individuals working together, highlights the importance of a team approach in developing and

implementing personalized dietary plans. This collaboration ensures a comprehensive

understanding of an individual's health and facilitates the creation of sustainable nutritional

strategies.
As precision nutrition continues to evolve, fueled by ongoing research and technological

advancements, it holds the promise of refining our understanding of individualized dietary

requirements further. This dynamic field has the potential to revolutionize how we approach

nutrition, moving away from one-size-fits-all recommendations to strategies that truly align with

the unique characteristics of each person.

In summary, precision nutrition is a transformative paradigm that recognizes the intricacies of

individual dietary needs, providing a pathway to optimize health, prevent diseases, and foster

well-being through personalized and evidence-based nutritional interventions.

RECOMMENDATION

 Engage with healthcare professionals, such as registered dietitians or nutritionists, who

specialize in precision nutrition. These experts can conduct thorough assessments,

analyze genetic and biomarker data, and provide personalized dietary recommendations

based on your unique needs and goals.

 Take an active role in understanding your own health and nutritional needs. Stay

informed about the latest research and advancements in precision nutrition. This

knowledge empowers you to make informed decisions and actively participate in the

development of your personalized nutrition plan.

 Regularly monitor your progress and be open to adjusting your nutrition plan as needed.

Lifestyle, health status, and dietary needs may change over time, and ongoing assessment

allows for timely adjustments to optimize the effectiveness of your precision nutrition

approach
 Understand that achieving optimal results with precision nutrition takes time. Be patient

and consistent in implementing the recommended dietary strategies. Consistency is key to

seeing sustainable improvements in health and well-being.

REFERENCES

Ordovas JM, Corella D. (2004) Nutritional genomics. Annu Rev Genomics Hum Genet. (2004)

5:71–118. doi: 10.1146/annurev.genom.5.061903.180008

U.S. Department of Agriculture and U.S. Department of Health and Human Services. Dietary

Guidelines for Americans, 2020-2025. (2020). Available online at: www.DietaryGuidelines.gov

(accessed June 9, 2022).

Berry SE, Valdes AM, et al. (2020) Human postprandial responses to food and potential for

precision nutrition. Nat Med. (2020) 26:264–73. doi: 10.1038/s41591-020-0934-0

Lopez-Miranda J, Williams C, Lairon D. (2007) Dietary, physiological, genetic and pathological

influences on postprandial lipid metabolism. Br J Nutr. (2007) 98:458–73. doi:

10.1017/S000711450774268X

Ordovas JM, Ferguson LR, Tai ES, Mathers JC (2018). Personalised nutrition and health. BMJ.

(2018) 13:361:bmj.k2173. doi: 10.1136/bmj.k2173


Berciano S, Lai CQ, Herranz J, et al. (2017) Behavior related genes, dietary preferences and

anthropometric traits. FASEB J. (2017) 31:299.1. doi: 10.1096/fasebj.31.1_supplement.299.1

Perez-Martinez P, Phillips. (2013) Nutrigenetics, metabolic syndrome risk and personalized

nutrition. Curr Vasc Pharmacol. (2013) 11:946–53. doi: 10.2174/157016111106140128120911

Rodgers GP, Collins FS. (2020) Precision nutrition—the answer to “What to Eat to Stay

Healthy”. JAMA. (2020) 324:735–6. doi: 10.1001/jama.2020.13601

Zeevi D, Korem T, Zmora N, et al. (2015) Personalized nutrition by prediction of glycemic

responses. Cell. (2015) 163:1079–94. doi: 10.1016/j.cell.2015.11.001

Fiamoncini J, Rundle M, (2018) Plasma metabolome analysis identifies distinct human

metabotypes in the postprandial state with different susceptibility to weight loss-mediated

metabolic improvements. FASEB J. (2018) 32:5447–58. doi: 10.1096/fj.201800330R

Gibney MJ, Walsh MC (2013) The future direction of personalised nutrition: my diet, my

phenotype, my genes. Proc Nutr Soc 2013;72:219-225.

Martínez JA (2014) Perspectives on personalized nutrition for obesity. J Nutrigenet

Nutrigenomics 2014;7:188-190.

Juma S, Imrham V, Vijayagopal P, Prasad C (2014) Prescribing personalized nutrition for

cardiovascular health: are we ready? J Nutrigenet Nutrigenomics 2014;7:153-160.

