Open access peer-reviewed chapter

Perspective Chapter: Predictive Genomics

Written By

Jörg Kriegsmann, Sanja Cirovic, Rita Casadonte, Torsten Hansen, Katharina Kriegsmann and Mark Kriegsmann

Submitted: 22 September 2023 Reviewed: 22 September 2023 Published: 03 January 2024

DOI: 10.5772/intechopen.1003246

From the Edited Volume

Electron Microscopes, Spectroscopy and Their Applications

Guillermo Huerta Cuellar

Chapter metrics overview

74 Chapter Downloads

View Full Metrics

Abstract

Predictive genomics can support treatment decisions by giving people the chance to act in time to prevent serious illness. Tests based on single nucleotide polymorphism (SNP) can be analyzed by various methods. Matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry technology detects genetic variants based on their individual mass. Standardized workflow, automation, sensitivity, quick turnaround time, and reliability are the main advantages of the MALDI-TOF use in molecular analysis. Beside pharmacogenetics, SNP variation plays a role in various fields of medicine. In the present article importance of various SNPs for nutrigenetics is presented. Especially, various aspects of fat metabolism, vitamin metabolism, and intolerances were discussed.

Keywords

  • MALDI-TOF (matrix-assisted laser desorption/ionization time-of-flight)
  • SNP (single nucleotide polymorphism)
  • nutrigenetics
  • predictive genomics
  • vitamins
  • intolerances

1. Introduction

Predictive genetics includes various areas of genetics, including nutrigenetics, fitness genetics, and pharmacogenetics. Some authors use the word “lifestyle genetics” because it is different from medical genetics.

Genetic and nongenetic information has to be combined to understand diseases and include this information into personalized preventive medicine.

For the investigation of genetic polymorphisms, mass spectrometry seems to be a very reliable and cost-efficient method compared to next generation sequencing (NGS) technology when investigating not more than 250 SNPs.

We have developed several genetic test panels for various areas of genetic diagnostics and discuss some aspects of nutrigenetics as fat metabolism, vitamins, and intolerances in this report.

Advertisement

2. Mass spectrometry in medicine

Mass spectrometry represents a technology, which is increasingly applied in medical diagnostics. Recently, review articles were analyzed concerning mass spectrometry consisting of microbiological pathogens, diagnosis of diseases, DNA analysis, and small molecules [1].

Matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry is used in everyday routine in clinical diagnostic of microorganism infections. MALDI-TOF technology has many advantages versus traditional techniques, especially fast turnaround time, low amount of hands-on time, and low cost [2]. Direct identification of viruses and bacteria is possible within minutes, allowing the administration of a targeted antimicrobial therapy [3]. Microorganisms were detected by mass spectrometry based on a mass spectrum identifying a characteristic spectrum, which is compared to a large database provided by the manufacturers of the mass spectrometers [4]. Furthermore, the technology may clarify microbial resistance mechanisms [5, 6]. Numerous reports have been published identifying bacteria, fungi [6], and various viruses [7, 8].

Mass spectrometry is able to identify drugs and other metabolites in various body fluids, tissues, and cells [9, 10]. This technique is not only able to identify molecular targets but also their spatial distribution providing a three-dimensional image of the targets. Spatial analysis of drug absorption, distribution, metabolism, and toxicology has been performed using mass spectrometry imaging (MSI) technique [11, 12]. One of the recent developments of MSI is the highly multiplexed immunohistochemistry (IHC) based on MALDI MSI (MALDI-IHC), where up to 30 different antibodies simultaneously can be detected and quantified within a tissue section [13].

Imaging technology was also used in tumor classification providing a tool to identify morphological features of a tissue combined with detection of proteins, glycans, or lipids directly without the limitations and expense of antibodies [14, 15].

The technique of mass spectrometry has been used for the detection of various molecules. The molecular targets for mass spectrometry include proteins [16], peptides [17], lipids [18], glycans [19, 20, 21, 22], and metabolites [23]. Application of mass spectrometry in nucleic acid analysis has been shown in various fields [24, 25, 26].

A new and growing class of medical tests, differing from conventional medical diagnostic tests, are tests in genetics [27, 28], including pharmacogenetics [29, 30].

Advertisement

3. Mass spectrometry in genetics

Predictive genetic tests represent a new and growing field in medicine that differs from conventional medical diagnostic tests. Unlike testing patients with a disease condition, predictive genetics is applied in asymptomatic people to predict the future risk of disease. Early identification of individuals at risk for a specific condition will lead to reduced morbidity and mortality. Unfortunately, predictive genetic tests carry a degree of uncertainty about whether a condition will develop, when it will develop, and its severity [31].

Various genetic tests were developed and integrated into medical diagnostics, especially in predictive medicine [32].

To date, the medical genetic tests offered are mainly for BRCA1/2 (59, 40%), Lynch syndrome (23, 16%), and newborn screening (18, 12%).

3.1 SNP and GWAS

It is well-known that the DNA sequence at each locus may contain nucleotide bases: A, C, G, and T, which can be similar (homozygous) or different (heterozygous) at each DNA strength.

Single-nucleotide polymorphisms (SNPs) are DNA polymorphisms caused by a single-nucleotide substitution mutation. SNPs are caused by mutation and are present one SNP per thousand bases [33].

SNPs may influence various disease conditions and may alter metabolism of various drugs. The difference between SNP and SNV (single nucleotide variation) is that for the first more than 1% of a population has carry a variant nucleotide at a specific position of the DNA. SNPs can be present in coding (exons) or noncoding regions of DNA (introns). SNPs may cause change in the encoded amino acid or not and may be, therefore, of utmost importance or does not have any effect [34]. Although a particular SNP may not cause a disorder, some SNPs are associated with a disease. Improving knowledge may provide useful SNP markers for medical testing and a safer individualized medication to treat the most common disorders [34].

SNPs may help to provide information to prevent diseases or to give the opportunity of personalized medicine to patients.

In pharmacogenetics specific, SNPs can be used for treatment decisions or to choose the appropriate dosage of a drug. This could save time and could prevent adverse drug effects in patients.

Improved knowledge of the meaning of SNPs comes from genome-wide association studies (GWASs). The principle of GWAS is to compare genetics of two or more different groups of individuals [35, 36]. In the last years, the number of GWAS meta-analysis increased to study various traits in different populations [35].

Genetic analysis of SNPs can be done using various body materials as a source of human DNA such as saliva/buccal smears [37], blood samples [27, 38, 39], bone marrow cell lines [40], cytological liquid samples [41], formalin-fixed paraffin-embedded (FFPE) tissue [24, 26, 42, 43, 44].

3.1.1 Mass spectrometry for the detection of SNP (technical considerations)

MALDI-TOF mass spectrometry allows high efficiency in gentying. An efficient analysis of SNPs is offered by the Agena Bioscience iPLEX® procedure. Automatic extraction of DNA can be performed using Chemagic 360 Instrument (Perkin Elmer). DNA from a variety of biospecimen types (blood, saliva, cells collected by cheek brush, and even from FFPE) can be used for genotyping. Extracted DNA is then processed following manufacturer’s instruction for SNP genotyping by Agena Bioscience, described in the multiplex (iPLEX®) assay procedure.

