1
|
Pierre-Ferrer S, Collins B, Lukacsovich D, Wen S, Cai Y, Winterer J, Yan J, Pedersen L, Földy C, Brown SA. A phosphate transporter in VIPergic neurons of the suprachiasmatic nucleus gates locomotor activity during the light/dark transition in mice. Cell Rep 2024; 43:114220. [PMID: 38735047 DOI: 10.1016/j.celrep.2024.114220] [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: 07/22/2023] [Revised: 02/23/2024] [Accepted: 04/25/2024] [Indexed: 05/14/2024] Open
Abstract
The suprachiasmatic nucleus (SCN) encodes time of day through changes in daily firing; however, the molecular mechanisms by which the SCN times behavior are not fully understood. To identify factors that could encode day/night differences in activity, we combine patch-clamp recordings and single-cell sequencing of individual SCN neurons in mice. We identify PiT2, a phosphate transporter, as being upregulated in a population of Vip+Nms+ SCN neurons at night. Although nocturnal and typically showing a peak of activity at lights off, mice lacking PiT2 (PiT2-/-) do not reach the activity level seen in wild-type mice during the light/dark transition. PiT2 loss leads to increased SCN neuronal firing and broad changes in SCN protein phosphorylation. PiT2-/- mice display a deficit in seasonal entrainment when moving from a simulated short summer to longer winter nights. This suggests that PiT2 is responsible for timing activity and is a driver of SCN plasticity allowing seasonal entrainment.
Collapse
Affiliation(s)
- Sara Pierre-Ferrer
- Chronobiology and Sleep Research Group, Institute of Pharmacology and Toxicology, Faculties of Medicine and Science, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland.
| | - Ben Collins
- Chronobiology and Sleep Research Group, Institute of Pharmacology and Toxicology, Faculties of Medicine and Science, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland; Department of Biology, Sacred Heart University, 5151 Park Ave., Fairfield, CT 06825, USA
| | - David Lukacsovich
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Shao'Ang Wen
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuchen Cai
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jochen Winterer
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Jun Yan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Lene Pedersen
- Department of Molecular Biology and Genetics, Aarhus University, Universitetsbyen 81, 8000 Aarhus, Denmark
| | - Csaba Földy
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland.
| | - Steven A Brown
- Chronobiology and Sleep Research Group, Institute of Pharmacology and Toxicology, Faculties of Medicine and Science, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| |
Collapse
|
2
|
Villa-Bellosta R. Vascular Calcification: A Passive Process That Requires Active Inhibition. BIOLOGY 2024; 13:111. [PMID: 38392329 PMCID: PMC10886409 DOI: 10.3390/biology13020111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/07/2024] [Accepted: 02/08/2024] [Indexed: 02/24/2024]
Abstract
The primary cause of worldwide mortality and morbidity stems from complications in the cardiovascular system resulting from accelerated atherosclerosis and arterial stiffening. Frequently, both pathologies are associated with the pathological calcification of cardiovascular structures, present in areas such as cardiac valves or blood vessels (vascular calcification). The accumulation of hydroxyapatite, the predominant form of calcium phosphate crystals, is a distinctive feature of vascular calcification. This phenomenon is commonly observed as a result of aging and is also linked to various diseases such as diabetes, chronic kidney disease, and several genetic disorders. A substantial body of evidence indicates that vascular calcification involves two primary processes: a passive process and an active process. The physicochemical process of hydroxyapatite formation and deposition (a passive process) is influenced significantly by hyperphosphatemia. However, the active synthesis of calcification inhibitors, including proteins and low-molecular-weight inhibitors such as pyrophosphate, is crucial. Excessive calcification occurs when there is a loss of function in enzymes and transporters responsible for extracellular pyrophosphate metabolism. Current in vivo treatments to prevent calcification involve addressing hyperphosphatemia with phosphate binders and implementing strategies to enhance the availability of pyrophosphate.
Collapse
Affiliation(s)
- Ricardo Villa-Bellosta
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Campus Vida, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Department of Biochemistry and Molecular Biology, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- The Health Research Institute of Santiago de Compostela (IDIS), Travesia da Choupana S/N, 15706 Santiago de Compostela, Spain
| |
Collapse
|
3
|
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: 1.0] [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
|
4
|
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: 6] [Impact Index Per Article: 6.0] [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
|
5
|
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
|
6
|
Jennings ML. Role of transporters in regulating mammalian intracellular inorganic phosphate. Front Pharmacol 2023; 14:1163442. [PMID: 37063296 PMCID: PMC10097972 DOI: 10.3389/fphar.2023.1163442] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 03/17/2023] [Indexed: 03/31/2023] Open
Abstract
This review summarizes the current understanding of the role of plasma membrane transporters in regulating intracellular inorganic phosphate ([Pi]In) in mammals. Pi influx is mediated by SLC34 and SLC20 Na+-Pi cotransporters. In non-epithelial cells other than erythrocytes, Pi influx via SLC20 transporters PiT1 and/or PiT2 is balanced by efflux through XPR1 (xenotropic and polytropic retrovirus receptor 1). Two new pathways for mammalian Pi transport regulation have been described recently: 1) in the presence of adequate Pi, cells continuously internalize and degrade PiT1. Pi starvation causes recycling of PiT1 from early endosomes to the plasma membrane and thereby increases the capacity for Pi influx; and 2) binding of inositol pyrophosphate InsP8 to the SPX domain of XPR1 increases Pi efflux. InsP8 is degraded by a phosphatase that is strongly inhibited by Pi. Therefore, an increase in [Pi]In decreases InsP8 degradation, increases InsP8 binding to SPX, and increases Pi efflux, completing a feedback loop for [Pi]In homeostasis. Published data on [Pi]In by magnetic resonance spectroscopy indicate that the steady state [Pi]In of skeletal muscle, heart, and brain is normally in the range of 1–5 mM, but it is not yet known whether PiT1 recycling or XPR1 activation by InsP8 contributes to Pi homeostasis in these organs. Data on [Pi]In in cultured cells are variable and suggest that some cells can regulate [Pi] better than others, following a change in [Pi]Ex. More measurements of [Pi]In, influx, and efflux are needed to determine how closely, and how rapidly, mammalian [Pi]In is regulated during either hyper- or hypophosphatemia.
Collapse
|
7
|
Kar P, Goswami R. Effect of calcitriol and calcium on basal ganglia calcification in hypoparathyroidism: experimental models. J Mol Endocrinol 2023; 70:JME-22-0108. [PMID: 36445941 DOI: 10.1530/jme-22-0108] [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: 11/02/2022] [Accepted: 11/29/2022] [Indexed: 12/02/2022]
Abstract
Basal ganglia calcification (BGC) is a common complication in hypoparathyroid patients, linked to hyperphosphatemia and altered vitamin-D and calcium homeostasis following conventional therapy. The pathogenesis of BGC in hypoparathyroidism is not clear. Recently, we developed an ex vivo model of BGC using rat-striatal cell culture in 10.0 mmol/L of β-glycerophosphate (31.8 mg/dL phosphate). However, the effect of 1,25(OH)2 D, calcium, and milder phosphate excess on BGC in hypoparathyroidism is not known. This study describes two modified ex vivo models investigating pathogenesis of BGC in 'drug-naïve' and 'conventionally treated' hypoparathyroid state. The first modification involved striatal cells cultured in low concentration 1,25(OH)2D (16.0 pg/mL), ionized calcium(0.99 mmol/L), hPTH(1-34) (6.0 pg/mL), and 2.68 mmol/L (8.3 mg/dL) of phosphate akin to 'drug-naïve' state for 24 days. In second modification, striatal cells were exposed to 46.0 pg/mL of 1,25(OH)2D, normal ionized calcium of 1.17 mmol/L, and 2.20 mmol/L (6.8 mg/dL) of phosphate akin to 'conventionally treated' state. Striatal cell culture under 'drug-naïve' state showed that even 16.0 pg/mL of 1,25(OH)2D enhanced the calcification. In 'conventionally treated' model, striatal cell calcification was enhanced in 54% cases over 'drug-naïve' state. Calcification in 'conventionally treated' state further increased on increasing phosphate to 8.3 mg/dL, suggesting importance of phosphatemic control in hypoparathyroid patients. Striatal cells in 'drug-naïve' state showed increased mRNA expression of pro-osteogenic Wnt3a, Cd133,Vglut-1-neuronal phosphate-transporters, calcium-ion channel-Trvp2,Alp, and Collagen-1α and decreased expression of Ca-II. These models suggest that in 'drug-naïve' state, 1,25(OH)2D along with moderately elevated phosphate increases the expression of pro-osteogenic molecules to induce BGC. Although normalization of calcium in 'conventionally treated' state increased the expression of Opg, Osterix, Alp, and Cav2, calcification increased only in a subset, akin to variation in progression of BGC in hypoparathyroid patients on conventional therapy.
Collapse
Affiliation(s)
- Parmita Kar
- 1Department of Endocrinology and Metabolism, All India Institute of Medical Sciences, New Delhi, India
| | - Ravinder Goswami
- 1Department of Endocrinology and Metabolism, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
8
|
The Pathology of Primary Familial Brain Calcification: Implications for Treatment. Neurosci Bull 2022; 39:659-674. [PMID: 36469195 PMCID: PMC10073384 DOI: 10.1007/s12264-022-00980-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 07/10/2022] [Indexed: 12/08/2022] Open
Abstract
AbstractPrimary familial brain calcification (PFBC) is an inherited neurodegenerative disorder mainly characterized by progressive calcium deposition bilaterally in the brain, accompanied by various symptoms, such as dystonia, ataxia, parkinsonism, dementia, depression, headaches, and epilepsy. Currently, the etiology of PFBC is largely unknown, and no specific prevention or treatment is available. During the past 10 years, six causative genes (SLC20A2, PDGFRB, PDGFB, XPR1, MYORG, and JAM2) have been identified in PFBC. In this review, considering mechanistic studies of these genes at the cellular level and in animals, we summarize the pathogenesis and potential preventive and therapeutic strategies for PFBC patients. Our systematic analysis suggests a classification for PFBC genetic etiology based on several characteristics, provides a summary of the known composition of brain calcification, and identifies some potential therapeutic targets for PFBC.
Collapse
|
9
|
Villa-Bellosta R. Role of the extracellular ATP/pyrophosphate metabolism cycle in vascular calcification. Purinergic Signal 2022:10.1007/s11302-022-09867-1. [PMID: 35511317 DOI: 10.1007/s11302-022-09867-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/19/2022] [Indexed: 10/18/2022] Open
Abstract
Conventionally, ATP is considered to be the principal energy source in cells. However, over the last few years, a novel role for ATP as a potent extracellular signaling molecule and the principal source of extracellular pyrophosphate, the main endogenous inhibitor of vascular calcification, has emerged. A large body of evidence suggests that two principal mechanisms are involved in the initiation and progression of ectopic calcification: high phosphate concentration and pyrophosphate deficiency. Pathologic calcification of cardiovascular structures, or vascular calcification, is a feature of several genetic diseases and a common complication of chronic kidney disease, diabetes, and aging. Previous studies have shown that the loss of function of several enzymes and transporters involved in extracellular ATP/pyrophosphate metabolism is associated with vascular calcification. Therefore, pyrophosphate homeostasis should be further studied to facilitate the design of novel therapeutic approaches for ectopic calcification of cardiovascular structures, including strategies to increase pyrophosphate concentrations by targeting the ATP/pyrophosphate metabolism cycle.
