Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Developmental mechanisms of stripe patterns in rodents

Abstract

Mammalian colour patterns are among the most recognizable characteristics found in nature and can have a profound impact on fitness. However, little is known about the mechanisms underlying the formation and subsequent evolution of these patterns. Here we show that, in the African striped mouse (Rhabdomys pumilio), periodic dorsal stripes result from underlying differences in melanocyte maturation, which give rise to spatial variation in hair colour. We identify the transcription factor ALX3 as a regulator of this process. In embryonic dorsal skin, patterned expression of Alx3 precedes pigment stripes and acts to directly repress Mitf, a master regulator of melanocyte differentiation, thereby giving rise to light-coloured hair. Moreover, Alx3 is upregulated in the light stripes of chipmunks, which have independently evolved a similar dorsal pattern. Our results show a previously undescribed mechanism for modulating spatial variation in hair colour and provide insights into how phenotypic novelty evolves.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Phenotypic characterization.
Figure 2: Alx3 is a candidate for regulating spatial differences in hair colour.
Figure 3: Alx3 decreases melanin synthesis in vivo.
Figure 4: Alx3 binds to the Mitf promoter directly.
Figure 5: Hair colour patterning mechanisms in rodents.

Similar content being viewed by others

References

  1. Barsh, G. S. The genetics of pigmentation: from fancy genes to complex traits. Trends Genet. 12, 299–305 (1996)

    Article  CAS  Google Scholar 

  2. Jackson, I. J. Molecular and developmental genetics of mouse coat color. Annu. Rev. Genet. 28, 189–217 (1994)

    Article  CAS  Google Scholar 

  3. Mills, M. G. & Patterson, L. B. Not just black and white: pigment pattern development and evolution in vertebrates. Semin. Cell Dev. Biol. 20, 72–81 (2009)

    Article  CAS  Google Scholar 

  4. Candille, S. I. et al. Dorsoventral patterning of the mouse coat by Tbx15. PLoS Biol. 2, E3 (2004)

    Article  Google Scholar 

  5. Morris, D. Animal Watching. A Field Guide to Animal Behavior. (Jonathan Cape Ltd, 1990)

  6. Cloudsley-Thompson, J. L. Multiple factors in the evolution of animal coloration. Naturwissenschaften 86, 123–132 (1999)

    Article  CAS  ADS  Google Scholar 

  7. Caro, T., Izzo, A., Reiner, R. C. Jr, Walker, H. & Stankowich, T. The function of zebra stripes. Nat. Commun. 5, 3535 (2014)

    Article  ADS  Google Scholar 

  8. Brodie, E. D. III . Correlational selection for color pattern and antipredator behavior in the garter snake Thamnophis ordinoides. Evolution 46, 1284–1298 (1992)

    Article  Google Scholar 

  9. King, R. B. Color pattern polymorphism in the Lake Erie water snake, Nerodia sipedon insularum. Evolution 41, 241–255 (1987)

    PubMed  Google Scholar 

  10. Caro, T. The adaptive significance of coloration in mammals. Bioscience 55, 125–136 (2005)

    Article  Google Scholar 

  11. Schradin, C. et al. Social flexibility and social evolution in mammals: a case study of the African striped mouse (Rhabdomys pumilio). Mol. Ecol. 21, 541–553 (2012)

    Article  Google Scholar 

  12. Steingrímsson, E., Copeland, N. G. & Jenkins, N. A. Melanocytes and the microphthalmia transcription factor network. Annu. Rev. Genet. 38, 365–411 (2004)

    Article  Google Scholar 

  13. ten Berge, D. et al. Mouse Alx3: an aristaless-like homeobox gene expressed during embryogenesis in ectomesenchyme and lateral plate mesoderm. Dev. Biol. 199, 11–25 (1998)

    Article  CAS  Google Scholar 

  14. Beverdam, A., Brouwer, A., Reijnen, M., Korving, J. & Meijlink, F. Severe nasal clefting and abnormal embryonic apoptosis in Alx3/Alx4 double mutant mice. Development 128, 3975–3986 (2001)