The multiplex (iPLEX®) assay procedure and MassARRAY based on MALDI-TOF mass spectrometry includes several steps [45, 46]. The various steps of this analysis include amplification of targeted DNA sequence by PCR. Then, PCR products are neutralized with shrimp alkaline phosphatase (SAP) for unbound nucleotides. PCR products are then extended by one base. The mass is then measured using a mass spectrometer to produce/calculate a specific mass spectrum of targeted SNPs.

The MassARRAY Analyzer System is built to detect DNA fragments within a mass range of approximately 4500–9000 Da and can easily distinguish between analytes separated by 16 Da. The assay design process is assisted by an online suite of programs that allow for design using “default” settings, as well as “user-defined” settings (more advanced manipulation). Up to two plates of 96 or 384 samples can be genotyped for about 40-plex assay in around 8–12 h, resulting in the generation of more than 30,000 genotypes. Data analysis is performed using MassARRAY Typer Analyzer software from Agena Bioscience. MassARRAY, iPLEX® and SpectroCHIP are registered trademarks of Agena Bioscience, Inc.

3.2 Nutrigenetics

Nutrigenetics attempts to characterize and integrate the relationship between food constituents and gene expression. These approaches for precision nutrition and their relation to disease risk help to identify genetic variants that could modify the effects of dietary intake, affect food metabolism, and influence food preferences [47].

The aim combining genomics and nutrigenomics with clinical data is to get information about genetic variants, which are the basis for personalized nutritional supplementation. The substances of interest for nutritional genetics include lipids, proteins, vitamins, glycose, and iron or calcium [48].

Additionally, in the future, nutrigenetics and nutrigenomics will be combined with data of other omics technologies, such as proteomics and metabolomics, as well as microbiome and data technology [49].

The basic knowledge of emerging nutrigenomics and nutrigenetics can be applied to optimize health, prevention, and treatment of diseases [50]. The increasing number of patients with diabetes and obesity has led focus to these diseases, including genetic risk factors in the last years [51].

Since, these diseases have at least in part a genetic background to explore gene-diet interactions on obesity and diabetes is of utmost interest [52].

Personalized nutrition seems to be necessary because of the substantial variation in the genetic pattern of various human subjects [53].

3.2.1 Fat mass and obesity (FTO) associated gene

FTO gene variants (fat mass and obesity-associated) gene were detected in a GWAS search for genes predisposing for diabetes [36].

This gene is involved in the expression of fat deposition and metabolism-related hormones and genes [54]. For these reasons, investigation of the polymorphism of this gene is included in nearly all specific nutrigenetics and nutrigenomics tests.

Various studies have shown that polymorphisms in this gene lead to a higher body mass index [55, 56].

It has been reported that polymorphisms in the FTO gene are associated with other genes involved in adipogenesis. Furthermore, their impact is not solely dependent on the expression of the polymorphisms themselves [57, 58].

The FTO rs9939609 has been found to relate to the hormone ghrelin, which is associated with digestive behavior [59].

Childhood metabolic syndrome is prevalent around the world and is associated with increased disease risk, especially of cardiovascular diseases, including hypertension and acute coronary syndrome. Some variants of the human FTO gene contribute to the early onset of childhood metabolic syndrome [60].

It must be emphasized that epigenetic influence on the FTO gene is possible as a new approach in the treatment and management of obesity depending on the genetic variant [61].

Figure 1 shows a representative genotyping result of the SNP FTO_ rs9939609 using the MassARRAY System.

Figure 1.

Representative genotyping result of the SNP FTO_ rs9939609 with the MassARRAY system. (A) Spectrum showing a representative multiplex assay of a patient with heterozygous genotype for the SNP FTO_ rs9939609 (AT). A zoomed view in the mass range 6700–6860 shows two peaks indicated with blue dotted lines representing the heterozygous alleles (AT). (B) Representative spectrum of multiplex assay of a patient with homozygous genotype for the SNP FTO_ rs9939609 (AA). A zoomed view in the mass range 6700–6860 shows the detection of one single peak of interest (blue dotted line).

FTO polymorphisms were reviewed in a minireview, which provides thorough inside into these genetic variants [61].

3.2.2 Lipids

Dyslipidemias are known risk factors, which could require precision nutrition designed according to characteristics, such as diet, phenotype, and genotype [62]. Increased intake of triglycerides and cholesterol is associated with an increased risk of metabolic diseases.

One of the best-studied genetic polymorphisms is that of the Apolipoprotein E (ApoE)-gene.

ApoE plays a key role in the transport of cholesterol. ApoE variants are associated with early-onset Alzheimer’s disease. Specifically, ApoE4 isoform may be responsible for alterations in insulin- and lipid metabolism, altered gray matter volume, and impaired cerebrovascular functions [63].

The ApoE2 isoform is generally the most favorable and ApoE4 the least favorable for cardiovascular and neurological health. Under metabolic stress, homozygosity for ApoE2 may result in dysbetalipoproteinemia [64].

It is of special interest that omega-3 fatty acid intake and physical activity may modify the impact of ApoE4 on Alzheimer’s disease and cardiovascular disease risk [65].

Genotyping for ApoE may help develop a targeted approach to disease prevention. Adherence to Mediterranean diet may lower Alzheimer’s disease-related anatomical or clinical symptoms in individuals without ApoE4 genotype [66].

The association of diet rich in saturated fatty acids may increase Alzheimer’s risks in ApoE4 carriers [67].

3.2.3 Nonalcoholic fatty liver disease NFLD

Nonalcoholic fatty liver disease (NAFLD) is a chronic condition associated with genetic and environmental factors, obesity, type 2 diabetes, and dyslipidemia in which fat abnormally accumulates in the liver. Different genetic polymorphisms seem to be involved in this context [68].

3.2.4 Vitamins

3.2.4.1 Vitamin A

Retinol (Vitamin A) plays a crucial role in the anti-aging industry, primarily due to its ability to neutralize free radicals in tissues, which subsequently leads to a reduced appearance of wrinkles.

β-carotene 15,15′-monooxygenase 1 (BCMO1) is the most critical enzyme involved in retinoid metabolism [69].

Especially, A379V TT variant was inversely related to vitamin A status [69]. Assessment of the responsiveness to beta-carotene confirmed that carriers of variant alleles had a reduced ability to convert beta-carotene [70].

Individual responsiveness was associated with genetic variants in SNP rs7501331 of the carotenoid metabolizing enzyme BCMO1, resulting of single nucleotide variation (SNV) from C to T [71]. Carriers of T nucleotide have lower ability to convert beta-carotene. Studies shown that only 5% of the population have TT genotypes, while 56% have CC. Having this on mined and knowing that lacking of normal retinol metabolism is responsible for several diseases, supplementation/treatment of vitamin A should be completely personalized in the future in regards to genetic variations in the BCO1 gene [72, 73].