Collapse
Affiliation(s)
- Ricardo Villa-Bellosta
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Av Barcelona, Campus Vida, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain. .,Department of Biochemistry and Molecular Biology, Universidade de Santiago de Compostela, Plaza do Obradoiro s/n, Santiago de Compostela, Spain.
| |
Collapse
|
10
|
Carbone MG, Della Rocca F. Neuropsychiatric Manifestations of Fahr's Disease, Diagnostic and Therapeutic Challenge: A Case Report and a Literature Review. CLINICAL NEUROPSYCHIATRY 2022; 19:121-131. [PMID: 35601245 PMCID: PMC9112992 DOI: 10.36131/cnfioritieditore20220206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Objective Calcifications in basal ganglia could be an incidental finding up to 20% of asymptomatic patients undergoing computed tomography (CT) or magnetic resonance imaging (MRI) scan. The presence of neuropsychiatric symptomatology associated with basal ganglia calcifications identifies a clinical entity defined as Fahr's Disease. This term is used in presence of calcifications secondary to a specific cause, but the variability of etiology, pathogenesis, and clinical picture underlying this condition have raised the question of the real existence of a syndrome. Several classifications based on the etiology, the location of brain calcifications and the clinical presentation have been proposed. Method In the present study, we describe the case of a 52 years old man with a Bipolar I disorder diagnosis and a recent onset of behavioral disinhibition and alcohol misuse. The patient came to our center, specialized for bipolar disorder, as a consequence of a progressive worsening of the clinical picture associated to behavioral disturbances (sexual disinhibition, episodes of binge-eating, alcohol misuse), initial degrees of deterioration in cognitive function, peculiar psychotic symptoms and a resistance to various psychopharmacological treatment. The patient underwent neuro-psychologic evaluation, laboratory examinations and neuroimaging. Results and Conclusions CT and MRI revealed basal ganglia calcification and, in presence of normal blood tests, a diagnosis of Fahr's syndrome was suggested. During the hospitalization, the patient showed a good clinical response to a psychopharmacological therapy constituted by two mood stabilizers (lithium carbonate and oxcarbazepine) and mild antipsychotics doses (quetiapine and aripiprazole). Finally, we performed a literature review on the complex and multifaceted neuropsychiatric clinical manifestations of Fahr's disease in order to provide useful elements in terms of etiology, clinical manifestation, diagnosis, and treatment.
Collapse
Affiliation(s)
- Manuel Glauco Carbone
- Department of Medicine and Surgery, Division of Psychiatry, University of Insubria, Viale Luigi Borri 57, 21100 Varese, Italy,Pisa-School of Experimental and Clinical Psychiatry, University of Pisa, Via Roma 57, 56100, Pisa, Italy.,Corresponding author Manuel Glauco Carbone, M.D. Department of Medicine and Surgery, Division of Psychiatry, University of Insubria, Viale Luigi Borri 57, 21100 Varese, Italy E-mail:
| | - Filippo Della Rocca
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 57, 56100, Pisa, Italy.,
| |
Collapse
|
11
|
Nguyen NT, Nguyen TT, Park KS. Oxidative Stress Related to Plasmalemmal and Mitochondrial Phosphate Transporters in Vascular Calcification. Antioxidants (Basel) 2022; 11:antiox11030494. [PMID: 35326144 PMCID: PMC8944874 DOI: 10.3390/antiox11030494] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/26/2022] [Accepted: 02/28/2022] [Indexed: 12/04/2022] Open
Abstract
Inorganic phosphate (Pi) is essential for maintaining cellular function but excess of Pi leads to serious complications, including vascular calcification. Accumulating evidence suggests that oxidative stress contributes to the pathogenic progression of calcific changes. However, the molecular mechanism underlying Pi-induced reactive oxygen species (ROS) generation and its detrimental consequences remain unclear. Type III Na+-dependent Pi cotransporter, PiT-1/-2, play a significant role in Pi uptake of vascular smooth muscle cells. Pi influx via PiT-1/-2 increases the abundance of PiT-1/-2 and depolarization-activated Ca2+ entry due to its electrogenic properties, which may lead to Ca2+ and Pi overload and oxidative stress. At least four mitochondrial Pi transporters are suggested, among which the phosphate carrier (PiC) is known to be mainly involved in mitochondrial Pi uptake. Pi transport via PiC may induce hyperpolarization and superoxide generation, which may lead to mitochondrial dysfunction and endoplasmic reticulum stress, together with generation of cytosolic ROS. Increase in net influx of Ca2+ and Pi and their accumulation in the cytosol and mitochondrial matrix synergistically increases oxidative stress and osteogenic differentiation, which could be prevented by suppressing either Ca2+ or Pi overload. Therapeutic strategies targeting plasmalemmal and mitochondrial Pi transports can protect against Pi-induced oxidative stress and vascular calcification.
Collapse
Affiliation(s)
- Nhung Thi Nguyen
- Department of Physiology, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea;
- Mitohormesis Research Center, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea
- Medical Doctor Program, College of Health Sciences, VinUniversity, Hanoi 12406, Vietnam
| | - Tuyet Thi Nguyen
- Department of Physiology, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea;
- Internal Medicine Residency Program, College of Health Sciences, VinUniversity, Hanoi 12406, Vietnam
- Correspondence: (T.T.N.); (K.-S.P.); Tel.: +84-247-108-9779 (T.T.N.); +82-33-741-0294 (K.-S.P.)
| | - Kyu-Sang Park
- Department of Physiology, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea;
- Mitohormesis Research Center, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea
- Correspondence: (T.T.N.); (K.-S.P.); Tel.: +84-247-108-9779 (T.T.N.); +82-33-741-0294 (K.-S.P.)
| |
Collapse
|
12
|
Maheshwari U, Huang SF, Sridhar S, Keller A. The Interplay Between Brain Vascular Calcification and Microglia. Front Aging Neurosci 2022; 14:848495. [PMID: 35309892 PMCID: PMC8924545 DOI: 10.3389/fnagi.2022.848495] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/11/2022] [Indexed: 12/17/2022] Open
Abstract
Vascular calcifications are characterized by the ectopic deposition of calcium and phosphate in the vascular lumen or wall. They are a common finding in computed tomography scans or during autopsy and are often directly related to a pathological condition. While the pathogenesis and functional consequences of vascular calcifications have been intensively studied in some peripheral organs, vascular calcification, and its pathogenesis in the central nervous system is poorly characterized and understood. Here, we review the occurrence of vessel calcifications in the brain in the context of aging and various brain diseases. We discuss the pathomechanism of brain vascular calcification in primary familial brain calcification as an example of brain vessel calcification. A particular focus is the response of microglia to the vessel calcification in the brain and their role in the clearance of calcifications.
Collapse
Affiliation(s)
- Upasana Maheshwari
- Department of Neurosurgery, Clinical Neuroscience Center, Zürich University Hospital, University of Zürich, Zurich, Switzerland
| | - Sheng-Fu Huang
- Department of Neurosurgery, Clinical Neuroscience Center, Zürich University Hospital, University of Zürich, Zurich, Switzerland
| | - Sucheta Sridhar
- Department of Neurosurgery, Clinical Neuroscience Center, Zürich University Hospital, University of Zürich, Zurich, Switzerland
- Neuroscience Center Zürich, University of Zürich and ETH Zürich, Zurich, Switzerland
| | - Annika Keller
- Department of Neurosurgery, Clinical Neuroscience Center, Zürich University Hospital, University of Zürich, Zurich, Switzerland
- Neuroscience Center Zürich, University of Zürich and ETH Zürich, Zurich, Switzerland
- *Correspondence: Annika Keller,
| |
Collapse
|
13
|
Characteristics and therapeutic potential of sodium-dependent phosphate cotransporters in relation to idiopathic basal ganglia calcification. J Pharmacol Sci 2021; 148:152-155. [PMID: 34924120 DOI: 10.1016/j.jphs.2021.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/30/2021] [Accepted: 11/08/2021] [Indexed: 12/18/2022] Open
Abstract
Type-III sodium-dependent phosphate transporters 1 and 2 (PiT 1 and PiT 2, respectively) are proteins encoded by SLC20A1 and SLC20A2, respectively. The ubiquitous distribution of SLC20A1 and SLC20A2 mRNAs in mammalian tissues supports the housekeeping maintenance and homeostasis of intracellular inorganic phosphate (Pi), which is absorbed from interstitial fluid for normal cellular functions. SLC20A2 variants have been found in patients with idiopathic basal ganglia calcification (IBGC), also known as Fahr's disease or primary familial brain calcification (PFBC). Thus, disrupted Pi homeostasis is considered one of the major factors in the pathogenic mechanism of IBGC. In this paper, among the causative genes of IBGC, we focused specifically on PiT2, and its potential for a therapeutic target of IBGC.
Collapse
|
14
|
Radvar E, Griffanti G, Tsolaki E, Bertazzo S, Nazhat SN, Addison O, Mata A, Shanahan CM, Elsharkawy S. Engineered In vitro Models for Pathological Calcification: Routes Toward Mechanistic Understanding. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Elham Radvar
- Centre for Oral, Clinical and Translational Sciences Faculty of Dentistry, Oral and Craniofacial Sciences King's College London London SE1 1UL UK
| | - Gabriele Griffanti
- Department of Mining and Materials Engineering Faculty of Engineering McGill University Montreal QC H3A 0C5 Canada
| | - Elena Tsolaki
- Department of Medical Physics and Biomedical Engineering University College London London WC1E 6BT UK
| | - Sergio Bertazzo
- Department of Medical Physics and Biomedical Engineering University College London London WC1E 6BT UK
| | - Showan N. Nazhat
- Department of Mining and Materials Engineering Faculty of Engineering McGill University Montreal QC H3A 0C5 Canada
| | - Owen Addison
- Centre for Oral, Clinical and Translational Sciences Faculty of Dentistry, Oral and Craniofacial Sciences King's College London London SE1 1UL UK
| | - Alvaro Mata
- School of Pharmacy University of Nottingham Nottingham NG7 2RD UK
| | - Catherine M. Shanahan
- BHF Centre of Research Excellence Cardiovascular Division James Black Centre King's College London London SE1 1UL UK
| | - Sherif Elsharkawy
- Centre for Oral, Clinical and Translational Sciences Faculty of Dentistry, Oral and Craniofacial Sciences King's College London London SE1 1UL UK
| |
Collapse
|
15
|
Suzuki T, Katada E, Mizuoka Y, Takagi S, Kazuki Y, Oshimura M, Shindo M, Hara T. A novel all-in-one conditional knockout system uncovered an essential role of DDX1 in ribosomal RNA processing. Nucleic Acids Res 2021; 49:e40. [PMID: 33503245 PMCID: PMC8053084 DOI: 10.1093/nar/gkaa1296] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/09/2020] [Accepted: 01/04/2021] [Indexed: 11/13/2022] Open
Abstract
Generation of conditional knockout (cKO) and various gene-modified cells is laborious and time-consuming. Here, we established an all-in-one cKO system, which enables highly efficient generation of cKO cells and simultaneous gene modifications, including epitope tagging and reporter gene knock-in. We applied this system to mouse embryonic stem cells (ESCs) and generated RNA helicase Ddx1 cKO ESCs. The targeted cells displayed endogenous promoter-driven EGFP and FLAG-tagged DDX1 expression, and they were converted to Ddx1 KO via FLP recombinase. We further established TetFE ESCs, which carried a reverse tetracycline transactivator (rtTA) expression cassette and a tetracycline response element (TRE)-regulated FLPERT2 cassette in the Gt(ROSA26)Sor locus for instant and tightly regulated induction of gene KO. By utilizing TetFE Ddx1F/F ESCs, we isolated highly pure Ddx1F/F and Ddx1−/− ESCs and found that loss of Ddx1 caused rRNA processing defects, thereby activating the ribosome stress–p53 pathway. We also demonstrated cKO of various genes in ESCs and homologous recombination-non-proficient human HT1080 cells. The frequency of cKO clones was remarkably high for both cell types and reached up to 96% when EGFP-positive clones were analyzed. This all-in-one cKO system will be a powerful tool for rapid and precise analyses of gene functions.
