Mouse Systems Genetics As a Prelude to Precision Medicine.
Hao Li,Johan Auwerx
DOI: https://doi.org/10.1016/j.tig.2020.01.004
IF: 11.4
2020-01-01
Trends in Genetics
Abstract:The mouse is the premier model organism for human biomedical research.So far, most of the research studies involving mouse models rely on a single or few genetic backgrounds and controlled external factors that limit the generalizability of the results.Systems genetics improves the translational potential of mouse studies in human.Systems genetics approaches in mouse panels could serve as the prototype and provide valuable insights for human precision medicine. Mouse models have been instrumental in understanding human disease biology and proposing possible new treatments. The precise control of the environment and genetic composition of mice allows more rigorous observations, but limits the generalizability and translatability of the results into human applications. In the era of precision medicine, strategies using mouse models have to be revisited to effectively emulate human populations. Systems genetics is one promising paradigm that may promote the transition to novel precision medicine strategies. Here, we review the state-of-the-art resources and discuss how mouse systems genetics helps to understand human diseases and to advance the development of precision medicine, with an emphasis on the existing resources and strategies. Mouse models have been instrumental in understanding human disease biology and proposing possible new treatments. The precise control of the environment and genetic composition of mice allows more rigorous observations, but limits the generalizability and translatability of the results into human applications. In the era of precision medicine, strategies using mouse models have to be revisited to effectively emulate human populations. Systems genetics is one promising paradigm that may promote the transition to novel precision medicine strategies. Here, we review the state-of-the-art resources and discuss how mouse systems genetics helps to understand human diseases and to advance the development of precision medicine, with an emphasis on the existing resources and strategies. Most complex traits and diseases, such as height, longevity, and diabetes, are heritable and influenced by various genetic factors [1.Boyle E.A. et al.An expanded view of complex traits: from polygenic to omnigenic.Cell. 2017; 169: 1177-1186Abstract Full Text Full Text PDF PubMed Scopus (585) Google Scholar], while being modulated by environmental stimuli. Due every individual's unique genetic makeup, response to drugs [2.Roden D.M. et al.Pharmacogenomics: the genetics of variable drug responses.Circulation. 2011; 123: 1661-1670Crossref PubMed Scopus (60) Google Scholar], nutrition [3.Zeevi D. et al.Personalized nutrition by prediction of glycemic responses.Cell. 2015; 163: 1079-1094Abstract Full Text Full Text PDF PubMed Scopus (590) Google Scholar], and lifestyle [4.Buford T.W. et al.Toward exercise as personalized medicine.Sports Med. 2013; 43: 157-165Crossref PubMed Scopus (57) Google Scholar] vary considerably from person to person. This uniqueness of every human being underpins the purpose of precision medicine, which posits that disease prediction, diagnosis, and treatment for each individual is based on personal genomic variations and external environments [5.Hamburg M.A. Collins F.S. The path to personalized medicine.N. Engl. J. Med. 2010; 363: 301-304Crossref PubMed Scopus (1068) Google Scholar]. Precision medicine is an innovative approach that takes the variability in genetics, environment, and lifestyle of each individual into account in disease prevention and treatment, and provides better prediction of effective treatments, while concurrently minimizing the possibility of drug side effects [6.Collins F.S. Varmus H. A new initiative on precision medicine.N. Engl. J. Med. 2015; 372: 793-795Crossref PubMed Scopus (2184) Google Scholar]. Therefore, precision medicine requires a good understanding of the genetic bases of variation in phenotypes and their interaction with the environment in health and disease. Despite the high expectations, there are several concerns with the implementation of precision medicine [7.Joyner M.J. Paneth N. Promises, promises, and precision medicine.J. Clin. Invest. 2019; 129: 946-948Crossref PubMed Scopus (13) Google Scholar,8.Duffy D.J. Problems, challenges and promises: perspectives on precision medicine.Brief. Bioinform. 2016; 17: 494-504Crossref PubMed Scopus (36) Google Scholar]. To date, the concept of precision medicine has been successful in the context of cancer, for example, the use of trastuzumab for breast cancer that is HER2 receptor positive [9.Slamon D.J. et al.Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2.N. Engl. J. Med. 2001; 344: 783-792Crossref PubMed Scopus (8227) Google Scholar], as well as for rare diseases, for example, the use of ivacaftor for cystic fibrosis patients with mutations in the CFTR gene [10.Ramsey B.W. et al.A CFTR potentiator in patients with cystic fibrosis and the G551D mutation.N. Engl. J. Med. 2011; 365: 1663-1672Crossref PubMed Scopus (1115) Google Scholar,11.Van Goor F. et al.Rescue of CF airway epithelial cell function in vitro by a CFTR potentiator, VX-770.Proc. Natl. Acad. Sci. U. S. A. 2009; 106: 18825-18830Crossref PubMed Scopus (665) Google Scholar]. One illustrative case for a precision medicine approach in the setting of rare disease is a female individual who was diagnosed with hereditary spastic paraplegia, but all the medical evaluations had been unsuccessful. Whole exome sequencing revealed a mutation in the GCH1 gene, which was reported to be causal for a dopa-responsive dystonia. The mutation suggested that she might respond to levodopa and the patient noticed improvements after a few days of such treatment [12.Fan Z. et al.GCH1 heterozygous mutation identified by whole-exome sequencing as a treatable condition in a patient presenting with progressive spastic paraplegia.J. Neurol. 2014; 261: 622-624Crossref PubMed Scopus (9) Google Scholar]. There are still doubts that precision medicine can achieve its full potential in complex diseases [13.Shendure J. et al.Genomic medicine-progress, pitfalls, and promise.Cell. 2019; 177: 45-57Abstract Full Text Full Text PDF PubMed Scopus (5) Google Scholar,14.Aronson S.J. Rehm H.L. Building the foundation for genomics in precision medicine.Nature. 2015; 526: 336-342Crossref PubMed Scopus (182) Google Scholar]. This could be partially explained by the fact that complex diseases are influenced by the combination of genetic variants and environmental factors, whereas most rare diseases are caused by a single genetic mutation. This complexity also contributes to the difficulty of generalizing findings from human groups to individuals. Some argue that the lack of group-to-individual generalizability of the statistical measures is a threat to human subject research [15.Adolf J.K. Fried E.I. Ergodicity is sufficient but not necessary for group-to-individual generalizability.Proc. Natl. Acad. Sci. U. S. A. 2019; 116: 6540-6541Crossref PubMed Scopus (4) Google Scholar,16.Fisher A.J. et al.Lack of group-to-individual generalizability is a threat to human subjects research.Proc. Natl. Acad. Sci. U. S. A. 2018; 115: E6106-E6115Crossref PubMed Scopus (90) Google Scholar]. Several recent studies have proposed advanced statistical methods to accurately predict complex traits or disease risks (or polygenic risk scores) based on genetic variants [17.Torkamani A. et al.The personal and clinical utility of polygenic risk scores.Nat. Rev. Genet. 2018; 19: 581-590Crossref PubMed Scopus (118) Google Scholar, 18.Chatterjee N. et al.Developing and evaluating polygenic risk prediction models for stratified disease prevention.Nat. Rev. Genet. 2016; 17: 392-406Crossref PubMed Scopus (157) Google Scholar, 19.de Los Campos G. et al.Complex-trait prediction in the era of big data.Trends Genet. 2018; 34: 746-754Abstract Full Text Full Text PDF PubMed Scopus (0) Google Scholar, 20.Khera A.V. et al.Genome-wide polygenic scores for common diseases identify individuals with risk equivalent to monogenic mutations.Nat. Genet. 2018; 50: 1219-1224Crossref PubMed Scopus (283) Google Scholar], especially when traits show a high heritability (see Glossary) (e.g., height) [21.Lello L. et al.Accurate genomic prediction of human height.Genetics. 