1
|
Caruso V, Rigoli L. Beyond Wolfram Syndrome 1: The WFS1 Gene's Role in Alzheimer's Disease and Sleep Disorders. Biomolecules 2024; 14:1389. [PMID: 39595565 PMCID: PMC11591713 DOI: 10.3390/biom14111389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/27/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
The WFS1 gene was first identified in Wolfram Syndrome 1 (WS1), a rare autosomal recessive genetic disorder characterized by severe and progressive neurodegenerative changes. WFS1's role in various cellular mechanisms, particularly in calcium homeostasis and the modulation of endoplasmic reticulum (ER) stress, suggests its potential involvement in the pathogenesis of Alzheimer's disease (AD) and sleep disorders. Because it is involved in maintaining ER balance, calcium signaling, and stress responses, WFS1 plays a multifaceted role in neuronal health. Numerous studies have shown that the absence or improper expression of WFS1 disrupts these cellular processes, leading to neurodegeneration and making neurons more vulnerable. In AD, WFS1 dysfunction is thought to contribute to the accumulation of amyloid-β (Aβ) plaques and tau tangles, thereby accelerating disease progression. Additionally, WFS1 plays an essential role in sleep regulation by influencing neuronal excitability and neurotransmitter release, which may explain the sleep disturbances frequently observed in neurodegenerative diseases. Due to its involvement in the pathological mechanisms of AD and sleep disorders, WFS1 is regarded as a potential early diagnostic marker for these diseases. Further research is required to fully elucidate WFS1's role in the cellular pathway, perhaps facilitating the development of new therapeutic strategies for Alzheimer's disease and sleep disorders.
Collapse
Affiliation(s)
- Valerio Caruso
- Department of Neuroscience, Psychiatric Section, Azienda Ospedaliera Universitaria Pisana (A.U.O.P.), 56126 Pisa, Italy;
| | - Luciana Rigoli
- Department of Human Pathology of Adulthood and Childhood G. Barresi, University of Messina, 98125 Messina, Italy
| |
Collapse
|
2
|
Wu L, Zhang J, Li D, Zhang Z, Ni Q, Han R, Ye L, Zhang Y, Hong J, Wang W, Ning G, Gu W. Novel WFS1 variants are associated with different diabetes phenotypes. Front Genet 2024; 15:1433060. [PMID: 39221226 PMCID: PMC11361961 DOI: 10.3389/fgene.2024.1433060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Background The WFS1 gene encodes the protein wolframin, which is crucial for maintaining endoplasmic reticulum homeostasis. Variants in this gene are predominantly associated with Wolfram syndrome and have been implicated in other disorders such as diabetes mellitus and psychiatric diseases, which increases the rate of clinical misdiagnosis. Methods Patients were diagnosed with early-onset unclassified diabetes according to their clinical and laboratory data. We performed whole-exome sequencing (WES) in 165 patients, interpreting variants according to the American College of Medical Genetics/Association for Molecular Pathology (ACMG/AMP) 2015 guidelines. Variant verification was done by Sanger sequencing. In vitro experiments were conducted to evaluate the effects of WFS1 compound heterozygous variants. Results We identified WFS1 compound heterozygous variants (p.A214fs*74/p.F329I and p.I427S/p.I304T) in two patients with Wolfram Syndrome-Like disorders (WSLD). Both WFS1 compound heterozygous variants were associated with increased ER stress, reduced cell viability, and decreased SERCA2b mRNA levels. Additionally, pathogenic or likely pathogenic WFS1 heterozygous variants were identified in the other three patients. Conclusion Our results underscore the importance of early genetic testing for diagnosing young-onset diabetes and highlight the clinical relevance of WFS1 variants in increasing ER stress and reducing cell viability. Incorporating these genetic insights into clinical practice can reduce misdiagnoses and improve treatment strategies for related disorders.
Collapse
Affiliation(s)
- Lei Wu
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Juan Zhang
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Danjie Li
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Zhongyun Zhang
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Qicheng Ni
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rulai Han
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Ye
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yifei Zhang
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Hong
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqing Wang
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqiong Gu
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Jung J, Jang SH, Won D, Gee HY, Choi JY, Jung J. Clinical Characteristics and Audiological Profiles of Patients with Pathogenic Variants of WFS1. J Clin Med 2024; 13:4851. [PMID: 39200993 PMCID: PMC11355604 DOI: 10.3390/jcm13164851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/06/2024] [Accepted: 08/14/2024] [Indexed: 09/02/2024] Open
Abstract
Background: Mutations in Wolfram syndrome 1 (WFS1) cause Wolfram syndrome and autosomal dominant non-syndromic hearing loss DFNA6/14/38. To date, more than 300 pathogenic variants of WFS1 have been identified. Generally, the audiological phenotype of Wolfram syndrome or DFNA6/14/38 is characterized by low-frequency hearing loss; however, this phenotype is largely variable. Hence, there is a need to better understand the diversity in audiological and vestibular profiles associated with WFS1 variants, as this can have significant implications for diagnosis and management. This study aims to investigate the clinical characteristics, audiological phenotypes, and vestibular function in patients with DFNA6/14/38. Methods: Whole-exome or targeted deafness gene panel sequencing was performed to confirm the pathogenic variants in patients with genetic hearing loss. Results: We identified nine independent families with affected individuals who carried a heterozygous pathogenic variant of WFS1. The onset of hearing loss varied from the first to the fifth decade. On a pure-tone audiogram, hearing loss was symmetrical, and the severity ranged from mild to severe. Notably, either both low-frequency and high-frequency or all-frequency-specific hearing loss was observed. However, hearing loss was non-progressive in all types. In addition, vestibular impairment was identified in patients with DFNA6/14/38, indicating that impaired WFS1 may also affect the vestibular organs. Conclusions: Diverse audiological and vestibular profiles were observed in patients with pathogenic variants of WFS1. These findings highlight the importance of comprehensive audiological and vestibular assessments in patients with WFS1 mutations for accurate diagnosis and management.
Collapse
Affiliation(s)
- Joonho Jung
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (J.J.); (S.H.J.); (J.Y.C.)
| | - Seung Hyun Jang
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (J.J.); (S.H.J.); (J.Y.C.)
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
| | - Dongju Won
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
| | - Heon Yung Gee
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
| | - Jae Young Choi
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (J.J.); (S.H.J.); (J.Y.C.)
| | - Jinsei Jung
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (J.J.); (S.H.J.); (J.Y.C.)
| |
Collapse
|
4
|
Caruso V, Raia A, Rigoli L. Wolfram Syndrome 1: A Neuropsychiatric Perspective on a Rare Disease. Genes (Basel) 2024; 15:984. [PMID: 39202345 PMCID: PMC11353439 DOI: 10.3390/genes15080984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 09/03/2024] Open
Abstract
Wolfram syndrome 1 (WS1) is an uncommon autosomal recessive neurological disorder that is characterized by diabetes insipidus, early-onset non-autoimmune diabetes mellitus, optic atrophy, and deafness (DIDMOAD). Other clinical manifestations are neuropsychiatric symptoms, urinary tract alterations, and endocrinological disorders. The rapid clinical course of WS1 results in death by the age of 30. Severe brain atrophy leads to central respiratory failure, which is the main cause of death in WS1 patients. Mutations in the WFS1 gene, located on chromosome 4p16, account for approximately 90% of WS1 cases. The gene produces wolframin, a transmembrane glycoprotein widely distributed and highly expressed in retinal, neural, and muscular tissues. Wolframin plays a crucial role in the regulation of apoptosis, insulin signaling, and ER calcium homeostasis, as well as the ER stress response. WS1 has been designated as a neurodegenerative and neurodevelopmental disorder due to the numerous abnormalities in the ER stress-mediated system. WS1 is a devastating neurodegenerative disease that affects patients and their families. Early diagnosis and recognition of the initial clinical signs may slow the disease's progression and improve symptomatology. Moreover, genetic counseling should be provided to the patient's relatives to extend multidisciplinary care to their first-degree family members. Regrettably, there are currently no specific drugs for the therapy of this fatal disease. A better understanding of the etiology of WS1 will make possible the development of new therapeutic approaches that may enhance the life expectancy of patients. This review will examine the pathogenetic mechanisms, development, and progression of neuropsychiatric symptoms commonly associated with WS1. A thorough understanding of WS1's neurophysiopathology is critical for achieving the goal of improving patients' quality of life and life expectancy.
Collapse
Affiliation(s)
- Valerio Caruso
- Department of Neuroscience, Psychiatric Section, Azienda Ospedaliera Universitaria Pisana (A.U.O.P.), 56126 Pisa, Italy;
| | - Accursio Raia
- Department of Neuroscience, Psychiatric Section, Azienda Ospedaliera Universitaria Pisana (A.U.O.P.), 56126 Pisa, Italy;
| | - Luciana Rigoli
- Department of Human Pathology of Adulthood and Childhood G. Barresi, University of Messina, 98125 Messina, Italy
| |
Collapse
|
5
|
Zhou X, Xiao Q, Liu Y, Chen S, Xu X, Zhang Z, Hong Y, Shao J, Chen Y, Chen Y, Wang L, Yang F, Tu J. Astrocyte-mediated regulation of BLA WFS1 neurons alleviates risk-assessment deficits in DISC1-N mice. Neuron 2024; 112:2197-2217.e7. [PMID: 38642554 DOI: 10.1016/j.neuron.2024.03.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 02/10/2024] [Accepted: 03/27/2024] [Indexed: 04/22/2024]
Abstract
Assessing and responding to threats is vital in everyday life. Unfortunately, many mental illnesses involve impaired risk assessment, affecting patients, families, and society. The brain processes behind these behaviors are not well understood. We developed a transgenic mouse model (disrupted-in-schizophrenia 1 [DISC1]-N) with a disrupted avoidance response in risky settings. Our study utilized single-nucleus RNA sequencing and path-clamp coupling with real-time RT-PCR to uncover a previously undescribed group of glutamatergic neurons in the basolateral amygdala (BLA) marked by Wolfram syndrome 1 (WFS1) expression, whose activity is modulated by adjacent astrocytes. These neurons in DISC1-N mice exhibited diminished firing ability and impaired communication with the astrocytes. Remarkably, optogenetic activation of these astrocytes reinstated neuronal excitability via D-serine acting on BLAWFS1 neurons' NMDA receptors, leading to improved risk-assessment behavior in the DISC1-N mice. Our findings point to BLA astrocytes as a promising target for treating risk-assessment dysfunctions in mental disorders.
Collapse
Affiliation(s)
- Xinyi Zhou
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Department of Neurology, The Second Clinical Medical College, Jinan University, Shenzhen People's Hospital, Shenzhen 518020, China; The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
| | - Qian Xiao
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yaohui Liu
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No. 88 East Wenhua Road, Jinan 250014, China
| | - Shuai Chen
- University of Chinese of Academy of Sciences, Beijing 100049, China
| | - Xirong Xu
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese of Academy of Sciences, Beijing 100049, China
| | - Zhigang Zhang
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yuchuan Hong
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese of Academy of Sciences, Beijing 100049, China
| | - Jie Shao
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Department of Neurology, The Second Clinical Medical College, Jinan University, Shenzhen People's Hospital, Shenzhen 518020, China; The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
| | - Yuewen Chen
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese of Academy of Sciences, Beijing 100049, China; Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yu Chen
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese of Academy of Sciences, Beijing 100049, China; Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Liping Wang
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese of Academy of Sciences, Beijing 100049, China; Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| | - Fan Yang
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese of Academy of Sciences, Beijing 100049, China; Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| | - Jie Tu
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese of Academy of Sciences, Beijing 100049, China; Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| |
Collapse
|
6
|
Li S, Li X, Qu J. A Wolfram-like syndrome family: Case report. Eur J Ophthalmol 2024; 34:NP51-NP57. [PMID: 38470317 DOI: 10.1177/11206721241237552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
BACKGROUND Wolfram-like syndrome (WFLS) is an autosomal dominant inherited disease characterized by a single heterozygous pathogenic variant in the WFS1 gene. Its clinical presentation is similar to autosomal recessive Wolfram syndrome. CASE PRESENTATION We reported a case of a 10-year-old boy and his family members who initially experienced hearing impairment (HI), followed by optic atrophy. Genetic testing revealed the presence of a WFS1 variant (chr4-6302385 exon8 NM_006005.3: c.2590G > A, p. Glu864Lys). CONCLUSION Wolfram-like syndrome, a rare neurodegenerative genetic disorder, manifested as deafness, optic atrophy, and diabetes mellitus. There hasn't been a definite treatment yet. Early identification of the variant in the WFS1 gene is beneficial for genetic counseling.
Collapse
Affiliation(s)
- Siying Li
- Department of Ophthalmology, Peking University People's Hospital, Beijing, China
- Eye Diseases and Optometry Institute, Beijing, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center, Beijing, China
| | - Xiaoxin Li
- Department of Ophthalmology, Peking University People's Hospital, Beijing, China
- Eye Diseases and Optometry Institute, Beijing, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center, Beijing, China
| | - Jinfeng Qu
- Department of Ophthalmology, Peking University People's Hospital, Beijing, China
- Eye Diseases and Optometry Institute, Beijing, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center, Beijing, China
| |
Collapse
|
7
|
Jurca AD, Galea-Holhos LB, Jurca AA, Atasie D, Petchesi CD, Severin E, Jurca CM. Wolfram Syndrome Type I Case Report and Review-Focus on Early Diagnosis and Genetic Variants. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1064. [PMID: 39064493 PMCID: PMC11278941 DOI: 10.3390/medicina60071064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 06/23/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024]
Abstract
Background and Objectives: Wolfram syndrome type 1 (OMIM# 222300; ORPHAcode 3463) is an extremely rare autosomal recessive syndrome with a 25% recurrence risk in children. It is characterized by the presence of juvenile-onset diabetes mellitus (DM), progressive optic atrophy (OA), diabetes insipidus (DI), and sensorineural deafness (D), often referred to by the acronym DIDMOAD. It is a severe neurodegenerative disease with a life expectancy of 39 years, with death occurring due to cerebral atrophy. For a positive diagnosis, the presence of diabetes mellitus and optic nerve atrophy is sufficient. The disease occurs because of pathogenic variants in the WFS1 gene. The aim of this article is to present a case report of Wolfram Syndrome Type I, alongside a review of genetic variants, clinical manifestations, diagnosis, therapy, and long-term management. Emphasizing the importance of early diagnosis and a multidisciplinary approach, the study aims to enhance understanding and improve outcomes for patients with this complex syndrome. Materials and Methods: A case of a 28-year-old patient diagnosed with DM at the age of 6 and with progressive optic atrophy at 26 years old is presented. Molecular diagnosis revealed the presence of a heterozygous nonsense variant WFS1 c.1943G>A (p.Trp648*), and a heterozygous missense variant WFS1 c.1675G>C (p.Ala559Pro). Results: The molecular diagnosis of the patient confirmed the presence of a heterozygous nonsense variant and a heterozygous missense variant in the WFS1 gene, correlating with the clinical presentation of Wolfram syndrome type 1. Both allelic variants found in our patient have been previously described in other patients, whilst this combination has not been described before. Conclusions: This case report and review underscores the critical role of early recognition and diagnosis in Wolfram syndrome, facilitated by genetic testing. By identifying pathogenic variants in the WFS1 gene, genetic testing not only confirms diagnosis but also guides clinical management and informs genetic counseling for affected families. Timely intervention based on genetic insights can potentially reduce the progressive multisystem manifestations of the syndrome, thereby improving the quality of life and outcomes for patients.
Collapse
Affiliation(s)
- Alexandru Daniel Jurca
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410081 Oradea, Romania; (A.D.J.); (C.D.P.); (C.M.J.)
| | - Larisa Bianca Galea-Holhos
- Department of Morphological Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410081 Oradea, Romania
| | | | - Diter Atasie
- Departament II Medical Clinic, Faculty of Medicine, University “Lucian Blaga of Sibiu”, Lucian Blaga Street 2A, 550169 Sibiu, Romania;
| | - Codruta Diana Petchesi
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410081 Oradea, Romania; (A.D.J.); (C.D.P.); (C.M.J.)
- Regional Center of Medical Genetics Bihor, County Emergency Clinical Hospital Oradea (Part of ERN-ITHACA), 410469 Oradea, Romania
| | - Emilia Severin
- Genetics Department, “Carol Davila” University of Medicine and Pharmacy, 020027 Bucharest, District 2, Romania
| | - Claudia Maria Jurca
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410081 Oradea, Romania; (A.D.J.); (C.D.P.); (C.M.J.)
- Regional Center of Medical Genetics Bihor, County Emergency Clinical Hospital Oradea (Part of ERN-ITHACA), 410469 Oradea, Romania
| |
Collapse
|
8
|
Hu R, Chen X, Su Q, Wang Z, Wang X, Gong M, Xu M, Le R, Gao Y, Dai P, Zhang ZN, Shao L, Li W. ISR inhibition reverses pancreatic β-cell failure in Wolfram syndrome models. Cell Death Differ 2024; 31:322-334. [PMID: 38321214 PMCID: PMC10923889 DOI: 10.1038/s41418-024-01258-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/08/2024] Open
Abstract
Pancreatic β-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 β cell specific Wfs1 knockout were applied to model β-cell failure in WS. We charted a high-resolution roadmap with single-cell RNA-seq (scRNA-seq) to investigate pathogenesis for WS β-cell failure, revealing two distinct cellular fates along pseudotime trajectory: maturation and stress branches. WFS1 deficiency disrupted β-cell fate trajectory toward maturation and directed it towards stress trajectory, ultimately leading to β-cell failure. Notably, further investigation of the stress trajectory identified activated integrated stress response (ISR) as a crucial mechanism underlying WS β-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 β-cell failure in WFS1-deficient SC-islets. We further validated therapeutic efficacy in vivo with β-cell specific Wfs1 knockout mice. Altogether, our study provides novel insights into WS pathogenesis and offers a strategy targeting ISR to treat WS diabetes.
Collapse
Affiliation(s)
- Rui Hu
- Medical Innovation Center and State Key Laboratory of Cardiology, Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xiangyi Chen
- Medical Innovation Center and State Key Laboratory of Cardiology, Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Qiang Su
- Medical Innovation Center and State Key Laboratory of Cardiology, Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Zhaoyue Wang
- Medical Innovation Center and State Key Laboratory of Cardiology, Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xushu Wang
- Medical Innovation Center and State Key Laboratory of Cardiology, Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Mengting Gong
- Medical Innovation Center and State Key Laboratory of Cardiology, Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Minglu Xu
- Medical Innovation Center and State Key Laboratory of Cardiology, Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Rongrong Le
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yawei Gao
- Medical Innovation Center and State Key Laboratory of Cardiology, Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Peng Dai
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Zhen-Ning Zhang
- Medical Innovation Center and State Key Laboratory of Cardiology, Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Li Shao
- Department of VIP Clinic, Shanghai East Hospital, Tongji University School of Medicine, No. 1800 Yuntai Road, Pudong District, Shanghai, 200123, China.
| | - Weida Li
- Medical Innovation Center and State Key Laboratory of Cardiology, Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
- Reg-Verse Therapeutics (Shanghai) Co. Ltd., Shanghai, 200120, China.
| |
Collapse
|
9
|
Frontino G, Di Tonno R, Stancampiano MR, Arrigoni F, Rigamonti A, Morotti E, Canarutto D, Bonfanti R, Russo G, Barera G, Piemonti L. Paediatric Wolfram syndrome Type 1: should gonadal dysfunction be part of the diagnostic criteria? Front Endocrinol (Lausanne) 2023; 14:1155644. [PMID: 37383390 PMCID: PMC10294676 DOI: 10.3389/fendo.2023.1155644] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/26/2023] [Indexed: 06/30/2023] Open
Abstract
Aims Wolfram Syndrome Spectrum Disorder (WFS1-SD), in its "classic" form, is a rare autosomal recessive disease with poor prognosis and wide phenotypic spectrum. Insulin dependent diabetes mellitus (DM), optic atrophy (OA) diabetes insipidus (DI) and sensorineural deafness (D) are the main features of WFS1-SD. Gonadal dysfunction (GD) has been described mainly in adults with variable prevalence and referred to as a minor clinical feature. This is the first case series investigating gonadal function in a small cohort of paediatric patients affected by WFS1-SD. Methods Gonadal function was investigated in eight patients (3 male and 5 female) between 3 and 16 years of age. Seven patients have been diagnosed with classic WFS1-SD and one with non-classic WFS1-SD. Gonadotropin and sex hormone levels were monitored, as well as markers of gonadal reserve (inhibin-B and anti-Mullerian hormone). Pubertal progression was assessed according to Tanner staging. Results Primary hypogonadism was diagnosed in 50% of patients (n=4), more specifically 67% (n=2) of males and 40% of females (n=2). Pubertal delay was observed in one female patient. These data confirm that gonadal dysfunction may be a frequent and underdiagnosed clinical feature in WFS1-SD. Conclusions GD may represent a frequent and earlier than previously described feature in WFS1-SD with repercussions on morbidity and quality of life. Consequently, we suggest that GD should be included amongst clinical diagnostic criteria for WFS1-SD, as has already been proposed for urinary dysfunction. Considering the heterogeneous and elusive presentation of WFS1-SD, this clinical feature may assist in an earlier diagnosis and timely follow-up and care of treatable associated diseases (i.e. insulin and sex hormone replacement) in these young patients.
Collapse
Affiliation(s)
- Giulio Frontino
- Department of Pediatrics, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) San Raffaele Hospital, Milan, Italy
- Diabetes Research Institute, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) San Raffaele Hospital, Milan, Italy
| | - Raffaella Di Tonno
- Department of Pediatrics, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) San Raffaele Hospital, Milan, Italy
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Marianna Rita Stancampiano
- Department of Pediatrics, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) San Raffaele Hospital, Milan, Italy
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Francesca Arrigoni
- Department of Pediatrics, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) San Raffaele Hospital, Milan, Italy
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Andrea Rigamonti
- Department of Pediatrics, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) San Raffaele Hospital, Milan, Italy
- Diabetes Research Institute, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) San Raffaele Hospital, Milan, Italy
| | - Elisa Morotti
- Department of Pediatrics, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) San Raffaele Hospital, Milan, Italy
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Daniele Canarutto
- San Raffaele Telethon Institute for Gene Therapy, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) San Raffaele Hospital, Milan, Italy
- Pediatric Immunohematology Unit and BMT Program, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) San Raffaele Hospital, Milan, Italy
| | - Riccardo Bonfanti
- Department of Pediatrics, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) San Raffaele Hospital, Milan, Italy
- Diabetes Research Institute, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) San Raffaele Hospital, Milan, Italy
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Gianni Russo
- Department of Pediatrics, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) San Raffaele Hospital, Milan, Italy
| | - Graziano Barera
- Department of Pediatrics, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) San Raffaele Hospital, Milan, Italy
| | - Lorenzo Piemonti
- Diabetes Research Institute, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) San Raffaele Hospital, Milan, Italy
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
10
|
Su Q, Yuan F, Li X, Wang X, Yang K, Shao L, Li W. Wfs1 loss-of-function disrupts the composition of mouse pancreatic endocrine cells from birth and impairs Glut2 localization to cytomembrane in pancreatic β cells. Biochem Biophys Res Commun 2023; 658:80-87. [PMID: 37027908 DOI: 10.1016/j.bbrc.2023.03.074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023]
Abstract
Wfs1 is an endoplasmic reticulum (ER) membrane located protein highly expressed in pancreatic β cells and brain. Wfs1 deficiency causes adult pancreatic β cells dysfunction following β cells apoptosis. Previous studies mainly focus on the Wfs1 function in adult mouse pancreatic β cells. However, whether Wfs1 loss-of-function impairs mouse pancreatic β 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 β 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 β 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 β cells.
Collapse
Affiliation(s)
- Qiang Su
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Fei Yuan
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xiaobo Li
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xuan Wang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Kaijiang Yang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Li Shao
- Department of VIP Clinic, Shanghai East Hospital, Tongji University School of Medicine, No. 1800 Yuntai Road, Pudong District, Shanghai, 200092, China.
| | - Weida Li
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China; Reg-Verse Therapeutics (Shanghai) Co. Ltd., Shanghai, 200120, China.
| |
Collapse
|
11
|
Lim HD, Lee SM, Yun YJ, Lee DH, Lee JH, Oh SH, Lee SY. WFS1 autosomal dominant variants linked with hearing loss: update on structural analysis and cochlear implant outcome. BMC Med Genomics 2023; 16:79. [PMID: 37041640 PMCID: PMC10088283 DOI: 10.1186/s12920-023-01506-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 04/02/2023] [Indexed: 04/13/2023] Open
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.
Collapse
Affiliation(s)
- Hui Dong Lim
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul, Republic of Korea
| | - So Min Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul, Republic of Korea
| | - Ye Jin Yun
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dae Hee Lee
- CTCELLS, Inc, 21, Yuseong-daero, 1205beon-gil, Yuseong-gu, Daejeon, Republic of Korea
| | - Jun Ho Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul, Republic of Korea
| | - Seung-Ha Oh
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sang-Yeon Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul, Republic of Korea.
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
| |
Collapse
|
12
|
Chan YH, Tsai CY, Ho CH, Lu YC, Lin PH, Chen TC, Chen YT, Huang CY, Liu TC, Hsu CJ, Wu CC. Generation of induced pluripotent stem cells (IBMSi027-A) from a patient with hearing loss carrying WFS1 c.2051C > T (p.Ala684Val) variant. Stem Cell Res 2023; 69:103068. [PMID: 36933359 DOI: 10.1016/j.scr.2023.103068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 03/07/2023] [Accepted: 03/11/2023] [Indexed: 03/18/2023] Open
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.
Collapse
Affiliation(s)
- Yen-Hui Chan
- Department of Otolaryngology, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan; Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan
| | - Cheng-Yu Tsai
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan; Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chang-Han Ho
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ying-Chang Lu
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan
| | - Pei-Hsuan Lin
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ta-Ching Chen
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - You-Tzung Chen
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Cheng-Yen Huang
- Gene Knockout/in Cell Line Modeling Core, Human Disease Modeling Center, First Core Laboratory, Branch Office of Research and Development, College of Medicine, National Taiwan University, Taiwan
| | - Tien-Chen Liu
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chuan-Jen Hsu
- Department of Otolaryngology, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan.
| | - Chen-Chi Wu
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
13
|
Serbis A, Rallis D, Giapros V, Galli-Tsinopoulou A, Siomou E. Wolfram Syndrome 1: A Pediatrician's and Pediatric Endocrinologist's Perspective. Int J Mol Sci 2023; 24:ijms24043690. [PMID: 36835101 PMCID: PMC9960967 DOI: 10.3390/ijms24043690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/21/2023] [Accepted: 02/10/2023] [Indexed: 02/15/2023] Open
Abstract
Wolfram syndrome 1 (WS1) is a rare autosomal recessive neurodegenerative disease caused by mutations in WFS1 and WFS2 genes that produce wolframin, a protein involved in endoplasmic reticulum calcium homeostasis and cellular apoptosis. Its main clinical features are diabetes insipidus (DI), early-onset non-autoimmune insulin-dependent diabetes mellitus (DM), gradual loss of vision due to optic atrophy (OA) and deafness (D), hence the acronym DIDMOAD. Several other features from different systems have been reported such as urinary tract, neurological, and psychiatric abnormalities. In addition, endocrine disorders that can appear during childhood and adolescence include primary gonadal atrophy and hypergonadotropic hypogonadism in males and menstrual cycle abnormalities in females. Further, anterior pituitary dysfunction with deficient GH and/or ACTH production have been described. Despite the lack of specific treatment for the disease and its poor life expectancy, early diagnosis and supportive care is important for timely identifying and adequately managing its progressive symptoms. The current narrative review focuses on the pathophysiology and the clinical features of the disease, with a special emphasis on its endocrine abnormalities that appear during childhood and adolescence. Further, therapeutic interventions that have been proven to be effective in the management of WS1 endocrine complications are discussed.
Collapse
Affiliation(s)
- Anastasios Serbis
- Department of Pediatrics, School of Medicine, University of Ioannina, 451 10 Ioannina, Greece
- Correspondence:
| | - Dimitrios Rallis
- Neonatal Intensive Care Unit, School of Medicine, University of Ioannina, 451 10 Ioannina, Greece
| | - Vasileios Giapros
- Neonatal Intensive Care Unit, School of Medicine, University of Ioannina, 451 10 Ioannina, Greece
| | - Assimina Galli-Tsinopoulou
- Second Department of Pediatrics, School of Medicine, Faculty of Health Sciences, AHEPA University General Hospital, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Ekaterini Siomou
- Department of Pediatrics, School of Medicine, University of Ioannina, 451 10 Ioannina, Greece
| |
Collapse
|
14
|
Yuan F, Li Y, Hu R, Gong M, Chai M, Ma X, Cha J, Guo P, Yang K, Li M, Xu M, Ma Q, Su Q, Zhang C, Sheng Z, Wu H, Wang Y, Yuan W, Bian S, Shao L, Zhang R, Li K, Shao Z, Zhang ZN, Li W. Modeling disrupted synapse formation in wolfram syndrome using hESCs-derived neural cells and cerebral organoids identifies Riluzole as a therapeutic molecule. Mol Psychiatry 2023; 28:1557-1570. [PMID: 36750736 DOI: 10.1038/s41380-023-01987-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/18/2023] [Accepted: 01/30/2023] [Indexed: 02/09/2023]
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.
Collapse
Affiliation(s)
- Fei Yuan
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.,Tsingtao Advanced Research Institute, Tongji University, Qingdao, 266071, China
| | - Yana Li
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Rui Hu
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.,Tsingtao Advanced Research Institute, Tongji University, Qingdao, 266071, China
| | - Mengting Gong
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.,Tsingtao Advanced Research Institute, Tongji University, Qingdao, 266071, China
| | - Mengyao Chai
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.,Tsingtao Advanced Research Institute, Tongji University, Qingdao, 266071, China
| | - Xuefei Ma
- QuietD Biotechnology, Ltd., Shanghai, 201210, China
| | - Jiaxue Cha
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Pan Guo
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Kaijiang Yang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.,Tsingtao Advanced Research Institute, Tongji University, Qingdao, 266071, China
| | - Mushan Li
- Department of Statistics, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Minglu Xu
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.,Tsingtao Advanced Research Institute, Tongji University, Qingdao, 266071, China
| | - Qing Ma
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.,Tsingtao Advanced Research Institute, Tongji University, Qingdao, 266071, China
| | - Qiang Su
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.,Tsingtao Advanced Research Institute, Tongji University, Qingdao, 266071, China
| | - Chuan Zhang
- School of Medicine, Tongji University, Shanghai, 200092, China
| | - Zhejin Sheng
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.,Tsingtao Advanced Research Institute, Tongji University, Qingdao, 266071, China
| | - Heng Wu
- Department of Psychosomatic Medicine, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200092, China
| | - Yuan Wang
- Department of Neurology and Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and National Collaborative Innovation Center, Chengdu, Sichuan, 610041, China
| | - Wen Yuan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Shan Bian
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.,Tsingtao Advanced Research Institute, Tongji University, Qingdao, 266071, China
| | - Li Shao
- Department of VIP Clinic, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092, China
| | - Ru Zhang
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Kaicheng Li
- QuietD Biotechnology, Ltd., Shanghai, 201210, China
| | - Zhen Shao
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Zhen-Ning Zhang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China. .,Tsingtao Advanced Research Institute, Tongji University, Qingdao, 266071, China.
| | - Weida Li
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China. .,Tsingtao Advanced Research Institute, Tongji University, Qingdao, 266071, China. .,Reg-Verse Therapeutics (Shanghai) Co. Ltd., Shanghai, 200120, China.
| |
Collapse
|
15
|
Wang Z, Wang X, Shi L, Cai Y, Hu B. Wolfram syndrome 1b mutation suppresses Mauthner-cell axon regeneration via ER stress signal pathway. Acta Neuropathol Commun 2022; 10:184. [PMID: 36527091 PMCID: PMC9758940 DOI: 10.1186/s40478-022-01484-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 12/23/2022] Open
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.
Collapse
Affiliation(s)
- Zongyi Wang
- grid.59053.3a0000000121679639Hefei National Research Center for Physical Sciences at the Microscale, Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026 China
| | - Xinliang Wang
- grid.59053.3a0000000121679639Hefei National Research Center for Physical Sciences at the Microscale, Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026 China
| | - Lingyu Shi
- grid.59053.3a0000000121679639Hefei National Research Center for Physical Sciences at the Microscale, Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026 China
| | - Yuan Cai
- grid.59053.3a0000000121679639Hefei National Research Center for Physical Sciences at the Microscale, Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026 China ,grid.59053.3a0000000121679639First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026 China
| | - Bing Hu
- grid.59053.3a0000000121679639Hefei National Research Center for Physical Sciences at the Microscale, Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026 China ,grid.59053.3a0000000121679639Research Institute of Frontier Cross Science and Biomedical Sciences, Hefei Comprehensive National Science Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026 China
| |
Collapse
|
16
|
Xie HB, Yan C, Adeola AC, Wang K, Huang CP, Xu MM, Qiu Q, Yin X, Fan CY, Ma YF, Yin TT, Gao Y, Deng JK, Okeyoyin AO, Oluwole OO, Omotosho O, Okoro VMO, Omitogun OG, Dawuda PM, Olaogun SC, Nneji LM, Ayoola AO, Sanke OJ, Luka PD, Okoth E, Lekolool I, Mijele D, Bishop RP, Han J, Wang W, Peng MS, Zhang YP. African Suid Genomes Provide Insights into the Local Adaptation to Diverse African Environments. Mol Biol Evol 2022; 39:6840307. [PMID: 36413509 PMCID: PMC9733430 DOI: 10.1093/molbev/msac256] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 10/21/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
African wild suids consist of several endemic species that represent ancient members of the family Suidae and have colonized diverse habitats on the African continent. However, limited genomic resources for African wild suids hinder our understanding of their evolution and genetic diversity. In this study, we assembled high-quality genomes of a common warthog (Phacochoerus africanus), a red river hog (Potamochoerus porcus), as well as an East Asian Diannan small-ear pig (Sus scrofa). Phylogenetic analysis showed that common warthog and red river hog diverged from their common ancestor around the Miocene/Pliocene boundary, putatively predating their entry into Africa. We detected species-specific selective signals associated with sensory perception and interferon signaling pathways in common warthog and red river hog, respectively, which contributed to their local adaptation to savannah and tropical rainforest environments, respectively. The structural variation and evolving signals in genes involved in T-cell immunity, viral infection, and lymphoid development were identified in their ancestral lineage. Our results provide new insights into the evolutionary histories and divergent genetic adaptations of African suids.
Collapse
Affiliation(s)
| | | | | | | | | | - Ming-Min Xu
- State Key Laboratory of Genetic Resources and Evolution & Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China
| | - Qiang Qiu
- School of Ecology and Environment, Northwestern Polytechnical University, Xi’an 710129, China
| | - Xue Yin
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Chen-Yu Fan
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Yun-Fei Ma
- State Key Laboratory of Genetic Resources and Evolution & Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China
| | - Ting-Ting Yin
- State Key Laboratory of Genetic Resources and Evolution & Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China
| | - Yun Gao
- State Key Laboratory of Genetic Resources and Evolution & Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China
| | - Jia-Kun Deng
- State Key Laboratory of Genetic Resources and Evolution & Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China
| | - Agboola O Okeyoyin
- National Park Service Headquarter, Federal Capital Territory, Abuja 900108, Nigeria
| | - Olufunke O Oluwole
- Institute of Agricultural Research and Training, Obafemi Awolowo University, Ibadan, Nigeria
| | - Oladipo Omotosho
- Department of Veterinary Medicine, University of Ibadan, Ibadan 200005, Nigeria
| | - Victor M O Okoro
- Department of Animal Science and Technology, School of Agriculture and Agricultural Technology, Federal University of Technology, Owerri 460114, Nigeria
| | - Ofelia G Omitogun
- Department of Animal Sciences, Obafemi Awolowo University, Ile-Ife 220282, Nigeria
| | - Philip M Dawuda
- Department of Veterinary Surgery and Theriogenology, College of Veterinary Medicine, University of Agriculture Makurdi, Makurdi 970001, Nigeria
| | - Sunday C Olaogun
- Department of Veterinary Medicine, University of Ibadan, Ibadan 200005, Nigeria
| | - Lotanna M Nneji
- State Key Laboratory of Genetic Resources and Evolution & Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China,Sino-Africa Joint Research Center, Chinese Academy of Sciences, Kunming 650204, China
| | - Adeola O Ayoola
- State Key Laboratory of Genetic Resources and Evolution & Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China,Sino-Africa Joint Research Center, Chinese Academy of Sciences, Kunming 650204, China
| | - Oscar J Sanke
- Taraba State Ministry of Agriculture and Natural Resources, Jalingo 660213, Nigeria
| | - Pam D Luka
- National Veterinary Research Institute, Vom 930103, Nigeria
| | - Edward Okoth
- International Livestock Research Institute (ILRI), Nairobi 00100, Kenya
| | | | | | - Richard P Bishop
- International Livestock Research Institute (ILRI), Nairobi 00100, Kenya
| | | | - Wen Wang
- Corresponding authors: E-mails: ; ; ;
| | | | | |
Collapse
|
17
|
Majander A, Jurkute N, Burté F, Brock K, João C, Huang H, Neveu MM, Chan CM, Duncan HJ, Kelly S, Burkitt-Wright E, Khoyratty F, Lai YT, Subash M, Chinnery PF, Bitner-Glindzicz M, Arno G, Webster AR, Moore AT, Michaelides M, Stockman A, Robson AG, Yu-Wai-Man P. WFS1-Associated Optic Neuropathy: Genotype-Phenotype Correlations and Disease Progression. Am J Ophthalmol 2022; 241:9-27. [PMID: 35469785 DOI: 10.1016/j.ajo.2022.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 04/11/2022] [Accepted: 04/13/2022] [Indexed: 11/21/2022]
Abstract
OBJECTIVE To evaluate the pattern of vision loss and genotype-phenotype correlations in WFS1-associated optic neuropathy (WON). DESIGN Multicenter cohort study. METHODS The study involved 37 patients with WON carrying pathogenic or candidate pathogenic WFS1 variants. Genetic and clinical data were retrieved from the medical records. Thirteen patients underwent additional comprehensive ophthalmologic assessment. Deep phenotyping involved visual electrophysiology and advanced psychophysical testing with a complementary metabolomic study. MAIN OUTCOME MEASURES WFS1 variants, functional and structural optic nerve and retinal parameters, and metabolomic profile. RESULTS Twenty-two recessive and 5 dominant WFS1 variants were identified. Four variants were novel. All WFS1 variants caused loss of macular retinal ganglion cells (RGCs) as assessed by optical coherence tomography (OCT) and visual electrophysiology. Advanced psychophysical testing indicated involvement of the major RGC subpopulations. Modeling of vision loss showed an accelerated rate of deterioration with increasing age. Dominant WFS1 variants were associated with abnormal reflectivity of the outer plexiform layer (OPL) on OCT imaging. The dominant variants tended to cause less severe vision loss compared with recessive WFS1 variants, which resulted in more variable phenotypes ranging from isolated WON to severe multisystem disease depending on the WFS1 alleles. The metabolomic profile included markers seen in other neurodegenerative diseases and type 1 diabetes mellitus. CONCLUSIONS WFS1 variants result in heterogenous phenotypes influenced by the mode of inheritance and the disease-causing alleles. Biallelic WFS1 variants cause more variable, but generally more severe, vision and RGC loss compared with heterozygous variants. Abnormal cleftlike lamination of the OPL is a distinctive OCT feature that strongly points toward dominant WON.
Collapse
Affiliation(s)
- Anna Majander
- From the UCL Institute of Ophthalmology (A.M., N.J., C.J., M.M.N., C.M.C., M.S., G.A., A.R.W., A.T.M., M.M., A.S., A.G.R., P.Y.-W.-M.), London, United Kingdom; Moorfields Eye Hospital (A.M., N.J., M.M.N., C.M.C., G.A., A.R.W., A.T.M., M.M., A.G.R., P.Y.-W.-M.), London, United Kingdom; Department of Ophthalmology, Helsinki University Hospital, University of Helsinki (A.M.), Helsinki, Finland.
| | - Neringa Jurkute
- From the UCL Institute of Ophthalmology (A.M., N.J., C.J., M.M.N., C.M.C., M.S., G.A., A.R.W., A.T.M., M.M., A.S., A.G.R., P.Y.-W.-M.), London, United Kingdom; Moorfields Eye Hospital (A.M., N.J., M.M.N., C.M.C., G.A., A.R.W., A.T.M., M.M., A.G.R., P.Y.-W.-M.), London, United Kingdom
| | - Florence Burté
- Biosciences Institute, International Centre for Life, Newcastle University (F.B.), Newcastle upon Tyne, United Kingdom
| | - Kristian Brock
- Cancer Research UK Clinical Trials Unit, University of Birmingham (K.B.), Birmingham, United Kingdom
| | - Catarina João
- From the UCL Institute of Ophthalmology (A.M., N.J., C.J., M.M.N., C.M.C., M.S., G.A., A.R.W., A.T.M., M.M., A.S., A.G.R., P.Y.-W.-M.), London, United Kingdom
| | - Houbin Huang
- Hainan Hospital of the General Hospital of Chinese People's Liberation Army (H.H.), Sanya, China
| | - Magella M Neveu
- From the UCL Institute of Ophthalmology (A.M., N.J., C.J., M.M.N., C.M.C., M.S., G.A., A.R.W., A.T.M., M.M., A.S., A.G.R., P.Y.-W.-M.), London, United Kingdom; Moorfields Eye Hospital (A.M., N.J., M.M.N., C.M.C., G.A., A.R.W., A.T.M., M.M., A.G.R., P.Y.-W.-M.), London, United Kingdom
| | - Choi Mun Chan
- From the UCL Institute of Ophthalmology (A.M., N.J., C.J., M.M.N., C.M.C., M.S., G.A., A.R.W., A.T.M., M.M., A.S., A.G.R., P.Y.-W.-M.), London, United Kingdom; Moorfields Eye Hospital (A.M., N.J., M.M.N., C.M.C., G.A., A.R.W., A.T.M., M.M., A.G.R., P.Y.-W.-M.), London, United Kingdom
| | - Holly J Duncan
- Newcastle Eye Centre, Royal Victoria Infirmary (H.J.D.), Newcastle upon Tyne, United Kingdom
| | - Simon Kelly
- Bolton NHS Foundation Trust (S.K., F.K., Y.T.L.), Bolton, United Kingdom
| | - Emma Burkitt-Wright
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust (E.B.-W.), Manchester, United Kingdom; Division of Evolution and Genomic Sciences, University of Manchester, Manchester Academic Health Sciences Centre (E.B.-W.), Manchester, United Kingdom
| | - Fadil Khoyratty
- Bolton NHS Foundation Trust (S.K., F.K., Y.T.L.), Bolton, United Kingdom
| | - Yoon Tse Lai
- Bolton NHS Foundation Trust (S.K., F.K., Y.T.L.), Bolton, United Kingdom
| | - Mala Subash
- From the UCL Institute of Ophthalmology (A.M., N.J., C.J., M.M.N., C.M.C., M.S., G.A., A.R.W., A.T.M., M.M., A.S., A.G.R., P.Y.-W.-M.), London, United Kingdom
| | - Patrick F Chinnery
- MRC Mitochondrial Biology Unit, Department of Clinical Neurosciences, University of Cambridge (P.F.C.), Cambridge, United Kingdom
| | | | - Gavin Arno
- From the UCL Institute of Ophthalmology (A.M., N.J., C.J., M.M.N., C.M.C., M.S., G.A., A.R.W., A.T.M., M.M., A.S., A.G.R., P.Y.-W.-M.), London, United Kingdom; Moorfields Eye Hospital (A.M., N.J., M.M.N., C.M.C., G.A., A.R.W., A.T.M., M.M., A.G.R., P.Y.-W.-M.), London, United Kingdom
| | - Andrew R Webster
- From the UCL Institute of Ophthalmology (A.M., N.J., C.J., M.M.N., C.M.C., M.S., G.A., A.R.W., A.T.M., M.M., A.S., A.G.R., P.Y.-W.-M.), London, United Kingdom; Moorfields Eye Hospital (A.M., N.J., M.M.N., C.M.C., G.A., A.R.W., A.T.M., M.M., A.G.R., P.Y.-W.-M.), London, United Kingdom
| | - Anthony T Moore
- From the UCL Institute of Ophthalmology (A.M., N.J., C.J., M.M.N., C.M.C., M.S., G.A., A.R.W., A.T.M., M.M., A.S., A.G.R., P.Y.-W.-M.), London, United Kingdom; Moorfields Eye Hospital (A.M., N.J., M.M.N., C.M.C., G.A., A.R.W., A.T.M., M.M., A.G.R., P.Y.-W.-M.), London, United Kingdom; Department of Ophthalmology, UCSF School of Medicine (A.T.M.), San Francisco, California, USA
| | - Michel Michaelides
- From the UCL Institute of Ophthalmology (A.M., N.J., C.J., M.M.N., C.M.C., M.S., G.A., A.R.W., A.T.M., M.M., A.S., A.G.R., P.Y.-W.-M.), London, United Kingdom; Moorfields Eye Hospital (A.M., N.J., M.M.N., C.M.C., G.A., A.R.W., A.T.M., M.M., A.G.R., P.Y.-W.-M.), London, United Kingdom
| | - Andrew Stockman
- From the UCL Institute of Ophthalmology (A.M., N.J., C.J., M.M.N., C.M.C., M.S., G.A., A.R.W., A.T.M., M.M., A.S., A.G.R., P.Y.-W.-M.), London, United Kingdom
| | - Anthony G Robson
- From the UCL Institute of Ophthalmology (A.M., N.J., C.J., M.M.N., C.M.C., M.S., G.A., A.R.W., A.T.M., M.M., A.S., A.G.R., P.Y.-W.-M.), London, United Kingdom; Moorfields Eye Hospital (A.M., N.J., M.M.N., C.M.C., G.A., A.R.W., A.T.M., M.M., A.G.R., P.Y.-W.-M.), London, United Kingdom
| | - Patrick Yu-Wai-Man
- From the UCL Institute of Ophthalmology (A.M., N.J., C.J., M.M.N., C.M.C., M.S., G.A., A.R.W., A.T.M., M.M., A.S., A.G.R., P.Y.-W.-M.), London, United Kingdom; Moorfields Eye Hospital (A.M., N.J., M.M.N., C.M.C., G.A., A.R.W., A.T.M., M.M., A.G.R., P.Y.-W.-M.), London, United Kingdom; John van Geest Centre for Brain Repair and MRC Mitochondrial Biology Unit, Department of Clinical Neurosciences, University of Cambridge (P.Y.-W.-M.), Cambridge, United Kingdom; and Cambridge Eye Unit, Addenbrooke's Hospital, Cambridge University Hospitals (P.Y.-W.-M.), Cambridge, United Kingdom
| |
Collapse
|
18
|
β-cyclodextrin based nano gene delivery using pharmaceutical applications to treat Wolfram syndrome. Ther Deliv 2022; 13:449-462. [PMID: 36748654 DOI: 10.4155/tde-2022-0036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Wolfram syndrome is a rare multisystem autosomal recessive neurodegenerative disorder that affects the brain and central nervous system. Currently, there is no cure or treatment for Wolfram syndrome. Therefore, new techniques are needed to target the loss of the WFS1 gene. Gene therapy approach to introduce a functional gene using a viral or a non-viral vector could be a treatment strategy for Wolfram syndrome 1 (WS1). Viral vectors have therapeutic benefits and greater efficiency; however, they pose a high health risk. Recently pharmaceutical therapeutic research has developed cell-penetrating non-viral nano molecules that could be used as vectors for gene delivery. Among nonviral vectors, the unique properties of β-cyclodextrin suggest that it can be a promising safe vector for gene delivery.
Collapse
|
19
|
Tepp K, Aid-Vanakova J, Puurand M, Timohhina N, Reinsalu L, Tein K, Plaas M, Shevchuk I, Terasmaa A, Kaambre T. Wolframin deficiency is accompanied with metabolic inflexibility in rat striated muscles. Biochem Biophys Rep 2022; 30:101250. [PMID: 35295995 PMCID: PMC8918847 DOI: 10.1016/j.bbrep.2022.101250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/28/2022] [Accepted: 03/08/2022] [Indexed: 11/26/2022] Open
Abstract
The protein 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. Mutations in Wfs1 gene cause autosomal recessive disorder Wolfram syndrome (WS). The first symptom of the WS is diabetes mellitus, so accurate diagnosis of the disease as WS is often delayed. In this study we aimed to characterize the role of the Wfs1 deficiency on bioenergetics of muscles. Alterations in the bioenergetic profiles of Wfs1-exon-5-knock-out (Wfs1KO) male rats in comparison with their wild-type male littermates were investigated using high-resolution respirometry, and enzyme activity measurements. The changes were followed in oxidative (cardiac and soleus) and glycolytic (rectus femoris and gastrocnemius) muscles. There were substrate-dependent alterations in the oxygen consumption rate in Wfs1KO rat muscles. In soleus muscle, decrease in respiration rate was significant in all the followed pathways. The relatively small alterations in muscle during development of WS, such as increased mitochondrial content and/or increase in the OxPhos-related enzymatic activity could be an adaptive response to changes in the metabolic environment. The significant decrease in the OxPhos capacity is substrate dependent indicating metabolic inflexibility when multiple substrates are available. Wolfram syndrome (WS) model rats have muscle type-dependent metabolic changes. Substrate-dependent modulation of OxPhos in WS model rat muscles. Metabolic inflexibility in early-stage WS rat muscle mitochondria.
Collapse
|
20
|
Rigoli L, Caruso V, Salzano G, Lombardo F. Wolfram Syndrome 1: From Genetics to Therapy. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:3225. [PMID: 35328914 PMCID: PMC8949990 DOI: 10.3390/ijerph19063225] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 02/01/2023]
Abstract
Wolfram syndrome 1 (WS1) is a rare neurodegenerative disease transmitted in an autosomal recessive mode. It is characterized by diabetes insipidus (DI), diabetes mellitus (DM), optic atrophy (OA), and sensorineural hearing loss (D) (DIDMOAD). The clinical picture may be complicated by other symptoms, such as urinary tract, endocrinological, psychiatric, and neurological abnormalities. WS1 is caused by mutations in the WFS1 gene located on chromosome 4p16 that encodes a transmembrane protein named wolframin. Many studies have shown that wolframin regulates some mechanisms of ER calcium homeostasis and therefore plays a role in cellular apoptosis. More than 200 mutations are responsible for WS1. However, abnormal phenotypes of WS with or without DM, inherited in an autosomal dominant mode and associated with one or more WFS1 mutations, have been found. Furthermore, recessive Wolfram-like disease without DM has been described. The prognosis of WS1 is poor, and the death occurs prematurely. Although there are no therapies that can slow or stop WS1, a careful clinical monitoring can help patients during the rapid progression of the disease, thus improving their quality of life. In this review, we describe natural history and etiology of WS1 and suggest criteria for a most pertinent approach to the diagnosis and clinical follow up. We also describe the hallmarks of new therapies for WS1.
Collapse
Affiliation(s)
- Luciana Rigoli
- Department of Human Pathology of Adulthood and Childhood G. Barresi, University of Messina, 98125 Messina, Italy; (G.S.); (F.L.)
| | - Valerio Caruso
- Psychiatry 2 Unit, Clinical and Experimental Medicine Department, University of Pisa, 56126 Pisa, Italy;
| | - Giuseppina Salzano
- Department of Human Pathology of Adulthood and Childhood G. Barresi, University of Messina, 98125 Messina, Italy; (G.S.); (F.L.)
| | - Fortunato Lombardo
- Department of Human Pathology of Adulthood and Childhood G. Barresi, University of Messina, 98125 Messina, Italy; (G.S.); (F.L.)
| |
Collapse
|
21
|
Rigoli L, Caruso V, Aloi C, Salina A, Maghnie M, d’Annunzio G, Lamacchia O, Salzano G, Lombardo F, Picca G. An Atypical Case of Late-Onset Wolfram Syndrome 1 without Diabetes Insipidus. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:2473. [PMID: 35206658 PMCID: PMC8872384 DOI: 10.3390/ijerph19042473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 12/04/2022]
Abstract
Wolfram syndrome 1, a rare autosomal recessive neurodegenerative disease, is caused by mutations in the WFS1 gene. It is characterized by diabetes insipidus, diabetes mellitus, optic atrophy, and deafness (DIDMOAD), and other clinical manifestations such as urological and neurological disorders. Here we described the case of a patient with an atypical late-onset Wolfram syndrome 1 without DI. Our WS1 patient was a c.1620_1622delGTG (p.Trp540del)/c.124 C > T (p.Arg42*) heterozygous compound. The p.Arg42* nonsense mutation was also found in heterozygosity in his sister and niece, both suffering from psychiatric disorders. The p.Arg42* nonsense mutation has never been found in WS1 and its pathogenicity is unclear so far. Our study underlined the need to study a greater number of WS1 cases in order to better understand the clinical significance of many WFS1 variants.
Collapse
Affiliation(s)
- Luciana Rigoli
- Department of Human Pathology of Adulthood and Childhood G. Barresi, University of Messina, 98125 Messina, Italy; (G.S.); (F.L.)
| | - Valerio Caruso
- Psychiatry 2 Unit, Clinical and Experimental Medicine Department, University of Pisa, 56126 Pisa, Italy;
| | - Concetta Aloi
- Pediatric Clinic, LABSIEM (Laboratory for the Study of Inborn Errors of Metabolism), IRCCS Institute Giannina Gaslini, 16147 Genoa, Italy; (C.A.); (A.S.); (M.M.); (G.d.)
| | - Alessandro Salina
- Pediatric Clinic, LABSIEM (Laboratory for the Study of Inborn Errors of Metabolism), IRCCS Institute Giannina Gaslini, 16147 Genoa, Italy; (C.A.); (A.S.); (M.M.); (G.d.)
| | - Mohamad Maghnie
- Pediatric Clinic, LABSIEM (Laboratory for the Study of Inborn Errors of Metabolism), IRCCS Institute Giannina Gaslini, 16147 Genoa, Italy; (C.A.); (A.S.); (M.M.); (G.d.)
| | - Giuseppe d’Annunzio
- Pediatric Clinic, LABSIEM (Laboratory for the Study of Inborn Errors of Metabolism), IRCCS Institute Giannina Gaslini, 16147 Genoa, Italy; (C.A.); (A.S.); (M.M.); (G.d.)
| | - Olga Lamacchia
- Unit of Endocrinology and Diabetology, Department of Medical and Surgical Sciences, University of Foggia, 71100 Foggia, Italy; (O.L.); (G.P.)
| | - Giuseppina Salzano
- Department of Human Pathology of Adulthood and Childhood G. Barresi, University of Messina, 98125 Messina, Italy; (G.S.); (F.L.)
| | - Fortunato Lombardo
- Department of Human Pathology of Adulthood and Childhood G. Barresi, University of Messina, 98125 Messina, Italy; (G.S.); (F.L.)
| | - Giuseppe Picca
- Unit of Endocrinology and Diabetology, Department of Medical and Surgical Sciences, University of Foggia, 71100 Foggia, Italy; (O.L.); (G.P.)
| |
Collapse
|
22
|
Other Hereditary Optic Neuropathy. Neuroophthalmology 2022. [DOI: 10.1007/978-981-19-4668-4_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
23
|
Barnabei A, Strigari L, Corsello A, Paragliola RM, Iannantuono GM, Salvatori R, Corsello SM, Torino F. Grading Central Diabetes Insipidus Induced by Immune Checkpoint Inhibitors: A Challenging Task. Front Endocrinol (Lausanne) 2022; 13:840971. [PMID: 35388297 PMCID: PMC8978963 DOI: 10.3389/fendo.2022.840971] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/08/2022] [Indexed: 12/26/2022] Open
Abstract
Central diabetes insipidus (CDI) is a rare endocrine disease deriving from an insufficient production or secretion of anti-diuretic hormone. Recently, CDI has been reported as a rare side effect triggered by immune checkpoint inhibitors (ICI) in cancer patients. Despite its current rarity, CDI triggered by ICI is expected to affect an increasing number of patients because of the expanding use of these effective drugs in a growing number of solid and hematologic malignancies. An appropriate assessment of the severity of adverse events induced by anticancer agents is crucial in their management, including dosing adjustment and temporary withdrawal or discontinuation treatment. However, assessment of the severity of CDI induced by ICI may be challenging, as its main signs and symptoms (polyuria, dehydration, weight loss, and hypernatremia) can be incompletely graded. Indeed, the current grading system of toxicity induced by anticancer treatments does not include polyuria. Additionally, dehydration in patients affected by diabetes insipidus, including ICI-induced CDI, is different in certain aspects from that due to other conditions seen in cancer patients, such as vomiting and diarrhea. This prompted us to reflect on the need to grade polyuria, and how to grade it, and to consider a specific grading system for dehydration associated with CDI induced by ICI. Here we propose a new grading system for polyuria and dehydration, as critical symptoms of the CDI syndrome occurring in patients on ICI treatment, to obtain better management of both the adverse event and the triggering drugs.
Collapse
Affiliation(s)
- Agnese Barnabei
- Endocrinology Unit, Presidio Ospedaliero Santo Spirito in Sassia, Azienda Sanitaria Locale Roma 1, Rome, Italy
| | - Lidia Strigari
- Medical Physics Department, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Andrea Corsello
- Department of Translational Medicine and Surgery, Unit of Endocrinology, Università Cattolica del Sacro Cuore-Fondazione Policlinico “Gemelli” Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Rosa Maria Paragliola
- Department of Translational Medicine and Surgery, Unit of Endocrinology, Università Cattolica del Sacro Cuore-Fondazione Policlinico “Gemelli” Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | | | - Roberto Salvatori
- Division of Endocrinology, Diabetes, and Metabolism and Pituitary Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Salvatore Maria Corsello
- Department of Translational Medicine and Surgery, Unit of Endocrinology, Università Cattolica del Sacro Cuore-Fondazione Policlinico “Gemelli” Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
- UniCamillus Chair of Endocrinology, Saint Camillus International University of Health Sciences, Rome, Italy
| | - Francesco Torino
- Department of Systems Medicine, Medical Oncology, Tor Vergata University of Rome, Rome, Italy
- *Correspondence: Francesco Torino,
| |
Collapse
|
24
|
Mirrahimi M, Safi S, Mohammadzadeh M, Doozandeh A, Suri F. Variable Expressivity of Wolfram Syndrome in a Family with Multiple Affected Subjects. J Ophthalmic Vis Res 2021; 16:602-610. [PMID: 34840683 PMCID: PMC8593543 DOI: 10.18502/jovr.v16i4.9750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/25/2020] [Indexed: 11/30/2022] Open
Abstract
Purpose To study the genetic basis and clinical manifestations of Wolfram syndrome in a multi-affected family. Methods Complete clinical examinations including urological, ophthalmic, neurological, and endocrinologic assessment were performed for three affected family members. Genomic DNA was extracted from peripheral blood leukocytes with salting out method and all WFS1 exons and their flanking regions were sequenced. Candidate variation was screened for segregation in the pedigree by Sanger sequencing. Results A known pathogenic missense mutation in WFS1 gene (c.1885C>T which leads to p.Arg629Trp in the encoded protein) was identified in all affected individuals. Both clinical and genetic investigations confirmed Wolfram syndrome diagnosis with variable phenotypic features Conclusion Identical mutations in the Wolfram syndrome causative gene can lead to variable manifestations of the syndrome even in the same family. Although the medical findings and clinical examination are imperative for the diagnosis of Wolfram syndrome, genetic testing is useful to confirm the diagnosis, especially in cases with possible reduced penetrance of the characteristic signs.
Collapse
Affiliation(s)
- Mehraban Mirrahimi
- Ocular Tissue Engineering Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sare Safi
- Ophthalmic Epidemiology Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Mohammadzadeh
- Ophthalmic Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azadeh Doozandeh
- Ophthalmic Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Suri
- Ophthalmic Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Jagomäe T, Seppa K, Reimets R, Pastak M, Plaas M, Hickey MA, Kukker KG, Moons L, De Groef L, Vasar E, Kaasik A, Terasmaa A, Plaas M. Early Intervention and Lifelong Treatment with GLP1 Receptor Agonist Liraglutide in a Wolfram Syndrome Rat Model with an Emphasis on Visual Neurodegeneration, Sensorineural Hearing Loss and Diabetic Phenotype. Cells 2021; 10:cells10113193. [PMID: 34831417 PMCID: PMC8623088 DOI: 10.3390/cells10113193] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/11/2021] [Accepted: 11/14/2021] [Indexed: 01/11/2023] Open
Abstract
Wolfram syndrome (WS), also known as a DIDMOAD (diabetes insipidus, early-onset diabetes mellitus, optic nerve atrophy and deafness) is a rare autosomal disorder caused by mutations in the Wolframin1 (WFS1) gene. Previous studies have revealed that glucagon-like peptide-1 receptor agonist (GLP1 RA) are effective in delaying and restoring blood glucose control in WS animal models and patients. The GLP1 RA liraglutide has also been shown to have neuroprotective properties in aged WS rats. WS is an early-onset, chronic condition. Therefore, early diagnosis and lifelong pharmacological treatment is the best solution to control disease progression. Hence, the aim of this study was to evaluate the efficacy of the long-term liraglutide treatment on the progression of WS symptoms. For this purpose, 2-month-old WS rats were treated with liraglutide up to the age of 18 months and changes in diabetes markers, visual acuity, and hearing sensitivity were monitored over the course of the treatment period. We found that treatment with liraglutide delayed the onset of diabetes and protected against vision loss in a rat model of WS. Therefore, early diagnosis and prophylactic treatment with the liraglutide may also prove to be a promising treatment option for WS patients by increasing the quality of life.
Collapse
Affiliation(s)
- Toomas Jagomäe
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, 14B Ravila Street, 50411 Tartu, Estonia; (K.S.); (R.R.); (K.G.K.); (A.T.)
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia;
- Correspondence: (T.J.); (M.P.)
| | - Kadri Seppa
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, 14B Ravila Street, 50411 Tartu, Estonia; (K.S.); (R.R.); (K.G.K.); (A.T.)
| | - Riin Reimets
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, 14B Ravila Street, 50411 Tartu, Estonia; (K.S.); (R.R.); (K.G.K.); (A.T.)
| | - Marko Pastak
- Eye Clinic of Tartu University Hospital, L. Puusepa 8 Street, 50406 Tartu, Estonia;
| | - Mihkel Plaas
- Ear Clinic of Tartu University Hospital, L. Puusepa 1a Street, 50406 Tartu, Estonia;
| | - Miriam A. Hickey
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; (M.A.H.); (A.K.)
| | - Kaia Grete Kukker
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, 14B Ravila Street, 50411 Tartu, Estonia; (K.S.); (R.R.); (K.G.K.); (A.T.)
| | - Lieve Moons
- Research Group Neural Circuit Development and Regeneration, Department of Biology, Belgium & Leuven Brain Institute, University of Leuven, Naamsestraat 61, Box 2464, 3000 Leuven, Belgium; (L.M.); (L.D.G.)
| | - Lies De Groef
- Research Group Neural Circuit Development and Regeneration, Department of Biology, Belgium & Leuven Brain Institute, University of Leuven, Naamsestraat 61, Box 2464, 3000 Leuven, Belgium; (L.M.); (L.D.G.)
| | - Eero Vasar
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia;
| | - Allen Kaasik
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; (M.A.H.); (A.K.)
| | - Anton Terasmaa
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, 14B Ravila Street, 50411 Tartu, Estonia; (K.S.); (R.R.); (K.G.K.); (A.T.)
| | - Mario Plaas
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, 14B Ravila Street, 50411 Tartu, Estonia; (K.S.); (R.R.); (K.G.K.); (A.T.)
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia;
- Correspondence: (T.J.); (M.P.)
| |
Collapse
|
26
|
Tunyasuvunakool K, Adler J, Wu Z, Green T, Zielinski M, Žídek A, Bridgland A, Cowie A, Meyer C, Laydon A, Velankar S, Kleywegt GJ, Bateman A, Evans R, Pritzel A, Figurnov M, Ronneberger O, Bates R, Kohl SAA, Potapenko A, Ballard AJ, Romera-Paredes B, Nikolov S, Jain R, Clancy E, Reiman D, Petersen S, Senior AW, Kavukcuoglu K, Birney E, Kohli P, Jumper J, Hassabis D. Highly accurate protein structure prediction for the human proteome. Nature 2021; 596:590-596. [PMID: 34293799 PMCID: PMC8387240 DOI: 10.1038/s41586-021-03828-1] [Citation(s) in RCA: 1711] [Impact Index Per Article: 427.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/16/2021] [Indexed: 02/07/2023]
Abstract
Protein structures can provide invaluable information, both for reasoning about biological processes and for enabling interventions such as structure-based drug development or targeted mutagenesis. After decades of effort, 17% of the total residues in human protein sequences are covered by an experimentally determined structure1. Here we markedly expand the structural coverage of the proteome by applying the state-of-the-art machine learning method, AlphaFold2, at a scale that covers almost the entire human proteome (98.5% of human proteins). The resulting dataset covers 58% of residues with a confident prediction, of which a subset (36% of all residues) have very high confidence. We introduce several metrics developed by building on the AlphaFold model and use them to interpret the dataset, identifying strong multi-domain predictions as well as regions that are likely to be disordered. Finally, we provide some case studies to illustrate how high-quality predictions could be used to generate biological hypotheses. We are making our predictions freely available to the community and anticipate that routine large-scale and high-accuracy structure prediction will become an important tool that will allow new questions to be addressed from a structural perspective.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Sameer Velankar
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, UK
| | - Gerard J Kleywegt
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, UK
| | - Alex Bateman
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Ewan Birney
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, UK
| | | | | | | |
Collapse
|
27
|
Gong Y, Xiong L, Li X, Su L, Xiao H. A novel mutation of WFS1 gene leading to increase ER stress and cell apoptosis is associated an autosomal dominant form of Wolfram syndrome type 1. BMC Endocr Disord 2021; 21:76. [PMID: 33879153 PMCID: PMC8059287 DOI: 10.1186/s12902-021-00748-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 04/12/2021] [Indexed: 12/15/2022] Open
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.
Collapse
Affiliation(s)
- Yingying Gong
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road 2, Guangzhou, 510080, P. R. China
- Department of Geriatrics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Li Xiong
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road 2, Guangzhou, 510080, P. R. China
| | - Xiujun Li
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road 2, Guangzhou, 510080, P. R. China
| | - Lei Su
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road 2, Guangzhou, 510080, P. R. China
- Department of Geriatrics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Haipeng Xiao
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road 2, Guangzhou, 510080, P. R. China.
| |
Collapse
|
28
|
Acharya A, Raza SI, Anwar MZ, Bharadwaj T, Liaqat K, Khokhar MAS, Everard JL, Nasir A, Nickerson DA, Bamshad MJ, Ansar M, Schrauwen I, Ahmad W, Leal SM. Wolfram-like syndrome with bicuspid aortic valve due to a homozygous missense variant in CDK13. J Hum Genet 2021; 66:1009-1018. [PMID: 33879837 PMCID: PMC8472924 DOI: 10.1038/s10038-021-00922-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 12/03/2022]
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.
Collapse
Affiliation(s)
- Anushree Acharya
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and the Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Syed Irfan Raza
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.,Department of Biochemistry, HBS Medical and Dental College, Islamabad, Pakistan
| | | | - Thashi Bharadwaj
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and the Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Khurram Liaqat
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | | | - Jenna L Everard
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and the Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Abdul Nasir
- Synthetic Protein Engineering Lab (SPEL), Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | | | | | - Michael J Bamshad
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.,Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Muhammad Ansar
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Isabelle Schrauwen
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and the Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Wasim Ahmad
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Suzanne M Leal
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, and the Department of Neurology, Columbia University Medical Center, New York, NY, USA. .,Taub Institute for Alzheimer's Disease and The Aging Brain, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
29
|
Zebrafish Models of Autosomal Recessive Ataxias. Cells 2021; 10:cells10040836. [PMID: 33917666 PMCID: PMC8068028 DOI: 10.3390/cells10040836] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/01/2021] [Accepted: 04/06/2021] [Indexed: 12/11/2022] Open
Abstract
Autosomal recessive ataxias are much less well studied than autosomal dominant ataxias and there are no clearly defined systems to classify them. Autosomal recessive ataxias, which are characterized by neuronal and multisystemic features, have significant overlapping symptoms with other complex multisystemic recessive disorders. The generation of animal models of neurodegenerative disorders increases our knowledge of their cellular and molecular mechanisms and helps in the search for new therapies. Among animal models, the zebrafish, which shares 70% of its genome with humans, offer the advantages of being small in size and demonstrating rapid development, making them optimal for high throughput drug and genetic screening. Furthermore, embryo and larval transparency allows to visualize cellular processes and central nervous system development in vivo. In this review, we discuss the contributions of zebrafish models to the study of autosomal recessive ataxias characteristic phenotypes, behavior, and gene function, in addition to commenting on possible treatments found in these models. Most of the zebrafish models generated to date recapitulate the main features of recessive ataxias.
Collapse
|
30
|
Reschke F, Rohayem J, Maffei P, Dassie F, Schwandt A, de Beaufort C, Toni S, Szypowska A, Cardona-Hernandez R, Datz N, Klee K, Danne T. Collaboration for rare diabetes: understanding new treatment options for Wolfram syndrome. Endocrine 2021; 71:626-633. [PMID: 33527330 DOI: 10.1007/s12020-021-02622-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/09/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Wolfram Syndrome is a very rare genetic disease causing diabetes mellitus, blindness, deafness, diabetes insipidus, and progressive brainstem degeneration. Neurologic symptoms of affected patients include ataxia, sleep apnea, loss of bladder control, dysphagia, loss of taste, and accompanying psychiatric symptoms as a sign of progressive neurodegeneration. Its genetic cause is mainly biallelic mutations of the Wolframin endoplasmatic reticulum transmembrane glycoprotein gene Wfs1. These result in increased ER stress, which in turn induces apoptosis and leads to the depletion of the corresponding cells and a loss of their physiological functions. Though diabetes mellitus is mostly treated by insulin, there is still no proven cure for the disease in general. It leads to premature death in affected individuals-usually within the 4th decade of live. CURRENT RESEARCH AND TREATMENT TRIALS Clinical studies are currently being conducted at various locations worldwide to test a therapy for the disease using various approaches. POTENTAIL OF VIRTUAL NETOWRKING As rare diseases in general represent a major challenge for individual clinicians and researchers due to the rarity of diagnosis, the lack of evidence and of value of existing research, international cooperation, coordination and networking leading to an alignment of different stakeholders is necessary to support patients and increase knowledge about these diseases, like wolfram syndrome. CONCLUSION ENDO-ERN and EURRECA are two EU-funded networks that aim to promote knowledge sharing, education and research on rare endocrine diseases.
Collapse
Affiliation(s)
- Felix Reschke
- Diabetes Center, Children's Hospital AUF DER BULT, Hannover, Germany.
| | - Julia Rohayem
- Centre of Reproductive Medicine and Andrology, Department of Clinical and Surgical Andrology, University of Münster, Muenster, Germany
| | - Pietro Maffei
- Department of Medicine (DIMED), Padua University Hospital, Padua, Italy.
| | - Francesca Dassie
- Department of Medicine (DIMED), Padua University Hospital, Padua, Italy
| | - Anke Schwandt
- Institute of Epidemiology and Medical Biometry, ZIBMT, Ulm University, Ulm, Germany
- German Center for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Carine de Beaufort
- Pediatric Clinic, DECCP/Center Hospitalier de Luxembourg, Luxembourg, Grand-Duché de Luxembourg
| | - Sonia Toni
- Ospedale Pediatrico Meyer Firenze, Florence, Italy
| | | | | | - Nicolin Datz
- Diabetes Center, Children's Hospital AUF DER BULT, Hannover, Germany
| | - Katharina Klee
- Diabetes Center, Children's Hospital AUF DER BULT, Hannover, Germany
| | - Thomas Danne
- Diabetes Center, Children's Hospital AUF DER BULT, Hannover, Germany
| |
Collapse
|
31
|
Mishra R, Chen BS, Richa P, Yu-Wai-Man P. Wolfram syndrome: new pathophysiological insights and therapeutic strategies. THERAPEUTIC ADVANCES IN RARE DISEASE 2021; 2:26330040211039518. [PMID: 37181110 PMCID: PMC10032446 DOI: 10.1177/26330040211039518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 07/23/2021] [Indexed: 05/16/2023]
Abstract
Wolfram Syndrome (WS) is an ultra-rare, progressive neurodegenerative disease characterized by early-onset diabetes mellitus and irreversible loss of vision, secondary to optic nerve degeneration. Visual loss in WS is an important cause of registrable blindness in children and young adults and the pathological hallmark is the preferential loss of retinal ganglion cells within the inner retina. In addition to optic atrophy, affected individuals frequently develop variable combinations of neurological, endocrinological, and psychiatric complications. The majority of patients carry recessive mutations in the WFS1 (4p16.1) gene that encodes for a multimeric transmembrane protein, wolframin, embedded within the endoplasmic reticulum (ER). An increasingly recognised subgroup of patients harbor dominant WFS1 mutations that usually cause a milder phenotype, which can be limited to optic atrophy. Wolframin is a ubiquitous protein with high levels of expression in retinal, neuronal, and muscle tissues. It is a multifunctional protein that regulates a host of cellular functions, in particular the dynamic interaction with mitochondria at mitochondria-associated membranes. Wolframin has been implicated in several crucial cellular signaling pathways, including insulin signaling, calcium homeostasis, and the regulation of apoptosis and the ER stress response. There is currently no cure for WS; management remains largely supportive. This review will cover the clinical, genetic, and pathophysiological features of WS, with a specific focus on disease models and the molecular pathways that could serve as potential therapeutic targets. The current landscape of therapeutic options will also be discussed in the context of the latest evidence, including the pipeline for repurposed drugs and gene therapy. Plain language summary Wolfram syndrome - disease mechanisms and treatment options Wolfram syndrome (WS) is an ultra-rare genetic disease that causes diabetes mellitus and progressive loss of vision from early childhood. Vision is affected in WS because of damage to a specialized type of cells in the retina, known as retinal ganglion cells (RGCs), which converge at the back of the eye to form the optic nerve. The optic nerve is the fast-conducting cable that transmits visual information from the eye to the vision processing centers within the brain. As RGCs are lost, the optic nerve degenerates and it becomes pale in appearance (optic atrophy). Although diabetes mellitus and optic atrophy are the main features of WS, some patients can develop more severe problems because the brain and other organs, such as the kidneys and the bladder, are also affected. The majority of patients with WS carry spelling mistakes (mutations) in the WFS1 gene, which is located on the short arm of chromosome 4 (4p16.1). This gene is highly expressed in the eye and in the brain, and it encodes for a protein located within a compartment of the cell known as the endoplasmic reticulum. For reasons that still remain unclear, WFS1 mutations preferentially affect RGCs, accounting for the prominent visual loss in this genetic disorder. There is currently no effective treatment to halt or slow disease progression and management remains supportive, including the provision of visual aids and occupational rehabilitation. Research into WS has been limited by its relative rarity and the inability to get access to eye and brain tissues from affected patients. However, major advances in our understanding of this disease have been made recently by making use of more accessible cells from patients, such as skin cells (fibroblasts), or animal models, such as mice and zebrafish. This review summarizes the mechanisms by which WFS1 mutations affect cells, impairing their function and eventually leading to their premature loss. The possible treatment strategies to block these pathways are also discussed, with a particular focus on drug repurposing (i.e., using drugs that are already approved for other diseases) and gene therapy (i.e., replacing or repairing the defective WFS1 gene).
Collapse
Affiliation(s)
- Ratnakar Mishra
- Cambridge Centre for Brain Repair and MRC
Mitochondrial Biology Unit, Department of Clinical Neurosciences, University
of Cambridge, Cambridge, UK
| | - Benson S. Chen
- Cambridge Centre for Brain Repair and MRC
Mitochondrial Biology Unit, Department of Clinical Neurosciences, University
of Cambridge, Cambridge, UK
- Cambridge Eye Unit, Addenbrooke’s Hospital,
Cambridge University Hospitals, Cambridge, UK
| | - Prachi Richa
- Department of Physiology, Development and
Neuroscience, University of Cambridge, Cambridge, UK
| | - Patrick Yu-Wai-Man
- Cambridge Centre for Brain Repair and MRC
Mitochondrial Biology Unit, Department of Clinical Neurosciences, University
of Cambridge, ED Adrian Building, Robinson Way, Cambridge, CB2 0PY, UK
- Cambridge Eye Unit, Addenbrooke’s Hospital,
Cambridge University Hospitals, Cambridge, UK
- Moorfields Eye Hospital, London, UK
- UCL Institute of Ophthalmology, University
College London, London, UK
| |
Collapse
|
32
|
Abu-El-Haija A, McGowan C, Vanderveen D, Bodamer O. Autosomal-dominant WFS1-related disorder-Report of a novel WFS1 variant and review of the phenotypic spectrum of autosomal recessive and dominant forms. Am J Med Genet A 2020; 185:528-533. [PMID: 33179441 DOI: 10.1002/ajmg.a.61945] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 10/04/2020] [Accepted: 10/12/2020] [Indexed: 12/17/2022]
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.
Collapse
Affiliation(s)
- Aya Abu-El-Haija
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA.,Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Caroline McGowan
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Deborah Vanderveen
- Department of Ophthalmology, Boston Children's Hospital, Boston, MA, USA
| | - Olaf Bodamer
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| |
Collapse
|
33
|
Li L, Venkataraman L, Chen S, Fu H. Function of WFS1 and WFS2 in the Central Nervous System: Implications for Wolfram Syndrome and Alzheimer's disease. Neurosci Biobehav Rev 2020; 118:775-783. [PMID: 32949681 DOI: 10.1016/j.neubiorev.2020.09.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 08/25/2020] [Accepted: 09/10/2020] [Indexed: 12/14/2022]
Abstract
L.P. Li, L. Venkataraman, S. Chen, and H.J. Fu. Function of WFS1 and WFS2 in the Central Nervous System: Implications for Wolfram Syndrome and Alzheimer's Disease. NEUROSCI BIOBEHAV REVXXX-XXX,2020.-Wolfram syndrome (WS) is a rare monogenetic spectrum disorder characterized by insulin-dependent juvenile-onset diabetes mellitus, diabetes insipidus, optic nerve atrophy, hearing loss, progressive neurodegeneration, and a wide spectrum of psychiatric manifestations. Most WS patients belong to Wolfram Syndrome type 1 (WS1) caused by mutations in the Wolfram Syndrome 1 (WFS1/Wolframin) gene, while a small fraction of patients belongs to Wolfram Syndrome type 2 (WS2) caused by pathogenic variants in the CDGSH Iron Sulfur Domain 2 (CISD2/WFS2) gene. Although currently there is no treatment for this life-threatening disease, the molecular mechanisms underlying the pathogenesis of WS have been proposed. Interestingly, Alzheimer's disease (AD), an age-dependent neurodegenerative disease, shares some common mechanisms with WS. In this review, we focus on the function of WFS1 and WFS2 in the central nervous system as well as their implications in WS and AD. We also propose three future directions for elucidating the role of WFS1 and WFS2 in WS and AD.
Collapse
Affiliation(s)
- Liangping Li
- Department of Neuroscience, Chronic Brain Injury, Discovery Themes, The Ohio State University, Columbus, OH, USA
| | - Lalitha Venkataraman
- Department of Neuroscience, Chronic Brain Injury, Discovery Themes, The Ohio State University, Columbus, OH, USA
| | - Shuo Chen
- Department of Neuroscience, Chronic Brain Injury, Discovery Themes, The Ohio State University, Columbus, OH, USA
| | - Hongjun Fu
- Department of Neuroscience, Chronic Brain Injury, Discovery Themes, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
34
|
Batjargal K, Tajima T, Jimbo EF, Yamagata T. Effect of 4-phenylbutyrate and valproate on dominant mutations of WFS1 gene in Wolfram syndrome. J Endocrinol Invest 2020; 43:1317-1325. [PMID: 32219690 DOI: 10.1007/s40618-020-01228-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 03/17/2020] [Indexed: 02/08/2023]
Abstract
PURPOSE Wolfram syndrome (WS) is a rare disorder caused by mutations in WFS1 that is characterized by diabetes mellitus, optic atrophy, sensorineural deafness, diabetes insipidus, and neurodegeneration. This disease is usually inherited as an autosomal recessive trait, but an autosomal dominant form has been reported. WFS1 encodes a transmembrane protein, which is a maintenance component of endoplasmic homeostasis. These dominant mutations were thought to increase endoplasmic reticulum (ER) stress. Recent studies suggest that 4-phenylbutyrate (PBA) and valproate (VPA) reduce ER stress. The objective of this study was to analyze the effect of PBA and VPA on dominant WFS1 mutants in vitro. METHODS We determined whether dominant WFS1 mutants (p.His313Tyr, p.Trp314Arg, p.Asp325_Ile328del, p.Glu809Lys, and p.Glu864Lys) have the dominant negative effect using a luciferase assay of ER stress response element marker as ER stress. Moreover, the rescue of cell apoptosis induced by dominant WFS1 mutants following treatment with PBA or VPA was determined by quantitative real-time PCR of C/EBP homologous protein (CHOP) mRNA expression. RESULTS These mutants showed the dominant negative effect on the wild-type WFS1. In addition, the levels of ER stress and CHOP mRNA were significantly elevated by all dominant WFS1 mutants. After treatment with PBA or VPA, ER stress and cell apoptosis were reduced in each mutant. CONCLUSIONS PBA and VPA could reduce the ER stress and cell apoptosis caused by dominant WFS1 mutants.
Collapse
Affiliation(s)
- K Batjargal
- Department of Pediatrics, Graduate School of Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-shi, Tochigi, 329-0498, Japan.
- Department of Pediatrics, School of Medicine, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia.
| | - T Tajima
- Department of Pediatrics, Graduate School of Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-shi, Tochigi, 329-0498, Japan
| | - E F Jimbo
- Department of Pediatrics, Graduate School of Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-shi, Tochigi, 329-0498, Japan
| | - T Yamagata
- Department of Pediatrics, Graduate School of Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-shi, Tochigi, 329-0498, Japan
| |
Collapse
|
35
|
Das L, Rai A, Mavuduru R, Vaiphei K, Sharma A, Gupta V, Bhadada SK, Lodha S, Panda N, Bhansali A, Singh P, Dutta P. Wolfram syndrome: clinical and genetic profiling of a cohort from a tertiary care centre with characterization of the primary gonadal failure. Endocrine 2020; 69:420-429. [PMID: 32350710 DOI: 10.1007/s12020-020-02320-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 04/16/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Wolfram syndrome (WFS) is a rare, monogenic neurodegenerative syndrome characterised by insulin requiring non-autoimmune diabetes mellitus (DM) and optic atrophy which are usually the earliest and commonest manifestations. However, there are other features which are under-recognized, adding to morbidity and premature mortality in these patients. METHODS Five patients (three males, two females) with genetically confirmed WFS at a single tertiary care centre were prospectively followed up. Their symptomatology, clinical profile, genetic analysis and radiology were analyzed. Multidisciplinary approach was used for comprehensive clinical care of this cohort. Patients with primary gonadal failure were subjected to biopsy and immunohistochemistry (IHC) for wolframin was performed. RESULTS DM was the earliest presenting manifestation at 6.2 ± 1.3 years followed by optic atrophy at 10.4 ± 2.3 years, diabetes insipidus at 12 ± 2.1 years and deafness at 12.8 ± 2.1 years. All patients were autoantibody negative with low C-peptide(<0.6 ng/ml). Hypoglycemic episodes were frequent (upto 60%) but there was no instance of diabetic ketoacidosis. Optic atrophy was present alongwith proliferative diabetic retinopathy and cataract in 40%. Uncommon manifestations included neuropsychiatric features, parasuicide, cystopathy, brainstem atrophy and hypergonadotropic hypogonadism only in adult males (n = 2). Testicular biopsy revealed partly hyalinised seminiferous tubules and prominence of Leydig cells. IHC confirmed the presence of mutated wolframin, which was not significantly different from normal testis specimen on protein quantification. CONCLUSIONS WFS requires a multidisciplinary approach with special emphasis on early diagnosis and management of other endocrine and non-endocrine features so as to improve long-term outcomes. Gonadal functions need periodic assessment, especially in adult males.
Collapse
Affiliation(s)
- Liza Das
- Department of Endocrinology, PGIMER, Chandigarh, India
| | - Ashutosh Rai
- Department of Translational and Regenerative Medicine, PGIMER, Chandigarh, India
| | | | - Kim Vaiphei
- Department of Histopathology, PGIMER, Chandigarh, India
| | | | - Vishali Gupta
- Department of Ophthalmology, PGIMER, Chandigarh, India
| | | | - Sailesh Lodha
- Department of Endocrinology, Eternal Heart Care Centre, Jaipur, India
| | - Naresh Panda
- Department of Otorhinolarygology, PGIMER, Chandigarh, India
| | - Anil Bhansali
- Department of Endocrinology, PGIMER, Chandigarh, India
| | | | - Pinaki Dutta
- Department of Endocrinology, PGIMER, Chandigarh, India.
| |
Collapse
|
36
|
Riachi M, Yilmaz S, Kurnaz E, Aycan Z, Çetinkaya S, Tranebjærg L, Rendtorff ND, Bitner-Glindzicz M, Bockenhauer D, Hussain K. Functional assessment of variants associated with Wolfram syndrome. Hum Mol Genet 2020; 28:3815-3824. [PMID: 31600780 DOI: 10.1093/hmg/ddz212] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 08/28/2019] [Accepted: 08/28/2019] [Indexed: 11/14/2022] Open
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.
Collapse
Affiliation(s)
- Melissa Riachi
- Genetics and Genomic Medicine, UCL GOS Institute of Child Health, London, UK
| | - Sebahat Yilmaz
- Dr. Sami Ulus Obstetrics and Gynecology, Pediatric Health and Disease Training and Research Hospital, Pediatric Endocrinology Clinic, Ankara, Turkey
| | - Erdal Kurnaz
- Dr. Sami Ulus Obstetrics and Gynecology, Pediatric Health and Disease Training and Research Hospital, Pediatric Endocrinology Clinic, Ankara, Turkey
| | - Zehra Aycan
- Dr. Sami Ulus Obstetrics and Gynecology, Pediatric Health and Disease Training and Research Hospital, Pediatric Endocrinology Clinic, Ankara, Turkey
| | - Semra Çetinkaya
- Dr. Sami Ulus Obstetrics and Gynecology, Pediatric Health and Disease Training and Research Hospital, Pediatric Endocrinology Clinic, Ankara, Turkey
| | - Lisbeth Tranebjærg
- Department of Clinical Genetics, The Kennedy Center, University Hospital, Copenhagen, Denmark.,Institute of Clinical Medicine (IKM), The Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nanna Dahl Rendtorff
- Department of Clinical Genetics, The Kennedy Center, University Hospital, Copenhagen, Denmark
| | | | - Detlef Bockenhauer
- Department of Renal Medicine, UCL, London, UK.,Renal Unit, Great Ormond Street Hospital for Children, London, UK
| | - Khalid Hussain
- Genetics and Genomic Medicine, UCL GOS Institute of Child Health, London, UK.,Department of Pediatrics, Division of Endocrinology, Sidra Medicine, Doha, Qatar
| |
Collapse
|
37
|
Torkamandi S, Rezaei S, Mirfakhraie R, Bayat S, Piltan S, Gholami M. A homozygous missense mutation of WFS1 gene causes Wolfram's syndrome without hearing loss in an Iranian family (a report of clinical heterogeneity). J Clin Lab Anal 2020; 34:e23358. [PMID: 32419160 PMCID: PMC7439424 DOI: 10.1002/jcla.23358] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 04/14/2020] [Accepted: 04/21/2020] [Indexed: 12/13/2022] Open
Abstract
Background Wolfram's syndrome (WFS) is a hereditary (autosomal recessive) neurodegenerative disorder. The clinical features are related to diabetes insipidus, diabetes mellitus, optic atrophy, and deafness (DIDMOAD) with other variable clinical manifestations. Pathogenic variants in the WFS1 gene, encoding wolframin, are known to be the main cause of Wolfram's syndrome. In this study, we present the clinical and genetic characteristics of two WFS patients from an Iranian family. Methods The mutation screening was performed by polymerase chain reaction (PCR) followed by direct Sanger sequencing of all exons from two affected WFS. Results The complete Sanger sequencing of the WFS1 gene detected a homozygous missense variant, c.2207G>A (p.Gly736Asp), in the eighth exon of the WFS1 gene. Both cases developed all the major symptoms of the disease, interestingly, except hearing loss. Conclusions Because of the rarity and clinical heterogeneity of WFS, the molecular genetic assay is essential to confirm the diagnosis and management of the WFS patients.
Collapse
Affiliation(s)
- Shahram Torkamandi
- Department of Medical Genetics and Immunology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Somaye Rezaei
- Department of Neurology, Imam Khomeini Hospital, Urmia University of Medical Sciences, Urmia, Iran
| | - Reza Mirfakhraie
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahar Bayat
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samira Piltan
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Milad Gholami
- Molecular and Medicine Research Center, Arak University of Medical Sciences, Arak, Iran.,Department of Biochemistry and Genetics, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| |
Collapse
|
38
|
Schäffer DE, Iyer LM, Burroughs AM, Aravind L. Functional Innovation in the Evolution of the Calcium-Dependent System of the Eukaryotic Endoplasmic Reticulum. Front Genet 2020; 11:34. [PMID: 32117448 PMCID: PMC7016017 DOI: 10.3389/fgene.2020.00034] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 01/10/2020] [Indexed: 01/30/2023] Open
Abstract
The origin of eukaryotes was marked by the emergence of several novel subcellular systems. One such is the calcium (Ca2+)-stores system of the endoplasmic reticulum, which profoundly influences diverse aspects of cellular function including signal transduction, motility, division, and biomineralization. We use comparative genomics and sensitive sequence and structure analyses to investigate the evolution of this system. Our findings reconstruct the core form of the Ca2+-stores system in the last eukaryotic common ancestor as having at least 15 proteins that constituted a basic system for facilitating both Ca2+ flux across endomembranes and Ca2+-dependent signaling. We present evidence that the key EF-hand Ca2+-binding components had their origins in a likely bacterial symbiont other than the mitochondrial progenitor, whereas the protein phosphatase subunit of the ancestral calcineurin complex was likely inherited from the asgard archaeal progenitor of the stem eukaryote. This further points to the potential origin of the eukaryotes in a Ca2+-rich biomineralized environment such as stromatolites. We further show that throughout eukaryotic evolution there were several acquisitions from bacteria of key components of the Ca2+-stores system, even though no prokaryotic lineage possesses a comparable system. Further, using quantitative measures derived from comparative genomics we show that there were several rounds of lineage-specific gene expansions, innovations of novel gene families, and gene losses correlated with biological innovation such as the biomineralized molluscan shells, coccolithophores, and animal motility. The burst of innovation of new genes in animals included the wolframin protein associated with Wolfram syndrome in humans. We show for the first time that it contains previously unidentified Sel1, EF-hand, and OB-fold domains, which might have key roles in its biochemistry.
Collapse
Affiliation(s)
- Daniel E Schäffer
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, United States.,Science, Mathematics, and Computer Science Magnet Program, Montgomery Blair High School, Silver Spring, MD, United States
| | - Lakshminarayan M Iyer
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, United States
| | - A Maxwell Burroughs
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, United States
| | - L Aravind
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
39
|
Rigoli L, Aloi C, Salina A, Di Bella C, Salzano G, Caruso R, Mazzon E, Maghnie M, Patti G, D'Annunzio G, Lombardo F. Wolfram syndrome 1 in the Italian population: genotype-phenotype correlations. Pediatr Res 2020; 87:456-462. [PMID: 31266054 DOI: 10.1038/s41390-019-0487-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 05/14/2019] [Accepted: 06/20/2019] [Indexed: 11/09/2022]
Abstract
OBJECTIVES We studied 45 patients with Wolfram syndrome 1 (WS1) to describe their clinical history and to search for possible genotype-phenotype correlations. METHODS Clinical criteria contributing to WS1 diagnosis were analyzed. The patients were classified into three genotypic classes according to type of detected mutations. RESULTS WS1 prevalence in Italy is 0.74/1,000,000. All four manifestations of DIDMOAD were found in 46.7% of patients. Differently combined WS1 clinical features were detected in 53.3% of patients. We found 35 WFS1 different mutations and a novel missense mutation, c.1523A>G. WS1 patients were homozygotes or compound heterozygotes for WFS1 mutations except for 2 heterozygote patients (4.5%). Each genotypic group exhibited a different age onset of DM, D, and DI but not of OA. Genotypic Group 2 patients manifested a lower number of clinical manifestations compared to Groups 1 and 3. Moreover, genotypic Group 1 patients tended to have a shorter survival time than the other groups. No differences were found regarding type of clinical pictures. CONCLUSIONS Our study suggested that molecular WFS1 typing is a useful tool for early assessment of clinical history, follow-up, and prognosis of WS1.
Collapse
Affiliation(s)
- Luciana Rigoli
- Department of Human Pathology, University of Messina, Messina, Italy
| | - Concetta Aloi
- LABSIEM (Laboratory for the Study of Inborn Errors of Metabolism), Istituto Giannina Gaslini, Genoa, Italy
| | - Alessandro Salina
- LABSIEM (Laboratory for the Study of Inborn Errors of Metabolism), Istituto Giannina Gaslini, Genoa, Italy
| | - Chiara Di Bella
- Department of Human Pathology, University of Messina, Messina, Italy
| | | | - Rosario Caruso
- Department of Human Pathology, University of Messina, Messina, Italy
| | | | - Mohamad Maghnie
- Department of Pediatrics, University of Genoa, Istituto Giannina Gaslini, Genova, Italy
| | - Giuseppa Patti
- Department of Pediatrics, University of Genoa, Istituto Giannina Gaslini, Genova, Italy
| | | | | |
Collapse
|
40
|
Clinical and genetic analysis of two wolfram syndrome families with high occurrence of wolfram syndrome and diabetes type II: a case report. BMC MEDICAL GENETICS 2020; 21:13. [PMID: 31937257 PMCID: PMC6961406 DOI: 10.1186/s12881-020-0950-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 01/07/2020] [Indexed: 12/21/2022]
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.
Collapse
|
41
|
Tepp K, Puurand M, Timohhina N, Aid-Vanakova J, Reile I, Shevchuk I, Chekulayev V, Eimre M, Peet N, Kadaja L, Paju K, Käämbre T. Adaptation of striated muscles to Wolframin deficiency in mice: Alterations in cellular bioenergetics. Biochim Biophys Acta Gen Subj 2020; 1864:129523. [PMID: 31935437 DOI: 10.1016/j.bbagen.2020.129523] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 12/12/2022]
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.
Collapse
Affiliation(s)
- Kersti Tepp
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia.
| | - Marju Puurand
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia
| | - Natalja Timohhina
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia
| | - Jekaterina Aid-Vanakova
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia
| | - Indrek Reile
- Laboratory of Chemical Physics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia
| | - Igor Shevchuk
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia
| | - Vladimir Chekulayev
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia
| | - Margus Eimre
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411 Tartu, Estonia
| | - Nadežda Peet
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411 Tartu, Estonia
| | - Lumme Kadaja
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411 Tartu, Estonia
| | - Kalju Paju
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411 Tartu, Estonia
| | - Tuuli Käämbre
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia
| |
Collapse
|
42
|
Abstract
Neonatal diabetes mellitus (DM) is defined by the onset of persistent hyperglycemia within the first six months of life but may present up to 12 months of life. A gene mutation affecting pancreatic beta cells or synthesis/secretion of insulin is present in more than 80% of the children with neonatal diabetes. Neonatal DM can be transient, permanent, or be a component of a syndrome. Genetic testing is important as a specific genetic mutation can significantly alter the treatment and outcome. Patients with mutations of either KCNJ11 or ABCC8 that encode subunits of the KATP channel gene mutation can be managed with sulfonylurea oral therapy while patients with other genetic mutations require insulin treatment.
Collapse
Affiliation(s)
- Amanda Dahl
- Division of Pediatric Endocrinology and Metabolism, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - Seema Kumar
- Division of Pediatric Endocrinology and Metabolism, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
- Correspondence: Seema Kumar Division of Pediatric Endocrinology and Metabolism, Department of Pediatric and Adolescent Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN55590, USATel +1 507-284-3300Fax +1 507-284-0727 Email
| |
Collapse
|
43
|
Black DJ, Tran QK, Keightley A, Chinawalkar A, McMullin C, Persechini A. Evaluating Calmodulin-Protein Interactions by Rapid Photoactivated Cross-Linking in Live Cells Metabolically Labeled with Photo-Methionine. J Proteome Res 2019; 18:3780-3791. [PMID: 31483676 DOI: 10.1021/acs.jproteome.9b00510] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
This work addresses the question of how the Ca2+ sensor protein calmodulin shapes cellular responses to Ca2+ signals. Proteins interacting with affinity tagged calmodulin were captured by rapid (t1/2 ≈ 7 s) photoactivated cross-linking under basal conditions, after brief removal of extracellular Ca2+ and during a cytosolic [Ca2+] transient in cells metabolically labeled with a photoreactive methionine analog. Tagged adducts were stringently enriched, and captured proteins were identified and quantified by LC-MS/MS. A set of 489 proteins including 27 known calmodulin interactors was derived. A threshold for fractional capture was applied to define a high specificity group of 170 proteins, including 22 known interactors, and a low specificity group of 319 proteins. Capture of ∼60% of the high specificity group was affected by manipulations of Ca2+, compared with ∼20% of the low specificity group. This suggests that the former is likely to contain novel interactors of physiological significance. The capture of 29 proteins, nearly all high specificity, was decreased by the removal of extracellular Ca2+, although this does not affect cytosolic [Ca2+]. Capture of half of these was unaffected by the cytosolic [Ca2+] transient, consistent with high local [Ca2+]. These proteins are hypothesized to reside in or near microdomains of high [Ca2+] supported by the Ca2+ influx.
Collapse
Affiliation(s)
- D J Black
- Division of Molecular Biology and Biochemistry , University of Missouri-Kansas City , Kansas City , Missouri 64110-2499 , United States
| | | | - Andrew Keightley
- Division of Molecular Biology and Biochemistry , University of Missouri-Kansas City , Kansas City , Missouri 64110-2499 , United States
| | - Ameya Chinawalkar
- Division of Molecular Biology and Biochemistry , University of Missouri-Kansas City , Kansas City , Missouri 64110-2499 , United States
| | - Cole McMullin
- Division of Molecular Biology and Biochemistry , University of Missouri-Kansas City , Kansas City , Missouri 64110-2499 , United States
| | - Anthony Persechini
- Division of Molecular Biology and Biochemistry , University of Missouri-Kansas City , 5007 Rockhill Road , Kansas City , Missouri 64110-2499 , United States
| |
Collapse
|
44
|
Zhang Y, Feng L, Kong X, Wu J, Chen Y, Tian G. Novel mutations and the ophthalmologic characters in Chinese patients with Wolfram Syndrome. Orphanet J Rare Dis 2019; 14:190. [PMID: 31391115 PMCID: PMC6686481 DOI: 10.1186/s13023-019-1161-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 07/19/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Wolfram Syndrome (WFS) is a rare autosomal recessive neurodegenerative disease which has a wide spectrum of manifestations including diabetes insipidus, diabetes mellitus, optic atrophy and deafness. WFS1 and CISD2 are two main causing genes of WFS. The aim of this study was to illustrate the ophthalmologic manifestations and determine the genotype of Chinese WFS patients. RESULTS Completed ophthalmic examinations and family investigations were performed on 4 clinically diagnosed WFS patients from 4 unrelated families. Genetic testing was done by the next generation sequencing of candidate genes. One patient carried a homozygous mutation (c.272_273del) in CISD2, two patients carried compound heterozygous mutations (c.1618 T > G + c.2020G > A and c.1048 T > A + c.2020G > A) in WFS1, and one patient carried a heterozygous mutation (c.937C > T) in WFS1. Three of them were novel mutations. CONCLUSIONS Our study indicated WFS in Chinese is a neurodegenerative disease with both wide spectrum of clinical features and genetic heterogeneity. We found three novel mutations in WFS patients, and to our best knowledge, this is the first report of Chinese WFS patient with mutation in CISD2.
Collapse
Affiliation(s)
- Youjia Zhang
- Department of Ophthalmology and Visual Science, Eye, Ear, Nose and Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China, 83 Fenyang Road, Shanghai, 200031, China
| | - Lili Feng
- Department of Ophthalmology and Visual Science, Eye, Ear, Nose and Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China, 83 Fenyang Road, Shanghai, 200031, China
| | - Xiangmei Kong
- Department of Ophthalmology and Visual Science, Eye, Ear, Nose and Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China, 83 Fenyang Road, Shanghai, 200031, China
| | - Jihong Wu
- Department of Ophthalmology and Visual Science, Eye, Ear, Nose and Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China, 83 Fenyang Road, Shanghai, 200031, China
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Eye Ear Nose and Throat Hospital of Fudan University, 83 Fenyang Road, Shanghai, 200031, China
| | - Yuhong Chen
- Department of Ophthalmology and Visual Science, Eye, Ear, Nose and Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China, 83 Fenyang Road, Shanghai, 200031, China.
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Eye Ear Nose and Throat Hospital of Fudan University, 83 Fenyang Road, Shanghai, 200031, China.
- NHC Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, Eye Ear Nose and Throat Hospital of Fudan University, 83 Fenyang Road, Shanghai, 200031, China.
- Shanghai Key Laboratory of Visual Impairment and Restoration (Fudan University), Eye Ear Nose and Throat Hospital of Fudan University, Shanghai, China, 83 Fenyang Road, Shanghai, 200031, China.
| | - Guohong Tian
- Department of Ophthalmology and Visual Science, Eye, Ear, Nose and Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China, 83 Fenyang Road, Shanghai, 200031, China.
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Eye Ear Nose and Throat Hospital of Fudan University, 83 Fenyang Road, Shanghai, 200031, China.
- NHC Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, Eye Ear Nose and Throat Hospital of Fudan University, 83 Fenyang Road, Shanghai, 200031, China.
- Shanghai Key Laboratory of Visual Impairment and Restoration (Fudan University), Eye Ear Nose and Throat Hospital of Fudan University, Shanghai, China, 83 Fenyang Road, Shanghai, 200031, China.
| |
Collapse
|
45
|
Pallotta MT, Tascini G, Crispoldi R, Orabona C, Mondanelli G, Grohmann U, Esposito S. Wolfram syndrome, a rare neurodegenerative disease: from pathogenesis to future treatment perspectives. J Transl Med 2019; 17:238. [PMID: 31337416 PMCID: PMC6651977 DOI: 10.1186/s12967-019-1993-1] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 07/17/2019] [Indexed: 02/06/2023] Open
Abstract
Background Wolfram syndrome (WS), a rare genetic disorder, is considered the best prototype of endoplasmic reticulum (ER) diseases. Classical WS features are childhood-onset diabetes mellitus, optic atrophy, deafness, diabetes insipidus, neurological signs, and other abnormalities. Two causative genes (WFS1 and WFS2) have been identified. The transmission of the disease takes place in an autosomal recessive mode but autosomal dominant mutations responsible for WS-related disorders have been described. Prognosis is poor, death occurs at the median age of 39 years with a major cause represented by respiratory failure as a consequence of brain stem atrophy and neurodegeneration. The aim of this narrative review is to focus on etiology, pathogenesis and natural history of WS for an adequate patient management and for the discussion of future therapeutic interventions. Main body WS requires a multidisciplinary approach in order to be successfully treated. A prompt diagnosis decreases morbidity and mortality through prevention and treatment of complications. Being a monogenic pathology, WS represents a perfect model to study the mechanisms of ER stress and how this condition leads to cell death, in comparison with other prevalent diseases in which multiple factors interact to produce the disease manifestations. WS is also an important disease prototype to identify drugs and molecules associated with ER homeostasis. Evidence indicates that specific metabolic diseases (type 1 and type 2 diabetes), neurodegenerative diseases, atherosclerosis, inflammatory pathologies and also cancer are closely related to ER dysfunction. Conclusions Therapeutic strategies in WS are based on drug repurposing (i.e., investigation of approved drugs for novel therapeutic indications) with the aim to stop the progression of the disease by reducing the endoplasmic reticulum stress. An extensive understanding of WS from pathophysiology to therapy is fundamental and more studies are necessary to better manage this devastating disease and guarantee the patients a better quality of life and longer life expectancy.
Collapse
Affiliation(s)
- Maria Teresa Pallotta
- Pharmacology Section, Department of Experimental Medicine, Università degli Studi di Perugia, Perugia, Italy
| | - Giorgia Tascini
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Piazza Menghini 1, 06129, Perugia, Italy
| | - Roberta Crispoldi
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Piazza Menghini 1, 06129, Perugia, Italy
| | - Ciriana Orabona
- Pharmacology Section, Department of Experimental Medicine, Università degli Studi di Perugia, Perugia, Italy
| | - Giada Mondanelli
- Pharmacology Section, Department of Experimental Medicine, Università degli Studi di Perugia, Perugia, Italy
| | - Ursula Grohmann
- Pharmacology Section, Department of Experimental Medicine, Università degli Studi di Perugia, Perugia, Italy
| | - Susanna Esposito
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Piazza Menghini 1, 06129, Perugia, Italy.
| |
Collapse
|
46
|
Ustaoglu M, Onder F, Karapapak M, Taslidere H, Guven D. Ophthalmic, systemic, and genetic characteristics of patients with Wolfram syndrome. Eur J Ophthalmol 2019; 30:1099-1105. [DOI: 10.1177/1120672119842489] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Purpose: To evaluate the ophthalmic, systemic, and genetic characteristics of patients with Wolfram syndrome. Methods: In total, 13 patients with suspected or clinically diagnosed Wolfram syndrome underwent ophthalmic and systemic examinations and genetic analyses for Wolfram syndrome between August and October 2018. Results: The mean age of the subjects was 24.2 ± 7.1 years, of which 5 (38.5%) subjects were male and 8 (61.5%) were female. The mean best-corrected visual acuity ranged from counting fingers to 20/40, with a mean of 20/250 (1.10 ± 0.69 logarithm of the minimum angle of resolution). Dyschromatopsia was present in all patients (100%). There was a severe decrease in the average peripapillary retinal nerve fiber layer and macular ganglion cell–inner plexiform layer thicknesses (54.7 ± 6.5 and 51.9 ± 4.8 µm, respectively). Optical coherence tomography angiography showed significantly lower whole-image, inside disk, and peripapillary vessel densities in the patients with Wolfram syndrome than in the healthy controls (p < 0.001 for all). All patients who underwent genetic analyses had mutations in the WFS1 gene. Moreover, two novel mutations, p.Met623Trpfs*2 (c.1867delA) and p.Arg611Profs*9 (c.1832_11847del16) at exon 8, were detected. The frequency of systemic findings was as follows: optic atrophy (100%), diabetes mellitus (92.3%), central diabetes insipidus (38.5%), sensorineural hearing loss (38.5%), and presence of urological (30.8%), psychiatric (30.8%), and neurological (23.1%) diseases. Conclusion: Wolfram syndrome is a rare genetic disorder that can be associated with severe ophthalmic and systemic abnormalities. All patients who present with unexplained optic atrophy should be evaluated for Wolfram syndrome, even if they do not have diabetes mellitus because optic atrophy can sometimes manifest before diabetes mellitus.
Collapse
Affiliation(s)
- Melih Ustaoglu
- Glaucoma Research Center, Wills Eye Hospital, Philadelphia, PA, USA
- Department of Ophthalmology, Sisli Hamidiye Etfal Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Feyza Onder
- Department of Ophthalmology, Haseki Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Murat Karapapak
- Department of Ophthalmology, Sisli Hamidiye Etfal Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Hasan Taslidere
- Department of Medical Genetics, Haseki Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Dilek Guven
- Department of Ophthalmology, Sisli Hamidiye Etfal Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| |
Collapse
|
47
|
Azaiez H, Booth KT, Ephraim SS, Crone B, Black-Ziegelbein EA, Marini RJ, Shearer AE, Sloan-Heggen CM, Kolbe D, Casavant T, Schnieders MJ, Nishimura C, Braun T, Smith RJ. Genomic Landscape and Mutational Signatures of Deafness-Associated Genes. Am J Hum Genet 2018; 103:484-497. [PMID: 30245029 PMCID: PMC6174355 DOI: 10.1016/j.ajhg.2018.08.006] [Citation(s) in RCA: 213] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 08/08/2018] [Indexed: 12/21/2022] Open
Abstract
The classification of genetic variants represents a major challenge in the post-genome era by virtue of their extraordinary number and the complexities associated with ascribing a clinical impact, especially for disorders exhibiting exceptional phenotypic, genetic, and allelic heterogeneity. To address this challenge for hearing loss, we have developed the Deafness Variation Database (DVD), a comprehensive, open-access resource that integrates all available genetic, genomic, and clinical data together with expert curation to generate a single classification for each variant in 152 genes implicated in syndromic and non-syndromic deafness. We evaluate 876,139 variants and classify them as pathogenic or likely pathogenic (more than 8,100 variants), benign or likely benign (more than 172,000 variants), or of uncertain significance (more than 695,000 variants); 1,270 variants are re-categorized based on expert curation and in 300 instances, the change is of medical significance and impacts clinical care. We show that more than 96% of coding variants are rare and novel and that pathogenicity is driven by minor allele frequency thresholds, variant effect, and protein domain. The mutational landscape we define shows complex gene-specific variability, making an understanding of these nuances foundational for improved accuracy in variant interpretation in order to enhance clinical decision making and improve our understanding of deafness biology.
Collapse
|
48
|
Splice-altering variant in COL11A1 as a cause of nonsyndromic hearing loss DFNA37. Genet Med 2018; 21:948-954. [PMID: 30245514 PMCID: PMC6431578 DOI: 10.1038/s41436-018-0285-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 08/16/2018] [Indexed: 02/07/2023] Open
Abstract
PURPOSE The aim of this study was to determine the genetic cause of autosomal dominant nonsyndromic hearing loss segregating in a multigenerational family. METHODS Clinical examination, genome-wide linkage analysis, and exome sequencing were carried out on the family. RESULTS Affected individuals presented with early-onset progressive mild hearing impairment with a fairly flat, gently downsloping or U-shaped audiogram configuration. Detailed clinical examination excluded any additional symptoms. Linkage analysis detected an interval on chromosome 1p21 with a logarithm of the odds (LOD) score of 8.29: designated locus DFNA37. Exome sequencing identified a novel canonical acceptor splice-site variant c.652-2A>C in the COL11A1 gene within the DFNA37 locus. Genotyping of all 48 family members confirmed segregation of this variant with the deafness phenotype in the extended family. The c.652-2A>C variant is novel, highly conserved, and confirmed in vitro to alter RNA splicing. CONCLUSION We have identified COL11A1 as the gene responsible for deafness at the DFNA37 locus. Previously, COL11A1 was solely associated with Marshall and Stickler syndromes. This study expands its phenotypic spectrum to include nonsyndromic deafness. The implications of this discovery are valuable in the clinical diagnosis, prognosis, and treatment of patients with COL11A1 pathogenic variants.
Collapse
|
49
|
Soares A, Mota Á, Fonseca S, Faria O, Brandão E, Falcão Dos Reis F, Gentil R, Guimarães S, Mendonça L. Ophthalmologic Manifestations of Wolfram Syndrome: Report of 14 Cases. Ophthalmologica 2018; 241:116-119. [PMID: 30056456 DOI: 10.1159/000490535] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 05/31/2018] [Indexed: 11/19/2022]
Abstract
PURPOSE The aim of this study was to describe ophthalmological abnormalities in 14 cases of Wolfram syndrome belonging to 9 different families. METHODS Patients were submitted to a complete ophthalmological, neurological, otorhinolaryngological, urological, and genetic evaluation. RESULTS Our sample comprised 14 Caucasian patients belonging to 9 different families. Their ages ranged from 10 to 38 years. The mean duration of known disease was 11.3 ± 8.7 years. Genetic confirmation was obtained in 7 families. There was a parental consanguinity history in 2 families. Five families were homozygous for a mutation of exon 8 of the WFS1 gene (Chr. 4), and 2 patients were heterozygous. Diabetes mellitus was the first manifestation in all except 1 patient. The mean age at diagnosis was 8.7 years (range 3-22). None had diabetic retinopathy. The mean age at diagnosis of optic atrophy was 11.1 years (range 8-35). The best-corrected visual acuity ranged from counting fingers to 20/50. CONCLUSIONS Association of optic atrophy with insulin-dependent diabetes mellitus should raise the suspicion of Wolfram syndrome.
Collapse
Affiliation(s)
| | - Ágata Mota
- Department of Ophthalmology, Hospital Pedro Hispano, Matosinhos, Portugal
| | - Sofia Fonseca
- Department of Ophthalmology, Centro Hospitalar de Vila Nova de Gaia, Gaia, Portugal
| | - Olinda Faria
- Department of Ophthalmology, Centro Hospitalar de São João, Porto, Portugal
| | - Elisete Brandão
- Department of Ophthalmology, Centro Hospitalar de São João, Porto, Portugal
| | | | - Rita Gentil
- Department of Ophthalmology, Hospital de Braga, Braga, Portugal
| | | | - Luís Mendonça
- Department of Ophthalmology, Hospital de Braga, Braga, Portugal
| |
Collapse
|
50
|
Rove KO, Vricella GJ, Hershey T, Thu MH, Lugar HM, Vetter J, Marshall BA, Austin PF. Lower Urinary Tract Dysfunction and Associated Pons Volume in Patients with Wolfram Syndrome. J Urol 2018; 200:1107-1113. [PMID: 29883657 DOI: 10.1016/j.juro.2018.06.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/29/2018] [Indexed: 01/23/2023]
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.
Collapse
Affiliation(s)
- Kyle O Rove
- Division of Pediatric Urology, St. Louis Children's Hospital, Washington University School of Medicine, St. Louis, Missouri.
| | - Gino J Vricella
- Division of Pediatric Urology, St. Louis Children's Hospital, Washington University School of Medicine, St. Louis, Missouri
| | - Tamara Hershey
- Department of Neurology and Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Muang H Thu
- Division of Pediatric Urology, St. Louis Children's Hospital, Washington University School of Medicine, St. Louis, Missouri
| | - Heather M Lugar
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri
| | - Joel Vetter
- Division of Pediatric Urology, St. Louis Children's Hospital, Washington University School of Medicine, St. Louis, Missouri
| | - Bess A Marshall
- Departments of Pediatrics and Cell Biology, St. Louis Children's Hospital, Washington University School of Medicine, St. Louis, Missouri
| | - Paul F Austin
- Scott Department of Urology, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas
| |
Collapse
|