    CAS  PubMed  Google Scholar 

  15. Twigg, S. R. F. et al. Frontorhiny, a distinctive presentation of frontonasal dysplasia caused by recessive mutations in the ALX3 homeobox gene. Am. J. Hum. Genet. 84, 698–705 (2009)

    Article  CAS  Google Scholar 

  16. Lakhwani, S., García-Sanz, P. & Vallejo, M. Alx3-deficient mice exhibit folic acid-resistant craniofacial midline and neural tube closure defects. Dev. Biol. 344, 869–880 (2010)

    Article  CAS  Google Scholar 

  17. Peters, E. M. J., Tobin, D. J., Botchkareva, N., Maurer, M. & Paus, R. Migration of melanoblasts into the developing murine hair follicle is accompanied by transient c-Kit expression. J. Histochem. Cytochem. 50, 751–766 (2002)

    Article  CAS  Google Scholar 

  18. Beronja, S., Livshits, G., Williams, S. & Fuchs, E. Rapid functional dissection of genetic networks via tissue-specific transduction and RNAi in mouse embryos. Nat. Med. 16, 821–827 (2010)

    Article  CAS  Google Scholar 

  19. Lee, M., Goodall, J., Verastegui, C., Ballotti, R. & Goding, C. R. Direct regulation of the Microphthalmia promoter by Sox10 links Waardenburg-Shah syndrome (WS4)-associated hypopigmentation and deafness to WS2. J. Biol. Chem. 275, 37978–37983 (2000)

    Article  CAS  Google Scholar 

  20. Potterf, S. B., Furumura, M., Dunn, K. J., Arnheiter, H. & Pavan, W. J. Transcription factor hierarchy in Waardenburg syndrome: regulation of MITF expression by SOX10 and PAX3. Hum. Genet. 107, 1–6 (2000)

    Article  CAS  Google Scholar 

  21. Elworthy, S., Lister, J. A., Carney, T. J., Raible, D. W. & Kelsh, R. N. Transcriptional regulation of mitfa accounts for the sox10 requirement in zebrafish melanophore development. Development 130, 2809–2818 (2003)

    Article  CAS  Google Scholar 

  22. Pérez-Villamil, B., Mirasierra, M. & Vallejo, M. The homeoprotein Alx3 contains discrete functional domains and exhibits cell-specific and selective monomeric binding and transactivation. J. Biol. Chem. 279, 38062–38071 (2004)

    Article  Google Scholar 

  23. Levy, C., Khaled, M. & Fisher, D. E. MITF: master regulator of melanocyte development and melanoma oncogene. Trends Mol. Med. 12, 406–414 (2006)

    Article  CAS  Google Scholar 

  24. García-Sanz, P., Fernández-Pérez, A. & Vallejo, M. Differential configurations involving binding of USF transcription factors and Twist1 regulate Alx3 promoter activity in mesenchymal and pancreatic cells. Biochem. J. 450, 199–208 (2013)

    Article  Google Scholar 

  25. Meredith, R. W. et al. Impacts of the Cretaceous Terrestrial Revolution and KPg extinction on mammal diversification. Science 334, 521–524 (2011)

    Article  CAS  ADS  Google Scholar 

  26. dos Reis, M. et al. Phylogenomic datasets provide both precision and accuracy in estimating the timescale of placental mammal phylogeny. Proc. R. Soc. B 279, 3491–3500 (2012)

    Article  Google Scholar 

  27. Huchon, D. et al. Multiple molecular evidences for a living mammalian fossil. Proc. Natl Acad. Sci. USA 104, 7495–7499 (2007)

    Article  CAS  ADS  Google Scholar 

  28. Rawls, J. F., Mellgren, E. M. & Johnson, S. L. How the zebrafish gets its stripes. Dev. Biol. 240, 301–314 (2001)

    Article  CAS  Google Scholar 

  29. Parichy, D. M. Pigment patterns: fish in stripes and spots. Curr. Biol. 13, R947–R950 (2003)

    Article  CAS  Google Scholar 

  30. Singh, A. P. & Nüsslein-Volhard, C. Zebrafish stripes as a model for vertebrate colour pattern formation. Curr. Biol. 25, R81–R92 (2015)

