Hostname: page-component-8448b6f56d-42gr6 Total loading time: 0 Render date: 2024-04-19T02:20:12.155Z Has data issue: false hasContentIssue false

Twin Registries Moving Forward and Meeting the Future: A Review

Published online by Cambridge University Press:  19 August 2019

Paul N. Baird*
Affiliation:
Department of Surgery, Ophthalmology, The University of Melbourne, Melbourne, Victoria, Australia
Pirro Hysi
Affiliation:
Department of Ophthalmology, King’s College London, St. Thomas’ Hospital, London, UK Department of Twin & Genetic Epidemiology, King’s College London, St. Thomas’ Hospital, London, UK
*
Author for correspondence: Paul N. Baird, Email: pbaird@unimelb.edu.au

Abstract

Twin registries have developed as a valuable resource for the study of many aspects of disease and society over the years in many different countries. A number of these registries include large numbers of twins with data collected at varying information levels for twin cohorts over the past several decades. More recent expansion of twin datasets has allowed for the collection of genetic data, together with many other levels of ‘omic’ information along with multiple demographic, physiological, health outcomes and other measures typically used in epidemiologic research. Other twin data sources outside these registries reflect research interests in particular aspects of disease or specific phenotypic assessment. Twin registries have the potential to play a key role in many aspects of the artificial intelligence/machine learning-driven projects of the future and will continue to keep adapting to the changing research landscape.

Type
Articles
Copyright
© The Author(s) 2019 

Background to Twin Registries

‘Mr Jones’s Case of Twins’ (Jones, Reference Jones1811) documented the earliest published article on twins in 1811 with the unexpected finding of a second fetus following labor and discussions on whether birth of the second twin child should be left and trusted to ‘nature’. While much of the literature for the following 60 years would center on obstetric considerations, it was not until Francis Galton’s study in 1875 when he recognized the potential of assessing shared environment and genetic differences through the study of twins. He made use of these findings in developing the first twin psychological questionnaire to query behavioral tendencies (Burbridge, Reference Burbridge2001). Since then, the recognition of twins to provide a powerful resource for exploring the role of genes and environment gathered speed through the establishment of twin registries.

The first reported use of twins in a registry was provided as part of the Danish Cancer Registry in 1942 (Busk et al., Reference Busk, Clemmesen and Nielsen1948) documenting 336 twin-pairs, but it was later in 1954 that the Danish Twin Registry was formally established. This national twin register represents the oldest in the world, having grown from the collection of twins born in Denmark from 1870 to 1910 to now having a total of more than 65,000 twin-pairs included in the registry (Skytthe et al., Reference Skytthe, Christiansen, Kyvik, Bodker, Hvidberg, Petersen and Christensen2013). This was followed by the Connecticut Twin Registry in 1961 (Honeyman, Reference Honeyman1961) and then the first mention of a twin registry dedicated to eye research in 1970, with the documented collection of over 650 twins in the Twin Eye Registry in the Washington DC area of the USA, aimed at studying diabetic retinopathy, glaucoma and myopia (Schwartz, Reference Schwartz1970). Since then, there has been a growth in twin registries across the globe, with over 30 of these currently listed by the International Society for Twin Studies (http://www.twinstudies.org/information/twinregisters/).

Approaches to Building Twin Registries

Varied study designs have been used by the different twin registries worldwide. For many twin registries, the initial approach to document twins was through linkage to national health population-based records. This population-based approach has been widely used by the Scandinavian countries when forming the Danish, Swedish, Finnish and Norwegian Twin Registries (Kaprio et al., Reference Kaprio, Koskenvuo and Rose1990). Other twin registries such as the Australian Twin Registry (ATR) are twin-volunteer-based and now house records on over 40,000 twin-pairs of all zygosity types and ages unselected for their health or medical history. The ATR facilitates researcher-led investigations rather than undertaking its own research studies (Hopper et al., Reference Hopper, Foley, White and Pollaers2013). These twin registries have allowed for the documentation of changing trends in disease in twins as well as prevalence changes in monozygotic (MZ) and dizygotic (DZ) twins over the years. Other twin registries have arisen through access to veteran affairs data. These include the Vietnam Era Twin Registry, composed of over 7,000 male–male twin-pairs of conscripts who served in the military during the time of the Vietnam conflict (1965–1975; Goldberg et al., Reference Goldberg, Curran, Vitek, Henderson and Boyko2002) and the The National Academy of Sciences-National Research Council Twin Registry (NAS-NRC Twin Registry) in the USA of 16,000 White male twin-pairs born in the years 1917–1927 who served in the armed forces (Gatz et al., Reference Gatz, Harris, Kaprio, McGue, Smith and Snieder2015). The above registries used a questionnaire to assess zygosity and health but have since followed up with additional questionnaires on education, employment history and earnings, as well as linkage to data records for mortality, disability and medical data. Other registries such as the Minnesota Twin Registry represent a birth-record-based twin registry, with data going back to 1904 with over 4,000 twins combined with questionnaire data on personality, occupational interests, demographics and leisure-time activities (Krueger & Johnson, Reference Krueger and Johnson2002).

Future Proofing Twin Registries

Twin registries provide a repository of information pertaining to twins allowing for qualitative assessment of twins through questionnaires that explore multiple facets of interest to researchers. In addition, they provide quantitative genetic methods to assess zygosity, concordance, correlations and heritability estimates. Investigation of the relative importance of genetic and environmental influences on a phenotype is also achieved through a number of twin designs, including the classical twin study, co-twin control study, within-pair twin family study, longitudinal twin studies, adopted twin study and randomized controlled trials (RCTs).

Through their life span, twin studies are usually designed to longitudinally follow up their participants from the moment of recruitment to older ages. This presents with huge opportunities to study a variety of life-course factors and address public health issues affecting different demographics and age groups. But, as is the case with all longitudinal information, the information housed in twin registries is potentially finite and restricted by normal factors determining continued participation such as attrition and longevity. This, therefore, poses a challenge in having an extremely valuable population or cohort resource at one level but the need to assure its continued use and relevance so that it can be future proofed in a rapidly changing environment. Longitudinal studies on registry twins provide valuable snapshots of information on a twin or twin cohort at varying timepoints in relation to disease phenotype and progression, but they are limited to questionnaires and standard twin statistics unless additional levels of data are available. Twin registries have established and collected other levels of information that can be used in the future, long after the normal life expectancy of their participants. These sources of information include several ‘omics’ data to enhance understanding of disease and extend this to complex diseases and enable identification of genetic risks, such as DNA genomic information through genome-wide association studies, next-generation sequencing, as well as explore other mechanisms of genetic changes such as gene expression regulation or various nonsequence-related epigenetic modifications (Kaminsky et al., Reference Kaminsky, Tang, Wang, Ptak, Oh, Wong and Petronis2009; Pedersen et al., Reference Pedersen, Lichtenstein and Svedberg2002; Skytthe et al., Reference Spector and Williams2006, Reference Skytthe, Christiansen, Kyvik, Bodker, Hvidberg, Petersen and Christensen2013; Spector & Williams, Reference Spector and Williams2006). Some twin registries such as Twins UK have extended beyond the collection of only a genetic sample to also include microbiome and biochemical data, some of which will be described in more detail below. These broad sample collections will ultimately allow for investigation of extensive clinical, genetic, physiological, behavioral and lifestyle data to provide a holistic approach to investigation of common diseases (Moayyeri et al., Reference Moayyeri, Hammond, Hart and Spector2013). The ability to undertake such studies comes from access to a range of tissue samples, including DNA, cell lines, serum, plasma, stool, urine, fat and skin, collected over multiple time points to provide the most comprehensive representation of an individual as possible.

Twin Studies in Epigenetics

In no other field of genetic research are natural twin experiments more relevant than in epigenetics. Methylation of a cytosine residue serves as a molecular interface with heritable, sequence variants of the DNA, which allows for environmental influences to alter transcription. Previous twin studies have demonstrated that population-wide variability of differentially methylated cytosine paired with guanidine (CpG) represents sites most readily affected by nonshared environmental exposures. This compares to one-third of the methylation variance determined by genetic additive effects (Busche et al., Reference Busche, Shao, Caron, Kwan, Allum, Cheung and Grundberg2015) that in addition to changes in DNA sequence can also affect the estimation of differentially methylated positions in the genome (Dyke et al., Reference Dyke, Cheung, Joly, Ammerpohl, Lutsik, Rothstein and Pastinen2015).

Epigenetic diversity arises throughout life, being reflected in differences in environmental exposures at early intrauterine life (Gordon et al., Reference Gordon, Joo, Andronikos, Ollikainen, Wallace, Umstad and Craig2011), growth during life course from childhood (Martino et al., Reference Martino, Loke, Gordon, Ollikainen, Cruickshank, Saffery and Craig2013) and through later life (Fraga et al., Reference Fraga, Ballestar, Paz, Ropero, Setien, Ballestar and Esteller2005). Therefore, differential methylation patterns over a person’s life can potentially explain larger proportions of phenotypic variability than DNA variants (Liang et al., Reference Liang, Willis-Owen, Laprise, Wong, Davies, Hudson and Cookson2015). Additionally, preexisting differential methylation inherited from parents/grandparents can also determine the patterns of subsequent interactions between an individual and environment (Klengel & Binder, Reference Klengel and Binder2015; Klengel et al., Reference Klengel, Mehta, Anacker, Rex-Haffner, Pruessner, Pariante and Binder2013). Thus, disentangling genetic from epigenetic marker effects of shared or nonshared environmental factors in epigenome-wide association studies (EWAS) is not a trivial exercise and presents a challenge even in well-powered large datasets that are currently generated through high-throughput platforms.

The Discordant MZ Twin Model

The discordant MZ twin model is a modern-day extension of the co-twin control methodology, first described in the early 20th century (Gesell, Reference Gesell1942). The purpose is to have a matched pairwise comparison between the epigenetic status and degree of phenotypic dissimilarity or discordance. MZ twins largely share the same genotypes and transgenerational epigenetic events (Wang et al., Reference Wang, Zhang, Duan, Gao, Zhu, Lu and Liu2014) from perinatal exposure onward (Gordon et al., Reference Gordon, Joo, Powell, Ollikainen, Novakovic, Li and Saffery2012; Kaati et al., Reference Kaati, Bygren and Edvinsson2002) that typically determines subsequent and stable patterns of gene expression profiles through the process of developmental epigenetic programming (Hochberg et al., Reference Hochberg, Feil, Constancia, Fraga, Junien, Carel and Albertsson-Wikland2011). For example, it is known that parental imprinting is modulated through epigenetic changes and is widespread across the genome (Baran et al., Reference Baran, Subramaniam, Biton, Tukiainen, Tsang, Rivas and Lappalainen2015; Zink et al., Reference Zink, Magnusdottir, Magnusson, Walker, Morris, Sigurdsson and Stefansson2018) with subsequent strong effect on health and disease (Begemann et al., Reference Begemann, Rezwan, Beygo, Docherty, Kolarova, Schroeder and Mackay2018). Epigenetic investigations in phenotypically discordant MZ twins have been able to show that aberrant imprinting in Beckwith–Wiedemann syndrome (Weksberg et al., Reference Weksberg, Shuman, Caluseriu, Smith, Fei, Nishikawa and Squire2002), Silver–Russell syndrome (Riess et al., Reference Riess, Binder, Ziegler, Begemann, Soellner and Eggermann2016) and others can exist.

Theoretical models show that the MZ power of discordant twin designs, compared to traditional unstructured cross-sectional regression-based models, rises proportionally with higher estimates of genetic heritability of the phenotypic traits (Li et al., Reference Li, Christiansen, Hjelmborg, Baumbach and Tan2018). This is apparent even after discounting for the presence of other confounders that are common in most diseases of complex etiology, such as demographic factors and life-course environmental exposures. MZ twin discordance models effectively remove the effect of most shared prenatal and early childhood heritable factors or exposures that are inherently difficult to model statistically. They also indirectly mitigate the impact of other confounding factors, such as those related to varied cell composition of sampled tissues included in EWAS analysis, thereby improving the uniformity of adjustment and performance of various deconvolution methods (Teschendorff & Zheng, Reference Teschendorff and Zheng2017).

Despite the improving availability of epigenetic information from large population-based unrelated-sample cohorts (whose results are being catalogued and curated in several repositories; Li, Zou et al., Reference Li, Zou, Li, Gao, Sang, Zhang and Zhang2019; Liu et al., Reference Liu, Zhao, Wang, Zhou, Fan, Li and Jiang2019), the discordant MZ twin model remains very popular (Table 1). Taking advantage of the additional power from this type of study design, important associations between differential methylation and several common traits, such as age-related macular degeneration (Wei et al., Reference Wei, Liu, Tuo, Shen, Chen, Li and Nussenblatt2012), rheumatoid arthritis (Svendsen et al., Reference Svendsen, Gervin, Lyle, Christiansen, Kyvik, Junker and Tan2016; Webster et al., Reference Webster, Plant, Ecker, Zufferey, Bell, Feber and Worthington2018), psychiatric (Chen et al., Reference Chen, Sudre, Sharp, Donovan, Chandrasekharappa, Hansen and Shaw2018; Córdova-Palomera et al., Reference Córdova-Palomera, Palma-Gudiel, Fores-Martos, Tabares-Seisdedos and Fananas2018; Kesselmeier et al., Reference Kesselmeier, Putter, Volckmar, Baurecht, Grallert and Illig2018; Peng et al., Reference Peng, Zhu, Strachan, Fowler, Bacus, Roy-Byrne and Zhao2018), metabolic phenotypes (Li, Zhang et al., Reference Li, Zou, Li, Gao, Sang, Zhang and Zhang2019; Peng et al., Reference Peng, Zhu, Goldberg, Vaccarino and Zhao2019) and others have been reported in the literature (Table 1). This trend is likely to continue in the short term, given the additional accuracy and increased popularity of pyrosequencing-based approaches when comparing the performance of multiple DNA methylation methods between different cell types (Blueprint Consortium, 2016), although the technology remains comparatively expensive.

Table 1. List of epigenetic studies that have used the discordant MZ twin model in the past five years

BMI, body mass index.

The Human Microbiome

The human microbiota consists of up to 100 trillion living cells (Turnbaugh et al., Reference Turnbaugh, Ley, Hamady, Fraser-Liggett, Knight and Gordon2007), a much higher number compared to the 37 trillion cells that make up the human body (Bianconi et al., Reference Bianconi, Piovesan, Facchin, Beraudi, Casadei, Frabetti and Canaider2013). At over 3 million unique genes (Qin et al., Reference Qin, Li, Raes, Arumugam, Burgdorf, Manichanh and Wang2010), the genetic complexity of the human microbiome is orders of magnitude higher than that of humans’ few tens of thousands (Salzberg, Reference Salzberg2018). These permanent inhabitants of the human body appear to play critical roles in both normal human (Le Chatelier et al., Reference Le Chatelier, Nielsen, Qin, Prifti, Hildebrand, Falony and Pedersen2013) and xenobiotic metabolism (Zimmermann et al., Reference Zimmermann, Zimmermann-Kogadeeva, Wegmann and Goodman2019) and may have profound effects over human health (Gilbert et al., Reference Gilbert, Quinn, Debelius, Xu, Morton, Garg and Knight2016). The human microbiota represents another layer of complexity in the quest to better understand mechanisms that contribute to our physiological homeostatic equilibria, and significant efforts are therefore being dedicated to research the interactions between host and microbiome.