2018; 210: 477-497Crossref PubMed Scopus (0) Google Scholar]. However, confounding factors, including demographic, environmental, and other factors, such as sex and age, limit the portability of the prediction within and across human populations [22.Martin A.R. et al.Human demographic history impacts genetic risk prediction across diverse populations.Am. J. Hum. Genet. 2017; 100: 635-649Abstract Full Text Full Text PDF PubMed Scopus (188) Google Scholar, 23.Kim M.S. et al.Genetic disease risks can be misestimated across global populations.Genome Biol. 2018; 19: 179Crossref PubMed Scopus (18) Google Scholar, 24.Mostafavi H. et al.Variable prediction accuracy of polygenic scores within an ancestry group.bioRxiv. 2019; (Published online May 7, 2019. https://doi.org/10.1101/629949)Google Scholar], emphasizing the importance of the external variables in such prediction. For practical and ethical issues, model organisms have been used as simplified models for humans to study the genetic, molecular, and physiological basis of complex traits and to find therapeutic targets for human diseases. Mice have been the most studied animal models for many reasons, including their similarity in genetics, anatomy, and physiology to humans and the possibility of controlling the environmental factors (Box 1). In recent years, more and more systems genetics studies have been performed on mouse populations and have shown that mice from different genetic backgrounds exhibit distinct phenotypic responses, corroborating the principles that form the basis of precision medicine. Previously, many genetic determinants of complex traits have been identified using mouse populations and verified in human cohorts [25.Li H. et al.An integrated systems genetics and omics toolkit to probe gene function.Cell Syst. 2018; 6: 90-102Abstract Full Text Full Text PDF PubMed Scopus (11) Google Scholar,26.Wang X. et al.Joint mouse-human phenome-wide association to test gene function and disease risk.Nat. Commun. 2016; 7: 10464Crossref PubMed Scopus (52) Google Scholar]. We review here the recent developments identified in mouse systems genetics studies on complex traits and diseases, summarize the existing resources and strategies, and discuss how they may help with the implementation of personalized and precision medicine approaches.Box 1Advantages of Using Mouse Systems Genetics to Study Human Diseases1.Mice are similar to humans in many aspects, including genetics, anatomy, and physiology. The pathophysiology of disease in mice is also similar to that in humans.2.The genomes of many commonly used mouse strains have been sequenced and there are developed tools to manipulate the mouse genome and record their phenotypes.3.There are well-established mouse models for many diseases, as well as genetic reference panels for systems genetics studies.4.Mice are cost effective due to their relatively short lifespan (2–3 years) and generation time and are easy to handle and breed.5.The external environment of mouse models can be well controlled and monitored, which also facilitates the study of gene–environment interactions.6.Studies using inbred mice allow resampling isogenic individuals to replicate the same experiment or perform multiple experiments to better estimate the influence of genetics and environment on phenotypes.7.Researchers have access to all tissues in mice, especially those highly relevant in diseases, including deep tissues, which is impossible in most human studies because of ethical issues.8.Mouse models can be used to capture the disease progression stages in longitudinal studies.9.Mouse genetic populations are able to model the genetic diversity of human populations and require fewer individuals for genetic association analyses.10.Unlike human genetic studies where data should always be kept highly confidential, data from mouse studies can be made publicly available to facilitate its reanalysis to the fullest extent. 1.Mice are similar to humans in many aspects, including genetics, anatomy, and physiology. The pathophysiology of disease in mice is also similar to that in humans.2.The genomes of many commonly used mouse strains have been sequenced and there are developed tools to manipulate the mouse genome and record their phenotypes.3.There are well-established mouse models for many diseases, as well as genetic reference panels for systems genetics studies.4.Mice are cost effective due to their relatively short lifespan (2–3 years) and generation time and are easy to handle and breed.5.The external environment of mouse models can be well controlled and monitored, which also facilitates the study of gene–environment interactions.6.Studies using inbred mice allow resampling isogenic individuals to replicate the same experiment or perform multiple experiments to better estimate the influence of genetics and environment on phenotypes.7.Researchers have access to all tissues in mice, especially those highly relevant in diseases, including deep tissues, which is impossible in most human studies because of ethical issues.8.Mouse models can be used to capture the disease progression stages in longitudinal studies.9.Mouse genetic populations are able to model the genetic diversity of human populations and require fewer individuals for genetic association analyses.10.Unlike human genetic studies where data should always be kept highly confidential, data from mouse studies can be made publicly available to facilitate its reanalysis to the fullest extent. For many decades, research studies using model organisms have been conducted to guide our understanding of biological processes, with the mouse being one of the most extensively used models. In 2011, 61% of all the animals used for experimental and other scientific purposes in the EU were mice (http://eara.eu/en/animal-research/animal-research-statistics-europe/#eu-statistical-report). A number of major breakthroughs in biomedical research and a large fraction of the current therapies were developed with the help of animal models, especially mice. In fact, 94 of the 106 Nobel Prizes in physiology or medicine were awarded to research using animal models (see the complete list at www.animalresearch.info/en/medical-advances/nobel-prizes/). One notable example is the discovery of cancer immunotherapy by James P. Allison and Tasuku Honjo, who received the Nobel Prize in 2018 [27.Smyth M.J. Teng M.W. 2018 Nobel Prize in physiology or medicine.Clin. Transl. Immunol. 2018; 7e1041Crossref PubMed Scopus (5) Google Scholar]. Allison and Honjo's work would not have been possible without the extensive use of mouse tumor and immune deficiency models to uncover the immune cell mechanism of action [28.Leach D.R. et al.Enhancement of antitumor immunity by CTLA-4 blockade.Science. 1996; 271: 1734-1736Crossref PubMed Google Scholar,29.Nishimura H. et al.Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor.Immunity. 1999; 11: 141-151Abstract Full Text Full Text PDF PubMed Scopus (1571) Google Scholar]. Recently, however, there have been increasing doubts about the translational potential of findings in mouse models [30.Seok J. et al.Genomic responses in mouse models poorly mimic human inflammatory diseases.Proc. Natl. Acad. Sci. U. S. A. 2013; 110: 3507-3512Crossref PubMed Scopus (1585) Google Scholar,31.Takao K. Miyakawa T. Genomic responses in mouse models greatly mimic human inflammatory diseases.Proc. Natl. Acad. Sci. U. S. A. 2015; 112: 1167-1172Crossref PubMed Scopus (247) Google Scholar]. In another review, we argued against this opinion and demonstrated, through evidence, the contributions of mouse studies in human drug discovery and in the general understanding of human biology [32.Nadeau J.H. Auwerx J. The virtuous cycle of human genetics and mouse models in drug discovery.Nat. Rev. Drug Discov. 2019; 18: 255-272Crossref PubMed Scopus (0) Google Scholar] (Box 1). Some of the successes of mouse studies for medical applications rely simply on access to intermediate phenotypes, as collected through transcriptomic, proteomic, and metabolomic studies of deep tissues. For instance, access to both serum and liver enabled a deep molecular dissection of lysine metabolism in the BXD mouse genetic reference population (GRP), which, through mRNA expression quantitative trait locus (eQTL), protein quantitative trait locus (pQTL), and metabolite quantitative trait locus (mQTL) mapping, unequivocally established that Dhtkd1 controls the levels of 2-aminoadipate (2-AA) and, as such, influences the onset of diabetes [33.Wu Y. et al.Multilayered genetic and omics dissection of mitochondrial activity in a mouse reference population.Cell. 2014; 158: 1415-1430Abstract Full Text Full Text PDF PubMed Scopus (0) Google Scholar]. Earlier human studies established 2-AA as a biomarker of diabetes risk, but failed to causally link this observation to DHTKD1 itself in the absence of eQTL data [34.Wang T.J. et al.2-Aminoadipic acid is a biomarker for diabetes risk.J. Clin. Invest. 2013; 123: 4309-4317Crossref PubMed Scopus (171) Google Scholar]. In human studies, it is furthermore difficult and even impossible to assess critical environmental factors influencing disease development, therefore limiting the ability to study the underlying genetic determinants of complex traits and diseases, as well as the gene–environment interactions (GxE) [35.Thomas D. Gene--environment-wide association studies: emerging approaches.Nat. Rev. Genet. 2010; 11: 259-272Crossref PubMed Scopus (418) Google Scholar]. By contrast, the environment of mice can be well controlled and modulated. Key examples are the identification of the complex risk factors contributing to the development of Alzheimer's disease (AD) and Parkinson's disease [36.Dunn A.R. et al.Gene-by-environment interactions in Alzheimer's disease and Parkinson's disease.Neurosci. Biobehav. Rev. 2019; 103: 73-80Crossref PubMed Scopus (0) Google Scholar], or the GxE effects favoring the onset of asthma [37.Maazi H. et al.A GWAS approach identifies Dapp1 as a determinant of air pollution-induced airway hyperreactivity.PLoS Genet. 2019; 15e1008528Crossref PubMed Scopus (0) Google Scholar]. Another case in point is the influence of genetic and dietary effects on body weight; the heritability of body weight is over 70% in the BXD mouse GRP fed with either normal diet or a high-fat diet, but the heritability drops to around 30% when combining individuals fed with different diets, highlighting the importance of controlling external factors [38.Williams E.G. et al.Systems proteomics of liver mitochondria function.Science. 2016; 352aad0189Crossref PubMed Google Scholar]. Such a dissection of GxE factors is nearly impossible for human studies, where confounders, such as diet and environment, cannot be standardized. Another application of mouse models is to examine the influence of genome in the response to nutrients or drugs in so-called nutri- and pharmaco-genomic studies. One such example is the study of the beneficial effects of calorie restriction (CR) on lifespan using mouse individuals with different genetic backgrounds. Although CR has been considered as one of the most robust life-extending interventions, studies from mouse cohorts showed that CR was not universally beneficial for all individuals [39.Liao C.Y. et al.Genetic variation in the murine lifespan response to dietary restriction: from life extension to life shortening.Aging Cell. 2010; 9: 92-95Crossref PubMed Scopus (267) Google Scholar]. Indeed, the majority of mouse strains exhibited no lifespan extension after CR, while 1/4 of the strains even showed a reduced lifespan with CR [39.Liao C.Y. et al.Genetic variation in the murine lifespan response to dietary restriction: from life extension to life shortening.Aging Cell. 2010; 9: 92-95Crossref PubMed Scopus (267) Google Scholar]. The genes underlying the response to CR still remain to be identified. Acetaminophen overdose is the leading cause of acute liver injury in humans; however, the genetic basis of the interindividual differences in susceptibility to acetaminophen hepatotoxicity are not well understood [40.Yoon E. et al.Acetaminophen-induced hepatotoxicity: a comprehensive update.J. Clin. Transl. Hepatol. 2016; 4: 131-142PubMed Google Scholar]. A genome-wide association analysis, using a panel of 36 inbred mouse strains, identified CD44 as the candidate gene for acetaminophen-induced liver injury, which was later confirmed in humans [41.Harrill A.H. et al.Mouse population-guided resequencing reveals that variants in CD44 contribute to acetaminophen-induced liver injury in humans.Genome Res. 2009; 19: 1507-1515Crossref PubMed Scopus (0) Google Scholar]. Similar approaches have also been conducted to identify the genomic loci that influence hematotoxicity induced by chemotherapy drugs, such as doxorubicin, cyclophosphamide, or docetaxel [42.Gatti D.M. et al.Genetic background influences susceptibility to chemotherapy-induced hematotoxicity.Pharm. J. 2018; 18: 319-330Google Scholar]. Such translational mouse studies highlight the importance of genetics for precision medicine and suggest that it is worth taking the personal genetic background of patients into consideration. One major criticism against mouse models is that results from mouse experiments do not always reflect human diseases. For example, there is no single model that recapitulates the pathophysiological and molecular aspects of nonalcoholic steatohepatitis [43.Teufel A. et al.Comparison of gene expression patterns between mouse models of nonalcoholic fatty liver disease and liver tissues from patients.Gastroenterology. 2016; 151: 513-525Abstract Full Text Full Text PDF PubMed Google Scholar], making expectations of the translatability of certain mouse studies unrealistic. However, the process of finding novel and refining existing mouse models is an ongoing iterative process [44.Kleinert M. et al.Animal models of obesity and diabetes mellitus.Nat. Rev. Endocrinol. 2018; 14: 140-162Crossref PubMed Scopus (87) Google Scholar, 45.Kebede M.A. Attie A.D. Insights into obesity and diabetes at the intersection of mouse and human genetics.Trends Endocrinol. Metab. 2014; 25: 493-501Abstract Full Text Full Text PDF PubMed Google Scholar, 46.von Scheidt M. et al.Applications and limitations of mouse models for understanding human atherosclerosis.Cell Metab. 2017; 25: 248-261Abstract Full Text Full Text PDF PubMed Scopus (45) Google Scholar]. For instance, new mouse models have been recently proposed for the most common form of heart failure in humans [47.Schiattarella G.G. et al.Nitrosative stress drives heart failure with preserved ejection fraction.Nature. 2019; 568: 351-356Crossref PubMed Scopus (34) Google Scholar]. In addition, the emergence of new technologies such as CRISPR/Cas9 could unlock novel and more refined mouse models [48.Zuberi A. Lutz C. Mouse models for drug discovery. Can new tools and technology improve translational power?.ILAR J. 2016; 57: 178-185Crossref PubMed Scopus (7) Google Scholar]. Furthermore, most mouse models are built on inbred strains with a fixed genome, while individuals with different genetic backgrounds may behave and progress differently under disease conditions [49.Sittig L.J. et al.Genetic background limits generalizability of genotype-phenotype relationships.Neuron. 2016; 91: 1253-1259Abstract Full Text Full Text PDF PubMed Scopus (73) Google Scholar,50.Neuner S.M. et al.Harnessing genetic complexity to enhance translatability of Alzheimer's disease mouse models: a path toward precision medicine.Neuron. 2019; 101: 399-411Abstract Full Text Full Text PDF PubMed Scopus (0) Google Scholar]. Hence, we argue that many of the shortcomings of existing mouse models can be attributed to the extreme standardization of mouse experiments, where most research is performed in mice from one or a few genetic backgrounds, usually inbred strains. Laboratory mice housed in well-controlled and 'hygienic' cages do not experience the dynamic real-life environment as wild mice or humans [51.Beura L.K. et al.Normalizing the environment recapitulates adult human immune traits in laboratory mice.Nature. 2016; 532: 512-516Crossref PubMed Scopus (343) Google Scholar]; laboratory mice, acquiring the natural microbes of wild mice, however, model well the immune responses in humans [51.Beura L.K. et al.Normalizing the environment recapitulates adult human immune traits in laboratory mice.Nature. 2016; 532: 512-516Crossref PubMed Scopus (343) Google Scholar,52.Rosshart S.P. et al.Laboratory mice born to wild mice have natural microbiota and model human immune responses.Science. 2019; 365eaaw4361Crossref PubMed Scopus (15) Google Scholar], highlighting the importance of microbial exposure and environmental challenges of laboratory animals in biomedical research. The choice of genetic background in biomedical research is a crucial but often neglected step. However, increasing evidence shows that individuals with different genetic backgrounds may behave and progress differently in disease conditions and can even react in opposite directions to external stimuli and treatments [49.Sittig L.J. et al.Genetic background limits generalizability of genotype-phenotype relationships.Neuron. 2016; 91: 1253-1259Abstract Full Text Full Text PDF PubMed Scopus (73) Google Scholar,50.Neuner S.M. et al.Harnessing genetic complexity to enhance translatability of Alzheimer's disease mouse models: a path toward precision medicine.Neuron. 2019; 101: 399-411Abstract Full Text Full Text PDF PubMed Scopus (0) Google Scholar]. The response to morphine or cocaine [53.Philip V.M. et al.High-throughput behavioral phenotyping in the expanded panel of BXD recombinant inbred strains.Genes Brain Behav. 2010; 9: 129-159Crossref PubMed Scopus (0) Google Scholar], body weight gain upon high-fat diet [38.Williams E.G. et al.Systems proteomics of liver mitochondria function.Science. 2016; 352aad0189Crossref PubMed Google Scholar], and lifespan changes after caloric restriction [39.Liao C.Y. et al.Genetic variation in the murine lifespan response to dietary restriction: from life extension to life shortening.Aging Cell. 2010; 9: 92-95Crossref PubMed Scopus (267) Google Scholar] are just a few examples. C57BL/6J is the most extensively used mouse strain in biomedical research [54.Johnson M. Laboratory mice and rats.Mater. Methods. 2012; 2: 113Google Scholar]; however, many of the findings from C57BL/6J are not even generalizable to its substrains, like C57BL/6N, which has only 51 coding variants different from C57BL/6J [55.Fontaine D.A. Davis D.B. Attention to background strain is essential for metabolic research: C57BL/6 and the International Knockout Mouse Consortium.Diabetes. 2016; 65: 25-33Crossref PubMed Scopus (100) Google Scholar,56.Simon M.M. et al.A comparative phenotypic and genomic analysis of C57BL/6J and C57BL/6N mouse strains.Genome Biol. 2013; 14: R82Crossref PubMed Scopus (192) Google Scholar]. From a genomic standpoint, C57BL/6J carries the minor alleles for 19% of the high-impact variants among the 30 sequenced inbred mouse strains [57.Lilue J. et al.Sixteen diverse laboratory mouse reference genomes define strain-specific haplotypes and novel functional loci.Nat. Genet. 2018; 50: 1574-1583Crossref PubMed Scopus (23) Google Scholar,58.Keane T.M. et al.Mouse genomic variation and its effect on phenotypes and gene regulation.Nature. 2011; 477: 289-294Crossref PubMed Scopus (792) Google Scholar] (Figure 1A ), demonstrating that studies focusing on genes with these variants using C57BL/6J might not be well translated to most of the other strains. Furthermore, other mouse strains possess the minor allele in similar numbers of high-impact variants (Figure 1A), implying that choosing one strain over the others may lead to serious biases due to these naturally occurring variants. This would be the equivalent to studying diseases using a single human individual and then extrapolating results to the entire population. In addition, high-impact genetic mutations in some strains lead to the disruption of genes crucial in certain biological processes (Figure 1B), therefore more attention should be paid when planning animal experiments. For example, C57BL/6J is known to carry a large deletion in the Nnt gene, which associates with impaired insulin secretion and glucose tolerance [56.Simon M.M. et al.A comparative phenotypic and genomic analysis of C57BL/6J and C57BL/6N mouse strains.Genome Biol. 2013; 14: R82Crossref PubMed Scopus (192) Google Scholar]. Some strains, including the widely used C57BL/6J and BALB/cJ strains, have a 6-bp deletion of Cox7a2l causing a two amino acid truncation and its inactivation, disrupting the formation of mitochondrial supercomplexes [38.Williams E.G. et al.Systems proteomics of liver mitochondria function.Science. 2016; 352aad0189Crossref PubMed Google Scholar,59.Lapuente-Brun E. et al.Supercomplex assembly determines electron flux in the mitochondrial electron transport chain.Science. 2013; 340: 1567-1570Crossref PubMed Scopus (347) Google Scholar]. DBA/2J possesses coding and noncoding variants in Oprm1, a primary opioid receptor, and therefore exhibits weaker morphine preference and response compared with C57BL/6J [25.Li H. et al.An integrated systems genetics and omics toolkit to probe gene function.Cell Syst. 2018; 6: 90-102Abstract Full Text Full Text PDF PubMed Scopus (11) Google Scholar,60.Berrettini W.H. et al.Quantitative trait loci mapping of three loci c