Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Hum Genet ; 108(11): 2171-2185, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34699745

ABSTRACT

Recent studies indicate that neurodegenerative processes that appear during childhood and adolescence in individuals with Wolfram syndrome (WS) occur in addition to early brain development alteration, which is clinically silent. Underlying pathological mechanisms are still unknown. We have used induced pluripotent stem cell-derived neural cells from individuals affected by WS in order to reveal their phenotypic and molecular correlates. We have observed that a subpopulation of Wolfram neurons displayed aberrant neurite outgrowth associated with altered expression of axon guidance genes. Selective inhibition of the ATF6α arm of the unfolded protein response prevented the altered phenotype, although acute endoplasmic reticulum stress response-which is activated in late Wolfram degenerative processes-was not detected. Among the drugs currently tried in individuals with WS, valproic acid was the one that prevented the pathological phenotypes. These results suggest that early defects in axon guidance may contribute to the loss of neurons in individuals with WS.


Subject(s)
Age of Onset , Induced Pluripotent Stem Cells/cytology , Neurites , Neurons/cytology , Wolfram Syndrome/pathology , CRISPR-Cas Systems , Case-Control Studies , Endoplasmic Reticulum Stress , Gene Expression Regulation , Humans , Neurites/drug effects , Valproic Acid/pharmacology , Wolfram Syndrome/genetics
2.
J Hum Genet ; 66(10): 1009-1018, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33879837

ABSTRACT

BACKGROUND: Wolfram syndrome (WFS) is characterized by deafness, diabetes mellitus, and diabetes insipidus along with optic atrophy. WFS has an autosomal recessive mode of inheritance and is due to variants in WFS1 and CISD2. METHODS: We evaluated the underlying molecular etiology of three affected members of a consanguineous family with hearing impairment, bicuspid aortic valve, diabetes mellitus and insipidus, clinodactyly, and gastrointestinal tract abnormalities via exome sequencing approach. We correlated clinical and imaging data with the genetic findings and their associated phenotypes. RESULTS: We identified a homozygous missense variant p.(Asn1097Lys) in CDK13, a gene previously associated with autosomal dominant congenital heart defects, dysmorphic facial features, clinodactyly, gastrointestinal tract abnormalities, intellectual developmental disorder, and seizures with variable phenotypic features. CONCLUSION: We report a homozygous variant in CDK13 and suggest that this gene causes an autosomal recessive disorder with hearing impairment, bicuspid aortic valve, diabetes mellitus and insipidus, clinodactyly, and gastrointestinal tract abnormalities.


Subject(s)
CDC2 Protein Kinase/genetics , Deafness/genetics , Genetic Predisposition to Disease , Optic Atrophy/genetics , Wolfram Syndrome/genetics , Adolescent , Adult , Bicuspid Aortic Valve Disease/genetics , Bicuspid Aortic Valve Disease/pathology , Child , Child, Preschool , Consanguinity , Deafness/complications , Deafness/pathology , Diabetes Mellitus/genetics , Female , Gastrointestinal Tract/abnormalities , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/pathology , Hearing Loss , Homozygote , Humans , Infant , Male , Mutation, Missense/genetics , Optic Atrophy/complications , Optic Atrophy/pathology , Wolfram Syndrome/complications , Wolfram Syndrome/epidemiology , Wolfram Syndrome/pathology , Young Adult
3.
Am J Med Genet A ; 185(2): 528-533, 2021 02.
Article in English | MEDLINE | ID: mdl-33179441

ABSTRACT

Wolfram syndrome was initially reported as an autosomal recessive (AR), progressive neurodegenerative disorder that leads to diabetes insipidus, childhood onset diabetes mellitus (DM), optic atrophy, and deafness (D) also known as DIDMOAD. However, heterozygous dominant pathogenic variants in Wolfram syndrome type 1 (WFS1) may lead to distinct, allelic conditions, described as isolated sensorineural hearing loss (SNHL), syndromic SNHL, congenital cataracts, or early onset DM. We report a family with a novel dominant, likely pathogenic variant in WFS1 (NM_006005.3) c.2605_2616del12 (p.Ser869_His872del), resulting in cataracts, SNHL, and DM in a female and her mother. A maternal aunt had cataracts, DM, and SNHL but was not tested for the familial WFS1 mutation. Both the mother and maternal aunt had early menopause by age 43 years and infertility which may be a coincidental finding that has not been associated with autosomal dominant AD WFS1-related disorder to the best of our knowledge. Screening at risk individuals in families with the AR Wolfram syndrome, for DM, SNHL, and for cataracts is indicated.


