Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
J Pediatr Endocrinol Metab ; 37(5): 434-440, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38465704

ABSTRACT

OBJECTIVES: Wolfram syndrome is characterised by insulin-dependent diabetes (IDDM), diabetes insipidus (DI), optic atrophy, sensorineural deafness and neurocognitive disorders. The DIDMOAD acronym has been recently modified to DIDMOAUD suggesting the rising awareness of the prevalence of urinary tract dysfunction (UD). End stage renal disease is the commonest cause of mortality in Wolfram syndrome. We present a case series with main objective of long term follow up in four children having Wolfram syndrome with evaluation of their urodynamic profile. METHODS: A prospective follow up of four genetically proven children with Wolfram syndrome presenting to a tertiary care pediatric diabetes clinic in Pune, India was conducted. Their clinical, and urodynamic parameters were reviewed. RESULTS: IDDM, in the first decade, was the initial presentation in all the four children (three male and one female). Three children had persistent polyuria and polydipsia despite having optimum glycemic control; hence were diagnosed to have DI and treated with desmopressin. All four patients entered spontaneous puberty. All patients had homozygous mutation in WFS1 gene; three with exon 8 and one with exon 6 novel mutations. These children with symptoms of lower urinary tract malfunction were further evaluated with urodynamic studies; two of them had hypocontractile detrusor and another had sphincter-detrusor dyssynergia. Patients with hypocontractile bladder were taught clean intermittent catheterization and the use of overnight drain. CONCLUSIONS: We report a novel homozygous deletion in exon 6 of WFS-1 gene. The importance of evaluation of lower urinary tract malfunction is highlighted by our case series. The final bladder outcome in our cases was a poorly contractile bladder in three patients.


Subject(s)
Urodynamics , Wolfram Syndrome , Adolescent , Child , Female , Humans , Male , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/physiopathology , Follow-Up Studies , Membrane Proteins/genetics , Mutation , Prognosis , Prospective Studies , Wolfram Syndrome/genetics , Wolfram Syndrome/complications , Wolfram Syndrome/physiopathology
2.
Sci Rep ; 12(1): 2103, 2022 02 08.
Article in English | MEDLINE | ID: mdl-35136185

ABSTRACT

To evaluate differences in macular and optic disc circulation in patients affected by Wolfram Syndrome (WS) employing optical coherence tomography-angiography (OCTA) imaging. In this retrospective study, 18 eyes from 10 WS patients, 16 eyes of 8 patients affected by type I diabetes and 17 eyes from 17 healthy controls were enrolled. All patients were imaged through OCT and OCTA and vascular parameters, as perfusion density (PD) and vessel length density (VLD) were measured. OCTA showed reduced PD in WS patients at the macular superficial capillary plexus (SCP, 27.8 ± 5.3%), deep vascular complex (DVC, 33.2 ± 1.9%) and optic nerve head (ONH, 21.2 ± 9.1%) compared to both diabetic patients (SCP 33.9 ± 1.9%, P < 0.0001; DVC 33.2 ± 0.7%, P = 1.0; ONH 33.9 ± 1.3, P < 0.0001) and healthy controls (SCP 31.6 ± 2.5, P = 0.002; DVC 34.0 ± 0.7%, P = 0.089; ONH 34.6 ± 0.8%, P < 0.0001). Similarly, VLD was lower in WS patients at the SCP (10.9 ± 2.7%) and ONH levels (7.5 ± 4.1%) compared to diabetic patients (SCP 13.8 ± 1.2%, P = 0.001; DVC 13.8 ± 0.2%, P < 0.0001; ONH 13.0 ± 0.7%, P = < 0.0001), but higher in DVC (15.7 ± 1.2%, P < 0.0001). Furthermore, VLD was lower in WS patients in all the vascular parameters compared to controls (SCP 13.8 ± 1.5%, P < 0.0001; DVC 17.3 ± 0.6%, P < 0.0001; ONH 15.7 ± 0.5%, P < 0.0001). A significant microvasculature impairment in the macular SCP and ONH microvasculature was demonstrated in eyes affected by WS. Microvascular impairment may be considered a fundamental component of the neurodegenerative changes in WS.


Subject(s)
Macula Lutea/blood supply , Microcirculation , Microvessels/pathology , Optic Disk/blood supply , Wolfram Syndrome/diagnostic imaging , Adolescent , Adult , Angiography , Child , Female , Humans , Male , Middle Aged , Retrospective Studies , Tomography, Optical Coherence , Wolfram Syndrome/pathology , Wolfram Syndrome/physiopathology , Young Adult
3.
Sci Rep ; 11(1): 20491, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34650143

ABSTRACT

Wolfram syndrome (WS) is an ultra-rare progressive neurodegenerative disorder defined by early-onset diabetes mellitus and optic atrophy. The majority of patients harbour recessive mutations in the WFS1 gene, which encodes for Wolframin, a transmembrane endoplasmic reticulum protein. There is limited availability of human ocular and brain tissues, and there are few animal models for WS that replicate the neuropathology and clinical phenotype seen in this disorder. We, therefore, characterised two wfs1 zebrafish knockout models harbouring nonsense wfs1a and wfs1b mutations. Both homozygous mutant wfs1a-/- and wfs1b-/- embryos showed significant morphological abnormalities in early development. The wfs1b-/- zebrafish exhibited a more pronounced neurodegenerative phenotype with delayed neuronal development, progressive loss of retinal ganglion cells and clear evidence of visual dysfunction on functional testing. At 12 months of age, wfs1b-/- zebrafish had a significantly lower RGC density per 100 µm2 (mean ± standard deviation; 19 ± 1.7) compared with wild-type (WT) zebrafish (25 ± 2.3, p < 0.001). The optokinetic response for wfs1b-/- zebrafish was significantly reduced at 8 and 16 rpm testing speeds at both 4 and 12 months of age compared with WT zebrafish. An upregulation of the unfolded protein response was observed in mutant zebrafish indicative of increased endoplasmic reticulum stress. Mutant wfs1b-/- zebrafish exhibit some of the key features seen in patients with WS, providing a versatile and cost-effective in vivo model that can be used to further investigate the underlying pathophysiology of WS and potential therapeutic interventions.


Subject(s)
Membrane Proteins/genetics , Membrane Proteins/metabolism , Wolfram Syndrome/genetics , Wolfram Syndrome/physiopathology , Animals , Codon, Nonsense , Disease Models, Animal , Gene Knockout Techniques , Mutation , Optic Atrophy , Unfolded Protein Response , Wolfram Syndrome/metabolism , Zebrafish
4.
JCI Insight ; 6(15)2021 08 09.
Article in English | MEDLINE | ID: mdl-34185708

ABSTRACT

BACKGROUNDWolfram syndrome is a rare ER disorder characterized by insulin-dependent diabetes mellitus, optic nerve atrophy, and progressive neurodegeneration. Although there is no treatment for Wolfram syndrome, preclinical studies in cell and rodent models suggest that therapeutic strategies targeting ER calcium homeostasis, including dantrolene sodium, may be beneficial.METHODSBased on results from preclinical studies on dantrolene sodium and ongoing longitudinal studies, we assembled what we believe is the first-ever clinical trial in pediatric and adult Wolfram syndrome patients with an open-label phase Ib/IIa trial design. The primary objective was to assess the safety and tolerability of dantrolene sodium in adult and pediatric Wolfram syndrome patients. Secondary objectives were to evaluate the efficacy of dantrolene sodium on residual pancreatic ß cell functions, visual acuity, quality-of-life measures related to vision, and neurological functions.RESULTSDantrolene sodium was well tolerated by Wolfram syndrome patients. Overall, ß cell functions were not significantly improved, but there was a significant correlation between baseline ß cell functions and change in ß cell responsiveness (R2, P = 0.004) after 6-month dantrolene therapy. Visual acuity and neurological functions were not improved by 6-month dantrolene sodium. Markers of inflammatory cytokines and oxidative stress, such as IFN-γ, IL-1ß, TNF-α, and isoprostane, were elevated in subjects.CONCLUSIONThis study justifies further investigation into using dantrolene sodium and other small molecules targeting the ER for treatment of Wolfram syndrome.TRIAL REGISTRATIONClinicalTrials.gov identifier NCT02829268FUNDINGNIH/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) (DK112921, DK113487, DK020579), NIH/National Center for Advancing Translational Sciences (NCATS) (TR002065, TR000448), NIH training grant (F30DK111070), Silberman Fund, Ellie White Foundation, Snow Foundation, Unravel Wolfram Syndrome Fund, Stowe Fund, Eye Hope Foundation, Feiock Fund, Washington University Institute of Clinical and Translational Sciences grant UL1TR002345 from NIH/NCATS, Bursky Center for Human Immunology & Immunotherapy Programs.


Subject(s)
Dantrolene , Insulin-Secreting Cells , Interleukin-18/analysis , Interleukin-1beta/analysis , Quality of Life , Visual Acuity/drug effects , Wolfram Syndrome , Adolescent , Adult , Biological Availability , Calcium Signaling/drug effects , Child , Dantrolene/administration & dosage , Dantrolene/adverse effects , Dantrolene/pharmacokinetics , Dose-Response Relationship, Drug , Drug Monitoring/methods , Humans , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/physiology , Molecular Targeted Therapy/methods , Molecular Targeted Therapy/statistics & numerical data , Muscle Relaxants, Central/administration & dosage , Muscle Relaxants, Central/adverse effects , Muscle Relaxants, Central/pharmacokinetics , Neurologic Examination/drug effects , Treatment Outcome , Wolfram Syndrome/diagnosis , Wolfram Syndrome/drug therapy , Wolfram Syndrome/metabolism , Wolfram Syndrome/physiopathology
5.
Hum Mol Genet ; 30(3-4): 265-276, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33693650

ABSTRACT

Mutations in the WFS1 gene, encoding wolframin (WFS1), cause endoplasmic reticulum (ER) stress and are associated with a rare autosomal-recessive disorder known as Wolfram syndrome (WS). WS is clinically characterized by childhood-onset diabetes mellitus, optic atrophy, deafness, diabetes insipidus and neurological signs. We identified two novel WFS1 mutations in a patient with WS, namely, c.316-1G > A (in intron 3) and c.757A > T (in exon 7). Both mutations, located in the N-terminal region of the protein, were predicted to generate a truncated and inactive form of WFS1. We found that although the WFS1 protein was not expressed in peripheral blood mononuclear cells (PBMCs) of the proband, no constitutive ER stress activation could be detected in those cells. In contrast, WS proband's PBMCs produced very high levels of proinflammatory cytokines (i.e. TNF-α, IL-1ß, and IL-6) in the absence of any stimulus. WFS1 silencing in PBMCs from control subjects by means of small RNA interference also induced a pronounced proinflammatory cytokine profile. The same cytokines were also significantly higher in sera from the WS patient as compared to matched healthy controls. Moreover, the chronic inflammatory state was associated with a dominance of proinflammatory T helper 17 (Th17)-type cells over regulatory T (Treg) lymphocytes in the WS PBMCs. The identification of a state of systemic chronic inflammation associated with WFS1 deficiency may pave the way to innovative and personalized therapeutic interventions in WS.


Subject(s)
Inflammation , Leukocytes, Mononuclear/metabolism , Membrane Proteins/genetics , Mutation , Wolfram Syndrome/metabolism , Child , Cytokines/genetics , Cytokines/metabolism , Female , Gene Expression Regulation , Humans , Leukocytes, Mononuclear/immunology , Sequence Analysis, DNA , Wolfram Syndrome/genetics , Wolfram Syndrome/immunology , Wolfram Syndrome/physiopathology
6.
Behav Brain Res ; 405: 113187, 2021 05 07.
Article in English | MEDLINE | ID: mdl-33610659

ABSTRACT

Wolfram syndrome (WFS) is a rare autosomal recessive disorder characterized by diabetes mellitus and insipidus, progressive optic atrophy and sensorineural deafness. An increased incidence of psychiatric disorders has also been reported in WFS patients. There are two subtypes of WFS. Type 1 (WFS1) is caused by mutations in the WFS1 gene and type 2 (WFS2) results from mutations in the CISD2 gene. Existing Wfs1 knockout mice exhibit many WFS1 cardinal symptoms including diabetic nephropathy, metabolic disruptions and optic atrophy. Far fewer studies have examined loss of Cisd2 function in mice. We identified B6.DDY-Cisd2m1Lmt, a mouse model with a spontaneous mutation in the Cisd2 gene. B6.DDY-Cisd2m1Lmt mice were initially identified based on the presence of audible sonic vocalizations as well as decreased body size and weight compared to unaffected wildtype littermates. Although Wfs1 knockout mice have been characterized for numerous behavioral phenotypes, similar studies have been lacking for Cisd2 mutant mice. We tested B6.DDY-Cisd2m1Lmt mice in a battery of behavioral assays that model phenotypes related to neurological and psychiatric disorders including anxiety, sensorimotor gating, stress response, social interaction and learning and memory. B6.DDY-Cisd2m1Lmt mice displayed hypoactivity across several behavioral tests, exhibited increased stress response and had deficits in spatial learning and memory and sensorimotor gating compared to wildtype littermates. Our data indicate that the B6.DDY-Cisd2m1Lmt mouse strain is a useful model to investigate potential mechanisms underlying the neurological and psychiatric symptoms observed in WFS.


Subject(s)
Autophagy-Related Proteins/genetics , Behavior, Animal/physiology , Cognitive Dysfunction , Disease Models, Animal , Motor Activity/physiology , Nerve Tissue Proteins/genetics , Prepulse Inhibition/physiology , Wolfram Syndrome , Animals , Cognitive Dysfunction/genetics , Cognitive Dysfunction/physiopathology , Corticosterone/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/genetics , Phenotype , Prepulse Inhibition/genetics , Vocalization, Animal/physiology , Wolfram Syndrome/genetics , Wolfram Syndrome/physiopathology
8.
J Clin Res Pediatr Endocrinol ; 13(1): 34-43, 2021 02 26.
Article in English | MEDLINE | ID: mdl-32938580

ABSTRACT

Objective: Bi-allelic mutations in the wolframin gene (WFS1) cause Wolfram syndrome 1 (WS1 or DIDMOAD) characterized by nonautoimmune diabetes mellitus, optic atrophy, diabetes insipidus, sensorineural deafness, urinary tract abnormalities, and neuropsychiatric disorders. Patients presenting with an incomplete phenotype of WS1 were evaluated using homozygosity mapping and subsequent whole-exome sequencing. Methods: Four unrelated consanguineous Turkish families, including seven affected children, and their unaffected parents and siblings were evaluated. Homozygosity mapping was performed, followed by whole-exome sequencing of WFS1. Mutations were classified according to results of "in silico" analyses, protein prediction, and functional consequences. Results: Homozygosity mapping confirmed shared homozygous regions on chromosome 4 (chr4p16.1) between the affected individuals, that was absent in their unaffected siblings. Exome sequencing identified three novel (c.1215T>A, c.554G>A, c.1525_1540dup) and one known (c.1522_1523delTA) mutations in WFS1. All mutations were predicted to cause stop codon leading to early termination of protein synthesis and complete loss-of-function. All patients were found to be homozygous for the change, with parents and other unaffected siblings being carriers. Conclusion: Our study expands the mutation spectrum of WSF1 mutations with three novel mutations. Homozygosity mapping may provide enrichment for molecular genetic analysis and early diagnosis of WS1 patients with incomplete phenotype, particularly in consanguineous pedigrees.


Subject(s)
Membrane Proteins/genetics , Wolfram Syndrome/genetics , Wolfram Syndrome/physiopathology , Adolescent , Adult , Child , Consanguinity , Female , Humans , Male , Pedigree , Turkey , Young Adult
9.
Lab Invest ; 100(9): 1197-1207, 2020 09.
Article in English | MEDLINE | ID: mdl-32366942

ABSTRACT

Endoplasmic reticulum (ER) stress-mediated cell death is an emerging target for human chronic disorders, including neurodegeneration and diabetes. However, there is currently no treatment for preventing ER stress-mediated cell death. Here, we show that mesencephalic astrocyte-derived neurotrophic factor (MANF), a neurotrophic factor secreted from ER stressed cells, prevents ER stress-mediated ß cell death and enhances ß cell proliferation in cell and mouse models of Wolfram syndrome, a prototype of ER disorders. Our results indicate that molecular pathways regulated by MANF are promising therapeutic targets for regenerative therapy of ER stress-related disorders, including diabetes, retinal degeneration, neurodegeneration, and Wolfram syndrome.


Subject(s)
Cell Proliferation/drug effects , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum/metabolism , Insulin-Secreting Cells/drug effects , Nerve Growth Factors/pharmacology , Wolfram Syndrome/prevention & control , Animals , Cell Line, Tumor , Cells, Cultured , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Mice, Transgenic , Rats , Wolfram Syndrome/metabolism , Wolfram Syndrome/physiopathology
10.
Am J Ophthalmol ; 217: 140-151, 2020 09.
Article in English | MEDLINE | ID: mdl-32335055

ABSTRACT

PURPOSE: To evaluate corneal morphology among patients with Wolfram syndrome (WFS). DESIGN: Comparative observational longitudinal case series of WFS patients with a laboratory approach in the WFS1 gene knockout (Wfs1KO) mouse model. METHODS: A group of 12 patients with biallelic mutations in the WFS1 gene recruited from the whole country and a control group composed of 30 individuals with type 1 diabetes (T1D) were evaluated in a national reference center for monogenic diabetes. All subjects (n = 42) underwent a complete ophthalmic examination, computer videokeratography, and corneal thickness and endothelial measurements. Additionally, WFS patients (n = 9) underwent longitudinal videokeratography and Pentacam evaluation. Corneal characteristics were assessed and compared between both groups. Human and mouse corneas were subjected to immunohistochemistry to detect wolframin expression and microscopic evaluation to study corneal morphology ex vivo. RESULTS: Clinical and topographic abnormalities similar to keratoconus were observed in 14 eyes (58.3%) of 8 WFS patients (66.7%). Flat keratometry, inferior-superior dioptric asymmetry, skewed radial axis, logarithm of keratoconus percentage index, index of surface variance, index of vertical asymmetry, keratoconus index, central keratoconus index, index of height asymmetry, and index of height decentration differed between WFS and T1D patients. Immunohistochemistry demonstrated wolframin expression in human and mouse corneas. Compared with Wfs1WT mice, Wfs1KO mice also presented corneal abnormalities. CONCLUSIONS: Patients with WFS present a high prevalence of changes in corneal morphology compatible with the diagnosis of early stages of keratoconus. Observations in a mouse model suggest that a mutation in the WFS1 gene may be responsible for corneal abnormalities similar to keratoconus.


Subject(s)
Cornea/pathology , Corneal Diseases/diagnosis , Corneal Topography/methods , Wolfram Syndrome/complications , Adolescent , Adult , Animals , Corneal Diseases/etiology , Corneal Diseases/physiopathology , Corneal Pachymetry , Female , Humans , Male , Mice , ROC Curve , Retrospective Studies , Wolfram Syndrome/diagnosis , Wolfram Syndrome/physiopathology , Young Adult
11.
Prog Urol ; 30(4): 205-208, 2020 Mar.
Article in French | MEDLINE | ID: mdl-31761518

ABSTRACT

Wolfram syndrome is a neurodegenerative disorder characterized by childhood onset diabetes mellitus, optic nerve atrophy, diabetes insipidus, hearing impairment, brainstem alteration and commonly bladder and bowel dysfunction. OBJECTIVE: We present here, 6 new cases of urinary dysfunction in this rare disease. RESULTS: All patients had urinary retention with overactive bladder. The urodynamic assessment found overactive detrusor in 3 cases. Five out of six patients performed self-catheterization and were treated with anticholinergics or intradetrusor injection of botulinum toxin. The follow-up at 5 years found an alteration of the upper urinary tracts and a renal failure 3/6. CONCLUSION: Urinary dysfunction is common in Wolfram syndrome, mainly characterized by overactive bladder and urinary retention. The urological risk is major requiring a systelmatic follow-up of these patients. LEVEL OF EVIDENCE: 4.


Subject(s)
Urinary Bladder, Overactive/etiology , Urinary Retention/etiology , Wolfram Syndrome/physiopathology , Adolescent , Adult , Botulinum Toxins/administration & dosage , Cholinergic Antagonists/administration & dosage , Female , Follow-Up Studies , Humans , Male , Middle Aged , Self Care/methods , Urinary Bladder, Overactive/therapy , Urinary Catheterization/methods , Urinary Retention/therapy , Urodynamics , Young Adult
12.
Hum Mol Genet ; 28(22): 3815-3824, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31600780

ABSTRACT

Wolfram syndrome (WS) is a heterogeneous multisystem neurodegenerative disorder with two allelic variations in addition to a separate subtype known as WS type 2. The wide phenotypic spectrum of WS includes diabetes mellitus and optic atrophy which is often accompanied by diabetes insipidus, deafness, urological and neurological complications in combination or in isolation. To date, the understanding of the genotype-phenotype relationship in this complex syndrome remains poorly understood. In this study, we identified and explored the functionality of rare and novel variants in the two causative WS genes WFS1 and CISD2 by assessing the effects of the mutations on the encoded proteins Wolframin and ERIS, in a cohort of 12 patients with autosomal recessive WS, dominant WS and WS type 2. The identified pathogenic variants included missense changes, frameshift deletions and insertions in WFS1 and an exonic deletion in CISD2 which all altered the respective encoded protein in a manner that did not correlate to the phenome previously described. These observations suggest the lack of genotype-phenotype correlation in this complex syndrome and the need to explore other molecular genetic mechanisms. Additionally, our findings highlight the importance of functionally assessing variants for their pathogenicity to tackle the problem of increasing variants of unknown significance in the public genetic databases.


Subject(s)
Membrane Proteins/genetics , Wolfram Syndrome/genetics , Adolescent , Adult , Alleles , Exons , Female , Frameshift Mutation , Genetic Association Studies , Humans , Male , Membrane Proteins/metabolism , Mutation , Optic Atrophy/genetics , Pedigree , Phenotype , Wolfram Syndrome/physiopathology
13.
Orphanet J Rare Dis ; 14(1): 188, 2019 08 02.
Article in English | MEDLINE | ID: mdl-31375124

ABSTRACT

BACKGROUND: Wolfram syndrome is a rare disorder associated with diabetes mellitus, diabetes insipidus, optic nerve atrophy, hearing and vision loss, and neurodegeneration. Sleep complaints are common but have not been studied with objective measures. Our goal was to assess rates of sleep apnea and objective and self-reported measures of sleep quality, and to determine the relationship of sleep pathology to other clinical variables in Wolfram syndrome patients. METHODS: Genetically confirmed Wolfram syndrome patients were evaluated at the 2015 and 2016 Washington University Wolfram Syndrome Research Clinics. Patients wore an actigraphy device and a type III ambulatory sleep study device and completed the Epworth Sleepiness Scale (ESS), the Pittsburgh Sleep Quality Index (PSQI) and/or the Pediatric Sleep Questionnaire (PSQ). PSQI and PSQ questionnaire data were compared to a previously collected group of controls. Patients were characterized clinically with the Wolfram Unified Rating Scale (WURS) and a subset underwent magnetic resonance imaging (MRI) for brain volume measurements. RESULTS: Twenty-one patients were evaluated ranging from age 8.9-29.7 years. Five of 17 (29%) adult patients fit the criteria for obstructive sleep apnea (OSA; apnea-hypopnea index [AHI] ≥ 5) and all 4 of 4 (100%) children aged 12 years or younger fit the criteria for obstructive sleep apnea (AHI's ≥ 1). Higher AHI was related to greater disease severity (higher WURS Physical scores). Higher mixed apnea scores were related to lower brainstem and cerebellar volumes. Patients' scores on the PSQ were higher than those of controls, indicating greater severity of childhood obstructive sleep-related breathing disorders. CONCLUSIONS: Wolfram syndrome patients had a high rate of OSA. Further study would be needed to assess how these symptoms change over time. Addressing sleep disorders in Wolfram syndrome patients would likely improve their overall health and quality of life.


Subject(s)
Sleep Wake Disorders/physiopathology , Wolfram Syndrome/physiopathology , Adolescent , Adult , Female , Humans , Magnetic Resonance Imaging , Male , Sleep Wake Disorders/diagnostic imaging , Surveys and Questionnaires , Wolfram Syndrome/diagnostic imaging , Young Adult
14.
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
15.
Orphanet J Rare Dis ; 13(1): 102, 2018 06 27.
Article in English | MEDLINE | ID: mdl-29945639

ABSTRACT

BACKGROUND: Wolfram syndrome (WFS) is a rare autosomal recessive disease with clinical manifestations of diabetes mellitus (DM), diabetes insipidus (DI), optic nerve atrophy (OA) and sensorineural hearing loss (SNHL). Although SNHL is a key symptom of WFS, there is limited information on its natural history using standardized measures. Such information is important for clinical care and determining its use as an outcome measure in clinical trials. METHODS: Standardized audiologic measures, including pure-tone testing, tympanometry, speech perception, and the unaided Speech Intelligibility Index (SII) were assessed in patients with confirmed WFS annually. Mixed model analyses were used to examine main effects of age, time or interactions for pure tone average (PTA), high frequency average (HFA) and SII. RESULTS: Forty WFS patients were evaluated between 1 and 6 times. Mean age at initial enrollment was 13.5 years (SD = 5.6). Patients were classified as having normal hearing (n = 10), mild-to-severe (n = 24) or profound SNHL (n = 6). Mean age of diagnosis for SNHL was 8.3 years (SD = 5.1) with 75% prevalence. HFA worsened over time for both ears, and SII worsened over time in the worse ear, with greater decline in both measures in younger patients. Average estimated change over 1 year for all measures was in the subclinical range and power analyses suggest that 100 patients would be needed per group (treatment vs. placebo) to detect a 60% reduction in annual change of HFA over 3 years. If trials focused on just those patients with SNHL, power estimates suggest 55 patients per group would be sufficient. CONCLUSIONS: Most patients had a slow progressive SNHL emerging in late childhood. Change over time with standard audiologic tests (HFA, SII) was small and would not be detectable for at least 2 years in an individual. Relatively large sample sizes would be necessary to detect significant impact on hearing progression in a clinical trial. Hearing function should be monitored clinically in WFS to provide appropriate intervention. Because SNHL can occur very early in WFS, audiologists and otolaryngologists should be aware of and refer for later emerging symptoms.


Subject(s)
Hearing Loss/physiopathology , Wolfram Syndrome/physiopathology , Adolescent , Adult , Child , Female , Humans , Male , Speech Perception/physiology , Young Adult
16.
J Fr Ophtalmol ; 41(5): 402-406, 2018 May.
Article in French | MEDLINE | ID: mdl-29779933

ABSTRACT

INTRODUCTION: Hereditary optic neuropathies (HON) often begin in adulthood. However, some of them can have an early onset. These may have specific clinical features and natural histories. PATIENTS AND METHODS: Retrospective study of HON patients with onset before the age of 14 years seen in a referral center. In addition to the age of onset, we evaluated the genetic etiology, visual acuity at 15 years, last best corrected visual acuity, optic disc appearance, visual field and extra-ophthalmological manifestations. RESULTS: Forty-four patients (16 women) were included; i.e. 27.8% of all patients followed for HON. The mean age of onset was 8.5±3.3 years, with an onset earlier than 3 years in 5 patients. An etiology was not found in 8 patients. Of the remaining 36 patients, 12 had Leber's hereditary optic neuropathy (LHON), 11 had dominant optic atrophy, 12 had WS/WS-like syndrome, 2 had recessive optic atrophy and 1 had spastic paraplegia type 7. For 78 eyes of 40 patients (mean age 26.9±14.5 years), the mean last visual acuity was 0.80±0.33 LogMAR, with differences according to genetic forms. Visual acuity was less than or equal to counting fingers for 7 eyes (29.1%) of 4 WS/WS-like patients and one LHON patient. CONCLUSION: Early onset NOH are not unusual. Their visual prognosis is as severe as adult onset NOH, with variations depending on the underlying genetic causes.


Subject(s)
Optic Nerve Diseases/epidemiology , Optic Nerve Diseases/genetics , Optic Nerve Diseases/physiopathology , Optic Nerve Diseases/therapy , Visual Acuity/physiology , Adolescent , Adult , Age of Onset , Child , Child, Preschool , Female , Humans , Male , Optic Atrophy, Hereditary, Leber/epidemiology , Optic Atrophy, Hereditary, Leber/genetics , Optic Atrophy, Hereditary, Leber/physiopathology , Optic Atrophy, Hereditary, Leber/therapy , Retrospective Studies , Visual Acuity/genetics , Wolfram Syndrome/epidemiology , Wolfram Syndrome/genetics , Wolfram Syndrome/physiopathology , Wolfram Syndrome/therapy , Young Adult
17.
Cell Death Dis ; 9(3): 364, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29511163

ABSTRACT

Wolfram syndrome (WS) is a rare neurodegenerative disease, the main pathological hallmarks of which associate with diabetes, optic atrophy, and deafness. Other symptoms may be identified in some but not all patients. Prognosis is poor, with death occurring around 35 years of age. To date, no treatment is available. WS was first described as a mitochondriopathy. However, the localization of the protein on the endoplasmic reticulum (ER) membrane challenged this hypothesis. ER contacts mitochondria to ensure effective Ca2+ transfer, lipids transfer, and apoptosis within stabilized and functionalized microdomains, termed "mitochondria-associated ER membranes" (MAMs). Two types of WS are characterized so far and Wolfram syndrome type 2 is due to mutation in CISD2, a protein mostly expressed in MAMs. The aim of the present review is to collect evidences showing that WS is indeed a mitochondriopathy, with established MAM dysfunction, and thus share commonalities with several neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis, as well as metabolic diseases, such as diabetes.


Subject(s)
Endoplasmic Reticulum/metabolism , Intracellular Membranes/metabolism , Mitochondria/metabolism , Wolfram Syndrome/metabolism , Wolfram Syndrome/pathology , Endoplasmic Reticulum Stress , Humans , Models, Biological , Wolfram Syndrome/physiopathology
18.
Pediatr Endocrinol Diabetes Metab ; 23(2): 107-110, 2017.
Article in English | MEDLINE | ID: mdl-29073294

ABSTRACT

Wolfram syndrome (WFS) is a rare autosomal recessive disease with non-autoimmune childhood onset insulin dependent diabetes and optic atrophy. WFS type 2 (WFS2) differs from WFS type 1 (WFS1) with upper intestinal ulcers, bleeding tendency and the lack ofdiabetes insipidus. Li-fespan is short due to related comorbidities. Only a few familieshave been reported with this syndrome with the CISD2 mutation. Here we report two siblings with a clinical diagnosis of WFS2, previously misdiagnosed with type 1 diabetes mellitus and diabetic retinopathy-related blindness. We report possible additional clinical and laboratory findings that have not been pre-viously reported, such as asymptomatic hypoparathyroidism, osteomalacia, growth hormone (GH) deficiency and hepatomegaly. Even though not a requirement for the diagnosis of WFS2 currently, our case series confirm hypogonadotropic hypogonadism to be also a feature of this syndrome, as reported before.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/etiology , Hypogonadism/physiopathology , Hypoparathyroidism/physiopathology , Osteomalacia/physiopathology , Wolfram Syndrome/complications , Wolfram Syndrome/physiopathology , Adolescent , Antidiuretic Agents/therapeutic use , Child , Female , Humans , Hypogonadism/genetics , Hypoparathyroidism/genetics , Insulin/therapeutic use , Male , Osteomalacia/genetics , Treatment Outcome , Wolfram Syndrome/genetics , Young Adult
19.
Gac Med Mex ; 153(4): 466-472, 2017.
Article in Spanish | MEDLINE | ID: mdl-28991271

ABSTRACT

Background: Wolfram syndrome (WS), also known by the acronym DIDMOAD, is a rare and progresive hereditary disease of autosomal recessive inheritance which minimum ascertainment diagnostic criteria are the occurrence together of diabetes mellitus and optic atrophy before 15 years of age. Objective: To describe the clinical, biochemical and molecular profile of WS in a tertiary care hospital in Mexico. Materials and Methods: We reviewed patients records who fulfill the minimum ascertainment diagnostic criteria of WS presenting between January 1987 and May 2015 in a tertiary care hospital in Mexico. Results: Five patients fulfill the inclusion criteria (three male and two female). Diabetes mellitus was the first manifestation of the syndrome in all of them, with a mean age at diagnosis of 5.8 ± 2.71 years, while the WS diagnosis was established at a mean age of 15.8 ± 8.37 years. All the patients had optic atrophy and two of them presented with the complete DIDMOAD spectrum. We found new associations with autoimmune hepatitis and testicular cancer. Conclusions: This study shows the variability of clinical presentation of WS, as well as two new associations.


Antecedentes: El síndrome de Wolfram (SW), también conocido por el acrónimo DIDMOAD, es una enfermedad hereditaria rara y progresiva, de transmisión autosómica recesiva, cuyos criterios diagnósticos mínimos son diabetes mellitus y atrofia óptica antes de los 15 años de edad. Objetivo: Describir la presentación clínica, bioquímica y molecular del SW en un hospital de tercer nivel en México. Material y Métodos: Se revisaron los expedientes de pacientes que cumplían con criterios diagnósticos clínicos mínimos de SW atendidos entre enero de 1987 y mayo de 2015 en un hospital de tercer nivel en México. Resultados: Cinco pacientes cumplieron con los criterios de inclusión (tres hombres y dos mujeres). La diabetes mellitus fue la primera manifestación del síndrome en todos ellos, con una media de edad al diagnóstico de 5.8 ± 2.71 años, mientras que el diagnóstico del SW se estableció en promedio a los 15.8 ± 8.37 años. Todos los pacientes tenían atrofia óptica y dos presentaron el espectro DIDMOAD completo. Se describen nuevas asociaciones con hepatitis autoinmunitaria y cáncer de testículo. Conclusiones: El presente estudio muestra la variabilidad de presentación clínica del SW y dos asociaciones no descritas previamente.


Subject(s)
Diabetes Mellitus/diagnosis , Optic Atrophy/diagnosis , Testicular Neoplasms/diagnosis , Wolfram Syndrome/diagnosis , Adolescent , Child , Child, Preschool , Diabetes Mellitus/epidemiology , Female , Hepatitis, Autoimmune/diagnosis , Hepatitis, Autoimmune/epidemiology , Humans , Male , Mexico , Optic Atrophy/epidemiology , Retrospective Studies , Testicular Neoplasms/epidemiology , Wolfram Syndrome/physiopathology , Young Adult
20.
Physiol Rep ; 4(7)2016 Apr.
Article in English | MEDLINE | ID: mdl-27053292

ABSTRACT

Wolfram syndrome, an autosomal recessive disorder characterized by juvenile-onset diabetes mellitus and optic atrophy, is caused by mutations in theWFS1gene.WFS1encodes an endoplasmic reticulum resident transmembrane protein. TheWfs1-null mice exhibit progressive insulin deficiency and diabetes. The aim of this study was to describe the insulin secretion and transcriptome of pancreatic islets inWFS1-deficient mice.WFS1-deficient (Wfs1KO) mice had considerably less pancreatic islets than heterozygous (Wfs1HZ) or wild-type (WT) mice. Wfs1KOpancreatic islets secreted less insulin after incubation in 2 and 10 mmol/L glucose and with tolbutamide solution compared toWTand Wfs1HZislets, but not after stimulation with 20 mmol/L glucose. Differences in proinsulin amount were not statistically significant although there was a trend that Wfs1KOhad an increased level of proinsulin. After incubation in 2 mmol/L glucose solution the proinsulin/insulin ratio in Wfs1KOwas significantly higher than that ofWTand Wfs1HZRNA-seq from pancreatic islets found melastatin-related transient receptor potential subfamily member 5 protein gene (Trpm5) to be downregulated inWFS1-deficient mice. Functional annotation ofRNAsequencing results showed thatWFS1 deficiency influenced significantly the pathways related to tissue morphology, endocrine system development and function, molecular transport network.


Subject(s)
Islets of Langerhans/metabolism , Membrane Proteins/deficiency , RNA/genetics , Sequence Analysis, RNA , Transcriptome , Wolfram Syndrome/genetics , Animals , Databases, Genetic , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Genetic Predisposition to Disease , Glucose/pharmacology , Heterozygote , Hypoglycemic Agents/pharmacology , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/pathology , Membrane Proteins/genetics , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Proinsulin/metabolism , RNA/metabolism , Real-Time Polymerase Chain Reaction , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism , Tissue Culture Techniques , Tolbutamide/pharmacology , Transcription, Genetic , Wolfram Syndrome/metabolism , Wolfram Syndrome/pathology , Wolfram Syndrome/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...