Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
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
2.
FEBS Open Bio ; 13(8): 1405-1414, 2023 08.
Article in English | MEDLINE | ID: mdl-37440664

ABSTRACT

Wolfram syndrome is a monogenic disease mainly caused by mutations in the WFS1 gene. Mutations in the WFS1 gene give rise to diabetes. Here, we characterized mutant WFS1 proteins by studying the stability of full-length wild-type (WT) WFS1, a missense mutant P724L, and two C-terminally truncated mutants, W837X and Y652X. We compared their stability by overexpressing them in MIN6 and HEK293T cells. The C-terminally truncated mutants W837X and Y652X are degraded more rapidly than the missense P724L mutant or wild-type WFS1 in MIN6 cells. In contrast, Y652X is more stable than WT or other mutant WFS1 proteins in HEK293T. In conclusion, we found that C-terminally truncated WFS1 mutants are selectively degraded in a cell type-specific manner.


Subject(s)
Insulin-Secreting Cells , Wolfram Syndrome , Humans , HEK293 Cells , Insulin-Secreting Cells/metabolism , Mutation/genetics , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Wolfram Syndrome/genetics , Wolfram Syndrome/metabolism
3.
Stem Cell Reports ; 18(5): 1090-1106, 2023 05 09.
Article in English | MEDLINE | ID: mdl-37163979

ABSTRACT

Mitochondrial dysfunction involving mitochondria-associated ER membrane (MAM) dysregulation is implicated in the pathogenesis of late-onset neurodegenerative diseases, but understanding is limited for rare early-onset conditions. Loss of the MAM-resident protein WFS1 causes Wolfram syndrome (WS), a rare early-onset neurodegenerative disease that has been linked to mitochondrial abnormalities. Here we demonstrate mitochondrial dysfunction in human induced pluripotent stem cell-derived neuronal cells of WS patients. VDAC1 is identified to interact with WFS1, whereas loss of this interaction in WS cells could compromise mitochondrial function. Restoring WFS1 levels in WS cells reinstates WFS1-VDAC1 interaction, which correlates with an increase in MAMs and mitochondrial network that could positively affect mitochondrial function. Genetic rescue by WFS1 overexpression or pharmacological agents modulating mitochondrial function improves the viability and bioenergetics of WS neurons. Our data implicate a role of WFS1 in regulating mitochondrial functionality and highlight a therapeutic intervention for WS and related rare diseases with mitochondrial defects.


Subject(s)
Induced Pluripotent Stem Cells , Neurodegenerative Diseases , Wolfram Syndrome , Humans , Wolfram Syndrome/genetics , Wolfram Syndrome/metabolism , Induced Pluripotent Stem Cells/metabolism , Neurodegenerative Diseases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neurons/metabolism , Mitochondria/metabolism , Mutation
4.
Biochem Biophys Res Commun ; 658: 80-87, 2023 05 28.
Article in English | MEDLINE | ID: mdl-37027908

ABSTRACT

Wfs1 is an endoplasmic reticulum (ER) membrane located protein highly expressed in pancreatic ß cells and brain. Wfs1 deficiency causes adult pancreatic ß cells dysfunction following ß cells apoptosis. Previous studies mainly focus on the Wfs1 function in adult mouse pancreatic ß cells. However, whether Wfs1 loss-of-function impairs mouse pancreatic ß cell from its early development is unknown. In our study, Wfs1 deficiency disrupts the composition of mouse pancreatic endocrine cells from early postnatal day 0 (P0) to 8 weeks old, with decreased percentage of ß cells and increased percentage of α and δ cells. Meanwhile, Wfs1 loss-of-function leads to reduced intracellular insulin content. Notably, Wfs1 deficiency impairs Glut2 localization and causes the accumulation of Glut2 in mouse pancreatic ß cell cytoplasm. In Wfs1-deficient mice, glucose homeostasis is disturbed from early 3 weeks old to 8 weeks old. This work reveals that Wfs1 is significantly required for the composition of pancreatic endocrine cells and is essential for Glut2 localization in mouse pancreatic ß cells.