Studies of host genetic mechanisms controlling the abundance and diversity of the microbiota have provided a very nuanced picture. While earlier studies conducted on small sample sizes of 20–30 twin-pairs suggested that correlation between microbiomes was highest among MZ compared with DZ twins or unrelated individuals (Stewart et al., Reference Stewart, Chadwick and Murray2005), a subsequent twin study using 16S ribosomal RNA metagenomic sequencing found no evidence in favor of heritability of the microbiota diversity (as measured by uniFrac, a commonly used metric of ecological diversity; Lozupone et al., Reference Lozupone, Hamady and Knight2006). Subsequent studies of the human microbiota, based on deep metagenomic pyrosequencing of 16S rRNA genes, have revealed that the majority of the metagenomic information is currently not mapped to specific reference genomes of known bacterial species (Turnbaugh et al., Reference Turnbaugh, Quince, Faith, McHardy, Yatsunenko, Niazi and Gordon2010). This can affect our ability to estimate the diversity of the microbiota in humans and make reliable comparisons difficult between twin and unrelated subjects. In addition, the amount of the microbiome complexity observed was directly dependent on the depth of the sequencing information (Turnbaugh et al., Reference Turnbaugh, Quince, Faith, McHardy, Yatsunenko, Niazi and Gordon2010).

Higher powered classic twin models have subsequently improved the estimates of the heritable component of the human microbiome. Using data from 416 twin-pairs, Goodrich et al. (Reference Goodrich, Waters, Poole, Sutter, Koren, Blekhman and Ley2014) concluded that while there is little support for genetic control over microbiome diversity in humans, there was substantial heritability for the numbers and abundance of operational taxonomic units (OTUs), the individual components of the microbiome in the gut. Consistent suggestions that coverage and statistical power affect microbiome heritability estimates were confirmed in a subsequent and improved classic twin model experiment involving 1,126 MZ and DZ twin-pairs, which provided increased estimates of heritability across all the OTUs assessed (Goodrich et al., Reference Goodrich, Davenport, Beaumont, Jackson, Knight, Ober and Ley2016). The authors also found association between OTU presence and genes that contribute to olfaction and diet (Goodrich et al., Reference Goodrich, Davenport, Beaumont, Jackson, Knight, Ober and Ley2016). Surprisingly, the gut microbiome preserves a degree of stability over decades of life, since the genetic variability appears to be more similar among MZ twins, even at older ages (Xie et al., Reference Xie, Guo, Zhong, Feng, Lan, Qin and Jia2016).

Although there is a better understanding of the genetic component determining diversity and composition of the human microbiome, several questions remain open. For example, the true nature of the interaction between the host genome and the microbiome remains unknown. Dietary preferences are heritable (Teucher et al., Reference Teucher, Skinner, Skidmore, Cassidy, Fairweather-Tait, Hooper and MacGregor2007) and can also directly cause changes in the gut microbiome (Cotillard et al., Reference Cotillard, Kennedy, Kong, Prifti, Pons, Le Chatelier and Ehrlich2013; Shoaie et al., Reference Shoaie, Ghaffari, Kovatcheva-Datchary, Mardinoglu, Sen, Pujos-Guillot and Nielsen2015). It is also unclear whether, and to what extent, innate (Huang, Reference Huang2015; Thaiss et al., Reference Thaiss, Zmora, Levy and Elinav2016; Yu et al., Reference Yu, Khodadadi and Baban2019) and adaptive immunity (Bonder et al., Reference Bonder, Kurilshikov, Tigchelaar, Mujagic, Imhann, Vila and Zhernakova2016) can affect the microbiome ecology. It is likely, however, that this is a two-way process (Slack et al., Reference Slack, Hapfelmeier, Stecher, Velykoredko, Stoel, Lawson and Macpherson2009), as the microbiome can induce immunological systemic changes, affecting organs and systems away from the gut (Erny et al., Reference Erny, Hrabe de Angelis, Jaitin, Wieghofer, Staszewski, David and Prinz2015). Finally, the ways in which the gut microbial ecology interacts with the microbiome are complex and need to be better understood. While dietary and host metabolic factors certainly influence the gut microbiome (Shoaie et al., Reference Shoaie, Ghaffari, Kovatcheva-Datchary, Mardinoglu, Sen, Pujos-Guillot and Nielsen2015), numerous products of human metabolism are processed in the human gut by the microbial flora (Tremaroli & Backhed, Reference Tremaroli and Backhed2012), which may interfere with the systemic wellbeing and therefore influence disease causation.

Twin Registries and Randomised Control Trials (RCTs)

Every clinical intervention is routinely assessed through RCTs that need to evaluate the efficacy of the intervention and its safety. RCTs need to be sufficiently well powered to capture the clinical and demographic complexities of the targeted population, which needs to be balanced against the time and material costs involved in running them. A very large clinical trial, in addition to the costs related to study setup and patient monitoring, may unduly delay the availability of potentially life-saving medication to the needing patients. Matching and randomization across the two arms of RCTs is critical to avoid bias in assessing the outcomes and allows a more realistic evaluation of cost-effectiveness of the treatment. The co-MZ twin model, due to its massive bias-reducing matching potential, is again thought to provide an ideal setting for RCTs (Plomin & Haworth, Reference Plomin and Haworth2010).

Experience has shown that this path, despite its theoretical potential, remains relatively unexplored, presumably due to lack of availability of twin siblings (Sumathipala et al., Reference Sumathipala, Yelland, Green, Shepherd, Jayaweera, Ferreira and Craig2018). In addition, twins participating in RCTs tend to be assigned to the same intervention arm (Sumathipala et al., Reference Sumathipala, Yelland, Green, Shepherd, Jayaweera, Ferreira and Craig2018), which may be a reflection of the genetically determined tendency toward phenotypic concordance in MZ twins, or due to a general preference toward being treated the same (Bernardo et al., Reference Bernardo, Nowacki, Martin, Fanaroff and Hibbs2015), perhaps due to parental preferences or psychological reasons ingrained in the participants’ twin identity.

Given the expected relatively low numbers of disease-discordant MZ twins available for therapies targeting rarer conditions, a more realistic option is to encourage participation of twins, MZ pairs in particular, to larger RCTs evaluating efficacy of large-scale public health interventions where assignment to any treatment arm is not constrained by the presence of a disease and where side effects will be reasonably negligible. For instance, several past and ongoing studies that continue to assess the efficacy of increased intake of antioxidants against arthritis (Canter et al., Reference Canter, Wider and Ernst2007), cardiovascular disease (Ashor et al., Reference Ashor, Siervo, Lara, Oggioni, Afshar and Mathers2015) and others would be well-suited instances where twin participation in opposing intervention arms could add power and improve the assessment of the true benefits of the intervention. Future clinical trials will, and should be able to, take advantage of the growing number of twin registries and the documented higher health awareness and willingness to participate in public good health research of the participants in these registries (Johnsson et al., Reference Johnsson, Helgesson, Rafnar, Halldorsdottir, Chia, Eriksson and Hansson2010).