Ahmadi K.R., Andrew T. Opportunism: A panacea for implementation of whole-genome

sequencing studies in nutrigenomics research? Genes Nutr. 2014;9:387. doi: 10.1007/s12263-

014-0387-5.
Allison D.B., Bassaganya-Riera J., Burlingame B., Brown A.W., le Coutre J., Dickson S.L., van

Eden W., Garssen J., Hontecillas R., Khoo C.S.H., et al. Goals in Nutrition Science 2015–2020.

Front. Nutr. 2015;2:1–13. doi: 10.3389/fnut.2015.00026.

Archer E., Pavela G., Lavie C.J. The Inadmissibility of What We Eat in America and NHANES

Dietary Data in Nutrition and Obesity Research and the Scientific Formulation of National

Dietary Guidelines. Mayo Clin. Proc. 2015;90:911–926. doi: 10.1016/j.mayocp.2015.04.009.

Azizi F., Ghanbarian A., Momenan A.A., Hadaegh F., Mirmiran P., Hedayati M., Mehrabi Y.,

Zahedi-Asl S. Tehran Lipid and Glucose Study Group. Prevention of non-communicable disease

in a population in nutrition transition: Tehran Lipid and Glucose Study phase II. Trials.

2009;10:5. doi: 10.1186/1745-6215-10-5.

Betts J.A., Gonzalez J.T. Personalised nutrition: What makes you so special? Nutr. Bull.

2016;41:353–359. doi: 10.1111/nbu.12238.

Brunkwall L., Chen Y., Hindy G., Rukh G., Ericson U., Barroso I., Johansson I., Franks P.W.,

Orho-Melander M., Renstrom F. Sugar-sweetened beverage consumption and genetic

predisposition to obesity in 2 Swedish cohorts. Am. J. Clin. Nutr. 2016;104:809–815. doi:

10.3945/ajcn.115.126052.

Casas-Agustench P., Arnett D.K., Smith C.E., Lai C.-Q., Parnell L.D., Borecki I.B., Frazier-

Wood A.C., Allison M., Chen Y.-D.I., Taylor K.D., et al. Saturated Fat Intake Modulates the

Association between an Obesity Genetic Risk Score and Body Mass Index in Two US

Populations. J. Acad. Nutr. Diet. 2014;114:1954–1966. doi: 10.1016/j.jand.2014.03.014.

Corella D., Coltell O., Mattingley G., Sorlí J.V., Ordovas J.M. Utilizing nutritional genomics to

tailor diets for the prevention of cardiovascular disease: A guide for upcoming studies and
implementations. Expert Rev. Mol. Diagn. 2017;17:495–513. doi:

10.1080/14737159.2017.1311208.

Corella D., Peloso G., Arnett D.K., Demissie S., Cupples L.A., Tucker K., Lai C.-Q., Parnell

L.D., Coltell O., Lee Y.-C., et al. APOA2, Dietary Fat, and Body Mass Index. Arch. Intern. Med.

2009;169:1897. doi: 10.1001/archinternmed.2009.343.

Corella D., Tai E.S., Sorlí J.V., Chew S.K., Coltell O., Sotos-Prieto M., García-Rios A., Estruch

R., Ordovas J.M. Association between the APOA2 promoter polymorphism and body weight in

Mediterranean and Asian populations: Replication of a gene-saturated fat interaction. Int. J.

Obes. (Lond.) 2011;35:666–675. doi: 10.1038/ijo.2010.187.

Cornelis M.C., El-Sohemy A., Campos H. Genetic polymorphism of the adenosine A2A receptor

is associated with habitual caffeine consumption. Am. J. Clin. Nutr. 2007;86:240–244.

Cornelis M.C., El-Sohemy A., Kabagambe E.K., Campos H. Coffee, CYP1A2 Genotype, and

Risk of Myocardial Infarction. JAMA. 2006;295:1135. doi: 10.1001/jama.295.10.1135.

Dhurandhar N.V., Schoeller D., Brown A.W., Heymsfield S.B., Thomas D., Sørensen T.I.A.,

Speakman J.R., Jeansonne M., Allison D.B. Energy balance measurement: When something is

not better than nothing. Int. J. Obes. 2015;39:1109–1113. doi: 10.1038/ijo.2014.199.

DiLella A.G., Huang W.M., Woo S.L. Screening for phenylketonuria mutations by DNA

amplification with the polymerase chain reaction. Lancet. 1988;1:497–499. doi: 10.1016/S0140-

6736(88)91295-0.
Estruch R., Ros E., Salas-Salvadó J., Covas M.-I., Corella D., Arós F., Gómez-Gracia E., Ruiz-

Gutiérrez V., Fiol M., Lapetra J., et al. Primary Prevention of Cardiovascular Disease with a

Mediterranean Diet. N. Engl. J. Med. 2013;368:1279–1290. doi: 10.1056/NEJMoa1200303.

Ferguson L.R., De Caterina R., Görman U., Allayee H., Kohlmeier M., Prasad C., Choi M.S.,

Curi R., de Luis D.A., Gil Á., et al. Guide and Position of the International Society of

Nutrigenetics/Nutrigenomics on Personalised Nutrition: Part 1 - Fields of Precision Nutrition. J.

Nutrigenet. Nutrigenomics. 2016;9:12–27. doi: 10.1159/000445350.

Fung T.T., McCullough M.L., Newby P.K., Manson J.E., Meigs J.B., Rifai N., Willett W.C., Hu

F.B. Diet-quality scores and plasma concentrations of markers of inflammation and endothelial

dysfunction. Am. J. Clin. Nutr. 2005;82:163–173.

Giner V., Poch E., Bragulat E., Oriola J., González D., Coca A., De La Sierra A. Renin-

angiotensin system genetic polymorphisms and salt sensitivity in essential hypertension.

Hypertension. 2000;35:512–517. doi: 10.1161/01.HYP.35.1.512.

Goni L., Cuervo M., Milagro F.I., Martínez J.A. A genetic risk tool for obesity predisposition

assessment and personalized nutrition implementation based on macronutrient intake. Genes

Nutr. 2015;10:1–10. doi: 10.1007/s12263-014-0445-z.

Hebert J.R., Frongillo E.A., Adams S.A., Turner-McGrievy G.M., Hurley T.G., Miller D.R.,

Ockene I.S. Perspective: Randomized Controlled Trials Are Not a Panacea for Diet-Related

Research. Adv. Nutr. An Int. Rev. J. 2016;7:423–432. doi: 10.3945/an.115.011023.

Konstantinidou V., Covas M.-I., Munoz-Aguayo D., Khymenets O., de la Torre R., Saez G.,

Tormos M.d.C., Toledo E., Marti A., Ruiz-Gutierrez V., et al. In vivo nutrigenomic effects of
virgin olive oil polyphenols within the frame of the Mediterranean diet: A randomized controlled

trial. FASEB J. 2010;24:2546–2557. doi: 10.1096/fj.09-148452.

Koochakpoor G., Daneshpour M.S., Mirmiran P., Hosseini S.A., Hosseini-Esfahani F.,

Sedaghatikhayat B., Azizi F. The effect of interaction between Melanocortin-4 receptor

polymorphism and dietary factors on the risk of metabolic syndrome. Nutr. Metab. (Lond.)

2016;13:35. doi: 10.1186/s12986-016-0092-z.

Loos R.J.F., Janssens A.C.J.W. Predicting Polygenic Obesity Using Genetic Information. Cell

Metab. 2017;25:535–543. doi: 10.1016/j.cmet.2017.02.013.

Ludvigsson J.F., Bai J.C., Biagi F., Card T.R., Ciacci C., Ciclitira P.J., Green P.H.R.,

Hadjivassiliou M., Holdoway A., van Heel D.A., et al. Diagnosis and management of adult

coeliac disease: Guidelines from the British Society of Gastroenterology. Gut. 2014;63:1210–

1228. doi: 10.1136/gutjnl-2013-306578.

Martínez-González M.A., Fernández-Jarne E., Serrano-Martínez M., Wright M., Gomez-Gracia

E. Development of a short dietary intake questionnaire for the quantitative estimation of

adherence to a cardioprotective Mediterranean diet. Eur. J. Clin. Nutr. 2004;58:1550–1552. doi:

10.1038/sj.ejcn.1602004.

Martínez-González M.A., García-Arellano A., Toledo E., Salas-Salvadó J., Buil-Cosiales P.,

Corella D., Covas M.I., Schröder H., Arós F., Gómez-Gracia E., et al. A 14-item Mediterranean

diet assessment tool and obesity indexes among high-risk subjects: The PREDIMED trial. PLoS

ONE. 2012;7:e43134. doi: 10.1371/journal.pone.0043134.

You might also like