1
|
Leerink CW, Aksnes H. Fahr's disease or primary familial brain calcification? TIDSSKRIFT FOR DEN NORSKE LEGEFORENING 2025; 145:24-0606. [PMID: 40272127 DOI: 10.4045/tidsskr.24.0606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025] Open
|
2
|
Magrinelli F, Jesuthasan A, Bhatia KP, Batla A. Basal ganglia calcification: 'Fahr's disease'. Pract Neurol 2025:pn-2024-004258. [PMID: 40169250 DOI: 10.1136/pn-2024-004258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2025] [Indexed: 04/03/2025]
Abstract
Brain calcification is often detected incidentally, but basal ganglia calcification has a wide differential diagnosis, including genetic and acquired causes. Primary familial brain calcification (PFBC) (formerly 'Fahr's disease') refers to neurological disorders characterised by bilateral, symmetrical deposition of calcium-hydroxyapatite crystals in the basal ganglia and other encephalic regions, with a presumed genetic basis. Its clinical picture encompasses motor, cognitive and psychiatric manifestations in various combinations. Seven genes have been linked to PFBC since 2012, with either autosomal dominant (SLC20A2, PDGFRB, PDGFB and XPR1) or recessive (MYORG, JAM2 and NAA60) mode of inheritance. Mendelian gene discovery has provided critical insights into the pathogenesis of PFBC. Dyshomeostasis of inorganic phosphate, impaired endothelial functions and disrupted blood-brain barrier integrity has been identified as converging pathomechanisms, which could highlight the targets of potential disease-modifying treatments. We provide a state-of-the-art overview on phenotypic features, diagnosis, aetiopathogenesis and management of PFBC.
Collapse
Affiliation(s)
- Francesca Magrinelli
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | | | - Kailash P Bhatia
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Amit Batla
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- Department of Neurology, Royal Free London NHS Foundation Trust Hospital, London, UK
| |
Collapse
|
3
|
Emmi A, Bonato G, Tushevski A, Bertolin C, Cavallieri F, Porzionato A, Antonini A, Salviati L, Carecchio M. Skin calcium deposits in primary familial brain calcification: A novel potential biomarker. Ann Clin Transl Neurol 2025; 12:737-745. [PMID: 39935191 PMCID: PMC12040503 DOI: 10.1002/acn3.52304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/29/2024] [Accepted: 12/22/2024] [Indexed: 02/13/2025] Open
Abstract
OBJECTIVE Primary Familial Brain Calcification (PFBC) is a rare neurodegenerative disorder characterized by small vessel calcifications in the basal ganglia. PFBC is caused by pathogenic variants in different genes and its physiopathology is still largely unknown. Skin vascular calcifications have been detected in single PFBC cases, suggesting that calcium deposition may not be limited to the brain, but it is unknown whether this is a hallmark of all PFBC genetic and clinical subtypes. This work aims at assessing anatomical and subcellular localization of calcium-phosphate deposits in skin biopsies from PFBC patients to ascertain the accuracy of histological calcium staining in differentiating PFBC from healthy controls (HC) and Parkinson's Disease (PD). METHODS Histopathology and light microscopy of skin biopsy from 20 PFBC, 7 HC and 10 PD subjects (3 mm ø-5 mm deep punch biopsies, Hematoxylin-Eosin and vonKossa staining, immunoperoxidase CD31 staining); clinical, genetic and radiological assessment. RESULTS Unlike HC and PD subjects, the majority of PFBC patients (17/20) showed a consistent pattern of granular argyrophilic calcium-phosphate deposits in the basal lamina and the cytoplasm of CD31+ endothelial cells and pericytes of dermal capillaries, and the basement membrane of sweat glands. This pattern was unrelated to the underlying mutated gene or clinical status. INTERPRETATION Skin biopsy may be a novel PFBC diagnostic tool and a potential biomarker for future therapies, and a tool to investigate PFBC disease mechanisms. Different findings in some patients could be due to skin sampling variability and biological consequences of specific PFBC gene variants.
Collapse
Affiliation(s)
- Aron Emmi
- Institute of Human Anatomy, Department of NeurosciencesUniversity of PadovaPadova35129Italy
- Parkinson and Movement Disorders Unit, Department of Neuroscience, Centre for Rare Neurological Diseases (ERN‐RND)University of PadovaPadova35128Italy
- Center for Neurodegenerative Disease Research (CESNE)University of PadovaPadova35128Italy
| | - Giulia Bonato
- Parkinson and Movement Disorders Unit, Department of Neuroscience, Centre for Rare Neurological Diseases (ERN‐RND)University of PadovaPadova35128Italy
- Center for Neurodegenerative Disease Research (CESNE)University of PadovaPadova35128Italy
| | - Aleksandar Tushevski
- Institute of Human Anatomy, Department of NeurosciencesUniversity of PadovaPadova35129Italy
| | - Cinzia Bertolin
- Clinical Genetics UnitUniversity Hospital of PadovaPadova35128Italy
| | - Francesco Cavallieri
- Neurology Unit, Neuromotor and Rehabilitation DepartmentAzienda USL‐IRCCS di Reggio EmiliaReggio Emilia42123Italy
| | - Andrea Porzionato
- Institute of Human Anatomy, Department of NeurosciencesUniversity of PadovaPadova35129Italy
| | - Angelo Antonini
- Parkinson and Movement Disorders Unit, Department of Neuroscience, Centre for Rare Neurological Diseases (ERN‐RND)University of PadovaPadova35128Italy
- Center for Neurodegenerative Disease Research (CESNE)University of PadovaPadova35128Italy
| | - Leonardo Salviati
- Clinical Genetics UnitUniversity Hospital of PadovaPadova35128Italy
- Clinical Genetics Unit, Department of Women's and Children's HealthUniversity of PadovaPadova35128Italy
| | - Miryam Carecchio
- Parkinson and Movement Disorders Unit, Department of Neuroscience, Centre for Rare Neurological Diseases (ERN‐RND)University of PadovaPadova35128Italy
- Center for Neurodegenerative Disease Research (CESNE)University of PadovaPadova35128Italy
| |
Collapse
|
4
|
Soleimani P, Khojasteh M, Ghasemi A, Heshmati A, Rohani M, Alavi A. Mutation spectrum and clinical features of MYORG in Iranian patients with Primary Familial Brain Calcification (PFBC). Neurol Sci 2025:10.1007/s10072-025-08105-x. [PMID: 40120050 DOI: 10.1007/s10072-025-08105-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 03/05/2025] [Indexed: 03/25/2025]
Abstract
INTRODUCTION Mutations in myogenesis regulating glycosidase (MYORG), result in autosomal recessive (AR) form of Primary Familial Brain Calcification (PFBC) which is a rare neurodegenerative disease. PFBC is characterized by symmetric brain calcifications, particularly in the thalami, cerebellum, basal ganglia, and subcortical white matter. To date, eight genes have been linked with PFBC, however, currently about half of people with PFBC remain without a genetic diagnosis. Among these genes, MYORG, JAM2, CMPK2, and NAA60 are associated with an AR-PFBC. Within AR-PFBCs, the frequency of mutations in MYORG and JAM2 is 13% and 2%, respectively. In this study, we present a comprehensive clinical and genetic analysis of a group of Iranian PFBC patients. METHODS Clinical and paraclinical assessments of all patients were done. Whole-exome sequencing was performed for all probands. Candidate variants were confirmed and checked in their family members. RESULTS Four homozygous variants in MYORG across four families were identified: two novel variants, c.1727G > A;p.Arg576His and c.1687del;p.The563Glnfs*191, in two families and two known mutations, c.176G > A;p.Gly59Asp and c.1092_1097del;p.Phe365_Asp366del in the remaining two families. A potential SNV/CNV in the PFBC-related genes that causes disease was not detected in one proband. CONCLUSION Our study expanded the clinical features and mutation spectrum of MYORG and emphasizes to genetic heterogeneity in different populations. While SLC20A2 mutations are the common cause of PFBC in other populations, MYORG and JAM2 mutations seem to be the main cause of this disease in Iran. This issue could prove to be advantageous in the process of gene prioritization for screening within this specific population.
Collapse
Affiliation(s)
- Parsa Soleimani
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Mana Khojasteh
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Aida Ghasemi
- Neuromuscular Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Heshmati
- School of Biology, University College of Science, University of Tehran, Tehran, Iran
| | - Mohammad Rohani
- Department of Neurology, The Five Senses Health Institute, Iran University of Medical Sciences, Tehran, Iran.
| | - Afagh Alavi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran.
- Neuromuscular Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Lin J, Wang Y, Gao X, Liu X. Novel Compound Heterozygous Mutation on MYORG Gene in a Chinese Patient with Primary Familial Brain Calcification. Neurol India 2025; 73:365-367. [PMID: 40176235 DOI: 10.4103/neurol-india.neurol-india-d-24-00551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 09/06/2024] [Indexed: 04/04/2025]
Affiliation(s)
- Jixiang Lin
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, People's Republic of China
| | - Yanfei Wang
- Department of Nursing, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, People's Republic of China
| | - Xiang Gao
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, People's Republic of China
| | - Xiaomin Liu
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, People's Republic of China
| |
Collapse
|
6
|
Bekkelund ÅK, Siggervåg A, Aksnes H. P i-ecing together brain calcification mechanisms for therapeutic advancement. Trends Mol Med 2025; 31:206-208. [PMID: 39755442 DOI: 10.1016/j.molmed.2024.12.003] [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: 11/18/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 01/06/2025]
Abstract
Seven primary familial brain calcification genes have been identified but their role in disease mechanisms has been less explored. Cheng et al. recently demonstrated that astrocyte-mediated regulation of brain phosphate (Pi) involves direct and functional interactions among three of these proteins, paving the way for new strategies to combat brain calcification.
Collapse
Affiliation(s)
- Åse K Bekkelund
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | - Henriette Aksnes
- Department of Biomedicine, University of Bergen, Bergen, Norway.
| |
Collapse
|
7
|
Yang D, Huang H, Zeng T, Wang L, Ying C, Chen X, Zhou X, Sun F, Chen Y, Li S, Wang B, Wu S, Xie F, Cen Z, Luo W. Unveiling distinct clinical manifestations of primary familial brain calcifications in Asian and European patients: A study based on 10-year individual-level data. Parkinsonism Relat Disord 2025; 132:107290. [PMID: 39827654 DOI: 10.1016/j.parkreldis.2025.107290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/06/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Primary Familial Brain Calcification (PFBC) can manifest clinically with a complex and heterogeneous array of symptoms, including parkinsonism, dysarthria, and cognitive impairment. However, the distinct presentations of PFBC in Asian and European populations remain unclear. METHODS We conducted a systematic search of PubMed for studies involving genetically confirmed PFBC patients. Demographic data, genetic information, radiological examinations, and clinical characteristics were extracted for each case. RESULTS The study included 120 publications and 564 genetically confirmed PFBC patients. Asian and European PFBC populations represented 54 % and 37 % of global patients, respectively. While calcification patterns showed no significant differences between Asian and European PFBC patients, European autosomal dominant PFBC variant carriers were more likely to exhibit clinical symptoms compared to their Asian counterparts (OR = 2.90, 95 % CI 1.55-5.60) and had an earlier estimated age of onset (median age 42 vs 58). CONCLUSION The interaction between regional differences and genetically determined calcification severity may collectively influence PFBC symptom progression. Future research should further explore the potential roles of gene modifiers, ethnic background, socioeconomic and environmental exposure factors underlying regional differences in PFBC progression.
Collapse
Affiliation(s)
- Dehao Yang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Honghao Huang
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tian Zeng
- The First School of Medicine, School of Information and Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lebo Wang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chenxin Ying
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xinhui Chen
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xinbo Zhou
- The First School of Medicine, School of Information and Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Fangyue Sun
- The First School of Medicine, School of Information and Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yilin Chen
- The First School of Medicine, School of Information and Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shengqi Li
- The First School of Medicine, School of Information and Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Bo Wang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Sheng Wu
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Fei Xie
- Department of Neurology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhidong Cen
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Wei Luo
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
8
|
Yang D, Jiang Z, Huang H, Wang L, Ying C, Chen Y, Lu Y, Zhang T, Zhu Y, Wang S, Wang Y, Guo Y, Wang H, Cen Z, Luo W. Genetic Mutations in Cell Junction Proteins Associated with Brain Calcification. Mov Disord 2025; 40:400-419. [PMID: 39620489 DOI: 10.1002/mds.30068] [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: 07/19/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 03/22/2025] Open
Abstract
Intracerebral calcium deposition, classified into primary familial brain calcification (PFBC) and secondary brain calcification, occurs within the brain parenchyma and vasculature. PFBC manifests with progressive motor decline, dysarthria, and cognitive impairment, with limited treatment options available. Recent research has suggested a link between dysfunction of the blood-brain barrier (BBB) and PFBC, with certain genetic variants potentially affecting neurovascular unit (NVU) function, thereby contributing to BBB integrity disruption and brain calcification. Cell junctions play an indispensable role in maintaining the function of NVUs. The pathogenic mechanisms of PFBC-causative genes, such as PDGFRB, PDGFB, MYORG, and JAM2, involve NVU disruption. Cell junctions, such as tight junctions, gap junctions, adherens junctions, desmosomes, hemidesmosomes, and focal adhesions, are vital for cell-cell and cell-extracellular matrix connections, maintaining barrier function, cell adhesion, and facilitating ion and metabolite exchange. Several recent studies have highlighted the role of mutations in genes encoding cell junction proteins in the onset and progression of brain calcification and its related phenotypes. This emerging body of research offers a unique perspective for investigating the underlying mechanisms driving brain calcification. In this review, we conducted an examination of the literature reporting on genetic variants in cell junction proteins associated with brain calcification to delineate potential molecular pathways and investigate genotype-phenotype correlations. This approach not only reinforces the rationale for molecular subtyping of brain calcification but also lays the groundwork for the discovery of novel causative genes involved in pathogenesis. © 2024 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Dehao Yang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zihan Jiang
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Honghao Huang
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lebo Wang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chenxin Ying
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiqun Chen
- The First School of Medicine, School of Information and Engineering, Wenzhou Medical University, Wenzhou, China
| | - Yangguang Lu
- The First School of Medicine, School of Information and Engineering, Wenzhou Medical University, Wenzhou, China
| | - Tingxuan Zhang
- Renji College, Wenzhou Medical University, Wenzhou, China
| | - Yusheng Zhu
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Shiyue Wang
- The First School of Medicine, School of Information and Engineering, Wenzhou Medical University, Wenzhou, China
| | - Yaoting Wang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Chu Kochen Honors College, Zhejiang University, Hangzhou, China
| | - Yuru Guo
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Chu Kochen Honors College, Zhejiang University, Hangzhou, China
| | - Haoyu Wang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Chu Kochen Honors College, Zhejiang University, Hangzhou, China
| | - Zhidong Cen
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Luo
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
9
|
Zhang W, Chen Y, Guan Z, Wang Y, Tang M, Du Z, Zhang J, Cheng M, Zuo J, Liu Y, Wang Q, Liu Y, Zhang D, Yin P, Ma L, Liu Z. Structural insights into the mechanism of phosphate recognition and transport by XPR1. Nat Commun 2025; 16:18. [PMID: 39747008 PMCID: PMC11696373 DOI: 10.1038/s41467-024-55471-9] [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: 08/27/2024] [Accepted: 12/11/2024] [Indexed: 01/04/2025] Open
Abstract
XPR1 is the sole protein known to transport inorganic phosphate (Pi) out of cells, a function conserved across species from yeast to mammals. Human XPR1 variants lead to cerebral calcium-phosphate deposition and primary familial brain calcification (PFBC), a hereditary neurodegenerative disorder. Here, we present the cryo-EM structure of human XPR1 in both its Pi-unbound and various Pi-bound states. XPR1 features 10 transmembrane α-helices forming an ion channel-like structure, with multiple Pi recognition sites along the channel. Pathogenic mutations in two arginine residues, which line the translocation channel, disrupt Pi transport. Molecular dynamics simulations reveal that Pi ion undergoes a stepwise transition through the sequential recognition sites during the transport process. Together with functional analyses, our results suggest that this sequential arrangement allows XPR1 to facilitate Pi ion passage via a "relay" process, and they establish a framework for the interpretation of disease-related mutations and for the development of future therapeutics.