Subject(s)
Insulin-Secreting Cells , Membrane Proteins , Wolfram Syndrome , Animals , Mice , Endoplasmic Reticulum/metabolism , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Wolfram Syndrome/metabolism , Membrane Proteins/genetics , Loss of Function Mutation
5.
Mol Psychiatry ; 28(4): 1557-1570, 2023 04.
Article in English | MEDLINE | ID: mdl-36750736

ABSTRACT

Dysregulated neurite outgrowth and synapse formation underlie many psychiatric disorders, which are also manifested by wolfram syndrome (WS). Whether and how the causative gene WFS1 deficiency affects synapse formation remain elusive. By mirroring human brain development with cerebral organoids, WFS1-deficient cerebral organoids not only recapitulate the neuronal loss in WS patients, but also exhibit significantly impaired synapse formation and function associated with reduced astrocytes. WFS1 deficiency in neurons autonomously delays neuronal differentiation with altered expressions of genes associated with psychiatric disorders, and impairs neurite outgrowth and synapse formation with elevated cytosolic calcium. Intriguingly, WFS1 deficiency in astrocytes decreases the expression of glutamate transporter EAAT2 by NF-κB activation and induces excessive glutamate. When co-cultured with wildtype neurons, WFS1-deficient astrocytes lead to impaired neurite outgrowth and increased cytosolic calcium in neurons. Importantly, disrupted synapse formation and function in WFS1-deficient cerebral organoids and impaired neurite outgrowth affected by WFS1-deficient astrocytes are efficiently reversed with Riluzole treatment, by restoring EAAT2 expression in astrocytes. Furthermore, Riluzole rescues the depressive-like behavior in the forced swimming test and the impaired recognition and spatial memory in the novel object test and water maze test in Wfs1 conditional knockout mice. Altogether, our study provides novel insights into how WFS1 deficiency affects synapse formation and function, and offers a strategy to treat this disease.


Subject(s)
Human Embryonic Stem Cells , Wolfram Syndrome , Animals , Mice , Humans , Wolfram Syndrome/drug therapy , Wolfram Syndrome/genetics , Wolfram Syndrome/metabolism , Riluzole/pharmacology , Riluzole/metabolism , Calcium/metabolism , Human Embryonic Stem Cells/metabolism , Neurons/metabolism , Mice, Knockout , Synapses/metabolism
6.
Elife ; 122023 01 16.
Article in English | MEDLINE | ID: mdl-36645345

ABSTRACT

Wolfram syndrome 1 (WS1) is a rare genetic disorder caused by mutations in the WFS1 gene leading to a wide spectrum of clinical dysfunctions, among which blindness, diabetes, and neurological deficits are the most prominent. WFS1 encodes for the endoplasmic reticulum (ER) resident transmembrane protein wolframin with multiple functions in ER processes. However, the WFS1-dependent etiopathology in retinal cells is unknown. Herein, we showed that Wfs1 mutant mice developed early retinal electrophysiological impairments followed by marked visual loss. Interestingly, axons and myelin disruption in the optic nerve preceded the degeneration of the retinal ganglion cell bodies in the retina. Transcriptomics at pre-degenerative stage revealed the STAT3-dependent activation of proinflammatory glial markers with reduction of the homeostatic and pro-survival factors glutamine synthetase and BDNF. Furthermore, label-free comparative proteomics identified a significant reduction of the monocarboxylate transport isoform 1 (MCT1) and its partner basigin that are highly enriched on retinal glia and myelin-forming oligodendrocytes in optic nerve together with wolframin. Loss of MCT1 caused a failure in lactate transfer from glial to neuronal cell bodies and axons leading to a chronic hypometabolic state. Thus, this bioenergetic impairment is occurring concurrently both within the axonal regions and cell bodies of the retinal ganglion cells, selectively endangering their survival while impacting less on other retinal cells. This metabolic dysfunction occurs months before the frank RGC degeneration suggesting an extended time-window for intervening with new therapeutic strategies focused on boosting retinal and optic nerve bioenergetics in WS1.