Collapse
Affiliation(s)
- Teruhiko Suzuki
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Eiji Katada
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.,Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Yuki Mizuoka
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.,Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Satoko Takagi
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.,Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Yasuhiro Kazuki
- Chromosome Engineering Research Center (CERC), Tottori University, 86 Nishicho, Yonago 683-8503, Japan.,Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Mitsuo Oshimura
- Chromosome Engineering Research Center (CERC), Tottori University, 86 Nishicho, Yonago 683-8503, Japan
| | - Mayumi Shindo
- Center for Basic Technology Research, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Takahiko Hara
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.,Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan.,Graduate School of Science, Department of Biological Science, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji-shi, Tokyo 192-0397, Japan
| |
Collapse
|
16
|
Bird RP, Eskin NAM. The emerging role of phosphorus in human health. ADVANCES IN FOOD AND NUTRITION RESEARCH 2021; 96:27-88. [PMID: 34112356 DOI: 10.1016/bs.afnr.2021.02.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Phosphorus, an essential nutrient, performs vital functions in skeletal and non-skeletal tissues and is pivotal for energy production. The last two decades of research on the physiological importance of phosphorus have provided several novel insights about its dynamic nature as a nutrient performing functions as a phosphate ion. Phosphorous also acts as a signaling molecule and induces complex physiological responses. It is recognized that phosphorus homeostasis is critical for health. The intake of phosphorus by the general population world-wide is almost double the amount required to maintain health. This increase is attributed to the incorporation of phosphate containing food additives in processed foods purchased by consumers. Research findings assessed the impact of excessive phosphorus intake on cells' and organs' responses, and highlighted the potential pathogenic consequences. Research also identified a new class of bioactive phosphates composed of polymers of phosphate molecules varying in chain length. These polymers are involved in metabolic responses including hemostasis, brain and bone health, via complex mechanism(s) with positive or negative health effects, depending on their chain length. It is amazing, that phosphorus, a simple element, is capable of exerting multiple and powerful effects. The role of phosphorus and its polymers in the renal and cardiovascular system as well as on brain health appear to be important and promising future research directions.
Collapse
Affiliation(s)
- Ranjana P Bird
- School of Health Sciences, University of Northern British Columbia, Prince George, BC, Canada.
| | - N A Michael Eskin
- Department of Food and Human Nutritional Sciences, Faculty of Agricultural and Food Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
17
|
Ren Y, Shen Y, Si N, Fan S, Zhang Y, Xu W, Shi L, Zhang X. Slc20a2-Deficient Mice Exhibit Multisystem Abnormalities and Impaired Spatial Learning Memory and Sensorimotor Gating but Normal Motor Coordination Abilities. Front Genet 2021; 12:639935. [PMID: 33889180 PMCID: PMC8056086 DOI: 10.3389/fgene.2021.639935] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 03/03/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Primary familial brain calcification (PFBC, OMIM#213600), also known as Fahr's disease, is a rare autosomal dominant or recessive neurodegenerative disorder characterized by bilateral and symmetrical microvascular calcifications affecting multiple brain regions, particularly the basal ganglia (globus pallidus, caudate nucleus, and putamen) and thalamus. The most common clinical manifestations include cognitive impairment, neuropsychiatric signs, and movement disorders. Loss-of-function mutations in SLC20A2 are the major genetic causes of PFBC. OBJECTIVE This study aimed to investigate whether Slc20a2 knockout mice could recapitulate the dynamic processes and patterns of brain calcification and neurological symptoms in patients with PFBC. We comprehensively evaluated brain calcifications and PFBC-related behavioral abnormalities in Slc20a2-deficient mice. METHODS Brain calcifications were analyzed using classic calcium-phosphate staining methods. The Morris water maze, Y-maze, and fear conditioning paradigms were used to evaluate long-term spatial learning memory, working memory, and episodic memory, respectively. Sensorimotor gating was mainly assessed using the prepulse inhibition of the startle reflex program. Spontaneous locomotor activity and motor coordination abilities were evaluated using the spontaneous activity chamber, cylinder test, accelerating rotor-rod, and narrowing balance beam tests. RESULTS Slc20a2 homozygous knockout (Slc20a2-HO) mice showed congenital and global developmental delay, lean body mass, skeletal malformation, and a high proportion of unilateral or bilateral eye defects. Brain calcifications were detected in the hypothalamus, ventral thalamus, and midbrain early at postnatal day 80 in Slc20a2-HO mice, but were seldom found in Slc20a2 heterozygous knockout (Slc20a2-HE) mice, even at extremely old age. Slc20a2-HO mice exhibited spatial learning memory impairments and sensorimotor gating deficits while exhibiting normal working and episodic memories. The general locomotor activity, motor balance, and coordination abilities were not statistically different between Slc20a2-HO and wild-type mice after adjusting for body weight, which was a major confounding factor in our motor function evaluations. CONCLUSION The human PFBC-related phenotypes were highly similar to those in Slc20a2-HO mice. Therefore, Slc20a2-HO mice might be suitable for the future evaluation of neuropharmacological intervention strategies targeting cognitive and neuropsychiatric impairments.
Collapse
Affiliation(s)
- Yaqiong Ren
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yuqi Shen
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Nuo Si
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Shiqi Fan
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yi Zhang
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, China
| | - Wanhai Xu
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, China
| | - Lei Shi
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, China
| | - Xue Zhang
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, China
| |
Collapse
|
18
|
Guérit E, Arts F, Dachy G, Boulouadnine B, Demoulin JB. PDGF receptor mutations in human diseases. Cell Mol Life Sci 2021; 78:3867-3881. [PMID: 33449152 PMCID: PMC11072557 DOI: 10.1007/s00018-020-03753-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/16/2020] [Accepted: 12/29/2020] [Indexed: 12/16/2022]
Abstract
PDGFRA and PDGFRB are classical proto-oncogenes that encode receptor tyrosine kinases responding to platelet-derived growth factor (PDGF). PDGFRA mutations are found in gastrointestinal stromal tumors (GISTs), inflammatory fibroid polyps and gliomas, and PDGFRB mutations drive myofibroma development. In addition, chromosomal rearrangement of either gene causes myeloid neoplasms associated with hypereosinophilia. Recently, mutations in PDGFRB were linked to several noncancerous diseases. Germline heterozygous variants that reduce receptor activity have been identified in primary familial brain calcification, whereas gain-of-function mutants are present in patients with fusiform aneurysms, Kosaki overgrowth syndrome or Penttinen premature aging syndrome. Functional analysis of these variants has led to the preclinical validation of tyrosine kinase inhibitors targeting PDGF receptors, such as imatinib, as a treatment for some of these conditions. This review summarizes the rapidly expanding knowledge in this field.
Collapse
Affiliation(s)
- Emilie Guérit
- De Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 75, Box B1.74.05, 1200, Brussels, Belgium
| | - Florence Arts
- De Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 75, Box B1.74.05, 1200, Brussels, Belgium
| | - Guillaume Dachy
- De Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 75, Box B1.74.05, 1200, Brussels, Belgium
| | - Boutaina Boulouadnine
- De Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 75, Box B1.74.05, 1200, Brussels, Belgium
| | - Jean-Baptiste Demoulin
- De Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 75, Box B1.74.05, 1200, Brussels, Belgium.
| |
Collapse
|
19
|
Ribet ABP, Ng PY, Pavlos NJ. Membrane Transport Proteins in Osteoclasts: The Ins and Outs. Front Cell Dev Biol 2021; 9:644986. [PMID: 33718388 PMCID: PMC7952445 DOI: 10.3389/fcell.2021.644986] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/09/2021] [Indexed: 12/12/2022] Open
Abstract
During bone resorption, the osteoclast must sustain an extraordinarily low pH environment, withstand immense ionic pressures, and coordinate nutrient and waste exchange across its membrane to sustain its unique structural and functional polarity. To achieve this, osteoclasts are equipped with an elaborate set of membrane transport proteins (pumps, transporters and channels) that serve as molecular ‘gatekeepers’ to regulate the bilateral exchange of ions, amino acids, metabolites and macromolecules across the ruffled border and basolateral domains. Whereas the importance of the vacuolar-ATPase proton pump and chloride voltage-gated channel 7 in osteoclasts has long been established, comparatively little is known about the contributions of other membrane transport proteins, including those categorized as secondary active transporters. In this Special Issue review, we provide a contemporary update on the ‘ins and outs’ of membrane transport proteins implicated in osteoclast differentiation, function and bone homeostasis and discuss their therapeutic potential for the treatment of metabolic bone diseases.
Collapse
Affiliation(s)
- Amy B P Ribet
- Bone Biology and Disease Laboratory, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Pei Ying Ng
- Bone Biology and Disease Laboratory, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Nathan J Pavlos
- Bone Biology and Disease Laboratory, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
20
|
Molecular Genetics and Modifier Genes in Pseudoxanthoma Elasticum, a Heritable Multisystem Ectopic Mineralization Disorder. J Invest Dermatol 2020; 141:1148-1156. [PMID: 33341249 DOI: 10.1016/j.jid.2020.10.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/19/2020] [Accepted: 10/23/2020] [Indexed: 01/08/2023]
Abstract
In the past two decades, there has been great progress in identifying the molecular basis and pathomechanistic details in pseudoxanthoma elasticum (PXE), a heritable multisystem ectopic mineralization disorder. Although the identification of pathogenic variants in ABCC6 has been critical for understanding the disease process, genetic modifiers have been disclosed that explain the phenotypic heterogeneity of PXE. Adding to the genetic complexity of PXE are PXE-like phenotypes caused by pathogenic variants in other ectopic mineralization-associated genes. This review summarizes the current knowledge of the genetics and candidate modifier genes in PXE, a multifactorial disease at the genome-environment interface.
Collapse
|
21
|
Snyder-Keller A, Bolivar VJ, Zink S, Kramer LD. Brain Iron Accumulation and the Formation of Calcifications After Developmental Zika Virus Infection. J Neuropathol Exp Neurol 2020; 79:767-776. [PMID: 32483612 DOI: 10.1093/jnen/nlaa043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 04/24/2020] [Indexed: 12/20/2022] Open
Abstract
Intracranial calcifications (ICC) are the most common neuropathological finding in the brains of children exposed in utero to the Zika virus (ZIKV). Using a mouse model of developmental ZIKV infection, we reported widespread calcifications in the brains of susceptible mice that correlated in multiple ways with the behavioral deficits observed. Here, we examined the time course of ICC development and the role of iron deposition in this process, in 3 genetically distinct inbred strains of mice. Brain iron deposits were evident by Perls' staining at 2 weeks post infection, becoming increasingly dense and coinciding with calcium buildup and the formation of ICCs. A regional analysis of the brains of susceptible mice (C57BL/6J and 129S1/SvImJ strains) revealed the presence of iron initially in regions containing many ZIKV-immunoreactive cells, but then spreading to regions containing few infected cells, most notably the thalamus and the fasciculus retroflexus. Microglial activation was widespread initially and later delineated the sites of ICC formation. Behavioral tests conducted at 5-6 weeks of age revealed greater deficits in mice with the most extensive iron deposition and calcification of subcortical regions, such as thalamus. These findings point to iron deposition as a key factor in the development of ICCs after developmental ZIKV infection.
Collapse
Affiliation(s)
- Abigail Snyder-Keller
- Wadsworth Center, New York State Department of Health.,Department of Biomedical Sciences, University at Albany School of Public Health, Albany, New York
| | - Valerie J Bolivar
- Wadsworth Center, New York State Department of Health.,Department of Biomedical Sciences, University at Albany School of Public Health, Albany, New York
| | - Steven Zink
- Wadsworth Center, New York State Department of Health
| | - Laura D Kramer
- Wadsworth Center, New York State Department of Health.,Department of Biomedical Sciences, University at Albany School of Public Health, Albany, New York
| |
Collapse
|
22
|
Rutsch F, Buers I, Nitschke Y. Hereditary Disorders of Cardiovascular Calcification. Arterioscler Thromb Vasc Biol 2020; 41:35-47. [PMID: 33176451 DOI: 10.1161/atvbaha.120.315577] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Arterial calcification is a common phenomenon in the elderly, in patients with atherosclerosis or renal failure and in diabetes. However, when present in very young individuals, it is likely to be associated with an underlying hereditary disorder of arterial calcification. Here, we present an overview of the few monogenic disorders presenting with early-onset cardiovascular calcification. These disorders can be classified according to the function of the respective disease gene into (1) disorders caused by an altered purine and phosphate/pyrophosphate metabolism, (2) interferonopathies, and (3) Gaucher disease. The finding of arterial calcification in early life should alert the clinician and prompt further genetic work-up to define the underlying genetic defect, to establish the correct diagnosis, and to enable appropriate therapy.
