Sie sind auf Seite 1von 36

0163-769X/07/$20.00/0 Printed in U.S.A.

Endocrine Reviews 28(1):48 83 Copyright 2007 by The Endocrine Society doi: 10.1210/er.2006-0035

The Genetic Landscape of Type 2 Diabetes in Mice


Susanne M. Clee and Alan D. Attie
Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706-1544
Inbred mouse strains provide genetic diversity comparable to that of the human population. Like humans, mice have a wide range of diabetes-related phenotypes. The inbred mouse strains differ in the response of their critical physiological functions, such as insulin sensitivity, insulin secretion, -cell proliferation and survival, and fuel partitioning, to diet and obesity. Most of the critical genes underlying these differences have not been identified, although many loci have been mapped. The dramatic improvements in genomic and bioinformatics resources are accelerating the pace of gene discovery. This review describes how mouse genetics can be used to discover diabetes-related genes, summarizes how the mouse strains differ in their diabetes-related phenotypes, and describes several examples of how loci identified in the mouse may directly relate to human diabetes. (Endocrine Reviews 28: 48 83, 2007)

I. Physiological Definition II. The High Prevalence of Diabetes: Implications for Genetics III. What Does Genetics Offer? IV. Advantages of Model Organisms such as the Mouse V. Origin of Inbred Mouse Strains VI. Experimental Strategies for Gene Mapping A. Qualitative vs. quantitative traits B. Backcross and intercross C. Recombinant inbred strains D. Chromosome substitution (consomic) strains E. Sensitized screens F. Outbred stocks VII. Mouse Strains Used in Diabetes Research A. C57BL/6 B. C57BLKS/J C. DBA/2 D. 129 E. BTBR F. A/J G. BALB/c H. C3H I. AKR J. CAST/Ei K. Nonobese diabetic (NOD) and nonobese nondiabetic (NON)
First Published Online October 3, 2006 Abbreviations: AIL, Advanced intercross line; BN, Brown Norway; CCK, cholecystokinin; Cpe, carboxypeptidase E; GK, Goto Kakizaki; ICR, Imperial Cancer Research; IPGTT, ip glucose tolerance test; LG, large strain; LH, Lyon hypertensive; LN, Lyon normotensive; LOD, logarithm of the odds; MODY, maturity onset diabetes of the young; NAD, nicotinamide-adenine dinucleotide; NADP, NAD phosphate; NOD, nonobese diabetic; NSY, Nagoya-Shibata-Yasuda; NZB, New Zealand Black; NZO, New Zealand obese; NZW, New Zealand White; OLETF, Otsuka Long-Evans Tokushima Fatty; QTL, quantitative trait loci; RCS, recombinant congenic strain; SDT, spontaneously diabetic Torii; SHR, spontaneously hypertensive rat; SJL, Swiss Jackson Laboratory; SM, small strain; SNP, single nucleotide polymorphism; TSOD, Tsumura, Suzuki, Obese Diabetes. Endocrine Reviews is published by The Endocrine Society (http:// www.endo-society.org), the foremost professional society serving the endocrine community.

L. New Zealand obese (NZO) M. FVB N. KK O. TallyHo P. Nagoya-Shibata-Yasuda (NSY) Q. ALR/Lt and ALS/Lt R. M16 S. LG and SM T. Tsumura, Suzuki, Obese Diabetes (TSOD) U. The Akita mouse VIII. Linkage Hot Spots A. Mouse chromosome 19, rat chromosome 1, and human chromosome 10q B. Mouse chromosome 9, rat chromosome 8, and human chromosome 11 C. Mouse chromosome 11, rat chromosome 10, and human chromosome 17p D. Mouse chromosome 12, rat chromosome 6, and human chromosome 14 E. Other type 2 diabetes susceptibility genes identified in the rat F. Hot spots in human linkage studies IX. Summary/Conclusion X. Appendix: Useful Web Site Resources for Mouse Genetics Research

I. Physiological Definition

IABETES MELLITUS IS defined by fasting hyperglycemia. In humans, fasting glucose levels of at least 126 mg/dl are the threshold for a diabetes diagnosis, with levels from 100 125 mg/dl considered impaired fasting glucose, or prediabetes (1). Impaired glucose tolerance is diagnosed in individuals with 2-h postload glucose levels exceeding 200 mg/dl (1). In the mouse, however, fasting glucose levels greater than 140 mg/dl are found in most inbred strains, ranging in males from approximately 140 to 220 mg/dl (2). Under the human criteria, nearly all inbred mice would be diabetic. This has led to discrepancies between reports as to what is considered diabetic, with no standardized criteria for the mouse. Fasting glucose exceeding 250 mg/dl is above the
48

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice

Endocrine Reviews, February 2007, 28(1):48 83 49

normal range of glucose generally observed in these animals and thus could be considered the threshold for a diagnosis of diabetes mellitus. Hyperglycemia may result from either a relative or absolute insulin deficiency. Absolute deficiency results from a loss of insulin production, e.g., the immunological destruction of -cells that occurs in type 1 diabetes. Relative deficiency, as seen in type 2 diabetes, may occur when there is an increased demand for insulin to compensate for a blunted tissue response to the hormone, termed insulin resistance. In this context, insulin production may be insufficient to maintain normoglycemia, despite a relatively robust insulin secretory response. Thus, diabetes can occur in an individual with higher than normal insulin levels. This failure of islets and/or individual -cells to maintain sufficient insulin production is an important component of type 2 diabetes in addition to insulin resistance. Thus, in our descriptions of the mouse models below, we have tried to distinguish models of insulin resistance from those that also exhibit -cell failure.

history. The thrifty gene hypothesis states that alleles that favor efficient fuel absorption and retention would experience positive selection in times of famine (23, 24). These same alleles would promote diabetes in times of nutritional excess. This hypothesis predicts that regions of the world with populations descended from those having experienced famine will experience a spike in diabetes as they adopt Westernstyle diets high in caloric density. Identification of the genetic variants underlying the world diabetes epidemic is tantamount to a genetic-sensitized screen. Millions of people are being exposed to an unprecedented level of metabolic stress in the form of caloric excess. Alleles that were hitherto phenotypically silent are now leading to diabetogenic phenotypes. Although tragic from a public health standpoint, this worldwide gene environment experiment will likely yield new insights into the key bottlenecks in pathways that maintain glucose homeostasis.

III. What Does Genetics Offer? II. The High Prevalence of Diabetes: Implications for Genetics

Type 2 diabetes is one of the most common metabolic diseases. In the United States, it afflicts over 20 million individuals, or 7% of the adult population (3), and the lifetime risk of developing diabetes is approximately 35% (4). The incidence of diabetes has increased dramatically in the past two decades, coinciding with an increase in obesity. With a lag of about a decade, we are now seeing the beginning of a far worse obesity/diabetes epidemic in Asia and South America (5), similar to what was observed as native American peoples adopted Westernized lifestyles (e.g., the Pima Indians of Arizona and New Mexico, Mexican-Americans living in Texas, and others). Approximately one half of the risk for type 2 diabetes is attributable to genetic variation. Twin studies have shown high concordance rates (80%) for monozygous twins; however the rate is not 100%, demonstrating the effects of environmental factors in the etiology of the disease (6 9). Individual phenotypes associated with type 2 diabetes, such as -cell function and insulin resistance, often have even greater heritabilities (7, 10 18). Despite the strong genetic component, however, genes with major effects do not account for the bulk of diabetes susceptibility alleles. Together, the genes underlying the rare monogenic forms of diabetes [e.g., maturity onset diabetes of the young (MODY)] account for only a small proportion of the genetic variation associated with type 2 diabetes (7, 19, 20). Instead, numerous genes each contribute a small increase in risk, although if present at high enough frequencies, they may contribute substantially to the population risk, e.g., the Pro12Ala substitution in PPARG (6, 21, 22). The rapidity of the increase in diabetes onset, coinciding with the adoption of Western lifestyles, suggests that environmental stressors are unmasking prior genetic predisposition. The high incidence of a disorder with a large genetic component suggests that diabetogenic alleles might have experienced positive selection at previous times in human

The great power of genetics resides in its ability to link genes to phenotypes, free of prior knowledge or a hypothesis about the underlying mechanism. This enables the discovery of novel genes and pathways and provides insight into points that are especially critical for resistance or susceptibility to disease. To date, dozens of candidate genes have been screened for their involvement in the development of type 2 diabetes, yet for very few has an association with the disease been detected (8, 19). Indeed, the genes that have been identified through positional cloning approaches would not have previously been thought to be candidates for diabetes. The correlation between biochemical measurements and disease suffers from the difficulty in establishing causality. A correlation can occur by chance, it can occur because of a common etiology, or there can be a true cause-effect relationship between the biochemical measure and disease. Genetics offers the power to establish unambiguous models of causality; genetic variation leads to phenotypic variation in a one-way direction. Linkage studies, demonstrating the coinheritance of a specific region of the genome along with the trait of interest, thus require analysis of related individuals and are limited in resolution primarily by the number of individuals. Cross-sectional population-based studies, where large cohorts can more easily be recruited, are necessarily correlational (i.e., showing a correlation between inheritance of a particular marker and phenotype), and do not establish direct causality. Recent advances in high-throughput single nucleotide polymorphism (SNP) genotyping have greatly increased the markers available for linkage analysis and fine-mapping and are allowing a better understanding of the haplotype structure of the genome (25). With this knowledge, it is possible to identify conserved haplotype blocks inherited from a more distant common ancestor that cosegregate with the trait of interest. As such, whole genome association studies with very dense SNP genotyping (hundreds of thousands of SNPs per individual) are being performed. This will allow interrogation of the genome at a heretofore unprecedented level

50

Endocrine Reviews, February 2007, 28(1):48 83

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice

of resolution. The assembly of very large, well-defined cohorts and collaborative efforts among multiple groups will also aid these efforts and, indeed, is already yielding success (26). These advances suggest that the pace of gene discovery in complex human disease will greatly accelerate. Several recent reviews of human diabetes genetics describe the progress to date (6 9, 19, 21, 27, 28).

IV. Advantages of Model Organisms such as the Mouse

Genetic studies in animal models offer several advantages over human studies. First, large numbers of offspring from defined crosses can be generated, allowing for linkage studies, which can greatly increase the power to detect quantitative trait loci (QTL) having minor effects on disease development. Also, because of the large number of animals that can be used in such linkage studies, traits can be mapped to a smaller interval. Once a QTL has been mapped, animal models also offer several strategies for continued fine-mapping of a mutated-gene or disease-associated allele. Once a linkage region has been identified, one is able to deconstruct a complex trait, breaking it into individual components, through the generation of congenic strains wherein a QTL region from one strain background is introgressed into the other strain background. This creates individuals that are genetically identical except at the locus of interest. For example, a type 2 diabetes-susceptible strain may have separate loci affecting insulin secretion, insulin resistance, and -cell survival. Congenics for each of these loci allow each trait to be studied independently. In addition to fine-mapping, this process can also separate multiple QTL lying under the same initial logarithm of the odds (LOD) peak. Nowhere has this been better demonstrated than with the type 1 diabetes susceptibility loci in the nonobese diabetic (NOD) mouse, or type 2 diabetes susceptibility loci in the Goto Kakizaki (GK) rat (29 32). For complex diseases like type 2 diabetes, where numerous factors, both genetic and environmental, can affect the trait being studied, the strength of the correlation between variation at a particular gene locus and a phenotype is affected by how far removed the underlying gene is from the phenotype. For example, fasting insulin levels are influenced by insulin sensitivity, insulin clearance, and insulin secretion. Insulin secretion, in turn, is affected by the total -cell mass and by the ability of individual -cells to sense and respond to glucose. Similarly, numerous processes underlie insulin resistance, and so forth. Thus, although these traits are still complex, they have less genetic heterogeneity than a broader trait, such as plasma insulin levels (14). Studies that focus on these individual components of the phenotype are more likely to detect genetic linkages than studies that merely measure fasting insulin. Mapping sensitivity and fine-mapping may also be increased by studying more detailed phenotypes that are closer to the underlying etiology of the phenotype, thereby reducing the noise from other genetic and environmental impacts on more downstream phenotypes. Extremely detailed phenotypic refinement may even lead to identification of the specific process affected

(e.g., glucose sensing vs. vesicle trafficking in models of insulin secretory defects). Animal models offer an advantage in that tissue samples are available for analysis to allow this greater phenotyping capability. In addition, phenotyping is performed in a controlled environment, reducing environmental noise. This also allows for greater flexibility in examining gene-environment (such as gene-diet) interactions on QTL. The mouse strains together contain as much genetic diversity as the human population; however, much less of this diversity is represented in the commonly used inbred strains (33). Recent SNP analysis suggests that all the commonly used strains are derived from a limited number of founder strains, setting an upper limit to the number of ancestral alleles at any given locus (34, 35). Nonetheless, the haplotype patterns across the strains are complex (36 39). The strains differ in their susceptibility to disease and in their response to environmental factors like diet. Strain background has been shown to influence most aspects of diabetes including insulin secretion, insulin resistance, and pancreatic characteristics such as -cell survival and islet number. Through the years, many spontaneous mutations have arisen and been preserved as valuable new animal models. The obese (ob) mouse with a mutation in the gene for leptin (Lep) and diabetes with a mutation in the leptin receptor (Lepr) are prominent examples (40 42). Similarly, other mutations result in spontaneous obesity and severe insulin resistance, such as the fat mutation in carboxypeptidase E (Cpe), the tubby mutation, and the ectopic expression induced by the yellow (y) allele of agouti (Ay) (43 46). However, the diabetic phenotype associated with these mutations is highly dependent upon the strain background in which they are studied (as described in the sections below). The Akita mouse represents the only spontaneous mutation in mouse models directly causing nonobese type 2 diabetes (47). We have summarized the features of this model later in this review. Only approximately 300 genes exist in humans that were not identified in the initial mouse genome assemblies (48, 49). There are large regions of synteny between the mouse and human genomes (49), thus linkages are easily translated between genomes. Under contract from the National Institutes of Health, Perlegen, Inc., is sequencing the genomes of 15 commonly used and genetically diverse mouse strains to capture the spectrum of genetic variation that exists within these strains. Additional projects are under way to genotype hundreds of thousands of SNPs in numerous strains to allow for haplotype construction and inference of genotypes of strains other than those being sequenced (35). The Mouse Phenome Database contains quantitative phenotype information for most of the commonly used mouse strains, with the ability to correlate these phenotypes with the available SNP data (50, 51). These valuable resources will greatly aid positional cloning projects in the mouse. A listing of these useful web sites is provided in an appendix to this review.

V. Origin of Inbred Mouse Strains

The development of agriculture around 10,000 yr ago was a windfall for wild mice. It provided them with an unprec-

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice

Endocrine Reviews, February 2007, 28(1):48 83 51

edented supply of food and shelter, beginning a long period of coexistence with human populations. Breeding of fancy mice was popular for centuries in China and Japan and became popular in Europe during the 19th century, leading to a trade in fancy mice at clubs, shows, and competitions. In the United States, Abbie Lathrop, a schoolteacher, began selling inbred mice in Granby, Massachusetts. Her mice were derived from the Asian musculus and European domesticus subspecies, which had evolved separately for over a million years. William Castle, a Harvard geneticist, purchased mice from Lathrop in 1902, thus collecting some of the first mouse strains still in use today. Castles student, Clarence Cook Little, carried out brother-sister mating to create inbred strains, i.e., mice that are genetically identical to one another. He created the first inbred strain, DBA, in 1909 (52). These strains have formed the foundation of mouse genetics. In 1929, with funding from the auto industrys Edsel Ford and Roscoe Jackson, Little founded The Jackson Laboratory in Bar Harbor, Maine, which today houses more than 2,000 inbred mouse strains. Most of the strains in use today derived from four subspecies of the house mouse (Mus musculus) that diverged approximately 1 million years ago: the domesticus subspecies that evolved in Western Europe; the musculus subspecies that evolved in Eastern Europe, Russia, and Northern China; the castaneus subspecies that evolved in Southeast Asia and Southern China; and the molossinus subspecies, which is a hybrid of the castaneus and musculus subspecies, from Japan (34, 53). The genealogies of the commonly used mouse strains have been described, and more recently, detailed strain haplotypes have been inferred from SNP analysis (33, 34, 36 38, 54 57). This has confirmed that the majority of inbred strains arose from a limited number of founders, primarily of the domesticus and musculus subspecies (34), and has also demonstrated the complex haplotype patterns that exist across strains (39, 57, 58). Each strain bears a unique combination of disease susceptibility and resistance loci, with the phenotype observed in each strain due to the composite effects of all these loci. Strains that are not diabetic may harbor diabetes-promoting alleles that are masked in an otherwise protective background, but the effects of these alleles may be observed when placed in the context of other diabetogenic genes. Thus, genetic analysis in animals whose genomes have been combined can reveal diabetogenic alleles contributed by the nondiabetic parental strain. Phenotypic differences across these strains exist for the majority, if not all, phenotypes that have been assessed, and this wealth of diversity has enabled genetic mapping of qualitative and quantitative trait loci controlling these differences. In Section VI, we describe the genetic and genomic methods that can lead to gene discovery. In Section VII, we summarize the phenotypes of the mouse strains used in diabetes research and the linkages that have been described in these models. We identify examples where specific genes related to diabetes have been identified. We have avoided mention of the numerous insights that have been obtained through transgenic and knockout models, because these have been reviewed extensively elsewhere (59 62), and we only refer to them where strain has been shown to modify the phenotype.

VI. Experimental Strategies for Gene Mapping A. Qualitative vs. quantitative traits

Mapping gene loci requires the identification of two animal groups that differ in the trait of interest. The genomes of these two samples are mixed, either intentionally or through the course of history, resulting in progeny that contain different combinations of alleles from each. In its simplest form, gene mapping involves the correlation of phenotype with genotype at markers that differ between the groups of individuals under study. This can be an association study in an outbred population, a linkage study in families, or an experimental cross. In all cases, alleles segregate and the question being asked is, does phenotype segregate with genotype at any locus? When a trait is scored in a binary fashion (affected vs. unaffected), then every new individual is informative and helps to narrow the position of the gene responsible for the trait. In an experimental cross for a dichotomous trait, the mapping resolution is simply a function of the sample size. But, for a quantitative trait like blood glucose or insulin, one cannot score animals as affected or unaffected because their values fall on a continuum. The mapping resolution is largely limited by the effect size of the locus (63). QTL often have small effect sizes. Thus, even with infinitely dense markers, mapping resolution is still limited for most quantitative traits. This helps us to understand why only about 1% of genes underlying the more than 2,000 rodent QTL have been identified (64).
B. Backcross and intercross

In mouse genetics, the most common method for gene mapping is to breed together the two inbred strains in which a phenotypic difference has been detected. These F1 offspring are then bred to one or both of the parental stains (backcross) or to each other (intercross) to generate an F2 sample (Fig. 1). Each time an F1 animal makes gametes, it generates recombinant chromosomes, with each gamete containing a unique distribution of the parental alleles. Thus, a backcross generates mice with discernible products of recombination on the haploid set of chromosomes inherited from the F1 parent, whereas in an intercross between two F1 animals, one can find recombinants on each transmitted chromosomal strand. The offspring of a backcross are either homozygous for one parental genotype or heterozygous. Thus, only alleles acting in a dominant fashion can be detected. In contrast, an F2 has all three possible genotypes at any locus, i.e., homozygotes for each parental allele and the heterozygote, and thus provides information on additivity and dominance. To obtain a picture of all the QTL, an F2 has a higher degree of power, requiring fewer individuals. However, for major dominant QTL, a backcross may have more power (65). Mapping resolution is limited by the number of recombination events, although with more recombination a greater marker density is needed to detect QTL. In an F2 sample, resolution is typically increased by increasing the number of F2 mice studied. Another way is by increasing the recombination within the sample, such as by intercrossing for ad-

52

Endocrine Reviews, February 2007, 28(1):48 83

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice

FIG. 1. The generation of an F2 intercross. Mice from two different parental strains (illustrated by single black and white chromosome pairs) are bred together (outcrossed) to produce F1 offspring that inherit a chromosome from parental strain (illustrated as a pair of chromosomes one of which is black and the other white). These mice are heterozygous at all loci. During gametogenesis, recombination between the chromosomal pairs occurs. In the F1 offspring, these recombination events are masked, because the chromosomal pairs within each parental strain were identical (both black or both white). However, when F1 animals are bred to each other (intercrossed) to produce the F2 generation, recombinant chromosomes are observed. Each chromosome in the gametes produced by the F1 is the product of a unique recombination event between the black and white chromosomes, resulting in an uneven mixture of chromosomal segments derived partially from one parental strain and partially from the other [part black and white, with distributions depending on the location of the recombination(s)]. Each resulting F2 offspring thus has a unique collection of recombinations on each of its chromosomes and can be heterozygous or homozygous for either parental allele at each locus.

FIG. 2. The generation of recombinant inbred strains. Pairs of F2 mice are chosen at random, separated from the other F2 mice, and mated. The offspring of each of these pairs are brother-sister mated for several generations. As inbreeding continues, at each generation the likelihood of being homozygous at any given location increases. By the 20th generation, each line is considered fully inbred. Because each pair of F2 mice had a unique combination of parental genotypes, each new line produced represents a unique, but fixed, mosaic of the two parental genomes.

ditional generations. Although it is a slower process, mapping accuracy is increased (65). Given the same sample size and QTL effect size, it has been estimated that the resolution using advanced intercross lines (AILs) where interbreeding has been carried out to the F10 generation increases approximately 5-fold from the corresponding F2 sample (66). This technique has been applied in the detection of QTL affecting glucose and insulin levels in F16 AILs (67). The approach was also successful in resolving multiple linked QTL affecting high-density lipoprotein-cholesterol concentrations into separate QTL (68). Another strategy is to use additional strains in the founding crosses. For example, Harper et al. (69) have recently used a heterogeneous F2 derived from (BALB/c C57BL/6) F1 mice mated to (C3H DBA/2) F1 mice in a mapping study for levels of hormones related to diabetes.
C. Recombinant inbred strains

One disadvantage of backcross and F2 samples is that each test animal is unique; thus there is no possibility of biological replication. Recombinant inbred (RI) strains are produced by brother-sister mating of pairs of F2 mice and their offspring (Fig. 2). After 20 generations, each new line of animals is essentially inbred and has fixed in their genomes a unique mosaic of alleles from the two parental strains. Unlike an F2 sample, a RI panel is homozygous at all loci. RI strains were first used in genetic mapping in 1971 (52). Owing largely to the efforts of Benjamin Taylor, The Jackson Laboratory created several panels of RI strains from widely used strains differing in susceptibility to various diseases and infections. Mapping a trait simply requires scoring that trait in the panel of strains and comparing the strain distribution pattern of the

trait with that of the marker genotypes. The panel need only be genotyped once, but because each strain is propagated, animals can be phenotyped on multiple occasions across time and even institutions. This allows all data obtained on an RI panel to be correlated to identify common loci affecting multiple phenotypes. Mapping resolution is limited by the number of recombination events contained within the panel (recombination within each strain and number of strains). However, for several panels, the number of strains is now large enough to permit high resolution mapping (70, 71). Furthermore, in light of the genome data and numerous available markers, concerted efforts are being made to fine-map the existing panels (72). A massively ambitious project, the Collaborative Cross, will begin with eight parental strains and produce 1,000 RI strains (73). Theoretically, with these strains, a locus exerting a modest effect could be detected and mapped with approximately 4 cM resolution (64). A second design that maintains the reproducibility of RI strains but creates more genetic diversity is the RI intercross, which creates F1 animals from each pair of RI strains in the panel (74). Finally, additional backcrossing before inbreeding generates recombinant congenic strains (RCS) in which each strain carries randomly distributed congenic segments of donor genome, inversely proportional in amount to the number of backcrosses performed. For example, each strain from an N3 backcross (an F1 backcrossed twice), will have a random distribution of roughly 12.5% of the donor genome in the recipient background. Several RCS panels have been created, including a reciprocal set between the commonly used A/J and C57BL/6 strains (75).