    Article  CAS  Google Scholar 

  31. Jackson, I. J. et al. Genetics and molecular biology of mouse pigmentation. Pigment Cell Res. 7, 73–80 (1994)

    Article  CAS  Google Scholar 

  32. Kaelin, C. B. et al. Specifying and sustaining pigmentation patterns in domestic and wild cats. Science 337, 1536–1541 (2012)

    Article  CAS  ADS  Google Scholar 

  33. Vrieling, H., Duhl, D. M., Millar, S. E., Miller, K. A. & Barsh, G. S. Differences in dorsal and ventral pigmentation result from regional expression of the mouse agouti gene. Proc. Natl Acad. Sci. USA 91, 5667–5671 (1994)

    Article  CAS  ADS  Google Scholar 

  34. Manceau, M., Domingues, V. S., Mallarino, R. & Hoekstra, H. E. The developmental role of Agouti in color pattern evolution. Science 331, 1062–1065 (2011)

    Article  CAS  ADS  Google Scholar 

  35. Schneider, C. A., Rasband, W. S. & Eliceiri, K. W. NIH Image to ImageJ: 25 years of image analysis. Nat. Methods 9, 671–675 (2012)

    Article  CAS  Google Scholar 

  36. McCloy, R. A. et al. Partial inhibition of Cdk1 in G 2 phase overrides the SAC and decouples mitotic events. Cell Cycle 13, 1400–1412 (2014)

    Article  CAS  Google Scholar 

  37. Spandidos, A., Wang, X., Wang, H. & Seed, B. PrimerBank: a resource of human and mouse PCR primer pairs for gene expression detection and quantification. Nucleic Acids Res. 38, D792–D799 (2010)

    Article  CAS  Google Scholar 

  38. Livak, K. J. & Schmittgen, T. D. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods 25, 402–408 (2001)

    Article  CAS  Google Scholar 

  39. Henrique, D. et al. Expression of a Delta homologue in prospective neurons in the chick. Nature 375, 787–790 (1995)

    Article  CAS  ADS  Google Scholar 

  40. Mallarino, R. et al. Two developmental modules establish 3D beak-shape variation in Darwin’s finches. Proc. Natl Acad. Sci. USA 108, 4057–4062 (2011)

    Article  CAS  ADS  Google Scholar 

  41. Moffat, J. et al. A lentiviral RNAi library for human and mouse genes applied to an arrayed viral high-content screen. Cell 124, 1283–1298 (2006)

    Article  CAS  Google Scholar 

  42. Kent, W. J. et al. The human genome browser at UCSC. Genome Res. 12, 996–1006 (2002)

    Article  CAS  Google Scholar 

  43. Schwartz, S. et al. PipMaker—a web server for aligning two genomic DNA sequences. Genome Res. 10, 577–586 (2000)

    Article  CAS  Google Scholar 

  44. Brudno, M. et al. LAGAN and Multi-LAGAN: efficient tools for large-scale multiple alignment of genomic DNA. Genome Res. 13, 721–731 (2003)

    Article  CAS  Google Scholar 

  45. Schreiber, E., Matthias, P., Müller, M. M. & Schaffner, W. Rapid detection of octamer binding proteins with ‘mini-extracts’, prepared from a small number of cells. Nucleic Acids Res. 17, 6419 (1989)

    Article  CAS  Google Scholar 

  46. Mirasierra, M. & Vallejo, M. Glucose-dependent downregulation of glucagon gene expression mediated by selective interactions between ALX3 and PAX6 in mouse alpha cells. Diabetologia 59, 766–775 (2016)

    Article  CAS  Google Scholar 

  47. de la Serna, I. L. et al. The microphthalmia-associated transcription factor requires SWI/SNF enzymes to activate melanocyte-specific genes. J. Biol. Chem. 281, 20233–20241 (2006)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank D. Mishkind, M. Omura, J. Chupasko, P. Walsh, T. Capellini, T. Linden, K. Turner and N. Hughes for providing technical and logistical support, and C. Perdomo, A. Bendesky, C. K. Hu, D. M. Kingsley and J. M. Lassance for discussions. M.Mi. and M.V. are supported by the Spanish Ministry of Economy and Competitiveness (MINECO grants BFU2011-24245 and BFU2014-52149-R) and Instituto de Salud Carlos III. CIBERDEM is an initiative of the Instituto de Salud Carlos III. H.E.H. is an Investigator of the Howard Hughes Medical Institute.