Subject(s)
Diabetes Mellitus/genetics , Hearing Loss, Sensorineural/genetics , Membrane Proteins/genetics , Wolfram Syndrome/genetics , Adult , Age of Onset , Cataract/complications , Cataract/genetics , Cataract/pathology , Diabetes Mellitus/pathology , Female , Genes, Recessive/genetics , Hearing Loss, Sensorineural/complications , Hearing Loss, Sensorineural/pathology , Humans , Mutation/genetics , Risk Factors , Wolfram Syndrome/complications , Wolfram Syndrome/pathology
4.
BMC Endocr Disord ; 21(1): 166, 2021 Aug 17.
Article in English | MEDLINE | ID: mdl-34404380

ABSTRACT

BACKGROUND: Wolfram syndrome (WFS) is a rare autosomal recessive genetic disease whose main cause is mutations in the WFS1 and CISD2 genes. Its characteristic clinical manifestations are diabetes insipidus, diabetes mellitus, optic atrophy and deafness. METHODS: In this study, two patients from this particular family underwent complete routine biochemical and ophthalmic tests. Blood, urine, routine stool test, visual acuity (VA) examination, visual field assessment, funduscope, optical coherence tomography and periorbital magnetic resonance imaging (MRI) scans were performed for each patient to evaluate whether the nerve fiber layer around the optic nerve head was atrophied and next-generation sequencing of target genes was performed in two patients. RESULTS: When the patients were diagnosed with Wolfram syndrome, their genetic analyses suggested unique three-site compound heterozygous mutations (c.2314C > T + c.2194C > T + c.2171C > T) in exon 8 of both patients' chromosome 4. One mutation (c.2314C > T) was a novel mutation in the known reports of Wolfram syndrome. As a degenerative genetic disease, the types of gene mutations in the Chinese population are generally homozygous mutations at the unit point or compound heterozygous mutations at two nucleotide change sites. However, the two patients reported in this study are the first known cases of compound heterozygous mutations with three mutation sites coexisting on the WFS1 gene in China or even globally. CONCLUSIONS: This study expands the phenotypic spectrum of Wolfram syndrome and may reveal a novel mutation pattern of pathogenesis of Wolfram syndrome. The implications of this discovery are valuable in the clinical diagnosis, prognosis, and treatment of patients with WFS1.


Subject(s)
Membrane Proteins/genetics , Mutation , Wolfram Syndrome/genetics , Wolfram Syndrome/pathology , Adolescent , Exons , Female , Homozygote , Humans , Male , Pedigree , Prognosis
5.
BMC Endocr Disord ; 21(1): 76, 2021 Apr 21.
Article in English | MEDLINE | ID: mdl-33879153

ABSTRACT

BACKGROUND: Wolfram syndrome (WS) is a rare autosomal recessive disorder characterized by diabetes insipidus, diabetes mellitus, optic atrophy and deafness. Mutations in Wolfram syndrome 1 (WFS1) gene may cause dysregulated endoplasmic reticulum (ER)-stress and cell apoptosis, contributing to WS symptoms. The aim of this study was to identify the molecular etiology of a case of WS and to explore the functional consequence of the mutant WFS1 gene in vitro. METHODS: A 27 years-old Chinese man was diagnosed as wolfram syndrome type 1 based on clinical data and laboratory data. DNA sequencing of WFS1 gene and mitochondrial m.3337G > A, m.3243A > G mutations were performed in the patient and his 4 family members. Functional analysis was performed to assessed the in vitro effect of the newly identified mutant. ER stress were evaluated by ER stress response element (ERSE)-luciferase assay. Cell apoptosis were performed by CCK-8, TUNEL staining and flow cytometric analysis. RESULTS: A novel heterozygous 10-base deletion (c. 2067_2076 del10, p.W690fsX706) was identified in the patient. In vitro studies showed that mutant p.W690fsX706 increased ERSE reporter activity in the presence or absence of thapsigargin instead of wild type WFS1. Knockdown of WFS1 activated the unfolded protein response (UPR) pathway and increased the cell apoptosis, which could not be restored by transfection with WFS1 mutant (p.W690fsX706) comparable to the wild type WFS1. CONCLUSIONS: A novel heterozygous mutation of WFS1 detected in the patient resulted in loss-of-function of wolframin, thereby inducing dysregulated ER stress signaling and cell apoptosis. These findings increase the spectrum of WFS1 gene mutations and broaden our insights into the roles of mutant WFS1 in the pathogenesis of WS.


Subject(s)
Apoptosis/genetics , Endoplasmic Reticulum Stress/genetics , Membrane Proteins/genetics , Wolfram Syndrome , Adult , China , Genes, Dominant , Heterozygote , Humans , Male , Mutation , Polymorphism, Single Nucleotide , Wolfram Syndrome/genetics , Wolfram Syndrome/metabolism , Wolfram Syndrome/pathology
6.
J Med Genet ; 57(2): 121-123, 2020 02.
Article in English | MEDLINE | ID: mdl-31363008

ABSTRACT

BACKGROUND: WFS1 was initially described as causative agent of autosomal recessive (AR) Wolfram syndrome, a childhood-onset disorder involving diabetes, optic atrophy, hearing loss and neurodegenerative features. However, the discovery of autosomal dominant (AD) disorders caused by this gene has resulted in clinical counselling and result interpretation challenges. OBJECTIVE: We seek to report a family that appears to segregate dominant and recessive forms of WFS1-related disease. METHODS/RESULTS: A 19-year-old woman presented with progressive childhood sensorineural hearing loss and recent optic atrophy, with biallelic mutations in WFS1: c.2486T>C (likely pathogenic) and c.2470G>A (uncertain significance). Her A1C was normal. Her sister carried the same variants and had a similar phenotype. Their father carried c.2486T>C and was found to have mild-moderate hearing loss but no optic atrophy or neurological symptoms. The mother carried c.2470G>A and had a normal audiogram and ophthalmological exam. Providing anticipatory guidance for this family was difficult given the phenotypic variability of WFS1-related disorders and the uncertainty surrounding whether the inheritance pattern was AR or AD. CONCLUSION: The clinical correlation of the variants identified in this family suggests an AR Wolfram-like syndrome, without the typical diabetes mellitus or diabetes insipidus nor neurological decline. To our knowledge, this is a novel WFS1-related phenotype.


Subject(s)
Hearing Loss, Sensorineural , Membrane Proteins , Optic Atrophy , Wolfram Syndrome , Adolescent , Adult , Aged , Child , Female , Humans , Male , Middle Aged , Young Adult , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/pathology , Membrane Proteins/genetics , Mutation/genetics , Optic Atrophy/genetics , Optic Atrophy/pathology , Pedigree , Phenotype , Wolfram Syndrome/genetics , Wolfram Syndrome/pathology
7.
BMC Med Genet ; 21(1): 13, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31937257

ABSTRACT

BACKGROUND: Mutations of the WFS1 gene are responsible for most cases of Wolfram syndrome (WS), a rare, recessively inherited neurodegenerative disorder characterized by juvenile-onset non-autoimmune diabetes mellitus and optic atrophy. Variants of WFS1 are also associated with non-syndromic hearing loss and type-2 diabetes mellitus (T2DM). Our study adds to literature significant associations between WS and T2DM. CASE PRESENTATION: In this study, we analyzed the clinical and genetic data of two families with high prevalence of WS and T2DM. Genetic linkage analysis and DNA sequencing were exploited to identify pathogenic variants. One novel pathogenic variant (c.2243-2244insC) and one known pathogenic (c.1232_1233delCT) (frameshift) variant were identified in exon eight of WFS1 gene. CONCLUSIONS: The mutational and phenotypic spectrum of WS is broadened by our report of novel WFS1 mutation. Our results reveal the value of molecular analysis of WFS1 in the improvement of clinical diagnostics for WS. This study also confirms the role of WFS1 in T2DM.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Genetic Testing , Membrane Proteins/genetics , Wolfram Syndrome/genetics , Adult , Child , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/pathology , Exons/genetics , Female , Frameshift Mutation/genetics , Genetic Linkage , Genetic Predisposition to Disease , Hearing Loss/complications , Hearing Loss/genetics , Hearing Loss/pathology , Humans , Iran/epidemiology , Male , Optic Atrophy/complications , Optic Atrophy/genetics , Optic Atrophy/pathology , Pedigree , Phenotype , Point Mutation/genetics , Wolfram Syndrome/complications , Wolfram Syndrome/pathology , Young Adult
8.
J Urol ; 200(5): 1107-1113, 2018 11.
Article in English | MEDLINE | ID: mdl-29883657

ABSTRACT

PURPOSE: Wolfram syndrome is a neurodegenerative disorder characterized by childhood onset diabetes mellitus, optic nerve atrophy, diabetes insipidus, hearing impairment, and commonly bladder and bowel dysfunction. We hypothesized that there is an association between a smaller pons, which contains the pontine micturition center, and abnormal lower urinary tract function. MATERIALS AND METHODS: Patients with genetically confirmed Wolfram syndrome attended an annual multidisciplinary research clinic. Subjects underwent noninvasive urodynamic testing and brain magnetic resonance imaging, and completed validated patient reported outcome measures. Bowel and bladder diaries were completed before visits. Age and gender corrected linear and logistic mixed effects models were used to correlate pons volume, corrected for whole brain size, to urodynamic and patient reported outcomes. RESULTS: A total of 36 patients attended 142 visits between 2010 and 2016. Mean age was 16.9 years (range 7 to 30) and 64% of patients were female. Functional bladder capacity was decreased in 31% of the patients, normal in 54% and increased in 14%. Of the patients 44% and 54% had abnormal uroflowmetry and post-void residual, respectively, on at least 1 occasion. There was no increase through time in incidence of lower urinary tract dysfunction. Decreased pons volume was associated with increased post-void residual (p = 0.048) and higher PinQ (Pediatric Incontinence Questionnaire) score (p = 0.011), indicating lower quality of life and higher levels of dysfunction. CONCLUSIONS: A significant number of children, adolescents and young adults with Wolfram syndrome have objective evidence of lower urinary tract dysfunction. Decreased pons volume is associated with more abnormal urinary function and lower quality of life in patients with Wolfram syndrome.


Subject(s)
Lower Urinary Tract Symptoms/etiology , Pons/pathology , Urinary Bladder/physiopathology , Urination/physiology , Wolfram Syndrome/complications , Adolescent , Adult , Age Factors , Child , Female , Humans , Image Processing, Computer-Assisted , Incidence , Longitudinal Studies , Lower Urinary Tract Symptoms/diagnosis , Lower Urinary Tract Symptoms/epidemiology , Lower Urinary Tract Symptoms/physiopathology , Magnetic Resonance Imaging , Male , Organ Size , Patient Reported Outcome Measures , Pons/diagnostic imaging , Pons/physiopathology , Quality of Life , Self Report/statistics & numerical data , Sex Factors , Urinary Bladder/innervation , Urodynamics/physiology , Wolfram Syndrome/diagnostic imaging , Wolfram Syndrome/pathology , Wolfram Syndrome/physiopathology , Young Adult
10.
Mol Biol Rep ; 41(11): 7499-505, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25173644

ABSTRACT

Wolfram syndrome also known as DIDMOAD (Diabetes Insipidus, Diabetes Mellitus, Optic Atrophy, and Deafness) is a rare neurodegenerative autosomal recessive disorder. There is evidence of variable expressivity both in patients and heterozygous carriers. In this study, we describe three Persian Wolfram syndrome families with differences in the age of onset, signs and symptoms of the disease. We clinically evaluated affected families for verifying WS clinical diagnosis. After linkage analysis via 5 STR markers, molecular analysis for WFS1 was performed by direct sequencing for patients and available family members. Three homozygous mutations were identified including c.1885 C>T, c.2205C>A both in exon 8 and c.460+1G>A in intron 4. The mutation c.2205C>A was found to be novel. We report interesting phenotype-genotype correlations: homozygous c.1885C>T and c.2205C>A variants were correlated with quite different disease severity and onset in the siblings. We report a rare case of WS with homozygous c.1885C>T who is married and has a healthy child. c.460+1G>A showed a possible partial dominant inheritance put forth by a heterozygous parent showing partial WS symptoms while her daughter displayed typical WS symptoms. Due to variable expressivity, detailed clinical examination and molecular diagnostics should be used to confirm WS and a more exact recurrence risk data.


Subject(s)
Membrane Proteins/genetics , Phenotype , Point Mutation/genetics , Wolfram Syndrome/genetics , Adult , Age of Onset , Base Sequence , Child , Female , Genes, Recessive , Genetic Linkage , Humans , Inheritance Patterns/genetics , Iran , Male , Molecular Sequence Data , Pedigree , Sequence Analysis, DNA , Wolfram Syndrome/pathology
11.
Cell Death Differ ; 31(3): 322-334, 2024 03.
Article in English | MEDLINE | ID: mdl-38321214

ABSTRACT

Pancreatic ß-cell failure by WFS1 deficiency is manifested in individuals with wolfram syndrome (WS). The lack of a suitable human model in WS has impeded progress in the development of new treatments. Here, human pluripotent stem cell derived pancreatic islets (SC-islets) harboring WFS1 deficiency and mouse model of ß cell specific Wfs1 knockout were applied to model ß-cell failure in WS. We charted a high-resolution roadmap with single-cell RNA-seq (scRNA-seq) to investigate pathogenesis for WS ß-cell failure, revealing two distinct cellular fates along pseudotime trajectory: maturation and stress branches. WFS1 deficiency disrupted ß-cell fate trajectory toward maturation and directed it towards stress trajectory, ultimately leading to ß-cell failure. Notably, further investigation of the stress trajectory identified activated integrated stress response (ISR) as a crucial mechanism underlying WS ß-cell failure, characterized by aberrant eIF2 signaling in WFS1-deficient SC-islets, along with elevated expression of genes in regulating stress granule formation. Significantly, we demonstrated that ISRIB, an ISR inhibitor, efficiently reversed ß-cell failure in WFS1-deficient SC-islets. We further validated therapeutic efficacy in vivo with ß-cell specific Wfs1 knockout mice. Altogether, our study provides novel insights into WS pathogenesis and offers a strategy targeting ISR to treat WS diabetes.


Subject(s)
Insulin-Secreting Cells , Wolfram Syndrome , Mice , Animals , Humans , Wolfram Syndrome/genetics , Wolfram Syndrome/metabolism , Wolfram Syndrome/pathology , Insulin-Secreting Cells/metabolism , Mice, Knockout
12.
Acta Neuropathol Commun ; 12(1): 140, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39198924

ABSTRACT

Wolfram syndrome (WS) is a rare childhood disease characterized by diabetes mellitus, diabetes insipidus, blindness, deafness, neurodegeneration and eventually early death, due to autosomal recessive mutations in the WFS1 (and WFS2) gene. While it is categorized as a neurodegenerative disease, it is increasingly becoming clear that other cell types besides neurons may be affected and contribute to the pathogenesis. MRI studies in patients and phenotyping studies in WS rodent models indicate white matter/myelin loss, implicating a role for oligodendroglia in WS-associated neurodegeneration. In this study, we sought to determine if oligodendroglia are affected in WS and whether their dysfunction may be the primary cause of the observed optic neuropathy and brain neurodegeneration. We demonstrate that 7.5-month-old Wfs1∆exon8 mice display signs of abnormal myelination and a reduced number of oligodendrocyte precursor cells (OPCs) as well as abnormal axonal conduction in the optic nerve. An MRI study of the brain furthermore revealed grey and white matter loss in the cerebellum, brainstem, and superior colliculus, as is seen in WS patients. To further dissect the role of oligodendroglia in WS, we performed a transcriptomics study of WS patient iPSC-derived OPCs and pre-myelinating oligodendrocytes. Transcriptional changes compared to isogenic control cells were found for genes with a role in ER function. However, a deep phenotyping study of these WS patient iPSC-derived oligodendroglia unveiled normal differentiation, mitochondria-associated endoplasmic reticulum (ER) membrane interactions and mitochondrial function, and no overt signs of ER stress. Overall, the current study indicates that oligodendroglia functions are largely preserved in the WS mouse and patient iPSC-derived models used in this study. These findings do not support a major defect in oligodendroglia function as the primary cause of WS, and warrant further investigation of neurons and neuron-oligodendroglia interactions as a target for future neuroprotective or -restorative treatments for WS.


Subject(s)
Induced Pluripotent Stem Cells , Oligodendroglia , Phenotype , Wolfram Syndrome , Animals , Induced Pluripotent Stem Cells/pathology , Wolfram Syndrome/pathology , Wolfram Syndrome/genetics , Oligodendroglia/pathology , Mice , Humans , Disease Models, Animal , Membrane Proteins/genetics , Membrane Proteins/metabolism , Male , Optic Nerve/pathology , Mice, Inbred C57BL , Female
13.
Autophagy ; 20(9): 2055-2066, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38651637

ABSTRACT

Dominant variants in WFS1 (wolframin ER transmembrane glycoprotein), the gene coding for a mitochondria-associated endoplasmic reticulum (ER) membrane (MAM) resident protein, have been associated with Wolfram-like syndrome (WLS). In vitro and in vivo, WFS1 loss results in reduced ER to mitochondria calcium (Ca2+) transfer, mitochondrial dysfunction, and enhanced macroautophagy/autophagy and mitophagy. However, in the WLS pathological context, whether the mutant protein triggers the same cellular processes is unknown. Here, we show that in human fibroblasts and murine neuronal cultures the WLS protein WFS1E864K leads to decreases in mitochondria bioenergetics and Ca2+ uptake, deregulation of the mitochondrial quality system mechanisms, and alteration of the autophagic flux. Moreover, in the Wfs1E864K mouse, these alterations are concomitant with a decrease of MAM number. These findings reveal pathophysiological similarities between WS and WLS, highlighting the importance of WFS1 for MAM's integrity and functionality. It may open new treatment perspectives for patients with WLS.Abbreviations: BafA1: bafilomycin A1; ER: endoplasmic reticulum; HSPA9/GRP75: heat shock protein family A (Hsp70) member 9; ITPR/IP3R: inositol 1,4,5-trisphosphate receptor; MAM: mitochondria-associated endoplasmic reticulum membrane; MCU: mitochondrial calcium uniporter; MFN2: mitofusin 2; OCR: oxygen consumption rate; ROS: reactive oxygen species; ROT/AA: rotenone+antimycin A; VDAC1: voltage dependent anion channel 1; WLS: Wolfram-like syndrome; WS: Wolfram syndrome; WT: wild-type.


Subject(s)
Autophagy , Calcium , Endoplasmic Reticulum , Membrane Proteins , Mitochondria , Mitophagy , Animals , Mitophagy/physiology , Humans , Membrane Proteins/metabolism , Endoplasmic Reticulum/metabolism , Autophagy/physiology , Mice , Mitochondria/metabolism , Calcium/metabolism , Wolfram Syndrome/metabolism , Wolfram Syndrome/pathology , Fibroblasts/metabolism , Neurons/metabolism
14.
Cell Metab ; 7(3): 269-76, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18316032

ABSTRACT

Endoplasmic reticulum (ER) stress-mediated apoptosis may play a crucial role in loss of pancreatic beta cell mass, contributing to the development of diabetes. Here we show that induction of 4E-BP1, the suppressor of the mRNA 5' cap-binding protein eukaryotic initiation factor 4E (eIF4E), is involved in beta cell survival under ER stress. 4E-BP1 expression was increased in islets under ER stress in several mouse models of diabetes. The Eif4ebp1 gene encoding 4E-BP1 was revealed to be a direct target of the transcription factor ATF4. Deletion of the Eif4ebp1 gene increased susceptibility to ER stress-mediated apoptosis in MIN6 beta cells and mouse islets, which was accompanied by deregulated translational control. Furthermore, Eif4ebp1 deletion accelerated beta cell loss and exacerbated hyperglycemia in mouse models of diabetes. Thus, 4E-BP1 induction contributes to the maintenance of beta cell homeostasis during ER stress and is a potential therapeutic target for diabetes.


Subject(s)
Activating Transcription Factor 4/metabolism , Apoptosis , Carrier Proteins/metabolism , Diabetes Mellitus/metabolism , Endoplasmic Reticulum/metabolism , Insulin-Secreting Cells/metabolism , Phosphoproteins/metabolism , Stress, Physiological/metabolism , Transcriptional Activation , Activating Transcription Factor 4/genetics , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/genetics , Cell Cycle Proteins , Cell Line, Tumor , Cell Survival , Diabetes Mellitus/genetics , Diabetes Mellitus/pathology , Disease Models, Animal , Endoplasmic Reticulum/pathology , Eukaryotic Initiation Factors , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Homeostasis , Insulin Resistance/genetics , Insulin-Secreting Cells/pathology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Phosphoproteins/genetics , Protein Folding , Stress, Physiological/pathology , Time Factors , Transduction, Genetic , Up-Regulation , Wolfram Syndrome/genetics , Wolfram Syndrome/metabolism , Wolfram Syndrome/pathology
15.
Hum Mol Genet ; 20(7): 1274-84, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21199859

ABSTRACT

Wolfram syndrome is an autosomal recessive disorder characterized by juvenile-onset insulin-dependent diabetes mellitus and optic atrophy. The gene responsible for the syndrome (WFS1) encodes an endoplasmic reticulum (ER) resident transmembrane protein. The Wfs1-null mouse exhibits progressive insulin deficiency causing diabetes. Previous work suggested that the function of the WFS1 protein is connected to unfolded protein response and to intracellular Ca(2+) homeostasis. However, its precise molecular function in pancreatic ß-cells remains elusive. In our present study, immunofluorescent and electron-microscopic analyses revealed that WFS1 localizes not only to ER but also to secretory granules in pancreatic ß-cells. Intragranular acidification was assessed by measuring intracellular fluorescence intensity raised by the acidotrophic agent, 3-[2,4-dinitroanilino]-3'-amino-N-methyldipropyramine. Compared with wild-type ß-cells, there was a 32% reduction in the intensity in WFS1-deficient ß-cells, indicating the impairment of granular acidification. This phenotype may, at least partly, account for the evidence that Wfs1-null islets have impaired proinsulin processing, resulting in an increased circulating proinsulin level. Morphometric analysis using electron microscopy evidenced that the density of secretory granules attached to the plasma membrane was significantly reduced in Wfs1-null ß-cells relative to that in wild-type ß-cells. This may be relevant to the recent finding that granular acidification is required for the priming of secretory granules preceding exocytosis and may partly explain the fact that glucose-induced insulin secretion is profoundly impaired in young prediabetic Wfs1-null mice. These results thus provide new insights into the molecular mechanisms of ß-cell dysfunction in patients with Wolfram syndrome.


Subject(s)
Endoplasmic Reticulum/metabolism , Exocytosis/physiology , Insulin-Secreting Cells/metabolism , Membrane Proteins/immunology , Proinsulin/metabolism , Secretory Vesicles/metabolism , Animals , Calcium/metabolism , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/ultrastructure , Exocytosis/drug effects , Glucose/pharmacology , Homeostasis/drug effects , Homeostasis/physiology , Humans , Insulin-Secreting Cells/ultrastructure , Membrane Proteins/genetics , Mice , Mice, Knockout , Proinsulin/genetics , Secretory Vesicles/genetics , Secretory Vesicles/ultrastructure , Sweetening Agents/pharmacology , Wolfram Syndrome/genetics , Wolfram Syndrome/metabolism , Wolfram Syndrome/pathology
16.
Sci Rep ; 13(1): 9127, 2023 06 05.
Article in English | MEDLINE | ID: mdl-37277527

ABSTRACT

Diabetes is one of the most common phenotypes of Wolfram syndrome owing to the presence of the variants of the WFS1 gene and is often misdiagnosed as other types of diabetes. We aimed to explore the prevalence of WFS1-related diabetes (WFS1-DM) and its clinical characteristics in a Chinese population with early-onset type 2 diabetes (EOD). We sequenced all exons of the WFS1 gene in 690 patients with EOD (age at diagnosis ≤ 40 years) for rare variants. Pathogenicity was defined according to the standards and guidelines of the American College of Medical Genetics and Genomics. We identified 33 rare variants predicted to be deleterious in 39 patients. The fasting [1.57(1.06-2.22) ng/ml] and postprandial C-peptide levels [2.8(1.75-4.46) ng/ml] of the patients with such WFS1 variations were lower than those of the patients without WFS1 variation [2.09(1.43-3.05) and 4.29(2.76-6.07) respectively, ng/ml]. Six (0.9%) patients carried pathogenic or likely pathogenic variants; they met the diagnostic criteria for WFS1-DM according to the latest guidelines, but typical phenotypes of Wolfram syndrome were seldom observed. They were diagnosed at an earlier age and usually presented with an absence of obesity, impaired beta cell function, and the need for insulin treatment. WFS1-DM is usually mistakenly diagnosed as type 2 diabetes, and genetic testing is helpful for individualized treatment.


Subject(s)
Diabetes Mellitus, Type 2 , Wolfram Syndrome , Humans , Diabetes Mellitus, Type 2/genetics , East Asian People , Genetic Testing , Phenotype , Wolfram Syndrome/diagnosis , Wolfram Syndrome/genetics , Wolfram Syndrome/pathology , Adult
17.
Stem Cell Res ; 69: 103068, 2023 06.
Article in English | MEDLINE | ID: mdl-36933359

ABSTRACT

Pathogenic variants of the WFS1 gene can cause recessive-inherited Wolfram syndrome or dominant-inherited Wolfram-like syndrome with optic atrophy and hearing impairment. Using the Sendai virus delivery system, we generated induced pluripotent stem cells from the peripheral blood mononuclear cells of a female patient with the WFS1 pathogenic variant c.2051C > T (p.Ala684Val). The resulting induced pluripotent stem cells exhibited a normal karyotype and pluripotency, as confirmed using immunofluorescence staining, and differentiated into three germ layers in vivo. This cellular model provides a useful platform for investigating the pathogenic mechanisms of both blindness and deafness related to WFS1 variants.


Subject(s)
Hearing Loss , Induced Pluripotent Stem Cells , Wolfram Syndrome , Humans , Female , Induced Pluripotent Stem Cells/pathology , Leukocytes, Mononuclear/pathology , Hearing Loss/genetics , Wolfram Syndrome/genetics , Wolfram Syndrome/pathology , Mutation
18.
BMC Med Genomics ; 16(1): 79, 2023 04 11.
Article in English | MEDLINE | ID: mdl-37041640

ABSTRACT

BACKGROUND: Wolfram syndrome type 1 gene (WFS1), which encodes a transmembrane structural protein (wolframin), is essential for several biological processes, including proper inner ear function. Unlike the recessively inherited Wolfram syndrome, WFS1 heterozygous variants cause DFNA6/14/38 and wolfram-like syndrome, characterized by autosomal dominant nonsyndromic hearing loss, optic atrophy, and diabetes mellitus. Here, we identified two WFS1 heterozygous variants in three DFNA6/14/38 families using exome sequencing. We reveal the pathogenicity of the WFS1 variants based on three-dimensional (3D) modeling and structural analysis. Furthermore, we present cochlear implantation (CI) outcomes in WFS1-associated DFNA6/14/38 and suggest a genotype-phenotype correlation based on our results and a systematic review. METHODS: We performed molecular genetic test and evaluated clinical phenotypes of three WFS1-associated DFNA6/14/38 families. A putative WFS1-NCS1 interaction model was generated, and the impacts of WFS1 variants on stability were predicted by comparing intramolecular interactions. A total of 62 WFS1 variants associated with DFNA6/14/38 were included in a systematic review. RESULTS: One variant is a known mutational hotspot variant in the endoplasmic reticulum (ER)-luminal domain WFS1(NM_006005.3) (c.2051 C > T:p.Ala684Val), and the other is a novel frameshift variant in transmembrane domain 6 (c.1544_1545insA:p.Phe515LeufsTer28). The two variants were pathogenic, based on the ACMG/AMP guidelines. Three-dimensional modeling and structural analysis show that non-polar, hydrophobic substitution of Ala684 (p.Ala684Val) destabilizes the alpha helix and contributes to the loss of WFS1-NCS1 interaction. Also, the p.Phe515LeufsTer28 variant truncates transmembrane domain 7-9 and the ER-luminal domain, possibly impairing membrane localization and C-terminal signal transduction. The systematic review demonstrates favorable outcomes of CI. Remarkably, p.Ala684Val in WFS1 is associated with early-onset severe-to-profound deafness, revealing a strong candidate variant for CI. CONCLUSIONS: We expanded the genotypic spectrum of WFS1 heterozygous variants underlying DFNA6/14/38 and revealed the pathogenicity of mutant WFS1, providing a theoretical basis for WFS1-NCS1 interactions. We presented a range of phenotypic traits for WFS1 heterozygous variants and demonstrated favorable functional CI outcomes, proposing p.Ala684Val a strong potential marker for CI candidates.