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice

Endocrine Reviews, February 2007, 28(1):48 83 53

D. Chromosome substitution (consomic) strains

Chromosome substitution strains (CSS) are created by introgressing an entire chromosome from a donor strain into a background strain. Nadeau and colleagues (76) created a panel of 22 strains that are genetically C57BL/6, except that each contains a single chromosome derived from the A/J strain, which differs from the C57BL/6 strain in many traits. A parallel effort is under way to produce two panels, in which the chromosomes of the Brown Norway strain are each being introgressed into either the Dahl salt-sensitive or the Fawn Hooded Hypertensive rat (77). Lusis and colleagues (78) have created a panel in which various large segments (not entire chromosomes, but overlapping large segments that span chromosomes) of DBA/2 have been introgressed into the C57BL/6 background and have shown that these strains differ in plasma glucose and insulin levels. Genome-tagged mice, in which overlapping segments spanning the entire genome of one strain are introgressed into a different strain have also been created in which DBA/2 or CAST/Ei segments have been introgressed into the C57BL/6 background (79). The power to detect QTL is higher in CSS panels than in an F2 intercross because there is less genetic variance introduced by background loci and because fewer markers need to be tested (80). However, epistatic interactions between donor-derived loci on different chromosomes cannot be detected, and extensive fine-mapping of the resultant loci is required to identify the underlying genetic variant(s).
E. Sensitized screens

FIG. 3. Outbred stock. Genetically diverse mice (e.g., several inbred strains or an inbred strain and outbred stock) are bred to produce F1 mice. Breeding at each generation continues, such that the probability of a locus being homozygous (identical by descent) increases by no more that 1% per generation. In practice, this can be achieved through the random mating of many (100) breeding pairs. Schemes utilizing fewer breeding pairs have also been developed to purposefully avoid mating related animals. We have illustrated such a scheme here. For simplicity we show only three breeding pairs, although in practice this would be carried out with a large number of breeding pairs (25). Maximum genetic diversity is maintained if each breeding male and female contributes a single male and female to the next breeding generation.

the added advantage of knowledge of the haplotype structure of the ancestral strains. An elegant variation of the association study is to cross outbred animals to an inbred strain to create an outbred-inbred F2 (83). Mapping can then be done in two steps, first at low-resolution with a small number of markers, then at higher resolution in selected regions of the genome showing evidence of linkage to the trait. Recent success with this approach has come in the positional cloning of a QTL for anxiety (84). For further details of these experimental strategies and the complexities of fine-mapping that result from each design, we refer the reader to recent reviews of this area (64, 65, 85).
VII. Mouse Strains Used in Diabetes Research

A sensitized screen is one in which the animals are rendered more sensitive to a stress by some prior manipulation, and then the response of the animals to the stressor is mapped. In diabetes, this is commonly performed by feeding normally non- or mildly insulin resistant animals high-fat or high-carbohydrate diets, or by studying the response of the animals to a genetic defect that induces insulin resistance, to identify alleles that modify this phenotype. The most commonly used models have been the monogenic obesity mutations, obese (ob) and diabetes (db) in the leptin (Lep) and leptin receptor (Lepr) genes, respectively. The classical studies of Coleman and Hummel (81) were the first to show that strain background affects diabetes susceptibility in mice.
F. Outbred stocks

A powerful alternative to crosses between inbred strains is the generation of outbred stocks (Fig. 3). In analogy to human populations, many generations of outbreeding yield a mosaic that is substantially more genetically fragmented than an F2 intercross. One set, consisting of eight founder strains, was bred for more than 60 generations and now has an average recombination distance of just 1.7 cM (82). An association study can be performed on a sample of 12,000 mice derived from this stock and have a power to detect a QTL with a resolution of 0.51.0 cM. These studies require dense genotyping, but this is now being facilitated by various automation schemes and genomewide SNP panels. This is similar to an association study in a human population, with

The Jackson Laboratory maintains web sites with a wealth of information on the various mouse strains. The Mouse Genome Informatics (MGI) site (www.informatics.jax.org) has information on the history of the strains, descriptions of their susceptibilities to various diseases, marker information for mapping, etc. The Mouse Phenome Database (MPD; www. jax.org/phenome) (50, 51) is systematically compiling phenotypic measurements across numerous mouse strains. For example, the glucose and insulin responses to high-fat feeding for a panel of 43 strains are available (2). This site is also linked to genomewide SNP genotype data for the strains, allowing for correlation of genetic markers with the phenotypic differences between strains. On a genomewide scale, this allows the possibility of haplotype mapping and mapping traits in silico (56, 86, 87).
A. C57BL/6

The C57BL/6 strain was created by Little in 1921 from a mating of female no. 57 and male no. 52 from Lathrops stock. SNP analysis indicates that the C57BL/6 strain shares some Mus musculus molossinus genomic regions and is somewhat removed evolutionarily from the other inbred strains, except the other C57-derived strains and C58 (36, 37, 88). The C57BL/6 strain is the most widely used of all inbred strains, accounting for some 14% of all such uses. Several RI panels

54

Endocrine Reviews, February 2007, 28(1):48 83

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice

have been generated with C57BL/6 as one strain (72). C57BL/6 is the most commonly used reference strain in studies of diabetes. There is some confusion about whether or not one should consider the C57BL/6 strain to be diabetes-susceptible or diabetes-resistant. Both protective and diabetes susceptibility alleles/phenotypes have been observed in this strain. Thus, the phenotype is largely relative to the strain it is being compared with and may depend on the criteria by which diabetes has been defined. C57BL/6 may be best described as a strain of intermediate susceptibility. Indeed, data from the Mouse Phenome Database shows that glucose and insulin levels of this strain are intermediate to those of the other strains (2). Several studies have provided evidence of increased diabetes susceptibility in the C57BL/6 strain. In parallel to studies of diet-induced atherosclerosis susceptibility, the C57BL/6 mouse emerged as susceptible to diet-induced obesity and hyperglycemia (89 92). Studies from the Surwit laboratory showed that a diet enriched with fat and carbohydrate (mainly from disaccharides) produced a roughly 50% increase in fasting plasma glucose and a 10.4-fold increase in plasma insulin in C57BL/6 mice (91). By contrast, glucose remained nearly unaltered, and insulin rose only 2.6-fold in A/J mice, demonstrating enhanced diet-induced insulin resistance and hyperglycemia in C57BL/6. Kaku et al. (93) found C57BL/6 to be the least glucose tolerant of six strains fed a normal chow diet. More recently, Goren et al. (94) compared glucose and insulin tolerance of C57BL/6, C57BLKS/J, DBA/2, and 129X1. Consistent with the aforementioned study of Kaku et al. (93), C57BL/6 was inherently less glucose tolerant in a glucose tolerance test. But, in an insulin tolerance test, the two C57 strains were more sensitive to the exogenously added insulin than DBA/2 or 129X1, suggesting that a defect in insulin secretion is the likely cause of the glucose intolerance of the C57BL/6 mice. An in vivo study that assessed insulin sensitivity and insulin secretion during the early and late stages of feeding a high-fat diet to C57BL/6 mice concluded that after longer-term feeding, although total insulin secretion was sufficient to compensate for insulin resistance, defective acute insulin secretion in response to glucose is a major contributor to glucose intolerance (95). Other investigators have also identified an insulin secretion defect in the C57BL/6 strain compared with other strains (93, 96 99). In perifusion experiments, islets from high-fat, high-simple carbohydrate diet-fed C57BL/6 mice had markedly impaired secretion of insulin in response to high (27.7 mm) glucose compared with A/J (100, 101). This was primarily attributed to a defect in the second-phase insulin secretion. Furthermore, C57BL/6 islets have been shown to be unresponsive to intermediate glucose levels (10 mm), due to incomplete closure of their KATP channels (98). These studies are consistent with the C57BL/6 mouse having a -cell defect in glucose responsiveness, which can, to a certain point in vivo, be offset through the generation of sufficient -cell mass. In vivo, -cells from this mouse strain have very high proliferative capacity (102). A study by Bock and co-workers surveyed the pancreatic architecture of several strains. In comparison to the others, lean C57BL/6 have

a relatively low total islet mass and -cell mass when expressed per body mass. But, C57BL/6 had the highest number of islets, albeit the smallest islets, consistent with a large foundation from which -cell mass can expand when stressed (103). However, this relatively small existing -cell pool may also contribute to the observations that C57BL/6 is more sensitive than other strains of mice to the -cell destructive effects of streptozotocin (104, 105). To identify genes involved in the glucose intolerance of the C57BL/6 strain, Toye et al. (98) created an F2 intercross from C57BL/6 and C3H/HeH. The mice were phenotyped for their response to glucose and insulin tolerance tests. Fasting insulin, postglucose insulin, and postinsulin glucose linked to distinct loci, confirming that variation in each of these parameters has a genetically distinct cause. A QTL on chromosome 13 that was linked to postglucose insulin levels contains the nicotinamide nucleotide transhydrogenase (Nnt) gene. Sequencing of the cDNA for this gene revealed a M35T polymorphism within the mitochondrial leader sequence. The C57BL/6 allele also has an in-frame deletion of exons 6 through 11, leading to a transcript of approximately 508 instead of approximately 1,261 bp. The gene is expressed at a much lower level in C57BL/6 than in C3H/HeH islets. The syntenic region to the mouse Nnt locus on human chromosome 5p15 is one of three loci linked with a genetic modifier of age of onset of diabetes in families with HNF1 (MODY3) mutations (106). Nnt is an inner mitochondrial membrane enzyme that catalyzes the exchange of reducing equivalents between nicotinamide-adenine dinucleotide (NAD) and NAD phosphate (NADP). The reaction is coupled to the transport of protons from the cytosol to the mitochondrial matrix: NADH NADP Hcytosolic 7 NAD NADPH Hmatrix. Together with the QTL mapping study, a very strong case implicates Nnt as the gene responsible for impaired insulin secretion in the C57BL/6 mouse. Knockdown of Nnt expression in the Min6 insulinoma cell line abolished the insulin secretion normally observed in response to 10 mm glucose (107). The same group screened an N-ethyl-N-nitrosourea (ENU)-mutagenized DNA archive and found gametes from two mutant mice with different Nnt nonconservative missense mutations, one in the NAD binding domain and the other in a transmembrane domain. Mice expressing either of these mutant alleles had impaired glucose tolerance, and islets from these mice had impaired glucose-stimulated insulin secretion. In short, the mutant mice reproduced the insulin secretion phenotype of the C57BL/6 mouse. Finally, transgenic expression of the 129S6/SvEvTac allele of the Nnt gene rescued the insulin secretion phenotype of the C57BL/6 mouse (108). It is noteworthy that this gene was identified and implicated in insulin secretion through genetic linkage rather than a biochemical hypothesis. How would loss of Nnt function reduce insulin secretion in response to glucose? Islets harboring the mutant Nnt allele produce a much higher amount of reactive oxygen species in response to 20 mm glucose (107), supporting the prior hypothesis that Nnt detoxifies reactive oxygen species (109). Because reactive oxygen species have been shown to increase uncoupling of oxidative phosphorylation, Nnt would be predicted to indirectly affect mito-

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice

Endocrine Reviews, February 2007, 28(1):48 83 55

chondrial ATP yield and thus the extent of the closure of the ATP-sensitive K channels. This prediction is consistent with the reduced glucose utilization seen in islets from the mutant mice (107). Another way to metabolically stress mice to find strain differences in diabetes susceptibility is to induce insulin resistance by introgressing null alleles in genes required for insulin signaling. When C57BL/6, DBA/2, and 129X1/Sv mice were created that were double heterozygotes for null alleles at the insulin receptor and insulin receptor substrate loci, all developed severe insulin resistance, as expected (110). The C57BL/6 mice developed glucose intolerance and dramatic hyperinsulinemia, which correlated with a striking 22-fold increase in -cell mass. The DBA/2 mice also developed hyperinsulinemia, but did not have as severe glucose intolerance or as strong a -cell proliferative response as the C57BL/6 mice. Thus, it was the C57BL/6 mice that had a much higher prevalence of diabetes. Despite the decreased insulin secretion and glucose intolerance of the C57BL/6 strain, C57BL/6 mice are relatively resistant to obesity-induced diabetes when compared with several other strains, as described below. This was revealed in the classic experiments of Douglas Coleman conducted over 30 yr ago. His experiments were the first to show that strain background affected the susceptibility of mice to obesity-induced diabetes. Coleman found that genetically obese mice (due to homozygosity for the ob mutation in the leptin gene; Lepob/ob) in the C57BLKS/J strain background are severely diabetic, whereas genetic obesity in the C57BL/6 background was associated with severe insulin resistance, but not the development of diabetes (111). In the C57BL/6 background, homozygosity for the Lepob mutation led to moderate (220 mg/dl), transient hyperglycemia between 8 and 12 wk of age, but then glucose returned to near normal levels (81). This return to normal glucose levels is due to a marked increase in insulin arising from a dramatic expansion of -cell number in the C57BL/6 mice that is not sustained in the C57BLKS/J background (112), consistent with the aforementioned high -cell proliferative capacity of C57BL/6 mice. C57BL/6 mice are also relatively resistant to the development of diabetic nephropathy (113).
B. C57BLKS/J

Lean C57BLKS/J are very sensitive to streptozotocin (117). C57BLKS/J mice have an approximate 50% reduction in -cell proliferative capacity compared with C57BL/6, and an increased -cell susceptibility to glucotoxicity (118, 119). Mean islet area is increased in male C57BLKS/J mice, but total pancreatic insulin content is reduced compared with C57BL/6 (119). Mapping studies have been performed in C57BL/6 C57BLKS/J Leprdb/db F2 mice. Coleman found concordance between the segregation of a malic enzyme regulatory locus on chromosome 12 and diabetes in a Leprdb/db F2 between C57BL/6 and C57BLKS/J, with the low activity allele associated with diabetes (111, 120). More recently, Paigen and colleagues (121) identified additional suggestive loci for increased glucose levels from C57BLKS/J alleles. A mapping study on C57BLKS/J DBA/2 Leprdb/db F2 mice identified a locus on chromosome 9 associated with impaired glucose tolerance (122). The DBA/2 allele at this locus contributes to the increased glucose levels, suggesting that the C57BLKS/J allele is a C57BL/6-derived protective modifier and that the DBA/2 strain has diabetogenic loci in addition to those introgressed into the C57BLKS/J strain. It is unclear, however, whether the effects of these alleles require the absence of effective leptin signaling. When the fat mutation in carboxypeptidase E (Cpe), which was originally identified on the HRS strain background, was introgressed onto the C57BLKS/J background, C57BLKS/J-Cpefat/fat males became diabetic despite reduced adiposity, whereas HRSCpefat/fat mice were more hyperinsulinemic and did not develop hyperglycemia (123). However, C57BLKS/J-Cpefat/fat mice were more insulin sensitive and showed less -cell/islet damage than C57BLKS/J-Leprdb/db mice (124). In fact, the hyperglycemia reached a plateau and eventually reversed in these animals, suggesting that the severe diabetes in C57BLKS/J-Leprdb/db mice results from an interaction between C57BLKS/J alleles and the Leprdb mutation. This could in part explain why high-fat, high-sucrose feeding showed C57BLKS/J to have a reduced response in terms of weight gain and glucose and insulin levels compared with C57BL/6 (125). A recent study in C57BLKS/J DBA/2 F2 mice, either Leprdb/db or Leprdb/, has shown that regulation of glucose levels is subject to complex interactions between sex and Leprdb genotype and epistatic interactions between loci (126).
C. DBA/2

The C57BLKS/J strain arose through the inadvertent genetic contamination of C57BL/6 (114). Although the nonC57BL/6 alleles are scattered across all the chromosomes and are primarily from the DBA/2 strain, recent SNP analysis indicates that there are contributions from at least three other strains, including C57BL/10, 129, and another unidentified strain that accounts for nearly 10% of the genome (36, 115, 116). As described above, the diabetes severity associated with homozygosity for the Lepob or Leprdb mutations is much greater on the C57BLKS/J than the C57BL/6 background (81). Because the genomic constitution of this strain derives primarily from C57BL/6 and this strain is more diabetesprone than C57BL/6, we can presume that the diabetogenic alleles are derived from the contaminating strains or perhaps the interaction between these alleles and diabetogenic C57BL/6 alleles.

The DBA strain was the first inbred strain created by Little, in 1909, deriving its name from its coat-color alleles dilute (d), brown (b), and non-agouti (a). Of the two substrains, it is the DBA/2 substrain that is commonly used in diabetes research. Given the strain differences between C57BLKS/J and C57BL/6 in diabetes susceptibility and the knowledge that C57BLKS/J and C57BL/6 differ genetically primarily due to the introgression of DBA/2-like alleles, this strain has been used as a model for genetic studies as well. Like C57BLKS/J-Leprdb/db, DBA-Leprdb/db mice become severely diabetic (111). DBA/2 Lepob/ob mice are also severely diabetic; however, unlike C57BLKS/J obese mice, not all the mice undergo pancreatic decompensation (127). Because the C57BLKS/J strain is primarily a combination of the C57BL/6

56

Endocrine Reviews, February 2007, 28(1):48 83

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice

and DBA/2 genomes, this suggests that either non-DBA/2 non-C57BL/6 alleles in C57BLKS/J or interactions between DBA/2 and C57BL/6 alleles promote the severe decompensation seen in the C57BLKS/J strain. However, as described above, there are also likely additional diabetogenic alleles in the DBA/2 strain not present in C57BLKS/J (122, 126). The DBA/2 strain is also susceptible to diabetic nephropathy (113). In contrast to genetic obesity, lean DBA/2 mice that are fed a high-fat diet are more glucose tolerant than C57BL/6 (98). In a survey of nine inbred strains, DBA/2 had the lowest glucose levels on high-fat feeding (92), and data from the Mouse Phenome Database show that DBA/2 mice have among the highest insulin levels (2). When insulin resistance is induced through double heterozygosity for insulin receptor (Insr) and insulin receptor substrate-1 (Irs1) knockout alleles, DBA/2 mice have reduced insulin resistance and a slower onset of hyperglycemia than C57BL/6 (110). DBA/2 mice have higher insulin levels in the fed state than C57BL/6 and other strains (93), which suggests that DBA/2 may have increased insulin secretion. Indeed, compared with C57BL/6, DBA/2 hypersecrete insulin in response to glucose from birth, which has been attributed to increased islet glucose utilization and GLUT2 levels (97). In a survey of pancreatic structure across seven mouse strains, DBA/2 also had the most islet and -cell mass of all the strains (103). Combined, these data suggest that DBA/2 -cells are acutely sensitive to obesity. The DBA/2 strain is genetically distinct from the other strains used in diabetes research (36, 37) and may provide important insight into factors underlying obesity-induced -cell failure. A set of very large insert congenic strains spanning the genome in which DBA/2 alleles have been introgressed into the C57BL/6 background have been developed (78) and will be an excellent resource for mapping these differences.
D. 129

The 129 strain has a complicated breeding history, which has resulted in numerous distinct substrains in existence today. Outcrossing among these substrains has led to several genetic differences between them, which have been extensively characterized, and a standardized nomenclature has been developed (128, 129). The majority of ES cell lines used in generating gene-targeted mice are derived from substrains of the 129/Sv strain, a heterogeneous strain which is the ancestor of many of todays substrains (128, 129). Thus, most knockouts are congenic strains in which the targeted allele and some amount of flanking sequence have been introgressed into other strain backgrounds. Depending on the amount of remaining flanking sequence, it is possible that nearby 129/Sv-derived alleles may contribute to the observed phenotype. For simplicity, here we refer to all substrains as 129, however it should be noted that the phenotypic effects may differ between the various substrains. On both chow and high-fat-diet feeding, the 129 strain maintains low insulin and is more glucose tolerant than other strains (2, 130, 131). This suggests that 129 mice have increased insulin sensitivity and is consistent with the observation that wild-type 129 mice have low -cell mass (103,

110). In a C57BL/6x 129 F2 study, five loci for hyperglycemia or hyperinsulinemia have been identified (130). At each of these loci, the 129 allele was associated with the lower insulin or glucose phenotype. Thus, the 129 strain is diabetes-resistant in the context of genetic obesity. Alleles from the 129 strain are capable of conferring protection to C57BLKS/J- Leprdb/db mice (132). Similarly, mice homozygous for a db allele (db3J) found on this strain have very mild and transient hyperglycemia, marked hyperinsulinemia, and dramatically elevated pancreatic insulin content. Although this is accompanied by progressive hyperglucagonemia, the mice ultimately develop hypoglycemia leading to sudden death (133). Similarly, lean 129 mice with genetically induced defects in insulin signaling due to double heterozygosity for Irs1 and Insr mutations have only mild hyperinsulinemia with little insulin resistance and do not increase -cell mass compared with wild-type 129 mice (110). This is in contrast to C57BL/6 and DBA/2 mice bearing the same mutations, which develop marked hyperinsulinemia with much more severe insulin resistance and resulting in diabetes (110, 131). In these double-heterozygous mice, linkages for plasma glucose levels have been identified on chromosomes 12 and 14, where C57BL/6 alleles are diabetogenic (131). In contrast to the insulin resistance evoked by Irs1-Insr double heterozygosity, heterozygosity for only a null allele of the Insr results in more severe hyperinsulinemia in the 129 than C57BL/6 background (134). When loci were mapped in an Insr/ C57BL/6x 129 F2 population, five loci (significant and suggestive) affecting plasma insulin levels were identified; however, at four of these loci, C57BL/6 alleles were linked to the higher serum insulin concentrations. Nonetheless, this suggests that the 129 strain may harbor at least one diabetogenic locus. Additional evidence of diabetes promoting alleles in the 129 strain come from other knockouts in the insulin signaling pathway. The insulin receptor substrate-2 (Irs2) gene is important for the normal survival and development of -cells (135). In a C57BL/6x 129/sv mixed background, Irs2/ mice have reduced Pdx1 expression and develop diabetes (136). However, mice with this mutation on the pure C57BL/6 background preserve the expression of the Pdx1 transcription factor and its downstream genes (137). This suggests that alleles from the 129 background contribute to the downregulation of Pdx1.
E. BTBR

The black and tan, brachyuric (BTBR) mouse strain was developed by L. C. Dunn from an outbred stock he obtained from Nadine Dobrovolskaia-Zavadskaia (138). The strain was maintained by Dorothea Bennett after 1962, and then by her graduate students, Flaherty and Artzt, who passed it on to Jean-Louis Guenet at the Pasteur Institute. Guenet maintained the strain for 45 generations, then passed it to Doves laboratory at the University of Wisconsin. During its breeding history, it was bred with 129 mice and selected for good breeding performance (139). Thus, it is genetically most related to the 129 strain (36, 57). However, in contrast to 129, BTBR is a diabetes-susceptible strain.

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice

Endocrine Reviews, February 2007, 28(1):48 83 57

Lean male F1 hybrids between C57BL/6 and BTBR are more glucose intolerant than either parental strain, suggesting that interactions between C57BL/6 and BTBR alleles can further promote diabetes susceptibility (140). Insulin-stimulated glucose uptake into isolated adipocytes from BTBR (141) and the BTBR C57BL/6 F1 mice is severely defective (142). Isolated skeletal muscles from the F1 mice also have defective insulin-stimulated glucose transport, although a much milder defect is observed in muscles from lean BTBR mice (140). In vivo, glucose uptake into muscle and adipose tissue in male BTBR mice under hyperinsulinemic clamp conditions is less than C57BL/6, although insulin-mediated suppression of hepatic glucose output is unaffected in BTBR (J. B. Flowers and A. D. Attie, unpublished observations). Islets isolated from BTBR mice hypersecrete insulin at low glucose concentrations and in response to nonglucose secretagogues (143). Similar to C57BLKS/J mice, when the Lepob mutation is bred into BTBR background, BTBR Lepob/ob mice develop severe diabetes (141, 144). Young BTBR Lepob/ob mice are hyperinsulinemic compared with C57BL/6 Lepob/ob. However, as these mice age, insulin levels decline, resulting in marked hyperglycemia (141). By 3 months of age, pancreatic insulin content is dramatically reduced, and islet architecture is severely disrupted in BTBR Lepob/ob mice (our unpublished observations). We have used this model for several genetic and genomic studies (144 147) and in a cross between BTBR and C57BL/6 mice, we have mapped several diabetes-related QTL in Lepob/ob F2 animals (144). Using overlapping interval-specific congenic (65) and subcongenic strains, Clee et al. (148) recently positionally cloned one of these loci on chromosome 19. The gene underlying the t2dm2 QTL was found to be SorCS1 (148), a receptor expressed in islets that binds to platelet-derived growth factor-BB (149). Platelet-derived growth factor-BB is required for pericyte recruitment to the islet microvasculature, a step critical for vessel formation and stability (150, 151). Thus, we believe that SorCS1 may play a role in the development or maintenance of the islet vasculature, which would be important for expanding -cell mass in response to insulin resistance. Interestingly, the diabetogenic allele at this locus was contributed from the C57BL/6 strain (144, 148) and was associated with reduced insulin secretion in vivo, suggesting that alterations in SorCS1 may also contribute to the impaired insulin delivery to the bloodstream of the C57BL/6 strain.
F. A/J

155). It is a common progenitor in RI and recombinant congenic panels.


G. BALB/c

The BALB/c strain is genetically most related to A/J (36, 37). Consistent with this, it is more glucose tolerant than C57BL/6 (98) and more resistant to the glucotoxic effects of streptozotocin (104, 117). BALB/c mice have a relatively high -cell mass due to a large number of islets (103). In the BALB/c background, Lepob/ob mice have increased insulin levels in the fed state compared with C57BL/6 Lepob/ob (156). Interestingly, BALB/c Lepob/ob mice have reduced adiposity, which correlates with increased thermogenesis. In contrast to other strains, BALB/c mice homozygous for the Lepob mutation remain fertile (156).
H. C3H

C3H/HeJ is a diabetes-resistant strain. This strain is related to BALB/c, A/J, and AKR (36). It is highly glucose tolerant and has a robust insulin secretory response, which is sufficient to overcome its slight insulin resistance (93, 98). C3H mice are resistant to the -cell toxic effects of streptozotocin, possibly due to the effects of a single gene (105, 117). -Cell mass in C3H is derived from relatively few islets of large size (103), resulting in increased -cell mass compared with C57BL/6, but less than other strains such as DBA/2 and BALB/c. As described above, mapping in F2 and F3 samples derived from C3H and C57BL/6 has identified loci on chromosomes 2 and 13 associated with improved glucose tolerance relative to C57BL/6 (99). In contrast, a C3H-derived allele associated with impaired glucose tolerance has recently been mapped in a cross with C57BLKS/J-db mice (157).
I. AKR

The A/J strain is the classic diabetes-resistant mouse strain and is among those with the lowest glucose levels (2). When fed a high-fat, cholesterol-containing diet, A/J mice maintain very low glucose and insulin levels. Compared with the C57BL/6 strain, A/J mice are resistant to high-fat, high-carbohydrate feeding-induced obesity, insulin resistance, and glucose intolerance (89 91, 125), in part due to increased insulin secretion and a sustained second-phase response (100, 101). Thus, A/J mice have reduced glucose even on chow (91, 152) and do not expand -cell mass when fed high-fat diets (100). However, even this resistant strain may harbor some alleles promoting glucose intolerance (153

AKR mice are closely related to the LG strain, described below, and are also related to C3H, BALB/c, and A/J (36). The AKR strain is very sensitive to diet-induced obesity (92), and adipocytes from this strain exhibit increased insulinstimulated glucose uptake compared with SWR/J mice (158). Unlike its related strains described above, when fed a highfat diet, AKR mice are more hyperinsulinemic than others (2). They are more insulin resistant than C57BL/6, primarily due to reduced glucose uptake into adipose tissue (96). Despite this, AKR are more glucose tolerant and have lower plasma glucose levels than C57BL/6 because of increased insulin secretion during a glucose challenge (96). AKR mice are relatively resistant to the effects of streptozotocin (117).
J. CAST/Ei

The CAST/Ei strain is an inbred line derived from wild Mus musculus castaneus mice from Thailand. Compared with other strains with data in the Mouse Phenome Database, CAST/Ei mice have relatively high glucose levels (2). The Lusis group (159) has made extensive use of this strain in studies of obesity and atherosclerosis. CAST/Ei mice are very lean, but, on a high-fat diet, hyperinsulinemic relative to C57BL/6. In an F2 derived from CAST/Ei and C57BL/6, three

58

Endocrine Reviews, February 2007, 28(1):48 83

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice

independent loci on chromosome 2 segregate with body weight loci and exhibit suggestive linkage to plasma insulin levels (159). Differences in glucose and insulin levels have been observed in congenic strains where CAST/Ei alleles have been transferred into the C57BL/6 background (160). In a recent study, a new selectively inbred strain, the Horio-Niki diabetic (HND) mouse, was derived from C57BL/6 and M. m. castaneus parents (161). These nonobese mice have impaired insulin secretion in vivo, and an altered response to 2-adrenergic stimulation, suggesting that this may be a model of impaired neurological control of insulin secretion (162).
K. Nonobese diabetic (NOD) and nonobese nondiabetic (NON)

diet (176). With 45% fat and simple carbohydrates, the mice become transiently hyperglycemic and very obese, but eventually go into remission from the hyperglycemia (E. H. Leiter, personal communication). The genetic mapping studies that mapped diabetes in the NON strain used an F2 intercross between this strain and NZO, described below. In contrast to either parental strain, F1 mice exhibit severe fasting hyperglycemia, and nearly 100% of males develop diabetes (173). Two NON-derived diabetogenic loci were identified in the F2 sample (173), and a third suggestive NON-derived locus was identified in a (NZOxNON) NON backcross (174).
L. New Zealand obese (NZO)

These strains have a complicated history. Originally, a mouse with spontaneous development of cataracts was observed in a closed outbred colony of Imperial Cancer Research (ICR) mice. This mouse was selected and inbred to develop the CTS (Cataract-Shiongi) line, however none of the offspring developed cataracts (29, 163). As cataracts can be a complication of diabetes, at the F6 generation, two lines were split, one with euglycemia (A) and one with mild hyperglycemia (B). At the F20 generation, a spontaneously diabetic mouse was observed in the euglycemic A line. The offspring of this mouse were inbred for several generations with selection for spontaneous diabetes to derive the NOD line, whereas mice from strain B did not develop spontaneous diabetes, and became the NON control line (163). The NOD strain is a model of type 1 autoimmune diabetes, with a complex genetic etiology. The genetics of this strain have been extensively studied and involve at least 20 different loci, none of which independently result in diabetes (29). The genetic complexities of this strain are also evident in that several of the mapped Idd loci have ultimately been dissected into multiple distinct loci (29, 164). Analysis of these loci often involves haplotype comparisons across several diabetes susceptible and resistant strains, in congenics where the genotype at several other loci is controlled (164, 165). Interestingly, alleles that further promote diabetes in NOD mice derived from the nondiabetic control strains, e.g., C57BL/6, have also been observed (166), and alleles associated with the sensitivity of this strain to streptozotocin have been mapped (167). We will not focus on the models of type 1 diabetes in this review, but point the reader to other reviews of the subject (168 172). Of more relevance to type 2 diabetes is the NON strain, which has been shown to have alleles promoting type 2 diabetes (173175). This should not be particularly surprising, given its selection for mild hyperglycemia during its breeding history and that several other models of -cell dysfunction and type 2 diabetes have been derived from the outbred ICR mice (see descriptions of the NSY and ALR and ALS strains below), suggesting that this stock has numerous alleles promoting diabetes (29, 163). As the genes are identified, it will be interesting to observe whether there is any overlap between type 1 and type 2 diabetes susceptibility alleles (29). NON mice are glucose intolerant and have impaired glucose-stimulated insulin secretion, but they do not transition to overt diabetes even when fed an 11% high-fat

