Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where David M. Kingsley is active.

Publication


Featured researches published by David M. Kingsley.


Nature | 2012

The genomic basis of adaptive evolution in threespine sticklebacks

Felicity C. Jones; Manfred Grabherr; Yingguang Frank Chan; Pamela Russell; Evan Mauceli; Jeremy A. Johnson; Ross Swofford; Mono Pirun; Michael C. Zody; Simon D. M. White; Ewan Birney; Stephen M. J. Searle; Jeremy Schmutz; Jane Grimwood; Mark Dickson; Richard M. Myers; Craig T. Miller; Brian R. Summers; Anne K. Knecht; Shannon D. Brady; Haili Zhang; Alex A. Pollen; Timothy R. Howes; Chris T. Amemiya; Eric S. Lander; Federica Di Palma; Kerstin Lindblad-Toh; David M. Kingsley

Marine stickleback fish have colonized and adapted to thousands of streams and lakes formed since the last ice age, providing an exceptional opportunity to characterize genomic mechanisms underlying repeated ecological adaptation in nature. Here we develop a high-quality reference genome assembly for threespine sticklebacks. By sequencing the genomes of twenty additional individuals from a global set of marine and freshwater populations, we identify a genome-wide set of loci that are consistently associated with marine–freshwater divergence. Our results indicate that reuse of globally shared standing genetic variation, including chromosomal inversions, has an important role in repeated evolution of distinct marine and freshwater sticklebacks, and in the maintenance of divergent ecotypes during early stages of reproductive isolation. Both coding and regulatory changes occur in the set of loci underlying marine–freshwater evolution, but regulatory changes appear to predominate in this well known example of repeated adaptive evolution in nature.


Nature | 2004

Genetic and developmental basis of evolutionary pelvic reduction in threespine sticklebacks.

Michael D. Shapiro; Melissa E. Marks; Catherine L. Peichel; Benjamin K. Blackman; Kirsten S. Nereng; Bjarni Jónsson; Dolph Schluter; David M. Kingsley

Hindlimb loss has evolved repeatedly in many different animals by means of molecular mechanisms that are still unknown. To determine the number and type of genetic changes underlying pelvic reduction in natural populations, we carried out genetic crosses between threespine stickleback fish with complete or missing pelvic structures. Genome-wide linkage mapping shows that pelvic reduction is controlled by one major and four minor chromosome regions. Pitx1 maps to the major chromosome region controlling most of the variation in pelvic size. Pelvic-reduced fish show the same left–right asymmetry seen in Pitx1 knockout mice, but do not show changes in Pitx1 protein sequence. Instead, pelvic-reduced sticklebacks show site-specific regulatory changes in Pitx1 expression, with reduced or absent expression in pelvic and caudal fin precursors. Regulatory mutations in major developmental control genes may provide a mechanism for generating rapid skeletal changes in natural populations, while preserving the essential roles of these genes in other processes.


Science | 2010

Adaptive evolution of pelvic reduction in sticklebacks by recurrent deletion of a Pitx1 enhancer

Yingguang Frank Chan; Melissa E. Marks; Felicity C. Jones; Guadalupe Villarreal; Michael D. Shapiro; Shannon D. Brady; Audrey Southwick; Devin Absher; Jane Grimwood; Jeremy Schmutz; Richard M. Myers; Dmitri A. Petrov; Bjarni Jónsson; Dolph Schluter; Michael A. Bell; David M. Kingsley

Adaptive Girdle Loss in Sticklebacks How do molecular changes give rise to phenotypic adaptation exemplified by the repeated reduction in the pelvic girdle observed in separate populations of sticklebacks? Now Chan et al. (p. 302, published online 10 December) have identified the specific DNA changes that control this major skeletal adaptation. The key locus controlling pelvic phenotypes mapped to a noncoding regulatory region upstream of the Pituitary homeobox transcription factor 1 gene, which drives a tissue-specific pelvic enhancer. Multiple populations showed independent deletions in this region and enhancer function was inactivated. Reintroduction of the enhancer restored pelvic development in a pelvic-reduced stickleback. Loss of a tissue-specific enhancer explains multiple parallel losses of the pelvic girdle in stickleback populations. The molecular mechanisms underlying major phenotypic changes that have evolved repeatedly in nature are generally unknown. Pelvic loss in different natural populations of threespine stickleback fish has occurred through regulatory mutations deleting a tissue-specific enhancer of the Pituitary homeobox transcription factor 1 (Pitx1) gene. The high prevalence of deletion mutations at Pitx1 may be influenced by inherent structural features of the locus. Although Pitx1 null mutations are lethal in laboratory animals, Pitx1 regulatory mutations show molecular signatures of positive selection in pelvic-reduced populations. These studies illustrate how major expression and morphological changes can arise from single mutational leaps in natural populations, producing new adaptive alleles via recurrent regulatory alterations in a key developmental control gene.


