1
|
Kavakli E, Gul N, Begentas OC, Kiris E. Astrocytes in Primary Familial Brain Calcification (PFBC): Emphasis on the Importance of Induced Pluripotent Stem Cell-Derived Human Astrocyte Models. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1479:19-38. [PMID: 39841380 DOI: 10.1007/5584_2024_840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
Primary familial brain calcification (PFBC) is a rare, progressive central nervous system (CNS) disorder without a cure, and the current treatment methodologies primarily aim to relieve neurological and psychiatric symptoms of the patients. The disease is characterized by abnormal bilateral calcifications in the brain, however, our mechanistic understanding of the biology of the disease is still limited. Determining the roles of the specific cell types and molecular mechanisms involved in the pathophysiological processes of the disease is of great importance for the development of novel and effective treatment methodologies. There is a growing interest in the involvement of astrocytes in PFBC, as recent studies have suggested that astrocytes play a central role in the disease and that functional defects in these cells are critical for the development and progression of the disease. This review aims to discuss recent findings on the roles of astrocytes in PFBC pathophysiology, with a focus on known expression and roles of PFBC genes in astrocytes. Additionally, we discuss the importance of human astrocytes for PFBC disease modeling, and astrocytes as a potential therapeutic target in PFBC. Utilization of species-specific and physiologically relevant PFBC model systems can open new avenues for basic research, drug development, and regenerative medicine.
Collapse
Affiliation(s)
- Ebru Kavakli
- Department of Biological Sciences, Middle East Technical University, Ankara, Türkiye
| | - Nazli Gul
- Department of Biological Sciences, Middle East Technical University, Ankara, Türkiye
| | - Onur Can Begentas
- Department of Biological Sciences, Middle East Technical University, Ankara, Türkiye
| | - Erkan Kiris
- Department of Biological Sciences, Middle East Technical University, Ankara, Türkiye.
| |
Collapse
|
2
|
Abitew YA, Reyer H, Hadlich F, Oster M, Trakooljul N, Sommerfeld V, Rodehutscord M, Wimmers K, Ponsuksili S. Transcriptional responses to diets without mineral phosphorus supplementation in the jejunum of two high-yielding laying hen strains. Poult Sci 2024; 103:104484. [PMID: 39515113 PMCID: PMC11584914 DOI: 10.1016/j.psj.2024.104484] [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: 07/22/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Phosphorus (P) is an essential mineral for all forms of life including laying hens, playing a crucial role in growth and efficient egg production. Recent studies suggest that current P recommendations might exceed the physiological demand, leading to unnecessarily high P excretions. This study on Lohmann Brown (LB) and Lohmann Selected Leghorn (LSL) laying hens (n=80; 10 replicates per strain, production period, and dietary group) investigates transcriptional changes in the jejunum, a critical intestinal segment for mineral absorption, in response to a diet either without (P-) or with (P+) a mineral supplement from monocalcium phosphate, administered over a 4-week period during the transition (15-19 weeks) or onset of laying (20-24 weeks). DESeq2 analysis of RNA sequencing data revealed that most differentially expressed genes (DEGs) varied between strains and age groups, with less pronounced effects from dietary mineral P content. The 19-week-old LB hens showed a stronger response to dietary mineral P removal, with transcripts affiliated with increased adaptation of the metabolism and decreased immune pathway activation. The identified pathways such as folate biosynthesis and p53 signaling, potentially link altered energy and amino acid metabolism (2-oxocarboxylic acid and arginine). Interestingly, genes involved in calcium transport (CALB1) and cellular signaling (PRKCA, STEAP4) along with tight junctions (CLDN2) were affected by complete removal of mineral P supplements, suggesting a promoted intestinal mineral uptake. Transcriptional regulation in the jejunum in response to low dietary mineral content is strain-specific when the laying phase begins, which may contribute to a physiological Ca:P ratio.
Collapse
Affiliation(s)
- Yosef A Abitew
- Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Henry Reyer
- Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Frieder Hadlich
- Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Michael Oster
- Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Nares Trakooljul
- Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Vera Sommerfeld
- University of Hohenheim, Institute of Animal Science, Stuttgart, Germany
| | | | - Klaus Wimmers
- Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany; University of Rostock, Faculty of Agricultural and Environmental Sciences, Rostock, Germany
| | | |
Collapse
|
3
|
Behzadi P, St Hilaire C. Metabolites and metabolism in vascular calcification: links between adenosine signaling and the methionine cycle. Am J Physiol Heart Circ Physiol 2024; 327:H1361-H1375. [PMID: 39453431 PMCID: PMC11588312 DOI: 10.1152/ajpheart.00267.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 10/21/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
The global population of individuals with cardiovascular disease is expanding, and a key risk factor for major adverse cardiovascular events is vascular calcification. The pathogenesis of cardiovascular calcification is complex and multifaceted, with external cues driving epigenetic, transcriptional, and metabolic changes that promote vascular calcification. This review provides an overview of some of the lesser understood molecular processes involved in vascular calcification and discusses the links between calcification pathogenesis and aspects of adenosine signaling and the methionine pathway; the latter of which salvages the essential amino acid methionine, but also provides the substrate critical for methylation, a modification that regulates the function and activity of DNA and proteins. We explore the complex and dynamic nature of osteogenic reprogramming underlying intimal atherosclerotic calcification and medial arterial calcification (MAC). Atherosclerotic calcification is more widely studied; however, emerging studies now show that MAC is a significant pathology independent from atherosclerosis. Furthermore, we emphasize metabolite and metabolic-modulating factors that influence vascular calcification pathogenesis. Although the contributions of these mechanisms are more well-define in relation to atherosclerotic intimal calcification, understanding these pathways may provide crucial mechanistic insights into MAC and inform future therapeutic approaches. Herein, we highlight the significance of adenosine and methyltransferase pathways as key regulators of vascular calcification pathogenesis.
Collapse
Affiliation(s)
- Parya Behzadi
- Division of Cardiology, Department of Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Cynthia St Hilaire
- Division of Cardiology, Department of Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
4
|
Lamessa A, Tesfaye K, Woyimo TG, Gebremichael EH. First-time seizure revealing late-onset Fahr's disease: a case report and brief literature review. Front Hum Neurosci 2024; 18:1456610. [PMID: 39651493 PMCID: PMC11621208 DOI: 10.3389/fnhum.2024.1456610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 10/23/2024] [Indexed: 12/11/2024] Open
Abstract
Fahr's disease (FD), otherwise known as primary familial brain calcification, is a rare neurodegenerative condition that involves intracerebral calcification at the level of the basal ganglia and other brain regions. It is an inherited neurologic disorder, although its molecular genetics have not been thoroughly defined. Patients usually present with a wide range of symptoms, predominantly movement disorders and cognitive changes. However, seizures are a rare initial presenting features of late-onset FD in adults. Herein, we present the case of a 60-year-old man with no known chronic illnesses who was admitted to a tertiary hospital after experiencing first-time generalized tonic-clonic seizures and loss of consciousness for two days. Basic laboratory results were within normal limits, and a non-contrast brain computed tomography (CT) scan showed intracerebral calcification. The patient was diagnosed with epilepsy secondary to FD based on its modified diagnostic criteria and responded well to antiepileptic treatment. The case highlights a rare association and emphasizes the importance of considering this diagnosis in patients experiencing an inaugural seizure; appropriate tests should be performed to confirm or rule out other relevant and secondary causes, and the treatment should be modified accordingly.
Collapse
|
5
|
Banga A, Aulakh R, Kumar P, Kaur R, Kaur J. Does ensuring optimum vitamin D levels result in early resolution of neurocysticercosis? Int J Neurosci 2023; 133:1285-1294. [PMID: 35574655 DOI: 10.1080/00207454.2022.2078207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 03/29/2022] [Accepted: 05/11/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Neurocysticercosis is a leading cause of acquired epilepsy. Calcified granulomas are known to cause seizure recurrence. Researchers have reported that vitamin D deficiency is associated with brain calcification and reduction in calcification occurs with vitamin D receptor agonist calcitriol through upregulation of SLC20A2. Based on these observations, a hypothesis was proposed that the occurrence of calcification could be reduced by optimizing vitamin D levels, resulting in early resolution of neurocysticercosis. METHODOLOGY A case-control (retrospective and prospective) study on 60 children with solitary intraparenchymal neurocysticercosis, 20 new cases prior to starting cysticidal therapy and other 40 resolved cases was carried out. Among new cases, children deficient in vitamin D were given megadose of vitamin D and vitamin D levels were rechecked after 30 days. Children having normal vitamin D were taken as cases and the deficient ones were taken as controls. Standard treatment for neurocysticercosis was given. Three monthly MRI scans were done. Outcome was evaluated as resolution/persistence of neurocysticercosis at 3, 6, 9 and 12 months. STATISTICS AND RESULTS Pearson chi square/Fisher's exact test was used along with Kaplan Meier and log rank test. Of 60 patients, at 6 months 3 cases and 4 controls (p value 0.43), at 9 months 2 cases and 6 controls (p value 0.037) and at 12 months 3 cases and 6 controls (p value 0.029) had complete resolution of NCC. CONCLUSION The results do not show that adequate vitamin D levels result in early resolution of neurocysticercosis.
Collapse
Affiliation(s)
- Akshay Banga
- Department of Pediatrics, Government Medical College and Hospital Sector 32, Chandigarh, India
| | - Roosy Aulakh
- Department of Pediatrics, Government Medical College and Hospital Sector 32, Chandigarh, India
| | - Pankaj Kumar
- Department of Pediatrics, Government Medical College and Hospital Sector 32, Chandigarh, India
| | - Ravinder Kaur
- Department of Radiodiagnosis, Government Medical College and Hospital Sector 32, Chandigarh, India
| | - Jasbinder Kaur
- Department of Biochemistry, Government Medical College and Hospital Sector 32, Chandigarh, India
| |
Collapse
|
6
|
Srichawla BS, Andrade E, Kipkorir V. Decoding brain calcifications: A single-center descriptive case series and examination of pathophysiological mechanisms. SAGE Open Med Case Rep 2023; 11:2050313X231197521. [PMID: 37663154 PMCID: PMC10474788 DOI: 10.1177/2050313x231197521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/10/2023] [Indexed: 09/05/2023] Open
Abstract
Brain calcifications, previously known as Fahr's disease, is a rare neurological disorder marked by various clinical symptoms, including movement disorders, cognitive impairment, and psychiatric disturbances. Despite its clinical importance, its pathophysiology is unclear and there are no specific treatments. We present four cases of brain calcifications from our tertiary care center, with three female patients (75%) and an average age of 63 years. Our cohort featured both genetic and endocrine etiologies, including one primary familial brain calcification case with a c.852del frameshift mutation in the SLC20A2 gene, and two endocrinopathy-related cases. One patient had an acute stroke which may have been contributed by brain calcifications. Computerized tomography and magnetic resonance imaging scans revealed basal ganglia and dentate nucleus calcifications. Treatment involved physical and occupational therapy in all patients. Hypoparathyroidism-related brain calcifications were treated with oral supplementation with calcitriol, calcium, and vitamin D. Three patients showed improvement or stability of their symptoms. This case series underscores the diverse clinical presentations and etiologies of brain calcifications. The complex pathophysiology involves disrupted Ca+2-PO43- homeostasis, deficient cellular PO43- transport, and vascular irregularities in genetic etiologies. Future research should focus on identifying novel genetic mutations, understanding molecular pathways, and refining diagnostic techniques. Integrating multidisciplinary approaches may improve diagnosis, management, and prognosis for patients with this intricate neurological disorder.
Collapse
Affiliation(s)
- Bahadar S Srichawla
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Eduardo Andrade
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | | |
Collapse
|
7
|
Uehara Y, Tanaka Y, Zhao S, Nikolaidis NM, Pitstick LB, Wu H, Yu JJ, Zhang E, Hasegawa Y, Noel JG, Gardner JC, Kopras EJ, Haffey WD, Greis KD, Guo J, Woods JC, Wikenheiser-Brokamp KA, Kyle JE, Ansong C, Teitelbaum SL, Inoue Y, Altinişik G, Xu Y, McCormack FX. Insights into pulmonary phosphate homeostasis and osteoclastogenesis emerge from the study of pulmonary alveolar microlithiasis. Nat Commun 2023; 14:1205. [PMID: 36864068 PMCID: PMC9981730 DOI: 10.1038/s41467-023-36810-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 02/17/2023] [Indexed: 03/04/2023] Open
Abstract
Pulmonary alveolar microlithiasis is an autosomal recessive lung disease caused by a deficiency in the pulmonary epithelial Npt2b sodium-phosphate co-transporter that results in accumulation of phosphate and formation of hydroxyapatite microliths in the alveolar space. The single cell transcriptomic analysis of a pulmonary alveolar microlithiasis lung explant showing a robust osteoclast gene signature in alveolar monocytes and the finding that calcium phosphate microliths contain a rich protein and lipid matrix that includes bone resorbing osteoclast enzymes and other proteins suggested a role for osteoclast-like cells in the host response to microliths. While investigating the mechanisms of microlith clearance, we found that Npt2b modulates pulmonary phosphate homeostasis through effects on alternative phosphate transporter activity and alveolar osteoprotegerin, and that microliths induce osteoclast formation and activation in a receptor activator of nuclear factor-κB ligand and dietary phosphate dependent manner. This work reveals that Npt2b and pulmonary osteoclast-like cells play key roles in pulmonary homeostasis and suggest potential new therapeutic targets for the treatment of lung disease.
Collapse
Affiliation(s)
- Yasuaki Uehara
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Yusuke Tanaka
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Shuyang Zhao
- Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Nikolaos M Nikolaidis
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Lori B Pitstick
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Huixing Wu
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jane J Yu
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Erik Zhang
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Yoshihiro Hasegawa
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - John G Noel
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jason C Gardner
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Elizabeth J Kopras
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Wendy D Haffey
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kenneth D Greis
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jinbang Guo
- Center for Pulmonary Imaging Research, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jason C Woods
- Center for Pulmonary Imaging Research, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | | | - Jennifer E Kyle
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Charles Ansong
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Steven L Teitelbaum
- Department of Pathology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yoshikazu Inoue
- Department of Diffuse Lung Diseases and Respiratory Failure, Clinical Research Center, National Hospital Organization Kinki-Chuo Chest Medical Center, Osaka, Japan
| | - Göksel Altinişik
- Department of Chest Diseases, Faculty of Medicine, Pamukkale University, Pamukkale, Turkey
| | - Yan Xu
- Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Departments of Pediatrics and Biomedical Informatics, University of Cincinnati School of Medicine, Cincinnati, OH, USA.
| | - Francis X McCormack
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
8
|
Carecchio M, Mainardi M, Bonato G. The clinical and genetic spectrum of primary familial brain calcification. J Neurol 2023; 270:3270-3277. [PMID: 36862146 DOI: 10.1007/s00415-023-11650-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023]
Abstract
Primary familial brain calcification (PFBC), formerly known as Fahr's disease, is a rare neurodegenerative disease characterized by bilateral progressive calcification of the microvessels of the basal ganglia and other cerebral and cerebellar structures. PFBC is thought to be due to an altered function of the Neurovascular Unit (NVU), where abnormal calcium-phosphorus metabolism, functional and microanatomical alterations of pericytes and mitochondrial alterations cause a dysfunction of the blood-brain barrier (BBB) and the generation of an osteogenic environment with surrounding astrocyte activation and progressive neurodegeneration. Seven causative genes have been discovered so far, of which four with dominant (SLC20A2, PDGFB, PDGFRB, XPR1) and three with recessive inheritance (MYORG, JAM2, CMPK2). Clinical presentation ranges from asymptomatic subjects to movement disorders, cognitive decline and psychiatric disturbances alone or in various combinations. Radiological patterns of calcium deposition are similar in all known genetic forms, but central pontine calcification and cerebellar atrophy are highly suggestive of MYORG mutations and extensive cortical calcification has been associated with JAM2 mutations. Currently, no disease-modifying drugs or calcium-chelating agents are available and only symptomatic treatments can be offered.
Collapse
Affiliation(s)
- Miryam Carecchio
- Department of Neuroscience, University of Padua, Via Niccolò Giustiniani, 5, 35128, Padua, Italy.
| | - Michele Mainardi
- Department of Neuroscience, University of Padua, Via Niccolò Giustiniani, 5, 35128, Padua, Italy
| | - Giulia Bonato
- Department of Neuroscience, University of Padua, Via Niccolò Giustiniani, 5, 35128, Padua, Italy
| |
Collapse
|
9
|
Golgi damage caused by dysfunction of PiT-2 in primary familial brain calcification. Biochem Biophys Res Commun 2023; 642:167-174. [PMID: 36584480 DOI: 10.1016/j.bbrc.2022.12.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 12/19/2022] [Indexed: 12/25/2022]
Abstract
The Golgi apparatus is vital for protein modification and molecular trafficking. It is essential for nerve development and activity, and damage thereof is implicated in many neurological diseases. Primary familial brain calcification (PFBC) is a rare inherited neurodegenerative disease characterized by multiple brain calcifications. SLC20A2, which encodes the inorganic phosphate transporter 2 (PiT-2) protein, is the main pathogenic gene in PFBC. The PiT-2 protein is a sodium-dependent phosphate type III transporter, and dysfunction leads to a deficit in the cellular intake of inorganic phosphate (Pi) and calcium deposits. Whether the impaired Golgi apparatus is involved in the PFBC procession requires elucidation. In this study, we constructed induced pluripotent stem cells (iPSCs) derived from two PFBC patients with different SLC20A2 gene mutations (c.613G > A or del exon10) and two healthy volunteers as dependable cell models for research on pathogenic mechanism. To study the mechanism, we differentiated iPSCs into neurons and astrocytes in vitro. Our study found disruptive Golgi structure and damaged autophagy in PFBC neurons with increased activity of mTOR. We also found damaged mitochondria and increased apoptosis in the PFBC dopaminergic neurons and astrocytes. In this study, we prove that dysfunctional PiT-2 leads to an imbalance of cellular Pi, which may disrupt the Golgi apparatus with impaired autophagy, mitochondria and apoptosis in PFBC. Our study provides a new avenue for understanding nerve damage and pathogenic mechanism in brain calcifications.
Collapse
|
10
|
Expression of phosphate and calcium transporters and their regulators in parotid glands of mice. Pflugers Arch 2023; 475:203-216. [PMID: 36274099 PMCID: PMC9849193 DOI: 10.1007/s00424-022-02764-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/07/2022] [Accepted: 10/13/2022] [Indexed: 02/01/2023]
Abstract
The concentration of inorganic phosphate (Pi) in plasma is under hormonal control, with deviations from normal values promptly corrected to avoid hyper- or hypophosphatemia. Major regulators include parathyroid hormone (PTH), fibroblast growth factor 23 (FGF-23), and active vitamin D3 (calcitriol). This control is achieved by mechanisms largely dependent on regulating intestinal absorption and renal excretion, whose combined actions stabilise plasma Pi levels at around 1-2 mM. Instead, Pi concentrations up to 13 and 40 mM have been measured in saliva from humans and ruminants, respectively, suggesting that salivary glands have the capacity to concentrate Pi. Here we analysed the transcriptome of parotid glands, ileum, and kidneys of mice, to investigate their potential differences regarding the expression of genes responsible for epithelial transport of Pi as well as their known regulators. Given that Pi and Ca2+ homeostasis are tightly connected, the expression of genes involved in Ca2+ homeostasis was also included. In addition, we studied the effect of vitamin D3 treatment on the expression of Pi and Ca2+ regulating genes in the three major salivary glands. We found that parotid glands are equipped preferentially with Slc20 rather than with Slc34 Na+/Pi cotransporters, are suited to transport Ca2+ through the transcellular and paracellular route and are potential targets for PTH and vitamin D3 regulation.
Collapse
|
11
|
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: 2.3] [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
|
12
|
Rai N, Thapa M, Pokharel M, Acharya J, Yadav D. Intracranial Calcification and Seizure with Down Syndrome: A Case Report. JNMA J Nepal Med Assoc 2022; 60:1063-1065. [PMID: 36705097 PMCID: PMC9795128 DOI: 10.31729/jnma.7950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Indexed: 12/03/2022] Open
Abstract
Down syndrome is a genetic disorder caused by an extra copy of chromosome number 21. New onset of seizure in adults with Down syndrome is rare. The exact pathogenesis of intracranial calcification and seizure in Down syndrome is unknown, however, a possible association between hypocalcemia and vitamin D deficiency in Down syndrome was reported. An 18-year-old girl with nasal bridge, mongoloid slants, clinodactyly and saddle gap of toes, and prominent Downs phenotypes was present with a low level of parathyroid hormone, calcium, and vitamin D. Due to a higher prevalence of intracranial calcification in people with Down syndrome, there is an increased possibility of hypocalcemia and vitamin D deficiency. Hence, serum levels of calcium and vitamin D should always be checked before starting treatment with anti-epileptic drugs. Keywords basal ganglia; Down syndrome; seizure; trisomy 21.
Collapse
Affiliation(s)
- Nilshan Rai
- KIST Medical College and Teaching Hospital, Gwarko, Lalitpur, Nepal,Correspondence: Mr Nilshan Rai, KIST Medical College and Teaching Hospital, Gwarko, Lalitpur, Nepal. , Phone: +977-9861304002
| | - Monika Thapa
- Department of Neurology, National Neuro Center, Maharajgunj, Kathmandu, Nepal
| | - Merina Pokharel
- Department of Neurology, National Neuro Center, Maharajgunj, Kathmandu, Nepal
| | - Jinee Acharya
- KIST Medical College and Teaching Hospital, Gwarko, Lalitpur, Nepal
| | - Dhirendra Yadav
- Patan Academy of Health Sciences, Lagankhel, Lalitpur, Nepal
| |
Collapse
|
13
|
Bezerra DP, de Aguiar JP, Keasey MP, Rodrigues CG, de Oliveira JRM. MiR-9-5p Regulates Genes Linked to Cerebral Calcification in the Osteogenic Differentiation Model and Induces Generalized Alteration in the Ion Channels. J Mol Neurosci 2021; 71:1897-1905. [PMID: 34041689 DOI: 10.1007/s12031-021-01830-w] [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: 08/15/2018] [Accepted: 03/15/2021] [Indexed: 12/01/2022]
Abstract
MicroRNA-9 (miR-9) modulates gene expression and demonstrates high structural conservation and wide expression in the central nervous system. Bioinformatics analysis predicts almost 100 ion channels, membrane transporters and receptors, including genes linked to primary familial brain calcification (PFBC), as possible miR-9-5p targets. PFBC is a neurodegenerative disorder, characterized by bilateral and symmetrical calcifications in the brain, associated with motor and behavioral disturbances. In this work, we seek to study the influence of miR-9-5p in regulating genes involved in PFBC, in an osteogenic differentiation model with SaOs-2 cells. During the induced calcification process, solute carrier family 20 member 2 (SLC20A2) and platelet-derived growth factor receptor beta (PDGFRB) were downregulated, while platelet-derived growth factor beta (PDGFB) showed no significant changes. Significantly decreased levels of SLC20A2 and PDGFRB were caused by the presence of miR-9-5p, while PDGFB showed no regulation. We confirmed the findings using an miR-9-5p inhibitor and also probed the cells in electrophysiological analysis to assess whether such microRNA might affect a broader range of ion channels, membrane transporters and receptors. Our electrophysiological data show that an increase of the miR-9-5p in SaOs-2 cells decreased the density and amplitude of the output ionic currents, indicating that it may influence the activity, and perhaps the expression, of some ionic channels. Additional investigations should determine whether such an effect is specific to miR-9-5p, and whether it could be used, together with the miR-9-5p inhibitor, as a therapeutic or diagnostic tool.
Collapse
Affiliation(s)
| | | | - Matthew Philip Keasey
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | | | - João Ricardo Mendes de Oliveira
- Keizo Asami Laboratory, Federal University of Pernambuco, Recife, PE, Brazil. .,Neuropsychiatry Department, Federal University of Pernambuco, Recife, PE, Brazil.
| |
Collapse
|
14
|
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: 2.3] [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
|
15
|
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: 0.6] [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
|
16
|
Abstract
Phosphate is an essential nutrient for life and is a critical component of bone formation, a major signaling molecule, and structural component of cell walls. Phosphate is also a component of high-energy compounds (i.e., AMP, ADP, and ATP) and essential for nucleic acid helical structure (i.e., RNA and DNA). Phosphate plays a central role in the process of mineralization, normal serum levels being associated with appropriate bone mineralization, while high and low serum levels are associated with soft tissue calcification. The serum concentration of phosphate and the total body content of phosphate are highly regulated, a process that is accomplished by the coordinated effort of two families of sodium-dependent transporter proteins. The three isoforms of the SLC34 family (SLC34A1-A3) show very restricted tissue expression and regulate intestinal absorption and renal excretion of phosphate. SLC34A2 also regulates the phosphate concentration in multiple lumen fluids including milk, saliva, pancreatic fluid, and surfactant. Both isoforms of the SLC20 family exhibit ubiquitous expression (with some variation as to which one or both are expressed), are regulated by ambient phosphate, and likely serve the phosphate needs of the individual cell. These proteins exhibit similarities to phosphate transporters in nonmammalian organisms. The proteins are nonredundant as mutations in each yield unique clinical presentations. Further research is essential to understand the function, regulation, and coordination of the various phosphate transporters, both the ones described in this review and the phosphate transporters involved in intracellular transport.
Collapse
Affiliation(s)
- Nati Hernando
- University of Zurich-Irchel, Institute of Physiology, Zurich, Switzerland; Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky; and Robley Rex VA Medical Center, Louisville, Kentucky
| | - Kenneth Gagnon
- University of Zurich-Irchel, Institute of Physiology, Zurich, Switzerland; Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky; and Robley Rex VA Medical Center, Louisville, Kentucky
| | - Eleanor Lederer
- University of Zurich-Irchel, Institute of Physiology, Zurich, Switzerland; Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky; and Robley Rex VA Medical Center, Louisville, Kentucky
| |
Collapse
|
17
|
Developmental Changes in Phosphate Homeostasis. Rev Physiol Biochem Pharmacol 2020; 179:117-138. [PMID: 33398502 DOI: 10.1007/112_2020_52] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Phosphate is a multivalent ion critical for a variety of physiological functions including bone formation, which occurs rapidly in the developing infant. In order to ensure maximal bone mineralization, young animals must maintain a positive phosphate balance. To accomplish this, intestinal absorption and renal phosphate reabsorption are greater in suckling and young animals relative to adults. This review discusses the known intestinal and renal adaptations that occur in young animals in order to achieve a positive phosphate balance. Additionally, we discuss the ontogenic changes in phosphotropic endocrine signalling as it pertains to intestinal and renal phosphate handling, including several endocrine factors not always considered in the traditional dogma of phosphotropic endocrine signalling, such as growth hormone, triiodothyronine, and glucocorticoids. Finally, a proposed model of how these factors may contribute to achieving a positive phosphate balance during development is proposed.
Collapse
|
18
|
Peters MEM, de Brouwer EJM, Bartstra JW, Mali WPTM, Koek HL, Rozemuller AJM, Baas AF, de Jong PA. Mechanisms of calcification in Fahr disease and exposure of potential therapeutic targets. Neurol Clin Pract 2019; 10:449-457. [PMID: 33299674 DOI: 10.1212/cpj.0000000000000782] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 10/10/2019] [Indexed: 11/15/2022]
Abstract
Purpose of review There is growing interest in disorders involved in ectopic mineralization. Fahr disease or idiopathic basal ganglia calcification can serve as a model for ectopic mineralization in the basal ganglia, which is fairly common in the general population. In this review, we will focus on causative gene mutations and corresponding pathophysiologic pathways in Fahr disease. Recent findings Patients with Fahr disease have a variability of symptoms, such as movement disorders, psychiatric signs, and cognitive impairment, but can also be asymptomatic. Fahr disease is mostly autosomal dominant inherited, and there are mutations found in 4 causative genes. Mutations in SLC20A2 and XPR1 lead to a disrupted phosphate metabolism involving brain-specific inorganic phosphate transporters. Mutations in PDGFB and PDGFRB are associated with disrupted blood-brain barrier integrity and dysfunctional pericyte maintenance. In addition, the MYORG gene has recently been discovered to be involved in the autosomal recessive inheritance of Fahr. Summary Knowledge about the mutations and corresponding pathways may expose therapeutic opportunities for patients with Fahr disease and vascular calcifications in the brain in general.
Collapse
Affiliation(s)
- Melissa E M Peters
- Departments of Radiology (MEMP, JWB, WPTMM, PAdJ), Geriatrics (EJMdB, HLK), Pathology (AJMR), and Genetics (AFB), University Medical Center Utrecht, The Netherlands
| | - Esther J M de Brouwer
- Departments of Radiology (MEMP, JWB, WPTMM, PAdJ), Geriatrics (EJMdB, HLK), Pathology (AJMR), and Genetics (AFB), University Medical Center Utrecht, The Netherlands
| | - Jonas W Bartstra
- Departments of Radiology (MEMP, JWB, WPTMM, PAdJ), Geriatrics (EJMdB, HLK), Pathology (AJMR), and Genetics (AFB), University Medical Center Utrecht, The Netherlands
| | - Willem P Th M Mali
- Departments of Radiology (MEMP, JWB, WPTMM, PAdJ), Geriatrics (EJMdB, HLK), Pathology (AJMR), and Genetics (AFB), University Medical Center Utrecht, The Netherlands
| | - Huiberdina L Koek
- Departments of Radiology (MEMP, JWB, WPTMM, PAdJ), Geriatrics (EJMdB, HLK), Pathology (AJMR), and Genetics (AFB), University Medical Center Utrecht, The Netherlands
| | - Annemieke J M Rozemuller
- Departments of Radiology (MEMP, JWB, WPTMM, PAdJ), Geriatrics (EJMdB, HLK), Pathology (AJMR), and Genetics (AFB), University Medical Center Utrecht, The Netherlands
| | - Annette F Baas
- Departments of Radiology (MEMP, JWB, WPTMM, PAdJ), Geriatrics (EJMdB, HLK), Pathology (AJMR), and Genetics (AFB), University Medical Center Utrecht, The Netherlands
| | - Pim A de Jong
- Departments of Radiology (MEMP, JWB, WPTMM, PAdJ), Geriatrics (EJMdB, HLK), Pathology (AJMR), and Genetics (AFB), University Medical Center Utrecht, The Netherlands
| |
Collapse
|
19
|
Lamquet S, Ramos EM, Legati A, Coppola G, Hemelsoet D, Vanakker OM. A likely pathogenic variant in the SLC20A2 gene presenting with progressive myoclonus. Ann Clin Transl Neurol 2019; 6:605-609. [PMID: 30911583 PMCID: PMC6414494 DOI: 10.1002/acn3.702] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 10/05/2018] [Accepted: 10/08/2018] [Indexed: 12/17/2022] Open
Abstract
A 60-year-old man is presented with progressive involuntary muscle movements and neuropsychiatric symptoms who developed a variety of additional complaints over 2 years. Brain imaging revealed bilateral basal ganglia calcifications suggesting primary familial brain calcification. Analysis of the SLC20A2 gene revealed a missense mutation (c.541C>T, p.(Arg181Trp)), in silico predicted to be deleterious and not found in available databases. Segregation analysis confirmed his asymptomatic father to harbor the same mutation, though on brain imaging basal ganglia calcifications were found. This report illustrates the intrafamilial variability of the phenotype and generalized myoclonus as the presenting symptom.
Collapse
Affiliation(s)
- Simon Lamquet
- Department of Neurology Ghent University Hospital Ghent Belgium
| | - Eliana M Ramos
- UCLA Jane & Terry Semel Institute for Neuroscience & Human Behavior Los Angeles California
| | - Andrea Legati
- UCLA Jane & Terry Semel Institute for Neuroscience & Human Behavior Los Angeles California
| | - Giovanni Coppola
- UCLA Jane & Terry Semel Institute for Neuroscience & Human Behavior Los Angeles California
| | | | | |
Collapse
|
20
|
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: 5.6] [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
|
21
|
Calcitriol Reverses the Down-Regulation Pattern of Tuberous Sclerosis Complex Genes in an In Vitro Calcification Model. J Mol Neurosci 2017; 64:140-143. [PMID: 29230685 DOI: 10.1007/s12031-017-1004-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 11/23/2017] [Indexed: 02/06/2023]
Abstract
Tuberous sclerosis complex (TSC) is a neurocutaneous syndrome with autosomal dominant inheritance, and most of the cases are related to loss of function of the TSC1 and TSC2 genes. TSC may occur with a wide range of clinical findings and skin, kidney, brain, and heart are the most commonly affected organs. Brain calcifications in TSC are also described and reported as diffuse and without pattern of symmetry or bilaterality. Recently, a new discovery opened the possibility of using vitamin D (VitD) for treating cerebral calcifications. Calcitriol, the active form of VitD, was able to reduce the calcification in an in vitro model, increasing expression of a gene related to primary familial brain calcification. We show that in the same experimental model, calcitriol was also able to restore and even increase expression of genes related to TSC. This article discusses the use of calcitriol supplementation in patients with TSC, which can be a very interesting strategy due to its low cost and because it is already used in various therapies.
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW We give an update on the etiology and potential treatment options of rare inherited monogenic disorders associated with arterial calcification and calcific cardiac valve disease. RECENT FINDINGS Genetic studies of rare inherited syndromes have identified key regulators of ectopic calcification. Based on the pathogenic principles causing the diseases, these can be classified into three groups: (1) disorders of an increased extracellular inorganic phosphate/inorganic pyrophosphate ratio (generalized arterial calcification of infancy, pseudoxanthoma elasticum, arterial calcification and distal joint calcification, progeria, idiopathic basal ganglia calcification, and hyperphosphatemic familial tumoral calcinosis; (2) interferonopathies (Singleton-Merten syndrome); and (3) others, including Keutel syndrome and Gaucher disease type IIIC. Although some of the identified causative mechanisms are not easy to target for treatment, it has become clear that a disturbed serum phosphate/pyrophosphate ratio is a major force triggering arterial and cardiac valve calcification. Further studies will focus on targeting the phosphate/pyrophosphate ratio to effectively prevent and treat these calcific disease phenotypes.
Collapse
MESH Headings
- Abnormalities, Multiple/drug therapy
- Abnormalities, Multiple/genetics
- Abnormalities, Multiple/metabolism
- Aortic Diseases/drug therapy
- Aortic Diseases/genetics
- Aortic Diseases/metabolism
- Basal Ganglia Diseases/drug therapy
- Basal Ganglia Diseases/genetics
- Basal Ganglia Diseases/metabolism
- Calcinosis/drug therapy
- Calcinosis/genetics
- Calcinosis/metabolism
- Cartilage Diseases/drug therapy
- Cartilage Diseases/genetics
- Cartilage Diseases/metabolism
- Dental Enamel Hypoplasia/drug therapy
- Dental Enamel Hypoplasia/genetics
- Dental Enamel Hypoplasia/metabolism
- Diphosphates/metabolism
- Enzyme Replacement Therapy
- Gaucher Disease/drug therapy
- Gaucher Disease/genetics
- Gaucher Disease/metabolism
- Hand Deformities, Congenital/drug therapy
- Hand Deformities, Congenital/genetics
- Hand Deformities, Congenital/metabolism
- Humans
- Hyperostosis, Cortical, Congenital/drug therapy
- Hyperostosis, Cortical, Congenital/genetics
- Hyperostosis, Cortical, Congenital/metabolism
- Hyperphosphatemia/drug therapy
- Hyperphosphatemia/genetics
- Hyperphosphatemia/metabolism
- Interferons/metabolism
- Metacarpus/abnormalities
- Metacarpus/metabolism
- Muscular Diseases/drug therapy
- Muscular Diseases/genetics
- Muscular Diseases/metabolism
- Odontodysplasia/drug therapy
- Odontodysplasia/genetics
- Odontodysplasia/metabolism
- Osteoporosis/drug therapy
- Osteoporosis/genetics
- Osteoporosis/metabolism
- Phosphates/metabolism
- Progeria/drug therapy
- Progeria/genetics
- Progeria/metabolism
- Pseudoxanthoma Elasticum/drug therapy
- Pseudoxanthoma Elasticum/genetics
- Pseudoxanthoma Elasticum/metabolism
- Pulmonary Valve Stenosis/drug therapy
- Pulmonary Valve Stenosis/genetics
- Pulmonary Valve Stenosis/metabolism
- Vascular Calcification/drug therapy
- Vascular Calcification/genetics
- Vascular Calcification/metabolism
Collapse
Affiliation(s)
- Yvonne Nitschke
- Department of General Pediatrics, Münster University Children's Hospital, Albert-Schweitzer-Campus 1, D-48149, Münster, Germany
| | - Frank Rutsch
- Department of General Pediatrics, Münster University Children's Hospital, Albert-Schweitzer-Campus 1, D-48149, Münster, Germany.
| |
Collapse
|
23
|
Phosphate Transporters Expression in Patients with Primary Familial Brain Calcifications. J Mol Neurosci 2017; 62:276-280. [DOI: 10.1007/s12031-017-0934-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 05/26/2017] [Indexed: 12/26/2022]
|
24
|
Three novel missense mutations in SLC20A2 associated with idiopathic basal ganglia calcification. J Neurol Sci 2017; 377:62-64. [PMID: 28477710 DOI: 10.1016/j.jns.2017.03.053] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/28/2017] [Accepted: 03/30/2017] [Indexed: 01/30/2023]
|
25
|
MiR-9-5p Down-Regulates PiT2, but not PiT1 in Human Embryonic Kidney 293 Cells. J Mol Neurosci 2017; 62:28-33. [PMID: 28303467 DOI: 10.1007/s12031-017-0906-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 02/27/2017] [Indexed: 10/20/2022]
Abstract
PiT1 (SLC20A1) and PiT2 (SLC20A2) are members of the mammalian type-III inorganic phosphate transporters and recent studies linked SLC20A2 mutations with primary brain calcifications. MicroRNAs (miRNAs) are endogenous noncoding regulatory RNAs and MicroRNA-9 (miR-9) modulates neurogenesis but is also involved with different types of cancer. We evaluated possible interactions between miR-9 and the phosphate transporters (PiT1 and PiT2). SLC20A2, platelet-derived growth factor receptor beta (PDGFRB) and Fibrillin-2 (FBN2) showed binding sites with high affinity for mir-9, In silico. miR-9 mimic was transfected into HEK293 cells and expression was confirmed by RT-qPCR. Overexpression of miR-9 in these cells caused a significant reduction in PiT2 and FBN2. PDGFRB appeared to be decreased, but was not significantly down-regulated. PiT1 showed no significant difference relative to controls. The down-regulation of PiT2 protein by miR-9 was confirmed by western blotting. In conclusion, we showed that miR-9 can down-regulate PiT2, in HEK293 cells. [corrected].
Collapse
|
26
|
Visavadiya NP, Keasey MP, Razskazovskiy V, Banerjee K, Jia C, Lovins C, Wright GL, Hagg T. Integrin-FAK signaling rapidly and potently promotes mitochondrial function through STAT3. Cell Commun Signal 2016; 14:32. [PMID: 27978828 PMCID: PMC5159999 DOI: 10.1186/s12964-016-0157-7] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 12/06/2016] [Indexed: 01/02/2023] Open
Abstract
Background STAT3 is increasingly becoming known for its non-transcriptional regulation of mitochondrial bioenergetic function upon activation of its S727 residue (S727-STAT3). Lengthy mitochondrial dysfunction can lead to cell death. We tested whether an integrin-FAK-STAT3 signaling pathway we recently discovered regulates mitochondrial function and cell survival, and treatments thereof. Methods Cultured mouse brain bEnd5 endothelial cells were treated with integrin, FAK or STAT3 inhibitors, FAK siRNA, as well as integrin and STAT3 activators. STAT3 null cells were transfected with mutant STAT3 plasmids. Outcome measures included oxygen consumption rate for mitochondrial bioenergetics, Western blotting for protein phosphorylation, mitochondrial membrane potential for mitochondrial integrity, ROS production, and cell counts. Results Vitronectin-dependent mitochondrial basal respiration, ATP production, and maximum reserve and respiratory capacities were suppressed within 4 h by RGD and αvβ3 integrin antagonist peptides. Conversely, integrin ligands vitronectin, laminin and fibronectin stimulated mitochondrial function. Pharmacological inhibition of FAK completely abolished mitochondrial function within 4 h while FAK siRNA treatments confirmed the specificity of FAK signaling. WT, but not S727A functionally dead mutant STAT3, rescued bioenergetics in cells made null for STAT3 using CRISPR-Cas9. STAT3 inhibition with stattic in whole cells rapidly reduced mitochondrial function and mitochondrial pS727-STAT3. Stattic treatment of isolated mitochondria did not reduce pS727 whereas more was detected upon phosphatase inhibition. This suggests that S727-STAT3 is activated in the cytoplasm and is short-lived upon translocation to the mitochondria. FAK inhibition reduced pS727-STAT3 within mitochondria and reduced mitochondrial function in a non-transcriptional manner, as shown by co-treatment with actinomycin. Treatment with the small molecule bryostatin-1 or hepatocyte growth factor (HGF), which indirectly activate S727-STAT3, preserved mitochondrial function during FAK inhibition, but failed in the presence of the STAT3 inhibitor. FAK inhibition induced loss of mitochondrial membrane potential, which was counteracted by bryostatin, and increased superoxide and hydrogen peroxide production. Bryostatin and HGF reduced the substantial cell death caused by FAK inhibition over a 24 h period. Conclusion These data suggest that extracellular matrix molecules promote STAT3-dependent mitochondrial function and cell survival through integrin-FAK signaling. We furthermore show a new treatment strategy for cell survival using S727-STAT3 activators.
Collapse
Affiliation(s)
- Nishant P Visavadiya
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Building 178, Maple Ave, PO Box 70582, Johnson City, TN37614, USA
| | - Matthew P Keasey
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Building 178, Maple Ave, PO Box 70582, Johnson City, TN37614, USA
| | - Vladislav Razskazovskiy
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Building 178, Maple Ave, PO Box 70582, Johnson City, TN37614, USA
| | - Kalpita Banerjee
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Building 178, Maple Ave, PO Box 70582, Johnson City, TN37614, USA
| | - Cuihong Jia
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Building 178, Maple Ave, PO Box 70582, Johnson City, TN37614, USA
| | - Chiharu Lovins
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Building 178, Maple Ave, PO Box 70582, Johnson City, TN37614, USA
| | - Gary L Wright
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Building 178, Maple Ave, PO Box 70582, Johnson City, TN37614, USA
| | - Theo Hagg
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Building 178, Maple Ave, PO Box 70582, Johnson City, TN37614, USA.
| |
Collapse
|