The New Zealand obese (NZO) strain was derived from an outbred stock from the Imperial Cancer Research (ICR) laboratory in England. It was originally selected for agouti coat color, along with the New Zealand Black (NZB) and New Zealand White (NZW) strains (177), and is a model of spontaneous polygenic obesity and insulin resistance. Leptin receptor polymorphisms have been detected in this strain; however, they do not appear to directly cause the obesity of the strain, because the related NZB strain shares these polymorphisms and is not obese (178). Furthermore, studies mapping obesity-related genes in this strain have not observed linkage to this region (179, 180). It is possible, however, that these polymorphisms may confer leptin resistance or impaired leptin transport across the blood-brain barrier (181) and interact with other obesity-promoting alleles to enhance the obesity of these mice (178, 180). The phenotype of NZO mice resembles the metabolic syndrome (182). Compared with the related NZB and NZC strains and the lean Swiss Jackson Laboratory (SJL) strain, NZO mice are markedly hyperinsulinemic, with males developing hyperglycemia (182, 183). The mice have reduced insulin-stimulated glucose uptake into muscle and adipose tissue, along with increased hepatic glucose production (183). Serum triglycerides and blood pressure are also increased in these mice (182). First-phase insulin secretion in response to a glucose bolus is impaired in NZO mice (183). After initial -cell hyperplasia, decompensation in males is accompanied by reduced -cell mass, degranulation, and ultrastructural changes (184). However, in various NZO substrains, not all NZO males decompensate, apparently needing to exceed a body weight threshold before diabetes develops (173, 174, 176). Like NZB mice, NZO mice have an autoimmunity phenotype; they have autoantibodies against the insulin receptor, which may contribute to their insulin resistance (185). Leukocyte infiltration of the pancreas has also been observed in NZO mice, and diabetes is worsened in NZO NZB F1 mice (177, 184). However, later studies have shown that this autoimmunity is not essential for the development of diabetes in these mice (176). Loci contributing to obesity and diabetes susceptibility in this strain have been detected on nearly every chromosome (175). Genetic mapping studies have also been performed in a (NZO SJL) SJL backcross panel (179). Interestingly, these studies identified a major locus on chromosome 4 from

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice TABLE 1. QTL for diabetes-related traits in mouse

Endocrine Reviews, February 2007, 28(1):48 83 59

Synteny of mouse positions (to the nearest 100 kb) was mapped to the nearest Mb position in human and rat using the comparative maps at NCBI (www.ncbi.nlm.nih.gov/Homology/) and chained alignments in the UCSC genome browser (www.genome.ucsc.edu) using the following genome assemblies: mm8 (Build 36), hg18 (Build 36.1), and rn3.4 (Build 3.4 based on RGSC) at www.genome.ucsc.edu. Adiposity and lipid-related QTL from these crosses are not described unless they co-mapped with an insulin or glucose locus. Sig?*, Significant at the genomewide level?; n, did not reach genomewide threshold; chr, chromosome; WTD, Western-type diet; HFD, high-fat diet; LFD, low-fat diet; CHO, high-carbohydrate diet; wt, weight; TG, triglycerides; AUC, area under the curve; B6, C57BL/6; BKS, C57BLKS/J; pos., position; incr, increased; decr, decreased; gluc, glucose; avg, average; het, heterozygotes. Table continues.

the SJL background that contributes to the development of diabetes (hypoinsulinemia and hyperglycemia) in these obese mice (179, 186). This locus, Nidd/SJL, is associated with hyperglycemia and relative hypoinsulinemia, due to a loss of -cells and islet disorganization in high-fat-fed mice (186). A strong interaction between this locus and one controlling obesity accelerates the progression of diabetes in these mice. Subsequent mapping in female mice demonstrated that NZO-derived alleles contribute to obesity and hyperinsulinemia (180). In an F2 derived from NZO and NON, Leiter et al. (173) identified three NZO-derived diabetogenic alleles. Two of these were suggestive loci affecting blood glucose concentrations. Interestingly, they were dependent on the direction of the original cross, i.e., whether the parental female or male was NZO. A second study, in an F1 NON backcross, identified a major NZO-derived diabetogenic QTL (174). This locus influenced body weight, plasma glucose, and early hyperinsulinemia. This study also detected complex interactions between various loci (e.g., those affecting obesity and plasma glucose levels), which may contribute to the increased diabetes in F1 males compared with either parental strain. Interactions were also detected between various loci and the maternal genotype (environment), illustrating the complexity of factors that may influence diabetes-related QTL (174).

Remarkable follow-up studies by this group have attempted to reconstitute the diabetes susceptibility of the F1 animals using RCSs containing various combinations of the diabetogenic QTL (175, 176). When numerous epistatic interactions are evident, this approach has an advantage over creating single-region congenics, which do not allow for interaction among multiple loci. These studies have elegantly demonstrated that the development of diabetes depends on the inheritance of unique combinations of these QTL. Some strains are obese without diabetes, whereas the prevalence of diabetes increases in strains bearing an increasing number of diabetogenic QTL (175). As predicted, these strains differ in numerous characteristics, including islet pathology (176).
M. FVB

Although there is relatively little information on lean FVB mice, sensitized screens show that the strain harbors diabetogenic alleles, particularly relating to peripheral insulin resistance. Glucose levels are higher in this strain than many others, accompanied by relatively low insulin levels (2). In the FVB background, Leprdb/db mice are more insulin resistant and have more severe hyperinsulinemia and hyperglycemia than C57BL/6 Leprdb/db mice. FVB-Leprdb/db mice have enlarged islets; however, islet destruction is not observed (127,

60

Endocrine Reviews, February 2007, 28(1):48 83

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice

TABLE 1. Continued

187), suggesting that these strains primarily differ in alleles promoting insulin resistance and not -cell failure. Similarly, a comparison of FVB Lepob/ob and C57BL/6 Lepob/ob mice revealed increased hyperglycemia and peripheral insulin resistance in FVB Lepob/ob, although hepatic insulin resistance was increased in the C57BL/6 Lepob/ob mice (188). An FVB C57BL/6-Leprdb/db F2 intercross mapped a locus on chromosome 5 where FVB alleles were associated with increased insulin. Congenic Leprdb/db mice inheriting C57BL/6 alleles in this region on the FVB background are unable to maintain insulin production and undergo pancreatic decompensation (187). This suggests that the insulin-resistant FVB background unmasks a C57BL/6-derived allele promoting -cell failure, which is otherwise silent in the C57BL/6 background. A second leptin-deficient model is a mouse made lipoatrophic by adipose expression of a dominant-negative protein, AZIP, that interferes with the C/EBP and Jun transcription factors (189). In the C57BL/6 background, the AZIP mice had transient hyperglycemia, returning to normal by 4 months of

age. In striking contrast, in the FVB strain background, the mice were severely diabetic with fed glucose levels of 600 mg/dl. Like the Lepob/ob mice, C57BL/6-AZIP mice had more severe liver insulin resistance, whereas FVB-AZIP mice have increased muscle insulin resistance (190). Under hyperinsulinemic conditions, the C57BL/6 AZIP mice had 40% higher glucose uptake into muscle, suggesting that the difference in glucose between these strains was likely due to a difference in peripheral insulin sensitivity (188).
N. KK

The KK strain was derived from wild-derived ddY mice in Japan in 1957 by Kondo (see Ref. 191). SNP analysis suggests that it is closely related to the NZO and NON strains (36). Because several breeding colonies of these mice have been maintained, there are now several substrains [T-KK (or Toronto-KK, KK/Upj), KK/HlLt, KK/Ta, KK/San]. For this review, we will refer to them all as KK. KK mice are obese and hyperleptinemic. A leptin receptor

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice TABLE 1. Continued

Endocrine Reviews, February 2007, 28(1):48 83 61

amino acid variant has been identified in this strain that may account for differences in adiposity in females, although this has not been observed in all studies (192, 193). Compared with its ddY progenitor, KK mice have increased glucose and HbA1c levels and impaired glucose tolerance (194 196). Similarly, compared with C57BL/6 or BALB/c, KK have increased body weight, accompanied by glucose intolerance (191, 195). KK mice develop severe hyperinsulinemia, comparable to the severely insulin resistant C57BLKS/J Leprdb/db mice (2, 192, 195). KK mice have both muscle and adipose insulin resistance (197). Total pancreatic insulin content is increased, associated with islet hypertrophy and hyperplasia but marked degranulation (reduced number and size of granules) (191). Dilated islet capillaries and increased vascular endothelial cells have also been noted in these mice (191). The KK strain is also susceptible to diabetic nephropathy (113). With the introgression of the Ay mutation in agouti, they exhibit further reduced glucose tolerance and become more severely diabetic (194, 196). KK-Ay mice are hyperinsulinemic, have increased HbA1c, and exhibit early stage nephropathy (196). Numerous linkages for diabetes-related

traits have been observed in several crosses with KK mice (193195, 198, 199).
O. TallyHo

The TallyHo mouse strain is a naturally occurring model of obesity and type 2 diabetes. The strain derived from two Theiler Original male mice with polyuria and hyperglycemia, which were selectively inbred for hyperglycemia (200). Mice from this strain have increased body weight and body fat and are hyperlipidemic and hyperinsulinemic. Hyperglycemia, accompanied by hypertrophied and degranulated islets, develops only in male mice (201). Several loci influencing plasma glucose levels and body weight have been mapped in this strain in crosses with either CAST/Ei or C57BL/6 (200, 202).
P. Nagoya-Shibata-Yasuda (NSY)

The NSY strain was developed by selective inbreeding for glucose intolerance from outbred JcI:ICR mice, the same

62

Endocrine Reviews, February 2007, 28(1):48 83

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice

TABLE 1. Continued

stock from which NOD and NON mice were derived (54, 203). Nearly all males have impaired glucose tolerance, which also develops in one third of females. Compared with the diabetes-resistant C3H strain, NSY males have increased fasting insulin levels and increased postprandial, but not fasting, glucose levels. These mice have impaired glucose tolerance and markedly impaired glucose-stimulated insulin secretion in vivo, which may account for the increased glucose levels in the fed state (203). NSY mice have increased pancreatic insulin content without an increase in mean islet size (203). Genetic analysis of this strain has mapped three primary loci [Nidd1nsy (Nidd1n), Nidd2nsy (Nidd2n), and Nidd3nsy (Nidd3n)] and one suggestive locus [Nidd4nsy (Nidd4n)] affecting insulin sensitivity and/or insulin secretion (204). Interestingly, the Nidd1n and Nidd2n loci overlap with regions contributing to -cell destruction or impaired regeneration in NOD mice derived from the same outbred population, suggesting that there may be common loci affecting susceptibility to both type 1 and type 2 diabetes (29, 205). Furthermore, this strain shares a locus affecting sensitivity to streptozotocin with the NOD strain (206).
Q. ALR/Lt and ALS/Lt

ALS/Lt males are hyperinsulinemic, hyperglycemic, and have impaired glucose tolerance, ultimately developing diabetes (209). ALS/Lt mice also have abnormal insulin secretion and develop diabetes when the agouti Ay mutation is introgressed into the strain (210). Because this occurs in the absence of alloxan, we might infer that genes protecting -cells from oxidative damage under normal conditions may be important mediators of type 2 diabetes. Recently, Mathews et al. (211) found that diabetes in the ALS/Lt strain was reduced when mitochondrial DNA was derived from the ALR/Lt strain. They identified a single SNP differing between the ALS/Lt and ALR/Lt mitochondrial genomes; a L276M polymorphism in the NADH dehydrogenase 2 gene, which they propose contributes to resistance to both chemical and autoimmune diabetes (211). The same mutation in humans is also correlated with protection from type 1 diabetes (212).
R. M16

The ALS/Lt and ALR/Lt inbred strains were also derived from CD-1 (ICR) outbred mice by selecting for susceptibility or resistance to alloxan (free-radical)-induced diabetes, respectively (207). These strains are related to the NOD and its related strains (36). The ALR/Lt and ALS/Lt strains also have differential susceptibility of the mice to streptozotocin and of their isolated islets to peroxide. ALR/Lt mice have reduced markers of oxidative stress, likely due to a single genetic difference (207). The effect is an intrinsic property of the islets because the phenotype is maintained in cultured islets (207, 208). In the absence of an oxidative challenge,

The M16 mouse strain was created through long-term selection of ICR outbred mice for body weight gain (213). Compared with the parental ICR stock, these mice have increased body weight and fat, due to increased food intake. Males and females are both hyperinsulinemic, but only males from this strain develop moderate hyperglycemia. Using an F2 comprised of nearly 1200 animals, Pomp and co-workers (214) identified numerous QTL affecting these phenotypes in an F2 cross with randomly selected ICR stock.
S. LG and SM

The LG (large) and SM (small) strains were independently derived, selecting for body size. LG was created in the 1930s by selection for large body size from a pool of albino mice

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice TABLE 1. Continued

Endocrine Reviews, February 2007, 28(1):48 83 63

(54), and it appears most closely related to AKR (36). SM was generated in the 1940s from a pool of mice derived from four crosses between seven strains, including dilute brown (dba), silver chocolate (sv ba), black and tan (at), pink-eyed shorteared dilute brown (psedba), albino (c), cinnamon spotted (bs), and agouti (a) (215). Thus, this strain has ancestors in common with both the DBA and BTBR strains, and it appears most closely related to DBA/2 (36). The LG and SM strains have been used for extensive genetic analysis of traits related to type 2 diabetes. The strains differ in many traits, including obesity, plasma glucose, glucose tolerance, and response to high-fat feeding (216). Compared with LG, SM mice have increased serum glucose on a high-fat diet. A RI strain panel has been created from these strains (215) and shows variation across strains in the development of hyperglycemia and hyperinsulinemia. Several QTL for insulin, glucose, and glucose tolerance have been identified in the LGXSM RI strains (217, 218). Interestingly, the majority of loci affecting glucose levels are distinct from those affecting obesity and are only weakly correlated with those affecting insulin, suggesting that the loci primarily

affecting glucose levels are not the same as those that affect insulin resistance (218). The SM strain has been used in another RI panel, the SMXA RI panel, derived from SM and A/J. The RI strains show differences in body weight and plasma insulin levels (219). When compared with A/J, SM mice fed a high carbohydrate diet have reduced body weight and improved glucose tolerance, which is also observed in response to high-fat feeding (153, 220). Although neither parental strain is diabetic, high-carbohydratefed F1 animals have further impaired glucose tolerance, which appears to result from the interaction of at least three loci, two from A/J and one from SM (153). These RI strains have been characterized for other diabetes-related phenotypes, and it has been suggested that one strain of the panel, SMXA-5, may be a useful diabetes model because it displays insulin resistance accompanied by increased -cell mass (220 222). To identify further the A/J-derived loci that contribute to these phenotypes, an F2 sample derived from the SMXA-5 and the parental SM strain was created. This identified several loci affecting plasma glucose and insulin levels and glucose tolerance (154). A major locus on mouse chromosome 2 was identified where

64

Endocrine Reviews, February 2007, 28(1):48 83

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice

TABLE 1. Continued

alleles from A/J contribute to impaired glucose tolerance. The phenotype was verified by a congenic strain in which A/J alleles were introgressed into the SM background (154).
T. Tsumura, Suzuki, Obese Diabetes (TSOD)

Like KK mice, the TSOD strain was derived as an inbred obese diabetic strain from the outbred Japanese ddY stock (223). Beginning in 1984, mice were selected for obesity and urinary glucose, then inbred, along with a nonobese/ nondiabetic control line (TSNO) derived from sibs (223). Compared with their parental ddY strain, the TSNO strain, or BALB/c, these mice are obese and insulin resistant, which results in hyperglycemia and hyperinsulinemia in males (223, 224). Severe diabetes does not develop because TSOD mice can increase -cell mass; however, there appears to be a defect in insulin secretion in response to an in vivo glucose challenge (223). Older mice from this strain develop islet hypertrophy, and also have evidence of neuropathy and nephropathy (225). Loci affecting plasma glucose and insulin levels and glucose intolerance have been mapped in a cross between this strain and BALB/c (224). Interestingly, a suggestive locus for increased glucose levels during an ip glucose tolerance test (IPGTT) from the BALB/c strain was also observed. Replication of the effects of the Nidd5 locus from this strain on body weight has been obtained in congenic mice, although these mice do not display the differences in insulin levels that were also mapped to this marker (226), suggesting that multiple genes may be responsible for the Nidd5 phenotype.
U. The Akita mouse

The Akita mouse strain derived from a C57BL/6 mouse with a spontaneous mutation causing severe insulin-dependent diabetes (47). The trait was mapped to telomeric chro-

mosome 7 in a backcross with C3H and an F2 intercross with M. m. castaneus (47). The hypoinsulinemia in these mice correlates with a reduced -cell mass. The phenotype of this strain is caused by a C96Y mutation in the insulin 2 (Ins2) gene, corresponding to the seventh amino acid on the insulin A chain (227). The mutation prevents formation of one of the disulfide bonds between the A and the B chains of the insulin molecule. This leads to reduced processing of proinsulin and a severe reduction in mature insulin. It was not immediately obvious how a mutation in the insulin gene would lead to a reduction in -cell mass. The answer to this conundrum came from an understanding of the way in which cells confront an overload of misfolded proteins in the ER, i.e., ER stress. Failure to properly respond to ER stress through activation of the unfolded protein response is the cause of several known inherited diseases. In the unfolded protein response, cells adapt by reducing the rate of synthesis of most proteins while increasing the synthesis of chaperonins (228). The inhibition of protein synthesis occurs when the ER kinase PERK phosphorylates translation initiation factor eIF2. PERK deficiency or a mutation abolishing eIF2 phosphorylation leads to a phenotype resembling that of the Akita mouse (229, 230). Although most protein synthesis is reduced upon eIF2 phosphorylation, the translation of the transcription factor ATF4 is increased, leading to increased expression of C/EBP homologous protein (CHOP). CHOP promotes cell death, presumably by reversing the attenuation of translation and by promoting excessively oxidizing conditions in the ER (231). A key observation is that deletion of the Chop gene delays the onset of diabetes in the Akita mouse, showing that at least in the earliest phase, this gene mediates the lethality of the protein folding defect (232). A recent study has mapped modifiers of the Akita phenotype in a cross with A/J mice

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice TABLE 1. Continued

Endocrine Reviews, February 2007, 28(1):48 83 65

and identified both A/J and C57BL/6-derived alleles that are associated with further glucose intolerance (155).
VIII. Linkage Hot Spots

In Table 1, we have summarized the linkages for glucose and insulin-related traits that have been identified in these mouse models. We have included the position of the peak marker but have not attempted to specify confidence intervals, unless these have been determined through a congenic strain. We also include a notation regarding whether the locus reached the genomewide significance threshold. Because several study design-related factors can influence whether a significant linkage is obtained, we have included all loci with at least nominal evidence of linkage, as they may still be real loci with smaller effects or effects that the study was underpowered to detect. Indeed, several of these loci overlap with each other and with other significant QTL. Additional loci identified only through analysis of epistatic interactions are described in Table 2. It can be seen immediately that linkages span the mouse genome, although some chromosomes have relatively more hits. Numerous linkages have been observed on chromosome 2, where many body weight/adiposity loci have also been mapped (233). Interestingly, although numerous body weight loci have been mapped on chromosome 7 (233), only one diabetes-related locus has been mapped to this chromosome, suggesting that these two phenotypes are separable. We observe four clusters (highlighted in Table 1), where QTL from at least four independent strains have peak markers residing within 20 Mb of each other. On chromosome 9, between 36 49 Mb, are loci for increased fasting glucose and glucose or insulin levels during an IPGTT in crosses involving the NZO, C57BL/6, DBA/2, and KK strains (98, 122, 126, 173, 194). On chromosome 11 from 6271 Mb, five loci have been identified from the NZO, C57BLKS/J, M16, SM, and NSY strains (123, 154,

173, 204, 214). Between 79 99 Mb on chromosome 12 are loci from the KK, NZO, 129, and C57BL/6 strains (131, 134, 173, 195). Finally, at the telomeric end of chromosome 19, from 4155 Mb, four loci have been described from the HRS, NZO, TallyHo, and C57BL/6 strains (123, 144, 148, 180, 200). These hot spots of linkage are described more in the next section. Because there are several regions of the mouse genome harboring multiple QTL, we asked whether these also correspond to diabetes-related QTL in rat models and examined their relationship to human diabetes QTL. To obtain a complete picture of the linkages observed in rodents that correspond to these loci, we have tabulated the diabetes-related QTL that have been observed in the rat (Supplemental Table 1, published as supplemental data on The Endocrine Societys Journals Online web site at http://edrv.endojournals.org), and compiled a list of all loci for type 2 diabetes and glucose and insulin-related traits mapped in various human populations, along with the positions of the MODY genes and the genes for which associations with type 2 diabetes have been validated by meta-analysis (Supplemental Table 2). We have presented all loci, whether they reach genomewide significance or not, because there are often numerous individual studies all pointing to the same area, which on their own do not meet strict statistical criteria but when taken together provide stronger evidence for a QTL in that region. The data are for illustrative purposes only, and the reader is strongly encouraged to read the original papers for details on the significance of the QTL, their confidence intervals, and details of the study design and analysis methods, because it is beyond the scope of this review to include these details here. Consistent with our analysis of the mouse data, we have also avoided inclusion of obesity-related loci, because these are extensively reviewed in the Obesity Gene Map (233). Examination of these data has revealed several hot spots containing multiple linkages across the species. Differences in the phenotypes mapped to these regions may either reflect differences in the assessed phenotypes, pleiotropic effects of

66

Endocrine Reviews, February 2007, 28(1):48 83

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice

TABLE 1. Continued

the underlying gene, or be indicative of multiple genes in the region. Indeed, it may often be the case that the strength of a QTL is increased if there are multiple closely linked genes affecting the phenotype, especially if the mapped phenotype was broad, such as type 2 diabetes. Here, we have only cross-referenced the regions directly syntenic to the LOD peak observed in mice. Thus, overlap of the entire confidence intervals may include additional loci not emphasized here (perhaps also on different chromosomes if there is a break in the syntenic block). Despite the narrow limits of our assessment, the data strongly suggest that one or more important genes for diabetes susceptibility reside in these locations and highlight the relevance of the rodent linkages to humans.
A. Mouse chromosome 19, rat chromosome 1, and human chromosome 10q

Arguably, the most success in type 2 diabetes genetics to date has come from studies of mouse chromosome 19, rat chromosome 1, and human chromosome 10q. Four studies have mapped diabetes-related traits to distal chromosome 19 in the mouse. We identified T2dm2 in a cross between C57BL/6 and

BTBR mice homozygous for the ob mutation (144). At this locus, the C57BL/6 strain contributes an allele associated with reduced insulin levels due to defective insulin secretion in vivo. As described above, we have recently identified the SorCS1 gene as underlying T2dm2 (148). The Tanidd1 locus, associated with increased glucose levels, was identified in the TallyHo mouse strain, in crosses between this strain and both the C57BL/6 and CAST/Ei strains (200). In the NZO strain, a locus associated with increased insulin levels was identified in an F2 cross between this strain and the Swiss/Jackson Laboratory (S/JL) strain (180). Most recently, a cross between the C57BLKS/J and HRS strains homozygous for the Cpefat mutation identified a locus, Find1, associated with increased plasma glucose levels (123). In rats, the distal end of chromosome 1 is syntenic to this region. The GK rat strain was derived by selective inbreeding of Wistar rats with the highest glucose levels (234). Although not obese, GK rats have hyperglycemia, insulin resistance, defective insulin secretion, a deficit of -cells, and diabetic complications, i.e., retinopathy, neuropathy, and later in life (1218 months), nephropathy (235). In an F2 cross between GK and nondiabetic Fisher 344 (F344) rats, Galli et al. (236)

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice TABLE 1. Continued

Endocrine Reviews, February 2007, 28(1):48 83 67

mapped multiple loci linked to fasting and postprandial glucose levels. The strongest linkage they observed, Niddm1, was on the distal region of chromosome 1, with a peak LOD of 11.0 for glucose at 60 min, at marker D1Mit7. Gauguier et al. (237) also mapped diabetes-related traits in the GK strain in a cross with the Brown Norway (BN) strain. They identified a peak linkage at D1Wox10, approximately 20 Mb centromeric to that of Galli and colleagues, for fasting glucose levels, postprandial glucose levels, insulin secretion, and adiposity, which they named Nidd/gk1. Galli et al. (30) dissected the chromosome 1 locus by creating congenic strains in which segments of the GK chromosome 1 were introgressed into the F344 background. A congenic strain with a 52-cM insert, Niddm1a, recapitulated the impaired postprandial glucose tolerance detected in the original F2 study. Two nonoverlapping subcongenic strains, Niddm1b and Niddm1i, had distinct phenotypes, indicating that at least two distinct loci can autonomously contribute to this phenotype. Interestingly, both strains are insulin resistant, and adipocytes from both subcongenic strains had defective insulin-stimulated glucose incorporation to lipids

(30). In addition, the strain with the more telomeric insertion, NIDDM1i, has defective postprandial insulin secretion in vivo (30) and severely defective glucose-stimulated insulin secretion in isolated islets (31). Additional fine-mapping studies by Gauguier and colleagues (238) have also identified an insulin secretion defect in this region. Very recently, Granhall et al. (32) have reported that the NIDDM1i locus itself contains multiple loci affecting plasma glucose concentrations. The SorCS1 gene is located within the NIDDM1i congenic strain, suggesting that it may be a candidate for the defective insulin secretion phenotype, although we do not see differences in insulin resistance in our mice. This gene is also the only gene contained within one of the glucose loci identified by Granhall et al. (32). The Niddm1b locus has been further fine-mapped with subcongenic strains to a region of approximately 2 Mb (239). This region contains the insulin degrading enzyme gene (Ide). The GK allele has two coding differences from the F-344 allele, H18R and A890V (239). COS cells transfected with the GK allele degraded insulin 31% less actively than those expressing the F344 variant. The results were replicated in

68

Endocrine Reviews, February 2007, 28(1):48 83

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice

TABLE 1. Continued

transfected fibroblasts (240). Neither of the amino acid substitutions individually reduced the activity of the IDE protein (239). Mice deficient in IDE have a 2.8-fold increase in fasting insulin and are glucose intolerant (241). Hence, the Ide gene is a likely diabetes susceptibility gene within the Niddm1b locus in the GK rat. Linkage to this region has also been detected in the Otsuka Long-Evans Tokushima Fatty (OLETF) rat, a strain that is derived from a spontaneously diabetic Long-Evans rat. Mice from this strain have mild obesity with early hypertriglyceridemia and impaired glucose tolerance, followed by hypergly-

cemia, pancreatic decompensation, and renal pathologies (242). Wei and colleagues (243, 244) identified a locus, Nidd6/of, affecting fasting and postprandial glucose levels and body weight in this region of chromosome 1 in an F2 intercross between OLETF and F344 rats. The authors so far have been unable to replicate the phenotype in a congenic strain (245). Independently, in F2 crosses between OLETF and both the BN and F344 strains, the Dmo1 locus, affecting body weight along with fasting and postprandial glucose levels was mapped to the same marker, D1Rat90 (246), the telomeric boundary of the Niddm1i subcongenic strain. Wa-

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice TABLE 2. Interacting QTL for diabetes-related traits in the mouse
Chromosomes Markers Peak LOD Phenotype

Endocrine Reviews, February 2007, 28(1):48 83 69

Genetic cross

Ref.

