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.

  • Letter
  • Published:

Human TGF-β1 deficiency causes severe inflammatory bowel disease and encephalopathy

Abstract

Transforming growth factor (TGF)-β1 (encoded by TGFB1) is the prototypic member of the TGF-β family of 33 proteins that orchestrate embryogenesis, development and tissue homeostasis1,2. Following its discovery3, enormous interest and numerous controversies have emerged about the role of TGF-β in coordinating the balance of pro- and anti-oncogenic properties4,5, pro- and anti-inflammatory effects6, or pro- and anti-fibrinogenic characteristics7. Here we describe three individuals from two pedigrees with biallelic loss-of-function mutations in the TGFB1 gene who presented with severe infantile inflammatory bowel disease (IBD) and central nervous system (CNS) disease associated with epilepsy, brain atrophy and posterior leukoencephalopathy. The proteins encoded by the mutated TGFB1 alleles were characterized by impaired secretion, function or stability of the TGF-β1–LAP complex, which is suggestive of perturbed bioavailability of TGF-β1. Our study shows that TGF-β1 has a critical and nonredundant role in the development and homeostasis of intestinal immunity and the CNS in humans.

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

Access options

Buy this article

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

Fig. 1: Identification of a biallelic TGFB1 mutation in patient 1 with very early-onset inflammatory bowel disease and global neurological defects.
Fig. 2: Effects of TGFB1 mutations on the biosynthesis and bioavailability of TGF-β1.

Similar content being viewed by others

References

  1. Blobe, G. C., Schiemann, W. P. & Lodish, H. F. Role of transforming growth factor-β in human disease. N. Engl. J. Med. 342, 1350–1358 (2000).

    Article  CAS  PubMed  Google Scholar 

  2. Wu, M. Y. & Hill, C. S. TGF-β superfamily signaling in embryonic development and homeostasis. Dev. Cell 16, 329–343 (2009).

    Article  CAS  PubMed  Google Scholar 

  3. Derynck, R. et al. Human transforming growth factor-β complementary DNA sequence and expression in normal and transformed cells. Nature 316, 701–705 (1985).

    Article  CAS  PubMed  Google Scholar 

  4. Principe, D. R. et al. TGF-β: duality of function between tumor prevention and carcinogenesis. J. Natl. Cancer Inst. 106, djt369 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Silberstein, G. B. & Daniel, C. W. Reversible inhibition of mammary gland growth by transforming growth factor-β. Science 237, 291–293 (1987).

    Article  CAS  PubMed  Google Scholar 

  6. Li, M. O., Wan, Y. Y., Sanjabi, S., Robertson, A. K. & Flavell, R. A. Transforming growth factor-β regulation of immune responses. Annu. Rev. Immunol. 24, 99–146 (2006).

    Article  CAS  PubMed  Google Scholar 

  7. Pohlers, D. et al. TGF-β and fibrosis in different organs—molecular pathway imprints. Biochim. Biophys. Acta 1792, 746–756 (2009).

    Article  CAS  PubMed  Google Scholar 

  8. Miyazono, K., Hellman, U., Wernstedt, C. & Heldin, C. H. Latent high-molecular-weight complex of transforming growth factor-β1. Purification from human platelets and structural characterization. J. Biol. Chem. 263, 6407–6415 (1988).

    CAS  PubMed  Google Scholar 

  9. Rifkin, D. B. Latent transforming growth factor-β (TGF-β)-binding proteins: orchestrators of TGF-β availability. J. Biol. Chem. 280, 7409–7412 (2005).

    Article  CAS  PubMed  Google Scholar 

  10. Li, M. O. & Flavell, R. A. TGF-β: a master of all T cell trades. Cell 134, 392–404 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Janssens, K. et al. Camurati–Engelmann disease: review of the clinical, radiological and molecular data of 24 families and implications for diagnosis and treatment. J. Med. Genet. 43, 1–11 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Loeys, B. L. et al. A syndrome of altered cardiovascular, craniofacial, neurocognitive and skeletal development caused by mutations in TGFBR1 or TGFBR2. Nat. Genet. 37, 275–281 (2005).

    Article  CAS  PubMed  Google Scholar 

  13. Shi, M. et al. Latent TGF-β structure and activation. Nature 474, 343–349 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Monteleone, G. et al. Blocking SMAD7 restores TGF-β1 signaling in chronic inflammatory bowel disease. J. Clin. Invest. 108, 601–609 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Glocker, E. O. et al. Inflammatory bowel disease and mutations affecting the interleukin-10 receptor. N. Engl. J. Med. 361, 2033–2045 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Kotlarz, D. et al. Loss of interleukin-10 signaling and infantile inflammatory bowel disease: implications for diagnosis and therapy. Gastroenterology 143, 347–355 (2012).

    Article  CAS  PubMed  Google Scholar 

  17. Shull, M. M. et al. Targeted disruption of the mouse transforming growth factor-β1 gene results in multifocal inflammatory disease. Nature 359, 693–699 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Li, M. O., Wan, Y. Y. & Flavell, R. A. T cell–produced transforming growth factor-β1 controls T cell tolerance and regulates TH1- and TH17 cell differentiation. Immunity 26, 579–591 (2007).

    Article  CAS  PubMed  Google Scholar 

  19. Gorelik, L. & Flavell, R. A. Abrogation of TGF-β signaling in T cells leads to spontaneous T cell differentiation and autoimmune disease. Immunity 12, 171–181 (2000).

    Article  CAS  PubMed  Google Scholar 

  20. Boirivant, M. et al. Inhibition of SMAD7 with a specific antisense oligonucleotide facilitates TGF-β1-mediated suppression of colitis. Gastroenterology 131, 1786–1798 (2006).

    Article  CAS  PubMed  Google Scholar 

  21. Monteleone, G. et al. Mongersen, an oral SMAD7 antisense oligonucleotide, and Crohn’s disease. N. Engl. J. Med. 372, 1104–1113 (2015).

    Article  CAS  PubMed  Google Scholar 

  22. Brionne, T. C., Tesseur, I., Masliah, E. & Wyss-Coray, T. Loss of TGF-β1 leads to increased neuronal cell death and microgliosis in mouse brain. Neuron 40, 1133–1145 (2003).

    Article  CAS  PubMed  Google Scholar 

  23. Koeglsperger, T. et al. Impaired glutamate recycling and GluN2B-mediated neuronal calcium overload in mice lacking TGF-β1 in the CNS. Glia 61, 985–1002 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  24. De Servi, B., La Porta, C. A., Bontempelli, M. & Comolli, R. Decrease of TGF-β1 plasma levels and increase of nitric oxide synthase activity in leukocytes as potential biomarkers of Alzheimer’s disease. Exp. Gerontol. 37, 813–821 (2002).

    Article  PubMed  Google Scholar 

  25. Tesseur, I. et al. Deficiency in neuronal TGF-β signaling promotes neurodegeneration and Alzheimer’s pathology. J. Clin. Invest. 116, 3060–3069 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Arosio, B. et al. + 10 T/C polymorphisms in the gene of transforming growth factor-β1 are associated with neurodegeneration and its clinical evolution. Mech. Ageing Dev. 128, 553–557 (2007).

    Article  CAS  PubMed  Google Scholar 

  27. Amir, E. D. et al. viSNE enables visualization of high dimensional single-cell data and reveals phenotypic heterogeneity of leukemia. Nat. Biotechnol. 31, 545–552 (2013).

  28. Li, H. & Durbin, R. Fast and accurate short-read alignment with Burrows–Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. McKenna, A. et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 20, 1297–1303 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Cingolani, P. et al. A program for annotating and predicting the effects of single-nucleotide polymorphisms, SnpEff: SNPs in the genome of Drosophila melanogaster strain w 1118; iso-2; iso-3. Fly 6, 80–92 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. McLaren, W. et al. Deriving the consequences of genomic variants with the Ensembl API and SNP Effect Predictor. Bioinformatics 26, 2069–2070 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Vigeland, M. D., Gjøtterud, K. S. & Selmer, K. K. FILTUS: a desktop GUI for fast and efficient detection of disease-causing variants, including a novel autozygosity detector. Bioinformatics 32, 1592–1594 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. 1000 Genomes Project Consortium. A global reference for human genetic variation. Nature 526, 68–74 (2015).

    Article  Google Scholar 

  34. Lek, M. et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature 536, 285–291 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Kumar, P., Henikoff, S. & Ng, P. C. Predicting the effects of coding nonsynonymous variants on protein function using the SIFT algorithm. Nat. Protoc. 4, 1073–1081 (2009).

    Article  CAS  PubMed  Google Scholar 

  36. Adzhubei, I., Jordan, D. M. & Sunyaev, S. R. Predicting functional effect of human missense mutations using PolyPhen-2. Curr. Protoc. Hum. Genet. 76, 7.20 (2013).

    Google Scholar 

  37. Kotlarz, D. et al. Loss-of-function mutations in the IL-21 receptor gene cause a primary immunodeficiency syndrome. J. Exp. Med. 210, 433–443 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Dennler, S. et al. Direct binding of SMAD3 and SMAD4 to critical TGF-β-inducible elements in the promoter of human plasminogen activator inhibitor type 1 gene. EMBO J. 17, 3091–3100 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Walton, K. L. et al. Two distinct regions of latency-associated peptide coordinate stability of the latent transforming growth factor-β1 complex. J. Biol. Chem. 285, 17029–17037 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are very grateful to our patients and their parents for allowing us to study their diseases. We thank the medical staff at the Dr. von Hauner Children’s Hospital, Oslo University Hospital and University Malaya Medical Center. In particular, we would like to acknowledge pathologist D. Klotz (Oslo University Hospital) for the histology of colonic biopsies. Whole-exome sequencing of family A was conducted at the Next-Generation Sequencing facility at the Dr. von Hauner Children’s Hospital under the supervision of M. Rohlfs. The sequencing service of family B was provided by the Norwegian Sequencing Centre, a national technology platform supported by the Functional Genomics and Infrastructure Programs of the Research Council of Norway and the Southeastern Regional Health Authorities, and the sequencing data of family B were analyzed by A. Holmgren. We acknowledge the assistance of the Flow Cytometry Core Facility at the Dr. von Hauner Children’s Hospital and of the Harvard Medical School CyTOF Core. Samples from the patient with CED were provided with support of the Oxford Gastrointestinal Illness Biobank and Biomedical Research Center Oxford. We gratefully acknowledge our bioinformatician, J. Puchalka, who died in a tragic accident during the course of the investigations. This work has been supported by The Leona M. and Harry B. Helmsley Charitable Trust, the Collaborative Research Consortium SFB1054 (DFG), PID-NET (BMBF), BioSysNet, the European Research Council, the Gottfried–Wilhelm–Leibniz Program (DFG), the DAAD network on ‘Rare Diseases and Personalized Therapies’, the German Center for Infection Research (DZIF) and the Care-for-Rare Foundation. W.S.L. was partly funded by University Malaya High Impact Research (UM.C/625/HIR/MOHE/CHAN/13/1). D.K. has been a scholar funded by the Else Kröner-Fresenius-Stiftung, the Daimler und Benz Stiftung and the Reinhard Frank-Stiftung.

Author information

Authors and Affiliations

Authors

Contributions

D.K. and C.K. designed and directed the study, managed recruitment of study participants, obtained clinical samples, supervised B.M. and interpreted the data; B.M. conducted and analyzed functional assays on heterologous cellular models; D.M., E.F. and P.S. supervised T.B. and E.M.S., initiated genetic analysis and drafted the clinical report of P2 and P3, and provided critical revision of the manuscript; T.B. acquired and interpreted genetic data from P2 and P3; R.C. conducted immunophenotypic analysis of PBMCs; T.M. and A.S.L. performed functional immunological assays; S.M.W. performed CyTOF analysis; L.K. supervised S.M.W. and analyzed the CyTOF results; S.H. performed the bioinformatics analysis of sequencing data; K.-P.H. conducted structural analysis of protein variants encoded by the identified TGFB1 mutations; W.S.L., I.B., F.H., P.B., E.M.S. and B.S.B. cared for the patients, collected patient samples and drafted clinical reports; C.W. examined histology; H.H.U. provided clinical information and a specimen from a patient with CED; A.M.M. and S.B.S. screened local cohorts of patients with very early-onset inflammatory bowel disease for mutations in TGFB1 and were instrumental in the interpretation of the human data; C.K. provided laboratory resources; and D.K. and C.K. wrote the manuscript with help from B.M. The manuscript was reviewed and approved by all co-authors.

Corresponding author

Correspondence to Christoph Klein.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Integrated supplementary information

Supplementary Figure 1 CD103 expression in lamina propria T cells from patient 1.

CyTOF analysis of CD103 expression in colonic lamina propria mononuclear cells derived from patients without IBD (controls: uninflamed; inflamed), a patient with Crohn’s disease and P1. The percentage of CD103-expressing cells is shown for CD3+CD4+ (top), CD3+CD8+ (middle) and CD127lo/–CD25+ (bottom) T cells.

Supplementary Figure 2 Sanger sequencing of patient 1.

Chromatograms of DNA Sanger sequencing identifying a compound heterozygous mutation in TGBF1 that segregates with the disease phenotype in P1.

Supplementary Figure 3 Clinical phenotype and mutational analysis of TGF-β1 deficiency in patients 2 and 3.

a, Pedigree of consanguineous Pakistani family B with two affected children. b, Sanger sequencing results confirming segregation of the identified biallelic TGFB1 missense mutation with the disease phenotype in pedigree B. c, Gastrointestinal findings in P3. Colonoscopy (top) revealed extensive colitis, and histology on colonic biopsies (bottom) showed chronic active inflammation accompanied by abscesses and crypt branching. d, Cerebral MRI images of P3 at the age of 2 years displaying gross cortical atrophy with widening of the subarachnoid spaces, delayed myelination and marked thinning of the corpus callosum.

Supplementary Figure 4 Normal STAT6 activity in lamina propria immune cells from patient 1.

CyTOF analysis of STAT6 phosphorylation (Tyr641; p-STAT6) in lamina propria mononuclear cells derived from patients without IBD (controls: uninflamed, blue; inflamed, orange), a patient with Crohn’s disease (CD) (green) and P1 (black). Histogram plots show baseline p-STAT6 in live cells that were gated on the indicated populations (left), and the heat map representation depicts the corresponding median expression values (MEV) for p-STAT6.

Supplementary Figure 5 Gating strategy for FACS analysis.

Gating strategy used in Fig. 1d.

Supplementary Figure 6 Gating strategy for CyTOF analysis.

a,b, Gating strategies used in Supplementary Fig. 1 (a) and in Fig. 2h and Supplementary Fig. 3 (b).

Supplementary Figure 7 Uncropped immunoblots.

Uncropped original immunoblots of Fig. 2d. The cropped areas are marked in red. Molecular weight markers are indicated in kDa.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 and Supplementary Tables 1–4

Life Sciences Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kotlarz, D., Marquardt, B., Barøy, T. et al. Human TGF-β1 deficiency causes severe inflammatory bowel disease and encephalopathy. Nat Genet 50, 344–348 (2018). https://doi.org/10.1038/s41588-018-0063-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-018-0063-6

This article is cited by

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