Collapse
Affiliation(s)
- Wenhui Zhang
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Yanke Chen
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Zeyuan Guan
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Yong Wang
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Meng Tang
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Zhangmeng Du
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Jie Zhang
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Meng Cheng
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Jiaqi Zuo
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Yan Liu
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Qiang Wang
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Yanjun Liu
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Delin Zhang
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Ping Yin
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Ling Ma
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Zhu Liu
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China.
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.
| |
Collapse
|
10
|
Kavakli E, Gul N, Begentas OC, Kiris E. Astrocytes in Primary Familial Brain Calcification (PFBC): Emphasis on the Importance of Induced Pluripotent Stem Cell-Derived Human Astrocyte Models. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1479:19-38. [PMID: 39841380 DOI: 10.1007/5584_2024_840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
Primary familial brain calcification (PFBC) is a rare, progressive central nervous system (CNS) disorder without a cure, and the current treatment methodologies primarily aim to relieve neurological and psychiatric symptoms of the patients. The disease is characterized by abnormal bilateral calcifications in the brain, however, our mechanistic understanding of the biology of the disease is still limited. Determining the roles of the specific cell types and molecular mechanisms involved in the pathophysiological processes of the disease is of great importance for the development of novel and effective treatment methodologies. There is a growing interest in the involvement of astrocytes in PFBC, as recent studies have suggested that astrocytes play a central role in the disease and that functional defects in these cells are critical for the development and progression of the disease. This review aims to discuss recent findings on the roles of astrocytes in PFBC pathophysiology, with a focus on known expression and roles of PFBC genes in astrocytes. Additionally, we discuss the importance of human astrocytes for PFBC disease modeling, and astrocytes as a potential therapeutic target in PFBC. Utilization of species-specific and physiologically relevant PFBC model systems can open new avenues for basic research, drug development, and regenerative medicine.
Collapse
Affiliation(s)
- Ebru Kavakli
- Department of Biological Sciences, Middle East Technical University, Ankara, Türkiye
| | - Nazli Gul
- Department of Biological Sciences, Middle East Technical University, Ankara, Türkiye
| | - Onur Can Begentas
- Department of Biological Sciences, Middle East Technical University, Ankara, Türkiye
| | - Erkan Kiris
- Department of Biological Sciences, Middle East Technical University, Ankara, Türkiye.
| |
Collapse
|
11
|
Bijnen M, Sridhar S, Keller A, Greter M. Brain macrophages in vascular health and dysfunction. Trends Immunol 2025; 46:46-60. [PMID: 39732528 DOI: 10.1016/j.it.2024.11.012] [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: 10/03/2024] [Revised: 11/08/2024] [Accepted: 11/19/2024] [Indexed: 12/30/2024]
Abstract
Diverse macrophage populations inhabit the rodent and human central nervous system (CNS), including microglia in the parenchyma and border-associated macrophages (BAMs) in the meninges, choroid plexus, and perivascular spaces. These innate immune phagocytes are essential in brain development and maintaining homeostasis, but they also play diverse roles in neurological diseases. In this review, we highlight the emerging roles of CNS macrophages in regulating vascular function in health and disease. We discuss that, in addition to microglia, BAMs, including perivascular macrophages, play roles in supporting vascular integrity and maintaining blood flow. We highlight recent advancements in understanding how these macrophages are implicated in protecting against vascular dysfunction and modulating the progression of cerebrovascular diseases, as seen in vessel-associated neurodegeneration.
Collapse
Affiliation(s)
- Mitchell Bijnen
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Sucheta Sridhar
- Department of Neurosurgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Annika Keller
- Department of Neurosurgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Melanie Greter
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
12
|
Sridhar S, Zhou Y, Ibrahim A, Bertazzo S, Wyss T, Swain A, Maheshwari U, Huang SF, Colonna M, Keller A. Targeting TREM2 signaling shows limited impact on cerebrovascular calcification. Life Sci Alliance 2025; 8:e202402796. [PMID: 39467636 PMCID: PMC11519321 DOI: 10.26508/lsa.202402796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/30/2024] Open
Abstract
Brain calcification, the ectopic mineral deposits of calcium phosphate, is a frequent radiological finding and a diagnostic criterion for primary familial brain calcification. We previously showed that microglia curtail the growth of small vessel calcification via the triggering receptor expressed in myeloid 2 (TREM2) in the Pdgfb ret/ret mouse model of primary familial brain calcification. Because boosting TREM2 function using activating antibodies has been shown to be beneficial in other disease conditions by aiding in microglial clearance of diverse pathologies, we investigated whether administration of a TREM2-activating antibody could mitigate vascular calcification in Pdgfb ret/ret mice. Single-nucleus RNA-sequencing analysis showed that calcification-associated microglia share transcriptional similarities to disease-associated microglia and exhibited activated TREM2 and TGFβ signaling. Administration of a TREM2-activating antibody increased TREM2-dependent microglial deposition of cathepsin K, a collagen-degrading protease, onto calcifications. However, this did not ameliorate the calcification load or alter the mineral composition and the microglial phenotype around calcification. We therefore conclude that targeting microglia with TREM2 agonistic antibodies is insufficient to demineralize and clear vascular calcifications.
Collapse
Affiliation(s)
- Sucheta Sridhar
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Neuroscience Centre Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Yingyue Zhou
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | | | - Sergio Bertazzo
- Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - Tania Wyss
- TDS-facility, AGORA Cancer Research Center, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Amanda Swain
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Upasana Maheshwari
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Sheng-Fu Huang
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Annika Keller
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Neuroscience Centre Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
13
|
Balck A, Klein C. First Evidence for Disease-Modifying Treatment of SLC20A2-Related Primary Familial Brain Calcification. Mov Disord 2024; 39:2180. [PMID: 39428678 DOI: 10.1002/mds.30034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/16/2024] [Accepted: 10/03/2024] [Indexed: 10/22/2024] Open
Affiliation(s)
- Alexander Balck
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
- Department of Neurology, University of Lübeck and University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| |
Collapse
|
14
|
Chen X, Shi Y, Fu F, Wang L, Yu H, Yang D, Wang X, Ying C, Wang H, Lin Z, Wang H, Zhang F, Zheng X, Guo Y, Wang Y, Zeng Y, Zhao M, Chen Y, Li J, Xia H, Chen J, Wang B, Wu S, Xie F, Feng J, Cen Z, Luo W. A Homozygous Variant in NAA60 Is Associated with Primary Familial Brain Calcification. Mov Disord 2024; 39:2190-2198. [PMID: 39229657 DOI: 10.1002/mds.30004] [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: 04/28/2024] [Revised: 07/04/2024] [Accepted: 08/08/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Primary familial brain calcification (PFBC) is a monogenic disorder characterized by bilateral calcifications in the brain. The genetic basis remains unknown in over half of the PFBC patients, indicating the existence of additional novel causative genes. NAA60 was a recently reported novel causative gene for PFBC. OBJECTIVE The aim was to identify the probable novel causative gene in an autosomal recessive inherited PFBC family. METHODS We performed a comprehensive genetic study on a consanguineous Chinese family with 3 siblings diagnosed with PFBC. We evaluated the effect of the variant in a probable novel causative gene on the protein level using Western blot, immunofluorescence, and coimmunoprecipitation. Possible downstream pathogenic mechanisms were further explored in gene knockout (KO) cell lines and animal models. RESULTS We identified a PFBC co-segregated homozygous variant of c.460_461del (p.D154Lfs*113) in NAA60. Functional assays showed that this variant disrupts NAA60 protein localization to Golgi and accelerated protein degradation. The mutant NAA60 protein alters its interaction with the PFBC-related proteins PiT2 and XPR1, affecting intracellular phosphate homeostasis. Further mass spectrometry analysis in NAA60 KO cell lines revealed decreased expression of multiple brain calcification-associated proteins, including reduced folate carrier (RFC), a folate metabolism-related protein. CONCLUSIONS Our study replicated the identification of NAA60 as a novel causative gene for autosomal recessive PFBC, demonstrating our causative variant leads to NAA60 loss of function. The NAA60 loss of function disrupts not only PFBC-related proteins (eg, PiT2 and XPR1) but also a wide range of other brain calcification-associated membrane protein substrates (eg, RFC), and provided a novel probable pathogenic mechanism for PFBC. © 2024 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Xinhui Chen
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yihua Shi
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Feng Fu
- Department of Neurology, Zhuji People's Hospital of Zhejiang Province, Shaoxing, China
| | - Lebo Wang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hongying Yu
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Neurology, Affiliated-Hospital of Shaoxing University, Shaoxing, China
| | - Dehao Yang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinchen Wang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chenxin Ying
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haoyu Wang
- Chu Kochen Honors College, Zhejiang University, Hangzhou, China
| | - Zhiru Lin
- Department of Neurology, Wenzhou Central Hospital, Wenzhou, China
| | - Haotian Wang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fan Zhang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaosheng Zheng
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuru Guo
- Chu Kochen Honors College, Zhejiang University, Hangzhou, China
| | - Yaoting Wang
- Chu Kochen Honors College, Zhejiang University, Hangzhou, China
| | - YiHeng Zeng
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Miao Zhao
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Yiling Chen
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaxiang Li
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haibin Xia
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiawen Chen
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bo Wang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Sheng Wu
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fei Xie
- Department of Neurology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianhua Feng
- Department of Paediatrics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhidong Cen
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Luo
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
15
|
El-Dehaibi F, Zamora R, Yin J, Namas RA, Billiar TR, Vodovotz Y. NETWORK ANALYSIS OF SINGLE-NUCLEOTIDE POLYMORPHISMS ASSOCIATED WITH ABERRANT INFLAMMATION IN TRAUMA PATIENTS SUGGESTS A ROLE FOR VESICLE-ASSOCIATED INFLAMMATORY PROGRAMS INVOLVING CD55. Shock 2024; 62:663-672. [PMID: 39178207 DOI: 10.1097/shk.0000000000002448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2024]
Abstract
ABSTRACT Background: Critical illness stemming from severe traumatic injury is a leading cause of morbidity and mortality worldwide and involves the dysfunction of multiple organ systems, driven, at least in part, by dysregulated inflammation. We and others have shown a key role for genetic predisposition to dysregulated inflammation and downstream adverse critical illness outcomes. Recently, we demonstrated an association among genotypes at the single-nucleotide polymorphism (SNP) rs10404939 in LYPD4 , dysregulated systemic inflammation, and adverse clinical outcomes in a broad sample of ~1,000 critically ill patients. Methods: We sought to gain mechanistic insights into the role of LYPD4 in critical illness by bioinformatically analyzing potential interactions among rs10404939 and other SNPs. We analyzed a dataset of common (i.e., not rare) SNPs previously defined to be associated with genotype-specific, significantly dysregulated systemic inflammation trajectories in trauma patients, in comparison to a control dataset of common SNPs determined to exhibit an absence of genotype-specific inflammatory responses. Results: In the control dataset, this analysis implicated SNPs associated with phosphatidylinositol and various membrane transport proteins, but not LYPD4. In the patient subset with genotypically dysregulated inflammation, our analysis suggested the co-localization to lipid rafts of LYPD4 and the complement receptor CD55, as well as the neurally related CNTNAP2 and RIMS4. Segregation of trauma patients based on genotype of the CD55 SNP rs11117564 showed distinct trajectories of organ dysfunction and systemic inflammation despite similar demographics and injury characteristics. Conclusion: These analyses define novel interactions among SNPs that could enhance our understanding of the response to traumatic injury and critical illness.