Notes

1, 12 1, 15 1, 17 2, 15 12, 15 15, 17 1, 8 2, 5 2, 8 2, 17 7, 19 8, 11 8, 15

D1Mit123 D12Mit150 D1Mit76 D15Mit159 D1Mit76 D17Mit61 D2Mit182 D15Mit26 D12Mit204 D15Mit159 D17Mit61 D15Mit159 D1Mit206 D8Mit42 D2Mit365 D5Mit372 D2Mit525 D8Mit47 D2Mit92 D17Mit139 D7Mit82 D19Mit35 D8Mit155 D11Mit99 D8Mit155 D15Mit239 9.75 6.72 7.53 8.62 8.15 8.48 9.31

Glucose Glucose Glucose Glucose Glucose Glucose Fasting glucose Fasting glucose IPGTT glucose Fed glucose IPGTT glucose 60-min IPGTT glucose 60-min IPGTT glucose

(NON/Lt NZO/HI) NON (NON/Lt NZO/HI) NON (NON/Lt NZO/HI) NON (NON/Lt NZO/HI) NON (NON/Lt NZO/HI) NON (NON/Lt NZO/HI) NON BKS DBA/2 F2 (Lepr-db/db) BKS DBA/2 F2 (Lepr-db/) BKS DBA/2 F2 (Lepr-db/db) BKS DBA/2 F2 (Lepr-db/db) BKS DBA/2 F2 (Lepr-db/db) BKS DBA/2 F2 (Lepr-db/) BKS DBA/2 F2 (Lepr-db/)

174 Also interaction with maternal environment 174 Also interaction with maternal environment 174 Also interaction with maternal environment 174 174 Also interaction with maternal environment 174 Also interaction with maternal environment 126 Females 126 Males 126 Males 126 Females 126 Males 126 Males 126 Males

tanabe and colleagues (247249) created congenic strains in which segments from the BN strain have been introgressed into the OLETF background. Their detailed finemapping of this region to an interval of approximately 500 kb recently led to the identification of Gpr10, a G proteincoupled receptor, as most likely the gene underlying this Dmo1 locus (248, 249). The OLETF strain has a mutation in the start codon, causing translation to initiate at one of two downstream ATG codons, only one of which is in-frame. However, the primary phenotype in rats with this mutant receptor appears to be increased food intake; effects on glucose tolerance are secondary to this defect (249), and thus it is most specifically an obesity QTL. The authors also identified this mutation in GK rats and Zucker Diabetic Fatty (ZDF) rats, suggesting that it may play a role in the development of insulin resistance secondary to body weight gain in several strains. Notably, this gene is likely contained within the Niddm1i strain; however, it is unknown at this point whether this gene contributes to the insulin resistance phenotypes of this strain. Gpr10 resides outside the body weight region recently fine-mapped within Niddm1i (32). These loci (mouse chromosome 19 and rat chromosome 1) are syntenic with human chromosome 10q, where several diabetes-related loci have also been mapped. In fact, 10q is one of the well-replicated human type 2 diabetes susceptibility regions (6, 7, 9). A study of Mexican-Americans in San Antonio found significant linkage for age of onset and risk of type 2 diabetes to a broad region, with a peak LOD at 150 Mb on chromosome 10q (10). Significant linkage of diabetes with the same region on chromosome 10q was also detected in a study of 719 Finnish sib pairs (250). Several other groups have also mapped loci affecting diabetes risk to this broad region, including loci in the Pima Indians, British, African-American, Icelandic, Dutch, and French cohorts (251263). Using a very high density of markers across this region, Grant et al. (26) have recently identified the TCF7L2 gene with a remarkably strong association with type 2 diabetes risk in Icelandic individuals. TCF7L2 (also known as TCF4) is a transcription factor involved in the Wnt signaling pathway through interactions with -cate-

nin, and it plays an important role in the development of the enteroendocrine system (264, 265). Thus, in addition to a gene affecting food intake, three potential type 2 diabetes susceptibility genes have been identified from these studies: SorCS1 in the mouse, Ide in the rat, and TCF7L2 in the Icelandic population (26, 148, 239). The corresponding Mb positions of these genes are: Ide (mouse chr19:37 Mb, human chr10:94 Mb, rat chr1:242 Mb), SorCS1 (mouse chr19:50 Mb, human chr10:109 Mb, rat chr1: 256 Mb), and Tcf7l2 (mouse chr19:56 Mb, human chr10:115 Mb, rat chr1:262 Mb). The IDE locus has been extensively surveyed in human genetic studies of diabetes. Several association studies offer the IDE locus as a candidate for diabetes susceptibility (266 268). However, a recent report that included both linkage and association studies in over 4000 individuals failed to implicate IDE in diabetes (269). Thus, the role of this gene in human type 2 diabetes susceptibility remains unclear. TCF7L2 and SorCS1 have both been reported recently, and very recently several studies have confirmed association of TCF7L2 SNPs and type 2 diabetes (270 274).
B. Mouse chromosome 9, rat chromosome 8, and human chromosome 11

Mouse chromosome 9 contains several diabetes-related loci between 36 and 49 Mb. Four studies have identified this region as associated with increased plasma glucose or increased glucose levels during an IPGTT. QTL for increased glucose were identified from the NZO strain in a cross with the NON strain (173) and in a cross between KK mice with the Ay agouti allele and C57BL/6 (194). Glucose intolerance QTL were mapped in crosses between C57BLKS/J and DBA/2 mice homozygous for the Leprdb mutation, whereas decreased insulin levels during a glucose tolerance test were mapped in a cross between C57BL/6 and C3H mice (98, 122, 126). The syntenic region in rat is chromosome 8 (38 53 Mb). Two rat models have identified linkages in this region. Moralejo et al. (275) identified a linkage to glucose levels during an oral glucose tolerance test in the OLETF rat. Interestingly, heterozygotes at this locus had the highest glucose levels, suggesting an

70

Endocrine Reviews, February 2007, 28(1):48 83

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice

interaction between the OLETF and F344 alleles. Linkage to postchallenge glucose levels was also observed in the Spontaneously Diabetic Torii (SDT) rat, a model of nonobese type 2 diabetes with severe retinopathy (276, 277). This region corresponds to human chromosome 11 (113 125 Mb). The peak markers of five different studies fall within this region. Hanson et al. (278) identified a significant linkage to type 2 diabetes susceptibility in the Pima Indians at 123 Mb. Putative linkages for type 2 diabetes were also observed in Caucasian families from Utah and the Netherlands (253, 279). Suggestive linkage for increased plasma glucose levels was detected in this region in individuals from the Framingham study (280), and studies of Mexican-Americans from San Antonio have identified a QTL for plasma insulin levels (281).
C. Mouse chromosome 11, rat chromosome 10, and human chromosome 17p

The 62- to 71-Mb region of mouse chromosome 11 harbors five loci for diabetes-related traits, identified in a diverse set of strains. A locus for decreased plasma insulin levels was identified in the NZO strain (173), whereas the M16 strain has a QTL for increased insulin levels in this region (214). Phenotypes related to glucose intolerance (increased glucose levels and area under the curve) were identified in studies of the SMXA-5 RI strain (154), and also in the NSY strain (204). Finally, a cross between C57BLKS/J and HRS mice homozygous for the fat mutation found a locus affecting plasma glucose levels in this region (123). The syntenic region of the rat genome is chromosome 10, approximately 48 58 Mb. The Niddm3 QTL for traits related to fasting glucose levels and glucose intolerance in the GK strain has been observed in a broad region that overlaps this syntenic region (236). This region is syntenic to the tip of the p-arm of human chromosome 17 (520 Mb), where several diabetes-related linkages have also been detected. Studies of Finnish sib pairs from the FUSION group have identified significant QTL affecting fasting insulin, the insulin/glucose ratio, 2-h insulin levels during a glucose tolerance test, and insulin sensitivity in this region (282). A meta-analysis of four European genome scans has identified significant evidence of a type 2 diabetes susceptibility locus at approximately 14 Mb (283). A significant QTL for leptin levels that interacts with a locus on chromosome 3 affecting insulin levels and the insulin/glucose ratio has also been found in this region (284). Six linkages that did not reach the genomewide significance threshold have been identified in this region in cohorts from Japan, the United Kingdom, the United States, Finland, the Netherlands, and West Africa (253, 263, 285288), providing additional evidence of the importance of this region in diabetes susceptibility.
D. Mouse chromosome 12, rat chromosome 6, and human chromosome 14

in 129 mice was detected in a cross of C57BL/6 129 Insr/ mice (134), whereas a C57BL/6-derived locus for increased fed glucose and diabetes was observed in a cross of Insr/ Irs1/ mice from these strains (131). It is unclear at this time whether these represent different descriptions of the same locus or are distinct loci. The corresponding region in the rat is chromosome 6, from approximately 110 125 Mb. Only one QTL on rat chromosome 6 has been detected: a locus affecting glucose intolerance in the SDT rat (276). The peak marker for this QTL is D6Mgh1 (at 136 Mb). Although this peak is not specifically in the region syntenic to the LOD peaks from the mouse studies, it is very likely that there is at least some overlap of the confidence intervals between these studies. Thus, it is possible that this represents a locus corresponding to those observed in mice. Five studies have identified linkages in the corresponding region of human chromosome 14 (70 90 Mb). A significant QTL for fasting insulin levels was detected in Finnish sib pairs (282). This region also houses a significant linkage to type 2 diabetes in the Pima (278), although a second study in this cohort suggesting linkage with maternal inheritance at this region did not reach statistical significance (289). Other studies, which did not reach genomewide significance levels, included a linkage to type 2 diabetes in Ashkenazi Jews (290), a QTL for glucose utilization in African-Americans and Mexican-Americans from the Insulin Resistance Atherosclerosis Study (13), and a complex metabolic syndrome trait that includes HOMA as a component in the National Heart, Lung and Blood Institute Family Heart Study (291).
E. Other type 2 diabetes susceptibility genes identified in the rat

Mouse chromosome 12 (79 99 Mb) also harbors several QTL for diabetes-related traits. Loci for increased glucose in the KK strain (195) and insulin levels in the NZO strain have been detected (173). A locus for increased fed insulin levels

Although not major hot spots in the mouse, two other potential diabetes susceptibility genes have been identified in rat studies. Work by Aitman et al. (292) has demonstrated that Cd36 accounts for part of the adipocyte insulin resistance locus mapped near D4Bro1 on rat chromosome 4 in the Spontaneously Hypertensive Rat (SHR). CD36 is a class B scavenger receptor and fatty acid translocase with numerous functions (293). The defect in the SHR rat results from a complex gene duplication/deletion event resulting in absent expression of the wild-type gene, and the phenotype could be rescued by transgenic overexpression of wild-type CD36 in the mutant strain (294). The importance of this gene was initially questioned because not all SHR strains have this Cd36 mutation, and the overall phenotype of the strains is similar, but the SHR/ Izm colony lacking the mutation shows little evidence of an insulin resistance QTL on this region of chromosome 4, consistent with the idea that Cd36 is an insulin resistance gene in some SHR strains (295, 296). In rats, this locus is associated with reduced insulin-stimulated glucose uptake into adipocytes and decreased catecholamine-stimulated free fatty acid secretion (292). Overexpression of Cd36 in these rats improved oral glucose tolerance and increased muscle glucose uptake (294). These findings predict that loss of Cd36 should be associated with

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice

Endocrine Reviews, February 2007, 28(1):48 83 71

insulin resistance. However, CD36-deficient mice have decreased, not increased, fasting (overnight) glucose and insulin levels, and on a chow diet, CD36 knockouts are more insulin sensitive (297, 298). Whole body glucose uptake is increased, primarily due to increased uptake in muscle; however, insulin fails to inhibit hepatic glucose production, and ketone bodies are elevated (299). These data are consistent with increased glucose utilization in muscle in the presence of impaired fatty acid uptake by this tissue, diverting excess fatty acids to the liver for oxidation and storage. In response to high-carbohydrate feeding, however, the knockout mice are more insulin resistant (298). None of these studies have examined adipocytes. Muscle-specific overexpression in mice was associated with increased fasting glucose and insulin (300). Thus, the differences with the rat may relate to tissue-specific effects of loss of CD36, differences in dietary conditions, or perhaps species differences in the relative utilization of free fatty acid and glucose. Nonetheless, these data support a role of Cd36 in the determination of insulin sensitivity. Mouse Cd36 is found at 17 Mb on chromosome 5. Given the apparent species differences in the effects of this gene, it is perhaps not surprising that no QTL have been mapped to this region in the mouse. CD36 is located on human chromosome 7, at approximately 80 Mb, where several QTL for diabetes-related traits have been observed (12, 262, 301, 302). Some, but not all, studies have shown increased plasma glucose levels, insulin resistance, and/or a relationship to diabetes in humans with CD36 deficiency (303307). Recently, Corpeleijn et al. (308) have described the first association of SNPs in this gene with insulin resistance, although these findings have not yet been replicated. As suggested from the rodent data, the role of CD36 in the development of insulin resistance may be dependent upon environmental factors such as diet. The cholecystokinin A receptor (Cckar, Cck1r) is another potential diabetes susceptibility gene that has been identified in the rat. Hirashima et al. (309) first reported a linkage for impaired glucose tolerance in the OLETF rat to chromosome 14, near marker D14Mit4, which they called Odb2. This linkage was independently replicated, and this region was also shown to be associated with reduced pancreatic proliferative capacity after pancreatectomy (243, 310). About the same time, it was discovered that the OLETF strain has an approximately 7-kb deletion that removes the promoter and first two exons of the nearby (17-Mb distal) Cckar gene (311). A role for Cckar genotype in glucose intolerance was suggested by showing that F2 rats homozygous for the deleted Cckar have impaired glucose tolerance compared with the cohort of F2 animals with the intact gene, but in comparing F2 to backcross rats both homozygous for the disrupted Cckar, it was also determined that additional OLETF alleles contribute to the glucose intolerance of this strain (312). Functionally, signaling through the Cckar is associated with secretion of insulin, somatostatin, and leptin, also with gastric emptying and gastric and intestinal motility, and with the acute inhibition of food intake, all of which might be related to the development of glucose intolerance (313). These data suggest that the Cckar is a candidate gene for this QTL, and

indeed numerous phenotypes observed in the OLETF strain have been attributed to the absence of the Cckar in these rats. Although intriguing, these studies do not provide genetic validation that the Cckar is the gene underlying this QTL or conclusively show which phenotypes in the OLETF rat are due to the absence of this receptor. Consistent with the F2 vs. backcross comparison, numerous additional QTL affecting body weight and glucose tolerance have been detected in this strain, including at least three others on rat chromosome 14 (233, 243246, 275, 314 316). Furthermore, the phenotype of Cckar knockout mice does not replicate that of OLETF rats (317, 318). Consistent with its role in food intake, Cckardeficient mice fail to decrease food intake in response to cholecystokinin (CCK); however, they show no difference in body weight, glucose levels, or pancreatic morphology (318). These differences may be due to species-specific effects or to interactions with other OLETF alleles, but it is also possible that the OLETF phenotype mapped to this region is not entirely due to the Cckar mutation. Notably, the Ppargc1a gene is less than 3 Mb from the Cckar gene and would also be an excellent candidate. In addition, because the Cckar mediates food intake, could the glucose intolerance phenotype be secondary to differences in obesity? Wei et al. (243) mapped body weight in their study and did not detect significant linkage to body weight here (Nidd10/of), although they did detect an interaction of this locus with body weight in determining postchallenge glucose levels. A large-insert congenic strain has been created, in which a greater than 40-Mb region of OLETF chromosome 14 has been introgressed into the F344 background (245). This F.O-Nidd10/of congenic strain maintains differences in postchallenge glucose levels but does not show differences in body weight, suggesting that the glucose intolerance phenotype is independent of body weight differences. The Cckar gene is located on mouse chromosome 5, at approximately 53 Mb. QTL near this region have been detected in the NZO (173) and FVB (187) strains. In humans, the CCKAR is located on chromosome 4 (26 Mb), and as described above is very near the PPARGC1A gene, which has been associated with type 2 diabetes susceptibility (21). A QTL for HbA1c levels in the Old Order Amish overlaps this region (12), and a type 2 diabetes QTL was observed nearby in Dutch individuals (253). A single study has suggested associations of SNPs in the promoter region of CCKAR with increased body fat and plasma insulin levels (319), and another study has detected amino acid variants in this gene in obese individuals (320). Thus, at this point, perhaps it is best to describe the Cckar as a candidate for diabetes susceptibility; however, additional evidence, either through further genetic studies or by restoring Cckar expression in OLETF rats, would greatly strengthen the argument that this is a diabetes susceptibility gene.
F. Hot spots in human linkage studies

What about the other major regions of human type 2 diabetes susceptibility genes on chromosomes 1q, 2q, 3p, 3q, 8p, 12q, 18, and 20q? Have any of these regions also been detected in the mouse? In short, the answer is yes. Mouse

72

Endocrine Reviews, February 2007, 28(1):48 83

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice

genetics has identified linkages in the syntenic regions corresponding to all these areas, with the 1q and 2q loci particularly evident. Nearly 50 QTL have been mapped on human chromosome 1q in the region from approximately 150 250 Mb (11, 12, 251, 252, 254, 256 258, 260 263, 278 280, 282, 283, 289, 321337). In mouse, this corresponds to regions of chromosome 3, (90 Mb), chromosome 1 (130 194 Mb), chromosome 8 (123125 Mb), and chromosome 13 (13 Mb). Six QTL for glucose or insulin levels in the regions of mouse chromosome 1 have been detected in the TSOD, KK, A/J, LG, and the wildderived PERA/Ei strains (155, 193, 198, 218, 224, 338), as has another for glucose intolerance in the small segment of chromosome 13 (67). Regions of rat chromosomes 2 (180 190 Mb), 13 (40 111 Mb), 17 (70 Mb), and 19 (5556 Mb) are syntenic to human chromosome 1q. The Nidd/gk2, Niddm2, and Nidd/ gk6 QTL in the GK rat (236, 237), the Ir2 QTL in the SHR strain (339), and the Insul1 QTL identified in a cross between the Lyon Hypertensive (LH) and normotensive (LN) strains are located within these regions (340). Human chromosome 2q (110 240 Mb) has roughly 25 QTL for diabetes and diabetes-related traits (12, 257, 258, 262, 263, 279, 281, 285, 291, 301, 321, 322, 327, 337, 341345). This region contains the CAPN10 gene, identified as a type 2 diabetes susceptibility gene through positional cloning, and for which growing evidence favors its role in this disease (21, 346 348). In addition, this region contains the NEUROD gene, which has been implicated in MODY (349), and the IRS1 gene, where SNPs have been significantly associated with type 2 diabetes in a meta-analysis (350). This broad region has syntenic regions primarily on mouse chromosomes 1 (48 94 Mb) and 2 (45 82 Mb), where several diabetes-related traits have been mapped in the KK, LG, SM, C57BL/6, TSOD, CAST/Ei, and A/J strains (67, 134, 154, 159, 160, 194, 214, 217, 219, 224, 226). In the rat, the majority of this region is syntenic to chromosomes 3 and 9 (20 70 and 4590 Mb, respectively). The region on chromosome 9 contains the Nidd8/of QTL from the OLETF strain (243245). Capn10 is within this region, and a Gly3 Ser mutation at amino acid 195 has been identified in the OLETF strain, although the functional consequences of this variant are unclear (351). Regions on both arms of human chromosome 3 have numerous QTL (10, 11, 252, 253, 256, 258, 261, 262, 280, 282, 284, 302, 328, 331333, 341, 342, 344, 352357). The p-arm (80 Mb), which contains PPARG, is syntenic to regions of mouse chromosomes 6, 9, 14, and 16. On mouse chromosome 6, a QTL for plasma glucose levels in the A/J strain has been detected (153, 155), and there is a potential overlap with the Nidd3/nsy locus (204). A QTL from C57BL/6 has been found in the syntenic region on chromosome 14 (130, 131). In the rat, this region has synteny to chromosomes 4, 8, 9, 15, and 16. A locus for increased postprandial glucose levels in the OLETF rat near D4Mgh7 may overlap with this region (246), and a QTL on chromosome 16, identified in a cross of Wistar and Zucker Leprfa/fa rats, is also in the corresponding region (358). The q-arm (140 200 Mb) corresponds to parts of mouse chromosomes 3, 9, and 16. Again, only a single mouse QTL associated with increased insulin in the SM strain is situated in the major syntenic regions (126). Segments of rat chromosomes 2, 8, and 11 are syntenic to this region. The

Nidd4/of QTL for plasma glucose levels in the OLETF strain is contained within the corresponding region of chromosome 11 (244, 245, 275). Additional QTL have been detected in the central region of human chromosome 3. Corresponding regions are found on mouse chromosomes 6, 9, 14, and 16. Human chromosome 8p (0 40 Mb), with roughly a dozen QTL (251, 253255, 258, 263, 279, 285, 301, 322, 328, 341, 353, 359, 360), has syntenic regions on mouse chromosomes 8 and 14. Three QTL for glucose intolerance have been detected in the KK, DBA/2 (chromosome 8), and C57BL/6 (chromosome 14) strains (126, 130, 194, 198). Rat chromosomes 15 and 16 harbor the syntenic regions; however, no QTL have been detected in the corresponding areas. Several loci reaching genomewide significance have been detected around approximately 70 Mb, and others near 120 Mb on human chromosome 12 (291, 352, 361364), along with many other suggestive QTL in these regions (13, 252, 257, 259, 263, 283, 286, 287, 301, 321, 328, 335, 337, 352, 359). In the mouse, the primary syntenic regions are chromosome 10 (100 120Mb) and chromosome 5 (115130 Mb). Diabetesrelated QTL have been found in both these locations from the SM, B6, and C57BLKS/J strains (123, 134, 153155, 219). Chromosomes 7 (50 60 Mb) and 12 (40 Mb) in the rat are syntenic to these loci; however, no QTL have been found in either of these areas. Linkages have been observed spanning human chromosome 18 (12, 252, 253, 255, 258, 259, 279, 280, 302, 323, 324, 332334, 337, 343345, 352, 354, 359). There is some evidence of mouse QTL in the corresponding syntenic regions that are primarily located on chromosomes 17 and 18 (153, 173). Rats have syntenic regions on chromosomes 9, 13, and 18, but we have not found any QTL reported in these locations. Thus, in contrast to the other hot spots, the QTL on human chromosome 18 have not been detected in the rodent models. Finally, human chromosome 20q is another hot spot containing several significant QTL. This region is entirely syntenic to mouse chromosome 2 (150 180 Mb). Somewhat surprisingly given the number of significant linkages in this region, and in contrast to the well-replicated regions on chromosomes 1q and 2q, only three mouse diabetes-related QTL have been observed in this region (99, 159, 214). Two of these are loci detected in the C57BL/6 strain, in crosses with either C3H or CAST/Ei, whereas the other was identified in the M16 strain. On the syntenic region of rat chromosome 3 is a QTL for response to pancreatectomy (365). The p-arm of human chromosome 20 also has several QTL (250, 253, 260, 261, 287, 290, 324, 337, 366). No QTL have been detected in the syntenic region of mouse chromosome 2, but in the rat, two additional QTL on chromosome 3 are syntenic to this region (339, 367).

IX. Summary/Conclusion

Genetic approaches to the identification of diabetes susceptibility genes are now becoming successful. These experiments are slow and painstaking. Sufficient time has only now elapsed since the original genome scans to begin

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice

Endocrine Reviews, February 2007, 28(1):48 83 73 3. National Diabetes Information Clearinghouse 2005 National Diabetes Statistics. No. 06-3892. Bethesda, MD: U.S. Department of Health and Human Services, National Institutes of Health (http:// diabetes.niddk.nih.gov/dm/pubs/statistics/index.htm#7) 4. Narayan KM, Boyle JP, Thompson TJ, Sorensen SW, Williamson DF 2003 Lifetime risk for diabetes mellitus in the United States. JAMA 290:1884 1890 5. Zimmet P, Alberti KG, Shaw J 2001 Global and societal implications of the diabetes epidemic. Nature 414:782787 6. Florez JC, Hirschhorn J, Altshuler D 2003 The inherited basis of diabetes mellitus: implications for the genetic analysis of complex traits. Annu Rev Genomics Hum Genet 4:257291 7. Permutt MA, Wasson J, Cox N 2005 Genetic epidemiology of diabetes. J Clin Invest 115:14311439 8. van Tilburg J, van Haeften TW, Pearson P, Wijmenga C 2001 Defining the genetic contribution of type 2 diabetes mellitus. J Med Genet 38:569 578 9. Das SK, Elbein SC 2006 The genetic basis of type 2 diabetes. Cellscience 2:100 131 10. Duggirala R, Blangero J, Almasy L, Dyer TD, Williams KL, Leach RJ, OConnell P, Stern MP 1999 Linkage of type 2 diabetes mellitus and of age at onset to a genetic location on chromosome 10q in Mexican Americans. Am J Hum Genet 64:11271140 11. Panhuysen CI, Cupples LA, Wilson PW, Herbert AG, Myers RH, Meigs JB 2003 A genome scan for loci linked to quantitative insulin traits in persons without diabetes: the Framingham Offspring Study. Diabetologia 46:579 587 12. Hsueh WC, St Jean PL, Mitchell BD, Pollin TI, Knowler WC, Ehm MG, Bell CJ, Sakul H, Wagner MJ, Burns DK, Shuldiner AR 2003 Genome-wide and fine-mapping linkage studies of type 2 diabetes and glucose traits in the Old Order Amish: evidence for a new diabetes locus on chromosome 14q11 and confirmation of a locus on chromosome 1q21 q24. Diabetes 52:550 557 13. Rich SS, Bowden DW, Haffner SM, Norris JM, Saad MF, Mitchell BD, Rotter JI, Langefeld CD, Wagenknecht LE, Bergman RN 2004 Identification of quantitative trait loci for glucose homeostasis: the Insulin Resistance Atherosclerosis Study (IRAS) Family Study. Diabetes 53:1866 1875 14. Baier LJ, Hanson RL 2004 Genetic studies of the etiology of type 2 diabetes in Pima Indians: hunting for pieces to a complicated puzzle. Diabetes 53:11811186 15. An P, Freedman BI, Hanis CL, Chen YD, Weder AB, Schork NJ, Boerwinkle E, Province MA, Hsiung CA, Wu X, Quertermous T, Rao DC 2005 Genome-wide linkage scans for fasting glucose, insulin, and insulin resistance in the National Heart, Lung, and Blood Institute Family Blood Pressure Program: evidence of linkages to chromosome 7q36 and 19q13 from meta-analysis. Diabetes 54:909 914 16. Poulsen P, Levin K, Petersen I, Christensen K, Beck-Nielsen H, Vaag A 2005 Heritability of insulin secretion, peripheral and hepatic insulin action, and intracellular glucose partitioning in young and old Danish twins. Diabetes 54:275283 17. Mills GW, Avery PJ, McCarthy MI, Hattersley AT, Levy JC, Hitman GA, Sampson M, Walker M 2004 Heritability estimates for -cell function and features of the insulin resistance syndrome in UK families with an increased susceptibility to type 2 diabetes. Diabetologia 47:732738 18. Poulsen P, Kyvik KO, Vaag A, Beck-Nielsen H 1999 Heritability of type II (non-insulin-dependent) diabetes mellitus and abnormal glucose tolerancea population-based twin study. Diabetologia 42:139 145 19. McCarthy MI 2004 Progress in defining the molecular basis of type 2 diabetes mellitus through susceptibility-gene identification. Hum Mol Genet 13 Spec No 1:R33R41 20. Bonnycastle LL, Willer CJ, Conneely KN, Jackson AU, Burrill CP, Watanabe RM, Chines PS, Narisu N, Scott LJ, Enloe ST, Swift AJ, Duren WL, Stringham HM, Erdos MR, Riebow NL, Buchanan TA, Valle TT, Tuomilehto J, Bergman RN, Mohlke KL, Boehnke M, Collins FS 2006 Common variants in maturity-onset diabetes of the young genes contribute to risk of type 2 diabetes in Finns. Diabetes 55:2534 2540 21. Parikh H, Groop L 2004 Candidate genes for type 2 diabetes. Rev Endocr Metab Disord 5:151176 22. Altshuler D, Hirschhorn JN, Klannemark M, Lindgren CM, Vohl

to allow identification of genes underlying these QTL. The identification of novel and relevant loci through mouse genetics holds great promise, because the QTL replicate the major loci observed in human genome scans. The identification of the underlying genes will be greatly facilitated by availability of the genome sequence of several strains and genomewide SNP genotypes in numerous strains, which will provide essentially unlimited markers for analysis. Identification of disease genes will also be facilitated through the projects aimed at identifying all the genes and transcribed sequences and their expression patterns. Furthermore, integrating data from multiple sources (e.g., high-density SNP genotyping, examination of multiple strains, microarray data, proteomic and metabolomic data) may prove especially useful (57, 368). We predict that in the next few years we will see a tremendous increase in the cloning and validation of novel diabetes genes, providing new insights into this disease process.
X. Appendix: Useful Web Site Resources for Mouse Genetics Research

UCSC mouse genome assembly: http://www.genome.ucsc.edu/ Ensembl genome assembly: http://www.ensembl.org/ Mouse Genome Informatics: http://www.informatics.jax.org/ Mouse Phenome Database: http://www.jax.org/phenome National Center for Biotechnology Information (NCBI) mouse genome resources: http://www.ncbi.nlm.nih.gov/genome/guide/mouse/ dbSNP for mouse SNP data: http://www.ncbi.nlm.nih.gov/SNP/MouseSNP.cgi Mouse-human-rat homology maps: http://www.ncbi.nlm.nih.gov/Homology/
Acknowledgments
We thank Drs. Edward H. Leiter, A. Jake Lusis, and Clayton E. Mathews for their careful reading of this manuscript and their insightful comments and corrections, and Adam Steinberg for his expert assistance with the preparation of the figures. Address all correspondence and requests for reprints to: Alan D. Attie, Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, Wisconsin 53706-1544. E-mail: attie@biochem.wisc.edu Our diabetes research is currently supported by National Institutes of Health Grants DK58037 and DK66369. S.M.C. is supported by an American Heart Association Scientist Development grant (0635234N). Disclosure Statement: The authors have nothing to disclose.

References
1. American Diabetes Association 2006 Diagnosis and classification of diabetes mellitus. Diabetes Care 29(Suppl 1):S43S48 2. Naggert JK, Svenson KL, Smith RV, Paigen B, Peters LL May 2006 Diet effects on bone mineral density and content, body composition, and plasma glucose, leptin, and insulin levels. MPD:143. Mouse Phenome Database (http://www.jax.org/phenome), The Jackson Laboratory, Bar Harbor, Maine

74

Endocrine Reviews, February 2007, 28(1):48 83 MC, Nemesh J, Lane CR, Schaffner SF, Bolk S, Brewer C, Tuomi T, Gaudet D, Hudson TJ, Daly M, Groop L, Lander ES 2000 The common PPAR Pro12Ala polymorphism is associated with decreased risk of type 2 diabetes. Nat Genet 26:76 80 Neel JV 1962 Diabetes mellitus: a thrifty genotype rendered detrimental by progress? Am J Hum Genet 14:353362 Diamond J 2003 The double puzzle of diabetes. Nature 423:599 602 International HapMap Consortium 2005 A haplotype map of the human genome. Nature 437:1299 1320 Grant SF, Thorleifsson G, Reynisdottir I, Benediktsson R, Manolescu A, Sainz J, Helgason A, Stefansson H, Emilsson V, Helgadottir A, Styrkarsdottir U, Magnusson KP, Walters GB, Palsdottir E, Jonsdottir T, Gudmundsdottir T, Gylfason A, Saemundsdottir J, Wilensky RL, Reilly MP, Rader DJ, Bagger Y, Christiansen C, Gudnason V, Sigurdsson G, Thorsteinsdottir U, Gulcher JR, Kong A, Stefansson K 2006 Variant of transcription factor 7-like 2 (TCF7L2) gene confers risk of type 2 diabetes. Nat Genet 38:320 323 McCarthy MI, Froguel P 2002 Genetic approaches to the molecular understanding of type 2 diabetes. Am J Physiol Endocrinol Metab 283:E217E225 ORahilly S, Barroso I, Wareham NJ 2005 Genetic factors in type 2 diabetes: the end of the beginning? Science 307:370 373 Ikegami H, Fujisawa T, Ogihara T 2004 Mouse models of type 1 and type 2 diabetes derived from the same closed colony: genetic susceptibility shared between two types of diabetes. ILAR J 45:268 277 Galli J, Fakhrai-Rad H, Kamel A, Marcus C, Norgren S, Luthman H 1999 Pathophysiological and genetic characterization of the major diabetes locus in GK rats. Diabetes 48:24632470 Lin JM, Ortsater H, Fakhrai-Rad H, Galli J, Luthman H, Bergsten P 2001 Phenotyping of individual pancreatic islets locates genetic defects in stimulus secretion coupling to Niddm1i within the major diabetes locus in GK rats. Diabetes 50:27372743 Granhall C, Park HB, Fakhrai-Rad H, Luthman H 1 September 2006 High-resolution QTL analysis reveals multiple diabetes susceptibility loci mapped to intervals less than 800-kb in the species conserved Niddm1i of the GK rat. Genetics 10.1534/genetics.106.062208 Ideraabdullah FY, de la Casa-Esperon E, Bell TA, Detwiler DA, Magnuson T, Sapienza C, de Villena FP 2004 Genetic and haplotype diversity among wild-derived mouse inbred strains. Genome Res 14:1880 1887 Wade CM, Kulbokas 3rd EJ, Kirby AW, Zody MC, Mullikin JC, Lander ES, Lindblad-Toh K, Daly MJ 2002 The mosaic structure of variation in the laboratory mouse genome. Nature 420:574 578 Wade CM, Daly MJ 2005 Genetic variation in laboratory mice. Nat Genet 37:11751180 Petkov PM, Ding Y, Cassell MA, Zhang W, Wagner G, Sargent EE, Asquith S, Crew V, Johnson KA, Robinson P, Scott VE, Wiles MV 2004 An efficient SNP system for mouse genome scanning and elucidating strain relationships. Genome Res 14:1806 1811 Wiltshire T, Pletcher MT, Batalov S, Barnes SW, Tarantino LM, Cooke MP, Wu H, Smylie K, Santrosyan A, Copeland NG, Jenkins NA, Kalush F, Mural RJ, Glynne RJ, Kay SA, Adams MD, Fletcher CF 2003 Genome-wide single-nucleotide polymorphism analysis defines haplotype patterns in mouse. Proc Natl Acad Sci USA 100:3380 3385 Yalcin B, Fullerton J, Miller S, Keays DA, Brady S, Bhomra A, Jefferson A, Volpi E, Copley RR, Flint J, Mott R 2004 Unexpected complexity in the haplotypes of commonly used inbred strains of laboratory mice. Proc Natl Acad Sci USA 101:9734 9739 Frazer KA, Wade CM, Hinds DA, Patil N, Cox DR, Daly MJ 2004 Segmental phylogenetic relationships of inbred mouse strains revealed by fine-scale analysis of sequence variation across 4.6 mb of mouse genome. Genome Res 14:14931500 Ingalls AM, Dickie MM, Snell GD 1950 Obese, a new mutation in the house mouse. J Hered 41:317318 Coleman DL 1978 Obese and diabetes: two mutant genes causing diabetes-obesity syndromes in mice. Diabetologia 14:141148 Chua Jr SC, Chung WK, Wu-Peng XS, Zhang Y, Liu SM, Tartaglia L, Leibel RL 1996 Phenotypes of mouse diabetes and rat fatty due to mutations in the OB (leptin) receptor. Science 271:994 996

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice 43. Bultman SJ, Michaud EJ, Woychik RP 1992 Molecular characterization of the mouse agouti locus. Cell 71:11951204 44. Coleman DL, Eicher EM 1990 Fat (fat) and tubby (tub): two autosomal recessive mutations causing obesity syndromes in the mouse. J Hered 81:424 427 45. Naggert JK, Fricker LD, Varlamov O, Nishina PM, Rouille Y, Steiner DF, Carroll RJ, Paigen BJ, Leiter EH 1995 Hyperproinsulinaemia in obese fat/fat mice associated with a carboxypeptidase E mutation which reduces enzyme activity. Nat Genet 10:135142 46. Noben-Trauth K, Naggert JK, North MA, Nishina PM 1996 A candidate gene for the mouse mutation tubby. Nature 380:534 538 47. Yoshioka M, Kayo T, Ikeda T, Koizumi A 1997 A novel locus, Mody4, distal to D7Mit189 on chromosome 7 determines early-onset NIDDM in nonobese C57BL/6 (Akita) mutant mice. Diabetes 46:887 894 48. Mural RJ, Adams MD, Myers EW, Smith HO, Miklos GL, Wides R, Halpern A, Li PW, Sutton GG, Nadeau J, Salzberg SL, Holt RA, Kodira CD, Lu F, Chen L, Deng Z, Evangelista CC, Gan W, Heiman TJ, Li J, Li Z, Merkulov GV, Milshina NV, Naik AK, Qi R, Shue BC, Wang A, Wang J, Wang X, Yan X, Ye J, Yooseph S, Zhao Q, Zheng L, Zhu SC, Biddick K, Bolanos R, Delcher AL, Dew IM, Fasulo D, Flanigan MJ, Huson DH, Kravitz SA, Miller JR, Mobarry CM, Reinert K, Remington KA, Zhang Q, Zheng XH, Nusskern DR, Lai Z, Lei Y, Zhong W, Yao A, Guan P, Ji RR, Gu Z, Wang ZY, Zhong F, Xiao C, Chiang CC, Yandell M, Wortman JR, Amanatides PG, Hladun SL, Pratts EC, Johnson JE, Dodson KL, Woodford KJ, Evans CA, Gropman B, Rusch DB, Venter E, Wang M, Smith TJ, Houck JT, Tompkins DE, Haynes C, Jacob D, Chin SH, Allen DR, Dahlke CE, Sanders R, Li K, Liu X, Levitsky AA, Majoros WH, Chen Q, Xia AC, Lopez JR, Donnelly MT, Newman MH, Glodek A, Kraft CL, Nodell M, Ali F, An HJ, Baldwin-Pitts D, Beeson KY, Cai S, Carnes M, Carver A, Caulk PM, Center A, Chen YH, Cheng ML, Coyne MD, Crowder M, Danaher S, Davenport LB, Desilets R, Dietz SM, Doup L, Dullaghan P, Ferriera S, Fosler CR, Gire HC, Gluecksmann A, Gocayne JD, Gray J, Hart B, Haynes J, Hoover J, Howland T, Ibegwam C, Jalali M, Johns D, Kline L, Ma DS, MacCawley S, Magoon A, Mann F, May D, McIntosh TC, Mehta S, Moy L, Moy MC, Murphy BJ, Murphy SD, Nelson KA, Nuri Z, Parker KA, Prudhomme AC, Puri VN, Qureshi H, Raley JC, Reardon MS, Regier MA, Rogers YH, Romblad DL, Schutz J, Scott JL, Scott R, Sitter CD, Smallwood M, Sprague AC, Stewart E, Strong RV, Suh E, Sylvester K, Thomas R, Tint NN, Tsonis C, Wang G, Wang G, Williams MS, Williams SM, Windsor SM, Wolfe K, Wu MM, Zaveri J, Chaturvedi K, Gabrielian AE, Ke Z, Sun J, Subramanian G, Venter JC, Pfannkoch CM, Barnstead M, Stephenson LD 2002 A comparison of whole-genome shotgun-derived mouse chromosome 16 and the human genome. Science 296:16611671 49. Waterston RH, Lindblad-Toh K, Birney E, Rogers J, Abril JF, Agarwal P, Agarwala R, Ainscough R, Alexandersson M, An P, Antonarakis SE, Attwood J, Baertsch R, Bailey J, Barlow K, Beck S, Berry E, Birren B, Bloom T, Bork P, Botcherby M, Bray N, Brent MR, Brown DG, Brown SD, Bult C, Burton J, Butler J, Campbell RD, Carninci P, Cawley S, Chiaromonte F, Chinwalla AT, Church DM, Clamp M, Clee C, Collins FS, Cook LL, Copley RR, Coulson A, Couronne O, Cuff J, Curwen V, Cutts T, Daly M, David R, Davies J, Delehaunty KD, Deri J, Dermitzakis ET, Dewey C, Dickens NJ, Diekhans M, Dodge S, Dubchak I, Dunn DM, Eddy SR, Elnitski L, Emes RD, Eswara P, Eyras E, Felsenfeld A, Fewell GA, Flicek P, Foley K, Frankel WN, Fulton LA, Fulton RS, Furey TS, Gage D, Gibbs RA, Glusman G, Gnerre S, Goldman N, Goodstadt L, Grafham D, Graves TA, Green ED, Gregory S, Guigo R, Guyer M, Hardison RC, Haussler D, Hayashizaki Y, Hillier LW, Hinrichs A, Hlavina W, Holzer T, Hsu F, Hua A, Hubbard T, Hunt A, Jackson I, Jaffe DB, Johnson LS, Jones M, Jones TA, Joy A, Kamal M, Karlsson EK, Karolchik D, Kasprzyk A, Kawai J, Keibler E, Kells C, Kent WJ, Kirby A, Kolbe DL, Korf I, Kucherlapati RS, Kulbokas EJ, Kulp D, Landers T, Leger JP, Leonard S, Letunic I, Levine R, Li J, Li M, Lloyd C, Lucas S, Ma B, Maglott DR, Mardis ER, Matthews L, Mauceli E, Mayer JH, McCarthy M, McCombie WR, McLaren S, McLay K, McPherson JD, Meldrim J, Meredith B, Mesirov JP, Miller W, Miner TL, Mongin E, Montgomery KT, Morgan M, Mott R, Mullikin JC, Muzny DM, Nash WE, Nelson JO, Nhan MN, Nicol R, Ning Z, Nusbaum C, OConnor MJ, Okazaki Y, Oliver K, OvertonLarty E, Pachter L, Parra G, Pepin KH, Peterson J, Pevzner P, Plumb

23. 24. 25. 26.

27. 28. 29. 30. 31.

32.

33.

34. 35. 36.

37.

38.

39.

40. 41. 42.

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice R, Pohl CS, Poliakov A, Ponce TC, Ponting CP, Potter S, Quail M, Reymond A, Roe BA, Roskin KM, Rubin EM, Rust AG, Santos R, Sapojnikov V, Schultz B, Schultz J, Schwartz MS, Schwartz S, Scott C, Seaman S, Searle S, Sharpe T, Sheridan A, Shownkeen R, Sims S, Singer JB, Slater G, Smit A, Smith DR, Spencer B, Stabenau A, Stange-Thomann N, Sugnet C, Suyama M, Tesler G, Thompson J, Torrents D, Trevaskis E, Tromp J, Ucla C, Ureta-Vidal A, Vinson JP, Von Niederhausern AC, Wade CM, Wall M, Weber RJ, Weiss RB, Wendl MC, West AP, Wetterstrand K, Wheeler R, Whelan S, Wierzbowski J, Willey D, Williams S, Wilson RK, Winter E, Worley KC, Wyman D, Yang S, Yang SP, Zdobnov EM, Zody MC, Lander ES 2002 Initial sequencing and comparative analysis of the mouse genome. Nature 420:520 562 Bogue M 2003 Mouse Phenome Project: understanding human biology through mouse genetics and genomics. J Appl Physiol 95:13351337 Grubb SC, Churchill GA, Bogue MA 2004 A collaborative database of inbred mouse strain characteristics. Bioinformatics 20:28572859 Paigen K 2003 One hundred years of mouse genetics: an intellectual history. I. The classical period (19021980). Genetics 163:17 Guenet JL, Bonhomme F 2003 Wild mice: an ever-increasing contribution to a popular mammalian model. Trends Genet 19:24 31 Beck JA, Lloyd S, Hafezparast M, Lennon-Pierce M, Eppig JT, Festing MF, Fisher EM 2000 Genealogies of mouse inbred strains. Nat Genet 24:2325 Lindblad-Toh K, Winchester E, Daly MJ, Wang DG, Hirschhorn JN, Laviolette JP, Ardlie K, Reich DE, Robinson E, Sklar P, Shah N, Thomas D, Fan JB, Gingeras T, Warrington J, Patil N, Hudson TJ, Lander ES 2000 Large-scale discovery and genotyping of singlenucleotide polymorphisms in the mouse. Nat Genet 24:381386 Pletcher MT, McClurg P, Batalov S, Su AI, Barnes SW, Lagler E, Korstanje R, Wang X, Nusskern D, Bogue MA, Mural RJ, Paigen B, Wiltshire T 2004 Use of a dense single nucleotide polymorphism map for in silico mapping in the mouse. PLoS Biol 2:e393 Cervino AC, Li G, Edwards S, Zhu J, Laurie C, Tokiwa G, Lum PY, Wang S, Castellini LW, Lusis AJ, Carlson S, Sachs AB, Schadt EE 2005 Integrating QTL and high-density SNP analyses in mice to identify Insig2 as a susceptibility gene for plasma cholesterol levels. Genomics 86:505517 Zhang J, Hunter KW, Gandolph M, Rowe WL, Finney RP, Kelley JM, Edmonson M, Buetow KH 2005 A high-resolution multistrain haplotype analysis of laboratory mouse genome reveals three distinctive genetic variation patterns. Genome Res 15:241249 Biddinger SB, Kahn CR 2006 From mice to men: insights into the insulin resistance syndromes. Annu Rev Physiol 68:123158 Nandi A, Kitamura Y, Kahn CR, Accili D 2004 Mouse models of insulin resistance. Physiol Rev 84:623 647 Plum L, Wunderlich FT, Baudler S, Krone W, Bruning JC 2005 Transgenic and knockout mice in diabetes research: novel insights into pathophysiology, limitations, and perspectives. Physiology (Bethesda) 20:152161 Terauchi Y, Kadowaki T 2002 Insights into molecular pathogenesis of type 2 diabetes from knockout mouse models. Endocr J 49:247263 Darvasi A, Soller M 1997 A simple method to calculate resolving power and confidence interval of QTL map location. Behav Genet 27:125132 Flint J, Valdar W, Shifman S, Mott R 2005 Strategies for mapping and cloning quantitative trait genes in rodents. Nat Rev Genet 6:271286 Darvasi A 1998 Experimental strategies for the genetic dissection of complex traits in animal models. Nat Genet 18:19 24 Darvasi A, Soller M 1995 Advanced intercross lines, an experimental population for fine genetic mapping. Genetics 141:1199 1207 Ehrich TH, Hrbek T, Kenney-Hunt JP, Pletscher LS, Wang B, Semenkovich CF, Cheverud JM 2005 Fine-mapping gene-by-diet interactions on chromosome 13 in a LG/J x SM/J murine model of obesity. Diabetes 54:18631872 Wang X, Le Roy I, Nicodeme E, Li R, Wagner R, Petros C, Churchill GA, Harris S, Darvasi A, Kirilovsky J, Roubertoux PL, Paigen B 2003 Using advanced intercross lines for high-resolution mapping of HDL cholesterol quantitative trait loci. Genome Res 13:1654 1664 Harper JM, Galecki AT, Burke DT, Pinkosky SL, Miller RA 2003 Quantitative trait loci for insulin-like growth factor I, leptin, thy-

Endocrine Reviews, February 2007, 28(1):48 83 75 roxine, and corticosterone in genetically heterogeneous mice. Physiol Genomics 15:44 51 Peirce JL, Lu L, Gu J, Silver LM, Williams RW 2004 A new set of BXD recombinant inbred lines from advanced intercross populations in mice. BMC Genet 5:7 Williams RW, Bennett B, Lu L, Gu J, DeFries JC, Carosone-Link PJ, Rikke BA, Belknap JK, Johnson TE 2004 Genetic structure of the LXS panel of recombinant inbred mouse strains: a powerful resource for complex trait analysis. Mamm Genome 15:637 647 Williams RW, Gu J, Qi S, Lu L 2001 The genetic structure of recombinant inbred mice: high-resolution consensus maps for complex trait analysis. Genome Biol 2:46.1 46.18 Churchill GA, Airey DC, Allayee H, Angel JM, Attie AD, Beatty J, Beavis WD, Belknap JK, Bennett B, Berrettini W, Bleich A, Bogue M, Broman KW, Buck KJ, Buckler E, Burmeister M, Chesler EJ, Cheverud JM, Clapcote S, Cook MN, Cox RD, Crabbe JC, Crusio WE, Darvasi A, Deschepper CF, Doerge RW, Farber CR, Forejt J, Gaile D, Garlow SJ, Geiger H, Gershenfeld H, Gordon T, Gu J, Gu W, de Haan G, Hayes NL, Heller C, Himmelbauer H, Hitzemann R, Hunter K, Hsu HC, Iraqi FA, Ivandic B, Jacob HJ, Jansen RC, Jepsen KJ, Johnson DK, Johnson TE, Kempermann G, Kendziorski C, Kotb M, Kooy RF, Llamas B, Lammert F, Lassalle JM, Lowenstein PR, Lu L, Lusis A, Manly KF, Marcucio R, Matthews D, Medrano JF, Miller DR, Mittleman G, Mock BA, Mogil JS, Montagutelli X, Morahan G, Morris DG, Mott R, Nadeau JH, Nagase H, Nowakowski RS, OHara BF, Osadchuk AV, Page GP, Paigen B, Paigen K, Palmer AA, Pan HJ, Peltonen-Palotie L, Peirce J, Pomp D, Pravenec M, Prows DR, Qi Z, Reeves RH, Roder J, Rosen GD, Schadt EE, Schalkwyk LC, Seltzer Z, Shimomura K, Shou S, Sillanpaa MJ, Siracusa LD, Snoeck HW, Spearow JL, Svenson K, Tarantino LM, Threadgill D, Toth LA, Valdar W, de Villena FP, Warden C, Whatley S, Williams RW, Wiltshire T, Yi N, Zhang D, Zhang M, Zou F 2004 The Collaborative Cross, a community resource for the genetic analysis of complex traits. Nat Genet 36:11331137 Zou F, Gelfond JA, Airey DC, Lu L, Manly KF, Williams RW, Threadgill DW 2005 Quantitative trait locus analysis using recombinant inbred intercrosses: theoretical and empirical considerations. Genetics 170:1299 1311 Fortin A, Diez E, Rochefort D, Laroche L, Malo D, Rouleau GA, Gros P, Skamene E 2001 Recombinant congenic strains derived from A/J and C57BL/6J: a tool for genetic dissection of complex traits. Genomics 74:2135 Singer JB, Hill AE, Burrage LC, Olszens KR, Song J, Justice M, OBrien WE, Conti DV, Witte JS, Lander ES, Nadeau JH 2004 Genetic dissection of complex traits with chromosome substitution strains of mice. Science 304:445 448 Cowley Jr AW, Liang M, Roman RJ, Greene AS, Jacob HJ 2004 Consomic rat model systems for physiological genomics. Acta Physiol Scand 181:585592 Davis RC, Schadt EE, Smith DJ, Hsieh EW, Cervino AC, van Nas A, Rosales M, Doss S, Meng H, Allayee H, Lusis AJ 2005 A genome-wide set of congenic mouse strains derived from DBA/2J on a C57BL/6J background. Genomics 86:259 270 Iakoubova OA, Olsson CL, Dains KM, Ross DA, Andalibi A, Lau K, Choi J, Kalcheva I, Cunanan M, Louie J, Nimon V, Machrus M, Bentley LG, Beauheim C, Silvey S, Cavalcoli J, Lusis AJ, West DB 2001 Genome-tagged mice (GTM): two sets of genome-wide congenic strains. Genomics 74:89 104 Singer GA, Lloyd AT, Huminiecki LB, Wolfe KH 2005 Clusters of co-expressed genes in mammalian genomes are conserved by natural selection. Mol Biol Evol 22:767775 Coleman DL, Hummel KP 1973 The influence of genetic background on the expression of the obese (Ob) gene in the mouse. Diabetologia 9:287293 Mott R, Talbot CJ, Turri MG, Collins AC, Flint J 2000 A method for fine mapping quantitative trait loci in outbred animal stocks. Proc Natl Acad Sci USA 97:12649 12654 Mott R, Flint J 2002 Simultaneous detection and fine mapping of quantitative trait loci in mice using heterogeneous stocks. Genetics 160:1609 1618 Yalcin B, Willis-Owen SA, Fullerton J, Meesaq A, Deacon RM, Rawlins JN, Copley RR, Morris AP, Flint J, Mott R 2004 Genetic

70. 71.

72. 73.

50. 51. 52. 53. 54. 55.

56.

57.

74.

58.

75.

59. 60. 61.

76.

77. 78.

62. 63. 64. 65. 66. 67.

79.

80. 81. 82. 83. 84.

68.

69.

76

Endocrine Reviews, February 2007, 28(1):48 83 dissection of a behavioral quantitative trait locus shows that Rgs2 modulates anxiety in mice. Nat Genet 36:11971202 Moore KJ, Nagle DL 2000 Complex trait analysis in the mouse: the strengths, the limitations and the promise yet to come. Annu Rev Genet 34:653 686 Grupe A, Germer S, Usuka J, Aud D, Belknap JK, Klein RF, Ahluwalia MK, Higuchi R, Peltz G 2001 In silico mapping of complex disease-related traits in mice. Science 292:19151918 Wang X, Korstanje R, Higgins D, Paigen B 2004 Haplotype analysis in multiple crosses to identify a QTL gene. Genome Res 14: 17671772 Abe K, Noguchi H, Tagawa K, Yuzuriha M, Toyoda A, Kojima T, Ezawa K, Saitou N, Hattori M, Sakaki Y, Moriwaki K, Shiroishi T 2004 Contribution of Asian mouse subspecies Mus musculus molossinus to genomic constitution of strain C57BL/6J, as defined by BAC-end sequence-SNP analysis. Genome Res 14:2439 2447 Rebuffe-Scrive M, Surwit R, Feinglos M, Kuhn C, Rodin J 1993 Regional fat distribution and metabolism in a new mouse model (C57BL/6J) of non-insulin-dependent diabetes mellitus. Metabolism 42:14051409 Surwit RS, Feinglos MN, Rodin J, Sutherland A, Petro AE, Opara EC, Kuhn CM, Rebuffe-Scrive M 1995 Differential effects of fat and sucrose on the development of obesity and diabetes in C57BL/6J and A/J mice. Metabolism 44:645 651 Surwit RS, Kuhn CM, Cochrane C, McCubbin JA, Feinglos MN 1988 Diet-induced type II diabetes in C57BL/6J mice. Diabetes 37:11631167 West DB, Boozer CN, Moody DL, Atkinson RL 1992 Dietary obesity in nine inbred mouse strains. Am J Physiol 262:R1025R1032 Kaku K, Fiedorek Jr FT, Province M, Permutt MA 1988 Genetic analysis of glucose tolerance in inbred mouse strains. Evidence for polygenic control. Diabetes 37:707713 Goren HJ, Kulkarni RN, Kahn CR 2004 Glucose homeostasis and tissue transcript content of insulin signaling intermediates in four inbred strains of mice: C57BL/6, C57BLKS/6, DBA/2, and 129X1. Endocrinology 145:33073323 Ahren B, Pacini G 2002 Insufficient islet compensation to insulin resistance vs. reduced glucose effectiveness in glucose-intolerant mice. Am J Physiol Endocrinol Metab 283:E738 E744 Rossmeisl M, Rim JS, Koza RA, Kozak LP 2003 Variation in type 2 diabetes-related traits in mouse strains susceptible to diet-induced obesity. Diabetes 52:1958 1966 Kooptiwut S, Zraika S, Thorburn AW, Dunlop ME, Darwiche R, Kay TW, Proietto J, Andrikopoulos S 2002 Comparison of insulin secretory function in two mouse models with different susceptibility to -cell failure. Endocrinology 143:20852092 Toye AA, Lippiat JD, Proks P, Shimomura K, Bentley L, Hugill A, Mijat V, Goldsworthy M, Moir L, Haynes A, Quarterman J, Freeman HC, Ashcroft FM, Cox RD 2005 A genetic and physiological study of impaired glucose homeostasis control in C57BL/6J mice. Diabetologia 48:675 686 Kayo T, Fujita H, Nozaki J, Xiaofei E, Koizumi A 2000 Identification of two chromosomal loci determining glucose intolerance in a C57BL/6 mouse strain. Comp Med 50:296 302 Lee SK, Opara EC, Surwit RS, Feinglos MN, Akwari OE 1995 Defective glucose-stimulated insulin release from perifused islets of C57BL/6J mice. Pancreas 11:206 211 Wencel HE, Smothers C, Opara EC, Kuhn CM, Feinglos MN, Surwit RS 1995 Impaired second phase insulin response of diabetes-prone C57BL/6J mouse islets. Physiol Behav 57:12151220 Hull RL, Kodama K, Utzschneider KM, Carr DB, Prigeon RL, Kahn SE 2005 Dietary-fat-induced obesity in mice results in -cell hyperplasia but not increased insulin release: evidence for specificity of impaired -cell adaptation. Diabetologia 48:1350 1358 Bock T, Pakkenberg B, Buschard K 2005 Genetic background determines the size and structure of the endocrine pancreas. Diabetes 54:133137 Cardinal JW, Allan DJ, Cameron DP 1998 Differential metabolite accumulation may be the cause of strain differences in sensitivity to streptozotocin-induced -cell death in inbred mice. Endocrinology 139:28852891 Kaku K, McGill J, Province M, Permutt MA 1989 A single major gene controls most of the difference in susceptibility to streptozo-

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice tocin-induced diabetes between C57BL/6J and C3H/HeJ mice. Diabetologia 32:716 723 Kim SH, Ma X, Klupa T, Powers C, Pezzolesi M, Warram JH, Rich SS, Krolewski AS, Doria A 2003 Genetic modifiers of the age at diagnosis of diabetes (MODY3) in carriers of hepatocyte nuclear factor-1 mutations map to chromosomes 5p15, 9q22, and 14q24. Diabetes 52:21822186 Freeman H, Shimomura K, Horner E, Cox RD, Ashcroft FM 2006 Nicotinamide nucleotide transhydrogenase: a key role in insulin secretion. Cell Metab 3:35 45 Freeman HC, Hugill A, Dear NT, Ashcroft FM, Cox RD 2006 Deletion of nicotinamide nucleotide transhydrogenase: a new quantitive trait locus accounting for glucose intolerance in C57BL/6J mice. Diabetes 55:21532156 Hoek JB, Rydstrom J 1988 Physiological roles of nicotinamide nucleotide transhydrogenase. Biochem J 254:110 Kulkarni RN, Almind K, Goren HJ, Winnay JN, Ueki K, Okada T, Kahn CR 2003 Impact of genetic background on development of hyperinsulinemia and diabetes in insulin receptor/insulin receptor substrate-1 double heterozygous mice. Diabetes 52:1528 1534 Coleman DL 1992 The influence of genetic background on the expression of mutations at the diabetes (db) locus in the mouse. VI: Hepatic malic enzyme activity is associated with diabetes severity. Metabolism 41:1134 1136 Bock T, Pakkenberg B, Buschard K 2003 Increased islet volume but unchanged islet number in ob/ob mice. Diabetes 52:1716 1722 Qi Z, Fujita H, Jin J, Davis LS, Wang Y, Fogo AB, Breyer MD 2005 Characterization of susceptibility of inbred mouse strains to diabetic nephropathy. Diabetes 54:2628 2637 Naggert JK, Mu JL, Frankel W, Bailey DW, Paigen B 1995 Genomic analysis of the C57BL/Ks mouse strain. Mamm Genome 6:131133 Mao HZ, Roussos ET, Peterfy M 2006 Genetic analysis of the diabetes-prone C57BLKS/J mouse strain reveals genetic contribution from multiple strains. Biochim Biophys Acta 1762:440 446 Davis RC, Schadt EE, Cervino AC, Peterfy M, Lusis AJ 2005 Ultrafine mapping of SNPs from mouse strains C57BL/6J, DBA/2J, and C57BLKS/J for loci contributing to diabetes and atherosclerosis susceptibility. Diabetes 54:11911199 Rossini AA, Appel MC, Williams RM, Like AA 1977 Genetic influence of the streptozotocin-induced insulitis and hyperglycemia. Diabetes 26:916 920 Swenne I, Andersson A 1984 Effect of genetic background on the capacity for islet cell replication in mice. Diabetologia 27:464 467 Leiter EH 1987 Analysis of differential survival of syngeneic islets transplanted into hyperglycemic C57BL/6J versus C57BL/KsJ mice. Transplantation 44:401 406 Coleman DL, Kuzava JE 1991 Genetic regulation of malic enzyme activity in the mouse. J Biol Chem 266:2199722002 Mu JL, Naggert JK, Svenson KL, Collin GB, Kim JH, McFarland C, Nishina PM, Levine DM, Williams KJ, Paigen B 1999 Quantitative trait loci analysis for the differences in susceptibility to atherosclerosis and diabetes between inbred mouse strains C57BL/6J and C57BLKS/J. J Lipid Res 40:1328 1335 Yaguchi H, Togawa K, Moritani M, Itakura M 2005 Identification of candidate genes in the type 2 diabetes modifier locus using expression QTL. Genomics 85:591599 Collin GB, Maddatu TP, Sen S, Naggert JK 2005 Genetic modifiers interact with Cpefat to affect body weight, adiposity, and hyperglycemia. Physiol Genomics 22:182190 Leiter EH, Kintner J, Flurkey K, Beamer WG, Naggert JK 1999 Physiologic and endocrinologic characterization of male sex-biased diabetes in C57BLKS/J mice congenic for the fat mutation at the carboxypeptidease E locus. Endocrine 10:57 66 Surwit RS, Seldin MF, Kuhn CM, Secor C, Feinglos MN 1994 Diet-induced obesity and diabetes in C57BL/6J and C57BL/KsJ mice. Mouse Genome 92:523525 Togawa K, Moritani M, Yaguchi H, Itakura M 2006 Multidimensional genome scans identify the combinations of genetic loci linked to diabetes-related phenotypes in mice. Hum Mol Genet 15:113128 Chua Jr S, Liu SM, Li Q, Yang L, Thassanapaff VT, Fisher P 2002 Differential -cell responses to hyperglycaemia and insulin resis-

85. 86. 87. 88.

106.

107. 108.

109. 110.

89.

90.

111.

91. 92. 93. 94.

112. 113. 114. 115. 116.

95. 96. 97.

117. 118. 119. 120. 121.

98.

99. 100. 101. 102.

122. 123. 124.

103. 104.

125. 126.

105.

127.

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice tance in two novel congenic strains of diabetes (FVB- Lepr (db)) and obese (DBA- Lep (ob)) mice. Diabetologia 45:976 990 Simpson EM, Linder CC, Sargent EE, Davisson MT, Mobraaten LE, Sharp JJ 1997 Genetic variation among 129 substrains and its importance for targeted mutagenesis in mice. Nat Genet 16:19 27 Festing MF, Simpson EM, Davisson MT, Mobraaten LE 1999 Revised nomenclature for strain 129 mice. Mamm Genome 10:836 Almind K, Kahn CR 2004 Genetic determinants of energy expenditure and insulin resistance in diet-induced obesity in mice. Diabetes 53:3274 3285 Almind K, Kulkarni RN, Lannon SM, Kahn CR 2003 Identification of interactive loci linked to insulin and leptin in mice with genetic insulin resistance. Diabetes 52:15351543 Kaku K, Province M, Permutt MA 1989 Genetic analysis of obesityinduced diabetes associated with a limited capacity to synthesize insulin in C57BL/KS mice: evidence for polygenic control. Diabetologia 32:636 643 Leiter EH, Coleman DL, Eisenstein AB, Strack I 1980 A new mutation (db3J) at the diabetes locus in strain 129/J mice. I. Physiological and histological characterization. Diabetologia 19:58 65 Kido Y, Philippe N, Schaffer AA, Accili D 2000 Genetic modifiers of the insulin resistance phenotype in mice. Diabetes 49:589 596 Withers DJ, Gutierrez JS, Towery H, Burks DJ, Ren JM, Previs S, Zhang Y, Bernal D, Pons S, Shulman GI, Bonner-Weir S, White MF 1998 Disruption of IRS-2 causes type 2 diabetes in mice. Nature 391:900 904 Kushner JA, Ye J, Schubert M, Burks DJ, Dow MA, Flint CL, Dutta S, Wright CV, Montminy MR, White MF 2002 Pdx1 restores -cell function in Irs2 knockout mice. J Clin Invest 109:11931201 Suzuki R, Tobe K, Terauchi Y, Komeda K, Kubota N, Eto K, Yamauchi T, Azuma K, Kaneto H, Taguchi T, Koga T, German MS, Watada H, Kawamori R, Wright CV, Kajimoto Y, Kimura S, Nagai R, Kadowaki T 2003 Pdx1 expression in Irs2-deficient mouse -cells is regulated in a strain-dependent manner. J Biol Chem 278:43691 43698 Korzh V, Grunwald D 2001 Nadine Dobrovolskaia-Zavadskaia and the dawn of developmental genetics. Bioessays 23:365371 Wahlsten D, Metten P, Crabbe JC 2003 Survey of 21 inbred mouse strains in two laboratories reveals that BTBR T/ tf/tf has severely reduced hippocampal commissure and absent corpus callosum. Brain Res 971:4754 Ranheim T, Dumke C, Schueler KL, Cartee GD, Attie AD 1997 Interaction between BTBR and C57BL/6J genomes produces an insulin resistance syndrome in (BTBR C57BL/6J) F1 mice. Arterioscler Thromb Vasc Biol 17:3286 3293 Clee SM, Nadler ST, Attie AD 2005 Genetic and genomic studies of the BTBR ob/ob mouse model of type 2 diabetes. Am J Ther 12:491 498 Nadler ST, Stoehr JP, Rabaglia ME, Schueler KL, Birnbaum MJ, Attie AD 2001 Normal Akt/PKB with reduced PI3K activation in insulin-resistant mice. Am J Physiol Endocrinol Metab 281:E1249 E1254 Rabaglia ME, Gray-Keller MP, Frey BL, Shortreed MR, Smith LM, Attie AD 2005 -Ketoisocaproate-induced hypersecretion of insulin by islets from diabetes-susceptible mice. Am J Physiol Endocrinol Metab 289:E218 E224 Stoehr JP, Nadler ST, Schueler KL, Rabaglia ME, Yandell BS, Metz SA, Attie AD 2000 Genetic obesity unmasks nonlinear interactions between murine type 2 diabetes susceptibility loci. Diabetes 49:1946 1954 Lan H, Chen M, Flowers JB, Yandell BS, Stapleton DS, Mata CM, Mui ET, Flowers MT, Schueler KL, Manly KF, Williams RW, Kendziorski C, Attie AD 2006 Combined expression trait correlations and expression quantitative trait locus mapping. PLoS Genet 2:e6 Lan H, Rabaglia ME, Stoehr JP, Nadler ST, Schueler KL, Zou F, Yandell BS, Attie AD 2003 Gene expression profiles of nondiabetic and diabetic obese mice suggest a role of hepatic lipogenic capacity in diabetes susceptibility. Diabetes 52:688 700 Lan H, Stoehr JP, Nadler ST, Schueler KL, Yandell BS, Attie AD 2003 Dimension reduction for mapping mRNA abundance as quantitative traits. Genetics 164:16071614 Clee SM, Yandell BS, Schueler KM, Rabaglia ME, Richards OC,

Endocrine Reviews, February 2007, 28(1):48 83 77 Raines SM, Kabara EA, Klass DM, Mui ET, Stapleton DS, GrayKeller MP, Young MB, Stoehr JP, Lan H, Boronenkov I, Raess PW, Flowers MT, Attie AD 2006 Positional cloning of Sorcs1, a type 2 diabetes quantitative trait locus. Nat Genet 38:688 693 Hermey G, Sjogaard SS, Petersen CM, Nykjaer A, Gliemann J 2006 Tumour necrosis factor -converting enzyme mediates ectodomain shedding of Vps10p-domain receptor family members. Biochem J 395:285293 Abramsson A, Lindblom P, Betsholtz C 2003 Endothelial and nonendothelial sources of PDGF-B regulate pericyte recruitment and influence vascular pattern formation in tumors. J Clin Invest 112:11421151 Song S, Ewald AJ, Stallcup W, Werb Z, Bergers G 2005 PDGFR perivascular progenitor cells in tumours regulate pericyte differentiation and vascular survival. Nat Cell Biol 7:870 879 Surwit RS, Seldin MF, Kuhn CM, Cochrane C, Feinglos MN 1991 Control of expression of insulin resistance and hyperglycemia by different genetic factors in diabetic C57BL/6J mice. Diabetes 40:82 87 Kobayashi M, Ohno T, Tsuji A, Nishimura M, Horio F 2003 Combinations of nondiabetic parental genomes elicit impaired glucose tolerance in mouse SMXA recombinant inbred strains. Diabetes 52:180 186 Kobayashi M, Io F, Kawai T, Kumazawa M, Ikegami H, Nishimura M, Ohno T, Horio F 2006 Major quantitative trait locus on chromosome 2 for glucose tolerance in diabetic SMXA-5 mouse established from non-diabetic SM/J and A/J strains. Diabetologia 49:486 495 Takeshita S, Moritani M, Kunika K, Inoue H, Itakura M 2006 Diabetic modifier QTLs identified in F2 intercrosses between Akita and A/J mice. Mamm Genome 17:927940 Qiu J, Ogus S, Mounzih K, Ewart-Toland A, Chehab FF 2001 Leptin-deficient mice backcrossed to the BALB/cJ genetic background have reduced adiposity, enhanced fertility, normal body temperature, and severe diabetes. Endocrinology 142:34213425 Moritani M, Togawa K, Yaguchi H, Fujita Y, Yamaguchi Y, Inoue H, Kamatani N, Itakura M 2006 Identification of diabetes susceptibility loci in db mice by combined quantitative trait loci analysis and haplotype mapping. Genomics 88:719 730 Eberhart GP, West DB, Boozer CN, Atkinson RL 1994 Insulin sensitivity of adipocytes from inbred mouse strains resistant or sensitive to diet-induced obesity. Am J Physiol 266:R1423R1428 Mehrabian M, Wen PZ, Fisler J, Davis RC, Lusis AJ 1998 Genetic loci controlling body fat, lipoprotein metabolism, and insulin levels in a multifactorial mouse model. J Clin Invest 101:24852496 Estrada-Smith D, Castellani LW, Wong H, Wen PZ, Chui A, Lusis AJ, Davis RC 2004 Dissection of multigenic obesity traits in congenic mouse strains. Mamm Genome 15:14 22 Anunciado RV, Imamura T, Ohno T, Horio F, Namikawa T 2000 Developing a new model for non-insulin dependent diabetes mellitus (NIDDM) by using the Philippine wild mouse, Mus musculus castaneus. Exp Anim 49:1 8 Horio F, Teradaira S, Imamura T, Anunciado RV, Kobayashi M, Namikawa T, Niki I 2005 The HND mouse, a nonobese model of type 2 diabetes mellitus with impaired insulin secretion. Eur J Endocrinol 153:971979 Makino S, Kunimoto K, Muraoka Y, Mizushima Y, Katagiri K, Tochino Y 1980 Breeding of a non-obese, diabetic strain of mice. Jikken Dobutsu 29:113 Lyons PA, Armitage N, Lord CJ, Phillips MS, Todd JA, Peterson LB, Wicker LS 2001 Mapping by genetic interaction: high-resolution congenic mapping of the type 1 diabetes loci Idd10 and Idd18 in the NOD mouse. Diabetes 50:26332637 Lyons PA, Armitage N, Argentina F, Denny P, Hill NJ, Lord CJ, Wilusz MB, Peterson LB, Wicker LS, Todd JA 2000 Congenic mapping of the type 1 diabetes locus, Idd3, to a 780-kb region of mouse chromosome 3: identification of a candidate segment of ancestral DNA by haplotype mapping. Genome Res 10:446 453 Brodnicki TC, Quirk F, Morahan G 2003 A susceptibility allele from a non-diabetes-prone mouse strain accelerates diabetes in NOD congenic mice. Diabetes 52:218 222 Gonzalez C, Cuvellier S, Hue-Beauvais C, Levi-Strauss M 2003 Genetic control of non obese diabetic mice susceptibility to highdose streptozotocin-induced diabetes. Diabetologia 46:12911295

128. 129. 130. 131. 132.

149.

150.

151. 152. 153.

133. 134. 135.

154.

136. 137.

155. 156.

157.

138. 139.

158. 159. 160. 161.

140.

141. 142.

162.

143.

163. 164.

144.

145.

165.

146.

166. 167.

147. 148.

78

Endocrine Reviews, February 2007, 28(1):48 83

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice ground genotype on muscle and liver insulin sensitivity of lipoatrophic mice. Role of triglyceride clearance. J Biol Chem 278:39923999 Nakamura M, Yamada K 1967 Studies on a diabetic (KK) strain of the mouse. Diabetologia 3:212221 Igel M, Taylor BA, Phillips SJ, Becker W, Herberg L, Joost HG 1998 Hyperleptinemia and leptin receptor variant Asp600Asn in the obese, hyperinsulinemic KK mouse strain. J Mol Endocrinol 21:337345 Taylor BA, Tarantino LM, Phillips SJ 1999 Gender-influenced obesity QTLs identified in a cross involving the KK type II diabetesprone mouse strain. Mamm Genome 10:963968 Suto J, Matsuura S, Imamura K, Yamanaka H, Sekikawa K 1998 Genetic analysis of non-insulin-dependent diabetes mellitus in KK and KK-Ay mice. Eur J Endocrinol 139:654 661 Shike T, Hirose S, Kobayashi M, Funabiki K, Shirai T, Tomino Y 2001 Susceptibility and negative epistatic loci contributing to type 2 diabetes and related phenotypes in a KK/Ta mouse model. Diabetes 50:19431948 Okazaki M, Saito Y, Udaka Y, Maruyama M, Murakami H, Ota S, Kikuchi T, Oguchi K 2002 Diabetic nephropathy in KK and KK-Ay mice. Exp Anim 51:191196 Dulin WE, Wyse BM 1970 Diabetes in the KK mouse. Diabetologia 6:317323 Suto J, Sekikawa K 2002 A quantitative trait locus that accounts for glucose intolerance maps to chromosome 8 in hereditary obese KK-A(y) mice. Int J Obes Relat Metab Disord 26:15171519 Shike T, Gohda T, Tanimoto M, Kobayashi M, Makita Y, Funabiki K, Horikoshi S, Hirose S, Shirai T, Tomino Y 2005 Chromosomal mapping of a quantitative trait locus for the development of albuminuria in diabetic KK/Ta mice. Nephrol Dial Transplant 20:879 885 Kim JH, Sen S, Avery CS, Simpson E, Chandler P, Nishina PM, Churchill GA, Naggert JK 2001 Genetic analysis of a new mouse model for non-insulin-dependent diabetes. Genomics 74:273286 Sung YY, Lee YS, Jung WH, Kim HY, Cheon HG, Yang SD, Rhee SD 2005 Glucose intolerance in young TallyHo mice is induced by leptin-mediated inhibition of insulin secretion. Biochem Biophys Res Commun 338:1779 1787 Kim JH, Stewart TP, Zhang W, Kim HY, Nishina PM, Naggert JK 2005 Type 2 diabetes mouse model TallyHo carries an obesity gene on chromosome 6 that exaggerates dietary obesity. Physiol Genomics 22:171181 Ueda H, Ikegami H, Yamato E, Fu J, Fukuda M, Shen G, Kawaguchi Y, Takekawa K, Fujioka Y, Fujisawa T, Nakagawa Y, Hamada Y, Shibata M, Ogihara T 1995 The NSY mouse: a new animal model of spontaneous NIDDM with moderate obesity. Diabetologia 38:503508 Ueda H, Ikegami H, Kawaguchi Y, Fujisawa T, Yamato E, Shibata M, Ogihara T 1999 Genetic analysis of late-onset type 2 diabetes in a mouse model of human complex trait. Diabetes 48:1168 1174 Leiter EH, Lee CH 2005 Mouse models and the genetics of diabetes: is there evidence for genetic overlap between type 1 and type 2 diabetes? Diabetes 54(Suppl 2):S151S158 Babaya N, Ikegami H, Fujisawa T, Nojima K, Itoi-Babaya M, Inoue K, Ohno T, Shibata M, Ogihara T 2005 Susceptibility to streptozotocin-induced diabetes is mapped to mouse chromosome 11. Biochem Biophys Res Commun 328:158 164 Mathews CE, Leiter EH 1999 Resistance of ALR/Lt islets to free radical-mediated diabetogenic stress is inherited as a dominant trait. Diabetes 48:2189 2196 Mathews CE, Suarez-Pinzon WL, Baust JJ, Strynadka K, Leiter EH, Rabinovitch A 2005 Mechanisms underlying resistance of pancreatic islets from ALR/Lt mice to cytokine-induced destruction. J Immunol 175:1248 1256 Mathews CE, Bagley R, Leiter EH 2004 ALS/Lt: a new type 2 diabetes mouse model associated with low free radical scavenging potential. Diabetes 53(Suppl 1):S125S129 Sekiguchi F, Ishibashi K, Kawamoto Y, Ino T 1991 [Diabetic peculiarity of the ALS-Ay and ALR-Ay strains]. Jikken Dobutsu 40:323329 Mathews CE, Leiter EH, Spirina O, Bykhovskaya Y, Gusdon AM, Ringquist S, Fischel-Ghodsian N 2005 mt-Nd2 Allele of the ALR/Lt mouse confers resistance against both chemically induced and autoimmune diabetes. Diabetologia 48:261267 Uchigata Y, Okada T, Gong JS, Yamada Y, Iwamoto Y, Tanaka M

168. Yang Y, Santamaria P 2006 Lessons on autoimmune diabetes from animal models. Clin Sci (Lond) 110:627 639 169. Todd JA, Wicker LS 2001 Genetic protection from the inflammatory disease type 1 diabetes in humans and animal models. Immunity 15:387395 170. Lang J, Bellgrau D 2004 Animal models of type 1 diabetes: genetics and immunological function. Adv Exp Med Biol 552:91116 171. Leiter EH 2005 Nonobese diabetic mice and the genetics of diabetes susceptibility. Curr Diab Rep 5:141148 172. Wong FS, Janeway Jr CA 1999 Insulin-dependent diabetes mellitus and its animal models. Curr Opin Immunol 11:643 647 173. Leiter EH, Reifsnyder PC, Flurkey K, Partke HJ, Junger E, Herberg L 1998 NIDDM genes in mice: deleterious synergism by both parental genomes contributes to diabetogenic thresholds. Diabetes 47:12871295 174. Reifsnyder PC, Churchill G, Leiter EH 2000 Maternal environment and genotype interact to establish diabesity in mice. Genome Res 10:1568 1578 175. Reifsnyder PC, Leiter EH 2002 Deconstructing and reconstructing obesity-induced diabetes (diabesity) in mice. Diabetes 51:825 832 176. Leiter EH, Reifsnyder PC 2004 Differential levels of diabetogenic stress in two new mouse models of obesity and type 2 diabetes. Diabetes 53(Suppl 1):S4 S11 177. Haskell BD, Flurkey K, Duffy TM, Sargent EE, Leiter EH 2002 The diabetes-prone NZO/HlLt strain. I. Immunophenotypic comparison to the related NZB/BlNJ and NZW/LacJ strains. Lab Invest 82:833 842 178. Igel M, Becker W, Herberg L, Joost HG 1997 Hyperleptinemia, leptin resistance, and polymorphic leptin receptor in the New Zealand obese mouse. Endocrinology 138:4234 4239 179. Plum L, Kluge R, Giesen K, Altmuller J, Ortlepp JR, Joost HG 2000 Type 2 diabetes-like hyperglycemia in a backcross model of NZO and SJL mice: characterization of a susceptibility locus on chromosome 4 and its relation with obesity. Diabetes 49:1590 1596 180. Kluge R, Giesen K, Bahrenberg G, Plum L, Ortlepp JR, Joost HG 2000 Quantitative trait loci for obesity and insulin resistance (Nob1, Nob2) and their interaction with the leptin receptor allele (LeprA720T/T1044I) in New Zealand obese mice. Diabetologia 43:15651572 181. Banks WA, DiPalma CR, Farrell CL 1999 Impaired transport of leptin across the blood-brain barrier in obesity. Peptides 20:13411345 182. Ortlepp JR, Kluge R, Giesen K, Plum L, Radke P, Hanrath P, Joost HG 2000 A metabolic syndrome of hypertension, hyperinsulinaemia and hypercholesterolaemia in the New Zealand obese mouse. Eur J Clin Invest 30:195202 183. Veroni MC, Proietto J, Larkins RG 1991 Evolution of insulin resistance in New Zealand obese mice. Diabetes 40:1480 1487 184. Junger E, Herberg L, Jeruschke K, Leiter EH 2002 The diabetesprone NZO/Hl strain. II. Pancreatic immunopathology. Lab Invest 82:843 853 185. Harrison LC, Itin A 1979 A possible mechanism for insulin resistance and hyperglycaemia in NZO mice. Nature 279:334 336 186. Plum L, Giesen K, Kluge R, Junger E, Linnartz K, Schurmann A, Becker W, Joost HG 2002 Characterisation of the mouse diabetes susceptibilty locus Nidd/SJL: islet cell destruction, interaction with the obesity QTL Nob1, and effect of dietary fat. Diabetologia 45: 823 830 187. Luo N, Liu SM, Liu H, Li Q, Xu Q, Sun X, Davis B, Li J, Chua Jr S 2006 Allelic variation on chromosome 5 controls -cell mass expansion during hyperglycemia in leptin receptor-deficient diabetes mice. Endocrinology 147:22872295 188. Haluzik M, Colombo C, Gavrilova O, Chua S, Wolf N, Chen M, Stannard B, Dietz KR, Le Roith D, Reitman ML 2004 Genetic background (C57BL/6J versus FVB/N) strongly influences the severity of diabetes and insulin resistance in ob/ob mice. Endocrinology 145:3258 3264 189. Moitra J, Mason MM, Olive M, Krylov D, Gavrilova O, MarcusSamuels B, Feigenbaum L, Lee E, Aoyama T, Eckhaus M, Reitman ML, Vinson C 1998 Life without white fat: a transgenic mouse. Genes Dev 12:3168 3181 190. Colombo C, Haluzik M, Cutson JJ, Dietz KR, Marcus-Samuels B, Vinson C, Gavrilova O, Reitman ML 2003 Opposite effects of back-

191. 192.

193. 194. 195.

196. 197. 198. 199.

200. 201.

202.

203.

204. 205. 206.

207. 208.

209. 210. 211.

212.

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice 2002 A mitochondrial genotype associated with the development of autoimmune-related type 1 diabetes. Diabetes Care 25:2106 Allan MF, Eisen EJ, Pomp D 2004 The M16 mouse: an outbred animal model of early onset polygenic obesity and diabesity. Obes Res 12:13971407 Allan MF, Eisen EJ, Pomp D 2005 Genomic mapping of direct and correlated responses to long-term selection for rapid growth rate in mice. Genetics 170:18631877 Hrbek T, de Brito RA, Wang B, Pletscher LS, Cheverud JM 2006 Genetic characterization of a new set of recombinant inbred lines (LGXSM) formed from the intercross of SM/J and LG/J inbred mouse strains. Mamm Genome 17:417 429 Ehrich TH, Kenney JP, Vaughn TT, Pletscher LS, Cheverud JM 2003 Diet, obesity, and hyperglycemia in LG/J and SM/J mice. Obes Res 11:1400 1410 Cheverud JM, Ehrich TH, Hrbek T, Kenney JP, Pletscher LS, Semenkovich CF 2004 Quantitative trait loci for obesity- and diabetesrelated traits and their dietary responses to high-fat feeding in LGXSM recombinant inbred mouse strains. Diabetes 53:3328 3336 Cheverud JM, Ehrich TH, Kenney JP, Pletscher LS, Semenkovich CF 2004 Genetic evidence for discordance between obesity- and diabetes-related traits in the LGXSM recombinant inbred mouse strains. Diabetes 53:2700 2708 Anunciado RV, Ohno T, Mori M, Ishikawa A, Tanaka S, Horio F, Nishimura M, Namikawa T 2000 Distribution of body weight, blood insulin and lipid levels in the SMXA recombinant inbred strains and the QTL analysis. Exp Anim 49:217224 Kobayashi M, Io F, Kawai T, Nishimura M, Ohno T, Horio F 2004 SMXA-5 mouse as a diabetic model susceptible to feeding a high-fat diet. Biosci Biotechnol Biochem 68:226 230 Kobayashi M, Ohno T, Kawada T, Ikegami H, Nishimura M, Horio F 2006 Serum adiponectin concentration: its correlation with diabetesrelated traits and quantitative trait loci analysis in mouse SMXA recombinant inbred strains. Biosci Biotechnol Biochem 70:677 683 Anunciado RV, Horio F, Ohno T, Tanaka S, Nishimura M, Namikawa T 2000 Characterization of hyperinsulinemic recombinant inbred (RI) strains (SMXA-5 and SMXA-9) derived from normoinsulinemic SM/J and A/J mice. Exp Anim 49:8390 Suzuki W, Iizuka S, Tabuchi M, Funo S, Yanagisawa T, Kimura M, Sato T, Endo T, Kawamura H 1999 A new mouse model of spontaneous diabetes derived from ddY strain. Exp Anim 48:181189 Hirayama I, Yi Z, Izumi S, Arai I, Suzuki W, Nagamachi Y, Kuwano H, Takeuchi T, Izumi T 1999 Genetic analysis of obese diabetes in the TSOD mouse. Diabetes 48:11831191 Iizuka S, Suzuki W, Tabuchi M, Nagata M, Imamura S, Kobayashi Y, Kanitani M, Yanagisawa T, Kase Y, Takeda S, Aburada M, Takahashi KW 2005 Diabetic complications in a new animal model (TSOD mouse) of spontaneous NIDDM with obesity. Exp Anim 54:71 83 Mizutani S, Gomi H, Hirayama I, Izumi T 2006 Chromosome 2 locus Nidd5 has a potent effect on adiposity in the TSOD mouse. Mamm Genome 17:375384 Wang J, Takeuchi T, Tanaka S, Kubo SK, Kayo T, Lu D, Takata K, Koizumi A, Izumi T 1999 A mutation in the insulin 2 gene induces diabetes with severe pancreatic -cell dysfunction in the Mody mouse. J Clin Invest 103:2737 Schroder M, Kaufman RJ 2005 The mammalian unfolded protein response. Annu Rev Biochem 74:739 789 Harding HP, Zeng H, Zhang Y, Jungries R, Chung P, Plesken H, Sabatini DD, Ron D 2001 Diabetes mellitus and exocrine pancreatic dysfunction in perk/ mice reveals a role for translational control in secretory cell survival. Mol Cell 7:11531163 Scheuner D, Song B, McEwen E, Liu C, Laybutt R, Gillespie P, Saunders T, Bonner-Weir S, Kaufman RJ 2001 Translational control is required for the unfolded protein response and in vivo glucose homeostasis. Mol Cell 7:11651176 Marciniak SJ, Yun CY, Oyadomari S, Novoa I, Zhang Y, Jungreis R, Nagata K, Harding HP, Ron D 2004 CHOP induces death by promoting protein synthesis and oxidation in the stressed endoplasmic reticulum. Genes Dev 18:3066 3077 Oyadomari S, Koizumi A, Takeda K, Gotoh T, Akira S, Araki E, Mori M 2002 Targeted disruption of the Chop gene delays endoplasmic reticulum stress-mediated diabetes. J Clin Invest 109:525532

Endocrine Reviews, February 2007, 28(1):48 83 79 233. Rankinen T, Zuberi A, Chagnon YC, Weisnagel SJ, Argyropoulos G, Walts B, Perusse L, Bouchard C 2006 The human obesity gene map: the 2005 update. Obesity 14:529 644 234. Goto Y, Kakizaki M, Masaki N 1976 Production of spontaneous diabetic rats by repetition of selective breeding. Tohoku J Exp Med 119:8590 235. Portha B 2005 Programmed disorders of -cell development and function as one cause for type 2 diabetes? The GK rat paradigm. Diabetes Metab Res Rev 21:495504 236. Galli J, Li LS, Glaser A, Ostenson CG, Jiao H, Fakhrai-Rad H, Jacob HJ, Lander ES, Luthman H 1996 Genetic analysis of non-insulin dependent diabetes mellitus in the GK rat. Nat Genet 12:3137 237. Gauguier D, Froguel P, Parent V, Bernard C, Bihoreau MT, Portha B, James MR, Penicaud L, Lathrop M, Ktorza A 1996 Chromosomal mapping of genetic loci associated with non-insulin dependent diabetes in the GK rat. Nat Genet 12:38 43 238. Collins SC, Wallis RH, Wilder SP, Wallace KJ, Argoud K, Kaisaki PJ, Bihoreau MT, Gauguier D 2006 Mapping diabetes QTL in an intercross derived from a congenic strain of the Brown Norway and Goto-Kakizaki rats. Mamm Genome 17:538 547 239. Fakhrai-Rad H, Nikoshkov A, Kamel A, Fernstrom M, Zierath JR, Norgren S, Luthman H, Galli J 2000 Insulin-degrading enzyme identified as a candidate diabetes susceptibility gene in GK rats. Hum Mol Genet 9:2149 2158 240. Farris W, Mansourian S, Leissring MA, Eckman EA, Bertram L, Eckman CB, Tanzi RE, Selkoe DJ 2004 Partial loss-of-function mutations in insulin-degrading enzyme that induce diabetes also impair degradation of amyloid -protein. Am J Pathol 164:14251434 241. Farris W, Mansourian S, Chang Y, Lindsley L, Eckman EA, Frosch MP, Eckman CB, Tanzi RE, Selkoe DJ, Guenette S 2003 Insulindegrading enzyme regulates the levels of insulin, amyloid -protein, and the -amyloid precursor protein intracellular domain in vivo. Proc Natl Acad Sci USA 100:4162 4167 242. Kawano K, Hirashima T, Mori S, Saitoh Y, Kurosumi M, Natori T 1992 Spontaneous long-term hyperglycemic rat with diabetic complications. Otsuka Long-Evans Tokushima Fatty (OLETF) strain. Diabetes 41:14221428 243. Wei S, Wei K, Moralejo DH, Ogino T, Koike G, Jacob HJ, Sugiura K, Sasaki Y, Yamada T, Matsumoto K 1999 Mapping and characterization of quantitative trait loci for non-insulin-dependent diabetes mellitus with an improved genetic map in the Otsuka Long-Evans Tokushima fatty rat. Mamm Genome 10:249 258 244. Sugiura K, Miyake T, Taniguchi Y, Yamada T, Moralejo DH, Wei S, Wei K, Sasaki Y, Matsumoto K 1999 Identification of novel non-insulin-dependent diabetes mellitus susceptibility loci in the Otsuka Long-Evans Tokushima fatty rat by MQM-mapping method. Mamm Genome 10:1126 1131 245. Kose H, Moralejo DH, Ogino T, Mizuno A, Yamada T, Matsumoto K 2002 Examination of OLETF-derived non-insulin-dependent diabetes mellitus QTL by construction of a series of congenic rats. Mamm Genome 13:558 562 246. Kanemoto N, Hishigaki H, Miyakita A, Oga K, Okuno S, Tsuji A, Takagi T, Takahashi E, Nakamura Y, Watanabe TK 1998 Genetic dissection of OLETF, a rat model for non-insulin-dependent diabetes mellitus. Mamm Genome 9:419 425 247. Watanabe TK, Okuno S, Ono T, Yamasaki Y, Oga K, MizoguchiMiyakita A, Miyao H, Suzuki M, Momota H, Goto Y, Shinomiya H, Hishigaki H, Hayashi I, Asai T, Wakitani S, Takagi T, Nakamura Y, Tanigami A 2001 Single-allele correction of the Dmo1 locus in congenic animals substantially attenuates obesity, dyslipidaemia and diabetes phenotypes of the OLETF rat. Clin Exp Pharmacol Physiol 28:28 42 248. Watanabe TK, Okuno S, Yamasaki Y, Ono T, Oga K, MizoguchiMiyakita A, Miyao H, Suzuki M, Momota H, Goto Y, Shinomiya H, Hishigaki H, Hayashi I, Asai T, Wakitani S, Takagi T, Nakamura Y, Tanigami A 2004 A 1.7 cM interval is responsible for Dmo1 obesity phenotypes in OLETF rats. Clin Exp Pharmacol Physiol 31:110 112 249. Watanabe TK, Suzuki M, Yamasaki Y, Okuno S, Hishigaki H, Ono T, Oga K, Mizoguchi-Miyakita A, Tsuji A, Kanemoto N, Wakitani S, Takagi T, Nakamura Y, Tanigami A 2005 Mutated G-protein-coupled receptor GPR10 is responsible for the hyperpha-

213. 214. 215.

216. 217.

218.

219.

220. 221.

222.

223. 224. 225.

226. 227.

228. 229.

230.

231.

232.

80

Endocrine Reviews, February 2007, 28(1):48 83 gia/dyslipidaemia/obesity locus of Dmo1 in the OLETF rat. Clin Exp Pharmacol Physiol 32:355366 Ghosh S, Watanabe RM, Valle TT, Hauser ER, Magnuson VL, Langefeld CD, Ally DS, Mohlke KL, Silander K, Kohtamaki K, Chines P, Balow Jr J, Birznieks G, Chang J, Eldridge W, Erdos MR, Karanjawala ZE, Knapp JI, Kudelko K, Martin C, MoralesMena A, Musick A, Musick T, Pfahl C, Porter R, Rayman JB 2000 The Finland-United States investigation of non-insulin-dependent diabetes mellitus genetics (FUSION) study. I. An autosomal genome scan for genes that predispose to type 2 diabetes. Am J Hum Genet 67:1174 1185 An P, Hong Y, Weisnagel SJ, Rice T, Rankinen T, Leon AS, Skinner JS, Wilmore JH, Chagnon YC, Bergman RN, Bouchard C, Rao DC 2003 Genomic scan of glucose and insulin metabolism phenotypes: the HERITAGE Family Study. Metabolism 52:246 253 An P, Teran-Garcia M, Rice T, Rankinen T, Weisnagel SJ, Bergman RN, Boston RC, Mandel S, Stefanovski D, Leon AS, Skinner JS, Rao DC, Bouchard C 2005 Genome-wide linkage scans for prediabetes phenotypes in response to 20 weeks of endurance exercise training in non-diabetic whites and blacks: the HERITAGE Family Study. Diabetologia 48:11421149 Aulchenko YS, Vaessen N, Heutink P, Pullen J, Snijders PJ, Hofman A, Sandkuijl LA, Houwing-Duistermaat JJ, Edwards M, Bennett S, Oostra BA, van Duijn CM 2003 A genome-wide search for genes involved in type 2 diabetes in a recently genetically isolated population from the Netherlands. Diabetes 52:30013004 Frayling TM, Wiltshire S, Hitman GA, Walker M, Levy JC, Sampson M, Groves CJ, Menzel S, McCarthy MI, Hattersley AT 2003 Young-onset type 2 diabetes families are the major contributors to genetic loci in the Diabetes UK Warren 2 genome scan and identify putative novel loci on chromosomes 8q21, 21q22, and 22q11. Diabetes 52:18571863 Freedman BI, Rich SS, Sale MM, Heiss G, Djousse L, Pankow JS, Province MA, Rao DC, Lewis CE, Chen YD, Beck SR 2005 Genomewide scans for heritability of fasting serum insulin and glucose concentrations in hypertensive families. Diabetologia 48:661 668 Hanson RL, Imperatore G, Narayan KM, Roumain J, Fagot-Campagna A, Pettitt DJ, Bennett PH, Knowler WC 2001 Family and genetic studies of indices of insulin sensitivity and insulin secretion in Pima Indians. Diabetes Metab Res Rev 17:296 303 Lakka TA, Rankinen T, Weisnagel SJ, Chagnon YC, Rice T, Leon AS, Skinner JS, Wilmore JH, Rao DC, Bouchard C 2003 A quantitative trait locus on 7q31 for the changes in plasma insulin in response to exercise training: the HERITAGE Family Study. Diabetes 52:15831587 Pratley RE, Thompson DB, Prochazka M, Baier L, Mott D, Ravussin E, Sakul H, Ehm MG, Burns DK, Foroud T, Garvey WT, Hanson RL, Knowler WC, Bennett PH, Bogardus C 1998 An autosomal genomic scan for loci linked to prediabetic phenotypes in Pima Indians. J Clin Invest 101:17571764 Reynisdottir I, Thorleifsson G, Benediktsson R, Sigurdsson G, Emilsson V, Einarsdottir AS, Hjorleifsdottir EE, Orlygsdottir GT, Bjornsdottir GT, Saemundsdottir J, Halldorsson S, Hrafnkelsdottir S, Sigurjonsdottir SB, Steinsdottir S, Martin M, Kochan JP, Rhees BK, Grant SF, Frigge ML, Kong A, Gudnason V, Stefansson K, Gulcher JR 2003 Localization of a susceptibility gene for type 2 diabetes to chromosome 5q34 q35.2. Am J Hum Genet 73:323335 Sale MM, Freedman BI, Langefeld CD, Williams AH, Hicks PJ, Colicigno CJ, Beck SR, Brown WM, Rich SS, Bowden DW 2004 A genome-wide scan for type 2 diabetes in African-American families reveals evidence for a locus on chromosome 6q. Diabetes 53:830 837 Silander K, Scott LJ, Valle TT, Mohlke KL, Stringham HM, Wiles KR, Duren WL, Doheny KF, Pugh EW, Chines P, Narisu N, White PP, Fingerlin TE, Jackson AU, Li C, Ghosh S, Magnuson VL, Colby K, Erdos MR, Hill JE, Hollstein P, Humphreys KM, Kasad RA, Lambert J, Lazaridis KN, Lin G, Morales-Mena A, Patzkowski K, Pfahl C, Porter R, Rha D, Segal L, Suh YD, Tovar J, Unni A, Welch C, Douglas JA, Epstein MP, Hauser ER, Hagopian W, Buchanan TA, Watanabe RM, Bergman RN, Tuomilehto J, Collins FS, Boehnke M 2004 A large set of Finnish affected sibling

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice pair families with type 2 diabetes suggests susceptibility loci on chromosomes 6, 11, and 14. Diabetes 53:821 829 Vionnet N, Hani El H, Dupont S, Gallina S, Francke S, Dotte S, De Matos F, Durand E, Lepretre F, Lecoeur C, Gallina P, Zekiri L, Dina C, Froguel P 2000 Genomewide search for type 2 diabetessusceptibility genes in French whites: evidence for a novel susceptibility locus for early-onset diabetes on chromosome 3q27-qter and independent replication of a type 2-diabetes locus on chromosome 1q21 q24. Am J Hum Genet 67:1470 1480 Wiltshire S, Hattersley AT, Hitman GA, Walker M, Levy JC, Sampson M, ORahilly S, Frayling TM, Bell JI, Lathrop GM, Bennett A, Dhillon R, Fletcher C, Groves CJ, Jones E, Prestwich P, Simecek N, Rao PV, Wishart M, Bottazzo GF, Foxon R, Howell S, Smedley D, Cardon LR, Menzel S, McCarthy MI 2001 A genomewide scan for loci predisposing to type 2 diabetes in a U.K. population (the Diabetes UK Warren 2 Repository): analysis of 573 pedigrees provides independent replication of a susceptibility locus on chromosome 1q. Am J Hum Genet 69:553569 Huelsken J, Birchmeier W 2001 New aspects of Wnt signaling pathways in higher vertebrates. Curr Opin Genet Dev 11:547553 Mulholland DJ, Dedhar S, Coetzee GA, Nelson CC 2005 Interaction of nuclear receptors with the Wnt/-catenin/Tcf signaling axis: Wnt you like to know? Endocr Rev 26:898 915 Gu HF, Efendic S, Nordman S, Ostenson CG, Brismar K, Brookes AJ, Prince JA 2004 Quantitative trait loci near the insulin-degrading enzyme (IDE) gene contribute to variation in plasma insulin levels. Diabetes 53:21372142 Groves CJ, Wiltshire S, Smedley D, Owen KR, Frayling TM, Walker M, Hitman GA, Levy JC, ORahilly S, Menzel S, Hattersley AT, McCarthy MI 2003 Association and haplotype analysis of the insulin-degrading enzyme (IDE) gene, a strong positional and biological candidate for type 2 diabetes susceptibility. Diabetes 52:1300 1305 Karamohamed S, Demissie S, Volcjak J, Liu C, Heard-Costa N, Liu J, Shoemaker CM, Panhuysen CI, Meigs JB, Wilson P, Atwood LD, Cupples LA, Herbert A 2003 Polymorphisms in the insulin-degrading enzyme gene are associated with type 2 diabetes in men from the NHLBI Framingham Heart Study. Diabetes 52: 15621567 Florez JC, Wiltshire S, Agapakis CM, Burtt NP, de Bakker PI, Almgren P, Bengtsson Bostrom K, Tuomi T, Gaudet D, Daly MJ, Hirschhorn JN, McCarthy MI, Altshuler D, Groop L 2006 Highdensity haplotype structure and association testing of the insulindegrading enzyme (IDE) gene with type 2 diabetes in 4,206 people. Diabetes 55:128 135 Florez JC, Jablonski KA, Bayley N, Pollin TI, de Bakker PI, Shuldiner AR, Knowler WC, Nathan DM, Altshuler D 2006 TCF7L2 polymorphisms and progression to diabetes in the Diabetes Prevention Program. N Engl J Med 355:241250 Damcott CM, Pollin TI, Reinhart LJ, Ott SH, Shen H, Silver KD, Mitchell BD, Shuldiner AR 2006 Polymorphisms in the transcription factor 7-like 2 (TCF7L2) gene are associated with type 2 diabetes in the Amish: replication and evidence for a role in both insulin secretion and insulin resistance. Diabetes 55:2654 2659 Groves CJ, Zeggini E, Minton J, Frayling TM, Weedon MN, Rayner NW, Hitman GA, Walker M, Wiltshire S, Hattersley AT, McCarthy MI 2006 Association analysis of 6,736 U.K. subjects provides replication and confirms TCF7L2 as a type 2 diabetes susceptibility gene with a substantial effect on individual risk. Diabetes 55:2640 2644 Scott LJ, Bonnycastle LL, Willer CJ, Sprau AG, Jackson AU, Narisu N, Duren WL, Chines PS, Stringham HM, Erdos MR, Valle TT, Tuomilehto J, Bergman RN, Mohlke KL, Collins FS, Boehnke M 2006 Association of transcription factor 7-like 2 (TCF7L2) variants with type 2 diabetes in a Finnish sample. Diabetes 55:2649 2653 Zhang C, Qi L, Hunter DJ, Meigs JB, Manson JE, van Dam RM, Hu FB 2006 Variant of transcription factor 7-like 2 (TCF7L2) gene and the risk of type 2 diabetes in large cohorts of U.S. women and men. Diabetes 55:26452648 Moralejo DH, Wei S, Wei K, Weksler-Zangen S, Koike G, Jacob HJ, Hirashima T, Kawano K, Sugiura K, Sasaki Y, Ogino T, Yamada T, Matsumoto K 1998 Identification of quantitative trait loci for non-insulin-dependent diabetes mellitus that interact with

250.

262.

263.

251.

252.

264. 265. 266.

253.

254.

267.

255.

268.

256.

269.

257.

258.

270.

271.

259.

272.

260.

273.

261.

274.

275.

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice body weight in the Otsuka Long-Evans Tokushima Fatty rat. Proc Assoc Am Physicians 110:545558 Masuyama T, Fuse M, Yokoi N, Shinohara M, Tsujii H, Kanazawa M, Kanazawa Y, Komeda K, Taniguchi K 2003 Genetic analysis for diabetes in a new rat model of nonobese type 2 diabetes, spontaneously diabetic Torii rat. Biochem Biophys Res Commun 304:196 206 Shinohara M, Masuyama T, Shoda T, Takahashi T, Katsuda Y, Komeda K, Kuroki M, Kakehashi A, Kanazawa Y 2000 A new spontaneously diabetic non-obese Torii rat strain with severe ocular complications. Int J Exp Diabetes Res 1:89 100 Hanson RL, Ehm MG, Pettitt DJ, Prochazka M, Thompson DB, Timberlake D, Foroud T, Kobes S, Baier L, Burns DK, Almasy L, Blangero J, Garvey WT, Bennett PH, Knowler WC 1998 An autosomal genomic scan for loci linked to type II diabetes mellitus and body-mass index in Pima Indians. Am J Hum Genet 63:1130 1138 Elbein SC, Hoffman MD, Teng K, Leppert MF, Hasstedt SJ 1999 A genome-wide search for type 2 diabetes susceptibility genes in Utah Caucasians. Diabetes 48:11751182 Jun G, Song Y, Stein CM, Iyengar SK 2003 An autosome-wide search using longitudinal data for loci linked to type 2 diabetes progression. BMC Genet 4(Suppl 1):S8 Duggirala R, Blangero J, Almasy L, Arya R, Dyer TD, Williams KL, Leach RJ, OConnell P, Stern MP 2001 A major locus for fasting insulin concentrations and insulin resistance on chromosome 6q with strong pleiotropic effects on obesity-related phenotypes in nondiabetic Mexican Americans. Am J Hum Genet 68:1149 1164 Watanabe RM, Ghosh S, Langefeld CD, Valle TT, Hauser ER, Magnuson VL, Mohlke KL, Silander K, Ally DS, Chines P, Blaschak-Harvan J, Douglas JA, Duren WL, Epstein MP, Fingerlin TE, Kaleta HS, Lange EM, Li C, McEachin RC, Stringham HM, Trager E, White PP, Balow Jr J, Birznieks G, Chang J, Eldridge W 2000 The Finland-United States investigation of non-insulin-dependent diabetes mellitus genetics (FUSION) study. II. An autosomal genome scan for diabetes-related quantitative-trait loci. Am J Hum Genet 67:1186 1200 Demenais F, Kanninen T, Lindgren CM, Wiltshire S, Gaget S, Dandrieux C, Almgren P, Sjogren M, Hattersley A, Dina C, Tuomi T, McCarthy MI, Froguel P, Groop LC 2003 A meta-analysis of four European genome screens (GIFT Consortium) shows evidence for a novel region on chromosome 17p11.2-q22 linked to type 2 diabetes. Hum Mol Genet 12:18651873 Kissebah AH, Sonnenberg GE, Myklebust J, Goldstein M, Broman K, James RG, Marks JA, Krakower GR, Jacob HJ, Weber J, Martin L, Blangero J, Comuzzie AG 2000 Quantitative trait loci on chromosomes 3 and 17 influence phenotypes of the metabolic syndrome. Proc Natl Acad Sci USA 97:14478 14483 Iwasaki N, Cox NJ, Wang YQ, Schwarz PE, Bell GI, Honda M, Imura M, Ogata M, Saito M, Kamatani N, Iwamoto Y 2003 Mapping genes influencing type 2 diabetes risk and BMI in Japanese subjects. Diabetes 52:209 213 Lindgren CM, Mahtani MM, Widen E, McCarthy MI, Daly MJ, Kirby A, Reeve MP, Kruglyak L, Parker A, Meyer J, Almgren P, Lehto M, Kanninen T, Tuomi T, Groop LC, Lander ES 2002 Genomewide search for type 2 diabetes mellitus susceptibility loci in Finnish families: the Botnia study. Am J Hum Genet 70:509 516 Rotimi CN, Chen G, Adeyemo AA, Furbert-Harris P, ParishGause D, Zhou J, Berg K, Adegoke O, Amoah A, Owusu S, Acheampong J, Agyenim-Boateng K, Eghan Jr BA, Oli J, Okafor G, Ofoegbu E, Osotimehin B, Abbiyesuku F, Johnson T, Rufus T, Fasanmade O, Kittles R, Daniel H, Chen Y, Dunston G, Collins FS 2004 A genome-wide search for type 2 diabetes susceptibility genes in West Africans: the Africa America Diabetes Mellitus (AADM) Study. Diabetes 53:838 841 Avery CL, Freedman BI, Kraja AT, Borecki IB, Miller MB, Pankow JS, Arnett D, Lewis CE, Myers RH, Hunt SC, North KE 2006 Genotype-by-sex interaction in the aetiology of type 2 diabetes mellitus: support for sex-specific quantitative trait loci in Hypertension Genetic Epidemiology Network participants. Diabetologia 49:2329 2336 Lindsay RS, Kobes S, Knowler WC, Bennett PH, Hanson RL 2001 Genome-wide linkage analysis assessing parent-of-origin effects in the inheritance of type 2 diabetes and BMI in Pima Indians. Diabetes 50:2850 2857 Permutt MA, Wasson JC, Suarez BK, Lin J, Thomas J, Meyer J,