Subject(s)
Optic Atrophy , Wolfram Syndrome , Animals , Mice , Nerve Degeneration/metabolism , Neuroinflammatory Diseases , Retinal Ganglion Cells/metabolism , Wolfram Syndrome/genetics , Wolfram Syndrome/metabolism
7.
Acta Neuropathol Commun ; 10(1): 184, 2022 12 17.
Article in English | MEDLINE | ID: mdl-36527091

ABSTRACT

Wolfram Syndrome (WS) is a fatal human inherited disease with symptoms of diabetes, vision decreasing, and neurodegeneration caused by mutations in the endoplasmic reticulum (ER)-resident protein WFS1. WFS1 has been reported to play an important role in glucose metabolism. However, the role of WFS1 in axonal regeneration in the central nervous system has so far remained elusive. Herein, we established a model of the wfs1b globally deficient zebrafish line. wfs1b deficiency severely impeded the Mauthner-cell (M-cell) axon regeneration, which was partly dependent on the ER stress response. The administration of ER stress inhibitor 4-Phenylbutyric acid (4-PBA) promoted M-cell axon regeneration in wfs1b-/- zebrafish larvae, while the ER stress activator Tunicamycin (TM) inhibited M-cell axon regeneration in wfs1b+/+ zebrafish larvae. Moreover, complementation of wfs1b at the single-cell level stimulated M-cell axon regeneration in the wfs1b-/- zebrafish larvae. Altogether, our results revealed that wfs1b promotes M-cell axon regeneration through the ER stress signal pathway and provide new evidence for a therapeutic target for WS and axon degeneration.


Subject(s)
Wolfram Syndrome , Animals , Humans , Wolfram Syndrome/genetics , Wolfram Syndrome/metabolism , Zebrafish/metabolism , Axons/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nerve Regeneration , Endoplasmic Reticulum Stress , Signal Transduction , Mutation/genetics
8.
Cells ; 11(12)2022 06 18.
Article in English | MEDLINE | ID: mdl-35741091

ABSTRACT

Calcium ions (Ca2+) operate as important messengers in the cell, indispensable for signaling the underlying numerous cellular processes in all of the cell types in the human body. In neurons, Ca2+ signaling is crucial for regulating synaptic transmission and for the processes of learning and memory formation. Hence, the dysregulation of intracellular Ca2+ homeostasis results in a broad range of disorders, including cancer and neurodegeneration. A major source for intracellular Ca2+ is the endoplasmic reticulum (ER), which has close contacts with other organelles, including mitochondria. In this review, we focus on the emerging role of Ca2+ signaling at the ER-mitochondrial interface in two different neurodegenerative diseases, namely Alzheimer's disease and Wolfram syndrome. Both of these diseases share some common hallmarks in the early stages, including alterations in the ER and mitochondrial Ca2+ handling, mitochondrial dysfunction and increased Reactive oxygen species (ROS) production. This indicates that similar mechanisms may underly these two disease pathologies and suggests that both research topics might benefit from complementary research.


Subject(s)
Alzheimer Disease , Wolfram Syndrome , Alzheimer Disease/metabolism , Calcium/metabolism , Endoplasmic Reticulum/metabolism , Homeostasis , Humans , Wolfram Syndrome/metabolism
9.
Front Endocrinol (Lausanne) ; 13: 849204, 2022.
Article in English | MEDLINE | ID: mdl-35399956

ABSTRACT

Wolfram syndrome is a rare genetic disorder characterized by juvenile-onset diabetes mellitus, optic nerve atrophy, hearing loss, diabetes insipidus, and progressive neurodegeneration. Pathogenic variants in the WFS1 gene are the main causes of Wolfram syndrome. WFS1 encodes a transmembrane protein localized to the endoplasmic reticulum (ER) and regulates the unfolded protein response (UPR). Loss of function of WFS1 leads to dysregulation of insulin production and secretion, ER calcium depletion, and cytosolic calpains activation, resulting in activation of apoptotic cascades. Although the terminal UPR has been shown to induce inflammation that accelerates pancreatic ß-cell dysfunction and death in diabetes, the contribution of pancreatic ß-cell inflammation to the development of diabetes in Wolfram syndrome has not been fully understood. Here we show that WFS1-deficiency enhances the gene expression of pro-inflammatory cytokines and chemokines, leading to cytokine-induced ER-stress and cell death in pancreatic ß-cells. PERK and IRE1α pathways mediate high glucose-induced inflammation in a ß-cell model of Wolfram syndrome. M1-macrophage infiltration and hypervascularization are seen in the pancreatic islets of Wfs1 whole-body knockout mice, demonstrating that WFS1 regulates anti-inflammatory responses in pancreatic ß-cells. Our results indicate that inflammation plays an essential role in the progression of ß-cell death and diabetes in Wolfram syndrome. The pathways involved in ER stress-mediated inflammation provide potential therapeutic targets for the treatment of Wolfram syndrome.


Subject(s)
Endoplasmic Reticulum Stress , Insulin-Secreting Cells , Membrane Proteins , Wolfram Syndrome , Animals , Endoribonucleases/metabolism , Inflammation/metabolism , Insulin-Secreting Cells/metabolism , Loss of Function Mutation , Membrane Proteins/genetics , Mice , Protein Serine-Threonine Kinases , Wolfram Syndrome/genetics , Wolfram Syndrome/metabolism , Wolfram Syndrome/pathology
10.
Hum Mol Genet ; 31(16): 2711-2727, 2022 08 23.
Article in English | MEDLINE | ID: mdl-35325133

ABSTRACT

Wolfram syndrome (WS) is a rare genetic disease characterized by diabetes, optic atrophy and deafness. Patients die at 35 years of age, mainly from respiratory failure or dysphagia. Unfortunately, there is no treatment to block the progression of symptoms and there is an urgent need for adequate research models. Here, we report on the phenotypical characterization of two loss-of-function zebrafish mutant lines: wfs1aC825X and wfs1bW493X. We observed that wfs1a deficiency altered the size of the ear and the retina of the fish. We also documented a decrease in the expression level of unfolded protein response (UPR) genes in basal condition and in stress condition, i.e. after tunicamycin treatment. Interestingly, both mutants lead to a decrease in their visual function measured behaviorally. These deficits were associated with a decrease in the expression level of UPR genes in basal and stress conditions. Interestingly, basal, ATP-linked and maximal mitochondrial respirations were transiently decreased in the wfs1b mutant. Taken together, these zebrafish lines highlight the critical role of wfs1a and wfs1b in UPR, mitochondrial function and visual physiology. These models will be useful tools to better understand the cellular function of Wfs1 and to develop novel therapeutic approaches for WS.


Subject(s)
Optic Atrophy , Wolfram Syndrome , Animals , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutation , Optic Atrophy/genetics , Phenotype , Wolfram Syndrome/genetics , Wolfram Syndrome/metabolism , Zebrafish/genetics , Zebrafish/metabolism
11.
J Med Genet ; 59(1): 65-74, 2022 01.
Article in English | MEDLINE | ID: mdl-34006618

ABSTRACT

BACKGROUND: Wolfram syndrome (WFS) is a rare disorder characterised by childhood-onset diabetes mellitus and progressive optic atrophy. Most patients have variants in the WFS1 gene. We undertook functional studies of WFS1 variants and correlated these with WFS1 protein expression and phenotype. METHODS: 9 patients with a clinical diagnosis of WFS were studied with quantitative PCR for markers of endoplasmic reticulum (ER) stress and immunoblotting of fibroblast protein extracts for WFS1 protein expression. Luciferase reporter assay was used to assess ATF-6 dependent unfolded protein response (UPR) activation. RESULTS: 6 patients with compound heterozygous nonsense mutations in WFS1 had no detectable WFS1 protein expression; 3 patients with missense variants had 4%, 45% and 48% WFS1 protein expression. One of these also had an OPA1 mutation and was reclassified as autosomal dominant optic atrophy-plus syndrome. There were no correlations between ER stress marker mRNA and WFS1 protein expression. ERSE-luciferase reporter indicated activation of the ATF6 branch of UPR in two patients tested. Patients with partial WFS1 expression showed milder visual acuity impairment (asymptomatic or colour blind only), compared with those with absent expression (registered severe vision impaired) (p=0.04). These differences remained after adjusting for duration of optic atrophy. CONCLUSIONS: Patients with WFS who have partial WFS1 protein expression present with milder visual impairment. This suggests a protective effect of partial WFS1 protein expression on the severity and perhaps progression of vision impairment and that therapies to increase residual WFS1 protein expression may be beneficial.