3.2.4.2 Vitamin B9 (folic acid)

Vitamin B9 is molecule responsible for normal cell growth and development. It is crucial supplement in the prevention of pregnancy complication. Enzyme, which plays a key role in vitamin D metabolisms, is 5,10-methylenetetrahydrofolate reductase (MTHFR), and it regulates around 60% of folic acid metabolism [48].

Mutations in the MTHFR gene can result in abnormal folate metabolism, which is associated with and may contribute to various pathological conditions, including stroke, depression, and reduced cognitive function etc.

Two mutations in SNPs, rs1801131 (SNV, T > G) and rs1801133 (SNV, G > A) have been reported that affect enzymatic activity of MTHFR [74].

Depending in which SNP is the mutation and the state of mutation (is it homozygous or not) several possible phenotypes can be detected. Each phenotype is associated with specific enzyme the activity and function.

Individuals with MTHFR (rs1801133) genotype CC has normal homocysteine levels, on the other hand, patients with TT genotype have high level of homocysteine and low folate levels. So, autosomal recessive MTHFR polymorphism led to wide range of vascular and neurological unfunctionally [75, 76].

The risk genotype of rs1801133 has been related with various pathological conditions such as deep vein thrombosis, various cardiovascular diseases (CVDs), then cancer, diabetes, etc. [76, 77, 78].

TT genotype is also known as C677T MTHFR polymorphism. Supposition of C with T will lead to amino acid change from alanine to valine. If a patient has two defective alleles of the MTHFR gene, enzyme activity can be reduced by 80 to 90%. Recent clinical studies have demonstrated that carriers of these alleles are at a significantly higher risk of ischemic stroke [79].

Studies on MTHFR have been successfully used to develop disease prevention strategies [80]. And therefore, future health education has to be based on personalized nutritional recommendations and prevention strategies in the field of vitamins supplementation.

3.2.4.3 Vitamin D

Vitamin D has been highlighted as a prime example of nutrigenomics. It is a molecule with multiple roles in the human body, including important functions in metabolism and various clinical applications [81, 82].

This vitamin plays a role in numerous system, such as in the immune system [83], skeletal system [84], reproductive system [85], insulin secretion [86], and intestinal system [87].

Vitamin D deficiency is very frequent, with almost 40% of the Europeans presenting levels below 50 nmol/L [88].

Vitamin D receptor (VDR) regulates several target gene transcription processes necessary for various biological functions of vitamin D. VDR will make hormone-receptor complex with active form of vitamin D (1,25(OH)2D3) in target cells. This complex interfere with specific DNA sequences of target genes to control the expression of numerous genes [89, 90].

Some genetic variants of the gene encoding VDR modify either its expression or function, with the consequent disruption of the vitamin D signaling pathway. Recent publications on the relationship between VDR genetic variants and the risk of type 2 diabetes, metabolic syndrome, overweight, and obesity were reviewed and give only partial answers to this question [91].

Vitamin D analogs bind to vitamin D receptors in tumor cells and activate downstream pathways to inhibit tumor growth. VDR expression is a prognostic indicator for digestive system tumors. That the intake of vitamin D analogs should be determined according to vitamin D receptor expression was stated in a comprehensive review on tumors of the digestive tract [92].

Findings considering gene polymorphisms in the VDR gene, which are based on the role of VDR SNPs in gene regulation and protein expression, will help to understand the detected role of VDR in various diseases [93].

For personalized medicine and pharmacogenomics new studies of VDR polymorphisms and vitamin D-VDR signaling are necessary for better understanding of role of this complex in various diseases [94].

Individuals who are genetically predisposed to low vitamin D benefit from foods rich in this vitamin [95, 96].

Individuals with genetic changes in the VDR gene may benefit from foods rich in vitamin D and from calcium and/or vitamin D supplementation [97, 98].

3.3 Intolerances

3.3.1 Gluten

Celiac disease (CD) is an autoimmune disorder, affecting about 1% of the population, where individuals are genetically predisposed to gluten intolerance [99, 100]. Gluten is a protein complex present in some cereals such as wheat, barley, and rye, in which gliadins and glutenin proteins are considered to be responsible for the inappropriate immune response of the genetically predisposed individuals. This condition can cause a localized complication in the mucosa of the intestine with toxic effects, leading to villous atrophy and lymphocyte infiltration in the small intestinal mucosa [101]. Typical symptoms include diarrhea, digestive tract pain and discomfort, weight loss, and malabsorption of nutrients [102].

There is a genetic basis at the origin of CD that determines susceptibility to the disease, which is correlated with genes in the human leukocyte antigen (HLA) system. More specifically, genetic testing for CD consists of determining the presence of the HLA DQ2 and DQ8 alleles [103].

Most CD cases (90%) are associated with the presence of the HLA-DQ2 haplotype encoded by (HLA-DQA1*05-DQB1*02). Some patients (5%) carry a second HLA DQ8 heterodimer encoded by (DQA1*03-DQB1*0302), and the remaining 5% of patients hold at least one of the two genes [104]. Six SNPs in HLA-DQ genes are responsible for CD, and in 75% patients CD has hereditary pattern. Sporadic, non -HLA related CD will occur in 68% patients [105].

Screening of single nucleotide polymorphisms by mass spectrometry within HLA region is an efficient method to accurately analyze multiple SNPs at the same time [106]. For example, individuals with CC (around 1% in Caucasian population) genotype in HLA-DQ8 gene (rs7454108) have high risk of gluten intolerance, while CT genotype indicates moderate risk of gluten intolerance However, the most frequent genotype for this polymorphism in Caucasian population is TT (no risk), around 80% (Figure 2), indicating that other genes. SNPs from HLA-DQ system plays key role in gluten intolerance.

Figure 2.

(A) Representative cluster plot displaying the SNP HLADQ8_rs7454108 genotypes of 70 samples assayed. Heterozygous region is close to the 45°-line (green squares represent heterozygous (TC) samples. Homozygous region for the low mass allele is close to the X coordinate (blue triangles represent homozygous (TT) samples for the same SNP. (B) Representative spectrum of homozygous genotype (TT) for the SNP HLADQ8_rs7454108 where one single peak of interest (blue dotted line) is indicated for the allele T. (C) Example of a spectrum of heterozygous genotype (TC) for the same SNP HLADQ8_rs7454108 where two peaks of interest are indicated with blue dotted lines representing the heterozygous alleles (AT).

3.3.2 Lactose

The most common carbohydrate, which is the main component of milk and milk products, is disaccharide lactose. Necessary enzymes in lactose metabolism (located at LCT/MCM6 gene) have role to cut lactose in glucose and galactose [107]. The ability to digest and have a normal metabolism of lactose in adults is called lactase persistence/persistent (LP). LP is regulated by five genetic polymorphisms, which have dominant distribution in humans [108].