Collapse
Affiliation(s)
- Fayten El-Dehaibi
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | | | | | | | | |
Collapse
|
16
|
Zhao M, Cheng X, Chen L, Zeng YH, Lin KJ, Li YL, Zheng ZH, Huang XJ, Zuo DD, Guo XX, Guo J, He D, Liu Y, Lin Y, Wang C, Lv WQ, Su HZ, Yao XP, Ye ZL, Chen XH, Lu YQ, Huang CW, Yang G, Zhang YX, Lin MT, Wang N, Xiong ZQ, Chen WJ. Antisense oligonucleotides enhance SLC20A2 expression and suppress brain calcification in a humanized mouse model. Neuron 2024; 112:3278-3294.e7. [PMID: 39121859 DOI: 10.1016/j.neuron.2024.07.013] [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: 07/18/2023] [Revised: 05/15/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024]
Abstract
Primary familial brain calcification (PFBC) is a genetic neurological disease, yet no effective treatment is currently available. Here, we identified five novel intronic variants in SLC20A2 gene from six PFBC families. Three of these variants increased aberrant SLC20A2 pre-mRNA splicing by altering the binding affinity of splicing machineries to newly characterized cryptic exons, ultimately causing premature termination of SLC20A2 translation. Inhibiting the cryptic-exon incorporation with splice-switching ASOs increased the expression levels of functional SLC20A2 in cells carrying SLC20A2 mutations. Moreover, by knocking in a humanized SLC20A2 intron 2 sequence carrying a PFBC-associated intronic variant, the SLC20A2-KI mice exhibited increased inorganic phosphate (Pi) levels in cerebrospinal fluid (CSF) and progressive brain calcification. Intracerebroventricular administration of ASOs to these SLC20A2-KI mice reduced CSF Pi levels and suppressed brain calcification. Together, our findings expand the genetic etiology of PFBC and demonstrate ASO-mediated splice modulation as a potential therapy for PFBC patients with SLC20A2 haploinsufficiency.
Collapse
Affiliation(s)
- Miao Zhao
- Department of Neurology, the First Affiliated Hospital, Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou 350005, China; Department of Neurology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350212, China
| | - Xuewen Cheng
- Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience and State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China; Lin Gang Laboratory, Shanghai 201602, China.
| | - Lei Chen
- Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience and State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China; School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi-Heng Zeng
- Department of Neurology, the First Affiliated Hospital, Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou 350005, China
| | - Kai-Jun Lin
- Department of Neurology, the First Affiliated Hospital, Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou 350005, China
| | - Yun-Lu Li
- Department of Neurology, the First Affiliated Hospital, Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou 350005, China
| | - Ze-Hong Zheng
- Department of Neurology, the First Affiliated Hospital, Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou 350005, China
| | - Xue-Jing Huang
- Department of Neurology, the First Affiliated Hospital, Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou 350005, China
| | - Dan-Dan Zuo
- Department of Neurology, the First Affiliated Hospital, Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou 350005, China
| | - Xin-Xin Guo
- Department of Neurology, the First Affiliated Hospital, Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou 350005, China
| | - Jun Guo
- Department of Neurology, Tangdu Hospital, Air Force Medical University, Xi'an 710038, China
| | - Dian He
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guiyang 550001, China
| | - Ying Liu
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guiyang 550001, China
| | - Yu Lin
- Department of Neurology, the First Affiliated Hospital, Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou 350005, China
| | - Chong Wang
- Department of Neurology, the First Affiliated Hospital, Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou 350005, China
| | - Wen-Qi Lv
- Department of Neurology, the First Affiliated Hospital, Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou 350005, China
| | - Hui-Zhen Su
- Department of Neurology, the First Affiliated Hospital, Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou 350005, China; Department of Neurology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350212, China
| | - Xiang-Ping Yao
- Department of Neurology, the First Affiliated Hospital, Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou 350005, China; Department of Neurology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350212, China
| | - Zi-Ling Ye
- Department of Neurology, the First Affiliated Hospital, Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou 350005, China
| | - Xiao-Hong Chen
- Department of Neurology, the First Affiliated Hospital, Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou 350005, China
| | - Ying-Qian Lu
- Department of Neurology, the First Affiliated Hospital, Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou 350005, China
| | - Chen-Wei Huang
- Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience and State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China; School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guang Yang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Yu-Xian Zhang
- Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience and State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China
| | - Min-Ting Lin
- Department of Neurology, the First Affiliated Hospital, Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou 350005, China; Department of Neurology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350212, China
| | - Ning Wang
- Department of Neurology, the First Affiliated Hospital, Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou 350005, China; Department of Neurology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350212, China
| | - Zhi-Qi Xiong
- Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience and State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China; School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, China; Shanghai Center for Brain Science and Brain-inspired Technology, Shanghai 201602, China.
| | - Wan-Jin Chen
- Department of Neurology, the First Affiliated Hospital, Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou 350005, China; Department of Neurology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350212, China.
| |
Collapse
|
17
|
Betsholtz C. Insights into mechanisms and therapeutic avenues for primary familial brain calcification. Neuron 2024; 112:3223-3225. [PMID: 39389009 DOI: 10.1016/j.neuron.2024.09.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
The diverse etiologies of the genetic neurodegenerative disorder known as primary familial brain calcification have dimmed hopes for curative therapies. However, two new papers in Neuron1,2 provide a reason for optimism by identifying mechanisms involved in brain phosphate transport and a promising target for restoring phosphate balance in the brain.
Collapse
Affiliation(s)
- Christer Betsholtz
- Department of Medicine Huddinge, Karolinska Institutet, Flemingsberg Campus, Huddinge, Sweden; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
18
|
Brand M, Westenberger A. Unraveling Primary Familial Brain Calcification Mechanisms: Are NAA60 and SLC20A2 Partners in Crime? Mov Disord 2024; 39:1731. [PMID: 39221593 DOI: 10.1002/mds.30000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Affiliation(s)
- Max Brand
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Ana Westenberger
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| |
Collapse
|
19
|
Cheng X, Zhao M, Chen L, Huang C, Xu Q, Shao J, Wang HT, Zhang Y, Li X, Xu X, Yao XP, Lin KJ, Xue H, Wang H, Chen Q, Zhu YC, Zhou JW, Ge WP, Zhu SJ, Liu JY, Chen WJ, Xiong ZQ. Astrocytes modulate brain phosphate homeostasis via polarized distribution of phosphate uptake transporter PiT2 and exporter XPR1. Neuron 2024; 112:3126-3142.e8. [PMID: 39019040 DOI: 10.1016/j.neuron.2024.06.020] [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: 04/10/2023] [Revised: 05/05/2024] [Accepted: 06/20/2024] [Indexed: 07/19/2024]
Abstract
Aberrant inorganic phosphate (Pi) homeostasis causes brain calcification and aggravates neurodegeneration, but the underlying mechanism remains unclear. Here, we found that primary familial brain calcification (PFBC)-associated Pi transporter genes Pit2 and Xpr1 were highly expressed in astrocytes, with importer PiT2 distributed over the entire astrocyte processes and exporter XPR1 localized to astrocyte end-feet on blood vessels. This polarized PiT2 and XPR1 distribution endowed astrocyte with Pi transport capacity competent for brain Pi homeostasis, which was disrupted in mice with astrocyte-specific knockout (KO) of either Pit2 or Xpr1. Moreover, we found that Pi uptake by PiT2, and its facilitation by PFBC-associated galactosidase MYORG, were required for the high Pi transport capacity of astrocytes. Finally, brain calcification was suppressed by astrocyte-specific PiT2 re-expression in Pit2-KO mice. Thus, astrocyte-mediated Pi transport is pivotal for brain Pi homeostasis, and elevating astrocytic Pi transporter function represents a potential therapeutic strategy for reducing brain calcification.
Collapse
Affiliation(s)
- Xuewen Cheng
- Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience and State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China; Lin Gang Laboratory, Shanghai 201602, China.
| | - Miao Zhao
- Department of Neurology, The First Affiliated Hospital, Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou 350005, China
| | - Lei Chen
- Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience and State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China; Lin Gang Laboratory, Shanghai 201602, China
| | - Chenwei Huang
- Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience and State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiwu Xu
- Lin Gang Laboratory, Shanghai 201602, China
| | - Jia Shao
- Lin Gang Laboratory, Shanghai 201602, China
| | - Hong-Tao Wang
- Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience and State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuxian Zhang
- Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience and State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xuequan Li
- Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience and State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xuan Xu
- Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience and State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiang-Ping Yao
- Department of Neurology, The First Affiliated Hospital, Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou 350005, China
| | - Kai-Jun Lin
- Department of Neurology, The First Affiliated Hospital, Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou 350005, China
| | - Hui Xue
- Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience and State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Han Wang
- Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience and State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qi Chen
- Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience and State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yong-Chuan Zhu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Jia-Wei Zhou
- Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience and State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Woo-Ping Ge
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Shu-Jia Zhu
- Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience and State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing-Yu Liu
- Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience and State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wan-Jin Chen
- Department of Neurology, The First Affiliated Hospital, Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou 350005, China.
| | - Zhi-Qi Xiong
- Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience and State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
20
|
Hobara T, Higuchi Y, Yoshida M, Suehara M, Ando M, Yuan JH, Yoshimura A, Kojima F, Matsuura E, Okamoto Y, Mitsui J, Tsuji S, Takashima H. Genetic and pathophysiological insights from autopsied patient with primary familial brain calcification: novel MYORG variants and astrocytic implications. Acta Neuropathol Commun 2024; 12:136. [PMID: 39180105 PMCID: PMC11342542 DOI: 10.1186/s40478-024-01847-3] [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: 06/19/2024] [Accepted: 08/04/2024] [Indexed: 08/26/2024] Open
Abstract
Primary familial brain calcification (PFBC) is a genetic neurological disorder characterized by symmetric brain calcifications that manifest with variable neurological symptoms. This study aimed to explore the genetic basis of PFBC and elucidate the underlying pathophysiological mechanisms. Six patients from four pedigrees with brain calcification were enrolled. Whole-exome sequencing identified two novel homozygous variants, c.488G > T (p.W163L) and c.2135G > A (p.W712*), within the myogenesis regulating glycosidase (MYORG) gene. Cerebellar ataxia (n = 5) and pyramidal signs (n = 4) were predominant symptoms, with significant clinical heterogeneity noted even within the same family. An autopsy of one patient revealed extensive brainstem calcifications, sparing the cerebral cortex, and marked by calcifications predominantly in capillaries and arterioles. The pathological study suggested morphological alterations characterized by shortened foot processes within astrocytes in regions with pronounced calcification and decreased immunoreactivity of AQP4. The morphology of astrocytes in regions without calcification remains preserved. Neuronal loss and gliosis were observed in the basal ganglia, thalamus, brainstem, cerebellum, and dentate nucleus. Notably, olivary hypertrophy, a previously undescribed feature in MYORG-PFBC, was discovered. Neuroimaging showed reduced blood flow in the cerebellum, highlighting the extent of cerebellar involvement. Among perivascular cells constituting the blood-brain barrier (BBB) and neurovascular unit, MYORG is most highly expressed in astrocytes. Astrocytes are integral components of the BBB, and their dysfunction can precipitate BBB disruption, potentially leading to brain calcification and subsequent neuronal loss. This study presents two novel homozygous variants in the MYORG gene and highlights the pivotal role of astrocytes in the development of brain calcifications, providing insights into the pathophysiological mechanisms underlying PFBC associated with MYORG variants.
Collapse
Affiliation(s)
- Takahiro Hobara
- Department of Neurology and Geriatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Yujiro Higuchi
- Department of Neurology and Geriatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan.
| | - Mari Yoshida
- Department of Neuropathology, Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan
| | - Masahito Suehara
- Department of Neurology, Fujimoto General Hospital, Miyazaki, Japan
| | - Masahiro Ando
- Department of Neurology and Geriatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Jun-Hui Yuan
- Department of Neurology and Geriatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Akiko Yoshimura
- Department of Neurology and Geriatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Fumikazu Kojima
- Department of Neurology and Geriatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Eiji Matsuura
- Department of Neurology and Geriatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Yuji Okamoto
- Department of Neurology and Geriatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
- Department of Physical Therapy, School of Health Sciences, Faculty of Medicine, Kagoshima University, Kagoshima, Japan
| | - Jun Mitsui
- Department of Precision Medicine Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shoji Tsuji
- Institute of Medical Genomics, International University of Health and Welfare, 4-3, Kozunomori, Chiba, Japan
- Department of Neurology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, Japan
| | - Hiroshi Takashima
- Department of Neurology and Geriatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| |
Collapse
|
21
|
Khojasteh M, Soleimani P, Ghasemi A, Taghizadeh P, Rohani M, Alavi A. JAM2 variants can be more common in primary familial brain calcification (PFBC) cases than those appear; may be due to a founder mutation. Neurol Sci 2024; 45:3829-3844. [PMID: 38441788 DOI: 10.1007/s10072-024-07419-6] [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: 11/13/2023] [Accepted: 02/20/2024] [Indexed: 07/18/2024]
Abstract
INTRODUCTION Mutations in JAM2 have been linked to ~ 2% of primary familial brain calcification (PFBC) cases. PFBC is a rare neurological disorder characterized by excessive calcium deposition in the brain. It causes movement disorders and psychiatric problems. Six other genes were identified as causing PFBC. However, the genetic basis of ~ 50% of PFBC cases remains unknown. This study presented the results of a comprehensive analysis of five unrelated Iranian PFBC families. METHODS Clinical and paraclinical features of all patients were recorded. Whole-exome sequencing (WES) was done on the DNAs of probands. Data was analyzed, and haplotypes were determined. RESULTS WES identified two homozygous variants in JAM2 across four families: a novel variant, c.426dup:p.Ser143Leufs*23, in one family and a known mutation, c.685C > T:p.Arg229*, in the remaining three families. Haplotype analysis using six intragenic single-nucleotide polymorphisms (SNPs) in JAM2 revealed an identical haplotype in probands who carried the same mutation, whereas two other probands presented diverse haplotypes. CONCLUSION Based on our results, p.Arg229* may be a founder mutation in the Iranian population. The variant has been detected in two out of seven other reported JAM2-related families who may originate from the Middle East and exhibit an identical haplotype. Even though this particular mutation may not be classified as a founder mutation, it does appear to be a hotspot, given that it has been observed in 45% of the 11 JAM2-associated families. Our study expanded the clinical features and mutation spectrum of JAM2 and revealed that mutations in JAM2 may be more common than previously reported.
Collapse
Affiliation(s)
- Mana Khojasteh
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Parsa Soleimani
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Aida Ghasemi
- Neuromuscular Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Peyman Taghizadeh
- School of Biology, University College of Science, University of Tehran, Tehran, Iran
| | - Mohammad Rohani
- Department of Neurology, Hazrat Rasool Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Afagh Alavi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran.
- Neuromuscular Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
22
|
Sun D, Wang Y, Wang J, Wang S, Zhu L, Xia K, Zhang Y, Wang X. Primary familial brain calcification presenting with parkinsonism and motor complications caused by a novel SLC20A2 variant: a case report. Front Neurol 2024; 15:1382534. [PMID: 39036637 PMCID: PMC11257840 DOI: 10.3389/fneur.2024.1382534] [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: 02/05/2024] [Accepted: 06/27/2024] [Indexed: 07/23/2024] Open
Abstract
Primary familial brain calcification (PFBC), also known as Fahr's disease, is a central nervous system calcium deposition disorder with symmetrical basal ganglia calcification. Most PFBC cases are caused by SLC20A2 gene variant. We report a Chinese female patient with PFBC and dopamine-responsive parkinsonism who had motor fluctuations and dyskinesia and recovered effectively after symptomatic medication adjustment. A novel heterozygous missense variant was found by whole-exome sequencing and proven harmful by family validation and genetic analysis. This example expands the phenotype of SLC20A2-associated PFBC patients and shows the clinical efficacy of dopaminergic replacement treatment.
Collapse
Affiliation(s)
- Dandan Sun
- Department of Graduate, Anhui University of Chinese Medicine, Hefei, China
- Institute of Neurology, Anhui University of Chinese Medicine, Hefei, China
| | - Yu Wang
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiawei Wang
- Institute of Neurology, Anhui University of Chinese Medicine, Hefei, China
| | - Shijing Wang
- Institute of Neurology, Anhui University of Chinese Medicine, Hefei, China
| | - Ling Zhu
- Institute of Neurology, Anhui University of Chinese Medicine, Hefei, China
| | - Kun Xia
- Institute of Neurology, Anhui University of Chinese Medicine, Hefei, China
| | - Yunyun Zhang
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xun Wang
- Institute of Neurology, Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
23
|
Rovelet-Lecrux A, Bonnevalle A, Quenez O, Delcroix W, Cassinari K, Richard AC, Boland A, Deleuze JF, Goizet C, Rucar A, Verny C, Nguyen K, Lecourtois M, Nicolas G. Upstream open reading frame-introducing variants in patients with primary familial brain calcification. Eur J Hum Genet 2024; 32:779-785. [PMID: 38433263 PMCID: PMC11219755 DOI: 10.1038/s41431-024-01580-4] [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: 12/13/2023] [Revised: 02/12/2024] [Accepted: 02/23/2024] [Indexed: 03/05/2024] Open
Abstract
More than 50% of patients with primary familial brain calcification (PFBC), a rare neurological disorder, remain genetically unexplained. While some causative genes are yet to be identified, variants in non-coding regions of known genes may represent a source of missed diagnoses. We hypothesized that 5'-Untranslated Region (UTR) variants introducing an AUG codon may initiate mRNA translation and result in a loss of function in some of the PFBC genes. After reannotation of exome sequencing data of 113 unrelated PFBC probands, we identified two upstream AUG-introducing variants in the 5'UTR of PDGFB. One, NM_002608.4:c.-373C>G, segregated with PFBC in the family. It was predicted to create an upstream open reading frame (ORF). The other one, NM_002608.4:c.-318C>T, was found in a simplex case. It was predicted to result in an ORF overlapping the natural ORF with a frameshift. In a GFP reporter assay, both variants were associated with a dramatic decrease in GFP levels, and, after restoring the reading frame with the GFP sequence, the c.-318C>T variant was associated with a strong initiation of translation as measured by western blotting. Overall, we found upstream AUG-introducing variants in the 5'UTR of PDGFB in 2/113 (1.7%) undiagnosed PFBC cases. Such variants thus represent a source of putative pathogenic variants.
Collapse
Affiliation(s)
- Anne Rovelet-Lecrux
- Univ Rouen Normandie, Inserm U1245 and CHU Rouen, Department of Genetics, CNRMAJ and Reference Center for Neurogenetics Disorders, F-76000, Rouen, France
| | - Antoine Bonnevalle
- Univ Rouen Normandie, Inserm U1245 and CHU Rouen, Department of Neurology, CNRMAJ and Reference Center for Neurogenetics Disorders, F-76000, Rouen, France
| | - Olivier Quenez
- Univ Rouen Normandie, Inserm U1245 and CHU Rouen, Department of Genetics, CNRMAJ and Reference Center for Neurogenetics Disorders, F-76000, Rouen, France
| | - Wandrille Delcroix
- Univ Rouen Normandie, Inserm U1245 and CHU Rouen, Department of Genetics, CNRMAJ and Reference Center for Neurogenetics Disorders, F-76000, Rouen, France
| | - Kévin Cassinari
- Univ Rouen Normandie, Inserm U1245 and CHU Rouen, Department of Genetics, CNRMAJ and Reference Center for Neurogenetics Disorders, F-76000, Rouen, France
| | - Anne-Claire Richard
- Univ Rouen Normandie, Inserm U1245 and CHU Rouen, Department of Genetics, CNRMAJ and Reference Center for Neurogenetics Disorders, F-76000, Rouen, France
| | - Anne Boland
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), 91057, Evry, France
| | - Jean-François Deleuze
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), 91057, Evry, France
| | - Cyril Goizet
- Department of Medical Genetics, National Reference Center for Rare Diseases 'Neurogenetic', Pellegrin Hospital, Bordeaux University Hospital, and University of Bordeaux, CNRS, INCIA, UMR 5287, NRGen Team, Bordeaux, France
| | - Alice Rucar
- Department of Neurology, University-Hospital of Angers, 49933, Angers, France
- Unité MitoVasc, UMR CNRS 6015, INSERM U1083, 49933, Angers, France
| | - Christophe Verny
- Department of Neurology, University-Hospital of Angers, 49933, Angers, France
- Unité MitoVasc, UMR CNRS 6015, INSERM U1083, 49933, Angers, France
| | - Karine Nguyen
- AP-HM, Hôpital Timone, Département de Génétique Médicale, Marseille, France
| | - Magalie Lecourtois
- Univ Rouen Normandie, Inserm U1245 and CHU Rouen, Department of Genetics, CNRMAJ and Reference Center for Neurogenetics Disorders, F-76000, Rouen, France
| | - Gaël Nicolas
- Univ Rouen Normandie, Inserm U1245 and CHU Rouen, Department of Genetics, CNRMAJ and Reference Center for Neurogenetics Disorders, F-76000, Rouen, France.
| |
Collapse
|
24
|
Bonato G, Cimino P, Pistonesi F, Salviati L, Bertolin C, Carecchio M. Non-Motor Symptoms in Primary Familial Brain Calcification. J Clin Med 2024; 13:3873. [PMID: 38999439 PMCID: PMC11242504 DOI: 10.3390/jcm13133873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 06/17/2024] [Accepted: 06/27/2024] [Indexed: 07/14/2024] Open
Abstract
Background/Objectives: Primary Familial Brain Calcification is a rare neurodegenerative disorder of adulthood characterized by calcium deposition in the basal ganglia and other brain areas; the main clinical manifestations include movement disorders, mainly parkinsonism. Non-motor symptoms are not well defined in PFBC. This work aims at defining the burden of non-motor symptoms in PFBC. Methods: A clinical, genetic and neuropsychological evaluation of a cohort of PFBC patients, COMPASS-31 scale administration. Results: A total of 50 PFBC patients were recruited; in 25, the genetic test was negative; 10 carried mutations in SLC20A2 gene, 8 in MYORG, 3 in PDGFB, 1 in PDGFRB, 2 in JAM2 (single mutations), and one test is still ongoing. The main motor manifestation was parkinsonism. Headache was reported in 26% of subjects (especially in PDGFB mutation carriers), anxiety or depression in 62%, psychosis or hallucinations in 10-12%, sleep disturbances in 34%; 14% of patients reported hyposmia, 32% constipation, and 34% urinary disturbances. A neuropsychological assessment revealed cognitive involvement in 56% (sparing memory functions, to some extent). The COMPASS-31 mean score was 20.6, with higher sub-scores in orthostatic intolerance and gastrointestinal problems. MYORG patients and subjects with cognitive decline tended to have higher scores and bladder involvement compared to other groups. Conclusions: The presence of non-motor symptoms is frequent in PFBC and should be systematically assessed to better meet patients' needs.
Collapse
Affiliation(s)
- Giulia Bonato
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases (ERN-RND), Department of Neuroscience, University of Padova, 35128 Padova, Italy
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, 35128 Padova, Italy
| | - Paola Cimino
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases (ERN-RND), Department of Neuroscience, University of Padova, 35128 Padova, Italy
| | - Francesca Pistonesi
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases (ERN-RND), Department of Neuroscience, University of Padova, 35128 Padova, Italy
| | - Leonardo Salviati
- Medical Genetics Unit, Department of Women and Children's Health, University of Padova, 35128 Padova, Italy
| | - Cinzia Bertolin
- Medical Genetics Unit, Department of Women and Children's Health, University of Padova, 35128 Padova, Italy
| | - Miryam Carecchio
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases (ERN-RND), Department of Neuroscience, University of Padova, 35128 Padova, Italy
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, 35128 Padova, Italy
| |
Collapse
|
25
|
Cao C, Luo J, Wang X. Case report: Primary familial brain calcification associated with a rare PDGFRB variant, coexisting with nontraumatic osteonecrosis of the femoral head. Front Neurosci 2024; 18:1381840. [PMID: 38859923 PMCID: PMC11163128 DOI: 10.3389/fnins.2024.1381840] [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: 02/04/2024] [Accepted: 05/15/2024] [Indexed: 06/12/2024] Open
Abstract
Primary familial brain calcification (PFBC) is a rare genetic neurodegenerative disorder characterized by bilateral calcifications in the brain. PFBC may manifest with a broad spectrum of motor, cognitive, and neuropsychiatric symptoms. Several causal genes have been identified in PFBC, which are inherited as both autosomal dominant and autosomal recessive traits. Herein, we present the case of a Chinese family diagnosed with PFBC. The family members carry a rare heterozygous variant (p. R334Q) in exon 7 of platelet-derived growth factor receptor β (PDGFRB) gene. The platelet-derived growth factor-B/PDGF receptor β (PDGF-B/PDGFRβ) signaling pathway plays a crucial role in pericyte development in various organs and tissues. Notably, this variant uniquely coexists with nontraumatic osteonecrosis of the femoral head. Additionally, we reviewed previous studies on PFBC-causing variants in PDGFRB.
Collapse
Affiliation(s)
- Conghui Cao
- Department of Endocrinology and Metabolism, Institute of Endocrinology, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang, China
| | - Jing Luo
- Department of Endocrinology and Metabolism, Institute of Endocrinology, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang, China
- Department of Endocrinology and Metabolism, Tieling Central Hospital, Tieling, China
| | - Xiaoli Wang
- Department of Endocrinology and Metabolism, Institute of Endocrinology, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
26
|
Sennfält S, Gustavsson P, Malmgren H, Gilland E, Almqvist H, Oscarson M, Engvall M, Björkhem I, Nilsson D, Lagerstedt-Robinson K, Svenningsson P, Paucar M. Novel findings in a Swedish primary familial brain calcification cohort. J Neurol Sci 2024; 460:123020. [PMID: 38642488 DOI: 10.1016/j.jns.2024.123020] [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: 08/30/2023] [Revised: 04/10/2024] [Accepted: 04/16/2024] [Indexed: 04/22/2024]
Abstract
INTRODUCTION Brain calcifications are frequent findings on imaging. In a small proportion of cases, these calcifications are associated with pathogenic gene variants, hence termed primary familial brain calcification (PFBC). The clinical penetrance is incomplete and phenotypic variability is substantial. This paper aims to characterize a Swedish PFBC cohort including 25 patients: 20 from seven families and five sporadic cases. METHODS Longitudinal clinical assessment and CT imaging were conducted, abnormalities were assessed using the total calcification score (TCS). Genetic analyses, including a panel of six known PFBC genes, were performed in all index and sporadic cases. Additionally, three patients carrying a novel pathogenic copy number variant in SLC20A2 had their cerebrospinal fluid phosphate (CSF-Pi) levels measured. RESULTS Among the 25 patients, the majority (76%) displayed varying symptoms during the initial assessment including motor (60%), psychiatric (40%), and/or cognitive abnormalities (24%). Clinical progression was observed in most patients (78.6%), but there was no significant difference in calcification between the first and second scans, with mean scores of 27.3 and 32.8, respectively. In three families and two sporadic cases, pathogenic genetic variants were identified, including a novel finding, in the SLC20A2 gene. In the three tested patients, the CSF-Pi levels were normal. CONCLUSIONS This report demonstrates the variable expressivity seen in PFBC and includes a novel pathogenic variant in the SLC20A2 gene. In four families and three sporadic cases, no pathogenic variants were found, suggesting that new PFBC genes remain to be discovered.
Collapse
Affiliation(s)
- Stefan Sennfält
- Department of Neurology, Karolinska University Hospital, Hälsovägen 13 R52, 141 86 Stockholm, Sweden; Department of Clinical Neuroscience, Karolinska Institutet, Nobels väg 6, 171 77 Stockholm, Sweden.
| | - Peter Gustavsson
- Department of Clinical Genetics, Karolinska University Hospital, Karolinska Vägen, 171 76500 Solna, Sweden; Department of Molecular Medicine and Surgery, Karolinska Institutet, Nobels väg 6, 171 77 Stockholm, Sweden.
| | - Helena Malmgren
- Department of Clinical Genetics, Karolinska University Hospital, Karolinska Vägen, 171 76500 Solna, Sweden; Department of Molecular Medicine and Surgery, Karolinska Institutet, Nobels väg 6, 171 77 Stockholm, Sweden.
| | - Eric Gilland
- Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Blå stråket 7, 413 46 Göteborg, Sweden.
| | - Håkan Almqvist
- Department of Clinical Neuroscience, Karolinska Institutet, Nobels väg 6, 171 77 Stockholm, Sweden; Department of Radiology, Capio S:t Goran Hospital, Sankt Göransplan 1, 112 19 Stockholm, Sweden.
| | - Mikael Oscarson
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Anna Steckséns g 47, 171 76 Solna, Sweden.
| | - Martin Engvall
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Anna Steckséns g 47, 171 76 Solna, Sweden.
| | - Ingemar Björkhem
- Science for Life Laboratory, Stockholm, Tomtebodavägen 23, 171 65 Solna, Sweden.
| | - Daniel Nilsson
- Department of Clinical Genetics, Karolinska University Hospital, Karolinska Vägen, 171 76500 Solna, Sweden; Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Alfred Nobels Allé 8, 141 52 Huddinge, Sweden.
| | - Kristina Lagerstedt-Robinson
- Department of Clinical Genetics, Karolinska University Hospital, Karolinska Vägen, 171 76500 Solna, Sweden; Department of Molecular Medicine and Surgery, Karolinska Institutet, Nobels väg 6, 171 77 Stockholm, Sweden.
| | - Per Svenningsson
- Department of Neurology, Karolinska University Hospital, Hälsovägen 13 R52, 141 86 Stockholm, Sweden; Department of Clinical Neuroscience, Karolinska Institutet, Nobels väg 6, 171 77 Stockholm, Sweden.
| | - Martin Paucar
- Department of Neurology, Karolinska University Hospital, Hälsovägen 13 R52, 141 86 Stockholm, Sweden; Department of Clinical Neuroscience, Karolinska Institutet, Nobels väg 6, 171 77 Stockholm, Sweden.
| |
Collapse
|
27
|
Burns D, Berlinguer-Palmini R, Werner A. XPR1: a regulator of cellular phosphate homeostasis rather than a Pi exporter. Pflugers Arch 2024; 476:861-869. [PMID: 38507112 PMCID: PMC11033234 DOI: 10.1007/s00424-024-02941-0] [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: 12/19/2023] [Revised: 02/19/2024] [Accepted: 03/04/2024] [Indexed: 03/22/2024]
Abstract
Phosphate (Pi) is an essential nutrient, and its plasma levels are under tight hormonal control. Uphill transport of Pi into cells is mediated by the two Na-dependent Pi transporter families SLC34 and SLC20. The molecular identity of a potential Pi export pathway is controversial, though XPR1 has recently been suggested by Giovannini and coworkers to mediate Pi export. We expressed XPR1 in Xenopus oocytes to determine its functional characteristics. Xenopus isoforms of proteins were used to avoid species incompatibility. Protein tagging confirmed the localization of XPR1 at the plasma membrane. Efflux experiments, however, failed to detect translocation of Pi attributable to XPR1. We tested various counter ions and export medium compositions (pH, plasma) as well as potential protein co-factors that could stimulate the activity of XPR1, though without success. Expression of truncated XPR1 constructs and individual domains of XPR1 (SPX, transmembrane core, C-terminus) demonstrated downregulation of the uptake of Pi mediated by the C-terminal domain of XPR1. Tethering the C-terminus to the transmembrane core changed the kinetics of the inhibition and the presence of the SPX domain blunted the inhibitory effect. Our observations suggest a regulatory role of XPR1 in cellular Pi handling rather than a function as Pi exporter. Accordingly, XPR1 senses intracellular Pi levels via its SPX domain and downregulates cellular Pi uptake via the C-terminal domain. The molecular identity of a potential Pi export protein remains therefore elusive.
Collapse
Affiliation(s)
- David Burns
- Biosciences Institute, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | | | - Andreas Werner
- Biosciences Institute, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK.
| |
Collapse
|
28
|
Chelban V, Aksnes H, Maroofian R, LaMonica LC, Seabra L, Siggervåg A, Devic P, Shamseldin HE, Vandrovcova J, Murphy D, Richard AC, Quenez O, Bonnevalle A, Zanetti MN, Kaiyrzhanov R, Salpietro V, Efthymiou S, Schottlaender LV, Morsy H, Scardamaglia A, Tariq A, Pagnamenta AT, Pennavaria A, Krogstad LS, Bekkelund ÅK, Caiella A, Glomnes N, Brønstad KM, Tury S, Moreno De Luca A, Boland-Auge A, Olaso R, Deleuze JF, Anheim M, Cretin B, Vona B, Alajlan F, Abdulwahab F, Battini JL, İpek R, Bauer P, Zifarelli G, Gungor S, Kurul SH, Lochmuller H, Da'as SI, Fakhro KA, Gómez-Pascual A, Botía JA, Wood NW, Horvath R, Ernst AM, Rothman JE, McEntagart M, Crow YJ, Alkuraya FS, Nicolas G, Arnesen T, Houlden H. Biallelic NAA60 variants with impaired n-terminal acetylation capacity cause autosomal recessive primary familial brain calcifications. Nat Commun 2024; 15:2269. [PMID: 38480682 PMCID: PMC10937998 DOI: 10.1038/s41467-024-46354-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 02/23/2024] [Indexed: 03/17/2024] Open
Abstract
Primary familial brain calcification (PFBC) is characterized by calcium deposition in the brain, causing progressive movement disorders, psychiatric symptoms, and cognitive decline. PFBC is a heterogeneous disorder currently linked to variants in six different genes, but most patients remain genetically undiagnosed. Here, we identify biallelic NAA60 variants in ten individuals from seven families with autosomal recessive PFBC. The NAA60 variants lead to loss-of-function with lack of protein N-terminal (Nt)-acetylation activity. We show that the phosphate importer SLC20A2 is a substrate of NAA60 in vitro. In cells, loss of NAA60 caused reduced surface levels of SLC20A2 and a reduction in extracellular phosphate uptake. This study establishes NAA60 as a causal gene for PFBC, provides a possible biochemical explanation of its disease-causing mechanisms and underscores NAA60-mediated Nt-acetylation of transmembrane proteins as a fundamental process for healthy neurobiological functioning.
Collapse
Affiliation(s)
- Viorica Chelban
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK.
- Neurobiology and Medical Genetics Laboratory, "Nicolae Testemitanu" State University of Medicine and Pharmacy, 165, Stefan cel Mare si Sfant Boulevard, MD, 2004, Chisinau, Republic of Moldova.
| | - Henriette Aksnes
- Department of Biomedicine, University of Bergen, Bergen, Norway.
| | - Reza Maroofian
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Lauren C LaMonica
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, USA
| | - Luis Seabra
- Université Paris Cité, Imagine Institute, Laboratory of Neurogenetics and Neuroinflammation, INSERM UMR 1163, Paris, France
| | | | - Perrine Devic
- Hospices Civils de Lyon, Groupement Hospitalier Sud, Service d'Explorations Fonctionnelles Neurologiques, Lyon, France
| | - Hanan E Shamseldin
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Jana Vandrovcova
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - David Murphy
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Anne-Claire Richard
- Univ Rouen Normandie, Inserm U1245, CHU Rouen, Department of Genetics and CNRMAJ, F-76000, Rouen, France
| | - Olivier Quenez
- Univ Rouen Normandie, Inserm U1245, CHU Rouen, Department of Genetics and CNRMAJ, F-76000, Rouen, France
| | - Antoine Bonnevalle
- Univ Rouen Normandie, Inserm U1245, CHU Rouen, Department of Genetics and CNRMAJ, F-76000, Rouen, France
| | - M Natalia Zanetti
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Rauan Kaiyrzhanov
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
- South Kazakhstan Medical Academy Shymkent, Shymkent, 160019, Kazakhstan
| | - Vincenzo Salpietro
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Stephanie Efthymiou
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Lucia V Schottlaender
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
- Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Av. Juan Domingo Perón 1500, B1629AHJ, Pilar, Argentina
- Instituto de medicina genómica (IMeG), Hospital Universitario Austral, Universidad Austral, Av. Juan Domingo Perón 1500, B1629AHJ, Pilar, Argentina
| | - Heba Morsy
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
- Department of Human Genetics, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Annarita Scardamaglia
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Ambreen Tariq
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Alistair T Pagnamenta
- Oxford NIHR Biomedical Research Centre, Wellcome Centre for Human Genetics, Oxford, United Kingdom
| | - Ajia Pennavaria
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Liv S Krogstad
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Åse K Bekkelund
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Alessia Caiella
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Nina Glomnes
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Department of Clinical Science, University of Bergen, 5020, Bergen, Norway
| | | | - Sandrine Tury
- Institut de Recherche en Infectiologie de Montpellier, Université de Montpellier, CNRS, Montpellier, France
| | - Andrés Moreno De Luca
- Department of Radiology, Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA, USA
- Department of Radiology, Neuroradiology Section, Kingston Health Sciences Centre, Queen's University Faculty of Health Sciences, Kingston, Ontario, Canada
| | - Anne Boland-Auge
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), 91057, Evry, France
| | - Robert Olaso
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), 91057, Evry, France
| | - Jean-François Deleuze
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), 91057, Evry, France
| | - Mathieu Anheim
- Neurology Department, Strasbourg University Hospital, Strasbourg, France
- Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg, France
- INSERM-U964; CNRS-UMR7104, University of Strasbourg, Illkirch-Graffenstaden, France
| | - Benjamin Cretin
- Neurology Department, Strasbourg University Hospital, Strasbourg, France
- Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg, France
- INSERM-U964; CNRS-UMR7104, University of Strasbourg, Illkirch-Graffenstaden, France
| | - Barbara Vona
- Institute of Human Genetics, University Medical Center Göttingen, 37073, Göttingen, Germany
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075, Göttingen, Germany
| | - Fahad Alajlan
- Department of Neuroscience Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Firdous Abdulwahab
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Jean-Luc Battini
- Institut de Recherche en Infectiologie de Montpellier, Université de Montpellier, CNRS, Montpellier, France
| | - Rojan İpek
- Paediatric Neurology, Faculty of Medicine, Dicle University, Diyarbakır, Turkey
| | - Peter Bauer
- Centogene GmbH, Am Strande 7, 18055, Rostock, Germany
| | | | - Serdal Gungor
- Inonu University, Faculty of Medicine, Turgut Ozal Research Center, Department of Pediatrics, Division of Pediatric Neurology, Malatya, Turkey
| | - Semra Hiz Kurul
- Dokuz Eylul University, School of Medicine, Department of Paediatric Neurology, Izmir, Turkey
| | - Hanns Lochmuller
- Children's Hospital of Eastern Ontario Research Institute and Division of Neurology, Department of Medicine, The Ottawa Hospital, Ottawa, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada
- Department of Neuropediatrics and Muscle Disorders, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Sahar I Da'as
- Department of Human Genetics, Sidra Medicine, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Khalid A Fakhro
- Department of Human Genetics, Sidra Medicine, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
- Weill Cornell Medical College, Doha, Qatar
| | - Alicia Gómez-Pascual
- Department of Information and Communications Engineering, University of Murcia, Campus Espinardo, 30100, Murcia, Spain
| | - Juan A Botía
- Department of Information and Communications Engineering, University of Murcia, Campus Espinardo, 30100, Murcia, Spain
| | - Nicholas W Wood
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
- Neurogenetics Laboratory, The National Hospital for Neurology and Neurosurgery, London, WC1N 3BG, UK
| | - Rita Horvath
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Andreas M Ernst
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, USA
- School of Biological Sciences, Department of Cell and Developmental Biology, University of California San Diego, La Jolla, CA, USA
| | - James E Rothman
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, USA
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Meriel McEntagart
- Medical Genetics Department, St George's University Hospitals, London, SWI7 0RE, UK
| | - Yanick J Crow
- Université Paris Cité, Imagine Institute, Laboratory of Neurogenetics and Neuroinflammation, INSERM UMR 1163, Paris, France
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Fowzan S Alkuraya
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
- Department of Anatomy and Cell Biology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Gaël Nicolas
- Univ Rouen Normandie, Inserm U1245, CHU Rouen, Department of Genetics and CNRMAJ, F-76000, Rouen, France
| | - Thomas Arnesen
- Department of Biomedicine, University of Bergen, Bergen, Norway.
- Department of Surgery, Haukeland University Hospital, Bergen, Norway.
| | - Henry Houlden
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK.
- Neurogenetics Laboratory, The National Hospital for Neurology and Neurosurgery, London, WC1N 3BG, UK.
| |
Collapse
|
29
|
Yoon S, Chung SJ, Kim YJ. Primary Familial Brain Calcification With XPR1 Mutation Presenting With Cognitive Dysfunction. J Clin Neurol 2024; 20:229-231. [PMID: 38433488 PMCID: PMC10921053 DOI: 10.3988/jcn.2023.0284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 09/19/2023] [Accepted: 11/06/2023] [Indexed: 03/05/2024] Open
Affiliation(s)
- Sojung Yoon
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
- Department of Neurology, Yongin Severance Hospital, Yonsei University Health System, Yongin, Korea
| | - Seok Jong Chung
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
- Department of Neurology, Yongin Severance Hospital, Yonsei University Health System, Yongin, Korea
- YONSEI BEYOND LAB, Yongin, Korea
| | - Yun Joong Kim
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
- Department of Neurology, Yongin Severance Hospital, Yonsei University Health System, Yongin, Korea
- YONSEI BEYOND LAB, Yongin, Korea.
| |
Collapse
|
30
|
Mathijssen G, van Valen E, de Jong PA, Golüke NMS, van Maren EA, Snijders BMG, Brilstra EH, Ruigrok YM, Bakker S, Goto RW, Emmelot-Vonk MH, Koek HL. The Association between Intracranial Calcifications and Symptoms in Patients with Primary Familial Brain Calcification. J Clin Med 2024; 13:828. [PMID: 38337525 PMCID: PMC10856178 DOI: 10.3390/jcm13030828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/23/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
(1) Background: Primary Familial Brain Calcification (PFBC) is a neurodegenerative disease characterized by bilateral calcifications of the basal ganglia and other intracranial areas. Many patients experience symptoms of motor dysfunction and cognitive disorders. The aim of this study was to investigate the association between the amount and location of intracranial calcifications with these symptoms. (2) Methods: Patients with suspected PFBC referred to our outpatient clinic underwent a clinical work-up. Intracranial calcifications were visualized on Computed Tomography (CT), and a Total Calcification Score (TCS) was constructed. Logistic and linear regression models were performed. (3) Results: Fifty patients with PFBC were included in this study (median age 64.0 years, 50% women). Of the forty-one symptomatic patients (82.0%), 78.8% showed motor dysfunction, and 70.7% showed cognitive disorders. In multivariate analysis, the TCS was associated with bradykinesia/hypokinesia (OR 1.07, 95%-CI 1.02-1.12, p < 0.01), gait ataxia (OR 1.06, 95%-CI 1.00-1.12, p = 0.04), increased fall risk (OR 1.04, 95%-CI 1.00-1.08, p = 0.03), and attention/processing speed disorders (OR 1.06, 95%-CI 1.01-1.12, p = 0.02). Calcifications of the lentiform nucleus and subcortical white matter were associated with motor and cognitive disorders. (4) Conclusions: cognitive and motor symptoms are common among patients with PFBC, and there is an association between intracranial calcifications and these symptoms.
Collapse
Affiliation(s)
- Gini Mathijssen
- Department of Geriatrics, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Evelien van Valen
- Department of Geriatrics, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Pim A de Jong
- Department of Radiology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Nienke M S Golüke
- Department of Geriatrics, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
- Department of Geriatrics, Hospital Gelderse Vallei, Willy Brandtlaan 10, 6716 RP Ede, The Netherlands
| | - Emiel A van Maren
- Department of Radiology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Birgitta M G Snijders
- Department of Geriatrics, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Eva H Brilstra
- Department of Genetics, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Ynte M Ruigrok
- Department of Neurology and Neurosurgery, University Medical Center Utrecht, Heidelberglaan 100, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Susan Bakker
- Department of Rehabilitation, Physical Therapy Science & Sports, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Renzo W Goto
- Department of Geriatrics, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Marielle H Emmelot-Vonk
- Department of Geriatrics, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Huiberdina L Koek
- Department of Geriatrics, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
31
|
Ramos-Brossier M, Romeo-Guitart D, Lanté F, Boitez V, Mailliet F, Saha S, Rivagorda M, Siopi E, Nemazanyy I, Leroy C, Moriceau S, Beck-Cormier S, Codogno P, Buisson A, Beck L, Friedlander G, Oury F. Slc20a1 and Slc20a2 regulate neuronal plasticity and cognition independently of their phosphate transport ability. Cell Death Dis 2024; 15:20. [PMID: 38195526 PMCID: PMC10776841 DOI: 10.1038/s41419-023-06292-z] [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: 04/03/2023] [Revised: 11/01/2023] [Accepted: 11/13/2023] [Indexed: 01/11/2024]
Abstract
In recent years, primary familial brain calcification (PFBC), a rare neurological disease characterized by a wide spectrum of cognitive disorders, has been associated to mutations in the sodium (Na)-Phosphate (Pi) co-transporter SLC20A2. However, the functional roles of the Na-Pi co-transporters in the brain remain still largely elusive. Here we show that Slc20a1 (PiT-1) and Slc20a2 (PiT-2) are the most abundant Na-Pi co-transporters expressed in the brain and are involved in the control of hippocampal-dependent learning and memory. We reveal that Slc20a1 and Slc20a2 are differentially distributed in the hippocampus and associated with independent gene clusters, suggesting that they influence cognition by different mechanisms. Accordingly, using a combination of molecular, electrophysiological and behavioral analyses, we show that while PiT-2 favors hippocampal neuronal branching and survival, PiT-1 promotes synaptic plasticity. The latter relies on a likely Otoferlin-dependent regulation of synaptic vesicle trafficking, which impacts the GABAergic system. These results provide the first demonstration that Na-Pi co-transporters play key albeit distinct roles in the hippocampus pertaining to the control of neuronal plasticity and cognition. These findings could provide the foundation for the development of novel effective therapies for PFBC and cognitive disorders.
Collapse
Affiliation(s)
- Mariana Ramos-Brossier
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Team 8, F-75015, Paris, France.
| | - David Romeo-Guitart
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Team 8, F-75015, Paris, France
| | - Fabien Lanté
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France
| | - Valérie Boitez
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Team 8, F-75015, Paris, France
| | - François Mailliet
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Team 8, F-75015, Paris, France
| | - Soham Saha
- Institut Pasteur, Perception & Memory Unit, F-75015, Paris, France
- MedInsights, 6 rue de l'église, F-02810, Veuilly la Poterie, France
| | - Manon Rivagorda
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Team 8, F-75015, Paris, France
| | - Eleni Siopi
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Team 8, F-75015, Paris, France
| | - Ivan Nemazanyy
- Platform for Metabolic Analyses, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UAR, 3633, Paris, France
| | - Christine Leroy
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Team 6, F-75015, Paris, France
| | - Stéphanie Moriceau
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Team 8, F-75015, Paris, France
- Platform for Neurobehavioural and metabolism, Structure Fédérative de Recherche Necker, INSERM, US24/CNRS UAR, 3633, Paris, France
- Institute of Genetic Diseases, Imagine, 75015, Paris, France
| | - Sarah Beck-Cormier
- Nantes Université, CNRS, Inserm, l'Institut du Thorax, F-44000, Nantes, France
| | - Patrice Codogno
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Team 6, F-75015, Paris, France
| | - Alain Buisson
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France
| | - Laurent Beck
- Nantes Université, CNRS, Inserm, l'Institut du Thorax, F-44000, Nantes, France.
| | - Gérard Friedlander
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Team 6, F-75015, Paris, France.
| | - Franck Oury
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Team 8, F-75015, Paris, France.
| |
Collapse
|
32
|
Timmi A, Morin A, Guillin O, Nicolas G. One Train May Hide Another: Two Cases of Co-Occurring Primary Familial Brain Calcification and Alzheimer's Disease. J Mol Neurosci 2024; 74:2. [PMID: 38180527 DOI: 10.1007/s12031-023-02184-1] [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: 10/04/2023] [Accepted: 12/02/2023] [Indexed: 01/06/2024]
Abstract
Primary familial brain calcification (PFBC) is a rare disorder that can manifest with a wide spectrum of motor, cognitive, and psychiatric symptoms or even remain asymptomatic. Alzheimer disease (AD) is a common condition that typically starts as a progressive amnestic disorder and progresses to major cognitive impairment. Accurately attributing an etiology to cognitive impairment can sometimes be challenging, especially when multiple pathologies with potentially overlapping symptomatology contribute to the clinical phenotype. Here, we present the case of two patients with autosomal dominant PFBC and non-monogenic AD. Cerebrospinal fluid (CSF) biomarker analysis combined with genetic testing permitted the dual diagnosis. We emphasize the importance of thoroughly characterizing the patient's phenotype at onset and during the follow-up. Particular attention is placed on psychiatric symptoms given that both patients had a history of mood disorder, a frequent condition in the general population and in neurological diseases. We also discuss and challenge the paradigm of seeking a single diagnosis explaining all symptoms, remembering the possibility of a rare disease co-occurring with a common one.
Collapse
Affiliation(s)
- Andrea Timmi
- Department of Psychiatry, Rouvray Hospital, Univ Rouen Normandie, F-76000, Rouen, France
| | - Alexandre Morin
- Department of Psychiatry, Rouvray Hospital, Univ Rouen Normandie, F-76000, Rouen, France
- Univ Rouen Normandie, Normandie Univ and CHU Rouen, Department of Neurology, F-76000, Rouen, France
| | - Olivier Guillin
- Department of Psychiatry, Rouvray Hospital, Univ Rouen Normandie, F-76000, Rouen, France
- Univ Rouen Normandie, Normandie Univ, Inserm U1245 and CHU Rouen, Department of Psychiatry, F-76000, Rouen, France
| | - Gaël Nicolas
- Univ Rouen Normandie, Normandie Univ, Inserm U1245 and CHU Rouen, Department of Genetics and CNR-MAJ, F-76000, Rouen, France.
| |
Collapse
|
33
|
Chelban V, Houlden H. White matter disorders with cerebral calcification in adulthood. HANDBOOK OF CLINICAL NEUROLOGY 2024; 204:111-131. [PMID: 39322374 DOI: 10.1016/b978-0-323-99209-1.00024-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
This chapter provides a comprehensive overview of adult-onset leukoencephalopathies with cerebral calcification (CC), emphasizing the importance of age at presentation, systemic clinical features, and neuroimaging patterns for accurate diagnosis. CC is a multifaceted phenomenon associated with various neurologic, developmental, metabolic, and genetic conditions, as well as normal aging. Here, we explore the distinction between primary familial brain calcification (PFBC) and secondary forms, including metabolic and mitochondrial causes. We discuss genetic causes, e.g., SLC20A2, XPR1, PDGFB, PDGFRB, MYORG, NAA60 and JAM2, in the context of autosomal dominant and recessive PFBC and other inherited conditions. The chapter delineates the diagnostic approach involving family history, clinical assessments, and detailed investigations of calcium-phosphate metabolism. Neuroimaging modalities, including computed tomography and magnetic resonance imaging, are crucial for assessing calcification patterns and localizations. Genetic testing, especially next-generation sequencing, plays a pivotal role in providing a final molecular diagnosis. The management of patients with CC encompasses symptomatic treatment and cause-specific approaches, requiring a multidisciplinary care approach. In conclusion, this chapter highlights the complexity of leukoencephalopathies with CC, emphasizing the need for integrated and evolving management to optimize patient care.
Collapse
Affiliation(s)
- Viorica Chelban
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom; Neurobiology and Medical Genetics Laboratory, "Nicolae Testemitanu" State University of Medicine and Pharmacy, Chisinau, Republic of Moldova
| | - Henry Houlden
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom; Neurogenetics Laboratory, National Hospital for Neurology and Neurosurgery, London, United Kingdom.
| |
Collapse
|
34
|
Zhao T, Xu S, Liu S, Xu J, Zhang X, Zhan Y. Fahr's disease linked to a novel mutation in MYORG variants manifesting as paroxysmal limb stiffness and dysarthria: Case report and literature review. Mol Genet Genomic Med 2023; 11:e2276. [PMID: 37680026 PMCID: PMC10724521 DOI: 10.1002/mgg3.2276] [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: 05/28/2023] [Revised: 07/24/2023] [Accepted: 08/17/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND Primary familial brain calcification (PFBC) is a rare hereditary neurodegenerative disorder associated with the MYORG gene; however, the clinical and radiological characteristics of MYORG-PFBC remain unclear. METHODS We present relevant medical data obtained from a patient affected by PFBC with a novel MYORG variant and conducted a mutational analysis of MYORG in her family members. We reviewed all reported PFBC cases with biallelic MYORG mutations until April 1, 2023, and summarized the associated clinical and radiological features and mutation sites. RESULTS The patient (22-year-old woman) exhibited paroxysmal limb stiffness and dysarthria for 3 years. Computed tomography revealed calcifications in the paraventricular white matter, basal ganglia, thalamus, and cerebellum. Whole-exome sequencing revealed a novel homozygous frameshift variant (c.743delG: p.G248Afs*32) in exon 2 of the MYORG gene (NM_020702.5). To date, 62 families and 64 mutation sites have been reported. Among the reported biallelic MYORG mutations, 57% were homozygous and 43% were compound heterozygous. Individuals with biallelic MYORG mutations experience more severe brain calcification with approximately 100% clinical penetrance. Ten single heterozygous mutation sites are associated with significant brain calcifications. CONCLUSION All patients with primary brain calcification, particularly younger patients without a family history of the disease, should be screened for MYORG mutations.
Collapse
Affiliation(s)
- Tianxue Zhao
- Department of Endocrinology, Affiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhouChina
| | - Shaokun Xu
- Department of GeriatricsZhejiang Provincial People's HospitalHangzhouChina
| | - Siyue Liu
- Department of Endocrinology, Affiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhouChina
| | - Juan Xu
- Department of Endocrinology, Affiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhouChina
| | - Xianfeng Zhang
- Department of Endocrinology, Affiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhouChina
| | - Yuhong Zhan
- Department of Endocrinology, Affiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhouChina
| |
Collapse
|
35
|
Maheshwari U, Mateos JM, Weber‐Stadlbauer U, Ni R, Tamatey V, Sridhar S, Restrepo A, de Jong PA, Huang S, Schaffenrath J, Stifter SA, Szeri F, Greter M, Koek HL, Keller A. Inorganic phosphate exporter heterozygosity in mice leads to brain vascular calcification, microangiopathy, and microgliosis. Brain Pathol 2023; 33:e13189. [PMID: 37505935 PMCID: PMC10580014 DOI: 10.1111/bpa.13189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
Calcification of the cerebral microvessels in the basal ganglia in the absence of systemic calcium and phosphate imbalance is a hallmark of primary familial brain calcification (PFBC), a rare neurodegenerative disorder. Mutation in genes encoding for sodium-dependent phosphate transporter 2 (SLC20A2), xenotropic and polytropic retrovirus receptor 1 (XPR1), platelet-derived growth factor B (PDGFB), platelet-derived growth factor receptor beta (PDGFRB), myogenesis regulating glycosidase (MYORG), and junctional adhesion molecule 2 (JAM2) are known to cause PFBC. Loss-of-function mutations in XPR1, the only known inorganic phosphate exporter in metazoans, causing dominantly inherited PFBC was first reported in 2015 but until now no studies in the brain have addressed whether loss of one functional allele leads to pathological alterations in mice, a commonly used organism to model human diseases. Here we show that mice heterozygous for Xpr1 (Xpr1WT/lacZ ) present with reduced inorganic phosphate levels in the cerebrospinal fluid and age- and sex-dependent growth of vascular calcifications in the thalamus. Vascular calcifications are surrounded by vascular basement membrane and are located at arterioles in the smooth muscle layer. Similar to previously characterized PFBC mouse models, vascular calcifications in Xpr1WT/lacZ mice contain bone matrix proteins and are surrounded by reactive astrocytes and microglia. However, microglial activation is not confined to calcified vessels but shows a widespread presence. In addition to vascular calcifications, we observed vessel tortuosity and transmission electron microscopy analysis revealed microangiopathy-endothelial swelling, phenotypic alterations in vascular smooth muscle cells, and thickening of the basement membrane.
Collapse
Affiliation(s)
- Upasana Maheshwari
- Department of Neurosurgery, Clinical Neuroscience CenterUniversity Hospital Zurich, University of ZurichZurichSwitzerland
| | - José M. Mateos
- Center for Microscopy and Image analysisUniversity of ZurichZurichSwitzerland
| | - Ulrike Weber‐Stadlbauer
- Institute of Veterinary Pharmacology and ToxicologyUniversity of Zurich‐Vetsuisse, University of ZurichZurichSwitzerland
- Neuroscience Center ZurichUniversity of Zurich and ETH ZurichZurichSwitzerland
| | - Ruiqing Ni
- Neuroscience Center ZurichUniversity of Zurich and ETH ZurichZurichSwitzerland
- Institute for Biomedical EngineeringUniversity of Zurich and ETH ZurichZurichSwitzerland
| | - Virgil Tamatey
- Research Centre for Natural SciencesInstitute of EnzymologyBudapestHungary
- Doctoral School of BiologyELTE Eotvos Lorand UniversityBudapestHungary
| | - Sucheta Sridhar
- Department of Neurosurgery, Clinical Neuroscience CenterUniversity Hospital Zurich, University of ZurichZurichSwitzerland
- Neuroscience Center ZurichUniversity of Zurich and ETH ZurichZurichSwitzerland
| | - Alejandro Restrepo
- Department of Neurosurgery, Clinical Neuroscience CenterUniversity Hospital Zurich, University of ZurichZurichSwitzerland
| | - Pim A. de Jong
- Department of RadiologyUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Sheng‐Fu Huang
- Department of Neurosurgery, Clinical Neuroscience CenterUniversity Hospital Zurich, University of ZurichZurichSwitzerland
| | - Johanna Schaffenrath
- Department of Neurosurgery, Clinical Neuroscience CenterUniversity Hospital Zurich, University of ZurichZurichSwitzerland
| | | | - Flora Szeri
- Research Centre for Natural SciencesInstitute of EnzymologyBudapestHungary
| | - Melanie Greter
- Institute of Experimental ImmunologyUniversity of ZurichZurichSwitzerland
| | - Huiberdina L. Koek
- Department of Geriatric MedicineUniversity Medical Centre Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Annika Keller
- Department of Neurosurgery, Clinical Neuroscience CenterUniversity Hospital Zurich, University of ZurichZurichSwitzerland
- Neuroscience Center ZurichUniversity of Zurich and ETH ZurichZurichSwitzerland
| |
Collapse
|
36
|
Al Ali J, Yang J, Phillips MS, Fink J, Mastrianni J, Seibert K. A case report of a patient with primary familial brain calcification with a PDGFRB genetic variant. Front Neurol 2023; 14:1235909. [PMID: 37780723 PMCID: PMC10538541 DOI: 10.3389/fneur.2023.1235909] [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: 06/06/2023] [Accepted: 08/30/2023] [Indexed: 10/03/2023] Open
Abstract
Fahr's disease, or primary familial brain calcification (PFBC), is a rare genetic neurologic disease characterized by abnormal calcification of the basal ganglia, subcortical white matter and cerebellum. Common clinical features include parkinsonism, neuropsychiatric symptoms, and cognitive decline. Genes implicated in Fahr's disease include PDGFB, PDGFRB, SLC20A2, XPR1, MYORG, and JAM2. We present the case of a 51-year-old woman who developed subacute cognitive and behavioral changes primarily affecting frontal-subcortical pathways and parkinsonism in association with extensive bilateral calcifications within the basal ganglia, subcortical white matter, and cerebellum on neuroimaging. Relevant family history included a paternal aunt with parkinsonism at age 50. Normal parathyroid hormone and calcium levels in the patient's serum ruled out hypoparathyroidism or pseudohypoparathyroidism as causes for the intracranial calcifications. Genetic panel sequencing revealed a variant of unknown significance in the PDGFRB gene resulting in a p.Arg919Gln substitution in the tyrosine kinase domain of PDGFRB protein. To our knowledge this is the first report of a p.Arg919Gln variant in the PDGFRB gene associated with PFBC. Although co-segregation studies were not possible in this family, the location of the variant is within the tyrosine kinase domain of PDGFRB and pathogenicity calculators predict it is likely to be pathogenic. This report adds to the list of genetic variants that warrant functional analysis and could underlie the development of PFBC, which may help to further our understanding of its pathogenesis and the development of targeted therapies for this disorder.
Collapse
Affiliation(s)
- Jamal Al Ali
- Department of Neurology, University of Chicago, Chicago, IL, United States
| | - Jessica Yang
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, United States
| | - Matthew S. Phillips
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, United States
| | - Joseph Fink
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, United States
| | - James Mastrianni
- Department of Neurology, University of Chicago, Chicago, IL, United States
| | - Kaitlin Seibert
- Department of Neurology, University of Chicago, Chicago, IL, United States
| |
Collapse
|
37
|
Orimo K, Kakumoto T, Hara R, Goto R, Ishiura H, Mitsui J, Yoshida C, Uesaka Y, Suzuki Y, Morishita S, Satake W, Tsuji S, Toda T. A Japanese family with idiopathic basal ganglia calcification carrying a novel XPR1 variant. J Neurol Sci 2023; 451:120732. [PMID: 37490806 DOI: 10.1016/j.jns.2023.120732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/30/2023] [Accepted: 07/10/2023] [Indexed: 07/27/2023]
Affiliation(s)
- Kenta Orimo
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toshiyuki Kakumoto
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Ryo Hara
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ryoji Goto
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Ishiura
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Jun Mitsui
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Molecular Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | | | - Yuta Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Shinichi Morishita
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Wataru Satake
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shoji Tsuji
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Institute of Medical Genomics, International University of Health and Welfare, Narita, Chiba, Japan
| | - Tatsushi Toda
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
38
|
Reyes NGD, Lang AE. Adult-Onset Tourettism in SLC20A2-Associated Primary Familial Brain Calcification. Mov Disord Clin Pract 2023; 10:1152-1154. [PMID: 37476312 PMCID: PMC10354603 DOI: 10.1002/mdc3.13766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 04/04/2023] [Accepted: 04/28/2023] [Indexed: 07/22/2023] Open
Affiliation(s)
- Nikolai Gil D. Reyes
- Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western HospitalTorontoOntarioCanada
| | - Anthony E. Lang
- Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western HospitalTorontoOntarioCanada
| |
Collapse
|
39
|
D'Onofrio G, Scala M, Severino M, Roberti R, Romano F, De Marco P, Iacomino M, Baldassari S, Uva P, Pavanello M, Gustincich S, Striano P, Zara F, Capra V. Expanding the phenotype associated with biallelic SLC20A2 variants. Eur J Hum Genet 2023; 31:725-729. [PMID: 36977836 PMCID: PMC10326077 DOI: 10.1038/s41431-023-01349-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Affiliation(s)
- Gianluca D'Onofrio
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16132, Genoa, Italy
- Pediatric Neurology and Muscular Diseases Unit, IRCCS Giannina Gaslini Institute, 16147, Genoa, Italy
| | - Marcello Scala
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16132, Genoa, Italy.
- Pediatric Neurology and Muscular Diseases Unit, IRCCS Giannina Gaslini Institute, 16147, Genoa, Italy.
- Unit of Medical Genetics, IRCCS Giannina Gaslini Institute, 16147, Genoa, Italy.
| | | | - Roberta Roberti
- Science of Health Department, School of Medicine, Magna Græcia University, 88100, Catanzaro, Italy
| | - Ferruccio Romano
- Unit of Medical Genetics, IRCCS Giannina Gaslini Institute, 16147, Genoa, Italy
| | - Patrizia De Marco
- Unit of Medical Genetics, IRCCS Giannina Gaslini Institute, 16147, Genoa, Italy
| | - Michele Iacomino
- Unit of Medical Genetics, IRCCS Giannina Gaslini Institute, 16147, Genoa, Italy
| | - Simona Baldassari
- Unit of Medical Genetics, IRCCS Giannina Gaslini Institute, 16147, Genoa, Italy
| | - Paolo Uva
- Clinical Bioinformatics Unit, IRCCS Giannina Gaslini Institute, 16147, Genoa, Italy
| | - Marco Pavanello
- Department of Neurosurgery, IRCCS Giannina Gaslini Institute, 16147, Genoa, Italy
| | - Stefano Gustincich
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, 16163, Genoa, Italy
| | - Pasquale Striano
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16132, Genoa, Italy
- Pediatric Neurology and Muscular Diseases Unit, IRCCS Giannina Gaslini Institute, 16147, Genoa, Italy
| | - Federico Zara
- Unit of Medical Genetics, IRCCS Giannina Gaslini Institute, 16147, Genoa, Italy
| | - Valeria Capra
- Unit of Medical Genetics, IRCCS Giannina Gaslini Institute, 16147, Genoa, Italy.
| |
Collapse
|
40
|
Chen SY, Ho CJ, Lu YT, Lin CH, Lan MY, Tsai MH. The Genetics of Primary Familial Brain Calcification: A Literature Review. Int J Mol Sci 2023; 24:10886. [PMID: 37446066 DOI: 10.3390/ijms241310886] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Primary familial brain calcification (PFBC), also known as Fahr's disease, is a rare inherited disorder characterized by bilateral calcification in the basal ganglia according to neuroimaging. Other brain regions, such as the thalamus, cerebellum, and subcortical white matter, can also be affected. Among the diverse clinical phenotypes, the most common manifestations are movement disorders, cognitive deficits, and psychiatric disturbances. Although patients with PFBC always exhibit brain calcification, nearly one-third of cases remain clinically asymptomatic. Due to advances in the genetics of PFBC, the diagnostic criteria of PFBC may need to be modified. Hitherto, seven genes have been associated with PFBC, including four dominant inherited genes (SLC20A2, PDGFRB, PDGFB, and XPR1) and three recessive inherited genes (MYORG, JAM2, and CMPK2). Nevertheless, around 50% of patients with PFBC do not have pathogenic variants in these genes, and further PFBC-associated genes are waiting to be identified. The function of currently known genes suggests that PFBC could be caused by the dysfunction of the neurovascular unit, the dysregulation of phosphate homeostasis, or mitochondrial dysfunction. An improved understanding of the underlying pathogenic mechanisms for PFBC may facilitate the development of novel therapies.
Collapse
Affiliation(s)
- Shih-Ying Chen
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kaohsiung 833401, Taiwan
| | - Chen-Jui Ho
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kaohsiung 833401, Taiwan
| | - Yan-Ting Lu
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kaohsiung 833401, Taiwan
| | - Chih-Hsiang Lin
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kaohsiung 833401, Taiwan
| | - Min-Yu Lan
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kaohsiung 833401, Taiwan
- Center for Parkinson's Disease, Kaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kaohsiung 833401, Taiwan
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kaohsiung 833401, Taiwan
| | - Meng-Han Tsai
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kaohsiung 833401, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kaohsiung 833401, Taiwan
- Genomics and Proteomics Core Laboratory, Kaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kaohsiung 833401, Taiwan
| |
Collapse
|
41
|
Monfrini E, Arienti F, Rinchetti P, Lotti F, Riboldi GM. Brain Calcifications: Genetic, Molecular, and Clinical Aspects. Int J Mol Sci 2023; 24:ijms24108995. [PMID: 37240341 DOI: 10.3390/ijms24108995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/21/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Many conditions can present with accumulation of calcium in the brain and manifest with a variety of neurological symptoms. Brain calcifications can be primary (idiopathic or genetic) or secondary to various pathological conditions (e.g., calcium-phosphate metabolism derangement, autoimmune disorders and infections, among others). A set of causative genes associated with primary familial brain calcification (PFBC) has now been identified, and include genes such as SLC20A2, PDGFB, PDGFRB, XPR1, MYORG, and JAM2. However, many more genes are known to be linked with complex syndromes characterized by brain calcifications and additional neurologic and systemic manifestations. Of note, many of these genes encode for proteins involved in cerebrovascular and blood-brain barrier functions, which both represent key anatomical structures related to these pathological phenomena. As a growing number of genes associated with brain calcifications is identified, pathways involved in these conditions are beginning to be understood. Our comprehensive review of the genetic, molecular, and clinical aspects of brain calcifications offers a framework for clinicians and researchers in the field.
Collapse
Affiliation(s)
- Edoardo Monfrini
- Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy
- Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neurology Unit, 20122 Milan, Italy
| | - Federica Arienti
- Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neurology Unit, 20122 Milan, Italy
| | - Paola Rinchetti
- Columbia University Irving Medical Center, Center for Motor Neuron Biology and Diseases, Departments of Pathology & Cell Biology and Neurology, New York, NY 10032, USA
| | - Francesco Lotti
- Columbia University Irving Medical Center, Center for Motor Neuron Biology and Diseases, Departments of Pathology & Cell Biology and Neurology, New York, NY 10032, USA
| | - Giulietta M Riboldi
- The Marlene and Paolo Fresco Institute for Parkinson's and Movement Disorders, Department of Neurology, NYU Langone Health, New York, NY 10017, USA
| |
Collapse
|
42
|
Burgunder JM. Mechanisms underlying phenotypic variation in neurogenetic disorders. Nat Rev Neurol 2023:10.1038/s41582-023-00811-4. [PMID: 37202496 DOI: 10.1038/s41582-023-00811-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2023] [Indexed: 05/20/2023]
Abstract
Neurological diseases associated with pathogenic variants in a specific gene, or even with a specific pathogenic variant, can show profound phenotypic variation with regard to symptom presentation, age at onset and disease course. Highlighting examples from a range of neurogenetic disorders, this Review explores emerging mechanisms that are involved in this variability, including environmental, genetic and epigenetic factors that influence the expressivity and penetrance of pathogenic variants. Environmental factors, some of which can potentially be modified to prevent disease, include trauma, stress and metabolic changes. Dynamic patterns of pathogenic variants might explain some of the phenotypic variations, for example, in the case of disorders caused by DNA repeat expansions such as Huntington disease (HD). An important role for modifier genes has also been identified in some neurogenetic disorders, including HD, spinocerebellar ataxia and X-linked dystonia-parkinsonism. In other disorders, such as spastic paraplegia, the basis for most of the phenotypic variability remains unclear. Epigenetic factors have been implicated in disorders such as SGCE-related myoclonus-dystonia and HD. Knowledge of the mechanisms underlying phenotypic variation is already starting to influence management strategies and clinical trials for neurogenetic disorders.
Collapse
|
43
|
Di Fonzo A, Jinnah HA, Zech M. Dystonia genes and their biological pathways. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 169:61-103. [PMID: 37482402 DOI: 10.1016/bs.irn.2023.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
High-throughput sequencing has been instrumental in uncovering the spectrum of pathogenic genetic alterations that contribute to the etiology of dystonia. Despite the immense heterogeneity in monogenic causes, studies performed during the past few years have highlighted that many rare deleterious variants associated with dystonic presentations affect genes that have roles in certain conserved pathways in neural physiology. These various gene mutations that appear to converge towards the disruption of interconnected cellular networks were shown to produce a wide range of different dystonic disease phenotypes, including isolated and combined dystonias as well as numerous clinically complex, often neurodevelopmental disorder-related conditions that can manifest with dystonic features in the context of multisystem disturbances. In this chapter, we summarize the manifold dystonia-gene relationships based on their association with a discrete number of unifying pathophysiological mechanisms and molecular cascade abnormalities. The themes on which we focus comprise dopamine signaling, heavy metal accumulation and calcifications in the brain, nuclear envelope function and stress response, gene transcription control, energy homeostasis, lysosomal trafficking, calcium and ion channel-mediated signaling, synaptic transmission beyond dopamine pathways, extra- and intracellular structural organization, and protein synthesis and degradation. Enhancing knowledge about the concept of shared etiological pathways in the pathogenesis of dystonia will motivate clinicians and researchers to find more efficacious treatments that allow to reverse pathologies in patient-specific core molecular networks and connected multipathway loops.
Collapse
Affiliation(s)
- Alessio Di Fonzo
- Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neurology Unit, Milan, Italy
| | - H A Jinnah
- Departments of Neurology, Human Genetics, and Pediatrics, Atlanta, GA, United States
| | - Michael Zech
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany; Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany.
| |
Collapse
|
44
|
Zhang H, Lu Y, Kramer PR, Benson MD, Cheng YSL, Qin C. Intracranial calcification in Fam20c-deficient mice recapitulates human Raine syndrome. Neurosci Lett 2023; 802:137176. [PMID: 36914045 PMCID: PMC11795667 DOI: 10.1016/j.neulet.2023.137176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/26/2023] [Accepted: 03/08/2023] [Indexed: 03/15/2023]
Abstract
FAM20C (family with sequence similarity 20-member C) is a protein kinase that phosphorylates secretory proteins, including the proteins that are essential to the formation and mineralization of calcified tissues. FAM20C loss-of-function mutations cause Raine syndrome in humans, characterized by generalized osteosclerosis, distinctive craniofacial dysmorphism, along with extensive intracranial calcification. Our previous studies revealed that inactivation of Fam20c in mice led to hypophosphatemic rickets. In this study, we examined the expression of Fam20c in the mouse brain and investigated brain calcification in Fam20c-deficient mice. Reverse transcription polymerase chain reaction (RT-PCR), Western-blotting and in situ hybridization analyses demonstrated the broad expression of Fam20c in the mouse brain tissue. X-ray and histological analyses showed that the global deletion of Fam20c (mediated by Sox2-cre) resulted in brain calcification in mice after postnatal 3 months and that the calcifications were bilaterally distributed within the brain. There was mild perifocal microgliosis as well as astrogliosis around calcospherites. The calcifications were first observed in the thalamus, and later in the forebrain and hindbrain. Furthermore, brain-specific deletion (mediated by Nestin-cre) of Fam20c in mice also led to cerebral calcification at an older age (postnatal 6 months), but no obvious skeletal or dental defects. Our results suggest that the local loss of FAM20C function in the brain may directly account for intracranial calcification. We propose that FAM20C plays an essential role in maintaining normal brain homeostasis and preventing ectopic brain calcification.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, TX 75246, USA.
| | - Yongbo Lu
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, TX 75246, USA
| | - Phillip R Kramer
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, TX 75246, USA
| | - M Douglas Benson
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, TX 75246, USA
| | - Yi-Shing L Cheng
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, TX 75246, USA
| | - Chunlin Qin
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, TX 75246, USA
| |
Collapse
|
45
|
Hashimoto Y, Greene C, Munnich A, Campbell M. The CLDN5 gene at the blood-brain barrier in health and disease. Fluids Barriers CNS 2023; 20:22. [PMID: 36978081 PMCID: PMC10044825 DOI: 10.1186/s12987-023-00424-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
The CLDN5 gene encodes claudin-5 (CLDN-5) that is expressed in endothelial cells and forms tight junctions which limit the passive diffusions of ions and solutes. The blood-brain barrier (BBB), composed of brain microvascular endothelial cells and associated pericytes and end-feet of astrocytes, is a physical and biological barrier to maintain the brain microenvironment. The expression of CLDN-5 is tightly regulated in the BBB by other junctional proteins in endothelial cells and by supports from pericytes and astrocytes. The most recent literature clearly shows a compromised BBB with a decline in CLDN-5 expression increasing the risks of developing neuropsychiatric disorders, epilepsy, brain calcification and dementia. The purpose of this review is to summarize the known diseases associated with CLDN-5 expression and function. In the first part of this review, we highlight the recent understanding of how other junctional proteins as well as pericytes and astrocytes maintain CLDN-5 expression in brain endothelial cells. We detail some drugs that can enhance these supports and are being developed or currently in use to treat diseases associated with CLDN-5 decline. We then summarise mutagenesis-based studies which have facilitated a better understanding of the physiological role of the CLDN-5 protein at the BBB and have demonstrated the functional consequences of a recently identified pathogenic CLDN-5 missense mutation from patients with alternating hemiplegia of childhood. This mutation is the first gain-of-function mutation identified in the CLDN gene family with all others representing loss-of-function mutations resulting in mis-localization of CLDN protein and/or attenuated barrier function. Finally, we summarize recent reports about the dosage-dependent effect of CLDN-5 expression on the development of neurological diseases in mice and discuss what cellular supports for CLDN-5 regulation are compromised in the BBB in human diseases.
Collapse
Affiliation(s)
- Yosuke Hashimoto
- Trinity College Dublin, Smurfit Institute of Genetics, Dublin, D02 VF25, Ireland.
| | - Chris Greene
- Trinity College Dublin, Smurfit Institute of Genetics, Dublin, D02 VF25, Ireland
| | - Arnold Munnich
- Institut Imagine, INSERM UMR1163, Université Paris Cité, Paris, F-75015, France
- Departments of Pediatric Neurology and Medical Genetics, Hospital Necker Enfants Malades, Université Paris Cité, Paris, F-75015, France
| | - Matthew Campbell
- Trinity College Dublin, Smurfit Institute of Genetics, Dublin, D02 VF25, Ireland.
| |
Collapse
|
46
|
Carecchio M, Mainardi M, Bonato G. The clinical and genetic spectrum of primary familial brain calcification. J Neurol 2023; 270:3270-3277. [PMID: 36862146 DOI: 10.1007/s00415-023-11650-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023]
Abstract
Primary familial brain calcification (PFBC), formerly known as Fahr's disease, is a rare neurodegenerative disease characterized by bilateral progressive calcification of the microvessels of the basal ganglia and other cerebral and cerebellar structures. PFBC is thought to be due to an altered function of the Neurovascular Unit (NVU), where abnormal calcium-phosphorus metabolism, functional and microanatomical alterations of pericytes and mitochondrial alterations cause a dysfunction of the blood-brain barrier (BBB) and the generation of an osteogenic environment with surrounding astrocyte activation and progressive neurodegeneration. Seven causative genes have been discovered so far, of which four with dominant (SLC20A2, PDGFB, PDGFRB, XPR1) and three with recessive inheritance (MYORG, JAM2, CMPK2). Clinical presentation ranges from asymptomatic subjects to movement disorders, cognitive decline and psychiatric disturbances alone or in various combinations. Radiological patterns of calcium deposition are similar in all known genetic forms, but central pontine calcification and cerebellar atrophy are highly suggestive of MYORG mutations and extensive cortical calcification has been associated with JAM2 mutations. Currently, no disease-modifying drugs or calcium-chelating agents are available and only symptomatic treatments can be offered.
Collapse
Affiliation(s)
- Miryam Carecchio
- Department of Neuroscience, University of Padua, Via Niccolò Giustiniani, 5, 35128, Padua, Italy.
| | - Michele Mainardi
- Department of Neuroscience, University of Padua, Via Niccolò Giustiniani, 5, 35128, Padua, Italy
| | - Giulia Bonato
- Department of Neuroscience, University of Padua, Via Niccolò Giustiniani, 5, 35128, Padua, Italy
| |
Collapse
|
47
|
Reyes NGD, Lang AE. Vertical Supranuclear Gaze Palsy in Primary Familial Brain Calcification Associated with a Novel SLC20A2 Mutation. Mov Disord Clin Pract 2023; 10:501-503. [PMID: 36989014 PMCID: PMC10026275 DOI: 10.1002/mdc3.13604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 10/12/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Nikolai Gil D. Reyes
- Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders ClinicToronto Western HospitalTorontoOntarioCanada
| | - Anthony E. Lang
- Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders ClinicToronto Western HospitalTorontoOntarioCanada
| |
Collapse
|
48
|
Song T, Zhao Y, Wen G, Du J, Xu Q. A novel MYORG mutation causes primary familial brain calcification with migraine: Case report and literature review. Front Neurol 2023; 14:1110227. [PMID: 36816548 PMCID: PMC9932805 DOI: 10.3389/fneur.2023.1110227] [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: 11/28/2022] [Accepted: 01/13/2023] [Indexed: 02/05/2023] Open
Abstract
Primary familial brain calcification (PFBC) is a disorder in which pathologic calcification of the basal ganglia, cerebellum, or other brain regions with bilateral symmetry occurs. Common clinical symptoms include dysarthria, cerebellar symptoms, motor deficits, and cognitive impairment. Genetic factors are an important cause of the disease; however autosomal recessive (AR) inheritance is rare. In 2018, the myogenesis-regulated glycosidase (MYORG) gene was the first to be associated with AR-PFBC. The present case is a 24-year-old woman with AR-PFBC that presented with migraine at the age of 16 years. Symmetrical patchy calcifications were seen in the bilateral cerebellopontine nuclei, thalamus, basal ganglia, and radiocoronal area on computed tomography and magnetic resonance imaging. AR-PFBC with migraine as the main clinical symptom is rare. Whole-exome sequencing revealed a compound heterozygous mutation in the MYORG gene, one of which has not been previously reported. Our case highlights the pathogenic profile of the MYORG gene, and demonstrates the need for exclusion of calcium deposits in the brain for migraine patients with AR inheritance.
Collapse
Affiliation(s)
- Tingwei Song
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Yuwen Zhao
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Guo Wen
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Juan Du
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Qian Xu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China,*Correspondence: Qian Xu ✉
| |
Collapse
|
49
|
Golgi damage caused by dysfunction of PiT-2 in primary familial brain calcification. Biochem Biophys Res Commun 2023; 642:167-174. [PMID: 36584480 DOI: 10.1016/j.bbrc.2022.12.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 12/19/2022] [Indexed: 12/25/2022]
Abstract
The Golgi apparatus is vital for protein modification and molecular trafficking. It is essential for nerve development and activity, and damage thereof is implicated in many neurological diseases. Primary familial brain calcification (PFBC) is a rare inherited neurodegenerative disease characterized by multiple brain calcifications. SLC20A2, which encodes the inorganic phosphate transporter 2 (PiT-2) protein, is the main pathogenic gene in PFBC. The PiT-2 protein is a sodium-dependent phosphate type III transporter, and dysfunction leads to a deficit in the cellular intake of inorganic phosphate (Pi) and calcium deposits. Whether the impaired Golgi apparatus is involved in the PFBC procession requires elucidation. In this study, we constructed induced pluripotent stem cells (iPSCs) derived from two PFBC patients with different SLC20A2 gene mutations (c.613G > A or del exon10) and two healthy volunteers as dependable cell models for research on pathogenic mechanism. To study the mechanism, we differentiated iPSCs into neurons and astrocytes in vitro. Our study found disruptive Golgi structure and damaged autophagy in PFBC neurons with increased activity of mTOR. We also found damaged mitochondria and increased apoptosis in the PFBC dopaminergic neurons and astrocytes. In this study, we prove that dysfunctional PiT-2 leads to an imbalance of cellular Pi, which may disrupt the Golgi apparatus with impaired autophagy, mitochondria and apoptosis in PFBC. Our study provides a new avenue for understanding nerve damage and pathogenic mechanism in brain calcifications.
Collapse
|
50
|
Sun H, Xu X, Luo J, Ma T, Cui J, Liu M, Xiong B, Zhu S, Liu JY. Mechanisms of PiT2-loop7 Missense Mutations Induced Pi Dyshomeostasis. Neurosci Bull 2023; 39:57-68. [PMID: 35713844 PMCID: PMC9849530 DOI: 10.1007/s12264-022-00893-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 04/14/2022] [Indexed: 01/25/2023] Open
Abstract
PiT2 is an inorganic phosphate (Pi) transporter whose mutations are linked to primary familial brain calcification (PFBC). PiT2 mainly consists of two ProDom (PD) domains and a large intracellular loop region (loop7). The PD domains are crucial for the Pi transport, but the role of PiT2-loop7 remains unclear. In PFBC patients, mutations in PiT2-loop7 are mainly nonsense or frameshift mutations that probably cause PFBC due to C-PD1131 deletion. To date, six missense mutations have been identified in PiT2-loop7; however, the mechanisms by which these mutations cause PFBC are poorly understood. Here, we found that the p.T390A and p.S434W mutations in PiT2-loop7 decreased the Pi transport activity and cell surface levels of PiT2. Furthermore, we showed that these two mutations attenuated its membrane localization by affecting adenosine monophosphate-activated protein kinase (AMPK)- or protein kinase B (AKT)-mediated PiT2 phosphorylation. In contrast, the p.S121C and p.S601W mutations in the PD domains did not affect PiT2 phosphorylation but rather impaired its substrate-binding abilities. These results suggested that missense mutations in PiT2-loop7 can cause Pi dyshomeostasis by affecting the phosphorylation-regulated cell-surface localization of PiT2. This study helps understand the pathogenesis of PFBC caused by PiT2-loop7 missense mutations and indicates that increasing the phosphorylation levels of PiT2-loop7 could be a promising strategy for developing PFBC therapies.
Collapse
Affiliation(s)
- Hao Sun
- College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan, 430074, China
| | - Xuan Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Junyu Luo
- College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan, 430074, China
| | - Tingbin Ma
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jiaming Cui
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Mugen Liu
- College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan, 430074, China
| | - Bo Xiong
- Department of Forensic Medicine, Tongji Medical College, HUST, Wuhan, 430030, China
| | - Shujia Zhu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Jing-Yu Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|