Non-twin Registry Resources

While twin registries provide opportunities for excellent epidemiological studies, it is clear that many other studies involving twins exist outside such registries and these also play an important and key role in clinical research. These studies may involve twins typically ascertained during the course of an ongoing study into a particular disease and thereby captured by in-house registries, studies that are seeking individuals of both a sporadic and familial nature, or through acquisition of twins for a specific task. In terms of family studies, these can range from collection of sib pairs/twins through collection of multiaffected individuals in a single generation through to affected members from multigenerational families. Reflecting recruitment, such family members may be given one or multiple psychological or health or lifestyle questionnaires providing medical history, as well as clinical examination and multiple imaging modalities to capture information of the organ/tissue of interest. Additionally, multiple samples will typically be obtained ranging from DNA/RNA, plasma, serum, saliva, blood, skin, microbiome and biochemical specimens that fall under the given human research ethics for the study. While numbers of recruited twins may not be large compared to twin registries, they do tend to provide highly targeted and perhaps extreme examples of clinical interests such as partial or total disease concordance, differences in biochemical or microbiome, genetic sequence, protein or epigenetic profile within MZ twins. These indicators can provide a wealth of information that may not be clearly visible in a twin registry unless there has been a deep level of phenotype interrogation across multiple traits/diseases within the collected twins.

A recent study – The National Aeronautics and Space Administration (NASA) twin study – is a case in point (Garrett-Bakelman et al., Reference Garrett-Bakelman, Darshi, Green, Gur, Lin, Macias and Turek2019). In that twin study, an MZ twin-pair was compared, consisting of a NASA astronaut who spent a year on the International Space Station and his brother who had remained on Earth. Multiple lines of research were undertaken, including physiological, telomeric, transcriptomic, epigenetic, proteomic, metabolomic, immune, microbiomic, cardiovascular, vision-related and cognitive. Measures were collected over 25 months prior, during and post return from the mission with the aim of elucidating changes in biological measures over extended time periods in space. While a number of initial changes were noted such as a lengthening of telomeres, gene regulation (epigenetic and transcriptional data), gut microbiome composition, body weight, carotid artery dimensions, subfoveal choroidal thickness and peripapillary total retinal thickness, and serum metabolites, most of these measures rapidly returned to preflight levels on return to Earth. However, the greatest change was noted for a series of ‘space genes’ where significant gene expression changes were still disrupted after 6 months return to Earth. These involved hypoxia, mitochondrial stress, telomere length, DNA damage, DNA repair, collagen, blood clotting, bone formation and hyperactive immune activity gene pathways, as well as increased DNA damage, increased numbers of short telomeres and attenuated cognitive function (Garrett-Bakelman et al., Reference Garrett-Bakelman, Darshi, Green, Gur, Lin, Macias and Turek2019).

Looking Into the Future: Twin Registries in the Personalized Medicine Era

There is an ever-increasing desire from researchers for increased amounts of data from a diverse variety of sources from twins. These demands for data will continue to evolve, especially given the intersection of artificial intelligence, machine learning algorithms and availability of big data. As such, twin registries will have to continually evolve with these demands. For instance, the recent PREDICT 1 Study investigated personal metabolomics in response to food using a multiomics approach in over 1,000 twins and singletons from the UK and USA (Berry et al., Reference Berry, Valdes, Davies, Delahanty, Drew, Chan and Spector2019). This study took measures on glycemic, insulinemic and lipemic postprandial responses combined with genetic, metabolic, metagenomics and meal-context contributions to postprandial responses and involved the use of 110 MZ and 25 DZ twin-pairs in interim analysis of genetic contributions. These findings were further integrated with metabolic burden and gut microbiome to predict how an individual would respond to food. The sources of data and outcomes were then assessed through the use of machine-learning algorithms to indicate that 46% of overall variation in glycemic responses could be predicted from meal content and baseline characteristics rather than genetic and microbiome factors. Such findings advance our understanding that there is growing evidence to indicate that the use of personalized nutritional guidelines rather than a population-based approach may be the most appropriate route for guiding individual health.

Conclusions

Twin registries worldwide are becoming more aware of the unique contributions that they can offer in many different research areas and are increasing efforts to improve their collaborative efforts (Ferreira et al., Reference Ferreira, Oliveira, Junqueira, Cisneros, Ferreira, Murphy and Teixeira-Salmela2016). Different twin registry models exist, with one extreme being that of the research-intensive twin registry such as Twins UK where research is typically driven from within. The opposite end of the scale is that of the more classic registry such as the ATR where information on twins is housed, and the registry provides the gateway to researchers wanting to access twins for their own studies following internal ethics approval. Currently, both of these registry approaches appear to work within their own national remits, but twin repositories rely on continued funding for their ongoing success and therefore can be influenced by other external factors and therefore need to remain relevant within society. Twin studies still have a unique part to play in advancing our understanding of disease or health and typically present at the forefront of many studies in exploring the intertwining of microbiologic factors with genetic, epigenetic, metabolic, physiological, biochemical and immune functions that are needed to maintain a good homeostasis. In the future, it is likely that the vast amounts of data available on twins combined with advances in artificial intelligence/machine learning will provide many research findings and pave the way for studies in wider sections of the community.

Acknowledgements

This study was supported by the Australian National Health and Medical Research Council Senior Research Fellowship (1138585 to PN Baird).