Subject(s)
Cochlear Implantation , Cochlear Implants , Deafness , Hearing Loss , Wolfram Syndrome , Humans , Wolfram Syndrome/complications , Wolfram Syndrome/genetics , Wolfram Syndrome/pathology , Pedigree , Hearing Loss/genetics
19.
JCI Insight ; 7(18)2022 09 22.
Article in English | MEDLINE | ID: mdl-36134655

ABSTRACT

Wolfram syndrome is a rare genetic disorder largely caused by pathogenic variants in the WFS1 gene and manifested by diabetes mellitus, optic nerve atrophy, and progressive neurodegeneration. Recent genetic and clinical findings have revealed Wolfram syndrome as a spectrum disorder. Therefore, a genotype-phenotype correlation analysis is needed for diagnosis and therapeutic development. Here, we focus on the WFS1 c.1672C>T, p.R558C variant, which is highly prevalent in the Ashkenazi Jewish population. Clinical investigation indicated that patients carrying the homozygous WFS1 c.1672C>T, p.R558C variant showed mild forms of Wolfram syndrome phenotypes. Expression of WFS1 p.R558C was more stable compared with the other known recessive pathogenic variants associated with Wolfram syndrome. Human induced pluripotent stem cell-derived (iPSC-derived) islets (SC-islets) homozygous for WFS1 c.1672C>T variant recapitulated genotype-related Wolfram syndrome phenotypes. Enhancing residual WFS1 function through a combination treatment of chemical chaperones mitigated detrimental effects caused by the WFS1 c.1672C>T, p.R558C variant and increased insulin secretion in SC-islets. Thus, the WFS1 c.1672C>T, p.R558C variant causes a mild form of Wolfram syndrome phenotypes, which can be remitted with a combination treatment of chemical chaperones. We demonstrate that our patient iPSC-derived disease model provides a valuable platform for further genotype-phenotype analysis and therapeutic development for Wolfram syndrome.


Subject(s)
Induced Pluripotent Stem Cells , Optic Atrophy , Wolfram Syndrome , Homozygote , Humans , Induced Pluripotent Stem Cells/metabolism , Membrane Proteins/genetics , Optic Atrophy/genetics , Optic Atrophy/pathology , Wolfram Syndrome/diagnosis , Wolfram Syndrome/genetics , Wolfram Syndrome/pathology
20.
Invest Ophthalmol Vis Sci ; 63(10): 9, 2022 09 01.
Article in English | MEDLINE | ID: mdl-36098976

ABSTRACT

Purpose: To identify the missing heritability of patients with Wolfram syndrome 1 (WFS1) in a Chinese cohort and to report their clinical and genetic features. Methods: We recruited 24 unrelated patients with suspected WFS1 who carried at least one variant in WFS1. All patients underwent ophthalmic examinations and comprehensive molecular genetic analyses, including Sanger-DNA sequencing of WFS1 and next-generation sequencing of the whole WFS1 sequence. Results: We identified 38 distinct pathogenic variants of WFS1 in the 24 probands, comprising 23 patients with biallelic variants and one patient with a monoallelic variant. Sanger-DNA sequencing of WFS1 initially detected 35 variants, and subsequent whole genome sequencing revealed three missing variants: one novel deep intronic variant (DIV), one copy number variant (CNV), and one variant in the promoter region. Minigene assays showed that the DIV activated cryptic splice sites, leading to the insertion of pseudoexons. Optic atrophy was observed in all patients, and diabetes mellitus (DM) was revealed in 21 patients (91.3%), hearing loss in nine patients (39.1%), renal tract abnormalities in nine patients (39.1%), and diabetes insipidus in five patients (21.7%). The mean onset age for DM was significantly younger in the patients with biallelic null variants than in the patients with biallelic missense variants. Conclusions: Our results extend the pathogenic variant spectrum of WFS1. DIVs and CNVs explained rare unresolved Chinese cases with WFS1. The patients showed a wide and variable clinical spectrum, supporting the importance of genetic analysis for patients with atypical WFS1.


Subject(s)
Optic Atrophy , Wolfram Syndrome , China/epidemiology , Genetic Testing , Humans , Membrane Proteins/genetics , Optic Atrophy/pathology , Wolfram Syndrome/diagnosis , Wolfram Syndrome/genetics , Wolfram Syndrome/pathology
SELECTION OF CITATIONS
SEARCH DETAIL