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.

  • Original Article
  • Published:

A homozygous loss-of-function mutation in inositol monophosphatase 1 (IMPA1) causes severe intellectual disability

Abstract

The genetic basis of intellectual disability (ID) is extremely heterogeneous and relatively little is known about the role of autosomal recessive traits. In a field study performed in a highly inbred area of Northeastern Brazil, we identified and investigated a large consanguineous family with nine adult members affected by severe ID associated with disruptive behavior. The Genome-Wide Human SNP Array 6.0 microarray was used to determine regions of homozygosity by descent from three affected and one normal family member. Whole-exome sequencing (WES) was performed in one affected patient using the Nextera Rapid-Capture Exome kit and Illumina HiSeq2500 system to identify the causative mutation. Potentially deleterious variants detected in regions of homozygosity by descent and not present in either 59 723 unrelated individuals from the Exome Aggregation Consortium (Browser) or 1484 Brazilians were subject to further scrutiny and segregation analysis by Sanger sequencing. Homozygosity-by-descent analysis disclosed a 20.7-Mb candidate region at 8q12.3-q21.2 (lod score: 3.11). WES identified a homozygous deleterious variant in inositol monophosphatase 1 (IMPA1) (NM_005536), consisting of a 5-bp duplication (c.489_493dupGGGCT; chr8: 82,583,247; GRCh37/hg19) leading to a frameshift and a premature stop codon (p.Ser165Trpfs*10) that cosegregated with the disease in 26 genotyped family members. The IMPA1 gene product is responsible for the final step of biotransformation of inositol triphosphate and diacylglycerol, two second messengers. Despite its many physiological functions, no clinical phenotype has been assigned to this gene dysfunction to date. Additionally, IMPA1 is the main target of lithium, a drug that is at the forefront of treatment for bipolar disorder.

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
Figure 2

Similar content being viewed by others

Stylianos E. Antonarakis, Brian G. Skotko, … Roger H. Reeves

References

  1. American Psychiatric Association Diagnostic and Statistical Manual of Mental Disorders. American Psychiatric Association: Washington, DC, USA, 2000.

  2. Leonard H, Wen X . The epidemiology of mental retardation: challenges and opportunities in the new millennium. Ment Retard Dev Disabil Res Rev 2002; 8: 117–134.

    Article  Google Scholar 

  3. Rauch A, Hoyer J, Guth S, Zweier C, Kraus C, Becker C et al. Diagnostic yield of various genetic approaches in patients with unexplained developmental delay or mental retardation. Am J Med Genet A 2006; 140: 2063–2074.

    Article  Google Scholar 

  4. Ellison JW, Rosenfeld JA, Shaffer LG . Genetic basis of intellectual disability. Annu Rev Med 2013; 64: 441–450.

    Article  CAS  Google Scholar 

  5. Resnick AC, Saiardi A . Inositol polyphosphate multikinase: metabolic architect of nuclear inositides. Front Biosci 2008; 13: 856–866.

    Article  CAS  Google Scholar 

  6. Hakim S, Bertucci MC, Conduit SE, Vuong DL, Mitchell CA . Inositol polyphosphate phosphatases in human disease. Curr Top Microbiol Immunol 2012; 362: 247–314.

    CAS  PubMed  Google Scholar 

  7. Atack JR, Broughton HB, Pollack SJ . Inositol monophosphatase—a putative target for Li+ in the treatment of bipolar disorder. Trends Neurosci 1995; 18: 343–349.

    Article  CAS  Google Scholar 

  8. Cryns K, Shamir A, Van Acker N, Levi I, Daneels G, Goris I et al. IMPA1 is essential for embryonic development and lithium-like pilocarpine sensitivity. Neuropsychopharmacology 2008; 33: 674–684.

    Article  CAS  Google Scholar 

  9. Seelow D, Schuelke M, Hildebrandt F, Nürnberg P . HomozygosityMapper—an interactive approach to homozygosity mapping. Nucleic Acids Res 2009; 37: 593–599.

    Article  Google Scholar 

  10. Ruschendorf F, Nurnberg P . ALOHOMORA: a tool for linkage analysis using 10k SNP array data. Bioinformatics 2005; 12: 2123–2125.

    Article  Google Scholar 

  11. Abecasis G, Cherny S, Cookson W, Cardon L . Merlin—rapid analysis of dense genetic maps using sparse gene flow trees. Nat Genet 2002; 30: 97–101.

    Article  CAS  Google Scholar 

  12. Li H, Durbin R . Fast and accurate long-read alignment with Burrows-Wheeler transform. Bioinformatics 2010; 26: 589–595.

    Article  Google Scholar 

  13. McKenna A, Hanna M, Banks E, Sivachenko A, Cibulskis K, Kernytsky A et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res 2010; 20: 1297–1303.

    Article  CAS  Google Scholar 

  14. Cingolani P, Platts A, Wang le L, Coon M, Nguyen T, Wang L et al. A program for annotating and predicting the effects of single nucleotide polymorphisms, SnpEff: SNPs in the genome of Drosophila melanogaster strain w1118; iso-2; iso-3. Fly (Austin) 2012; 6: 80–92.