References

Allione, A., Marcon, F., Fiorito, G., Guarrera, S., Siniscalchi, E., Zijno, A., … Matullo, G. (2015). Novel epigenetic changes unveiled by monozygotic twins discordant for smoking habits. PLoS One, 10, e0128265.CrossRefGoogle ScholarPubMed
Ashor, A. W., Siervo, M., Lara, J., Oggioni, C., Afshar, S., & Mathers, J. C. (2015). Effect of vitamin C and vitamin E supplementation on endothelial function: A systematic review and meta-analysis of randomised controlled trials. British Journal of Nutrition, 113, 11821194.CrossRefGoogle ScholarPubMed
Bahl, A., Pöllänen, E., Ismail, K., Sipilä, S., Mikkola, T. M., Berglund, E., … Ollikainen, M. (2015). Hormone Replacement Therapy Associated White Blood Cell DNA Methylation and Gene Expression are Associated With Within-Pair Differences of Body Adiposity and Bone Mass. Twin Research and Human Genetics, 18, 647661.CrossRefGoogle ScholarPubMed
Baran, Y., Subramaniam, M., Biton, A., Tukiainen, T., Tsang, E. K., Rivas, M. A., … Lappalainen, T. (2015). The landscape of genomic imprinting across diverse adult human tissues. Genome Research, 25, 927936.Google ScholarPubMed
Begemann, M., Rezwan, F. I., Beygo, J., Docherty, L. E., Kolarova, J., Schroeder, C., … Mackay, D. J. G. (2018). Maternal variants in NLRP and other maternal effect proteins are associated with multilocus imprinting disturbance in offspring. Journal of Medical Genetics, 55, 497504.CrossRefGoogle ScholarPubMed
Bernardo, J., Nowacki, A., Martin, R., Fanaroff, J. M., & Hibbs, A. M. (2015). Multiples and parents of multiples prefer same arm randomization of siblings in neonatal trials. Journal of Perinatology, 35, 310.CrossRefGoogle ScholarPubMed
Berry, S., Valdes, A., Davies, R., Delahanty, L., Drew, D., Chan, A. T., … Spector, T. (2019). Predicting personal metabolic responses to food using multi-omics machine learning in over 1000 twins and singletons from the UK and US: The PREDICT I Study (OR31-01-19). Current Developments in Nutrition, 3(Suppl. 1), nzz037.OR31-01-19.CrossRefGoogle Scholar
Bianconi, E., Piovesan, A., Facchin, F., Beraudi, A., Casadei, R., Frabetti, F., … Canaider, S. (2013). An estimation of the number of cells in the human body. Annals of Human Biology, 40, 463471.CrossRefGoogle ScholarPubMed
Blueprint Consortium. (2016). Quantitative comparison of DNA methylation assays for biomarker development and clinical applications. Nature Biotechnology, 34, 726737.CrossRefGoogle Scholar
Bonder, M. J., Kurilshikov, A., Tigchelaar, E. F., Mujagic, Z., Imhann, F., Vila, A. V., … Zhernakova, A. (2016). The effect of host genetics on the gut microbiome. Nature Genetics, 48, 14071412.CrossRefGoogle ScholarPubMed
Burbridge, D. (2001). Francis Galton on twins, heredity and social class. British Journal for the History of Science, 34, 323340.CrossRefGoogle Scholar
Busche, S., Shao, X., Caron, M., Kwan, T., Allum, F., Cheung, W. A., … Grundberg, E. (2015). Population whole-genome bisulfite sequencing across two tissues highlights the environment as the principal source of human methylome variation. Genome Biology, 16, 290308.CrossRefGoogle ScholarPubMed
Busk, T., Clemmesen, J., & Nielsen, A. (1948). Twin studies and other genetical investigations in the Danish Cancer Registry. British Journal of Cancer, 2, 156163.CrossRefGoogle ScholarPubMed
Canter, P. H., Wider, B., & Ernst, E. (2007). The antioxidant vitamins A, C, E and selenium in the treatment of arthritis: A systematic review of randomized clinical trials. Rheumatology (Oxford), 46, 12231233.CrossRefGoogle Scholar
Casey, KF, Levesque, ML, Szyf, M, Ismaylova, E, Verner, MP, Suderman, M, … Booij, L. (2017). Birth weight discordance, DNA methylation, and cortical morphology of adolescent monozygotic twins. Human Brain Mapping, 38, 20372050.CrossRefGoogle ScholarPubMed
Castellani, C. A., Melka, M. G., Gui, J. L., O’Reilly, R. L., & Singh, S M. (2015). Integration of DNA sequence and DNA methylation changes in monozygotic twin pairs discordant for schizophrenia. Schizophrenia Research, 169, 433440.CrossRefGoogle Scholar
Chen, Y. C., Sudre, G., Sharp, W., Donovan, F., Chandrasekharappa, S. C., Hansen, N., … Shaw, P. (2018). Neuroanatomic, epigenetic and genetic differences in monozygotic twins discordant for attention deficit hyperactivity disorder. Molecular Psychiatry, 23, 683690.CrossRefGoogle ScholarPubMed
Conroy, J., McGettigan, P. A., McCreary, D., Shah, N., Collins, K., Parry-Fielder, B., … King, M. D. (2014). Towards the identification of a genetic basis for Landau-Kleffner syndrome. Epilepsia, 55, 858865.CrossRefGoogle ScholarPubMed
Córdova-Palomera, A., Fatjó-Vilas, M., Palma-Gudiel, H., Blasco-Fontecilla, H., Kebir, O., & Fañanás, L. (2015). Further evidence of DEPDC7 DNA hypomethylation in depression: A study in adult twins. European Psychiatry, 30, 715718.Google ScholarPubMed
Córdova-Palomera, A., Palma-Gudiel, H., Fores-Martos, J., Tabares-Seisdedos, R., & Fananas, L. (2018). Epigenetic outlier profiles in depression: A genome-wide DNA methylation analysis of monozygotic twins. PLoS One, 13, e0207754.CrossRefGoogle ScholarPubMed
Cotillard, A., Kennedy, S. P., Kong, L. C., Prifti, E., Pons, N., Le Chatelier, E., … Ehrlich, S. D. (2013). Dietary intervention impact on gut microbial gene richness. Nature, 500, 585588.CrossRefGoogle ScholarPubMed
Dempster, E. L., Wong, C. C., Lester, K. J., Burrage, J., Gregory, A. M., Mill, J., & Eley, T. C. (2014). Genome-wide methylomic analysis of monozygotic twins discordant for adolescent depression. Biological Psychiatry, 6, 977983.CrossRefGoogle Scholar
Dyke, S. O., Cheung, W. A., Joly, Y., Ammerpohl, O., Lutsik, P., Rothstein, M. A., … Pastinen, T. (2015). Epigenome data release: A participant-centered approach to privacy protection. Genome Biology, 16, 142.CrossRefGoogle ScholarPubMed
Elboudwarej, E., Cole, M., Briggs, F. B., Fouts, A., Fain, P. R., Quach, H., … Noble, J. A. (2016). Hypomethylation within gene promoter regions and type 1 diabetes in discordant monozygotic twins. Journal of Autoimmunity, 68, 2329.CrossRefGoogle Scholar
Erny, D., Hrabe de Angelis, A. L., Jaitin, D., Wieghofer, P., Staszewski, O., David, E., … Prinz, M. (2015). Host microbiota constantly control maturation and function of microglia in the CNS. Nature Neuroscience, 18, 965977.CrossRefGoogle ScholarPubMed
Ferreira, P. H., Oliveira, V. C., Junqueira, D. R., Cisneros, L. C., Ferreira, L. C., Murphy, K., … Teixeira-Salmela, L. F. (2016). The Brazilian Twin Registry. Twin Research and Human Genetics, 19, 687691.CrossRefGoogle ScholarPubMed