Collapse
Affiliation(s)
- Frank Rutsch
- Department of General Pediatrics, Muenster University Children's Hospital, Germany
| | - Insa Buers
- Department of General Pediatrics, Muenster University Children's Hospital, Germany
| | - Yvonne Nitschke
- Department of General Pediatrics, Muenster University Children's Hospital, Germany
| |
Collapse
|
23
|
Zhuo MQ, Lv WH, Xu YH, Luo Z. Isolation and Characterization of Three Sodium-Phosphate Cotransporter Genes and Their Transcriptional Regulation in the Grass Carp Ctenopharyngodon idella. Int J Mol Sci 2020; 21:E8228. [PMID: 33153158 PMCID: PMC7662828 DOI: 10.3390/ijms21218228] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/31/2020] [Accepted: 11/02/2020] [Indexed: 12/20/2022] Open
Abstract
It is important to explore the regulatory mechanism of phosphorus homeostasis in fish, which help avoid the risk of P toxicity and prevent P pollution in aquatic environment. The present study obtained the full-length cDNA sequences and the promoters of three SLC20 members (slc20a1a, slc20a1b and slc20a2) from grass carp Ctenopharyngodon idella, and explored their responses to inorganic phosphorus (Pi). Grass carp SLC20s proteins possessed conservative domains and amino acid sites relevant with phosphorus transport. The mRNAs of three slc20s appeared in the nine tissues, but their expression levels were tissue-dependent. The binding sites of three transcription factors (SREBP1, NRF2 and VDR) were predicted on the slc20s promoters. The mutation and EMSA analysis indicated that: (1) SREBP1 binding site (-783/-771 bp) negatively but VDR (-260/-253 bp) binding site positively regulated the activities of slc20a1a promoter; (2) SREBP1 (-1187/-1178 bp), NRF2 (-572/-561 bp) and VDR(615/-609 bp) binding sites positively regulated the activities of slc20a1b promoter; (3) SREBP1 (-987/-977 bp), NRF2 (-1469/-1459 bp) and VDR (-1124/-1117 bp) binding sites positively regulated the activities of the slc20a2 promoter. Moreover, Pi incubation significantly reduced the activities of three slc20s promoters, and Pi-induced transcriptional inactivation of slc20s promoters abolished after the mutation of the VDR element but not SREBP1 and NRF2 elements. Pi incubation down-regulated the mRNA levels of three slc20s. For the first time, our study elucidated the transcriptional regulatory mechanisms of SLC20s and their responses to Pi, which offered new insights into the Pi homeostatic regulation and provided the basis for reducing phosphorus discharge into the waters.
Collapse
Affiliation(s)
- Mei-Qin Zhuo
- Laboratory of Molecular Nutrition for Aquatic Economic Animals, Fishery College, Huazhong Agricultural University, Wuhan 430070, China; (M.-Q.Z.); (W.-H.L.); (Y.-H.X.)
- School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Wu-Hong Lv
- Laboratory of Molecular Nutrition for Aquatic Economic Animals, Fishery College, Huazhong Agricultural University, Wuhan 430070, China; (M.-Q.Z.); (W.-H.L.); (Y.-H.X.)
| | - Yi-Huan Xu
- Laboratory of Molecular Nutrition for Aquatic Economic Animals, Fishery College, Huazhong Agricultural University, Wuhan 430070, China; (M.-Q.Z.); (W.-H.L.); (Y.-H.X.)
| | - Zhi Luo
- Laboratory of Molecular Nutrition for Aquatic Economic Animals, Fishery College, Huazhong Agricultural University, Wuhan 430070, China; (M.-Q.Z.); (W.-H.L.); (Y.-H.X.)
| |
Collapse
|
24
|
Machado A, Pouzolles M, Gailhac S, Fritz V, Craveiro M, López-Sánchez U, Kondo T, Pala F, Bosticardo M, Notarangelo LD, Petit V, Taylor N, Zimmermann VS. Phosphate Transporter Profiles in Murine and Human Thymi Identify Thymocytes at Distinct Stages of Differentiation. Front Immunol 2020; 11:1562. [PMID: 32793218 PMCID: PMC7387685 DOI: 10.3389/fimmu.2020.01562] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/15/2020] [Indexed: 12/22/2022] Open
Abstract
Thymocyte differentiation is dependent on the availability and transport of metabolites in the thymus niche. As expression of metabolite transporters is a rate-limiting step in nutrient utilization, cell surface transporter levels generally reflect the cell's metabolic state. The GLUT1 glucose transporter is upregulated on actively dividing thymocytes, identifying thymocytes with an increased metabolism. However, it is not clear whether transporters of essential elements such as phosphate are modulated during thymocyte differentiation. While PiT1 and PiT2 are both phosphate transporters in the SLC20 family, we show here that they exhibit distinct expression profiles on both murine and human thymocytes. PiT2 expression distinguishes thymocytes with high metabolic activity, identifying immature murine double negative (CD4−CD8−) DN3b and DN4 thymocyte blasts as well as immature single positive (ISP) CD8 thymocytes. Notably, the absence of PiT2 expression on RAG2-deficient thymocytes, blocked at the DN3a stage, strongly suggests that high PiT2 expression is restricted to thymocytes having undergone a productive TCRβ rearrangement at the DN3a/DN3b transition. Similarly, in the human thymus, PiT2 was upregulated on early post-β selection CD4+ISP and TCRαβ−CD4hiDP thymocytes co-expressing the CD71 transferrin receptor, a marker of metabolic activity. In marked contrast, expression of the PiT1 phosphate importer was detected on mature CD3+ murine and human thymocytes. Notably, PiT1 expression on CD3+DN thymocytes was identified as a biomarker of an aging thymus, increasing from 8.4 ± 1.5% to 42.4 ± 9.4% by 1 year of age (p < 0.0001). We identified these cells as TCRγδ and, most significantly, NKT, representing 77 ± 9% of PiT1+DN thymocytes by 1 year of age (p < 0.001). Thus, metabolic activity and thymic aging are associated with distinct expression profiles of the PiT1 and PiT2 phosphate transporters.
Collapse
Affiliation(s)
- Alice Machado
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States.,Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Marie Pouzolles
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Sarah Gailhac
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Vanessa Fritz
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Marco Craveiro
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Uriel López-Sánchez
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Taisuke Kondo
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Francesca Pala
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, United States
| | - Marita Bosticardo
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, United States
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, United States
| | | | - Naomi Taylor
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States.,Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Valérie S Zimmermann
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States.,Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| |
Collapse
|
25
|
Zhang Y, Zhang Y, Sun K, Meng Z, Chen L. The SLC transporter in nutrient and metabolic sensing, regulation, and drug development. J Mol Cell Biol 2020; 11:1-13. [PMID: 30239845 PMCID: PMC6359923 DOI: 10.1093/jmcb/mjy052] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 09/18/2018] [Indexed: 02/07/2023] Open
Abstract
The prevalence of metabolic diseases is growing worldwide. Accumulating evidence suggests that solute carrier (SLC) transporters contribute to the etiology of various metabolic diseases. Consistent with metabolic characteristics, the top five organs in which SLC transporters are highly expressed are the kidney, brain, liver, gut, and heart. We aim to understand the molecular mechanisms of important SLC transporter-mediated physiological processes and their potentials as drug targets. SLC transporters serve as ‘metabolic gate’ of cells and mediate the transport of a wide range of essential nutrients and metabolites such as glucose, amino acids, vitamins, neurotransmitters, and inorganic/metal ions. Gene-modified animal models have demonstrated that SLC transporters participate in many important physiological functions including nutrient supply, metabolic transformation, energy homeostasis, tissue development, oxidative stress, host defense, and neurological regulation. Furthermore, the human genomic studies have identified that SLC transporters are susceptible or causative genes in various diseases like cancer, metabolic disease, cardiovascular disease, immunological disorders, and neurological dysfunction. Importantly, a number of SLC transporters have been successfully targeted for drug developments. This review will focus on the current understanding of SLCs in regulating physiology, nutrient sensing and uptake, and risk of diseases.
Collapse
Affiliation(s)
- Yong Zhang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.,Advanced Biotechnology and Application Research Center, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China
| | - Yuping Zhang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Kun Sun
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Ziyi Meng
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Ligong Chen
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW In the last 7 years, changes in five genes [SLC20A2, PDGFRB, PDGFB, XPR1, and MYORG] have been implicated in the pathogenesis of primary familial brain calcification (PFBC), allowing for genetic delineation of this phenotypically complex neurodegenerative disorder. This review explores how the ensuing plethora of reported PFBC patients and their disease-causing variants improved our understanding of disease, pathogenesis, clinical manifestation, and penetrance. RECENT FINDINGS In PFBC patients, pathogenic changes have been most frequently described in SLC20A2, accounting for approximately the same number of patients as the variants in the other four PFBC genes combined. There is no appreciable relationship between any combination of the following three variables: the type of disease-causing change, the pattern or extent of calcifications, and the presence or nature of clinical manifestation in PFBC patients. Nevertheless, elucidation of underlying genetic factors provided important recent insights into the pathogenic mechanisms of PFBC, which collectively point toward a compromised neurovascular unit. SUMMARY The ongoing clinical and molecular research increases our understanding of PFBC facilitating diagnosis and identifying potential therapeutic targets for this multifaceted and likely underdiagnosed condition.
Collapse
|
27
|
Motta SE, Imenez Silva PH, Daryadel A, Haykir B, Pastor-Arroyo EM, Bettoni C, Hernando N, Wagner CA. Expression of NaPi-IIb in rodent and human kidney and upregulation in a model of chronic kidney disease. Pflugers Arch 2020; 472:449-460. [PMID: 32219532 DOI: 10.1007/s00424-020-02370-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 02/29/2020] [Accepted: 03/20/2020] [Indexed: 01/07/2023]
Abstract
Na+-coupled phosphate cotransporters from the SLC34 and SLC20 families of solute carriers mediate transepithelial transport of inorganic phosphate (Pi). NaPi-IIa/Slc34a1, NaPi-IIc/Slc34a3, and Pit-2/Slc20a2 are all expressed at the apical membrane of renal proximal tubules and therefore contribute to renal Pi reabsorption. Unlike NaPi-IIa and NaPi-IIc, which are rather kidney-specific, NaPi-IIb/Slc34a2 is expressed in several epithelial tissues, including the intestine, lung, testis, and mammary glands. Recently, the expression of NaPi-IIb was also reported in kidneys from rats fed on high Pi. Here, we systematically quantified the mRNA expression of SLC34 and SLC20 cotransporters in kidneys from mice, rats, and humans. In all three species, NaPi-IIa mRNA was by far the most abundant renal transcript. Low and comparable mRNA levels of the other four transporters, including NaPi-IIb, were detected in kidneys from rodents and humans. In mice, the renal expression of NaPi-IIa transcripts was restricted to the cortex, whereas NaPi-IIb mRNA was observed in medullary segments. Consistently, NaPi-IIb protein colocalized with uromodulin at the luminal membrane of thick ascending limbs of the loop of Henle segments. The abundance of NaPi-IIb transcripts in kidneys from mice was neither affected by dietary Pi, the absence of renal NaPi-IIc, nor the depletion of intestinal NaPi-IIb. In contrast, it was highly upregulated in a model of oxalate-induced kidney disease where all other SLC34 phosphate transporters were downregulated. Thus, NaPi-IIb may contribute to renal phosphate reabsorption, and its upregulation in kidney disease might promote hyperphosphatemia.
Collapse
Affiliation(s)
- Sarah E Motta
- Institute of Physiology, Switzerland and National Center of Competence in Research NCCR Kidney, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Pedro Henrique Imenez Silva
- Institute of Physiology, Switzerland and National Center of Competence in Research NCCR Kidney, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Arezoo Daryadel
- Institute of Physiology, Switzerland and National Center of Competence in Research NCCR Kidney, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Betül Haykir
- Institute of Physiology, Switzerland and National Center of Competence in Research NCCR Kidney, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Eva Maria Pastor-Arroyo
- Institute of Physiology, Switzerland and National Center of Competence in Research NCCR Kidney, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Carla Bettoni
- Institute of Physiology, Switzerland and National Center of Competence in Research NCCR Kidney, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Nati Hernando
- Institute of Physiology, Switzerland and National Center of Competence in Research NCCR Kidney, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Carsten A Wagner
- Institute of Physiology, Switzerland and National Center of Competence in Research NCCR Kidney, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland.
| |
Collapse
|
28
|
Villa-Bellosta R. New insights into endogenous mechanisms of protection against arterial calcification. Atherosclerosis 2020; 306:68-74. [PMID: 32209233 DOI: 10.1016/j.atherosclerosis.2020.03.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 02/24/2020] [Accepted: 03/11/2020] [Indexed: 12/31/2022]
Abstract
Cardiovascular complications due to accelerated atherosclerosis and arterial stiffening are the leading cause of morbidity and mortality in the Western society. Both pathologies are frequently associated with vascular calcification. Deposits of calcium phosphate salts, mainly in form of hydroxyapatite, is the hallmark of vascular calcification. Calcification is frequently observed in atherosclerotic lesions (intimal calcification) associated with vascular smooth muscle cells (VSMCs) and macrophages. By contrast, medial calcification, occurring in the elastic region of the arteries, is almost exclusively associated with VSMCs, and is common in arteriosclerosis related to aging, diabetes, and chronic kidney disease. In extracellular fluids, a range of endogenous low- and high-molecular weight calcification inhibitors are present, including osteopontin, matrix-Gla proteins and Fetuin A. Moreover, pyrophosphate deficiency plays a key role in vascular calcification. Pyrophosphate is produced by extracellular hydrolysis of ATP and is degraded to phosphate by tissue non-specific alkaline phosphatase. Loss of function in the enzymes and transporters involved in the extracellular pyrophosphate metabolism leads to excessive deposition of calcium-phosphate salts. This review summarizes the current knowledge about endogenous mechanisms of protection against calcification in the aortic wall, focusing on the role of extracellular pyrophosphate metabolism in vascular smooth muscle cells and macrophages.
Collapse
Affiliation(s)
- Ricardo Villa-Bellosta
- Fundación Instituto de Investigación Sanitaria, Fundación Jiménez Díaz (FIIS-FJD), Avenida Reyes Católicos 2, 28040, Madrid, Spain.
| |
Collapse
|
29
|
Chande S, Caballero D, Ho BB, Fetene J, Serna J, Pesta D, Nasiri A, Jurczak M, Chavkin NW, Hernando N, Giachelli CM, Wagner CA, Zeiss C, Shulman GI, Bergwitz C. Slc20a1/Pit1 and Slc20a2/Pit2 are essential for normal skeletal myofiber function and survival. Sci Rep 2020; 10:3069. [PMID: 32080237 PMCID: PMC7033257 DOI: 10.1038/s41598-020-59430-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 01/29/2020] [Indexed: 01/25/2023] Open
Abstract
Low blood phosphate (Pi) reduces muscle function in hypophosphatemic disorders. Which Pi transporters are required and whether hormonal changes due to hypophosphatemia contribute to muscle function is unknown. To address these questions we generated a series of conditional knockout mice lacking one or both house-keeping Pi transporters Pit1 and Pit2 in skeletal muscle (sm), using the postnatally expressed human skeletal actin-cre. Simultaneous conditional deletion of both transporters caused skeletal muscle atrophy, resulting in death by postnatal day P13. smPit1-/-, smPit2-/- and three allele mutants are fertile and have normal body weights, suggesting a high degree of redundance for the two transporters in skeletal muscle. However, these mice show a gene-dose dependent reduction in running activity also seen in another hypophosphatemic model (Hyp mice). In contrast to Hyp mice, grip strength is preserved. Further evaluation of the mechanism shows reduced ERK1/2 activation and stimulation of AMP kinase in skeletal muscle from smPit1-/-; smPit2-/- mice consistent with energy-stress. Similarly, C2C12 myoblasts show a reduced oxygen consumption rate mediated by Pi transport-dependent and ERK1/2-dependent metabolic Pi sensing pathways. In conclusion, we here show that Pit1 and Pit2 are essential for normal myofiber function and survival, insights which may improve management of hypophosphatemic myopathy.
Collapse
Affiliation(s)
- Sampada Chande
- Department of Internal Medicine, Section Endocrinology, Yale University School of Medicine, New Haven, CT, USA
| | - Daniel Caballero
- Department of Internal Medicine, Section Endocrinology, Yale University School of Medicine, New Haven, CT, USA
| | - Bryan B Ho
- Department of Internal Medicine, Section Endocrinology, Yale University School of Medicine, New Haven, CT, USA
| | - Jonathan Fetene
- Department of Internal Medicine, Section Endocrinology, Yale University School of Medicine, New Haven, CT, USA
| | - Juan Serna
- Department of Internal Medicine, Section Endocrinology, Yale University School of Medicine, New Haven, CT, USA
| | - Dominik Pesta
- Department of Cellular&Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
- German Diabetes Center, Düsseldorf, Germany, University of Washington, Box 355061, Foege Hall Seattle, WA, 98195, USA
| | - Ali Nasiri
- Department of Cellular&Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Michael Jurczak
- Department of Internal Medicine, Section Endocrinology, Yale University School of Medicine, New Haven, CT, USA
- Department of Medicine, Division of Endocrinology, University of Pittsburgh, University of Washington, Box 355061, Foege Hall Seattle, WA, 98195, USA
| | - Nicholas W Chavkin
- Department of Bioengineering, University of Washington, Box 355061, Foege Hall Seattle, WA, 98195, USA
| | - Nati Hernando
- Institute of Physiology, University of Zürich, Switzerland and National Center of Competence in Research NCCR Kidney.CH, Zürich, Switzerland
| | - Cecilia M Giachelli
- Department of Bioengineering, University of Washington, Box 355061, Foege Hall Seattle, WA, 98195, USA
| | - Carsten A Wagner
- Institute of Physiology, University of Zürich, Switzerland and National Center of Competence in Research NCCR Kidney.CH, Zürich, Switzerland
| | - Caroline Zeiss
- Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Gerald I Shulman
- Department of Internal Medicine, Section Endocrinology, Yale University School of Medicine, New Haven, CT, USA
- Department of Cellular&Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Clemens Bergwitz
- Department of Internal Medicine, Section Endocrinology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
30
|
Primary familial brain calcification presenting as paroxysmal kinesigenic dyskinesia: Genetic and functional analyses. Neurosci Lett 2020; 714:134543. [DOI: 10.1016/j.neulet.2019.134543] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 10/03/2019] [Accepted: 10/09/2019] [Indexed: 12/27/2022]
|
31
|
Nahar K, Lebouvier T, Andaloussi Mäe M, Konzer A, Bergquist J, Zarb Y, Johansson B, Betsholtz C, Vanlandewijck M. Astrocyte-microglial association and matrix composition are common events in the natural history of primary familial brain calcification. Brain Pathol 2019; 30:446-464. [PMID: 31561281 PMCID: PMC7317599 DOI: 10.1111/bpa.12787] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/21/2019] [Indexed: 02/06/2023] Open
Abstract
Primary familial brain calcification (PFBC) is an age-dependent and rare neurodegenerative disorder characterized by microvascular calcium phosphate deposits in the deep brain regions. Known genetic causes of PFBC include loss-of-function mutations in genes involved in either of three processes-platelet-derived growth factor (PDGF) signaling, phosphate homeostasis or protein glycosylation-with unclear molecular links. To provide insight into the pathogenesis of PFBC, we analyzed murine models of PFBC for the first two of these processes in Pdgfbret/ret and Slc20a2-/- mice with regard to the structure, molecular composition, development and distribution of perivascular calcified nodules. Analyses by transmission electron microscopy and immunofluorescence revealed that calcified nodules in both of these models have a multilayered ultrastructure and occur in direct contact with reactive astrocytes and microglia. However, whereas nodules in Pdgfbret/ret mice were large, solitary and smooth surfaced, the nodules in Slc20a2-/- mice were multi-lobulated and occurred in clusters. The regional distribution of nodules also differed between the two models. Proteomic analysis and immunofluorescence stainings revealed a common molecular composition of the nodules in the two models, involving proteins implicated in bone homeostasis, but also proteins not previously linked to tissue mineralization. While the brain vasculature of Pdgfbret/ret mice has been reported to display reduced pericyte coverage and abnormal permeability, we found that Slc20a2-/- mice have a normal pericyte coverage and no overtly increased permeability. Thus, lack of pericytes and increase in permeability of the blood-brain barrier are likely not the causal triggers for PFBC pathogenesis. Instead, gene expression and spatial correlations suggest that astrocytes are intimately linked to the calcification process in PFBC.
Collapse
Affiliation(s)
- Khayrun Nahar
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Thibaud Lebouvier
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Department of Neurology, CHRU Lille, Lille, France.,Inserm U1171, Lille, France
| | - Maarja Andaloussi Mäe
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Anne Konzer
- Scientific Service Group Mass Spectrometry, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Jonas Bergquist
- Department of Chemistry, Biomedical Centre, Uppsala University, Uppsala, Sweden
| | - Yvette Zarb
- Department of Neurosurgery, Clinical Neuroscience Center, Zurich University Hospital, Zurich University, Zurich, Switzerland.,Neuroscience Center Zurich (ZNZ), University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Bengt Johansson
- Electron Microscopy Unit, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Integrated Cardio Metabolic Centre (ICMC), Karolinska Institute, Huddinge, Sweden
| | - Michael Vanlandewijck
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Integrated Cardio Metabolic Centre (ICMC), Karolinska Institute, Huddinge, Sweden
| |
Collapse
|
32
|
Giorgio E, Garelli E, Carando A, Bellora S, Rubino E, Quarello P, Sirchia F, Marrama F, Gallone S, Grosso E, Pasini B, Massa R, Brussino A, Brusco A. Design of a multiplex ligation-dependent probe amplification assay for SLC20A2: identification of two novel deletions in primary familial brain calcification. J Hum Genet 2019; 64:1083-1090. [PMID: 31501477 DOI: 10.1038/s10038-019-0668-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 08/16/2019] [Accepted: 08/19/2019] [Indexed: 01/13/2023]
Abstract
Primary familial brain calcification (PFBC) is a rare disease characterized by brain calcifications that mainly affect the basal ganglia, thalamus, and cerebellum. Among the four autosomal-dominant genes known to be associated with the disease, SLC20A2 pathogenic variants are the most common, accounting for up to 40% of PFBC dominant cases; variants include both point mutations, small insertions/deletions and intragenic deletions. Over the last 7 years, we have collected a group of 50 clinically diagnosed PFBC patients, who were screened for single nucleotide changes and small insertions/deletions in SLC20A2 by Sanger sequencing. We found seven pathogenic/likely pathogenic variants: four were previously described by our group, and three are reported here (c.303delG, c.21delG, and c.1795-1G>A). We developed and validated a synthetic Multiplex Ligation-dependent Probe Amplification (MLPA) assay for SLC20A2 deletions, covering all ten coding exons and the 5' UTR (SLC20A2-MLPA). Using this method, we screened a group of 43 PFBC-patients negative for point mutations and small insertions/deletions, and identified two novel intragenic deletions encompassing exon 6 NC_000008.10:g.(42297172_42302163)_(423022281_42317413)del, and exons 7-11 including the 3'UTR NC_000008.10:g.(?_42275320)_(42297172_42302163)del. Overall, SLC20A2 deletions may be highly underestimated PFBC cases, and we suggest MLPA should be included in the routine molecular test for PFBC diagnosis.
Collapse
Affiliation(s)
- Elisa Giorgio
- Department of Medical Sciences, University of Torino, Turin, Italy
| | - Emanuela Garelli
- Department of Public Health and Pediatric Sciences, University of Torino, Turin, Italy
| | - Adriana Carando
- Department of Public Health and Pediatric Sciences, University of Torino, Turin, Italy
| | - Stefania Bellora
- Pediatric Neuropsychiatry Unit, "SS Antonio e Biagio e Cesare Arrigo" Hospital, Alessandria, Italy
| | - Elisa Rubino
- Department of Neuroscience and Mental Health, AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | - Paola Quarello
- Department of Public Health and Pediatric Sciences, University of Torino, Turin, Italy
| | - Fabio Sirchia
- Institute for Maternal and Child Health IRCCS Burlo Garofalo, Trieste, Italy
| | - Federico Marrama
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Salvatore Gallone
- Department of Neuroscience "Rita Levi Montalcini", University of Torino, Turin, Italy
| | - Enrico Grosso
- Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Turin, Italy
| | - Barbara Pasini
- Department of Medical Sciences, University of Torino, Turin, Italy.,Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Turin, Italy
| | - Roberto Massa
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | | | - Alfredo Brusco
- Department of Medical Sciences, University of Torino, Turin, Italy. .,Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Turin, Italy.
| |
Collapse
|
33
|
Huang YT, Zhang LH, Li MF, Cheng L, Zou GY, Zhou HH. A splice site mutation causing exon 6 skipping in SLC20A2 gene in a primary familial brain calcification family. Brain Res Bull 2019; 150:261-265. [PMID: 30634018 DOI: 10.1016/j.brainresbull.2019.01.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 12/24/2018] [Accepted: 01/03/2019] [Indexed: 12/17/2022]
|
34
|
Castelblanco M, Nasi S, Pasch A, So A, Busso N. The role of the gasotransmitter hydrogen sulfide in pathological calcification. Br J Pharmacol 2019; 177:778-792. [PMID: 31231793 DOI: 10.1111/bph.14772] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/29/2019] [Accepted: 06/05/2019] [Indexed: 12/11/2022] Open
Abstract
Calcification is the deposition of minerals, mainly hydroxyapatite, inside the cell or in the extracellular matrix. Physiological calcification is central for many aspects of development including skeletal and tooth growth; conversely, pathological mineralization occurs in soft tissues and is significantly associated with malfunction and impairment of the tissue where it is located. Various mechanisms have been proposed to explain calcification. However, this research area lacks a more integrative, systemic, and global perspective that could explain both physiological and pathological processes. In this review, we propose such an integrated explanation. Hydrogen sulfide (H2 S) is a newly recognized multifunctional gasotransmitters and tis actions have been studied in different physiological and pathological contexts, but little is known about its potential role on calcification. Interestingly, we found that H2 S promotes calcification under physiological conditions and has an inhibitory effect on pathological processes. This makes H2 S a potential therapy for diseases related to pathological calcification. LINKED ARTICLES: This article is part of a themed section on Hydrogen Sulfide in Biology & Medicine. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.4/issuetoc.
Collapse
Affiliation(s)
- Mariela Castelblanco
- Service of Rheumatology, DAL, Lausanne University Hospital (CHUV), University of Lausanne, Lausanne, Switzerland
| | - Sonia Nasi
- Service of Rheumatology, DAL, Lausanne University Hospital (CHUV), University of Lausanne, Lausanne, Switzerland
| | | | - Alexander So
- Service of Rheumatology, DAL, Lausanne University Hospital (CHUV), University of Lausanne, Lausanne, Switzerland
| | - Nathalie Busso
- Service of Rheumatology, DAL, Lausanne University Hospital (CHUV), University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
35
|
Quantitative Trait Locus and Integrative Genomics Revealed Candidate Modifier Genes for Ectopic Mineralization in Mouse Models of Pseudoxanthoma Elasticum. J Invest Dermatol 2019; 139:2447-2457.e7. [PMID: 31207231 DOI: 10.1016/j.jid.2019.04.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/28/2019] [Accepted: 04/26/2019] [Indexed: 02/06/2023]
Abstract
Pseudoxanthoma elasticum, a prototype of heritable multisystem ectopic mineralization disorders, is caused by mutations in the ABCC6 gene encoding a putative efflux transporter, ABCC6. The phenotypic spectrum of pseudoxanthoma elasticum varies, and the correlation between genotype and phenotype has not been established. To identify genetic modifiers, we performed quantitative trait locus analysis in inbred mouse strains that carry the same hypomorphic allele in Abcc6 yet with highly variable ectopic mineralization phenotypes of pseudoxanthoma elasticum. Abcc6 was confirmed as a major determinant for ectopic mineralization in multiple tissues. Integrative analysis using functional genomics tools that included GeneWeaver, String, and Mouse Genome Informatics identified a total of nine additional candidate modifier genes that could influence the organ-specific ectopic mineralization phenotypes. Integration of the candidate genes into the existing ectopic mineralization gene network expands the current knowledge on the complexity of the network that, as a whole, governs ectopic mineralization in soft connective tissues.
Collapse
|
36
|
Characterization of XPR1/SLC53A1 variants located outside of the SPX domain in patients with primary familial brain calcification. Sci Rep 2019; 9:6776. [PMID: 31043717 PMCID: PMC6494797 DOI: 10.1038/s41598-019-43255-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 04/17/2019] [Indexed: 12/17/2022] Open
Abstract
Primary familial brain calcification (PFBC) is a rare neurological disease characterized by deposits of calcium phosphate in the basal ganglia and other regions of the brain. Pathogenic variants in the XPR1/SLC53A1 gene, which encodes the only known inorganic phosphate exporter, cause an autosomal dominant form of PFBC. These variants are typically located in the SPX N-terminal domain of the protein. Here, we characterize three XPR1 variants outside of SPX in three PFBC patients with an apparently sporadic presentation: c.1375C > T p.(R459C), c.1855A > G p.(N619D) and c.1886T > G p.(I629S), with the latter identified as the first XPR1/SLC53A1 de novo mutation to occur in a PFBC proband. When tested in an in vitro physiological complementation assay, the three XPR1 variants were impaired in phosphate export function, although they were normally expressed at the cell surface and could serve as functional receptors for retrovirus entry. Moreover, peripheral blood cells from the p.N619D patient could be assayed ex vivo and displayed significantly impaired phosphate export. Our results establish for the first time the clinical and molecular characteristics of XPR1 variants located outside the SPX domain and assert a direct link between these variants, deficient phosphate export, and PFBC. Moreover, we unveiled new structural features in XPR1 C-terminal domain that play a role in phosphate export and disease.
Collapse
|
37
|
Guo X, Su H, Zou X, Lai L, Lu Y, Wang C, Li Y, Hong J, Zhao M, Lin K, Lin J, Zeng Y, Yao X, Wang N, Chen W. Identification of
SLC20A2
deletions in patients with primary familial brain calcification. Clin Genet 2019; 96:53-60. [PMID: 30891739 DOI: 10.1111/cge.13540] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 02/20/2019] [Accepted: 03/13/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Xin‐Xin Guo
- Department of Neurology and Institute of NeurologyThe First Affiliated Hospital of Fujian Medical University Fuzhou China
| | - Hui‐Zhen Su
- Department of Neurology and Institute of NeurologyThe First Affiliated Hospital of Fujian Medical University Fuzhou China
| | - Xiao‐Huan Zou
- Department of Neurology and Institute of NeurologyThe First Affiliated Hospital of Fujian Medical University Fuzhou China
| | - Lu‐Lu Lai
- Department of Neurology and Institute of NeurologyThe First Affiliated Hospital of Fujian Medical University Fuzhou China
| | - Ying‐Qian Lu
- Department of Neurology and Institute of NeurologyThe First Affiliated Hospital of Fujian Medical University Fuzhou China
| | - Chong Wang
- Department of Neurology and Institute of NeurologyThe First Affiliated Hospital of Fujian Medical University Fuzhou China
| | - Yun‐Lu Li
- Department of Neurology and Institute of NeurologyThe First Affiliated Hospital of Fujian Medical University Fuzhou China
| | - Jing‐Mei Hong
- Department of Neurology and Institute of NeurologyThe First Affiliated Hospital of Fujian Medical University Fuzhou China
| | - Miao Zhao
- Department of Neurology and Institute of NeurologyThe First Affiliated Hospital of Fujian Medical University Fuzhou China
| | - Kun‐Xin Lin
- Department of Neurology and Institute of NeurologyThe First Affiliated Hospital of Fujian Medical University Fuzhou China
| | - Jie Lin
- Department of Neurology and Institute of NeurologyThe First Affiliated Hospital of Fujian Medical University Fuzhou China
| | - Yi‐Heng Zeng
- Department of Neurology and Institute of NeurologyThe First Affiliated Hospital of Fujian Medical University Fuzhou China
| | - Xiang‐Ping Yao
- Department of Neurology and Institute of NeurologyThe First Affiliated Hospital of Fujian Medical University Fuzhou China
| | - Ning Wang
- Department of Neurology and Institute of NeurologyThe First Affiliated Hospital of Fujian Medical University Fuzhou China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University Fuzhou China
| | - Wan‐Jin Chen
- Department of Neurology and Institute of NeurologyThe First Affiliated Hospital of Fujian Medical University Fuzhou China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University Fuzhou China
| |
Collapse
|
38
|
Lack of Major Ophthalmic Findings in Patients with Primary Familial Brain Calcification Linked to SLC20A2 and PDGFB. J Mol Neurosci 2019; 67:441-444. [PMID: 30607898 DOI: 10.1007/s12031-018-1250-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 12/21/2018] [Indexed: 12/13/2022]
Abstract
Primary familial brain calcification (PFBC) is a rare neurodegenerative disorder characterized by symmetrical and bilateral brain calcification. It is typically inherited as an autosomal dominant disorder, and de novo variants have also been described. Interestingly, just recent studies have reported the first autosomal recessive PFBC-causative gene. PFBC patients exhibit high clinical heterogeneity including Parkinsonism, dystonia, ataxia, depression, and migraine. Mice studies, an important research tool, have been a breakthrough in increasing the understanding of PFBC's main signs and symptoms, and many findings reported in these mice have been subsequently reported in patients. One phenotype that has been observed in PFBC mice models but not in PFBC patients, however, is the development of ophthalmic abnormalities. This way, this report focused on performing an ophthalmic assessment in six Brazilian patients genetically diagnosed with PFBC. The assessments showed that none of the PFBC individuals included presented any of the ophthalmic abnormalities reported in mice models, such as cataracts, ocular calcification, abnormal iris and lens morphology, and retinal deterioration. Additionally, of the six PFBC patients described, two SLC20A2 mutation carriers showed physiological excavation of the optic nerve head and partial vitreous detachment, while just one individual presented bilateral narrowing of retinal arterioles. In summary, no evidence of similar ophthalmological abnormalities found in mice were found in our patients; nonetheless, further studies in larger sample size are warranted to corroborate with our findings. To our knowledge, this study is the first to focus on investigating, in PFBC patients, the ophthalmological phenotypes described in the PFBC mice models.
Collapse
|
39
|
Pericytes in Primary Familial Brain Calcification. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1147:247-264. [PMID: 31147881 DOI: 10.1007/978-3-030-16908-4_11] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pericytes are perivascular cells along capillaries that are critical for the development of a functional vascular bed in the central nervous system and other organs. Pericyte functions in the adult brain are less well understood. Pericytes have been suggested to mediate functional hyperemia at the capillary level, regulate the blood-brain barrier and to give rise to scar tissue after spinal cord injury. Furthermore, pericyte loss has been suggested to precede cognitive decline in mouse models of Alzheimer's disease. Despite this observation, there is no convincing causality between pericyte loss and the pathogenesis of Alzheimer's disease. However, recent loss-of-function mutations in PDGFB and PDGFRB genes have implicated pericytes as the principle cell type affected in primary familiar brain calcification (PFBC), a neuropsychiatric disorder with dominant inheritance. Here we review the role of the PDGFB/PDGFRB signaling pathway in pericyte development and briefly discuss homeostatic functions of pericytes in the brain. We provide an overview of recent studies with mouse models of PFBC and discuss suggested pathogenic mechanisms for PFBC with special reference to pericytes.
Collapse
|
40
|
Michigami T, Kawai M, Yamazaki M, Ozono K. Phosphate as a Signaling Molecule and Its Sensing Mechanism. Physiol Rev 2018; 98:2317-2348. [DOI: 10.1152/physrev.00022.2017] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
In mammals, phosphate balance is maintained by influx and efflux via the intestines, kidneys, bone, and soft tissue, which involves multiple sodium/phosphate (Na+/Pi) cotransporters, as well as regulation by several hormones. Alterations in the levels of extracellular phosphate exert effects on both skeletal and extra-skeletal tissues, and accumulating evidence has suggested that phosphate itself evokes signal transduction to regulate gene expression and cell behavior. Several in vitro studies have demonstrated that an elevation in extracellular Piactivates fibroblast growth factor receptor, Raf/MEK (mitogen-activated protein kinase/ERK kinase)/ERK (extracellular signal-regulated kinase) pathway and Akt pathway, which might involve the type III Na+/Picotransporter PiT-1. Excessive phosphate loading can lead to various harmful effects by accelerating ectopic calcification, enhancing oxidative stress, and dysregulating signal transduction. The responsiveness of mammalian cells to altered extracellular phosphate levels suggests that they may sense and adapt to phosphate availability, although the precise mechanism for phosphate sensing in mammals remains unclear. Unicellular organisms, such as bacteria and yeast, use some types of Pitransporters and other molecules, such as kinases, to sense the environmental Piavailability. Multicellular animals may need to integrate signals from various organs to sense the phosphate levels as a whole organism, similarly to higher plants. Clarification of the phosphate-sensing mechanism in humans may lead to the development of new therapeutic strategies to prevent and treat diseases caused by phosphate imbalance.
Collapse
Affiliation(s)
- Toshimi Michigami
- Department of Bone and Mineral Research, Research Institute, Osaka Women’s and Children’s Hospital, Osaka Prefectural Hospital Organization, Izumi, Osaka, Japan; and Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Masanobu Kawai
- Department of Bone and Mineral Research, Research Institute, Osaka Women’s and Children’s Hospital, Osaka Prefectural Hospital Organization, Izumi, Osaka, Japan; and Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Miwa Yamazaki
- Department of Bone and Mineral Research, Research Institute, Osaka Women’s and Children’s Hospital, Osaka Prefectural Hospital Organization, Izumi, Osaka, Japan; and Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Keiichi Ozono
- Department of Bone and Mineral Research, Research Institute, Osaka Women’s and Children’s Hospital, Osaka Prefectural Hospital Organization, Izumi, Osaka, Japan; and Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
41
|
Yamada S, Leaf EM, Chia JJ, Cox TC, Speer MY, Giachelli CM. PiT-2, a type III sodium-dependent phosphate transporter, protects against vascular calcification in mice with chronic kidney disease fed a high-phosphate diet. Kidney Int 2018; 94:716-727. [PMID: 30041812 PMCID: PMC6211801 DOI: 10.1016/j.kint.2018.05.015] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 05/08/2018] [Accepted: 05/10/2018] [Indexed: 12/11/2022]
Abstract
PiT-2, a type III sodium-dependent phosphate transporter, is a causative gene for the brain arteriolar calcification in people with familial basal ganglion calcification. Here we examined the effect of PiT-2 haploinsufficiency on vascular calcification in uremic mice using wild-type and global PiT-2 heterozygous knockout mice. PiT-2 haploinsufficiency enhanced the development of vascular calcification in mice with chronic kidney disease fed a high-phosphate diet. No differences were observed in the serum mineral biomarkers and kidney function between the wild-type and PiT-2 heterozygous knockout groups. Micro computed tomography analyses of femurs showed that haploinsufficiency of PiT-2 decreased trabecular bone mineral density in uremia. In vitro, sodium-dependent phosphate uptake was decreased in cultured vascular smooth muscle cells isolated from PiT-2 heterozygous knockout mice compared with those from wild-type mice. PiT-2 haploinsufficiency increased phosphate-induced calcification of cultured vascular smooth muscle cells compared to the wild-type. Furthermore, compared to wild-type vascular smooth muscle cells, PiT-2 deficient vascular smooth muscle cells had lower osteoprotegerin levels and increased matrix calcification, which was attenuated by osteoprotegerin supplementation. Thus, PiT-2 in vascular smooth muscle cells protects against phosphate-induced vascular calcification and may be a therapeutic target in the chronic kidney disease population.
Collapse
Affiliation(s)
- Shunsuke Yamada
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| | - Elizabeth M Leaf
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| | - Jia Jun Chia
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| | - Timothy C Cox
- Department of Pediatrics, University of Washington, Seattle, Washington, USA; Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Mei Y Speer
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| | - Cecilia M Giachelli
- Department of Bioengineering, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
42
|
Ramos EM, Carecchio M, Lemos R, Ferreira J, Legati A, Sears RL, Hsu SC, Panteghini C, Magistrelli L, Salsano E, Esposito S, Taroni F, Richard AC, Tranchant C, Anheim M, Ayrignac X, Goizet C, Vidailhet M, Maltete D, Wallon D, Frebourg T, Pimentel L, Geschwind DH, Vanakker O, Galasko D, Fogel BL, Innes AM, Ross A, Dobyns WB, Alcantara D, O'Driscoll M, Hannequin D, Campion D, Oliveira JR, Garavaglia B, Coppola G, Nicolas G. Primary brain calcification: an international study reporting novel variants and associated phenotypes. Eur J Hum Genet 2018; 26:1462-1477. [PMID: 29955172 PMCID: PMC6138755 DOI: 10.1038/s41431-018-0185-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 03/21/2018] [Accepted: 05/08/2018] [Indexed: 12/17/2022] Open
Abstract
Primary familial brain calcification (PFBC) is a rare cerebral microvascular calcifying disorder with a wide spectrum of motor, cognitive, and neuropsychiatric symptoms. It is typically inherited as an autosomal-dominant trait with four causative genes identified so far: SLC20A2, PDGFRB, PDGFB, and XPR1. Our study aimed at screening the coding regions of these genes in a series of 177 unrelated probands that fulfilled the diagnostic criteria for primary brain calcification regardless of their family history. Sequence variants were classified as pathogenic, likely pathogenic, or of uncertain significance (VUS), based on the ACMG-AMP recommendations. We identified 45 probands (25.4%) carrying either pathogenic or likely pathogenic variants (n = 34, 19.2%) or VUS (n = 11, 6.2%). SLC20A2 provided the highest contribution (16.9%), followed by XPR1 and PDGFB (3.4% each), and PDGFRB (1.7%). A total of 81.5% of carriers were symptomatic and the most recurrent symptoms were parkinsonism, cognitive impairment, and psychiatric disturbances (52.3%, 40.9%, and 38.6% of symptomatic individuals, respectively), with a wide range of age at onset (from childhood to 81 years). While the pathogenic and likely pathogenic variants identified in this study can be used for genetic counseling, the VUS will require additional evidence, such as recurrence in unrelated patients, in order to be classified as pathogenic.
Collapse
Affiliation(s)
- Eliana Marisa Ramos
- Department of Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Miryam Carecchio
- Molecular Neurogenetics Unit, Movement Disorders Section, IRCCS Foundation Carlo Besta Neurological Institute, Via L. Temolo n. 4, Milan, 20116, Italy
- Department of Pediatric Neurology, IRCCS Foundation Carlo Besta Neurological Institute, Via Celoria 11, Milan, 20131, Italy
- PhD Programme in Translational and Molecular Medicine, Milan Bicocca University, Monza, Italy
| | - Roberta Lemos
- Keizo Asami Laboratory, Universidade Federal de Pernambuco, Recife, Brazil
| | - Joana Ferreira
- Keizo Asami Laboratory, Universidade Federal de Pernambuco, Recife, Brazil
| | - Andrea Legati
- Department of Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Renee Louise Sears
- Department of Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Sandy Chan Hsu
- Department of Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Celeste Panteghini
- Molecular Neurogenetics Unit, Movement Disorders Section, IRCCS Foundation Carlo Besta Neurological Institute, Via L. Temolo n. 4, Milan, 20116, Italy
| | - Luca Magistrelli
- Department of Neurology, University of Eastern Piedmont, C.so Mazzini 18, Novara, 28100, Italy
| | - Ettore Salsano
- Department of Clinical Neurosciences, IRCCS Foundation Carlo Besta Neurological Institute, Via Celoria 11, Milan, 20131, Italy
| | - Silvia Esposito
- Department of Pediatric Neurology, IRCCS Foundation Carlo Besta Neurological Institute, Via Celoria 11, Milan, 20131, Italy
| | - Franco Taroni
- IRCCS Foundation Carlo Besta Neurological Institute, Via Amadeo 42, Milan, 20133, Italy
| | - Anne-Claire Richard
- Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, Department of Genetics and CNR-MAJ, F 76000, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | - Christine Tranchant
- Service de Neurologie, Hôpitaux Universitaires de Strasbourg, Hôpital de Hautepierre; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Strasbourg, Illkirch, France
| | - Mathieu Anheim
- Service de Neurologie, Hôpitaux Universitaires de Strasbourg, Hôpital de Hautepierre; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Strasbourg, Illkirch, France
| | - Xavier Ayrignac
- Department of Neurology, Montpellier University Hospital, Montpellier, France
| | - Cyril Goizet
- CHU Bordeaux, Service de Génétique Médicale, 33000, Bordeaux, France
- INSERM U1211, Univ Bordeaux, Laboratoire Maladies Rares, Génétique et Métabolisme, 33000, Bordeaux, France
| | - Marie Vidailhet
- Département de neurologie, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, UPMC Univ Paris 06, Inserm U1127, CNRS UMR 7225, ICM, F-75013, Sorbonne Universites, Paris, France
| | - David Maltete
- Normandie Univ, UNIROUEN, Inserm U1073, Rouen University Hospital, Department of Neurology, F 76000, Rouen, France
| | - David Wallon
- Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, Department of Neurology and CNR-MAJ, F 76000, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | - Thierry Frebourg
- Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, Department of Genetics and CNR-MAJ, F 76000, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | - Lylyan Pimentel
- Keizo Asami Laboratory, Universidade Federal de Pernambuco, Recife, Brazil
| | - Daniel H Geschwind
- Department of Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Olivier Vanakker
- Center for Medical Genetics, Ghent University Hospital, De Pintelaan 185, B-9000, Ghent, Belgium
| | - Douglas Galasko
- Veterans Affairs Medical Center, San Diego and University of California, San Diego, USA
| | - Brent L Fogel
- Departments of Neurology and Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - A Micheil Innes
- Department of Medical Genetics and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Alison Ross
- Department of Clinical Genetics, Ashgrove House, Foresterhill, Aberdeen, UK
| | - William B Dobyns
- Departments of Pediatrics and Neurology, University of Washington; and Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Diana Alcantara
- Genome Damage & Stability Centre, University of Sussex, Brighton, UK
| | - Mark O'Driscoll
- Genome Damage & Stability Centre, University of Sussex, Brighton, UK
| | - Didier Hannequin
- Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, Department of Neurology, Department of Genetics and CNR-MAJ, F 76000, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | - Dominique Campion
- Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, Department of Genetics and CNR-MAJ, F 76000, Normandy Center for Genomic and Personalized Medicine, Rouen, France
- Department of Research, Rouvray Psychiatric Hospital, Sotteville-lès-Rouen, Rouen, France
| | - João R Oliveira
- Keizo Asami Laboratory, Universidade Federal de Pernambuco, Recife, Brazil
| | - Barbara Garavaglia
- Molecular Neurogenetics Unit, Movement Disorders Section, IRCCS Foundation Carlo Besta Neurological Institute, Via L. Temolo n. 4, Milan, 20116, Italy
| | - Giovanni Coppola
- Department of Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| | - Gaël Nicolas
- Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, Department of Genetics and CNR-MAJ, F 76000, Normandy Center for Genomic and Personalized Medicine, Rouen, France.
| |
Collapse
|
43
|
Mice Knocked Out for the Primary Brain Calcification–Associated Gene Slc20a2 Show Unimpaired Prenatal Survival but Retarded Growth and Nodules in the Brain that Grow and Calcify Over Time. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:1865-1881. [DOI: 10.1016/j.ajpath.2018.04.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 03/30/2018] [Accepted: 04/19/2018] [Indexed: 12/17/2022]
|
44
|
Hernando N, Wagner CA. Mechanisms and Regulation of Intestinal Phosphate Absorption. Compr Physiol 2018; 8:1065-1090. [PMID: 29978897 DOI: 10.1002/cphy.c170024] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
States of hypo- and hyperphosphatemia have deleterious consequences including rickets/osteomalacia and renal/cardiovascular disease, respectively. Therefore, the maintenance of appropriate plasma levels of phosphate is an essential requirement for health. This control is executed by the collaborative action of intestine and kidney whose capacities to (re)absorb phosphate are regulated by a number of hormonal and metabolic factors, among them parathyroid hormone, fibroblast growth factor 23, 1,25(OH)2 vitamin D3 , and dietary phosphate. The molecular mechanisms responsible for the transepithelial transport of phosphate across enterocytes are only partially understood. Indeed, whereas renal reabsorption entirely relies on well-characterized active transport mechanisms of phosphate across the renal proximal epithelia, intestinal absorption proceeds via active and passive mechanisms, with the molecular identity of the passive component still unknown. The active absorption of phosphate depends mostly on the activity and expression of the sodium-dependent phosphate cotransporter NaPi-IIb (SLC34A2), which is highly regulated by many of the factors, mentioned earlier. Physiologically, the contribution of NaPi-IIb to the maintenance of phosphate balance appears to be mostly relevant during periods of low phosphate availability. Therefore, its role in individuals living in industrialized societies with high phosphate intake is probably less relevant. Importantly, small increases in plasma phosphate, even within normal range, associate with higher risk of cardiovascular disease. Therefore, therapeutic approaches to treat hyperphosphatemia, including dietary phosphate restriction and phosphate binders, aim at reducing intestinal absorption. Here we review the current state of research in the field. © 2017 American Physiological Society. Compr Physiol 8:1065-1090, 2018.
Collapse
Affiliation(s)
- Nati Hernando
- National Center for Competence in Research NCCR Kidney.CH, Institute of Physiology, University Zurich-Irchel, Zurich, Switzerland
| | - Carsten A Wagner
- National Center for Competence in Research NCCR Kidney.CH, Institute of Physiology, University Zurich-Irchel, Zurich, Switzerland
| |
Collapse
|
45
|
Konno T, Blackburn PR, Rozen TD, van Gerpen JA, Ross OA, Atwal PS, Wszolek ZK. Anticipation in a family with primary familial brain calcification caused by an SLC20A2 variant. Neurol Neurochir Pol 2018; 52:386-389. [DOI: 10.1016/j.pjnns.2018.03.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 03/31/2018] [Indexed: 11/17/2022]
|
46
|
Hozumi I, Kurita H, Ozawa K, Furuta N, Inden M, Sekine SI, Yamada M, Hayashi Y, Kimura A, Inuzuka T, Seishima M. Inorganic phosphorus (Pi) in CSF is a biomarker for SLC20A2-associated idiopathic basal ganglia calcification (IBGC1). J Neurol Sci 2018; 388:150-154. [PMID: 29627011 DOI: 10.1016/j.jns.2018.03.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 02/07/2018] [Accepted: 03/06/2018] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Idiopathic basal ganglia calcification (IBGC), also called Fahr's disease or recently primary familial brain calcification (PFBC), is characterized by abnormal deposits of minerals including calcium mainly and phosphate in the brain. Mutations in SLC20A2 (IBGC1 (merged with former IBGC2 and IBGC3)), which encodes PiT-2, a phosphate transporter, is the major cause of IBGC. Recently, Slc20a2-KO mice have been showed to have elevated levels of inorganic phosphorus (Pi) in cerebrospinal fluid (CSF); however, CSF Pi levels in patients with IBGC have not been fully examined. METHODS We investigated the cases of 29 patients with IBGC including six patients with SLC20A2 mutation and three patients with PDGFB mutation, and 13 controls. The levels of sodium (Na), potassium (K), chloride (Cl), calcium (Ca), and Pi in sera and CSF were determined by potentiometry and colorimetry. Moreover, clinical manifestations were investigated in the IBGC patients with high Pi levels in CSF. RESULTS The study revealed that the average level of Pi in the CSF of the total group of patients with IBGC is significantly higher than that of the control group, and the levels of Pi in CSF of the IBGC patients with SLC20A2 mutations are significantly higher than those of the IBGC patients with PDGFB mutations, the other IBGC patients and controls. CONCLUSION Results of this study suggest that the levels of CSF Pi will be a good biomarker for IBGC1.
Collapse
Affiliation(s)
- Isao Hozumi
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan.
| | - Hisaka Kurita
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan
| | - Kazuhiro Ozawa
- Nursing Collaboration Center, Gifu College Nursing, 3047-1, Hashima, Gifu 501-6295, Japan
| | - Nobuyuki Furuta
- Department of Informative Clinical Medicine, Gifu University, Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Masatoshi Inden
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan
| | - Shin-Ichiro Sekine
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan
| | - Megumi Yamada
- Department of Neurology and Geriatrics, Gifu University, Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Yuichi Hayashi
- Department of Neurology and Geriatrics, Gifu University, Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Akio Kimura
- Department of Neurology and Geriatrics, Gifu University, Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Takashi Inuzuka
- Department of Neurology and Geriatrics, Gifu University, Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Mitsuru Seishima
- Department of Informative Clinical Medicine, Gifu University, Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| |
Collapse
|
47
|
Greenwood AD, Ishida Y, O'Brien SP, Roca AL, Eiden MV. Transmission, Evolution, and Endogenization: Lessons Learned from Recent Retroviral Invasions. Microbiol Mol Biol Rev 2018; 82:e00044-17. [PMID: 29237726 PMCID: PMC5813887 DOI: 10.1128/mmbr.00044-17] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Viruses of the subfamily Orthoretrovirinae are defined by the ability to reverse transcribe an RNA genome into DNA that integrates into the host cell genome during the intracellular virus life cycle. Exogenous retroviruses (XRVs) are horizontally transmitted between host individuals, with disease outcome depending on interactions between the retrovirus and the host organism. When retroviruses infect germ line cells of the host, they may become endogenous retroviruses (ERVs), which are permanent elements in the host germ line that are subject to vertical transmission. These ERVs sometimes remain infectious and can themselves give rise to XRVs. This review integrates recent developments in the phylogenetic classification of retroviruses and the identification of retroviral receptors to elucidate the origins and evolution of XRVs and ERVs. We consider whether ERVs may recurrently pressure XRVs to shift receptor usage to sidestep ERV interference. We discuss how related retroviruses undergo alternative fates in different host lineages after endogenization, with koala retrovirus (KoRV) receiving notable interest as a recent invader of its host germ line. KoRV is heritable but also infectious, which provides insights into the early stages of germ line invasions as well as XRV generation from ERVs. The relationship of KoRV to primate and other retroviruses is placed in the context of host biogeography and the potential role of bats and rodents as vectors for interspecies viral transmission. Combining studies of extant XRVs and "fossil" endogenous retroviruses in koalas and other Australasian species has broadened our understanding of the evolution of retroviruses and host-retrovirus interactions.
Collapse
Affiliation(s)
- Alex D Greenwood
- Department of Wildlife Diseases, Leibniz Institute for Zoo and Wildlife Research (IZW) in the Forschungsverbund Berlin e.V., Berlin, Germany
| | - Yasuko Ishida
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Sean P O'Brien
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Alfred L Roca
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Maribeth V Eiden
- Department of Wildlife Diseases, Leibniz Institute for Zoo and Wildlife Research (IZW) in the Forschungsverbund Berlin e.V., Berlin, Germany
| |
Collapse
|
48
|
Quintáns B, Oliveira J, Sobrido MJ. Primary familial brain calcifications. HANDBOOK OF CLINICAL NEUROLOGY 2018; 147:307-317. [PMID: 29325620 DOI: 10.1016/b978-0-444-63233-3.00020-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Primary familial brain calcification (PFBC) is a neurodegenerative disease with characteristic calcium deposits in the basal ganglia and other brain regions. The disease usually presents as a combination of abnormal movements, cognitive and psychiatric manifestations, clinically indistinguishable from other adult-onset neurodegenerative disorders. The differential diagnosis must be established with genetic and nongenetic disorders that can also lead to calcium deposits in encephalic structures. In the past years PFBC causal mutations have been discovered in genes related to calcium phosphate homeostasis (SLC20A2, XPR1) and in genes involved with endothelial function and integrity (PDGFB, PDGFRB). The most frequently mutated gene is SLC20A2, where mutations can affect any domain of the protein. There is no clearcut relationship between the specific mutation/gene, onset age, neuroimaging pattern, and severity of clinical manifestations. The discovery of the genetic basis of PFBC provides not only a diagnostic tool, but also an insight into the pathomechanisms and potential therapeutic trials for this rare disease.
Collapse
Affiliation(s)
- Beatriz Quintáns
- Instituto de Investigación Sanitaria (IDIS), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Santiago de Compostela, Spain
| | | | - María-Jesús Sobrido
- Instituto de Investigación Sanitaria (IDIS), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Santiago de Compostela, Spain.
| |
Collapse
|
49
|
Borges-Medeiros R, Mendes de Oliveira JR. A commentary on band-like calcification with simplified gyration and polymicrogyria: report of 10 new families and identification of five novel OCLN mutations. J Hum Genet 2017; 63:255-256. [PMID: 29192239 DOI: 10.1038/s10038-017-0390-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 10/25/2017] [Indexed: 11/09/2022]
Affiliation(s)
- Rayssa Borges-Medeiros
- Keizo Asami Laboratory and Neuropsychiatric Department, Federal University of Pernambuco, Recife, Brazil
| | | |
Collapse
|
50
|
Koyama S, Sato H, Kobayashi R, Kawakatsu S, Kurimura M, Wada M, Kawanami T, Kato T. Clinical and radiological diversity in genetically confirmed primary familial brain calcification. Sci Rep 2017; 7:12046. [PMID: 28935882 PMCID: PMC5608910 DOI: 10.1038/s41598-017-11595-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 08/25/2017] [Indexed: 01/03/2023] Open
Abstract
Primary familial brain calcification (PFBC) is a rare neuropsychiatric disorder with characteristic symmetrical brain calcifications. Patients with PFBC may have a variety of symptoms, although they also may be clinically asymptomatic. Parkinsonism is one of the most common movement disorders; however, the underlying mechanism remains unclear. This condition is typically transmitted in an autosomal dominant fashion. To date, mutations in SLC20A2, PDGFRB, PDGFB, and XPR1 have been reported to cause PFBC. The aim of the study was to identify the genetic cause of brain calcification in probands from three PFBC families and in 8 sporadic patients and to perform clinical and radiological assessments focusing on parkinsonism in mutation carriers. Three familial PFBC probands and their relatives and eight sporadic patients affected with brain calcifications were enrolled in this study. Whole-exome sequencing identified three novel mutations: c.269G > T, p.(Gly90Val) and c.516+1G > A in SLC20A2 in familial cases, and c.602-1G > T in PDGFB in a sporadic patient. The c.516+1G > A mutation resulted in exon 4 skipping in SLC20A2 (p.Val144Glyfs*85). Dopamine transporter single photon emission computed tomography using 123I-ioflupane and 123I-metaiodobenzylguanidine cardiac scintigraphy revealed pre-synaptic dopaminergic deficit and cardiac sympathetic nerve dysfunction in two SLC20A2-related PFBC patients with parkinsonism.
Collapse
Affiliation(s)
- Shingo Koyama
- Department of Neurology, Hematology, Metabolism, Endocrinology, and Diabetology, Yamagata University Faculty of Medicine, 2-2-2 Iida-nishi, Yamagata, 990-9585, Japan.
| | - Hidenori Sato
- Genomic Information Analysis Unit, Department of Genomic Cohort Research, Yamagata University Faculty of Medicine, 2-2-2 Iida-nishi, Yamagata, 990-9585, Japan
| | - Ryota Kobayashi
- Department of Psychiatry, Yamagata University Faculty of Medicine, 2-2-2 Iida-nishi, Yamagata, 990-9585, Japan
| | - Shinobu Kawakatsu
- Department of Neuropsychiatry, Aizu Medical Center, Fukushima Medical University, 21-2 Maeda, Tanisawa, Kawahigashi, Aizuwakamatsu, Fukushima, 969-3492, Japan
| | - Masayuki Kurimura
- Department of Neurology, Okitama Public General Hospital, 2000 Nishi-otsuka, Kawanishi-machi, Higashi-okitama-gun, Yamagata, 992-0601, Japan
| | - Manabu Wada
- Department of Neurology, Hematology, Metabolism, Endocrinology, and Diabetology, Yamagata University Faculty of Medicine, 2-2-2 Iida-nishi, Yamagata, 990-9585, Japan
| | - Toru Kawanami
- Department of Neurology, Hematology, Metabolism, Endocrinology, and Diabetology, Yamagata University Faculty of Medicine, 2-2-2 Iida-nishi, Yamagata, 990-9585, Japan
| | - Takeo Kato
- Department of Neurology, Hematology, Metabolism, Endocrinology, and Diabetology, Yamagata University Faculty of Medicine, 2-2-2 Iida-nishi, Yamagata, 990-9585, Japan
| |
Collapse
|