Lactase non-persistence/intolerance is a worldwide phenomenon, but it affects people with varying degrees of severity. Completely inactive lactose gene is very rare and present in patients with alactasia [109].

Lactose intolerance can manifest as secondary lactase deficiency, which can be either temporary or chronic, depending on the duration and nature of the harmful mediator affecting the small intestinal mucosa cells. Alternatively, it can appear as primary lactase deficiency, typically emerging in adolescence or early adulthood [110].

In the natural condition lactase non-persistence (LPN) in primary lactase deficiency, the activity of the enzyme LCT decreases with age. Various studies have been done and shown that genetic alterations are responsible for type of reduced lactase enzyme activity [111].

In the European population polymorphism in gene MCM6 (rs4988235, C > T), which is placed in the promoter region of MCM6 gene is mainly responsible for tolerance or intolerance for lactose.

Lactase intolerance in adults is triggered by a recessively congenital polymorphism of the MCM6 gene, and this phenotype is inherited as an autosomal dominant characteristic. Individuals who carry C alle are likely that will develop lactose intolerance during lifetime, meaning that CT individuals have more chance that they can digest milk in older ages, while TT individuals can digest lactose during all lifetime [112].

The decreased ability of the body to hydrolyze lactose is due to a programmed regulatory phenomenon involving the MCM6 gene intron, 13,14 kb upstream of the MCM6 gene. This gene has several single nucleotide base polymorphisms including the (rs4988235) for which the Thymine (T) allele forms an haplotype that is commonly evaluated in LP studies [113].

The −13,910∗T allele is commonly distributed in the European population with an average frequency of 50.8%. However, genetic distribution of T allele is most prevalent in northern Europe, especially England and Scotland (72.0% of the population), while it progressively decreases in the southern Europe with a frequency of 8.9% in Tuscany, Italy [114].

Mutations in the SNP rs4988235 cause intolerance problems [115]. Specifically, a genotype that has two (TT) at position –13,910 results in a lactase persistence (LP), while a homozygous CC genotype in the same position results in a lactase non-persistent (LNP) phenotype [116, 117].

The undigested lactose that remains in the intestine is metabolized by intestinal bacteria with the generation of an osmotic effect causing a recall of water, resulting in symptoms such as diarrhea, cramping, meteorism, intestinal discomfort, and sometimes nausea and vomiting [118, 119].

Twenty-nine percentage of individuals reported symptoms attributed to the ingestion of fresh milk, with abdominal pain, bloating, and flatulence being the most frequent [120].

Previous studies have shown that gut microbiota may be capable of adapting to lactose consumption in LNP individuals [121].

Mass spectrometry seems to be an ideal method to detect SNPs in the MCM-Gene. By excluding the genetic predisposition to lactose intolerance, people can avoid unnecessary dietary restrictions on dairy products [122].

Advertisement

4. Conclusions

This book chapter aimed to present the application of mass spectrometry for DNA analysis. After a small introduction of application of mass spectrometry in modern medicine methods for the detection of SNPs were discussed. Furthermore, an overview about studies using SNPs as genetic markers related to nutrigenetics, including fat metabolism, vitamins, and intolerances, were provided.