Author information

Authors and Affiliations

Authors

Contributions

R.M., M.Ma. and H.E.H. conceived the project. R.M., G.S.B. and H.E.H. designed experiments. R.M. performed cell proliferation assays, immunohistochemistry, quantitative PCR, in situ hybridizations, comparative sequence analysis, in vitro gain- and loss-of-function experiments and luciferase reporter assays. R.M. and M.Ma. collected samples and performed phenotypic characterization; R.M. and S.B. performed in vivo ultrasound-guided lentiviral injections. C.H. carried out the large-scale RNA experiments, including construction and annotation of the de novo transcriptome, and design and analysis of the RNA-seq work. M.Mi. and M.V. performed protein–DNA binding assays. C.S. provided the first embryos for pilot studies and founding members for the striped mice laboratory colony. R.M., G.S.B. and H.E.H. wrote the paper with input from all authors.

Corresponding author

Correspondence to Hopi E. Hoekstra.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Reviewer Information Nature thanks H. Arnheiter, T. Caro, M. Levine and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Extended data figures and tables

Extended Data Figure 1 Hair characterization in adult striped mice.

a, Striped mice have three different phenotypic categories of hair (light, black and banded) based on individual pigment pattern. All hair types have a black tip, which corresponds to structural hair features (not pigment). b, Relative proportion of light, black and banded guard and awl hair along the striped mouse dorsoventral axis (n = 5; error bars represent s.e.m.). a, Scale bar, 100 μm.

Source data

Extended Data Figure 2 Stripe-like differences in hair length along the dorsum in striped mouse embryos and pups.

ac, Flat-mount skin preparations (dermis up) of embryos at E16 (a) and E19 (b) and P2 pups (c). Middle axis is indicated in all cases (midline). White dashed lines mark regions differing in hair length at E19 (b) and regions differing in pigmentation at P2 (c). Incipient pigmentation stripes are shown in (b). d, Skin punches (1 mm) and length measurements show differences between hair in the dark and light stripe of P2 individuals. Hair length differences in (b) (incipient stripes) correlate with those seen when pigment differences arise (c, d). Differences among dorsal regions were evaluated by ANOVA followed by a Tukey–Kramer test; n = 15 per region; **P < 0.01; ***P < 0.001. Red lines depict the mean. e, Hair length measurements taken from guard, awl and zigzag hair found along the dorsum of adults. Differences among dorsal regions were evaluated by ANOVA followed by a Tukey–Kramer test; n = 24 (guard), 12 (awl) and 12 (zigzag) per region; P = 0.1736 (guard hair), P = 0.8006 (awl hair), P = 0.1038 (zigzag hair). Red lines depict the mean. f, Predicted probabilities of the observed stripe-like phenotypes, as inferred by supervised learning models built and trained to recognize time-point-specific gene expression signatures of the stripes. Bars reflect average probabilities ± s.e.m. computed from 30 consecutive iterations of the predictive model in each analysis. Labels indicate, as a ratio, the time point at which the randomForests model was applied to predict the stripe-like phenotype (the left term of the ratio), and the time point at which the model was built and trained (the right term of the ratio). The dotted line indicates the prior probability of either stripe phenotype (that is, 50% in case of only two distinct phenotypes). See Methods for details.

Source data

Extended Data Figure 3 Cell proliferation and hair follicle density in P2 striped mice.

a, Counts of proliferating cells, as determined by EdU labelling, in the epidermis and inside hair follicles (epidermal cells, dark stripe 1 versus light stripe, two-tailed t-test; n = 15 images per region; P = 0.5417; cells counted: 402 (dark stripe 1) and 444 (light stripe); intrafollicular cells, dark stripe 1 versus light stripe, two-tailed t-test; n = 12 images per region; P = 0.7537; cells counted: 724 (dark stripe 1) and 680 (light stripe)). Dotted lines delineate the epidermal compartment and the hair bulb. b, Number of hair follicles per surface area along the dorsoventral axis. Differences among dorsal regions were evaluated by ANOVA; n = 10 images per region; P = 0.4391; number of hair follicles counted: 139 (light stripe), 141 (dark stripe 1), 132 (dark stripe 2) and 128 (flank). Bright field images in a depict pigment. Boxes represent the 25th to 75th percentile, whiskers show the minimum and maximum value, and the horizontal line indicates the median. Red bars indicate the mean. Scale bars in a 100 μm, b 200 μm.