Cell | 1992

The mouse short ear skeletal morphogenesis locus is associated with defects in a bone morphogenetic member of the TGFβ superfamily

David M. Kingsley; Adrienne E. Bland; Janet M. Grubber; Paul C. Marker; Liane B. Russell; Neal G. Copeland; Nancy A. Jenkins

The mouse short ear gene is required for normal growth and patterning of skeletal structures, and for repair of bone fractures in adults. We have carried out an extensive chromosome walk in the chromosome region that surrounds this locus. Here we show that the short ear region contains the gene for a TGF beta-related protein called bone morphogenetic protein 5 (Bmp-5). This gene is deleted or rearranged in several independent mutations at the short ear locus. Mice homozygous for large deletions of the Bmp-5 coding region are viable and fertile. Mutations at the short ear locus provide an important new tool for defining the normal functions of BMPs in mammals. The specific skeletal defects seen in short-eared animals, which occur against a background of otherwise normal skeletal structures, suggest that particular aspects of skeletal morphology may be determined by individual members of a family of signaling factors that can induce the formation of cartilage and bone in vivo.


Nature | 2004

Evidence for ecology's role in speciation

Jeffrey S. McKinnon; Seiichi Mori; Benjamin K. Blackman; Lior David; David M. Kingsley; Leia Jamieson; Jennifer Chou; Dolph Schluter

A principal challenge in testing the role of natural selection in speciation is to connect the build-up of reproductive isolation between populations to divergence of ecologically important traits. Demonstrations of ‘parallel speciation’, or assortative mating by selective environment, link ecology and isolation, but the phenotypic traits mediating isolation have not been confirmed. Here we show that the parallel build-up of mating incompatibilities between stickleback populations can be largely accounted for by assortative mating based on one trait, body size, which evolves predictably according to environment. In addition to documenting the influence of body size on reproductive isolation for stickleback populations spread across the Northern Hemisphere, we have confirmed its importance through a new experimental manipulation. Together, these results suggest that speciation may arise largely as a by-product of ecological differences and divergent selection on a small number of phenotypic traits.


Journal of Heredity | 2009

Genome 10K: A Proposal to Obtain Whole-Genome Sequence for 10 000 Vertebrate Species

David Haussler; Stephen J. O'Brien; Oliver A. Ryder; F. Keith Barker; Michele Clamp; Andrew J. Crawford; Robert Hanner; Olivier Hanotte; Warren E. Johnson; Jimmy A. McGuire; Webb Miller; Robert W. Murphy; William J. Murphy; Frederick H. Sheldon; Barry Sinervo; Byrappa Venkatesh; E. O. Wiley; Fred W. Allendorf; George Amato; C. Scott Baker; Aaron M. Bauer; Albano Beja-Pereira; Eldredge Bermingham; Giacomo Bernardi; Cibele R. Bonvicino; Sydney Brenner; Terry Burke; Joel Cracraft; Mark Diekhans; Scott V. Edwards

The human genome project has been recently complemented by whole-genome assessment sequence of 32 mammals and 24 nonmammalian vertebrate species suitable for comparative genomic analyses. Here we anticipate a precipitous drop in costs and increase in sequencing efficiency, with concomitant development of improved annotation technology and, therefore, propose to create a collection of tissue and DNA specimens for 10,000 vertebrate species specifically designated for whole-genome sequencing in the very near future. For this purpose, we, the Genome 10K Community of Scientists (G10KCOS), will assemble and allocate a biospecimen collection of some 16,203 representative vertebrate species spanning evolutionary diversity across living mammals, birds, nonavian reptiles, amphibians, and fishes (ca. 60,000 living species). In this proposal, we present precise counts for these 16,203 individual species with specimens presently tagged and stipulated for DNA sequencing by the G10KCOS. DNA sequencing has ushered in a new era of investigation in the biological sciences, allowing us to embark for the first time on a truly comprehensive study of vertebrate evolution, the results of which will touch nearly every aspect of vertebrate biological enquiry.