In the last years, MALDI-TOF mass spectrometry technique has been proven to be a versatile tool for the characterization of point mutations. This method allows the detection of SNPs in a rapid, precise, cost-effective, and high-throughput way. MALDI-TOF is a technique, which allows assessment of up to 250 SNPs. For the analysis of a larger amount of SNPs or genome-wide association studies, next-generation sequencing is the method of choice.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Li D, Yi J, Han G, Qiao L. MALDI-TOF mass spectrometry in clinical analysis and research. ACS Measurement Science Au. 2022;2(5):385-404
  2. 2. Chen XF, Hou X, Xiao M, Zhang L, Cheng JW, Zhou ML, et al. Matrix-assisted laser desorption/ionization time of flight mass spectrometry (MALDI-TOF MS). Analysis for the identification of pathogenic microorganisms: A review. Microorganisms. 2021;9(7):1536
  3. 3. Oviano M, Rodriguez-Sanchez B, Gomara M, Alcala L, Zvezdanova E, Ruiz A, et al. Direct identification of clinical pathogens from liquid culture media by MALDI-TOF MS analysis. Clinical Microbiology and Infection. 2018;24(6):624-629
  4. 4. Hou TY, Chiang-Ni C, Teng SH. Current status of MALDI-TOF mass spectrometry in clinical microbiology. Journal of Food and Drug Analysis. 2019;27(2):404-414
  5. 5. Oviano M, Bou G. Matrix-assisted laser desorption ionization-time of flight mass spectrometry for the rapid detection of antimicrobial resistance mechanisms and beyond. Clinical Microbiology Reviews. 2019;32(1):e00037-18
  6. 6. Vrioni G, Tsiamis C, Oikonomidis G, Theodoridou K, Kapsimali V, Tsakris A. MALDI-TOF mass spectrometry technology for detecting biomarkers of antimicrobial resistance: Current achievements and future perspectives. Annals of Translational Medicine. 2018;6(12):240
  7. 7. Wierz M, Sauerbrei B, Wandernoth P, Kriegsmann M, Casadonte R, Kriegsmann K, et al. Detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) including variant analysis by mass spectrometry in placental tissue. Viruses. 2022;14(3):604
  8. 8. Calderaro A, Arcangeletti MC, Rodighiero I, Buttrini M, Montecchini S, Vasile Simone R, et al. Identification of different respiratory viruses, after a cell culture step, by matrix assisted laser desorption/ionization time of flight mass spectrometry (MALDI-TOF MS). Scientific Reports. 2016;6:36082
  9. 9. Ren JL, Zhang AH, Kong L, Wang XJ. Advances in mass spectrometry-based metabolomics for investigation of metabolites. RSC Advances. 2018;8(40):22335-22350
  10. 10. Fresnais M, Yildirim E, Karabulut S, Jager D, Zornig I, Benzel J, et al. Rapid MALDI-MS assays for drug quantification in biological matrices: Lessons learned, new developments, and future perspectives. Molecules. 2021;26(5)
  11. 11. Spruill ML, Maletic-Savatic M, Martin H, Li F, Liu X. Spatial analysis of drug absorption, distribution, metabolism, and toxicology using mass spectrometry imaging. Biochemical Pharmacology. 2022;201:115080
  12. 12. Karlsson O, Hanrieder J. Imaging mass spectrometry in drug development and toxicology. Archives of Toxicology. 2017;91(6):2283-2294
  13. 13. Yagnik G, Liu Z, Rothschild KJ, Lim MJ. Highly multiplexed Immunohistochemical MALDI-MS imaging of biomarkers in tissues. Journal of the American Society for Mass Spectrometry. 2021;32(4):977-988
  14. 14. Casadonte R, Kriegsmann M, Kriegsmann K, Hauk I, Meliss RR, Muller CSL, et al. Imaging mass spectrometry-based proteomic analysis to differentiate melanocytic nevi and malignant melanoma. Cancers (Basel). 2021;13(13):3197
  15. 15. Kriegsmann M, Zgorzelski C, Casadonte R, Schwamborn K, Muley T, Winter H, et al. Mass spectrometry imaging for reliable and fast classification of non-small cell lung cancer subtypes. Cancers (Basel). 2020;12(9):1-14
  16. 16. Yang HC, Li W, Sun J, Gross ML. Advances in mass spectrometry on membrane proteins. Membranes (Basel). 2023;13(5):457
  17. 17. Taha HB, Chawla E, Bitan G. IM-MS and ECD-MS/MS provide insight into modulation of amyloid proteins self-assembly by peptides and small molecules. Journal of the American Society for Mass Spectrometry. 2023;34(10):2066-2086
  18. 18. Holzlechner M, Eugenin E, Prideaux B. Mass spectrometry imaging to detect lipid biomarkers and disease signatures in cancer. Cancer Report (Hoboken). 2019;2(6):e1229
  19. 19. Scott DA, Casadonte R, Cardinali B, Spruill L, Mehta AS, Carli F, et al. Increases in tumor N-glycan polylactosamines associated with advanced HER2-positive and triple-negative breast cancer tissues. Proteomics Clinical Applications. 2019;13(1):e1800014
  20. 20. Li Y, Wang J, Chen W, Lu H, Zhang Y. Comprehensive review of MS-based studies on N-glycoproteome and N-glycome of extracellular vesicles. Proteomics. 2023;2023:e2300065
  21. 21. McDowell CT, Lu X, Mehta AS, Angel PM, Drake RR. Applications and continued evolution of glycan imaging mass spectrometry. Mass Spectrometry Reviews. 2023;42(2):674-705
  22. 22. Peng W, Kobeissy F, Mondello S, Barsa C, Mechref Y. MS-based glycomics: An analytical tool to assess nervous system diseases. Frontiers in Neuroscience. 2022;16:1000179
  23. 23. Wang Z, Zhu H, Xiong W. Advances in mass spectrometry-based multi-scale metabolomic methodologies and their applications in biological and clinical investigations. Science Bulletin (Beijing). 2023;68(19):2268-2284
  24. 24. Kriegsmann M, Arens N, Endris V, Weichert W, Kriegsmann J. Detection of KRAS, NRAS and BRAF by mass spectrometry – a sensitive, reliable, fast and cost-effective technique. Diagnostic Pathology. 2015;10:132
  25. 25. Su KY, Tseng JS, Liao KM, Yang TY, Chen KC, Hsu KH, et al. Mutational monitoring of EGFR T790M in cfDNA for clinical outcome prediction in EGFR-mutant lung adenocarcinoma. PLoS One. 2018;13(11):e0207001
  26. 26. Sutton BC, Birse RT, Maggert K, Ray T, Hobbs J, Ezenekwe A, et al. Assessment of common somatic mutations of EGFR, KRAS, BRAF, NRAS in pulmonary non-small cell carcinoma using iPLEX(R) HS, a new highly sensitive assay for the MassARRAY(R) system. PLoS One. 2017;12(9):e0183715
  27. 27. Lin Y, Lin CH, Yin X, Zhu L, Yang J, Shen Y, et al. Newborn screening for spinal muscular atrophy in China using DNA mass spectrometry. Frontiers in Genetics. 2019;10:1255
  28. 28. Lin Y, Zheng W, Chen Y, Huang C, Fu Q , Chen D, et al. Incorporating second-tier genetic screening for multiple acyl-CoA dehydrogenase deficiency. Clinica Chimica Acta. 2022;537:181-187
  29. 29. Williams GR, Cook L, Lewis LD, Tsongalis GJ, Nerenz RD. Clinical validation of a 106-SNV MALDI-ToF MS pharmacogenomic panel. Journal of Applied Lab Medicine. 2020;5(3):454-466