Subject(s)
Gene Expression Regulation , Membrane Proteins/genetics , Mutation , Optic Atrophy/genetics , Phenotype , Wolfram Syndrome/genetics , Adolescent , Adult , Codon, Nonsense , Female , Humans , Male , Mutation, Missense , Pedigree , Wolfram Syndrome/metabolism , Young Adult
12.
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
13.
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
14.
J Pediatr Endocrinol Metab ; 34(8): 1049-1053, 2021 Aug 26.
Article in English | MEDLINE | ID: mdl-33882198

ABSTRACT

OBJECTIVES: Wolfram syndrome (WS) is a rarely seen autosomal recessive multisystem neurodegenerative disorder caused by mutations in the WFS1 gene. CASE PRESENTATION: Three sisters with WS had diabetes mellitus (DM) at 4 years of age and optic atrophy. In addition, the first case had hearing impairment, and the second case had diabetes insipidus and urinary incontinence. Linagliptin was administered to the first case as add-on therapy to intensive insulin treatment 15 years after the onset of DM, and her insulin need showed a dramatic decrease. The third case had a remission phase one month after the onset of DM. CONCLUSIONS: Even in cases with the same mutation, symptoms and findings may widely vary in WS. Remission of diabetes has rarely been reported in WS. Also, this report describes the first trial of a dipeptidyl peptidase-4 inhibitor in a patient with WS which provided a decrease in exogenous insulin need.


Subject(s)
Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Insulin/therapeutic use , Membrane Proteins/genetics , Mutation , Wolfram Syndrome/drug therapy , Adolescent , Child , Child, Preschool , Drug Therapy, Combination , Female , Humans , Hypoglycemic Agents/therapeutic use , Male , Pedigree , Prognosis , Remission Induction , Wolfram Syndrome/metabolism , Wolfram Syndrome/pathology
15.
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
16.
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
17.
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
18.
Sci Rep ; 10(1): 4785, 2020 03 16.
Article in English | MEDLINE | ID: mdl-32179840

ABSTRACT

Wolfram syndrome (WS) is a recessive multisystem disorder defined by the association of diabetes mellitus and optic atrophy, reminiscent of mitochondrial diseases. The role played by mitochondria remains elusive, with contradictory results on the occurrence of mitochondrial dysfunction. We evaluated 13 recessive WS patients by deep clinical phenotyping, including optical coherence tomography (OCT), serum lactic acid at rest and after standardized exercise, brain Magnetic Resonance Imaging, and brain and muscle Magnetic Resonance Spectroscopy (MRS). Finally, we investigated mitochondrial bioenergetics, network morphology, and calcium handling in patient-derived fibroblasts. Our results do not support a primary mitochondrial dysfunction in WS patients, as suggested by MRS studies, OCT pattern of retinal nerve fiber layer loss, and, in fibroblasts, by mitochondrial bioenergetics and network morphology results. However, we clearly found calcium mishandling between endoplasmic reticulum (ER) and mitochondria, which, under specific metabolic conditions of increased energy requirements and in selected tissue or cell types, may turn into a secondary mitochondrial dysfunction. Critically, we showed that Wolframin (WFS1) protein is enriched at mitochondrial-associated ER membranes and that in patient-derived fibroblasts WFS1 protein is completely absent. These findings support a loss-of-function pathogenic mechanism for missense mutations in WFS1, ultimately leading to defective calcium influx within mitochondria.


Subject(s)
Calcium/metabolism , Energy Metabolism , Mitochondria/metabolism , Wolfram Syndrome/diagnosis , Wolfram Syndrome/genetics , Adolescent , Adult , Biomarkers/blood , Child , Endoplasmic Reticulum/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Humans , Lactic Acid , Loss of Function Mutation , Magnetic Resonance Imaging , Male , Membrane Proteins/genetics , Middle Aged , Mitochondria/pathology , Mutation, Missense , Tomography, Optical Coherence , Wolfram Syndrome/etiology , Wolfram Syndrome/metabolism , Young Adult
19.
Biochim Biophys Acta Gen Subj ; 1864(4): 129523, 2020 04.
Article in English | MEDLINE | ID: mdl-31935437

ABSTRACT

BACKGROUND: Wolfram syndrome (WS), caused by mutations in WFS1 gene, is a multi-targeting disease affecting multiple organ systems. Wolframin is localized in the membrane of the endoplasmic reticulum (ER), influencing Ca2+ metabolism and ER interaction with mitochondria, but the exact role of the protein remains unclear. In this study we aimed to characterize alterations in energy metabolism in the cardiac and in the oxidative and glycolytic skeletal muscles in Wfs1-deficiency. METHODS: Alterations in the bioenergetic profiles in the cardiac and skeletal muscles of Wfs1-knock-out (KO) male mice and their wild type male littermates were determined using high resolution respirometry, quantitative RT-PCR, NMR spectroscopy, and immunofluorescence confocal microscopy. RESULTS: Oxygen consumption without ATP synthase activation (leak) was significantly higher in the glycolytic muscles of Wfs1 KO mice compared to wild types. ADP-stimulated respiration with glutamate and malate was reduced in the Wfs1-deficient cardiac as well as oxidative and glycolytic skeletal muscles. CONCLUSIONS: Wfs1-deficiency in both cardiac and skeletal muscles results in functional alterations of energy transport from mitochondria to ATP-ases. There was a substrate-dependent decrease in the maximal Complex I -linked respiratory capacity of the electron transport system in muscles of Wfs1 KO mice. Moreover, in cardiac and gastrocnemius white muscles a decrease in the function of one pathway were balanced by the increase in the activity of the parallel pathway. GENERAL SIGNIFICANCE: This work provides new insights to the muscle involvement at early stages of metabolic syndrome like WS as well as developing glucose intolerance.


Subject(s)
Energy Metabolism , Muscle, Skeletal/metabolism , Muscle, Striated/metabolism , Wolfram Syndrome/metabolism , Animals , Disease Models, Animal , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/pathology , Wolfram Syndrome/pathology
20.
Orphanet J Rare Dis ; 14(1): 279, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31796109

ABSTRACT

Wolfram syndrome is a rare multisystem disorder caused by mutations in WFS1 or CISD2 genes leading to brain structural abnormalities and neurological symptoms. These abnormalities appear in early stages of the disease. The pathogenesis of Wolfram syndrome involves abnormalities in the endoplasmic reticulum (ER) and mitochondrial dynamics, which are common features in several other neurodegenerative disorders. Mutations in WFS1 are responsible for the majority of Wolfram syndrome cases. WFS1 encodes for an endoplasmic reticulum (ER) protein, wolframin. It is proposed that wolframin deficiency triggers the unfolded protein response (UPR) pathway resulting in an increased ER stress-mediated neuronal loss. Recent neuroimaging studies showed marked alteration in early brain development, primarily characterized by abnormal white matter myelination. Interestingly, ER stress and the UPR pathway are implicated in the pathogenesis of some inherited myelin disorders like Pelizaeus-Merzbacher disease, and Vanishing White Matter disease. In addition, exploratory gene-expression network-based analyses suggest that WFS1 expression occurs preferentially in oligodendrocytes during early brain development. Therefore, we propose that Wolfram syndrome could belong to a category of neurodevelopmental disorders characterized by ER stress-mediated myelination impairment. Further studies of myelination and oligodendrocyte function in Wolfram syndrome could provide new insights into the underlying mechanisms of the Wolfram syndrome-associated brain changes and identify potential connections between neurodevelopmental disorders and neurodegeneration.


Subject(s)
Neuroimaging/methods , Wolfram Syndrome/diagnostic imaging , Wolfram Syndrome/metabolism , Animals , Brain/diagnostic imaging , Brain/metabolism , Endoplasmic Reticulum , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Unfolded Protein Response/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...