    Article  CAS  Google Scholar 

  15. Plagnol V, Curtis J, Epstein M, Mok KY, Stebbings E, Grigoriadou S et al. A robust model for read count data in exome sequencing experiments and implications for copy number variant calling. Bioinformatics 2012; 28: 2747–2754.

    Article  CAS  Google Scholar 

  16. Figueiredo T, Melo US, Pessoa AL, Nobrega PR, Kitajima JP, Correia I et al. Homozygous missense mutation in MED25 segregates with syndromic intellectual disability in a large consanguineous family. J Med Genet 2015; 52: 123–127.

    Article  CAS  Google Scholar 

  17. OMIM. OMIM database. http://www.ncbi.nlm.nih.gov/omim/.

  18. Berry GT, Wu S, Buccafusca R, Ren J, Gonzales LW, Ballard PL et al. Loss of murine Na+/myo-inositol cotransporter leads to brain myo-inositol depletion and central apnea. J Bio Chem 2003; 278: 18297–18302.

    Article  CAS  Google Scholar 

  19. Ohnishi T, Murata T, Watanabe A, Hida A, Ohba H, Iwayama Y et al. Defective craniofacial development and brain function in a mouse model for depletion of intracellular inositol synthesis. J Biol Chem 2014; 289: 10785–10796.

    Article  CAS  Google Scholar 

  20. Andreassi C, Zimmermann C, Mitter R, Fusco S, De Vita S, Saiardi A et al. An NGF-responsive element targets myo-inositol monophosphatase-1 mRNA to sympathetic neuron axons. Nat Neurosci 2010; 13: 291–301.

    Article  CAS  Google Scholar 

  21. Ohnishi T, Ohba H, Seo KC, Im J, Sato Y, Iwayama Y et al. Spatial expression patterns and biochemical properties distinguish a second myo-inositol monophosphatase IMPA2 from IMPA1. J Biol Chem 2007; 282: 637–646.

    Article  CAS  Google Scholar 

  22. Youngs RM, Chu MS, Meloni EG, Naydenov A, Carlezon WA Jr, Konradi C . Lithium administration to preadolescent rats causes long-lasting increases in anxiety-LIKE behavior and has molecular consequences. J Neurosci 2006; 26: 6031–6039.

    Article  CAS  Google Scholar 

  23. Tanizawa Y, Kuhara A, Inada H, Kodama E, Mizuno T, Mori I . Inositol monophosphatase regulates localization of synaptic components and behavior in the mature nervous system of C. elegans. Genes Dev 2006; 20: 3296–3310.

    Article  CAS  Google Scholar 

  24. Kimata T, Tanizawa Y, Can Y, Ikeda S, Kuhara A, Mori I . Synaptic polarity depends on phosphatidylinositol signaling regulated by myo-inositol monophosphatase in Caenorhabditis elegans. Genetics 2012; 191: 509–521.

    Article  CAS  Google Scholar 

  25. Ohnishi T, Tanizawa Y, Watanabe A, Nakamura T, Ohba H, Hirata H . Human myo-inositol monophosphatase 2 rescues the nematode thermotaxis mutant ttx-7 more efficiently than IMPA1: functional and evolutionary considerations of the two mammalian myo-inositol monophosphatase genes. J Neurochem 2013; 124: 685–694.

    Article  CAS  Google Scholar 

  26. Fisher SK, Novak JE, Agranoff BW . Inositol and higher inositol phosphates in neural tissues: homeostasis, metabolism and functional significance. J Neurochem 2012; 82: 736–754.

    Article  Google Scholar 

  27. Najmabadi H, Hu H, Garshasbi M, Zemojtel T, Abedini SS, Chen W et al. Deep sequencing reveals 50 novel genes for recessive cognitive disorders. Nature 2011; 478: 57–63.

    Article  CAS  Google Scholar 

  28. Arimura N, Kaibuchi K . Key regulators in neuronal polarity. Neuron 2005; 48: 881–884.

    Article  CAS  Google Scholar 

  29. Arimura N, Kaibuchi K . Neuronal polarity: from extracellular signals to intracellular mechanisms. Nat Rev Neurosci 2007; 8: 194–205.

    Article  CAS  Google Scholar 

  30. Wen PJ, Osborne SL, Meunier FA . Phosphoinositides in neuroexocytosis and neuronal diseases. In: Falasca M (ed) Phosphoinositides and Disease. Springer: London, UK, 2012 pp 87–98.

    Book  Google Scholar 

Download references

Acknowledgements

This investigation would not have been possible without the contribution of many Public Health Agents from the Family’s Health Program of several municipalities of Paraiba State, Brazil. This manuscript is dedicated to all families involved in this study. This study was financially supported by the UEPB/PROPESQ, FAPESQ/CNPq/PPSUS, CAPES, CNPq/INCT and FAPESP/CEPID.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to F Kok.

Ethics declarations

Competing interests

The authors declare no conflict of interest.

Additional information

Supplementary Information accompanies the paper on the Molecular Psychiatry website

Supplementary information

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Figueiredo, T., Melo, U., Pessoa, A. et al. A homozygous loss-of-function mutation in inositol monophosphatase 1 (IMPA1) causes severe intellectual disability. Mol Psychiatry 21, 1125–1129 (2016). https://doi.org/10.1038/mp.2015.150

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/mp.2015.150

This article is cited by

Search

Quick links