  30. 30. Wollmann BM, Storset E, Kringen MK, Molden E, Smith RL. Prediction of CYP2D6 poor metabolizers by measurements of solanidine and metabolites-a study in 839 patients with known CYP2D6 genotype. European Journal of Clinical Pharmacology. 2023;79(4):523-531
  31. 31. Evans JP, Skrzynia C, Burke W. The complexities of predictive genetic testing. BMJ. 2001;322(7293):1052-1056
  32. 32. Unim B, Pitini E, Lagerberg T, Adamo G, De Vito C, Marzuillo C, et al. Current genetic service delivery models for the provision of genetic testing in Europe: A systematic review of the literature. Frontiers in Genetics. 2019;10:552
  33. 33. International HapMap C, Frazer KA, Ballinger DG, Cox DR, Hinds DA, Stuve LL, et al. A second generation human haplotype map of over 3.1 million SNPs. Nature. 2007;449(7164):851-861
  34. 34. Shastry BS. SNPs: Impact on gene function and phenotype. Methods in Molecular Biology. 2009;578:3-22
  35. 35. Defo J, Awany D, Ramesar R. From SNP to pathway-based GWAS meta-analysis: Do current meta-analysis approaches resolve power and replication in genetic association studies? Briefings in Bioinformatics. 2023;24(1):bbac600
  36. 36. Frayling TM, Timpson NJ, Weedon MN, Zeggini E, Freathy RM, Lindgren CM, et al. A common variant in the FTO gene is associated with body mass index and predisposes to childhood and adult obesity. Science. 2007;316(5826):889-894
  37. 37. Hernandez MM, Banu R, Shrestha P, Patel A, Chen F, Cao L, et al. RT-PCR/MALDI-TOF mass spectrometry-based detection of SARS-CoV-2 in saliva specimens. Journal of Medical Virology. 2021;93(9):5481-5486
  38. 38. Ramos-Levi A, Barabash A, Valerio J, Garcia de la Torre N, Mendizabal L, Zulueta M, et al. Genetic variants for prediction of gestational diabetes mellitus and modulation of susceptibility by a nutritional intervention based on a Mediterranean diet. Frontier in Endocrinology (Lausanne). 2022;13:1036088
  39. 39. Ryan DJ, Toomey S, Smyth R, Madden SF, Workman J, Cummins R, et al. Exhaled breath condensate (EBC) analysis of circulating tumour DNA (ctDNA) using a lung cancer specific UltraSEEK oncogene panel. Lung Cancer. 2022;168:67-73
  40. 40. Allinson LM, Potts A, Goodman A, Bown N, Bashton M, Thompson D, et al. Loss of ALK hotspot mutations in relapsed neuroblastoma. Genes, Chromosomes & Cancer. 2022;61(12):747-753
  41. 41. Pedersen H, Ejegod DM, Quint W, Xu L, Arbyn M, Bonde J. Clinical performance of the full genotyping Agena MassARRAY HPV assay using SurePath screening samples within the VALGENT4 framework. The Journal of Molecular Diagnostics. 2022;24(4):365-373
  42. 42. Giannoudis A, Sartori A, Eastoe L, Zakaria R, Charlton C, Hickson N, et al. Genomic profiling using the UltraSEEK panel identifies discordancy between paired primary and breast cancer brain metastases and an association with brain metastasis-free survival. Breast Cancer Research and Treatment. 2021;190(2):241-253
  43. 43. Sirivisoot S, Kasantikul T, Techangamsuwan S, Radtanakatikanon A, Chen K, Lin TY, et al. Evaluation of 41 single nucleotide polymorphisms in canine diffuse large B-cell lymphomas using MassARRAY. Scientific Reports. 2022;12(1):5120
  44. 44. Tian HX, Zhang XC, Wang Z, Chen JG, Chen SL, Guo WB, et al. Establishment and application of a multiplex genetic mutation-detection method of lung cancer based on MassARRAY platform. Cancer Biology & Medicine. 2016;13(1):68-76
  45. 45. Ellis JA, Ong B. The MassARRAY((R)) system for targeted SNP genotyping. Methods in Molecular Biology. 2017;1492:77-94
  46. 46. Oeth P, del Mistro G, Marnellos G, Shi T, van den Boom D. Qualitative and quantitative genotyping using single base primer extension coupled with matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MassARRAY). Methods in Molecular Biology. 2009;578:307-343
  47. 47. Kiani AK, Bonetti G, Donato K, Kaftalli J, Herbst KL, Stuppia L, et al. Polymorphisms, diet and nutrigenomics. Journal of Preventive Medicine and Hygiene. 2022;63(2 Suppl 3):E125-EE41
  48. 48. Wang F, Zheng J, Cheng J, Zou H, Li M, Deng B, et al. Personalized nutrition: A review of genotype-based nutritional supplementation. Frontiers in Nutrition. 2022;9:992986
  49. 49. Singh V. Current challenges and future implications of exploiting the omics data into nutrigenetics and nutrigenomics for personalized diagnosis and nutrition-based care. Nutrition. 2023;110:112002
  50. 50. Lal MK, Sharma E, Tiwari RK, Devi R, Mishra UN, Thakur R, et al. Nutrient-mediated perception and signalling in human metabolism: A perspective of nutrigenomics. International Journal of Molecular Sciences. 2022;23(19):11305
  51. 51. Guevara-Ramirez P, Cadena-Ullauri S, Ruiz-Pozo VA, Tamayo-Trujillo R, Paz-Cruz E, Simancas-Racines D, et al. Genetics, genomics, and diet interactions in obesity in the Latin American environment. Frontiers in Nutrition. 2022;9:1063286
  52. 52. Sekar P, Ventura EF, Dhanapal A, Cheah ESG, Loganathan A, Quen PL, et al. Gene-diet interactions on metabolic disease-related outcomes in southeast Asian populations: A systematic review. Nutrients. 2023;15(13)
  53. 53. Mitchelson KAJ, Ni Chathail MB, Roche HM. Systems biology approaches to inform precision nutrition. The Proceedings of the Nutrition Society. 2023;82(2):208-218
  54. 54. Xu ZY, Jing X, Xiong XD. Emerging role and mechanism of the FTO gene in cardiovascular diseases. Biomolecules. 2023;13(5):850
  55. 55. Scuteri A, Sanna S, Chen WM, Uda M, Albai G, Strait J, et al. Genome-wide association scan shows genetic variants in the FTO gene are associated with obesity-related traits. PLoS Genetics. 2007;3(7):e115
  56. 56. Sentinelli F, Incani M, Coccia F, Capoccia D, Cambuli VM, Romeo S, et al. Association of FTO polymorphisms with early age of obesity in obese Italian subjects. Experimental Diabetes Research. 2012;2012:872176
  57. 57. Grunnet LG, Nilsson E, Ling C, Hansen T, Pedersen O, Groop L, et al. Regulation and function of FTO mRNA expression in human skeletal muscle and subcutaneous adipose tissue. Diabetes. 2009;58(10):2402-2408
  58. 58. Smemo S, Tena JJ, Kim KH, Gamazon ER, Sakabe NJ, Gomez-Marin C, et al. Obesity-associated variants within FTO form long-range functional connections with IRX3. Nature. 2014;507(7492):371-375
  59. 59. Karra E, O'Daly OG, Choudhury AI, Yousseif A, Millership S, Neary MT, et al. A link between FTO, ghrelin, and impaired brain food-cue responsivity. The Journal of Clinical Investigation. 2013;123(8):3539-3551
  60. 60. Song Y, Wade H, Zhang B, Xu W, Wu R, Li S, et al. Polymorphisms of fat mass and obesity-associated gene in the pathogenesis of child and adolescent metabolic syndrome. Nutrients. 2023;15(12):2643
  61. 61. Popovic AM, Hudek Turkovic A, Zuna K, Bacun-Druzina V, Rubelj I, Matovinovic M. FTO gene polymorphisms at the crossroads of metabolic pathways of obesity and epigenetic influences. Food Technology and Biotechnology. 2023;61(1):14-26