Source data

Extended Data Figure 4 RNA-seq analysis.

ad, Venn diagrams showing numbers of differentially expressed genes identified using either the M. musculus reference or the striped mouse de novo transcriptome assembly in light versus dark stripes (a), light versus flank (b), and flank versus dark (c), or differentially expressed genes in light or dark stripes versus the other skin region (light or dark stripes and the flank) (d). Genes that are specifically upregulated only in the dark or light stripes are highlighted in red. e, f, RNA-seq transcript levels (normalized gene counts) plotted as a function of differential expression (log2 fold-change). e, The 1,777 genes demonstrating significant (FDR < 0.1) differential expression in the light stripe versus the flank are shown in yellow (higher expression in the light stripe) or blue (higher expression in the flank). Four differentially expressed pigmentation-related genes are highlighted (dark yellow or dark blue), while 11 additional pigmentation-related genes that are not differentially expressed are shown in pink (Supplementary Table 1b). f, The 1,148 genes demonstrating significant (FDR < 0.1) differential expression in the flank versus the dark stripe are shown in yellow (higher expression in the flank) or blue (higher expression in the dark stripe). Eleven differentially expressed pigmentation-related genes are highlighted (dark yellow or dark blue), while six additional pigmentation-related genes that are not differentially expressed are shown in pink (Supplementary Table 1c).

Extended Data Figure 5 Stage-specific gene expression.

ac, Quantitative PCR of the relative mRNA levels of pigment-type switching genes Asip (a), Edn3 (b), and melanin synthesis genes Tyr and Tyrp1 (c) in different regions of the striped mouse skin and at different time points. Differences among stripes within each time point were evaluated by ANOVA followed by a Tukey–Kramer test; n = 4 per time point; *P < 0.05; **P < 0.01; ***P < 0.001. Red bars indicate the mean.

Source data

Extended Data Figure 6 Gain- and loss-of-function experiments in cultured cells.

a, Lentiviral constructs were modified from PLKO.1, a generic vector for expressing human RNU6-1 promoter-driven short hairpin RNAs (red loop). LTR, long terminal repeat; ψ, retroviral packaging element, RRE, Rev response element; cPPT, central polypurine tract; PGK, phosphoglycerate kinase promoter; H2B–GFP, Hist2h2be fused to GFP cDNA; P2A, 2A peptide. b, Western blot for expression of ALX3 in nuclear extracts of B16-F1 cells. Positive controls were extracts from mouse embryonic mesenchyme (MEM) or COS cells transfected with a pcDNA–ALX3 expression vector. COS cells transfected with an empty pcDNA vector served as negative controls. Actin immunoreactivity is shown below for the same extracts as a control. For gel source images, see Supplementary Fig. 1. c, d, Quantitative PCR of Alx3 (white), Mitf (black) and Silver (grey) mRNA levels in cells transduced with LV–Alx3:GFP (c), relative to cells transduced with the LV–GFP control (LV–Alx3:GFP versus LV–GFP, two-tailed t-test; n = 3) or shRNA lentiviral constructs (d), relative to cells transduced with a scrambled control (shRNA1, 2, 3 or 4 versus shRNA scrambled, two-tailed t-test; n = 3). **P < 0.01; ***P < 0.001. Red bars indicate the mean.

Source data

Extended Data Figure 7 Co-culture experiments.

ad, Wild-type B16 melanocytes (B16 wild-type (WT)) were exposed to keratinocytes (a) or melanocytes (c) stably transduced with either LV–Alx3:GFP (LV–Alx3 in graphs) or LV–GFP. b, d, Quantitative PCR of levels of Alx3 mRNA in cells carrying the lentiviral constructs (grey panel) and of Mitf, Tyr and Silver mRNA in B16 wild-type melanocytes exposed to keratinocytes (b) or melanocytes (d) transduced with LV–Alx3:GFP (blue panels) or LV–GFP (red panels) (LV–Alx3:GFP versus LV–GFP, two-tailed t-test; n = 3, ***P < 0.001; NS, not significant). Red bars in b and d indicate the mean.

Source data

Extended Data Figure 8 Ultrasound-guided in utero lentiviral injections.

af, Hair follicles from embryos injected at E8.5 with lentiviruses stained for SOX10 (a, d), virus transduced cells (b, e) and merged images with arrowheads showing SOX+ GFP+ cells (c, f). Dotted lines (af) delineate the hair bulb. g, Number of detectable SOX10+ cells (LV–Alx3:GFP versus LV–GFP, two-tailed t-test; n = 60; P = 0.1173; cells counted: 426 cells (LV–Alx3:GFP) and 398 cells (LV–GFP)). ho, Effect of Alx3 on skin. Hair follicles from samples injected with LV–GFP control and LV–Alx3:GFP depicting immunohistochemistry for K14 (h, k), virus transduced cells (i, l), and merged images showing K14+ GFP+ cells (j, m). n, Number of detectable K14+ GFP+ cells per follicular area (LV–Alx3:GFP versus LV–GFP, two-tailed t-test; n = 40, P = 0.275; average: 52.809 cells per hair follicle area (LV–Alx3:GFP) and 55.123 cells per hair follicle area (LV–GFP)). o, Hair follicle density in samples injected with viruses (LV–Alx3:GFP versus LV–GFP, two-tailed t-test; n = 30; P = 0.103; average: 0.84 hair follicles per surface area (LV–Alx3:GFP) and 0.794 hair follicles per surface area (LV–GFP)). Boxes represent the 25th to 75th percentile, whiskers show the minimum and maximum value, and the horizontal line indicates the median. Scale bars in af and hm are 50 μM.

Source data

Extended Data Figure 9 Alignment of a ~1.5-kb region of the Mitf M promoter in M. musculus and striped mouse.

Black boxes represent conserved sequences. Mapped onto the sequences are evolutionary conserved regions of the mammalian Mitf M promoter identified in silico (http://genome.ucsc.edu) (yellow), regions from which the EMSA probes were designed (red), and the transcription start site (green). The ten TAAT binding sites, conserved between M. musculus and striped mice (blue), which were tested are labelled sequentially.

Extended Data Figure 10 EMSA and luciferase assays.

a, EMSAs show the binding of nuclear proteins from B16-F1 cells to candidate-binding sites 3, 5 and 10. The absence (−) or presence of non-specific (NSC; 500-fold molar excess) or specific (SC; indicated fold molar excess) competing oligonucleotides, or the addition of ALX3 antibodies or control (NRS or IgG) is indicated. Arrows indicate complexes containing ALX3, arrowhead shows supershift for site 3 and asterisk highlights an artefact in the gel. b, EMSAs showing the binding of recombinant Alx3 synthesized using a rabbit reticulocyte lysate system to the indicated sites (sites 4–9). The absence (−) or presence of non-specific (NSC) or specific (SC) competing oligonucleotides (500-fold molar excess) is indicated. Note that site 5 is the only one showing sequence-specific binding. For gel source images, see Supplementary Fig. 1. c, Relative levels of luciferase activity in B16-F1 cells stably expressing GFP (dark circles) or Alx3 (light circles) for indicated binding sites. Luciferase activity was normalized relative to cells transfected with the empty reporter vector and values are given as a fraction of luminescence for GFP-transfected cells. Differences between cells transfected with LV–Alx3:GFP and LV–GFP for each plasmid were evaluated using two-tailed t-tests; n = 5; *P < 0.05; NS, not significant. Red lines depict the mean. Labels of mutated binding sites correspond to those described in Fig. 4a.

Source data

Supplementary information

Supplementary Information

This file contains the uncropped gels and Supplementary Tables 1-3. (PDF 2478 kb)

PowerPoint slides

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mallarino, R., Henegar, C., Mirasierra, M. et al. Developmental mechanisms of stripe patterns in rodents. Nature 539, 518–523 (2016). https://doi.org/10.1038/nature20109

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature20109

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing