Complex clinical manifestations and new insights in RNA sequencing of children with diabetes and WFS1 variants

Background WFS1-related disorders involve a wide range of clinical phenotypes, including diabetes mellitus and neurodegeneration. Inheritance patterns of pathogenic variants of this gene can be autosomal recessive or dominant, and differences in penetrance present challenges for accurate diagnosis and genetic counselling. Methods Three probands and one elder brother from three families were systematically evaluated and the clinical data of other family members were collected from the medical history. Whole-exome sequencing was performed on the probands, and RNA sequencing was performed on four patients, their parents with WFS1 variants, and four gender- and age-matched children with type 1 diabetes mellitus. Results There were six patients with diabetes. Dilated cardiomyopathy, a rare manifestation of WFS1-related disease, was identified in one patient, along with MRI findings of brain atrophy at age 7 years and 3 months, the earliest age of discovery we know of. Whole-exome sequencing revealed five pathogenic or likely pathogenic variants in the WFS1 gene, including c.1348dupC (p.His450Profs*93), c.1381A>C (p.Thr461pro), c.1329C>G (p.Ser443Arg), c.2081delA (p.Glu694Glyfs*16), c.1350-1356delinsGCA (p.His450Glnfs*26), of which 3 variants (c.1348dupC, c.2081delA, c.1350-1356delinsGCA) were novel that have not been previously reported. The differentially expressed genes were mainly associated with immune-related pathways according to the Gene Ontology enrichment analysis of the RNA sequencing data. The exon 1 region of HLA-DRB1 in two patients was not transcribed, while the transcription of the region in their parents was normal. Conclusion This study emphasizes the clinical and genetic heterogeneity in patients, even in the same family with WFS1 variants. MRI evaluation of the brain should be considered when WFS1-related disorder is first diagnosed.

[1]  M. Rivas,et al.  A cross-population atlas of genetic associations for 220 human phenotypes , 2021, Nature Genetics.

[2]  Jian Wang,et al.  Clinical and genetic analysis in a Chinese cohort of children and adolescents with diabetes/persistent hyperglycemia , 2020, Journal of diabetes investigation.

[3]  G. Savini,et al.  Calcium mishandling in absence of primary mitochondrial dysfunction drives cellular pathology in Wolfram Syndrome , 2020, Scientific Reports.

[4]  Autosomal dominant , 2020, Definitions.

[5]  I. Krantz,et al.  Diagnosing Cornelia de Lange syndrome and related neurodevelopmental disorders using RNA sequencing , 2019, Genetics in Medicine.

[6]  Christopher D. Brown,et al.  The GTEx Consortium atlas of genetic regulatory effects across human tissues , 2019, Science.

[7]  J. Vengoechea,et al.  Segregation of two variants suggests the presence of autosomal dominant and recessive forms of WFS1-related disease within the same family: expanding the phenotypic spectrum of Wolfram Syndrome , 2019, Journal of Medical Genetics.

[8]  A. Rajab,et al.  Clinical and molecular assessment of 13 Iranian families with Wolfram syndrome , 2019, Endocrine.

[9]  G. d’Annunzio,et al.  Wolfram syndrome 1 in the Italian population: genotype–phenotype correlations , 2019, Pediatric Research.

[10]  Benjamin J. Strober,et al.  Identification of rare-disease genes using blood transcriptome sequencing and large control cohorts , 2019, Nature Medicine.

[11]  Jean M. Davidson,et al.  Identification of rare-disease genes using blood transcriptome sequencing and large control cohorts , 2019, Nature Medicine.

[12]  M. Atkinson,et al.  Clinical features, biochemistry and HLA-DRB1 status in youth-onset type 1 diabetes in Pakistan. , 2019, Diabetes research and clinical practice.

[13]  L. Wain,et al.  Moderate-to-severe asthma in individuals of European ancestry: a genome-wide association study , 2019, The Lancet. Respiratory medicine.

[14]  D. Swarbreck,et al.  Efficient and accurate detection of splice junctions from RNA-seq with Portcullis , 2017, bioRxiv.

[15]  Diego Garrido-Martín,et al.  ggsashimi: Sashimi plot revised for browser- and annotation-independent splicing visualization , 2018, PLoS Comput. Biol..

[16]  Cristina Has,et al.  Faculty of 1000 evaluation for Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. , 2018 .

[17]  T. Ariga,et al.  A novel heterozygous mutation of the WFS1 gene leading to constitutive endoplasmic reticulum stress is the cause of Wolfram syndrome , 2017, Pediatric diabetes.

[18]  V. Tillmann,et al.  Monogenic diabetes syndromes: Locus‐specific databases for Alström, Wolfram, and Thiamine‐responsive megaloblastic anemia , 2017, Human mutation.

[19]  P. Y. Man Monogenic diabetes syndromes: Locus-specific databases for Alström, Wolfram, and Thiamine-responsive megaloblastic anemia , 2017 .

[20]  K. Ogbonnaya,et al.  Pathophysiology, Diagnosis, and Management , 2017 .

[21]  Yiping Shen,et al.  A New Subtype of Multiple Synostoses Syndrome Is Caused by a Mutation in GDF6 That Decreases Its Sensitivity to Noggin and Enhances Its Potency as a BMP Signal , 2016, Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research.

[22]  S. Kõks,et al.  RNA‐sequencing of WFS1‐deficient pancreatic islets , 2016, Physiological reports.

[23]  F. Urano,et al.  Maternal uniparental disomy of chromosome 4 and homozygous novel mutation in the WFS1 gene in a paediatric patient with Wolfram syndrome. , 2015, Diabetes & metabolism.

[24]  田原 康玄,et al.  生活習慣病とgenome-wide association study , 2015 .

[25]  W. Huber,et al.  Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2 , 2014, Genome Biology.

[26]  Rachel L. Goldfeder,et al.  Autosomal Dominant Diabetes Arising From a Wolfram Syndrome 1 Mutation , 2013, Diabetes.

[27]  L. Schalkwyk,et al.  Silencing of the WFS1 gene in HEK cells induces pathways related to neurodegeneration and mitochondrial damage. , 2013, Physiological genomics.

[28]  P. Sham,et al.  Meta-analysis followed by replication identifies loci in or near CDKN1B, TET3, CD80, DRAM1, and ARID5B as associated with systemic lupus erythematosus in Asians. , 2013, American journal of human genetics.

[29]  M. Koenig,et al.  Mitochondrial cardiomyopathy: pathophysiology, diagnosis, and management. , 2013, Texas Heart Institute journal.

[30]  Thomas R. Gingeras,et al.  STAR: ultrafast universal RNA-seq aligner , 2013, Bioinform..

[31]  Guangchuang Yu,et al.  clusterProfiler: an R package for comparing biological themes among gene clusters. , 2012, Omics : a journal of integrative biology.

[32]  V. Tillmann,et al.  Hypothalamic gene expression profile indicates a reduction in G protein signaling in the Wfs1 mutant mice. , 2011, Physiological genomics.

[33]  B. Laway,et al.  Presentation and clinical course of Wolfram (DIDMOAD) syndrome from North India , 2011, Diabetic medicine : a journal of the British Diabetic Association.

[34]  Colin N. Dewey,et al.  RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome , 2011, BMC Bioinformatics.

[35]  G. Salzano,et al.  Diabetes and Neurodegeneration in Wolfram Syndrome , 2011, Diabetes Care.

[36]  V. Tillmann,et al.  Wfs1 gene deletion causes growth retardation in mice and interferes with the growth hormone pathway. , 2009, Physiological genomics.

[37]  B. Vialettes,et al.  Autosomal dominant transmission of diabetes and congenital hearing impairment secondary to a missense mutation in the WFS1 gene , 2008, Diabetic medicine : a journal of the British Diabetic Association.

[38]  G. Rutter,et al.  Sodium-potassium ATPase 1 subunit is a molecular partner of Wolframin, an endoplasmic reticulum protein involved in ER stress. , 2007, Human molecular genetics.

[39]  R. Sakakibara,et al.  Wolfram syndrome presenting marked brain MR imaging abnormalities with few neurologic abnormalities. , 2007, AJNR. American journal of neuroradiology.

[40]  A. Nava,et al.  Genotype-Phenotype Correlations , 2007 .

[41]  C. Schmitz,et al.  Wolfram syndrome 1 (WFS1) protein expression in retinal ganglion cells and optic nerve glia of the cynomolgus monkey. , 2006, Experimental eye research.

[42]  T. Hansen,et al.  Autosomal dominant optic atrophy associated with hearing impairment and impaired glucose regulation caused by a missense mutation in the WFS1 gene , 2005, Journal of Medical Genetics.

[43]  Yuil Kim,et al.  Two families with nonsyndromic low-frequency hearing loss harbor novel mutations in Wolfram syndrome gene 1 , 2005, Journal of Molecular Medicine.

[44]  N. Karabulut,et al.  Cranial magnetic resonance imaging of Wolfram (DIDMOAD) syndrome. , 2005, Australasian radiology.

[45]  K. Gerbitz,et al.  Wolfram syndrome: structural and functional analyses of mutant and wild-type wolframin, the WFS1 gene product. , 2003, Human molecular genetics.

[46]  M. King,et al.  Non-syndromic progressive hearing loss DFNA38 is caused by heterozygous missense mutation in the Wolfram syndrome gene WFS1. , 2001, Human molecular genetics.