  62. 62. Rivera-Iniguez I, Gonzalez- Becerra K, Ramos-Lopez O, Perez- Beltran YE, Chaguen-Hernandez MS, Martinez-Lopez E, et al. Lipid-related genetic variants for personalized dietary interventions: A systematic review. Molecular Nutrition & Food Research. 2023;67(14):e2200675
  63. 63. Jabeen K, Rehman K, Akash MSH. Genetic mutations of APOEepsilon4 carriers in cardiovascular patients lead to the development of insulin resistance and risk of Alzheimer’s disease. Journal of Biochemical and Molecular Toxicology. 2022;36(2):e22953
  64. 64. Marais AD. Apolipoprotein E in lipoprotein metabolism, health and cardiovascular disease. Pathology. 2019;51(2):165-176
  65. 65. Bos MM, Noordam R, Blauw GJ, Slagboom PE, Rensen PCN, van Heemst D. The ApoE epsilon4 isoform: Can the risk of diseases be reduced by environmental factors? The Journals of Gerontology. Series A, Biological Sciences and Medical Sciences. 2019;74(1):99-107
  66. 66. Martinez-Lapiscina EH, Galbete C, Corella D, Toledo E, Buil-Cosiales P, Salas-Salvado J, et al. Genotype patterns at CLU, CR1, PICALM and APOE, cognition and Mediterranean diet: The PREDIMED-NAVARRA trial. Genes & Nutrition. 2014;9(3):393
  67. 67. Kivipelto M, Rovio S, Ngandu T, Kareholt I, Eskelinen M, Winblad B, et al. Apolipoprotein E epsilon4 magnifies lifestyle risks for dementia: A population-based study. Journal of Cellular and Molecular Medicine. 2008;12(6B):2762-2771
  68. 68. Vasconcellos C, Ferreira O, Lopes MF, Ribeiro AF, Vasques J, Guerreiro CS. Nutritional genomics in nonalcoholic fatty liver disease. Biomedicine. 2023;11(2):319
  69. 69. Zumaraga MPP, Arquiza J, Concepcion MA, Perlas L, Alcudia-Catalma MN, Rodriguez M. Genotype effects on beta-carotene conversion to vitamin A: Implications on reducing vitamin A deficiency in the Philippines. Food and Nutrition Bulletin. 2022;43(1):25-34
  70. 70. Leung WC, Hessel S, Meplan C, Flint J, Oberhauser V, Tourniaire F, et al. Two common single nucleotide polymorphisms in the gene encoding beta-carotene 15,15′-monoxygenase alter beta-carotene metabolism in female volunteers. The FASEB Journal. 2009;23(4):1041-1053
  71. 71. Wang TT, Edwards AJ, Clevidence BA. Strong and weak plasma response to dietary carotenoids identified by cluster analysis and linked to beta-carotene 15,15′-monooxygenase 1 single nucleotide polymorphisms. The Journal of Nutritional Biochemistry. 2013;24(8):1538-1546
  72. 72. Moran NE, Thomas-Ahner JM, Fleming JL, McElroy JP, Mehl R, Grainger EM, et al. Single nucleotide polymorphisms in beta-carotene oxygenase 1 are associated with plasma lycopene responses to a tomato-soy juice intervention in men with prostate cancer. The Journal of Nutrition. 2019;149(3):381-397
  73. 73. Feigl B, Morris CP, Voisey J, Kwan A, Zele AJ. The relationship between BCMO1 gene variants and macular pigment optical density in persons with and without age-related macular degeneration. PLoS One. 2014;9(2):e89069
  74. 74. Goyette P, Christensen B, Rosenblatt DS, Rozen R. Severe and mild mutations in cis for the methylenetetrahydrofolate reductase (MTHFR) gene, and description of five novel mutations in MTHFR. American Journal of Human Genetics. 1996;59(6):1268-1275
  75. 75. Goyette P, Frosst P, Rosenblatt DS, Rozen R. Seven novel mutations in the methylenetetrahydrofolate reductase gene and genotype/phenotype correlations in severe methylenetetrahydrofolate reductase deficiency. American Journal of Human Genetics. 1995;56(5):1052-1059
  76. 76. Li WX, Cheng F, Zhang AJ, Dai SX, Li GH, Lv WW, et al. Folate deficiency and gene polymorphisms of MTHFR, MTR and MTRR elevate the hyperhomocysteinemia risk. Clinical Laboratory. 2017;63(3):523-533
  77. 77. Liew SC, Gupta ED. Methylenetetrahydrofolate reductase (MTHFR) C677T polymorphism: Epidemiology, metabolism and the associated diseases. European Journal of Medical Genetics. 2015;58(1):1-10
  78. 78. Raghubeer S, Matsha TE. Methylenetetrahydrofolate (MTHFR), the one-carbon cycle, and cardiovascular risks. Nutrients. 2021;13(12):4562
  79. 79. Zhao L, Li T, Dang M, Li Y, Fan H, Hao Q , et al. Association of methylenetetrahydrofolate reductase (MTHFR) rs1801133 (677C>T) gene polymorphism with ischemic stroke risk in different populations: An updated meta-analysis. Frontiers in Genetics. 2022;13:1021423
  80. 80. Huo Y, Li J, Qin X, Huang Y, Wang X, Gottesman RF, et al. Efficacy of folic acid therapy in primary prevention of stroke among adults with hypertension in China: The CSPPT randomized clinical trial. Journal of the American Medical Association. 2015;313(13):1325-1335
  81. 81. Bikle DD. Vitamin D metabolism, mechanism of action, and clinical applications. Chemistry & Biology. 2014;21(3):319-329
  82. 82. Szymczak-Pajor I, Miazek K, Selmi A, Balcerczyk A, Sliwinska A. The action of vitamin D in adipose tissue: Is there the link between vitamin D deficiency and adipose tissue-related metabolic disorders? International Journal of Molecular Sciences. 2022;23(2):956
  83. 83. Wimalawansa SJ. Infections and autoimmunity-the immune system and vitamin D: A systematic review. Nutrients. 2023;15(17):3842
  84. 84. Wu F, Fuleihan GE, Cai G, Lamberg-Allardt C, Viljakainen HT, Rahme M, et al. Vitamin D supplementation for improving bone density in vitamin D-deficient children and adolescents: Systematic review and individual participant data meta-analysis of randomized controlled trials. The American Journal of Clinical Nutrition. 2023;118(3):498-506
  85. 85. Mohan A, Haider R, Fakhor H, Hina F, Kumar V, Jawed A, et al. Vitamin D and polycystic ovary syndrome (PCOS): A review. Annals of Medical Surgery (Lond). 2023;85(7):3506-3511
  86. 86. Upadhyay PK, Thakur N, Vishwakarma VK, Srivastav RK, Ansari TM. Role of vitamin D in management of diabetes and unresolved cardiovascular diseases. Current Diabetes Reviews. 2024;20:e010923220647 [Online ahead of print]
  87. 87. Santa K, Watanabe K, Kumazawa Y, Nagaoka I. Phytochemicals and vitamin D for a healthy life and prevention of diseases. International Journal of Molecular Sciences. 2023;24(15)
  88. 88. Cashman KD, Dowling KG, Skrabakova Z, Gonzalez-Gross M, Valtuena J, De Henauw S, et al. Vitamin D deficiency in Europe: Pandemic? The American Journal of Clinical Nutrition. 2016;103(4):1033-1044
  89. 89. Nurminen V, Seuter S, Carlberg C. Primary vitamin D target genes of human monocytes. Frontiers in Physiology. 2019;10:194
  90. 90. Valdivielso JM, Fernandez E. Vitamin D receptor polymorphisms and diseases. Clinica Chimica Acta. 2006;371(1-2):1-12
  91. 91. Fronczek M, Osadnik T, Banach M. Impact of vitamin D receptor polymorphisms in selected metabolic disorders. Current Opinion in Clinical Nutrition and Metabolic Care. 2023;26(4):316-322
  92. 92. Zhao M, Liu Z, Shi H, Song J. Prognostic role of vitamin D receptor in digestive system tumours: A systematic review and preliminary meta-analysis. PLoS One. 2023;18(8):e0289598
  93. 93. Morrison NA, Qi JC, Tokita A, Kelly PJ, Crofts L, Nguyen TV, et al. Prediction of bone density from vitamin D receptor alleles. Nature. 1994;367(6460):284-287
  94. 94. Tourkochristou E, Mouzaki A, Triantos C. Gene polymorphisms and biological effects of vitamin D receptor on nonalcoholic fatty liver disease development and progression. International Journal of Molecular Sciences. 2023;24(9):8288
  95. 95. Bouillon R. Comparative analysis of nutritional guidelines for vitamin D. Nature Reviews. Endocrinology. 2017;13(8):466-479
  96. 96. Wang TJ, Zhang F, Richards JB, Kestenbaum B, van Meurs JB, Berry D, et al. Common genetic determinants of vitamin D insufficiency: A genome-wide association study. Lancet. 2010;376(9736):180-188
  97. 97. Arabi A, Zahed L, Mahfoud Z, El-Onsi L, Nabulsi M, Maalouf J, et al. Vitamin D receptor gene polymorphisms modulate the skeletal response to vitamin D supplementation in healthy girls. Bone. 2009;45(6):1091-1097
  98. 98. Morrison NA, George PM, Vaughan T, Tilyard MW, Frampton CM, Gilchrist NL. Vitamin D receptor genotypes influence the success of calcitriol therapy for recurrent vertebral fracture in osteoporosis. Pharmacogenetics and Genomics. 2005;15(2):127-135
  99. 99. Caio G, Volta U, Sapone A, Leffler DA, De Giorgio R, Catassi C, et al. Celiac disease: A comprehensive current review. BMC Medicine. 2019;17(1):142
  100. 100. Fasano A, Catassi C. Current approaches to diagnosis and treatment of celiac disease: An evolving spectrum. Gastroenterology. 2001;120(3):636-651
  101. 101. Kagnoff MF. Celiac disease: Pathogenesis of a model immunogenetic disease. The Journal of Clinical Investigation. 2007;117(1):41-49
  102. 102. Sapone A, Bai JC, Ciacci C, Dolinsek J, Green PH, Hadjivassiliou M, et al. Spectrum of gluten-related disorders: Consensus on new nomenclature and classification. BMC Medicine. 2012;10:13
  103. 103. Wolters VM, Wijmenga C. Genetic background of celiac disease and its clinical implications. The American Journal of Gastroenterology. 2008;103(1):190-195
  104. 104. Louka AS, Moodie SJ, Karell K, Bolognesi E, Ascher H, Greco L, et al. A collaborative European search for non-DQA1*05-DQB1*02 celiac disease loci on HLA-DR3 haplotypes: Analysis of transmission from homozygous parents. Human Immunology. 2003;64(3):350-358
  105. 105. Kuja-Halkola R, Lebwohl B, Halfvarson J, Wijmenga C, Magnusson PK, Ludvigsson JF. Heritability of non-HLA genetics in coeliac disease: A population-based study in 107 000 twins. Gut. 2016;65(11):1793-1798
  106. 106. Tsai SH, Chang PY, Wen YH, Lin WT, Hsu FP, Chen DP. Screening of single nucleotide polymorphisms within HLA region related to hematopoietic stem cell transplantation using MassARRAY technology. Scientific Reports. 2023;13(1):5913
  107. 107. Montgomery RK, Mulberg AE, Grand RJ. Development of the human gastrointestinal tract: Twenty years of progress. Gastroenterology. 1999;116(3):702-731
  108. 108. Fabre A, Fabre A, Bon C, Guerry P, Segurel L. Proposed mechanism for the selection of lactase persistence in childhood. BioEssays. 2023;45(7):e2200243
  109. 109. Kowalowka M, Kosewski G, Lipinski D, Przyslawski J. A comprehensive look at the −13910 C>T LCT gene polymorphism as a molecular marker for vitamin D and calcium levels in young adults in central and Eastern Europe: A preliminary study. International Journal of Molecular Sciences. 2023;24(12):10191
  110. 110. Porzi M, Burton-Pimentel KJ, Walther B, Vergeres G. Development of personalized nutrition: Applications in lactose intolerance diagnosis and management. Nutrients. 2021;13(5):1503
  111. 111. Lukito W, Malik SG, Surono IS, Wahlqvist ML. From ‘lactose intolerance’ to ‘lactose nutrition’. Asia Pacific Journal of Clinical Nutrition. 2015;24(Suppl 1):S1-S8
  112. 112. Ugidos-Rodriguez S, Matallana- Gonzalez MC, Sanchez-Mata MC. Lactose malabsorption and intolerance: A review. Food & Function. 2018;9(8):4056-4068
  113. 113. Guimaraes Alves AC, Sukow NM, Adelman Cipolla G, Mendes M, Leal TP, Petzl-Erler ML, et al. Tracing the distribution of European lactase persistence genotypes along the Americas. Frontiers in Genetics. 2021;12:671079
  114. 114. Auton A, Brooks LD, Durbin RM, Garrison EP, Kang HM, Korbel JO, et al. A global reference for human genetic variation. Nature. 2015;526(7571):68-74
  115. 115. Catanzaro R, Sciuto M, Marotta F. Lactose intolerance: An update on its pathogenesis, diagnosis, and treatment. Nutrition Research. 2021;89:23-34
  116. 116. De Luca P, Iaconis D, Biffali E, Enza C, de Magistris L, Riegler G, et al. Development of a novel SNP assay to detect lactase persistence associated genetic variants. Molecular Biology Reports. 2021;48(11):7087-7093
  117. 117. Enattah NS, Sahi T, Savilahti E, Terwilliger JD, Peltonen L, Jarvela I. Identification of a variant associated with adult-type hypolactasia. Nature Genetics. 2002;30(2):233-237
  118. 118. Hammer HF, Fox MR, Keller J, Salvatore S, Basilisco G, Hammer J, et al. European guideline on indications, performance, and clinical impact of hydrogen and methane breath tests in adult and pediatric patients: European Association for Gastroenterology, Endoscopy and Nutrition, European Society of Neurogastroenterology and Motility, and European Society for Paediatric Gastroenterology Hepatology and Nutrition consensus. United European Gastroenterology Journal. 2022;10(1):15-40
  119. 119. Segurel L, Bon C. On the evolution of lactase persistence in humans. Annual Review of Genomics and Human Genetics. 2017;18:297-319
  120. 120. Gaudin RGN, Figueiro G, Flores-Gutierrez S, Mut P, Vega-Requena Y, Luna-Andrada L, et al. DNA polymorphisms associated with lactase persistence, self-perceived symptoms of lactose intolerance, milk and dairy consumption, and ancestry, in the Uruguayan population. American Journal of Human Biology. 2023;35(6):e23868
  121. 121. Kable ME, Chin EL, Huang L, Stephensen CB, Lemay DG. Association of estimated daily lactose consumption, lactase persistence genotype (rs4988235), and gut microbiota in healthy adults in the United States. The Journal of Nutrition. 2023;153(8):2163-2173
  122. 122. Chengolova Z, Ivanova R, Gabrovska K. Lactose intolerance – single nucleotide polymorphisms and treatment. Journal of American Nutrition Association. 2023;2023:1-8

Written By

Jörg Kriegsmann, Sanja Cirovic, Rita Casadonte, Torsten Hansen, Katharina Kriegsmann and Mark Kriegsmann

Submitted: 22 September 2023 Reviewed: 22 September 2023 Published: 03 January 2024