Endocrine Reviews, February 2007, 28(1):48 83 81 Lewitzky S, Rennich JS, Parker A, DuPrat L, Maruti S, Chayen S, Glaser B 2001 A genome scan for type 2 diabetes susceptibility loci in a genetically isolated population. Diabetes 50:681 685 Tang W, Miller MB, Rich SS, North KE, Pankow JS, Borecki IB, Myers RH, Hopkins PN, Leppert M, Arnett DK 2003 Linkage analysis of a composite factor for the multiple metabolic syndrome: the National Heart, Lung, and Blood Institute Family Heart Study. Diabetes 52:2840 2847 Aitman TJ, Glazier AM, Wallace CA, Cooper LD, Norsworthy PJ, Wahid FN, Al-Majali KM, Trembling PM, Mann CJ, Shoulders CC, Graf D, St Lezin E, Kurtz TW, Kren V, Pravenec M, Ibrahimi A, Abumrad NA, Stanton LW, Scott J 1999 Identification of Cd36 (Fat) as an insulin-resistance gene causing defective fatty acid and glucose metabolism in hypertensive rats. Nat Genet 21:76 83 Febbraio M, Hajjar DP, Silverstein RL 2001 CD36: a class B scavenger receptor involved in angiogenesis, atherosclerosis, inflammation, and lipid metabolism. J Clin Invest 108:785791 Pravenec M, Landa V, Zidek V, Musilova A, Kren V, Kazdova L, Aitman TJ, Glazier AM, Ibrahimi A, Abumrad NA, Qi N, Wang JM, St Lezin EM, Kurtz TW 2001 Transgenic rescue of defective Cd36 ameliorates insulin resistance in spontaneously hypertensive rats. Nat Genet 27:156 158 Watanabe T, Iizuka Y, Liang YQ, Inomata H, Gotoda T, Yanai K, Isobe M, Kato N 2005 Evaluation of insulin resistance linkage to rat chromosome 4 in SHR of a Japanese colony. Biochem Biophys Res Commun 329:879 887 Gotoda T, Iizuka Y, Kato N, Osuga J, Bihoreau MT, Murakami T, Yamori Y, Shimano H, Ishibashi S, Yamada N 1999 Absence of Cd36 mutation in the original spontaneously hypertensive rats with insulin resistance. Nat Genet 22:226 228 Febbraio M, Abumrad NA, Hajjar DP, Sharma K, Cheng W, Pearce SF, Silverstein RL 1999 A null mutation in murine CD36 reveals an important role in fatty acid and lipoprotein metabolism. J Biol Chem 274:1905519062 Hajri T, Han XX, Bonen A, Abumrad NA 2002 Defective fatty acid uptake modulates insulin responsiveness and metabolic responses to diet in CD36-null mice. J Clin Invest 109:13811389 Goudriaan JR, Dahlmans VE, Teusink B, Ouwens DM, Febbraio M, Maassen JA, Romijn JA, Havekes LM, Voshol PJ 2003 CD36 deficiency increases insulin sensitivity in muscle, but induces insulin resistance in the liver in mice. J Lipid Res 44:2270 2277 Ibrahimi A, Bonen A, Blinn WD, Hajri T, Li X, Zhong K, Cameron R, Abumrad NA 1999 Muscle-specific overexpression of FAT/ CD36 enhances fatty acid oxidation by contracting muscle, reduces plasma triglycerides and fatty acids, and increases plasma glucose and insulin. J Biol Chem 274:2676126766 An P, Freedman BI, Rich SS, Mandel SA, Arnett DK, Myers RH, Chen YD, Hunt SC, Rao DC 2006 Quantitative trait loci on chromosome 8q24 for pancreatic -cell function and 7q11 for insulin sensitivity in obese nondiabetic white and black families: evidence from genome-wide linkage scans in the NHLBI Hypertension Genetic Epidemiology Network (HyperGEN) study. Diabetes 55:551558 Lindgren CM, Widen E, Tuomi T, Li H, Almgren P, Kanninen T, Melander O, Weng J, Lehto M, Groop LC 2002 Contribution of known and unknown susceptibility genes to early-onset diabetes in scandinavia: evidence for heterogeneity. Diabetes 51:1609 1617 Furuhashi M, Ura N, Nakata T, Shimamoto K 2003 Insulin sensitivity and lipid metabolism in human CD36 deficiency. Diabetes Care 26:471 474 Furuhashi M, Ura N, Nakata T, Tanaka T, Shimamoto K 2004 Genotype in human CD36 deficiency and diabetes mellitus. Diabet Med 21:952953 Lepretre F, Linton KJ, Lacquemant C, Vatin V, Samson C, Dina C, Chikri M, Ali S, Scherer P, Seron K, Vasseur F, Aitman T, Froguel P 2004 Genetic study of the CD36 gene in a French diabetic population. Diabetes Metab 30:459 463 Lepretre F, Vasseur F, Vaxillaire M, Scherer PE, Ali S, Linton K, Aitman T, Froguel P 2004 A CD36 nonsense mutation associated with insulin resistance and familial type 2 diabetes. Hum Mutat 24:104 Miyaoka K, Kuwasako T, Hirano K, Nozaki S, Yamashita S, Matsuzawa Y 2001 CD36 deficiency associated with insulin resistance. Lancet 357:686 687 Corpeleijn E, van der Kallen CJ, Kruijshoop M, Magagnin MG,

276.

291.

277.

292.

278.

293. 294.

279. 280. 281.

295.

282.

296.

297.

298. 299.

283.

284.

300.

285.

301.

286.

302.

287.

303. 304. 305.

288.

306. 307. 308.

289.

290.

82

Endocrine Reviews, February 2007, 28(1):48 83 de Bruin TW, Feskens EJ, Saris WH, Blaak EE 2006 Direct association of a promoter polymorphism in the CD36/FAT fatty acid transporter gene with type 2 diabetes mellitus and insulin resistance. Diabet Med 23:907911 Hirashima T, Kawano K, Mori S, Natori T 1996 A diabetogenic gene, ODB2, identified on chromosome 14 of the OLETF rat and its synergistic action with ODB1. Biochem Biophys Res Commun 224:420 425 Moralejo DH, Ogino T, Zhu M, Toide K, Wei S, Wei K, Yamada T, Mizuno A, Matsumoto K, Shima K 1998 A major quantitative trait locus co-localizing with cholecystokinin type A receptor gene influences poor pancreatic proliferation in a spontaneously diabetogenic rat. Mamm Genome 9:794 798 Takiguchi S, Takata Y, Funakoshi A, Miyasaka K, Kataoka K, Fujimura Y, Goto T, Kono A 1997 Disrupted cholecystokinin type-A receptor (CCKAR) gene in OLETF rats. Gene 197:169 175 Takiguchi S, Takata Y, Takahashi N, Kataoka K, Hirashima T, Kawano K, Miyasaka K, Funakoshi A, Kono A 1998 A disrupted cholecystokinin A receptor gene induces diabetes in obese rats synergistically with ODB1 gene. Am J Physiol 274:E265E270 Dufresne M, Seva C, Fourmy D 2006 Cholecystokinin and gastrin receptors. Physiol Rev 86:805 847 Hirashima T, Kawano K, Mori S, Matsumoto K, Natori T 1995 A diabetogenic gene (ODB-1) assigned to the X-chromosome in OLETF rats. Diabetes Res Clin Pract 27:9196 Nakaya A, Hishigaki H, Morishita S 2000 Mining the quantitative trait loci associated with oral glucose tolerance in the OLETF rat. Pac Symp Biocomput 5:367379 Akhi M, Kose H, Matsumoto K 2005 Fine mapping of the hyperglycemic and obesity QTL by congenic strains suggests multiple loci on rat chromosome 14. J Med Invest 52:109 113 Bi S, Scott KA, Kopin AS, Moran TH 2004 Differential roles for cholecystokinin a receptors in energy balance in rats and mice. Endocrinology 145:38733880 Kopin AS, Mathes WF, McBride EW, Nguyen M, Al-Haider W, Schmitz F, Bonner-Weir S, Kanarek R, Beinborn M 1999 The cholecystokinin-A receptor mediates inhibition of food intake yet is not essential for the maintenance of body weight. J Clin Invest 103:383391 Funakoshi A, Miyasaka K, Matsumoto H, Yamamori S, Takiguchi S, Kataoka K, Takata Y, Matsusue K, Kono A, Shimokata H 2000 Gene structure of human cholecystokinin (CCK) type-A receptor: body fat content is related to CCK type-A receptor gene promoter polymorphism. FEBS Lett 466:264 266 Inoue H, Iannotti CA, Welling CM, Veile R, Donis-Keller H, Permutt MA 1997 Human cholecystokinin type A receptor gene: cytogenetic localization, physical mapping, and identification of two missense variants in patients with obesity and non-insulindependent diabetes mellitus (NIDDM). Genomics 42:331335 Arya R, Blangero J, Williams K, Almasy L, Dyer TD, Leach RJ, OConnell P, Stern MP, Duggirala R 2002 Factors of insulin resistance syndrome: related phenotypes are linked to genetic locations on chromosomes 6 and 7 in nondiabetic Mexican-Americans. Diabetes 51:841 847 Avery CL, Freedman BI, Heiss G, Kraja A, Rice T, Arnett D, Miller MB, Pankow JS, Lewis CE, Myers RH, Hunt SC, Almasy L, North KE 2004 Linkage analysis of diabetes status among hypertensive families: the Hypertension Genetic Epidemiology Network study. Diabetes 53:33073312 Cheng LS, Davis RC, Raffel LJ, Xiang AH, Wang N, Quinones M, Wen PZ, Toscano E, Diaz J, Pressman S, Henderson PC, Azen SP, Hsueh WA, Buchanan TA, Rotter JI 2001 Coincident linkage of fasting plasma insulin and blood pressure to chromosome 7q in hypertensive hispanic families. Circulation 104:12551260 Chiu YF, Chuang LM, Hsiao CF, Hung YJ, Lin MW, Chen YT, Grove J, Jorgenson E, Quertermous T, Risch N, Hsiung CA 2005 An autosomal genome-wide scan for loci linked to pre-diabetic phenotypes in nondiabetic Chinese subjects from the Stanford Asia-Pacific Program of Hypertension and Insulin Resistance Family Study. Diabetes 54:1200 1206 Das SK, Hasstedt SJ, Zhang Z, Elbein SC 2004 Linkage and association mapping of a chromosome 1q21 q24 type 2 diabetes susceptibility locus in northern European Caucasians. Diabetes 53:492 499 Du W, Sun H, Wang H, Qiang B, Shen Y, Yao Z, Gu J, Xiong M,

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice Huang W, Chen Z, Zuo J, Hua X, Gao W, Sun Q, Fang F 2001 Confirmation of susceptibility gene loci on chromosome 1 in northern China Han families with type 2 diabetes. Chin Med J (Engl) 114:876 878 Francke S, Manraj M, Lacquemant C, Lecoeur C, Lepretre F, Passa P, Hebe A, Corset L, Yan SL, Lahmidi S, Jankee S, Gunness TK, Ramjuttun US, Balgobin V, Dina C, Froguel P 2001 A genome-wide scan for coronary heart disease suggests in Indo-Mauritians a susceptibility locus on chromosome 16p13 and replicates linkage with the metabolic syndrome on 3q27. Hum Mol Genet 10:27512765 Hunt KJ, Lehman DM, Arya R, Fowler S, Leach RJ, Goring HH, Almasy L, Blangero J, Dyer TD, Duggirala R, Stern MP 2005 Genome-wide linkage analyses of type 2 diabetes in Mexican Americans: the San Antonio Family Diabetes/Gallbladder Study. Diabetes 54:26552662 Langefeld CD, Wagenknecht LE, Rotter JI, Williams AH, Hokanson JE, Saad MF, Bowden DW, Haffner S, Norris JM, Rich SS, Mitchell BD 2004 Linkage of the metabolic syndrome to 1q23 q31 in Hispanic families: the Insulin Resistance Atherosclerosis Study Family Study. Diabetes 53:1170 1174 Loos RJ, Katzmarzyk PT, Rao DC, Rice T, Leon AS, Skinner JS, Wilmore JH, Rankinen T, Bouchard C 2003 Genome-wide linkage scan for the metabolic syndrome in the HERITAGE Family Study. J Clin Endocrinol Metab 88:59355943 Meigs JB, Panhuysen CI, Myers RH, Wilson PW, Cupples LA 2002 A genome-wide scan for loci linked to plasma levels of glucose and HbA(1c) in a community-based sample of Caucasian pedigrees: the Framingham Offspring Study. Diabetes 51:833 840 Ng MC, So WY, Cox NJ, Lam VK, Cockram CS, Critchley JA, Bell GI, Chan JC 2004 Genome-wide scan for type 2 diabetes loci in Hong Kong Chinese and confirmation of a susceptibility locus on chromosome 1q21 q25. Diabetes 53:1609 1613 Ng MC, So WY, Lam VK, Cockram CS, Bell GI, Cox NJ, Chan JC 2004 Genome-wide scan for metabolic syndrome and related quantitative traits in Hong Kong Chinese and confirmation of a susceptibility locus on chromosome 1q21 q25. Diabetes 53:2676 2683 Parker A, Meyer J, Lewitzky S, Rennich JS, Chan G, Thomas JD, Orho-Melander M, Lehtovirta M, Forsblom C, Hyrkko A, Carlsson M, Lindgren C, Groop LC 2001 A gene conferring susceptibility to type 2 diabetes in conjunction with obesity is located on chromosome 18p11. Diabetes 50:675 680 Wiltshire S, Frayling TM, Groves CJ, Levy JC, Hitman GA, Sampson M, Walker M, Menzel S, Hattersley AT, Cardon LR, McCarthy MI 2004 Evidence from a large U.K. family collection that genes influencing age of onset of type 2 diabetes map to chromosome 12p and to the MODY3/NIDDM2 locus on 12q24. Diabetes 53:855 860 Xiang K, Wang Y, Zheng T, Jia W, Li J, Chen L, Shen K, Wu S, Lin X, Zhang G, Wang C, Wang S, Lu H, Fang Q, Shi Y, Zhang R, Xu J, Weng Q 2004 Genome-wide search for type 2 diabetes/ impaired glucose homeostasis susceptibility genes in the Chinese: significant linkage to chromosome 6q21 q23 and chromosome 1q21 q24. Diabetes 53:228 234 Zhao JY, Xiong MM, Huang W, Wang H, Zuo J, Wu GD, Chen Z, Qiang BQ, Zhang ML, Chen JL, Ding W, Yuan WT, Xu HY, Jin L, Li YX, Sun Q, Liu QY, Boerwinkle E, Fang FD 2005 An autosomal genomic scan for loci linked to type 2 diabetes in northern Han Chinese. J Mol Med 83:209 215 Seidelmann SB, De Luca C, Leibel RL, Breslow JL, Tall AR, Welch CL 2005 Quantitative trait locus mapping of genetic modifiers of metabolic syndrome and atherosclerosis in low-density lipoprotein receptor-deficient mice: identification of a locus for metabolic syndrome and increased atherosclerosis on chromosome 4. Arterioscler Thromb Vasc Biol 25:204 210 Pravenec M, Zidek V, Musilova A, Simakova M, Kostka V, Mlejnek P, Kren V, Krenova D, Bila V, Mikova B, Jachymova M, Horky K, Kazdova L, St Lezin E, Kurtz TW 2002 Genetic analysis of metabolic defects in the spontaneously hypertensive rat. Mamm Genome 13:253258 Bilusic M, Bataillard A, Tschannen MR, Gao L, Barreto NE, Vincent M, Wang T, Jacob HJ, Sassard J, Kwitek AE 2004 Mapping the genetic determinants of hypertension, metabolic diseases, and related phenotypes in the lyon hypertensive rat. Hypertension 44:695701 Busfield F, Duffy DL, Kesting JB, Walker SM, Lovelock PK, Good

309. 310.

327.

328.

311. 312.

329.

313. 314. 315. 316. 317. 318.

330.

331.

332.

333.

334.

319.

335.

320.

336.

321.

322.

337.

323.

338.

324.

339.

325.

340.

326.

341.

Clee and Attie Genetic Landscape of Type 2 Diabetes in Mice D, Tate H, Watego D, Marczak M, Hayman N, Shaw JT 2002 A genomewide search for type 2 diabetes-susceptibility genes in indigenous Australians. Am J Hum Genet 70:349 357 Hanis CL, Boerwinkle E, Chakraborty R, Ellsworth DL, Concannon P, Stirling B, Morrison VA, Wapelhorst B, Spielman RS, Gogolin-Ewens KJ, Shepard JM, Williams SR, Risch N, Hinds D, Iwasaki N, Ogata M, Omori Y, Petzold C, Rietzch H, Schroder HE, Schulze J, Cox NJ, Menzel S, Boriraj VV, Chen X, Lim LR, Lindner T, Mereu LE, Wang YQ, Xiang K, Yamagata K, Yang Y, Bell GI 1996 A genome-wide search for human non-insulin-dependent (type 2) diabetes genes reveals a major susceptibility locus on chromosome 2. Nat Genet 13:161166 Li WD, Dong C, Li D, Garrigan C, Price RA 2004 A quantitative trait locus influencing fasting plasma glucose in chromosome region 18q2223. Diabetes 53:24872491 Mori Y, Otabe S, Dina C, Yasuda K, Populaire C, Lecoeur C, Vatin V, Durand E, Hara K, Okada T, Tobe K, Boutin P, Kadowaki T, Froguel P 2002 Genome-wide search for type 2 diabetes in Japanese affected sib-pairs confirms susceptibility genes on 3q, 15q, and 20q and identifies two new candidate loci on 7p and 11p. Diabetes 51:12471255 van Tilburg JH, Sandkuijl LA, Strengman E, van Someren H, Rigters-Aris CA, Pearson PL, van Haeften TW, Wijmenga C 2003 A genome-wide scan in type 2 diabetes mellitus provides independent replication of a susceptibility locus on 18p11 and suggests the existence of novel loci on 2q12 and 19q13. J Clin Endocrinol Metab 88:22232230 Cox NJ, Hayes MG, Roe CA, Tsuchiya T, Bell GI 2004 Linkage of calpain 10 to type 2 diabetes: the biological rationale. Diabetes 53(Suppl 1):S19 S25 Horikawa Y, Oda N, Cox NJ, Li X, Orho-Melander M, Hara M, Hinokio Y, Lindner TH, Mashima H, Schwarz PE, del BosquePlata L, Horikawa Y, Oda Y, Yoshiuchi I, Colilla S, Polonsky KS, Wei S, Concannon P, Iwasaki N, Schulze J, Baier LJ, Bogardus C, Groop L, Boerwinkle E, Hanis CL, Bell GI 2000 Genetic variation in the gene encoding calpain-10 is associated with type 2 diabetes mellitus. Nat Genet 26:163175 Tsuchiya T, Schwarz PE, Bosque-Plata LD, Geoffrey Hayes M, Dina C, Froguel P, Wayne Towers G, Fischer S, TemelkovaKurktschiev T, Rietzsch H, Graessler J, Vcelak J, Palyzova D, Selisko T, Bendlova B, Schulze J, Julius U, Hanefeld M, Weedon MN, Evans JC, Frayling TM, Hattersley AT, Orho-Melander M, Groop L, Malecki MT, Hansen T, Pedersen O, Fingerlin TE, Boehnke M, Hanis CL, Cox NJ, Bell GI 2006 Association of the calpain-10 gene with type 2 diabetes in Europeans: results of pooled and meta-analyses. Mol Genet Metab 89:174 184 Malecki MT, Jhala US, Antonellis A, Fields L, Doria A, Orban T, Saad M, Warram JH, Montminy M, Krolewski AS 1999 Mutations in NEUROD1 are associated with the development of type 2 diabetes mellitus. Nat Genet 23:323328 Jellema A, Zeegers MP, Feskens EJ, Dagnelie PC, Mensink RP 2003 Gly972Arg variant in the insulin receptor substrate-1 gene and association with type 2 diabetes: a meta-analysis of 27 studies. Diabetologia 46:990 995 Muramatsu Y, Taniguchi Y, Kose H, Matsumoto K, Yamada T, Sasaki Y 2003 Capn10, a candidate gene responsible for type 2 diabetes mellitus in the OLETF rat. IUBMB Life 55:533537 Ehm MG, Karnoub MC, Sakul H, Gottschalk K, Holt DC, Weber JL, Vaske D, Briley D, Briley L, Kopf J, McMillen P, Nguyen Q, Reisman M, Lai EH, Joslyn G, Shepherd NS, Bell C, Wagner MJ, Burns DK 2000 Genomewide search for type 2 diabetes susceptibility genes in four American populations. Am J Hum Genet 66: 18711881 Hegele RA, Sun F, Harris SB, Anderson C, Hanley AJ, Zinman B 1999 Genome-wide scanning for type 2 diabetes susceptibility in Canadian Oji-Cree, using 190 microsatellite markers. J Hum Genet 44:10 14 North KE, Almasy L, Goring HH, Cole SA, Diego VP, Laston S, Cantu T, Williams JT, Howard BV, Lee ET, Best LG, Fabsitz RR,

Endocrine Reviews, February 2007, 28(1):48 83 83 MacCluer JW 2005 Linkage analysis of factors underlying insulin resistance: Strong Heart Family Study. Obes Res 13:18771884 Mitchell BD, Cole SA, Hsueh WC, Comuzzie AG, Blangero J, MacCluer JW, Hixson JE 2000 Linkage of serum insulin concentrations to chromosome 3p in Mexican Americans. Diabetes 49:513516 Bowden DW, Rudock M, Ziegler J, Lehtinen AB, Xu J, Wagenknecht LE, Herrington D, Rich SS, Freedman BI, Carr JJ, Langefeld CD 2006 Coincident linkage of type 2 diabetes, metabolic syndrome, and measures of cardiovascular disease in a genome scan of the diabetes heart study. Diabetes 55:19851994 Cai G, Cole SA, Freeland-Graves JH, MacCluer JW, Blangero J, Comuzzie AG 2004 Principal component for metabolic syndrome risk maps to chromosome 4p in Mexican Americans: the San Antonio Family Heart Study. Hum Biol 76:651 665 Chung WK, Zheng M, Chua M, Kershaw E, Power-Kehoe L, Tsuji M, Wu-Peng XS, Williams J, Chua Jr SC, Leibel RL 1997 Genetic modifiers of Leprfa associated with variability in insulin production and susceptibility to NIDDM. Genomics 41:332344 Cai G, Cole SA, Freeland-Graves JH, MacCluer JW, Blangero J, Comuzzie AG 2004 Genome-wide scans reveal quantitative trait loci on 8p and 13q related to insulin action and glucose metabolism: the San Antonio Family Heart Study. Diabetes 53:1369 1374 Pezzolesi MG, Nam M, Nagase T, Klupa T, Dunn JS, Mlynarski WM, Rich SS, Warram JH, Krolewski AS 2004 Examination of candidate chromosomal regions for type 2 diabetes reveals a susceptibility locus on human chromosome 8p23.1. Diabetes 53:486 491 Bektas A, Hughes JN, Warram JH, Krolewski AS, Doria A 2001 Type 2 diabetes locus on 12q15. Further mapping and mutation screening of two candidate genes. Diabetes 50:204 208 Bektas A, Suprenant ME, Wogan LT, Plengvidhya N, Rich SS, Warram JH, Krolewski AS, Doria A 1999 Evidence of a novel type 2 diabetes locus 50 cM centromeric to NIDDM2 on chromosome 12q. Diabetes 48:2246 2251 Shaw JT, Lovelock PK, Kesting JB, Cardinal J, Duffy D, Wainwright B, Cameron DP 1998 Novel susceptibility gene for lateonset NIDDM is localized to human chromosome 12q. Diabetes 47:17931796 Mahtani MM, Widen E, Lehto M, Thomas J, McCarthy M, Brayer J, Bryant B, Chan G, Daly M, Forsblom C, Kanninen T, Kirby A, Kruglyak L, Munnelly K, Parkkonen M, Reeve-Daly MP, Weaver A, Brettin T, Duyk G, Lander ES, Groop LC 1996 Mapping of a gene for type 2 diabetes associated with an insulin secretion defect by a genome scan in Finnish families. Nat Genet 14:90 94 Ogino T, Moralejo DH, Zhu M, Toide K, Wei S, Wei K, Yamada T, Mizuno A, Matsumoto K, Shima K 1999 Identification of possible quantitative trait loci responsible for hyperglycaemia after 70% pancreatectomy using a spontaneously diabetogenic rat. Genet Res 73:29 36 Ghosh S, Watanabe RM, Hauser ER, Valle T, Magnuson VL, Erdos MR, Langefeld CD, Balow Jr J, Ally DS, Kohtamaki K, Chines P, Birznieks G, Kaleta HS, Musick A, Te C, Tannenbaum J, Eldridge W, Shapiro S, Martin C, Witt A, So A, Chang J, Shurtleff B, Porter R, Kudelko K, Unni A, Segal L, Sharaf R, Blaschak-Harvan J, Eriksson J, Tenkula T, Vidgren G, Ehnholm C, Tuomilehto-Wolf E, Hagopian W, Buchanan TA, Tuomilehto J, Bergman RN, Collins FS, Boehnke M 1999 Type 2 diabetes: evidence for linkage on chromosome 20 in 716 Finnish affected sib pairs. Proc Natl Acad Sci USA 96:2198 2203 Kovacs P, van den Brandt J, Kloting I 2000 Genetic dissection of the syndrome X in the rat. Biochem Biophys Res Commun 269: 660 665 Mehrabian M, Allayee H, Stockton J, Lum PY, Drake TA, Castellani LW, Suh M, Armour C, Edwards S, Lamb J, Lusis AJ, Schadt EE 2005 Integrating genotypic and expression data in a segregating mouse population to identify 5-lipoxygenase as a susceptibility gene for obesity and bone traits. Nat Genet 37:1224 1233 Leiter EH 1988 Control of spontaneous glucose intolerance, hyperinsulinemia, and islet hyperplasia in nonobese C3H.SW male mice by Y-linked locus and adrenal gland. Metabolism 37:689 696

355. 356.

342.

357.

343. 344.

358.

359.

345.

360.

361. 362.

346. 347.

363.

348.

364.

365.

349.

366.

350.

351. 352.

367. 368.

353.

369.

354.

Endocrine Reviews is published by The Endocrine Society (http://www.endo-society.org), the foremost professional society serving the endocrine community.

Das könnte Ihnen auch gefallen