Fraga, M. F., Ballestar, E., Paz, M. F., Ropero, S., Setien, F., Ballestar, M. L., … Esteller, M. (2005). Epigenetic differences arise during the lifetime of monozygotic twins. Proceedings of the National Academy of Sciences of the United States of America, 102, 1060410609.CrossRefGoogle ScholarPubMed
Garrett-Bakelman, F. E., Darshi, M., Green, S. J., Gur, R. C., Lin, L., Macias, B. R., … Turek, F. W. (2019). The NASA Twins Study: A multidimensional analysis of a year-long human spaceflight. Science, 364, eaau8650.Google ScholarPubMed
Gatz, M., Harris, J. R., Kaprio, J., McGue, M., Smith, N. L., Snieder, H., … Institute of Medicine Committee on Twins. (2015). Cohort profile: The National Academy of Sciences-National Research Council Twin Registry (NAS-NRC Twin Registry). International Journal of Epidemiology, 44, 819825.CrossRefGoogle Scholar
Gesell, A. (1942). The method of co-twin control. Science, 95, 446448.CrossRefGoogle ScholarPubMed
Gilbert, J. A., Quinn, R. A., Debelius, J., Xu, Z. Z., Morton, J., Garg, N., … Knight, R. (2016). Microbiome-wide association studies link dynamic microbial consortia to disease. Nature, 535, 94103.CrossRefGoogle ScholarPubMed
Goldberg, J., Curran, B., Vitek, M. E., Henderson, W. G., & Boyko, E. J. (2002). The Vietnam Era Twin Registry. Twin Research, 5, 476481.CrossRefGoogle ScholarPubMed
Goodrich, J. K., Davenport, E. R., Beaumont, M., Jackson, M. A., Knight, R., Ober, C., … Ley, R. E. (2016). Genetic determinants of the gut microbiome in UK twins. Cell Host & Microbe, 19, 731743.CrossRefGoogle ScholarPubMed
Goodrich, J. K., Waters, J. L., Poole, A. C., Sutter, J. L., Koren, O., Blekhman, R., … Ley, R. E. (2014). Human genetics shape the gut microbiome. Cell, 159, 789799.CrossRefGoogle ScholarPubMed
Gordon, L., Joo, J. E., Powell, J. E., Ollikainen, M., Novakovic, B., Li, X., … Saffery, R. (2012). Neonatal DNA methylation profile in human twins is specified by a complex interplay between intrauterine environmental and genetic factors, subject to tissue-specific influence. Genome Research, 22, 13951406.CrossRefGoogle ScholarPubMed
Gordon, L., Joo, J. H., Andronikos, R., Ollikainen, M., Wallace, E. M., Umstad, M. P., … Craig, J. M. (2011). Expression discordance of monozygotic twins at birth: Effect of intrauterine environment and a possible mechanism for fetal programming. Epigenetics, 6, 579592.CrossRefGoogle Scholar
Hochberg, Z., Feil, R., Constancia, M., Fraga, M., Junien, C., Carel, J. C., … Albertsson-Wikland, K. (2011). Child health, developmental plasticity, and epigenetic programming. Endocrine Reviews, 32, 159224.CrossRefGoogle ScholarPubMed
Honeyman, M. S. (1961). The Connecticut Twin Registry. Eugenics Quarterly, 8, 97.CrossRefGoogle ScholarPubMed
Hopper, J. L., Foley, D. L., White, P. A., & Pollaers, V. (2013). Australian Twin Registry: 30 years of progress. Twin Research and Human Genetics, 16, 3442.CrossRefGoogle ScholarPubMed
Huang, Y. J. (2015). The respiratory microbiome and innate immunity in asthma. Current Opinion in Pulmonary Medicine, 21, 2732.CrossRefGoogle ScholarPubMed
Hwang, J. Y., Lee, H. J., Go, M. J., Jang, H. B., Choi, N. H., Bae, J. B., … Kim, B. J. (2018). Genome-wide methylation analysis identifies ELOVL5 as an epigenetic biomarker for the risk of type 2 diabetes mellitus. Scientific Reports, 8, 14862.CrossRefGoogle Scholar
Jiao, Z., Jiang, Z., Wang, J., Xu, H., Zhang, Q., Liu, S., … Qiu, H. (2017). Whole-genome scale identification of methylation markers specific for cerebral palsy in monozygotic discordant twins. Molecular Medicine Reports, 16, 94239430.CrossRefGoogle Scholar
Johnsson, L., Helgesson, G., Rafnar, T., Halldorsdottir, I., Chia, K. S., Eriksson, S., & Hansson, M. G. (2010). Hypothetical and factual willingness to participate in biobank research. European Journal of Human Genetics, 18, 12611264.CrossRefGoogle ScholarPubMed
Jones, J. (1811). Mr. Jones’s case of twins. The Medical and Physical Journal, 25, 311313.Google Scholar
Kaati, G., Bygren, L. O., & Edvinsson, S. (2002). Cardiovascular and diabetes mortality determined by nutrition during parents’ and grandparents’ slow growth period. European Journal of Human Genetics, 10, 682688.CrossRefGoogle ScholarPubMed
Kaminsky, Z. A., Tang, T., Wang, S. C., Ptak, C., Oh, G. H., Wong, A. H., … Petronis, A. (2009). DNA methylation profiles in monozygotic and dizygotic twins. Nature Genetics, 41, 240245.CrossRefGoogle ScholarPubMed
Kaprio, J., Koskenvuo, M., & Rose, R. J. (1990). Population-based twin registries: Illustrative applications in genetic epidemiology and behavioral genetics from the Finnish Twin Cohort Study. Acta geneticae Medicae et Gemellologiae (Roma), 39, 427439.CrossRefGoogle ScholarPubMed
Kaut, O., Sharma, A., Schmitt, I., & Wüllner, U. (2017). DNA methylation of imprinted loci of autosomal chromosomes and IGF2 is not affected in Parkinson’s disease patients’ peripheral blood mononuclear cells. Neurological Research, 39, 281284.CrossRefGoogle Scholar
Kesselmeier, M., Putter, C., Volckmar, A. L., Baurecht, H., Grallert, H., Illig, T., … WTCCC3. (2018). High-throughput DNA methylation analysis in anorexia nervosa confirms TNXB hypermethylation. World Journal of Biological Psychiatry, 19, 187199.CrossRefGoogle ScholarPubMed
Klengel, T., & Binder, E. B. (2015). FKBP5 allele-specific epigenetic modification in gene by environment interaction. Neuropsychopharmacology, 40, 244246.CrossRefGoogle ScholarPubMed
Klengel, T., Mehta, D., Anacker, C., Rex-Haffner, M., Pruessner, J. C., Pariante, C. M., … Binder, E. B. (2013). Allele-specific FKBP5 DNA demethylation mediates gene-childhood trauma interactions. Nature Neuroscience, 16, 3341.CrossRefGoogle ScholarPubMed
Krueger, R. F., & Johnson, W. (2002). The Minnesota Twin Registry: Current status and future directions. Twin Research, 5, 488492.CrossRefGoogle ScholarPubMed
Le Chatelier, E., Nielsen, T., Qin, J., Prifti, E., Hildebrand, F., Falony, G., … Pedersen, O. (2013). Richness of human gut microbiome correlates with metabolic markers. Nature, 500, 541546.CrossRefGoogle ScholarPubMed
Lévesque, M. L., Casey, K. F., Szyf, M., Ismaylova, E., Ly, V., Verner, M. P., … Booij, L. (2014). Genome-wide DNA methylation variability in adolescent monozygotic twins followed since birth. Epigenetics, 9, 14101421 CrossRefGoogle ScholarPubMed
Li, M., Zou, D., Li, Z., Gao, R., Sang, J., Zhang, Y., … Zhang, Z. (2019). EWAS Atlas: A curated knowledgebase of epigenome-wide association studies. Nucleic Acids Research, 47, D983D988.CrossRefGoogle ScholarPubMed
Li, W., Zhang, D., Wang, W., Wu, Y., Mohammadnejad, A., Lund, J., … Tan, Q. (2019). DNA methylome profiling in identical twin pairs discordant for body mass index. International Journal of Obesity. doi:10.1038/s41366-019-0382-4CrossRefGoogle Scholar
Li, W., Christiansen, L., Hjelmborg, J., Baumbach, J., & Tan, Q. (2018). On the power of epigenome-wide association studies using a disease-discordant twin design. Bioinformatics, 34, 40734078.CrossRefGoogle ScholarPubMed
Liang, L., Willis-Owen, S. A. G., Laprise, C., Wong, K. C. C., Davies, G. A., Hudson, T. J., … Cookson, W. (2015). An epigenome-wide association study of total serum immunoglobulin E concentration. Nature, 520, 670674.CrossRefGoogle ScholarPubMed
Liu, D., Zhao, L., Wang, Z., Zhou, X., Fan, X., Li, Y., … Jiang, Y. (2019). EWASdb: Epigenome-wide association study database. Nucleic Acids Research, 47, D989D993.CrossRefGoogle ScholarPubMed
Lozupone, C., Hamady, M., & Knight, R. (2006). UniFrac — An online tool for comparing microbial community diversity in a phylogenetic context. BMC Bioinformatics, 7, 371.CrossRefGoogle Scholar
Malki, K., Koritskaya, E., Harris, F., Bryson, K., Herbster, M., & Tosto, M. G. (2016). Epigenetic differences in monozygotic twins discordant for major depressive disorder. Translational Psychiatry, 6, e839.CrossRefGoogle ScholarPubMed
Martino, D., Loke, Y. J., Gordon, L., Ollikainen, M., Cruickshank, M. N., Saffery, R., & Craig, J. M. (2013). Longitudinal, genome-scale analysis of DNA methylation in twins from birth to 18 months of age reveals rapid epigenetic change in early life and pair-specific effects of discordance. Genome Biology, 14, R42.CrossRefGoogle ScholarPubMed
Moayyeri, A., Hammond, C. J., Hart, D. J., & Spector, T. D. (2013). The UK Adult Twin Registry (TwinsUK Resource). Twin Research and Human Genetics, 16, 144149.CrossRefGoogle Scholar
Mohandas, N., Loke, Y. J., Hopkins, S., Mackenzie, L., Bennett, C., Berkovic, S. F., Vadlamudi, L., & Craig, J. M. (2019). Evidence for type-specific DNA methylation patterns in epilepsy: A discordant monozygotic twin approach. Epigenomics, 11, 951968.CrossRefGoogle ScholarPubMed
Murphy, T. M., Wong, C. C., Arseneault, L., Burrage, J., Macdonald, R., Hannon, E., … Mill, J. (2015). Methylomic markers of persistent childhood asthma: A longitudinal study of asthma-discordant monozygotic twins. Clinical Epigenetics, 7, 130.CrossRefGoogle ScholarPubMed
Ollikainen, M., Ismail, K., Gervin, K., Kyllönen, A., Hakkarainen, A., Lundbom, J., … Kaprio, J. (2015). Genome-wide blood DNA methylation alterations at regulatory elements and heterochromatic regions in monozygotic twins discordant for obesity and liver fat. Clinical Epigenetics, 7, 39.CrossRefGoogle ScholarPubMed
Oudejans, C., Poutsma, A., Michel, O., Mulders, J., Visser, A., van Dijk, M., … de Groot, C. J. (2016). Genome-wide identification of epigenetic hotspots potentially related to cardiovascular risk in adult women after a complicated pregnancy. PLoS One, 11, e0148313.CrossRefGoogle ScholarPubMed
Pedersen, N. L., Lichtenstein, P., & Svedberg, P. (2002). The Swedish Twin Registry in the third millennium. Twin Research, 5, 427432.CrossRefGoogle ScholarPubMed
Peng, H., Zhu, Y., Goldberg, J., Vaccarino, V., & Zhao, J. (2019). DNA methylation of five core circadian genes jointly contributes to glucose metabolism: A gene-set analysis in monozygotic twins. Frontiers in Genetics, 10, 329.CrossRefGoogle ScholarPubMed
Peng, H., Zhu, Y., Strachan, E., Fowler, E., Bacus, T., Roy-Byrne, P., … Zhao, J. (2018). Childhood trauma, DNA methylation of stress-related genes, and depression: Findings from two monozygotic twin studies. Psychosomatic Medicine, 80, 599608.CrossRefGoogle ScholarPubMed
Pietiläinen, K. H., Ismail, K., Järvinen, E., Heinonen, S., Tummers, M., Bollepalli, S., … Ollikainen, M. (2016). DNA methylation and gene expression patterns in adipose tissue differ significantly within young adult monozygotic BMI-discordant twin pairs. International Journal of Obesity, 40, 654661.CrossRefGoogle ScholarPubMed
Plomin, R., & Haworth, C. M. (2010). Genetics and intervention research. Perspectives on Psychological Science, 5, 557563.CrossRefGoogle ScholarPubMed
Qin, J., Li, R., Raes, J., Arumugam, M., Burgdorf, K. S., Manichanh, C., … Wang, J. (2010). A human gut microbial gene catalogue established by metagenomic sequencing. Nature, 464, 5965.CrossRefGoogle ScholarPubMed
Ramos, P. S., Zimmerman, K. D., Haddad, S., Langefeld, C. D., Medsger, T. A. Jr., & Feghali-Bostwick, C. A. (2019). Integrative analysis of DNA methylation in discordant twins unveils distinct architectures of systemic sclerosis subsets. Clinical Epigenetics, 11, 58.CrossRefGoogle ScholarPubMed
Riess, A., Binder, G., Ziegler, J., Begemann, M., Soellner, L., & Eggermann, T. (2016). First report on concordant monozygotic twins with Silver-Russell syndrome and ICR1 hypomethylation. European Journal of Medical Genetics, 59, 14.CrossRefGoogle ScholarPubMed
Salzberg, S. L. (2018). Open questions: How many genes do we have? BMC Biology, 16, 94.CrossRefGoogle Scholar
Schwartz, J. T. (1970). A twin register for eye studies and need for collaboration. Acta geneticae Medicae et Gemellologiae (Roma), 19, 344348.CrossRefGoogle ScholarPubMed
Shoaie, S., Ghaffari, P., Kovatcheva-Datchary, P., Mardinoglu, A., Sen, P., Pujos-Guillot, E., … Nielsen, J. (2015). Quantifying diet-induced metabolic changes of the human gut microbiome. Cell Metabolism, 22, 320331.CrossRefGoogle ScholarPubMed
Skytthe, A., Christiansen, L., Kyvik, K. O., Bodker, F. L., Hvidberg, L., Petersen, I., … Christensen, K. (2013). The Danish Twin Registry: Linking surveys, national registers, and biological information. Twin Research and Human Genetics, 16, 104111.CrossRefGoogle ScholarPubMed
Skytthe, A., Kyvik, K., Bathum, L., Holm, N., Vaupel, J. W., & Christensen, K. (2006). The Danish Twin Registry in the new millennium. Twin Research and Human Genetics, 9, 763771.CrossRefGoogle ScholarPubMed
Slack, E., Hapfelmeier, S., Stecher, B., Velykoredko, Y., Stoel, M., Lawson, M. A., … Macpherson, A. J. (2009). Innate and adaptive immunity cooperate flexibly to maintain host-microbiota mutualism. Science, 325, 617620.CrossRefGoogle ScholarPubMed
Souren, N. Y., Gerdes, L. A., Lutsik, P., Gasparoni, G., Beltrán, E., … Walter, J. (2019). DNA methylation signatures of monozygotic twins clinically discordant for multiple sclerosis. Nature Communications, 10, 2094.CrossRefGoogle ScholarPubMed
Spector, T. D., & Williams, F. M. (2006). The UK Adult Twin Registry (TwinsUK). Twin Research and Human Genetics, 9, 899906.CrossRefGoogle Scholar
Stefan, M., Zhang, W., Concepcion, E., Yi, Z., & Tomer, Y. (2014). DNA methylation profiles in type 1 diabetes twins point to strong epigenetic effects on etiology. Journal of Autoimmunity, 50, 3337.CrossRefGoogle ScholarPubMed
Stewart, J. A., Chadwick, V. S., & Murray, A. (2005). Investigations into the influence of host genetics on the predominant eubacteria in the faecal microflora of children. Journal of Medical Microbiology, 54, 12391242.CrossRefGoogle ScholarPubMed
Sumathipala, A., Yelland, L., Green, D., Shepherd, T., Jayaweera, K., Ferreira, P., & Craig, J. M. (2018). Twins as participants in randomized controlled trials: A review of published literature. Twin Research and Human Genetics, 21, 5156.CrossRefGoogle ScholarPubMed
Svendsen, A. J., Gervin, K., Lyle, R., Christiansen, L., Kyvik, K., Junker, P., … Tan, Q. (2016). Differentially methylated DNA regions in monozygotic twin pairs discordant for rheumatoid arthritis: An epigenome-wide study. Frontiers in Immunology, 7, 510.CrossRefGoogle ScholarPubMed
Tarr, I. S., McCann, E. P., Benyamin, B., Peters, T. J., Twine, N. A., Zhang, K. Y., … Williams, K. L. (2019). Monozygotic twins and triplets discordant for amyotrophic lateral sclerosis display differential methylation and gene expression. Scientific Reports, 9, 8254.CrossRefGoogle ScholarPubMed
Teschendorff, A. E., & Zheng, S. C. (2017). Cell-type deconvolution in epigenome-wide association studies: A review and recommendations. Epigenomics, 9, 757768.CrossRefGoogle ScholarPubMed
Teucher, B., Skinner, J., Skidmore, P. M., Cassidy, A., Fairweather-Tait, S. J., Hooper, L., … MacGregor, A. J. (2007). Dietary patterns and heritability of food choice in a UK female twin cohort. Twin Research and Human Genetics, 10, 734748.CrossRefGoogle Scholar
Thaiss, C. A., Zmora, N., Levy, M., & Elinav, E. (2016). The microbiome and innate immunity. Nature, 535, 6574.CrossRefGoogle ScholarPubMed
Tremaroli, V., & Backhed, F. (2012). Functional interactions between the gut microbiota and host metabolism. Nature, 489, 242249.CrossRefGoogle ScholarPubMed
Tsai, P. C., Van Dongen, J., Tan, Q., Willemsen, G., Christiansen, L., Boomsma, D. I., … Bell, J. T. (2015). DNA methylation changes in the IGF1R gene in birth weight discordant adult monozygotic twins. Twin Research and Human Genetics, 18, 635646.CrossRefGoogle ScholarPubMed
Turnbaugh, P. J., Ley, R. E., Hamady, M., Fraser-Liggett, C. M., Knight, R., & Gordon, J. I. (2007). The human microbiome project. Nature, 449, 804810.CrossRefGoogle ScholarPubMed
Turnbaugh, P. J., Quince, C., Faith, J. J., McHardy, A. C., Yatsunenko, T., Niazi, F., … Gordon, J. I. (2010). Organismal, genetic, and transcriptional variation in the deeply sequenced gut microbiomes of identical twins. Proceedings of the National Academy of Sciences of the United States of America, 107, 75037508.CrossRefGoogle ScholarPubMed
Ulff-Møller, C. J., Asmar, F., Liu, Y., Svendsen, A. J., Busato, F., Grønbaek, K., Tost, J., & Jacobsen, S. (2018). Twin DNA methylation profiling reveals flare-dependent interferon signature and B cell promoter hypermethylation in systemic lupus erythematosus. Arthritis & Rheumatology, 70, 878890.CrossRefGoogle Scholar
van Dongen, J., Nivard, M. G., Baselmans, B. M., Zilhão, N. R., Ligthart, L., BIOS Consortium, … Boomsma, D. I. (2015). Epigenome-wide association study of aggressive behavior. Twin Research and Human Genetics, 18, 686698.CrossRefGoogle ScholarPubMed
Wang, J., Van Den Berg, D., Hwang, A. E., Weisenberger, D., Triche, T., Nathwani, B. N., … Cozen, W. (2019). DNA methylation patterns of adult survivors of adolescent/young adult Hodgkin lymphoma compared to their unaffected monozygotic twin. Leukemia & Lymphoma, 60, 14291437.CrossRefGoogle ScholarPubMed
Wang, L., Zhang, J., Duan, J., Gao, X., Zhu, W., Lu, X., … Liu, J. (2014). Programming and inheritance of parental DNA methylomes in mammals. Cell, 157, 979991.CrossRefGoogle ScholarPubMed
Webster, A. P., Plant, D., Ecker, S., Zufferey, F., Bell, J. T., Feber, A., … Worthington, J. (2018). Increased DNA methylation variability in rheumatoid arthritis-discordant monozygotic twins. Genome Medicine, 10, 64.CrossRefGoogle ScholarPubMed
Wei, L., Liu, B., Tuo, J., Shen, D., Chen, P., Li, Z., … Nussenblatt, R. B. (2012). Hypomethylation of the IL17RC promoter associates with age-related macular degeneration. Cell Reports, 2, 11511158.CrossRefGoogle ScholarPubMed
Weksberg, R., Shuman, C., Caluseriu, O., Smith, A. C., Fei, Y. L., Nishikawa, J., … Squire, J. (2002). Discordant KCNQ1OT1 imprinting in sets of monozygotic twins discordant for Beckwith-Wiedemann syndrome. Human Molecular Genetics, 11, 13171325.CrossRefGoogle ScholarPubMed
Wolber, L. E., Steves, C. J., Tsai, P. C., Deloukas, P., Spector, T. D., Bell, J. T., & Williams, F. M. (2014). Epigenome-wide DNA methylation in hearing ability: New mechanisms for an old problem. PLoS One, 9, e105729.CrossRefGoogle ScholarPubMed
Wong, C. C., Meaburn, E. L., Ronald, A., Price, T. S., Jeffries, A. R., Schalkwyk, L. C., … Mill, J. (2014). Methylomic analysis of monozygotic twins discordant for autism spectrum disorder and related behavioural traits. Molecular Psychiatry, 19, 495503.CrossRefGoogle ScholarPubMed
Wong, C. C., Parsons, M. J., Lester, K. J., Burrage, J., Eley, T. C., Mill, J., … Gregory, A. M. (2015). Epigenome-Wide dna methylation analysis of monozygotic twins discordant for diurnal preference. Twin Research and Human Genetics, 18, 662669.CrossRefGoogle ScholarPubMed
Xie, H., Guo, R., Zhong, H., Feng, Q., Lan, Z., Qin, B., … Jia, H. (2016). Shotgun Metagenomics of 250 Adult twins reveals genetic and environmental impacts on the gut microbiome. Cell Systems, 3, 572584 e573.CrossRefGoogle ScholarPubMed
Yu, J. C., Khodadadi, H., & Baban, B. (2019). Innate immunity and oral microbiome: A personalized, predictive, and preventive approach to the management of oral diseases. EPMA Journal, 10, 4350.CrossRefGoogle ScholarPubMed
Yuan, W., Xia, Y., Bell, C. G., Yet, I., Ferreira, T., Ward, K. J., … Spector, T. D. (2014). An integrated epigenomic analysis for type 2 diabetes susceptibility loci in monozygotic twins. Nature Communications, 5, 5719.CrossRefGoogle ScholarPubMed
Yuan, Y. (2017). Study of global DNA methylation in monozygotic twins with cerebral palsy. Pakistan Journal of Pharmaceutical Sciences, 30, S1467S1473.Google ScholarPubMed
Zimmermann, M., Zimmermann-Kogadeeva, M., Wegmann, R., & Goodman, A. L. (2019). Mapping human microbiome drug metabolism by gut bacteria and their genes. Nature, 570, 462467.CrossRefGoogle ScholarPubMed
Zink, F., Magnusdottir, D. N., Magnusson, O. T., Walker, N. J., Morris, T. J., Sigurdsson, A., … Stefansson, K. (2018). Insights into imprinting from parent-of-origin phased methylomes and transcriptomes. Nature Genetics, 50, 15421552.CrossRefGoogle ScholarPubMed
Figure 0

Table 1. List of epigenetic studies that have used the discordant MZ twin model in the past five years