PLOS Biology | 2004

The Genetic Architecture of Parallel Armor Plate Reduction in Threespine Sticklebacks

Pamela F. Colosimo; Catherine L. Peichel; Kirsten S. Nereng; Benjamin K. Blackman; Michael D. Shapiro; Dolph Schluter; David M. Kingsley

How many genetic changes control the evolution of new traits in natural populations? Are the same genetic changes seen in cases of parallel evolution? Despite long-standing interest in these questions, they have been difficult to address, particularly in vertebrates. We have analyzed the genetic basis of natural variation in three different aspects of the skeletal armor of threespine sticklebacks (Gasterosteus aculeatus): the pattern, number, and size of the bony lateral plates. A few chromosomal regions can account for variation in all three aspects of the lateral plates, with one major locus contributing to most of the variation in lateral plate pattern and number. Genetic mapping and allelic complementation experiments show that the same major locus is responsible for the parallel evolution of armor plate reduction in two widely separated populations. These results suggest that a small number of genetic changes can produce major skeletal alterations in natural populations and that the same major locus is used repeatedly when similar traits evolve in different locations.


Current Biology | 2004

The Master Sex-Determination Locus in Threespine Sticklebacks Is on a Nascent Y Chromosome

Catherine L. Peichel; Joseph A. Ross; Clinton K. Matson; Mark Dickson; Jane Grimwood; Jeremy Schmutz; Richard M. Myers; Seiichi Mori; Dolph Schluter; David M. Kingsley

BACKGROUND Many different environmental and genetic sex-determination mechanisms are found in nature. Closely related species can use different master sex-determination switches, suggesting that these developmental pathways can evolve very rapidly. Previous cytological studies suggest that recently diverged species of stickleback fish have different sex chromosome complements. Here, we investigate the genetic and chromosomal mechanisms that underlie sex determination in the threespine stickleback (Gasterosteus aculeatus). RESULTS Genome-wide linkage mapping identifies a single chromosome region at the distal end of linkage group (LG) 19, which controls male or female sexual development in threespine sticklebacks. Although sex chromosomes are not cytogenetically visible in this species, several lines of evidence suggest that LG 19 is an evolving sex chromosome system, similar to the XX female/XY male system in many other species: (1) males are consistently heterozygous for unique alleles in this region; (2) recombination between loci linked to the sex-determination region is reduced in male meiosis relative to female meiosis; (3) sequence analysis of X- and Y-specific bacterial artificial chromosome (BAC) clones from the sex-determination region reveals many sequence differences between the X- and Y-specific clones; and (4) the Y chromosome has accumulated transposable elements and local duplications. CONCLUSIONS Taken together, our data suggest that threespine sticklebacks have a simple chromosomal mechanism for sex determination based on a nascent Y chromosome that is less than 10 million years old. Further analysis of the stickleback system will provide an exciting window into the evolution of sex-determination pathways and sex chromosomes in vertebrates.


Trends in Genetics | 1994

What do BMPs do in mammals? Clues from the mouse short-ear mutation.

David M. Kingsley

Bone morphogenetic proteins (BMPs) are a family of secreted signaling molecules that were originally isolated on the basis of their remarkable ability to induce the formation of ectopic bones when implanted into adult animals. The first mutations identified in a mammalian BMP gene suggest that members of this family induce the formation, patterning and repair of particular morphological features in higher animals.


Cell | 2007

Cis-regulatory changes in Kit ligand expression and parallel evolution of pigmentation in sticklebacks and humans

Craig T. Miller; Sandra Beleza; Alex A. Pollen; Dolph Schluter; Rick A. Kittles; Mark D. Shriver; David M. Kingsley

Dramatic pigmentation changes have evolved within most vertebrate groups, including fish and humans. Here we use genetic crosses in sticklebacks to investigate the parallel origin of pigmentation changes in natural populations. High-resolution mapping and expression experiments show that light gills and light ventrums map to a divergent regulatory allele of the Kit ligand (Kitlg) gene. The divergent allele reduces expression in gill and skin tissue and is shared by multiple derived freshwater populations with reduced pigmentation. In humans, Europeans and East Asians also share derived alleles at the KITLG locus. Strong signatures of selection map to regulatory regions surrounding the gene, and admixture mapping shows that the KITLG genomic region has a significant effect on human skin color. These experiments suggest that regulatory changes in Kitlg contribute to natural variation in vertebrate pigmentation, and that similar genetic mechanisms may underlie rapid evolutionary change in fish and humans.

Collaboration


Dive into the David M. Kingsley's collaboration.

Top Co-Authors

Avatar

Dolph Schluter

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Catherine L. Peichel

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nancy A. Jenkins

Houston Methodist Hospital

View shared research outputs
Top Co-Authors

Avatar

Neal G. Copeland

Houston Methodist Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Monty Krieger

Massachusetts Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge