101
|
Jeitner TM. Cofactors and Coenzymes | Cysteamine. ENCYCLOPEDIA OF BIOLOGICAL CHEMISTRY III 2021:346-355. [DOI: 10.1016/b978-0-12-819460-7.00141-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
102
|
Ouhenach M, Zrhidri A, Jaouad IC, Smaili W, Sefiani A. Application of next generation sequencing in genetic counseling a case of a couple at risk of cystinosis. BMC MEDICAL GENETICS 2020; 21:240. [PMID: 33308164 PMCID: PMC7733293 DOI: 10.1186/s12881-020-01167-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 11/10/2020] [Indexed: 11/25/2022]
Abstract
Background In Morocco, consanguinity rate is very high; which lead to an increase in the birth prevalence of infants with autosomal recessive disorders. Previously, it was difficult to diagnose rare autosomal recessive diseases. Next Generation Sequencing (NGS) techniques have considerably improved clinical diagnostics. A genetic diagnosis showing biallelic causative mutations is the requirement for targeted carrier testing in parents, prenatal and preimplantation genetic diagnosis in further pregnancies, and also for targeted premarital testing in future couples at risk of producing affected children by a known autosomal recessive disease. Methods In this report, we present our strategy to advise a future couple of first cousins, whose descendants would risk cystinosis; an autosomal recessive lysosomal disease caused by mutations in the CTNS gene. Indeed, our future husband’s sister is clinically and biochemically diagnosed with cystinosis in early childhood. First, we opted to identify the patient’s CTNS gene abnormality by using (NGS), then we searched for heterozygosity in the couple’s DNA, which allows us to predict the exact risk of this familial disease in the future couple’s offspring. Results We have shown that the future husband, brother of the patient is heterozygous for the familial mutation. On the other hand, his future wife did not inherit the familial mutation. Therefore, genetic counseling was reassuring for the risk of familial cystinosis in this couple’s offspring. Conclusions We report in this study, one of the major applications of (NGS), an effective tool to improve clinical diagnosis and to provide the possibility of targeted premarital carrier testing in couples at risk.
Collapse
Affiliation(s)
- Mouna Ouhenach
- Department of Medical Genetics, National Institute of Health, 27, Avenue Ibn Battouta, BP 769,, Rabat, Morocco. .,Human Genomics Center, Faculty of Medicine and Pharmacy, Rabat, Morocco.
| | - Abdelali Zrhidri
- Department of Medical Genetics, National Institute of Health, 27, Avenue Ibn Battouta, BP 769,, Rabat, Morocco.,Human Genomics Center, Faculty of Medicine and Pharmacy, Rabat, Morocco
| | - Imane Cherkaoui Jaouad
- Department of Medical Genetics, National Institute of Health, 27, Avenue Ibn Battouta, BP 769,, Rabat, Morocco.,Human Genomics Center, Faculty of Medicine and Pharmacy, Rabat, Morocco
| | - Wiam Smaili
- Department of Medical Genetics, National Institute of Health, 27, Avenue Ibn Battouta, BP 769,, Rabat, Morocco
| | - Abdelaziz Sefiani
- Department of Medical Genetics, National Institute of Health, 27, Avenue Ibn Battouta, BP 769,, Rabat, Morocco.,Human Genomics Center, Faculty of Medicine and Pharmacy, Rabat, Morocco
| |
Collapse
|
103
|
Lemaire M. Novel Fanconi renotubular syndromes provide insights in proximal tubule pathophysiology. Am J Physiol Renal Physiol 2020; 320:F145-F160. [PMID: 33283647 DOI: 10.1152/ajprenal.00214.2020] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The various forms of Fanconi renotubular syndromes (FRTS) offer significant challenges for clinicians and present unique opportunities for scientists who study proximal tubule physiology. This review will describe the clinical characteristics, genetic underpinnings, and underlying pathophysiology of the major forms of FRST. Although the classic forms of FRTS will be presented (e.g., Dent disease or Lowe syndrome), particular attention will be paid to five of the most recently discovered FRTS subtypes caused by mutations in the genes encoding for L-arginine:glycine amidinotransferase (GATM), solute carrier family 34 (type Ii sodium/phosphate cotransporter), member 1 (SLC34A1), enoyl-CoAhydratase/3-hydroxyacyl CoA dehydrogenase (EHHADH), hepatocyte nuclear factor 4A (HNF4A), or NADH dehydrogenase complex I, assembly factor 6 (NDUFAF6). We will explore how mutations in these genes revealed unexpected mechanisms that led to compromised proximal tubule functions. We will also describe the inherent challenges associated with gene discovery studies based on findings derived from small, single-family studies by focusing the story of FRTS type 2 (SLC34A1). Finally, we will explain how extensive alternative splicing of HNF4A has resulted in confusion with mutation nomenclature for FRTS type 4.
Collapse
Affiliation(s)
- Mathieu Lemaire
- Division of Nephrology and Cell Biology Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Pediatrics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
104
|
Adelmann CH, Traunbauer AK, Chen B, Condon KJ, Chan SH, Kunchok T, Lewis CA, Sabatini DM. MFSD12 mediates the import of cysteine into melanosomes and lysosomes. Nature 2020; 588:699-704. [PMID: 33208952 PMCID: PMC7770032 DOI: 10.1038/s41586-020-2937-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 08/28/2020] [Indexed: 12/12/2022]
Abstract
Dozens of genes contribute to the wide variation in human pigmentation. Many of these genes encode proteins that localize to the melanosome-the organelle, related to the lysosome, that synthesizes pigment-but have unclear functions1,2. Here we describe MelanoIP, a method for rapidly isolating melanosomes and profiling their labile metabolite contents. We use this method to study MFSD12, a transmembrane protein of unknown molecular function that, when suppressed, causes darker pigmentation in mice and humans3,4. We find that MFSD12 is required to maintain normal levels of cystine-the oxidized dimer of cysteine-in melanosomes, and to produce cysteinyldopas, the precursors of pheomelanin synthesis made in melanosomes via cysteine oxidation5,6. Tracing and biochemical analyses show that MFSD12 is necessary for the import of cysteine into melanosomes and, in non-pigmented cells, lysosomes. Indeed, loss of MFSD12 reduced the accumulation of cystine in lysosomes of fibroblasts from patients with cystinosis, a lysosomal-storage disease caused by inactivation of the lysosomal cystine exporter cystinosin7-9. Thus, MFSD12 is an essential component of the cysteine importer for melanosomes and lysosomes.
Collapse
Affiliation(s)
- Charles H Adelmann
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Anna K Traunbauer
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Brandon Chen
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kendall J Condon
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sze Ham Chan
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tenzin Kunchok
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Caroline A Lewis
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - David M Sabatini
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
105
|
Huizing M, Gahl WA. Inherited disorders of lysosomal membrane transporters. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2020; 1862:183336. [PMID: 32389669 PMCID: PMC7508925 DOI: 10.1016/j.bbamem.2020.183336] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/01/2020] [Accepted: 04/28/2020] [Indexed: 02/07/2023]
Abstract
Disorders caused by defects in lysosomal membrane transporters form a distinct subgroup of lysosomal storage disorders (LSDs). To date, defects in only 10 lysosomal membrane transporters have been associated with inherited disorders. The clinical presentations of these diseases resemble the phenotypes of other LSDs; they are heterogeneous and often present in children with neurodegenerative manifestations. However, for pathomechanistic and therapeutic studies, lysosomal membrane transport defects should be distinguished from LSDs caused by defective hydrolytic enzymes. The involved proteins differ in function, localization, and lysosomal targeting, and the diseases themselves differ in their stored material and therapeutic approaches. We provide an overview of the small group of disorders of lysosomal membrane transporters, emphasizing discovery, pathomechanism, clinical features, diagnostic methods and therapeutic aspects. We discuss common aspects of lysosomal membrane transporter defects that can provide the basis for preclinical research into these disorders.
Collapse
Affiliation(s)
- Marjan Huizing
- Human Biochemical Genetics Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - William A Gahl
- Human Biochemical Genetics Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
106
|
Iyob-Tessema H, Wang CS, Kennedy S, Reyes L, Shin S, Greenbaum LA, Hogan J. Grip Strength in Adults and Children with Cystinosis. Kidney Int Rep 2020; 6:389-395. [PMID: 33615064 PMCID: PMC7879123 DOI: 10.1016/j.ekir.2020.11.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/30/2020] [Accepted: 11/17/2020] [Indexed: 11/05/2022] Open
Abstract
Introduction Chronic kidney disease (CKD) is associated with impaired muscle strength. Patients with cystinosis have an increased risk for impaired muscle strength because of early development of CKD and cystinosis-induced myopathy. This study assesses muscle strength in patients with cystinosis and investigates risk factors of decreased muscle strength. Methods Adult and pediatric patients were recruited from Cystinosis Research Network conferences and a large pediatric nephrology clinic between 2017 and 2019. Patients and caregivers completed questionnaires on demographic characteristics, disease course, daily physical activity, and neuromuscular symptoms. Grip strength was assessed using a dynameter and calculated z-scores for age and sex were assessed for associations with patient characteristics. Results We included 76 patients with a mean grip strength z-score of −2.1 (SD, 1.1), which was lower than seen in patients with CKD without cystinosis. Male sex and delayed cysteamine initiation were independently associated with impaired grip strength. Among adults, a low level of physical activity was associated with lower grip strength z score, but no association was found in children. A third of the patients reported neuromuscular symptoms, with swallowing issues associated with lower grip strength. There was no significant correlation between eGFR and grip strength z-score. Conclusion Patients with cystinosis have impaired muscle strength compared with healthy control subjects and patients with CKD. This impairment is greater in male patients and in patients with late initiation of cysteamine therapy and is associated with lower physical activity. Further studies investigating the effect of different types of physical activities, optimizing cysteamine therapy, and other interventions are needed.
Collapse
Affiliation(s)
- Helina Iyob-Tessema
- Department of Pediatrics, Emory School of Medicine and Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Chia-Shi Wang
- Department of Pediatrics, Emory School of Medicine and Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Sabina Kennedy
- Department of Pediatrics, Emory School of Medicine and Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Loretta Reyes
- Department of Pediatrics, Emory School of Medicine and Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Stella Shin
- Department of Pediatrics, Emory School of Medicine and Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Larry A Greenbaum
- Department of Pediatrics, Emory School of Medicine and Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Julien Hogan
- Emory Transplant Center, Department of Surgery, Emory School of Medicine, Atlanta, Georgia, USA.,Pediatric Nephology Department, Robert Debré Hospital, Paris, France
| |
Collapse
|
107
|
Tokhmafshan F, Dickinson K, Akpa MM, Brasell E, Huertas P, Goodyer PR. A no-nonsense approach to hereditary kidney disease. Pediatr Nephrol 2020; 35:2031-2042. [PMID: 31807928 DOI: 10.1007/s00467-019-04394-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 07/05/2019] [Accepted: 10/07/2019] [Indexed: 01/12/2023]
Abstract
The advent of a new class of aminoglycosides with increased translational readthrough of nonsense mutations and reduced toxicity offers a new therapeutic strategy for a subset of patients with hereditary kidney disease. The renal uptake and retention of aminoglycosides at a high intracellular concentration makes the kidney an ideal target for this approach. In this review, we explore the potential of aminoglycoside readthrough therapy in a number of hereditary kidney diseases and discuss the therapeutic window of opportunity for subclasses of each disease, when caused by nonsense mutations.
Collapse
Affiliation(s)
- Fatima Tokhmafshan
- Research Institute of the McGill University Health Center, 1001 Décarie Boulevard, EM1.2232, Montreal, QC, H4A 3J1, Canada
| | - Kyle Dickinson
- Research Institute of the McGill University Health Center, 1001 Décarie Boulevard, EM1.2232, Montreal, QC, H4A 3J1, Canada.,Department of Experimental Medicine, McGill University, Montreal, Canada
| | - Murielle M Akpa
- Research Institute of the McGill University Health Center, 1001 Décarie Boulevard, EM1.2232, Montreal, QC, H4A 3J1, Canada
| | - Emma Brasell
- Department of Human Genetics, McGill University, Montreal, Canada
| | | | - Paul R Goodyer
- Research Institute of the McGill University Health Center, 1001 Décarie Boulevard, EM1.2232, Montreal, QC, H4A 3J1, Canada. .,Department of Experimental Medicine, McGill University, Montreal, Canada. .,Department of Human Genetics, McGill University, Montreal, Canada. .,Department of Pediatrics, McGill University, Montreal, Canada.
| |
Collapse
|
108
|
Cheung WW, Ding W, Hoffman HM, Wang Z, Hao S, Zheng R, Gonzalez A, Zhan JY, Zhou P, Li S, Esparza MC, Lieber RL, Mak RH. Vitamin D ameliorates adipose browning in chronic kidney disease cachexia. Sci Rep 2020; 10:14175. [PMID: 32843714 PMCID: PMC7447759 DOI: 10.1038/s41598-020-70190-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 07/21/2020] [Indexed: 12/15/2022] Open
Abstract
Patients with chronic kidney disease (CKD) are often 25(OH)D3 and 1,25(OH)2D3 insufficient. We studied whether vitamin D repletion could correct aberrant adipose tissue and muscle metabolism in a mouse model of CKD-associated cachexia. Intraperitoneal administration of 25(OH)D3 and 1,25(OH)2D3 (75 μg/kg/day and 60 ng/kg/day respectively for 6 weeks) normalized serum concentrations of 25(OH)D3 and 1,25(OH)2D3 in CKD mice. Vitamin D repletion stimulated appetite, normalized weight gain, and improved fat and lean mass content in CKD mice. Vitamin D supplementation attenuated expression of key molecules involved in adipose tissue browning and ameliorated expression of thermogenic genes in adipose tissue and skeletal muscle in CKD mice. Furthermore, repletion of vitamin D improved skeletal muscle fiber size and in vivo muscle function, normalized muscle collagen content and attenuated muscle fat infiltration as well as pathogenetic molecular pathways related to muscle mass regulation in CKD mice. RNAseq analysis was performed on the gastrocnemius muscle. Ingenuity Pathway Analysis revealed that the top 12 differentially expressed genes in CKD were correlated with impaired muscle and neuron regeneration, enhanced muscle thermogenesis and fibrosis. Importantly, vitamin D repletion normalized the expression of those 12 genes in CKD mice. Vitamin D repletion may be an effective therapeutic strategy for adipose tissue browning and muscle wasting in CKD patients.
Collapse
MESH Headings
- Adipocytes, Beige/drug effects
- Adipocytes, Beige/metabolism
- Adipocytes, Brown/metabolism
- Adipocytes, White/metabolism
- Animals
- Cachexia/drug therapy
- Cachexia/etiology
- Cachexia/physiopathology
- Calcifediol/blood
- Calcifediol/deficiency
- Calcifediol/pharmacology
- Calcifediol/therapeutic use
- Calcitriol/blood
- Calcitriol/deficiency
- Calcitriol/pharmacology
- Calcitriol/therapeutic use
- Disease Models, Animal
- Eating/drug effects
- Fibrosis/genetics
- Gene Expression Regulation/drug effects
- Hand Strength
- Mice
- Mice, Inbred C57BL
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/pathology
- Nephrectomy
- Parathyroid Hormone/blood
- RNA, Messenger/biosynthesis
- Renal Insufficiency, Chronic/blood
- Renal Insufficiency, Chronic/complications
- Renal Insufficiency, Chronic/drug therapy
- Rotarod Performance Test
- Sequence Analysis, RNA
- Thermogenesis/drug effects
- Weight Gain/drug effects
Collapse
Affiliation(s)
- Wai W Cheung
- Pediatric Nephrology, Rady Children's Hospital San Diego, University of California, San Diego, USA
| | - Wei Ding
- Division of Nephrology, School of Medicine, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Hal M Hoffman
- Department of Pediatrics, University of California, San Diego, USA
| | - Zhen Wang
- Department of Pediatrics, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Sheng Hao
- Department of Nephrology and Rheumatology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ronghao Zheng
- Department of Pediatrics, Hubei Maternal and Child Health Hospital, Wuhan, China
| | - Alex Gonzalez
- Pediatric Nephrology, Rady Children's Hospital San Diego, University of California, San Diego, USA
| | - Jian-Ying Zhan
- Children's Hospital, Zhejiang University, Hangzhou, China
| | - Ping Zhou
- Department of Pediatrics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shiping Li
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, China
| | - Mary C Esparza
- Department of Orthopedic Surgery, University of California, San Diego, USA
| | - Richard L Lieber
- Shirley Ryan AbilityLab and Northwestern University, Chicago, USA
| | - Robert H Mak
- Pediatric Nephrology, Rady Children's Hospital San Diego, University of California, San Diego, USA.
- Division of Pediatric Nephrology, Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, MC 0831, La Jolla, CA, 92093-0831, USA.
| |
Collapse
|
109
|
Nitrogen coordinated import and export of arginine across the yeast vacuolar membrane. PLoS Genet 2020; 16:e1008966. [PMID: 32776922 PMCID: PMC7440668 DOI: 10.1371/journal.pgen.1008966] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 08/20/2020] [Accepted: 06/30/2020] [Indexed: 11/19/2022] Open
Abstract
The vacuole of the yeast Saccharomyces cerevisiae plays an important role in nutrient storage. Arginine, in particular, accumulates in the vacuole of nitrogen-replete cells and is mobilized to the cytosol under nitrogen starvation. The arginine import and export systems involved remain poorly characterized, however. Furthermore, how their activity is coordinated by nitrogen remains unknown. Here we characterize Vsb1 as a novel vacuolar membrane protein of the APC (amino acid-polyamine-organocation) transporter superfamily which, in nitrogen-replete cells, is essential to active uptake and storage of arginine into the vacuole. A shift to nitrogen starvation causes apparent inhibition of Vsb1-dependent activity and mobilization of stored vacuolar arginine to the cytosol. We further show that this arginine export involves Ypq2, a vacuolar protein homologous to the human lysosomal cationic amino acid exporter PQLC2 and whose activity is detected only in nitrogen-starved cells. Our study unravels the main arginine import and export systems of the yeast vacuole and suggests that they are inversely regulated by nitrogen. The lysosome-like vacuole of the yeast Saccharomyces cerevisiae is an important storage compartment for diverse nutrients, including the cationic amino acid arginine, which accumulates at high concentrations in this organelle in nitrogen-replete cells. When these cells are transferred to a nitrogen-free medium, vacuolar arginine is mobilized to the cytosol, where it is used as an alternative nitrogen source to sustain growth. Although this phenomenon has been observed since the 1980s, the identity of the vacuolar transporters involved in the accumulation and the mobilization of arginine is not well established, and whether these processes are regulated according to nutritional cues remains unknown. In this study, we exploited in vitro and in vivo uptake assays in vacuoles to identify and characterize Vsb1 and Ypq2 as vacuolar membrane proteins mediating import and export of arginine, respectively. We further provide evidence that Vsb1 and Ypq2 are inversely regulated according to the nitrogen status of the cell. Our study sheds new light on the poorly studied topic of the diversity and metabolic control of vacuolar transporters. It also raises novel questions about the molecular mechanisms underlying their coordinated regulation and, by extension, the regulation of lysosomal transporters in human cells.
Collapse
|
110
|
Ewert A, Leifheit-Nestler M, Hohenfellner K, Büscher A, Kemper MJ, Oh J, Billing H, Thumfart J, Stangl G, Baur AC, Föller M, Feger M, Weber LT, Acham-Roschitz B, Arbeiter K, Tönshoff B, Zivicnjak M, Haffner D. Bone and Mineral Metabolism in Children with Nephropathic Cystinosis Compared with other CKD Entities. J Clin Endocrinol Metab 2020; 105:5837710. [PMID: 32413117 DOI: 10.1210/clinem/dgaa267] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 05/08/2020] [Indexed: 02/06/2023]
Abstract
CONTEXT Children with nephropathic cystinosis (NC) show persistent hypophosphatemia, due to Fanconi syndrome, as well as mineral and bone disorders related to chronic kidney disease (CKD); however, systematic analyses are lacking. OBJECTIVE To compare biochemical parameters of bone and mineral metabolism between children with NC and controls across all stages of CKD. DESIGN Cross-sectional multicenter study. SETTING Hospital clinics. PATIENTS Forty-nine children with NC, 80 CKD controls of the same age and CKD stage. MAIN OUTCOME MEASURES Fibroblast growth factor 23 (FGF23), soluble Klotho, bone alkaline phosphatase (BAP), tartrate-resistant acid phosphatase 5b (TRAP5b), sclerostin, osteoprotegerin (OPG), biochemical parameters related to mineral metabolism, and skeletal comorbidity. RESULTS Despite Fanconi syndrome medication, NC patients showed an 11-fold increased risk of short stature, bone deformities, and/or requirement for skeletal surgery compared with CKD controls. This was associated with a higher frequency of risk factors such as hypophosphatemia, hypocalcemia, low parathyroid hormone (PTH), metabolic acidosis, and a specific CKD stage-dependent pattern of bone marker alterations. Pretransplant NC patients in mild to moderate CKD showed a delayed increase or lacked an increase in FGF23 and sclerostin, and increased BAP, TRAP5b, and OPG concentrations compared with CKD controls. Post-transplant, BAP and OPG returned to normal, TRAP5b further increased, whereas FGF23 and PTH were less elevated compared with CKD controls and associated with higher serum phosphate. CONCLUSIONS Patients with NC show more severe skeletal comorbidity associated with distinct CKD stage-dependent alterations of bone metabolism than CKD controls, suggesting impaired mineralization and increased bone resorption, which is only partially normalized after renal transplantation.
Collapse
Affiliation(s)
- Annika Ewert
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School Children's Hospital, Hannover, Germany
| | - Maren Leifheit-Nestler
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School Children's Hospital, Hannover, Germany
| | | | - Anja Büscher
- Department of Pediatrics II, University Hospital Essen, Essen, Germany
| | | | - Jun Oh
- Division of Pediatric Nephrology, University Children's Hospital Hamburg, Hamburg, Germany
| | - Heiko Billing
- Division of Pediatric Nephrology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Julia Thumfart
- Department of Pediatric Gastroenterology, Nephrology and Metabolism, Charite Hospital, Berlin, Germany
| | - Gabriele Stangl
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Anja C Baur
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Michael Föller
- Institute of Physiology, University of Hohenheim, Stuttgart, Germany
| | - Martina Feger
- Institute of Physiology, University of Hohenheim, Stuttgart, Germany
| | - Lutz T Weber
- Division of Pediatric Nephrology, Children´s and Adolescents´ Hospital, University of Cologne, Faculty of Medicine and University Hospital, Cologne, Germany
| | | | - Klaus Arbeiter
- Division of Pediatric Nephrology and Gastroenterology, Medical University Vienna, Austria
| | - Burkhard Tönshoff
- Department of Pediatrics I, University Children's Hospital Heidelberg, Heidelberg, Germany
| | - Miroslav Zivicnjak
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School Children's Hospital, Hannover, Germany
| | - Dieter Haffner
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School Children's Hospital, Hannover, Germany
| |
Collapse
|
111
|
Topaloglu R, Keser AG, Gülhan B, Ozaltin F, Demir H, Çiftci T, Demir N, Temucin ÇM, Yuce A, Akhan O. Cystinosis beyond kidneys: gastrointestinal system and muscle involvement. BMC Gastroenterol 2020; 20:242. [PMID: 32727395 PMCID: PMC7392712 DOI: 10.1186/s12876-020-01385-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 07/15/2020] [Indexed: 11/10/2022] Open
Abstract
Background Cystinosis is a multisystemic disease resulting from cystine accumulation primarily in kidney and many other tissues. We intended to study the evolution of less commonly seen extrarenal complications of cystinosis in a group of patients who have periods without cysteamine treatment. Methods Gastrointestinal and muscular complications of cystinosis were studied in a group of 21 patients. Results Twenty one patients were included in the study. Among them, 14 were homozygous and 3 were compound heterozygous for CTNS mutations. The median age of diagnosis was 15 months (range; 5 months-14 years) and the mean age at last visit was 11.3 ± 6.5 years. Nine patients (42%) had end stage renal disease at a mean age of 10.6 years (6.5–17 years). Abdominal ultrasonography and portal vein doppler ultrasonography were performed in19 patients, 14 of them (74%) had hepatomegaly, 10 patients (53%) had granular pattern or heterogeneity of liver. Only one patient had high transaminase levels and liver biopsy showed cystine crystals in the liver. Eleven patients (58%) had borderline or increased portal vein minimum and maximum flow velocities. One patient had CK level of 9024 U/L and electromyographic study showed active myopathic involvement. Two patients were found to have gastroesaphageal reflux only and 4 patients were found to have esophageal remnants in addition to reflux. Conclusions In addition to renal functions, extrarenal organs may be affected from cystine accumulation even in childhood, especially in patients who are incompliant to treatment, resulting in complications such as swallowing difficulty, hepatomegaly and portal hypertension.
Collapse
Affiliation(s)
- Rezan Topaloglu
- Division of Pediatric Nephrology, Hacettepe University School of Medicine, 06100 Sıhhiye, Ankara, Turkey.
| | | | - Bora Gülhan
- Division of Pediatric Nephrology, Hacettepe University School of Medicine, 06100 Sıhhiye, Ankara, Turkey
| | - Fatih Ozaltin
- Division of Pediatric Nephrology, Hacettepe University School of Medicine, 06100 Sıhhiye, Ankara, Turkey.,Nephrogenetics Laboratory, Hacettepe University School of Medicine, Ankara, Turkey
| | - Hülya Demir
- Division of Pediatric Gastroenterology, Hacettepe University School of Medicine, Ankara, Turkey
| | - Türkmen Çiftci
- Department of Radiology, Hacettepe University School of Medicine, Ankara, Turkey
| | - Numan Demir
- Hacettepe University School of Physiotherapy, Ankara, Turkey
| | - Çağrı Mesut Temucin
- Department of Neurology, Hacettepe University School of Medicine, Ankara, Turkey
| | - Aysel Yuce
- Division of Pediatric Gastroenterology, Hacettepe University School of Medicine, Ankara, Turkey
| | - Okhan Akhan
- Department of Radiology, Hacettepe University School of Medicine, Ankara, Turkey
| |
Collapse
|
112
|
Amick J, Tharkeshwar AK, Talaia G, Ferguson SM. PQLC2 recruits the C9orf72 complex to lysosomes in response to cationic amino acid starvation. J Cell Biol 2020; 219:132798. [PMID: 31851326 PMCID: PMC7039192 DOI: 10.1083/jcb.201906076] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 10/07/2019] [Accepted: 10/25/2019] [Indexed: 12/13/2022] Open
Abstract
This study reveals that PQLC2, a lysosomal transporter of cationic amino acids, coordinates cellular responses to cationic amino acid availability via the regulated recruitment of a heterotrimeric protein complex containing C9orf72, SMCR8, and WDR41 to the surface of lysosomes. The C9orf72 protein is required for normal lysosome function. In support of such functions, C9orf72 forms a heterotrimeric complex with SMCR8 and WDR41 that is recruited to lysosomes when amino acids are scarce. These properties raise questions about the identity of the lysosomal binding partner of the C9orf72 complex and the amino acid–sensing mechanism that regulates C9orf72 complex abundance on lysosomes. We now demonstrate that an interaction with the lysosomal cationic amino acid transporter PQLC2 mediates C9orf72 complex recruitment to lysosomes. This is achieved through an interaction between PQLC2 and WDR41. The interaction between PQLC2 and the C9orf72 complex is negatively regulated by arginine, lysine, and histidine, the amino acids that PQLC2 transports across the membrane of lysosomes. These results define a new role for PQLC2 in the regulated recruitment of the C9orf72 complex to lysosomes and reveal a novel mechanism that allows cells to sense and respond to changes in the availability of cationic amino acids within lysosomes.
Collapse
Affiliation(s)
- Joseph Amick
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT.,Department of Neuroscience, Yale University School of Medicine, New Haven, CT.,Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT
| | - Arun Kumar Tharkeshwar
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT.,Department of Neuroscience, Yale University School of Medicine, New Haven, CT.,Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT
| | - Gabriel Talaia
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT.,Department of Neuroscience, Yale University School of Medicine, New Haven, CT.,Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT
| | - Shawn M Ferguson
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT.,Department of Neuroscience, Yale University School of Medicine, New Haven, CT.,Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
113
|
Zhang W, Li X, Wang S, Chen Y, Liu H. Regulation of TFEB activity and its potential as a therapeutic target against kidney diseases. Cell Death Discov 2020; 6:32. [PMID: 32377395 PMCID: PMC7195473 DOI: 10.1038/s41420-020-0265-4] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/20/2020] [Accepted: 04/09/2020] [Indexed: 12/24/2022] Open
Abstract
The transcription factor EB (TFEB) regulates the expression of target genes bearing the Coordinated Lysosomal Expression and Regulation (CLEAR) motif, thereby modulating autophagy and lysosomal biogenesis. Furthermore, TFEB can bind to the promoter of autophagy-associated genes and induce the formation of autophagosomes, autophagosome-lysosome fusion, and lysosomal cargo degradation. An increasing number of studies have shown that TFEB stimulates the intracellular clearance of pathogenic factors by enhancing autophagy and lysosomal function in multiple kidney diseases, such as cystinosis, acute kidney injury, and diabetic nephropathy. Taken together, this highlights the importance of developing novel therapeutic strategies against kidney diseases based on TFEB regulation. In this review, we present an overview of the current data on TFEB and its implication in kidney disease.
Collapse
Affiliation(s)
- Weihuang Zhang
- Institute of Nephrology, and Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Affiliated Hospital of Guangdong Medical University, 524001 Zhanjiang, Guangdong China
| | - Xiaoyu Li
- Institute of Nephrology, and Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Affiliated Hospital of Guangdong Medical University, 524001 Zhanjiang, Guangdong China
| | - Shujun Wang
- Institute of Nephrology, and Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Affiliated Hospital of Guangdong Medical University, 524001 Zhanjiang, Guangdong China
| | - Yanse Chen
- Institute of Nephrology, and Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Affiliated Hospital of Guangdong Medical University, 524001 Zhanjiang, Guangdong China
| | - Huafeng Liu
- Institute of Nephrology, and Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Affiliated Hospital of Guangdong Medical University, 524001 Zhanjiang, Guangdong China
| |
Collapse
|
114
|
Yarwood R, Hellicar J, Woodman PG, Lowe M. Membrane trafficking in health and disease. Dis Model Mech 2020; 13:13/4/dmm043448. [PMID: 32433026 PMCID: PMC7197876 DOI: 10.1242/dmm.043448] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Membrane trafficking pathways are essential for the viability and growth of cells, and play a major role in the interaction of cells with their environment. In this At a Glance article and accompanying poster, we outline the major cellular trafficking pathways and discuss how defects in the function of the molecular machinery that mediates this transport lead to various diseases in humans. We also briefly discuss possible therapeutic approaches that may be used in the future treatment of trafficking-based disorders. Summary: This At a Glance article and poster summarise the major intracellular membrane trafficking pathways and associated molecular machineries, and describe how defects in these give rise to disease in humans.
Collapse
Affiliation(s)
- Rebecca Yarwood
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - John Hellicar
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Philip G Woodman
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Martin Lowe
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| |
Collapse
|
115
|
Thoene JG, DelMonte MA, Mullet J. Microvesicle delivery of a lysosomal transport protein to ex vivo rabbit cornea. Mol Genet Metab Rep 2020; 23:100587. [PMID: 32280591 PMCID: PMC7138922 DOI: 10.1016/j.ymgmr.2020.100587] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/31/2020] [Accepted: 04/02/2020] [Indexed: 11/17/2022] Open
Abstract
Therapeutic use of transmembrane proteins is limited because of irreversible denaturation when away from their native lipid membrane. Mutations in lysosomal membrane transport proteins cause many lethal disorders including cystinosis which results from mutations in CTNS, which codes for the lysosomal cystine transport protein, cystinosin. Cystinosin-deficient fibroblasts, including keratocytes (corneal fibroblasts) accumulate lysosomal cystine. Cystinosis patients develop highly painful corneal cystine crystals, resulting in severe visually debilitating photophobia. The only available therapy is daily treatment with cysteamine eye drops. We have previously shown that microvesicles containing functional cystinosin are spontaneously produced by infecting Spodoptera frugiperda cells (Sf9) with baculovirus containing human wt CTNS. Infecting Sf9 cells for 3 days at a MOI of 1 yields 1011microvesicles /ml with a modal diameter of 90 nm. Addition of these vesicles to cultures of cystinotic fibroblasts produces cystine depletion over the course of 96 h, which persists for 2 weeks. In this paper we show that addition of such microvesicles containing cystinosinGFP to ex vivo rabbit ocular globes yields punctate perinuclear green fluorescence in the corneal keratocytes. These results support potential therapeutic use of these cystinosin containing microvesicles in treating cystinotic corneal keratopathy with the advantage of administering twice/month instead of daily topical administration.
Collapse
Affiliation(s)
- Jess G Thoene
- Department of Pediatrics, Division of Pediatric Genetics, Metabolism and Genomic Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Monte A DelMonte
- Department of Ophthalmology and Visual Sciences, Division of Pediatric Ophthalmology, Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA
| | - Jodi Mullet
- Department of Pediatrics, Division of Pediatric Genetics, Metabolism and Genomic Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
116
|
Cheung WW, Hao S, Wang Z, Ding W, Zheng R, Gonzalez A, Zhan J, Zhou P, Li S, Esparza MC, Hoffman HM, Lieber RL, Mak RH. Vitamin D repletion ameliorates adipose tissue browning and muscle wasting in infantile nephropathic cystinosis-associated cachexia. J Cachexia Sarcopenia Muscle 2020; 11:120-134. [PMID: 31721480 PMCID: PMC7015252 DOI: 10.1002/jcsm.12497] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 07/30/2019] [Accepted: 08/25/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Ctns-/- mice, a mouse model of infantile nephropathic cystinosis, exhibit hypermetabolism with adipose tissue browning and profound muscle wasting. Ctns-/- mice are 25(OH)D3 and 1,25(OH)2 D3 insufficient. We investigated whether vitamin D repletion could ameliorate adipose tissue browning and muscle wasting in Ctns-/- mice. METHODS Twelve-month-old Ctns-/- mice and wild-type controls were treated with 25(OH)D3 and 1,25(OH)2 D3 (75 μg/kg/day and 60 ng/kg/day, respectively) or an ethylene glycol vehicle for 6 weeks. Serum chemistry and parameters of energy homeostasis were measured. We quantitated total fat mass and studied expression of molecules regulating adipose tissue browning, energy metabolism, and inflammation. We measured lean mass content, skeletal muscle fibre size, in vivo muscle function (grip strength and rotarod activity), and expression of molecules regulating muscle metabolism. We also analysed the transcriptome of skeletal muscle in Ctns-/- mice using RNAseq. RESULTS Supplementation of 25(OH)D3 and 1,25(OH)2 D3 normalized serum concentration of 25(OH)D3 and 1,25(OH)2 D3 in Ctns-/- mice, respectively. Repletion of vitamin D partially or fully normalized food intake, weight gain, gain of fat, and lean mass, improved energy homeostasis, and attenuated perturbations of uncoupling proteins and adenosine triphosphate content in adipose tissue and muscle in Ctns-/- mice. Vitamin D repletion attenuated elevated expression of beige adipose cell biomarkers (UCP-1, CD137, Tmem26, and Tbx1) as well as aberrant expression of molecules implicated in adipose tissue browning (Cox2, Pgf2α, and NF-κB pathway) in inguinal white adipose tissue in Ctns-/- mice. Vitamin D repletion normalized skeletal muscle fibre size and improved in vivo muscle function in Ctns-/- mice. This was accompanied by correcting the increased muscle catabolic signalling (increased protein contents of IL-1β, IL-6, and TNF-α as well as an increased gene expression of Murf-2, atrogin-1, and myostatin) and promoting the decreased muscle regeneration and myogenesis process (decreased gene expression of Igf1, Pax7, and MyoD) in skeletal muscles of Ctns-/- mice. Muscle RNAseq analysis revealed aberrant gene expression profiles associated with reduced muscle and neuron regeneration, increased energy metabolism, and fibrosis in Ctns-/- mice. Importantly, repletion of 25(OH)D3 and 1,25(OH)2 D3 normalized the top 20 differentially expressed genes in Ctns-/- mice. CONCLUSIONS We report the novel findings that correction of 25(OH)D3 and 1,25(OH)2 D3 insufficiency reverses cachexia and may improve quality of life by restoring muscle function in an animal model of infantile nephropathic cystinosis. Mechanistically, vitamin D repletion attenuates adipose tissue browning and muscle wasting in Ctns-/- mice via multiple cellular and molecular mechanisms.
Collapse
Affiliation(s)
- Wai W. Cheung
- Pediatric NephrologyRady Children's Hospital—San Diego, University of California, San DiegoSan DiegoCAUSA
| | - Sheng Hao
- Department of Nephrology and RheumatologyShanghai Children's Hospital, Shanghai Jiao Tong UniversityShanghaiChina
| | - Zhen Wang
- Department of PediatricsShanghai General Hospital, Shanghai Jiao Tong UniversityShanghaiChina
| | - Wei Ding
- Division of NephrologyShanghai 9th People's Hospital, Shanghai Jiao Tong UniversityShanghaiChina
| | - Ronghao Zheng
- Department of Pediatric Nephrology, Rheumatology, and ImmunologyMaternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Alex Gonzalez
- Pediatric NephrologyRady Children's Hospital—San Diego, University of California, San DiegoSan DiegoCAUSA
| | | | - Ping Zhou
- Department of PediatricsThe 2 Hospital of Harbin Medical UniversityHarbinChina
| | - Shiping Li
- College of Bioscience and BiotechnologyYangzhou UniversityYangzhouChina
| | - Mary C. Esparza
- Department of Orthopedic SurgeryUniversity of California, San DiegoSan DiegoCAUSA
| | - Hal M. Hoffman
- Department of PediatricsUniversity of California, San DiegoSan DiegoCAUSA
| | - Richard L. Lieber
- Department of Orthopedic SurgeryUniversity of California, San DiegoSan DiegoCAUSA
- Rehabilitation Institute of ChicagoChicagoILUSA
| | - Robert H. Mak
- Pediatric NephrologyRady Children's Hospital—San Diego, University of California, San DiegoSan DiegoCAUSA
| |
Collapse
|
117
|
Francisco AA, Foxe JJ, Horsthuis DJ, Molholm S. Impaired auditory sensory memory in Cystinosis despite typical sensory processing: A high-density electrical mapping study of the mismatch negativity (MMN). NEUROIMAGE-CLINICAL 2020; 25:102170. [PMID: 31954986 PMCID: PMC6965721 DOI: 10.1016/j.nicl.2020.102170] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 12/13/2019] [Accepted: 01/10/2020] [Indexed: 11/24/2022]
Abstract
Children and adolescents with Cystinosis show similar N1 responses to their age-matched peers. Children and adolescents with Cystinosis show reduced MMNs for longer SOAs. Our results suggest typical auditory processing, but impaired sensory memory in Cystinosis.
Cystinosis, a genetic rare disease characterized by cystine accumulation and crystallization, results in significant damage in a multitude of tissues and organs, such as the kidney, thyroid, eye, and brain. While Cystinosis’ impact on brain function is relatively mild compared to its effects on other organs, the increased lifespan of this population and thus potential for productive societal contributions have led to increased interest on the effects on brain function. Nevertheless, and despite some evidence of structural brain differences, the neural impact of the mutation is still not well characterized. Here, using a passive duration oddball paradigm (with different stimulus onset asynchronies (SOAs), representing different levels of demand on memory) and high-density electrophysiology, we tested basic auditory processing in a group of 22 children and adolescents diagnosed with Cystinosis (age range: 6-17 years old) and in neurotypical age-matched controls (N = 24). We examined whether the N1 and mismatch negativity (MMN) significantly differed between the groups and if those neural measures correlated with verbal and non-verbal IQ. Individuals diagnosed with Cystinosis presented similar N1 responses to their age-matched peers, indicating typical basic auditory processing in this population. However, whereas both groups showed similar MMN responses for the shortest (450 ms) SOA, suggesting intact change detection and sensory memory, individuals diagnosed with Cystinosis presented clearly reduced responses for the longer (900 ms and 1800 ms) SOAs. This could indicate reduced duration auditory sensory memory traces, and thus sensory memory impairment, in children and adolescents diagnosed with Cystinosis. Future work addressing other aspects of sensory and working memory is needed to understand the underlying bases of the differences described here, and their implication for higher order processing.
Collapse
Affiliation(s)
- Ana A Francisco
- The Cognitive Neurophysiology Laboratory, Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York, USA.; Department of Neuroscience, Rose F. Kennedy Center, Albert Einstein College of Medicine, Bronx, New York, USA..
| | - John J Foxe
- The Cognitive Neurophysiology Laboratory, Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York, USA.; Department of Neuroscience, Rose F. Kennedy Center, Albert Einstein College of Medicine, Bronx, New York, USA.; The Cognitive Neurophysiology Laboratory, Department of Neuroscience, The Ernest J. Del Monde Institute for Neuroscience, School of Medicine and Dentistry, University of Rochester, Rochester, New York, USA
| | - Douwe J Horsthuis
- The Cognitive Neurophysiology Laboratory, Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Sophie Molholm
- The Cognitive Neurophysiology Laboratory, Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York, USA.; Department of Neuroscience, Rose F. Kennedy Center, Albert Einstein College of Medicine, Bronx, New York, USA.; The Cognitive Neurophysiology Laboratory, Department of Neuroscience, The Ernest J. Del Monde Institute for Neuroscience, School of Medicine and Dentistry, University of Rochester, Rochester, New York, USA..
| |
Collapse
|
118
|
Neves ACDO, Galván I, Van den Abeele D. Impairment of mixed melanin-based pigmentation in parrots. J Exp Biol 2020:jeb.225912. [PMID: 34005594 DOI: 10.1242/jeb.225912] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/05/2020] [Indexed: 11/20/2022]
Abstract
Parrots and allies (Order Psittaciformes) have evolved an exclusive capacity to synthesize polyene pigments called psittacofulvins at feather follicles, which allows them to produce a striking diversity of pigmentation phenotypes. Melanins are polymers constituting the most abundant pigments in animals, and the sulphurated form (pheomelanin) produces colors that are similar to those produced by psittacofulvins. However, the differential contribution of these pigments to psittaciform phenotypic diversity has not been investigated. Given the color redundancy, and physiological limitations associated to pheomelanin synthesis, we hypothesized that the latter would be avoided by psittaciform birds. Here we test this by using Raman spectroscopy to identify pigments in feathers exhibiting colors suspicious of being produced by pheomelanin (i.e., dull red, yellow and grey- and green-brownish) in 26 species from the three main lineages of Psittaciformes. We detected the non-sulphurated melanin form (eumelanin) in black, grey and brown plumage patches, and psittacofulvins in red, yellow and green patches, but no evidence of pheomelanin. As natural melanins are assumed to be composed of eumelanin and pheomelanin in varying ratios, our results represent the first report of impairment of mixed melanin-based pigmentation in animals. Given that psittaciforms also avoid the uptake of circulating carotenoid pigments, these birds seem to have evolved a capacity to avoid functional redundancy between pigments, likely by regulating follicular gene expression. Ours study provides the first vibrational characterization of different psittacofulvin-based colors and thus helps to determine the relative polyene chain length in these pigments, which is related to their antireductant protection activity.
Collapse
Affiliation(s)
| | - Ismael Galván
- Department of Evolutionary Ecology, Doñana Biological Station, CSIC, Sevilla, Spain
| | | |
Collapse
|
119
|
De Rasmo D, Signorile A, De Leo E, Polishchuk EV, Ferretta A, Raso R, Russo S, Polishchuk R, Emma F, Bellomo F. Mitochondrial Dynamics of Proximal Tubular Epithelial Cells in Nephropathic Cystinosis. Int J Mol Sci 2019; 21:ijms21010192. [PMID: 31888107 PMCID: PMC6982165 DOI: 10.3390/ijms21010192] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/20/2019] [Accepted: 12/24/2019] [Indexed: 12/27/2022] Open
Abstract
Nephropathic cystinosis is a rare lysosomal storage disorder caused by mutations in CTNS gene leading to Fanconi syndrome. Independent studies reported defective clearance of damaged mitochondria and mitochondrial fragmentation in cystinosis. Proteins involved in the mitochondrial dynamics and the mitochondrial ultrastructure were analyzed in CTNS-/- cells treated with cysteamine, the only drug currently used in the therapy for cystinosis but ineffective to treat Fanconi syndrome. CTNS-/- cells showed an overexpression of parkin associated with deregulation of ubiquitination of mitofusin 2 and fission 1 proteins, an altered proteolytic processing of optic atrophy 1 (OPA1), and a decreased OPA1 oligomerization. According to molecular findings, the analysis of electron microscopy images showed a decrease of mitochondrial cristae number and an increase of cristae lumen and cristae junction width. Cysteamine treatment restored the fission 1 ubiquitination, the mitochondrial size, number and lumen of cristae, but had no effect on cristae junction width, making CTNS-/- tubular cells more susceptible to apoptotic stimuli.
Collapse
Affiliation(s)
- Domenico De Rasmo
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnology (IBIOM), National Research Council (CNR), 70124 Bari, Italy;
- Correspondence: (D.D.R.); (F.B.); Tel.: +39-080-5448516 (D.D.R.); +39-06-68592997 (F.B)
| | - Anna Signorile
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.S.); (S.R.)
| | - Ester De Leo
- Renal Diseases Research Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital—IRCCS, 00146 Rome, Italy; (E.D.L.); (R.R.)
| | - Elena V. Polishchuk
- Telethon Institute of Genetics and Medicine, 80078 Pozzuoli, Italy; (E.V.P.); (R.P.)
| | - Anna Ferretta
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnology (IBIOM), National Research Council (CNR), 70124 Bari, Italy;
| | - Roberto Raso
- Renal Diseases Research Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital—IRCCS, 00146 Rome, Italy; (E.D.L.); (R.R.)
| | - Silvia Russo
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.S.); (S.R.)
| | - Roman Polishchuk
- Telethon Institute of Genetics and Medicine, 80078 Pozzuoli, Italy; (E.V.P.); (R.P.)
| | - Francesco Emma
- Division of Nephrology, Department of Pediatric Subspecialties, Bambino Gesù Children’s Hospital—IRCCS, 00165 Rome, Italy;
| | - Francesco Bellomo
- Renal Diseases Research Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital—IRCCS, 00146 Rome, Italy; (E.D.L.); (R.R.)
- Correspondence: (D.D.R.); (F.B.); Tel.: +39-080-5448516 (D.D.R.); +39-06-68592997 (F.B)
| |
Collapse
|
120
|
Paul BD, Snyder SH. Therapeutic Applications of Cysteamine and Cystamine in Neurodegenerative and Neuropsychiatric Diseases. Front Neurol 2019; 10:1315. [PMID: 31920936 PMCID: PMC6920251 DOI: 10.3389/fneur.2019.01315] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 11/27/2019] [Indexed: 12/31/2022] Open
Abstract
Current medications for neurodegenerative and neuropsychiatric diseases such as Alzheimer's disease (AD), Huntington's disease (HD), Parkinson's disease (PD), and Schizophrenia mainly target disease symptoms. Thus, there is an urgent need to develop novel therapeutics that can delay, halt or reverse disease progression. AD, HD, PD, and schizophrenia are characterized by elevated oxidative and nitrosative stress, which play a central role in pathogenesis. Clinical trials utilizing antioxidants to counter disease progression have largely been unsuccessful. Most antioxidants are relatively non-specific and do not adequately target neuroprotective pathways. Accordingly, a search for agents that restore redox balance as well as halt or reverse neuronal loss is underway. The small molecules, cysteamine, the decarboxylated derivative of the amino acid cysteine, and cystamine, the oxidized form of cysteamine, respectively, mitigate oxidative stress and inflammation and upregulate neuroprotective pathways involving brain-derived neurotrophic factor (BDNF) and Nuclear factor erythroid 2-related factor 2 (Nrf2) signaling. Cysteamine can traverse the blood brain barrier, a desirable characteristic of drugs targeting neurodegeneration. This review addresses recent developments in the use of these aminothiols to counter neurodegeneration and neuropsychiatric deficits.
Collapse
Affiliation(s)
- Bindu D Paul
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Solomon H Snyder
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
121
|
Brasell EJ, Chu LL, Akpa MM, Eshkar-Oren I, Alroy I, Corsini R, Gilfix BM, Yamanaka Y, Huertas P, Goodyer P. The novel aminoglycoside, ELX-02, permits CTNSW138X translational read-through and restores lysosomal cystine efflux in cystinosis. PLoS One 2019; 14:e0223954. [PMID: 31800572 PMCID: PMC6892560 DOI: 10.1371/journal.pone.0223954] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 10/02/2019] [Indexed: 12/19/2022] Open
Abstract
Background Cystinosis is a rare disorder caused by recessive mutations of the CTNS gene. Current therapy decreases cystine accumulation, thus slowing organ deterioration without reversing renal Fanconi syndrome or preventing eventual need for a kidney transplant.15-20% of cystinosis patients harbour at least one nonsense mutation in CTNS, leading to premature end of translation of the transcript. Aminoglycosides have been shown to permit translational read-through but have high toxicity level, especially in the kidney and inner ear. ELX-02, a modified aminoglycoside, retains it read-through ability without the toxicity. Methods and findings We ascertained the toxicity of ELX-02 in cells and in mice as well as the effect of ELX-02 on translational read-through of nonsense mutations in cystinotic mice and human cells. ELX-02 was not toxic in vitro or in vivo, and permitted read-through of nonsense mutations in cystinotic mice and human cells. Conclusions ELX-02 has translational read-through activity and produces a functional CTNS protein, as evidenced by reduced cystine accumulation. This reduction is comparable to cysteamine treatment. ELX-02 accumulates in the kidney but neither cytotoxicity nor nephrotoxicity was observed.
Collapse
Affiliation(s)
- Emma J. Brasell
- McGill University, Department of Human Genetics, Montreal, Canada
| | - Lee Lee Chu
- Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Murielle M. Akpa
- Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Idit Eshkar-Oren
- McGill University, Department of Experimental Medicine, Montreal, Canada
| | - Iris Alroy
- McGill University, Department of Experimental Medicine, Montreal, Canada
| | - Rachel Corsini
- Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Brian M. Gilfix
- Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Yojiro Yamanaka
- McGill University, Department of Human Genetics, Montreal, Canada
| | - Pedro Huertas
- McGill University, Department of Experimental Medicine, Montreal, Canada
| | - Paul Goodyer
- McGill University, Department of Human Genetics, Montreal, Canada
- Research Institute of the McGill University Health Centre, Montreal, Canada
- Montreal Children’s Hospital, Department of Nephrology, Montreal, Canada
- Eloxx Pharmaceuticals, Inc., Waltham, United States of America
- * E-mail:
| |
Collapse
|
122
|
Hohenfellner K, Bergmann C, Fleige T, Janzen N, Burggraf S, Olgemöller B, Gahl WA, Czibere L, Froschauer S, Röschinger W, Vill K, Harms E, Nennstiel U. Molecular based newborn screening in Germany: Follow-up for cystinosis. Mol Genet Metab Rep 2019; 21:100514. [PMID: 31641587 PMCID: PMC6796768 DOI: 10.1016/j.ymgmr.2019.100514] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Accepted: 08/29/2019] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Newborn screening (NBS) programs for treatable metabolic disorders have been enormously successful, but molecular-based screening has not been broadly implemented so far. METHODS This prospective pilot study was performed within the German NBS framework. DNA, extracted from dried blood cards was collected as part of the regular NBS program. As cystinosis has a prevalence of only 1:100,000-1:200,000, a molecular genetic assay for detection of the SMN1 gene mutation with a higher prevalence was also included in the screening process, a genetic defect that leads to spinal muscular atrophy (SMA). First tier multiplex PCR was employed for both diseases. The cystinosis screening employed assays for the three most common CTNS mutations covering 75% of German patients; in case of heterozygosity for one of these mutations, samples were screened by next generation sequencing (NGS) of the CTNS exons for 101 CTNS mutations. A detection rate of 98.5% is predicted using this approach. RESULTS Between January 15, 2018 and May 31, 2019, 257,734 newborns were screened in Germany for cystinosis. One neonate was diagnosed with cystinosis, consistent with the known incidence of the disease. No false positive or false negatives were detected so far. Screening, communication of findings to parents, and confirmation of diagnosis were accomplished in a multi-disciplinary setting. This program was accomplished with the cooperation of hospitals, physicians, and parents. In the neonate diagnosed with cystinosis, oral cysteamine treatment began on day 18. After 16 months of treatment the child has no clinical signs of renal tubular Fanconi syndrome. CONCLUSIONS This pilot study demonstrates the efficacy of a molecular-based neonatal screening program for cystinosis using an existing national screening framework.
Collapse
Affiliation(s)
- Katharina Hohenfellner
- Department of Pediatrics, Pediatric Nephrology, RoMed Kliniken, Pettenkoferstr. 10, Rosenheim 83022, Germany
| | - Carsten Bergmann
- Department of Medicine, University Hospital Freiburg, Hugstetter Str. 55, Freiburg 79106, Germany
| | - Tobias Fleige
- Laboratory Becker & Colleagues, Führichstr. 70, Munich 81671, Germany
| | - Nils Janzen
- Screening-Laboratory Hannover, Am Steinweg 11A/13B, Ronnenberg 30952, Germany
- Department of Clinical Chemistry, Hannover Medical School, Carl-Neuberg-str.1, Hannover 30625, Germany
| | | | - Bernd Olgemöller
- Formally Laboratory Becker, Olgemöller & Colleagues, Führichstr. 70, Munich 81671, Germany
| | | | - Ludwig Czibere
- Laboratory Becker & Colleagues, Führichstr. 70, Munich 81671, Germany
| | - Sonja Froschauer
- Cystinosis Foundation, Germany c/o Haus des Stiftens gGmbH, Landshuter Allee 11, Munich 80637, Germany
| | - Wulf Röschinger
- Laboratory Becker & Colleagues, Führichstr. 70, Munich 81671, Germany
| | - Katharina Vill
- Dr. v. Hauner Children's Hospital, Department of Pediatric Neurology and Developmental Medicine, LMU – University of Munich, Lindwurmstrasse 4, Munich 80337, Germany
| | - Erik Harms
- Formally University Hospital Münster, Children's Hospital, Albert-Schweitzer-Campus 1, Münster 48149, Germany
| | - Uta Nennstiel
- Screening Center, Bavarian Health and Food Safety Authority (LGL), Veterinaerstrasse 2, 85764 Oberschleissheim, Germany
| |
Collapse
|
123
|
Papizh S, Serzhanova V, Filatova A, Skoblov M, Tabakov V, van den Heuvel L, Levtchenko E, Prikhodina L. CTNS mRNA molecular analysis revealed a novel mutation in a child with infantile nephropathic cystinosis: a case report. BMC Nephrol 2019; 20:400. [PMID: 31672123 PMCID: PMC6822415 DOI: 10.1186/s12882-019-1589-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 10/13/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cystinosis is an autosomal recessive lysosomal storage disorder characterized by accumulation of cystine in lysosomes throughout the body. Cystinosis is caused by mutations in the CTNS gene that encodes the lysosomal cystine carrier protein cystinosin. CTNS mutations result in either complete absence or reduced cystine transporting function of the protein. The diagnosis of nephropathic cystinosis is generally based on measuring leukocyte cystine level, demonstration of corneal cystine crystals by the slit lamp examination and confirmed by genetic analysis of the CTNS gene. CASE PRESENTATION A boy born to consanguineous Caucasian parents had the characteristic clinical features of the infantile nephropathic cystinosis including renal Fanconi syndrome (polydipsia/polyuria, metabolic acidosis, hypokalemia, hypophosphatemia, low molecular weight proteinuria, glycosuria, cystine crystals in the cornea) and elevated WBC cystine levels. Initially we performed RFLP analysis of the common in the Northern European population 57-kb deletion of proband's DNA, then a direct Sanger sequencing which revealed no mutations in the coding part of the CTNS gene. To confirm the diagnosis we performed RT-PCR analysis of total RNA obtained from patient-derived fibroblasts in combination with cDNA sequencing. This revealed the skipping of exon 4 and exon 5 in the CTNS in our patient. Therefore, we detected a novel 9-kb homozygous deletion in the CTNS gene at genomic DNA level, spanning region from intron 3 to intron 5. In order to identify the inheritance pattern of the deletion we analyzed DNA of proband's mother and father. Both parents were found to be heterozygous carriers of the CTNS mutation. CONCLUSIONS Analysis of CTNS gene transcript allowed to identify a large homozygous deletion in the patient with infantile nephropathic cystinosis. Mutational detection at RNA level may be an efficient tool to establish the genetic defect in some cystinosis patients.
Collapse
Affiliation(s)
- Svetlana Papizh
- Department of hereditary and acquired kidney diseases, Research and Clinical Institute for Pediatrics at the Pirogov Russian National Research Medical University, 125412, Taldomskaya st., 2, Moscow, Russia.
| | | | | | - Mikhail Skoblov
- Research Centre for Medical Genetics, 115522, Russia, Moscow
| | | | - Lambert van den Heuvel
- Department of Pediatrics/Pediatric Nephrology, University Hospital Gasthuisberg, Leuven, Belgium
| | - Elena Levtchenko
- Department of Pediatrics/Pediatric Nephrology, University Hospital Gasthuisberg, Leuven, Belgium
| | - Larisa Prikhodina
- Department of hereditary and acquired kidney diseases, Research and Clinical Institute for Pediatrics at the Pirogov Russian National Research Medical University, 125412, Taldomskaya st., 2, Moscow, Russia
| |
Collapse
|
124
|
van der Wijst J, Belge H, Bindels RJM, Devuyst O. Learning Physiology From Inherited Kidney Disorders. Physiol Rev 2019; 99:1575-1653. [PMID: 31215303 DOI: 10.1152/physrev.00008.2018] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The identification of genes causing inherited kidney diseases yielded crucial insights in the molecular basis of disease and improved our understanding of physiological processes that operate in the kidney. Monogenic kidney disorders are caused by mutations in genes coding for a large variety of proteins including receptors, channels and transporters, enzymes, transcription factors, and structural components, operating in specialized cell types that perform highly regulated homeostatic functions. Common variants in some of these genes are also associated with complex traits, as evidenced by genome-wide association studies in the general population. In this review, we discuss how the molecular genetics of inherited disorders affecting different tubular segments of the nephron improved our understanding of various transport processes and of their involvement in homeostasis, while providing novel therapeutic targets. These include inherited disorders causing a dysfunction of the proximal tubule (renal Fanconi syndrome), with emphasis on epithelial differentiation and receptor-mediated endocytosis, or affecting the reabsorption of glucose, the handling of uric acid, and the reabsorption of sodium, calcium, and magnesium along the kidney tubule.
Collapse
Affiliation(s)
- Jenny van der Wijst
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; Institute of Physiology, University of Zurich , Zurich , Switzerland ; and Division of Nephrology, Institute of Experimental and Clinical Research (IREC), Medical School, Université catholique de Louvain, Brussels, Belgium
| | - Hendrica Belge
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; Institute of Physiology, University of Zurich , Zurich , Switzerland ; and Division of Nephrology, Institute of Experimental and Clinical Research (IREC), Medical School, Université catholique de Louvain, Brussels, Belgium
| | - René J M Bindels
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; Institute of Physiology, University of Zurich , Zurich , Switzerland ; and Division of Nephrology, Institute of Experimental and Clinical Research (IREC), Medical School, Université catholique de Louvain, Brussels, Belgium
| | - Olivier Devuyst
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; Institute of Physiology, University of Zurich , Zurich , Switzerland ; and Division of Nephrology, Institute of Experimental and Clinical Research (IREC), Medical School, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
125
|
Hohenfellner K, Rauch F, Ariceta G, Awan A, Bacchetta J, Bergmann C, Bechtold S, Cassidy N, Deschenes G, Elenberg E, Gahl WA, Greil O, Harms E, Herzig N, Hoppe B, Koeppl C, Lewis MA, Levtchenko E, Nesterova G, Santos F, Schlingmann KP, Servais A, Soliman NA, Steidle G, Sweeney C, Treikauskas U, Topaloglu R, Tsygin A, Veys K, v. Vigier R, Zustin J, Haffner D. Management of bone disease in cystinosis: Statement from an international conference. J Inherit Metab Dis 2019; 42:1019-1029. [PMID: 31177550 PMCID: PMC7379238 DOI: 10.1002/jimd.12134] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/02/2019] [Accepted: 06/03/2019] [Indexed: 01/10/2023]
Abstract
Cystinosis is an autosomal recessive storage disease due to impaired transport of cystine out of lysosomes. Since the accumulation of intracellular cystine affects all organs and tissues, the management of cystinosis requires a specialized multidisciplinary team consisting of pediatricians, nephrologists, nutritionists, ophthalmologists, endocrinologists, neurologists' geneticists, and orthopedic surgeons. Treatment with cysteamine can delay or prevent most clinical manifestations of cystinosis, except the renal Fanconi syndrome. Virtually all individuals with classical, nephropathic cystinosis suffer from cystinosis metabolic bone disease (CMBD), related to the renal Fanconi syndrome in infancy and progressive chronic kidney disease (CKD) later in life. Manifestations of CMBD include hypophosphatemic rickets in infancy, and renal osteodystrophy associated with CKD resulting in bone deformities, osteomalacia, osteoporosis, fractures, and short stature. Assessment of CMBD involves monitoring growth, leg deformities, blood levels of phosphate, electrolytes, bicarbonate, calcium, and alkaline phosphatase, periodically obtaining bone radiographs, determining levels of critical hormones and vitamins, such as thyroid hormone, parathyroid hormone, 25(OH) vitamin D, and testosterone in males, and surveillance for nonrenal complications of cystinosis such as myopathy. Treatment includes replacement of urinary losses, cystine depletion with oral cysteamine, vitamin D, hormone replacement, physical therapy, and corrective orthopedic surgery. The recommendations in this article came from an expert meeting on CMBD that took place in Salzburg, Austria, in December 2016.
Collapse
Affiliation(s)
| | - Frank Rauch
- Shriners Hospital for Children, McGill UniversityMontrealCanada
| | - Gema Ariceta
- Service of Pediatric NephrologyUniversity Hospital Vall d’ HebronBarcelonaSpain
| | - Atif Awan
- Department of NephrologyChildren's University HospitalDublinIreland
| | - Justine Bacchetta
- Référence Center for Rare Renal DiseasesHôpital Femme‐Mère‐EnfantBronFrance
| | - Carsten Bergmann
- Department of MedicineUniversity Hospital FreiburgFreiburgGermany
| | - Susanne Bechtold
- Division of Pediatric EndocrinologyChildren's Hospital and Polyclinic iSPZ, Dr. v. Haunerschen Kinderspital, University Hospital MunichMunichGermany
| | - Noelle Cassidy
- Department of Orthopaedic SurgeryChildren's University HospitalDublinIreland
| | - Geroges Deschenes
- Department of Pediatric NephrologyHôpital Robert‐Debré and University of Paris DiderotParisFrance
| | - Ewa Elenberg
- Department of PediatricsBaylor College of Medicine and Texas Children's HospitalHoustonTexas
| | - William A. Gahl
- National Human Genome Research InstituteNational Institutes of Health Undiagnosed Diseases ProgramBethesdaMaryland
| | - Oliver Greil
- Department of Diagnostic and Interventional RadiologyKlinikum TraunsteinTraunsteinGermany
| | - Erik Harms
- Children‘s University Hospital MuensterMuensterGermany
| | - Nadine Herzig
- Schoen Clinic Munich HarlachingSpecialist Centre for Paediatric and Neuro‐OrthopaedicsMunichGermany
| | - Bernd Hoppe
- Division of Pediatric NephrologyUniversity Children's HospitalBonnGermany
| | - Christian Koeppl
- Kliniken Südostbayern AG, Sozialpädiatrisches ZentrumTraunsteinGermany
| | - Malcolm A. Lewis
- Department of NephrologyChildren's University HospitalDublinIreland
| | - Elena Levtchenko
- Department of Pediatrics & Development and RegenerationUniversity Hospitals Leuven & Katholieke Universiteit LeuvenLeuvenBelgium
| | - Galina Nesterova
- National Institutes of Health, National Human Genome Research Institute (NHGRI)BethesdaMaryland
| | - Fernando Santos
- Hospital Universitario Central de AsturiasPediatríaOviedoSpain
| | - Karl P. Schlingmann
- Department of General PediatricsUniversity Children's Hospital MünsterMünsterGermany
| | - Aude Servais
- Reference Center of Inherited Metabolic Diseases, Nephrology Unit, Hospital Necker Enfants Malades, APHPUniversity Paris DescartesParisFrance
| | - Neveen A. Soliman
- Department of Pediatrics, Center of Pediatric Nephrology and Transplantation (CPNT), Kasr Al Ainy Faculty of MedicineCairo UniversityCairoEgypt
| | - Guenther Steidle
- Kliniken Südostbayern AG, Sozialpädiatrisches ZentrumTraunsteinGermany
| | - Clodagh Sweeney
- Department of NephrologyChildren's University HospitalDublinIreland
| | - Ulrike Treikauskas
- Department of Pediatrics, Department of Pediatric NephrologyRo‐Med KlinikenRosenheimGermany
| | - Rezan Topaloglu
- Department of Pediatric NephrologyHacettepe University Faculty of MedicineAnkaraTurkey
| | - Alexey Tsygin
- Department of NephrologyNational Medical and Research Center for Children's HealthMoscowRussia
| | - Koenraad Veys
- Department of Pediatrics & Development and RegenerationUniversity Hospitals Leuven & Katholieke Universiteit LeuvenLeuvenBelgium
| | - Rodo v. Vigier
- Pediatric ClinicWildermeth Children's HospitalBiel‐BienneSwitzerland
| | - Jozef Zustin
- Institute of Osteology and BiomechanicsUniversity Medical Center Hamburg‐Eppendorf, University of HamburgHamburgGermany
| | - Dieter Haffner
- Department of Pediatric Kidney, Liver and Metabolic DiseasesHannover Medical SchoolHannoverGermany
| |
Collapse
|
126
|
Rodríguez-Martínez S, Galván I. Exposure to a competitive social environment activates an epigenetic mechanism that limits pheomelanin synthesis in zebra finches. Mol Ecol 2019; 28:3698-3708. [PMID: 31290207 DOI: 10.1111/mec.15171] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/07/2019] [Accepted: 06/17/2019] [Indexed: 12/17/2022]
Abstract
Competitive environments promote high testosterone levels, produce oxidative stress and, consequently, impair cellular homeostasis. The regulation of genes involved in the synthesis of the pigment pheomelanin in melanocytes seems to help to maintain homeostasis against environmental oxidative stress. Here, we experimentally increased social interactions in some zebra finch (Taeniopygia guttata) males by keeping them in groups of six birds during feather growth, while others were kept alone, to test if melanocytes show epigenetic lability under a competitive social environment. As these changes may depend on the oxidative status, we administrated buthionine sulfoximine (BSO) to decrease the antioxidant capacity of some birds. The competitive environment downregulated a gene involved in pheomelanin synthesis (Slc7a11) by changing the level of DNA methylation in feather melanocytes. In other genes involved in pheomelanin synthesis (Slc45a2, MC1R and AGRP), DNA methylation was also affected, but no changes in expression were detected. Exposure to the competitive environment did not affect systemic oxidative stress and damage, indicating that a protective epigenetic mechanism that changes the expression of Slc7a11 may have been activated. However, no changes to the pigmentation phenotype of birds were found, probably due to the short duration or low intensity of the competitive environment. BSO treatment did not affect the epigenetic mechanism, suggesting that the antioxidant capacity of birds was high enough to deal with the competitive environment. An epigenetic mechanism limiting pheomelanin synthesis therefore becomes activated under exposure to a competitive environment in male zebra finches, which may help to avoid damage caused by competitive interactions.
Collapse
Affiliation(s)
| | - Ismael Galván
- Departamento de Ecología Evolutiva, Estación Biológica de Doñana, CSIC, Sevilla, Spain
| |
Collapse
|
127
|
Rocca CJ, Cherqui S. Potential use of stem cells as a therapy for cystinosis. Pediatr Nephrol 2019; 34:965-973. [PMID: 29789935 PMCID: PMC6250595 DOI: 10.1007/s00467-018-3974-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 04/26/2018] [Accepted: 04/30/2018] [Indexed: 01/02/2023]
Abstract
Cystinosis is an autosomal recessive metabolic disease that belongs to the family of lysosomal storage disorders (LSDs). Initial symptoms of cystinosis correspond to the renal Fanconi syndrome. Patients then develop chronic kidney disease and multi-organ failure due to accumulation of cystine in all tissue compartments. LSDs are commonly characterized by a defective activity of lysosomal enzymes. Hematopoietic stem and progenitor cell (HSPC) transplantation is a treatment option for several LSDs based on the premise that their progeny will integrate in the affected tissues and secrete the functional enzyme, which will be recaptured by the surrounding deficient cells and restore physiological activity. However, in the case of cystinosis, the defective protein is a transmembrane lysosomal protein, cystinosin. Thus, cystinosin cannot be secreted, and yet, we showed that HSPC transplantation can rescue disease phenotype in the mouse model of cystinosis. In this review, we are describing a different mechanism by which HSPC-derived cells provide cystinosin to diseased cells within tissues, and how HSPC transplantation could be an effective one-time treatment to treat cystinosis but also other LSDs associated with a lysosomal transmembrane protein dysfunction.
Collapse
Affiliation(s)
- Celine J Rocca
- Department of Pediatrics, Division of Genetics, University of California, 9500 Gilman Drive, MC 0734, La Jolla, San Diego, CA, 92093-0734, USA
| | - Stephanie Cherqui
- Department of Pediatrics, Division of Genetics, University of California, 9500 Gilman Drive, MC 0734, La Jolla, San Diego, CA, 92093-0734, USA.
| |
Collapse
|
128
|
Brasell EJ, Chu L, El Kares R, Seo JH, Loesch R, Iglesias DM, Goodyer P. The aminoglycoside geneticin permits translational readthrough of the CTNS W138X nonsense mutation in fibroblasts from patients with nephropathic cystinosis. Pediatr Nephrol 2019; 34:873-881. [PMID: 30413946 DOI: 10.1007/s00467-018-4094-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/13/2018] [Accepted: 09/19/2018] [Indexed: 12/24/2022]
Abstract
BACKGROUND Cystinosis is an ultrarare disorder caused by mutations of the cystinosin (CTNS) gene, encoding a cystine-selective efflux channel in the lysosomes of all cells of the body. Oral therapy with cysteamine reduces intralysosomal cystine accumulation and slows organ deterioration but cannot reverse renal Fanconi syndrome nor prevent the eventual need for renal transplantation. A definitive therapeutic remains elusive. About 15% of cystinosis patients worldwide carry one or more nonsense mutations that halt translation of the CTNS protein. Aminoglycosides such as geneticin (G418) can bind to the mammalian ribosome, relax translational fidelity, and permit readthrough of premature termination codons to produce full-length protein. METHODS To ascertain whether aminoglycosides permit readthrough of the most common CTNS nonsense mutation, W138X, we studied the effect of G418 on patient fibroblasts. RESULTS G418 treatment induced translational readthrough of CTNSW138X constructs transfected into HEK293 cells and expression of full-length endogenous CTNS protein in homozygous W138X fibroblasts. CONCLUSIONS Reduction in intracellular cystine indicates that the CTNS protein produced is functional as a cystine transporter. Interestingly, similar effects were seen even in W138X compound heterozygotes. These studies establish proof-of-principle for the potential of aminoglycosides to treat cystinosis and possibly other monogenic diseases caused by nonsense mutations.
Collapse
Affiliation(s)
- Emma J Brasell
- Department of Human Genetics, McGill University, Montreal, Québec, Canada
| | - LeeLee Chu
- The Research Institute of the McGill University Health Centre, 1001 Decarie Boulevard, Montreal, Québec, Canada
| | - Reyhan El Kares
- The Research Institute of the McGill University Health Centre, 1001 Decarie Boulevard, Montreal, Québec, Canada
| | - Jung Hwa Seo
- The Research Institute of the McGill University Health Centre, 1001 Decarie Boulevard, Montreal, Québec, Canada
| | | | | | - Paul Goodyer
- Department of Human Genetics, McGill University, Montreal, Québec, Canada. .,The Research Institute of the McGill University Health Centre, 1001 Decarie Boulevard, Montreal, Québec, Canada. .,Department of Experimental Medicine, McGill University, Montreal, Canada.
| |
Collapse
|
129
|
Li XQ, Wu D, Liang XJ, Li WJ, Liu M, Cao BY, Su C, Meng X, Gong CX. The diagnosis of cystinosis in patients reveals new CTNS gene mutations in the Chinese population. J Pediatr Endocrinol Metab 2019; 32:375-382. [PMID: 30849045 DOI: 10.1515/jpem-2018-0263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 01/30/2019] [Indexed: 11/15/2022]
Abstract
Background Cystinosis is a rare autosomal-recessive disorder caused by a defective transport of cystine across the lysosomal membrane. Previous studies have mapped cystinosis to the CTNS gene which is located on chromosome 17p13, and various CTNS mutations have been identified to correlate them with this disease. Methods We analyzed six patients from five unrelated families who were diagnosed with cystinosis in our hospital. We described the diagnostic procedures for all the patients and proposed alternative therapies for cystinosis patients instead of using cysteamine, an orphan drug which was commercially unavailable in China. Moreover, genetic analysis of all patients' samples was carried out to identify novel CTNS gene mutations. Results and conclusions The patients in this study were followed up from 1 to more than 10 years to monitor their growth and development, which indicated that the alternative therapies we used were helpful to ameliorate the complications of the cystinosis patients without cysteamine. Furthermore, by sequencing the patients' genome, we identified novel mutations in the CTNS gene including: c.477C > G (p.S159R), c.274C > T (p.Q92X) and c.680A > T (p.E227V); these mutations were only observed in cystinosis patients and had never been reported in any other populations, suggesting they might be specific to Chinese cystinosis patients.
Collapse
Affiliation(s)
- Xiao-Qiao Li
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Di Wu
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Xue-Jun Liang
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Wen-Jing Li
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Min Liu
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Bing-Yan Cao
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Chang Su
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Xi Meng
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Chun-Xiu Gong
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| |
Collapse
|
130
|
Rodríguez‐Martínez S, Márquez R, Inácio Â, Galván I. Changes in melanocyte RNA and DNA methylation favour pheomelanin synthesis and may avoid systemic oxidative stress after dietary cysteine supplementation in birds. Mol Ecol 2019; 28:1030-1042. [DOI: 10.1111/mec.15024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 01/11/2019] [Accepted: 01/14/2019] [Indexed: 12/27/2022]
Affiliation(s)
| | - Rafael Márquez
- Departamento de Ecología Evolutiva Estación Biológica de Doñana, CSIC Sevilla Spain
| | - Ângela Inácio
- Laboratório de GenéticaInstituto de Saúde AmbientalFaculdade de MedicinaUniversidade de Lisboa Lisboa Portugal
| | - Ismael Galván
- Departamento de Ecología Evolutiva Estación Biológica de Doñana, CSIC Sevilla Spain
| |
Collapse
|
131
|
Jeung Y, Lee K, Lee HJ, Kim E, Son MJ, Ahn J, Kim H, Kim W, Lee H, Kim JM, Chung K. Cationic amino acid transporter PQLC2 is a potential therapeutic target in gastric cancer. Cancer Sci 2019; 110:1453-1463. [PMID: 30729615 PMCID: PMC6447956 DOI: 10.1111/cas.13966] [Citation(s) in RCA: 5] [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: 09/27/2018] [Revised: 01/25/2019] [Accepted: 02/05/2019] [Indexed: 12/11/2022] Open
Abstract
Tumor cells overexpress amino acid transporters to meet the increased demand for amino acids. PQ loop repeat-containing (PQLC)2 is a cationic amino acid transporter that might be involved in cancer progression. Here, we show that upregulation of PQLC2 is critical to gastric cancer (GC) development in vitro and in vivo. Both PQLC2 mRNA and protein were overexpressed in GC tissues, especially of the diffuse type. Overexpression of PQLC2 promoted cell growth, anchorage independence, and tumor formation in nude mice. This was due to activation of MEK/ERK1/2 and PI3K/AKT signaling. Conversely, PQLC2 knockdown caused growth arrest and cell death of cancer cells and suppressed tumor growth in a mouse xenograft model. These results suggest that targeting PQLC2 is an effective strategy for GC treatment.
Collapse
Affiliation(s)
- Yun‐Ji Jeung
- Biomedical Translational Research CenterKRIBBDaejeonKorea
- Department of Pathology and Medical ScienceChungnam National University College of MedicineDaejeonKorea
| | - Kyeong Lee
- College of PharmacyDongguk University‐SeoulGoyangKorea
| | - Hyo Jin Lee
- Department of Internal MedicineChungnam National University College of MedicineDaejeonKorea
| | - Eunah Kim
- Stem Cell Convergence Research CenterKRIBBDaejeonKorea
| | - Myung Jin Son
- Stem Cell Convergence Research CenterKRIBBDaejeonKorea
- Department of Functional GenomicsKRIBB School of BioscienceKorea University of Science and Technology (UST)DaejeonKorea
| | - Jiwon Ahn
- Biomedical Translational Research CenterKRIBBDaejeonKorea
| | - Han‐Gyeul Kim
- Biomedical Translational Research CenterKRIBBDaejeonKorea
- Department of Functional GenomicsKRIBB School of BioscienceKorea University of Science and Technology (UST)DaejeonKorea
| | - Wantae Kim
- Biomedical Translational Research CenterKRIBBDaejeonKorea
| | - Ho‐Joon Lee
- Stem Cell Convergence Research CenterKRIBBDaejeonKorea
| | - Jin Man Kim
- Department of Internal MedicineChungnam National University College of MedicineDaejeonKorea
| | - Kyung‐Sook Chung
- Biomedical Translational Research CenterKRIBBDaejeonKorea
- Stem Cell Convergence Research CenterKRIBBDaejeonKorea
- Department of Functional GenomicsKRIBB School of BioscienceKorea University of Science and Technology (UST)DaejeonKorea
| |
Collapse
|
132
|
Battafarano G, Rossi M, Rega LR, Di Giovamberardino G, Pastore A, D'Agostini M, Porzio O, Nevo N, Emma F, Taranta A, Del Fattore A. Intrinsic Bone Defects in Cystinotic Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1053-1064. [PMID: 30794806 DOI: 10.1016/j.ajpath.2019.01.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 01/09/2019] [Accepted: 01/24/2019] [Indexed: 01/22/2023]
Abstract
Cystinosis is a rare lysosomal storage disorder caused by loss-of-function mutations of the CTNS gene, encoding cystinosin, a symporter that mediates cystine efflux from lysosomes. Approximately 95% of patients with cystinosis display renal Fanconi syndrome, short stature, osteopenia, and rickets. In this study, we investigated whether the absence of cystinosin primarily affects bone remodeling activity, apart from the influences of the Fanconi syndrome on bone mineral metabolism. Using micro-computed tomography and histomorphometric and bone serum biomarker analysis, we evaluated the bone phenotype of 1-month-old Ctns-/- knockout (KO) male mice without tubulopathy. An in vitro study was performed to characterize the effects of cystinosin deficiency on osteoblasts and osteoclasts. Micro-computed tomography analysis showed a reduction of trabecular bone volume, bone mineral density, and number and thickness in KO mice compared with wild-type animals; histomorphometric analysis revealed a reduction of osteoblast and osteoclast parameters in tibiae of cystinotic mice. Decreased levels of serum procollagen type 1 amino-terminal propeptide and tartrate-resistant acid phosphatase in KO mice confirmed reduced bone remodeling activity. In vitro experiments showed an impairment of Ctns-/- osteoblasts and osteoclasts. In conclusion, cystinosin deficiency primarily affects bone cells, leading to a bone loss phenotype of KO mice, independent from renal failure.
Collapse
Affiliation(s)
- Giulia Battafarano
- Bone Physiopathology Group, Multifactorial Disease and Complex Phenotype Research Area, Bambino Gesù Children's Hospital, Rome, Italy
| | - Michela Rossi
- Bone Physiopathology Group, Multifactorial Disease and Complex Phenotype Research Area, Bambino Gesù Children's Hospital, Rome, Italy
| | - Laura R Rega
- Laboratories of Nephrology, Bambino Gesù Children's Hospital, Rome, Italy
| | | | - Anna Pastore
- Molecular Genetics and Functional Genomics, Division of Genetic and Rare Diseases, Bambino Gesù Children's Hospital, Rome, Italy
| | | | - Ottavia Porzio
- Clinical Laboratory, Bambino Gesù Children's Hospital, Rome, Italy; Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Nathalie Nevo
- INSERM U1163, Université Paris Descartes, Institut Imagine, Hôpital Necker Enfants Malades, Paris, France
| | - Francesco Emma
- Laboratories of Nephrology, Bambino Gesù Children's Hospital, Rome, Italy; Division of Nephrology, Department of Pediatric Subspecialities, Bambino Gesù Children's Hospital, Rome, Italy
| | - Anna Taranta
- Laboratories of Nephrology, Bambino Gesù Children's Hospital, Rome, Italy.
| | - Andrea Del Fattore
- Bone Physiopathology Group, Multifactorial Disease and Complex Phenotype Research Area, Bambino Gesù Children's Hospital, Rome, Italy
| |
Collapse
|
133
|
Abstract
Fanconi syndrome, also known as the DeToni, Debré, Fanconi syndrome is a global dysfunction of the proximal tubule characterized by glucosuria, phosphaturia, generalized aminoaciduria, and type II renal tubular acidosis. Often there is hypokalemia, sodium wasting, and dehydration. In children, it typically is caused by inborn errors of metabolism, principally cystinosis. In adults, it is mainly caused by medications, exogenous toxins, and heavy metals. Treatment consists of treating the underlying cause and replacing the lost electrolytes and volume.
Collapse
Affiliation(s)
- John W Foreman
- Department of Pediatrics, Duke University School of Medicine, Erwin Road, Durham, NC 27710, USA.
| |
Collapse
|
134
|
Signorile A, Sgaramella G, Bellomo F, De Rasmo D. Prohibitins: A Critical Role in Mitochondrial Functions and Implication in Diseases. Cells 2019; 8:cells8010071. [PMID: 30669391 PMCID: PMC6356732 DOI: 10.3390/cells8010071] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/10/2019] [Accepted: 01/15/2019] [Indexed: 12/11/2022] Open
Abstract
Prohibitin 1 (PHB1) and prohibitin 2 (PHB2) are proteins that are ubiquitously expressed, and are present in the nucleus, cytosol, and mitochondria. Depending on the cellular localization, PHB1 and PHB2 have distinctive functions, but more evidence suggests a critical role within mitochondria. In fact, PHB proteins are highly expressed in cells that heavily depend on mitochondrial function. In mitochondria, these two proteins assemble at the inner membrane to form a supra-macromolecular structure, which works as a scaffold for proteins and lipids regulating mitochondrial metabolism, including bioenergetics, biogenesis, and dynamics in order to determine the cell fate, death, or life. PHB alterations have been found in aging and cancer, as well as neurodegenerative, cardiac, and kidney diseases, in which significant mitochondrial impairments have been observed. The molecular mechanisms by which prohibitins regulate mitochondrial function and their role in pathology are reviewed and discussed herein.
Collapse
Affiliation(s)
- Anna Signorile
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari "Aldo Moro", 70124 Bari, Italy.
| | - Giuseppe Sgaramella
- Water Research Institute (IRSA), National Research Council (CNR), Viale F. De Blasio, 5, 70132 Bari, Italy.
| | - Francesco Bellomo
- Laboratory of Nephrology, Department of Rare Diseases, Bambino Gesù Children's Hospital, Viale di S. Paolo, 15, 00149 Rome, Italy.
| | - Domenico De Rasmo
- Institute of Biomembrane, Bioenergetics and Molecular Biotechnology (IBIOM), National Research Council (CNR), 70126 Bari, Italy.
| |
Collapse
|
135
|
Dixon P, Chauhan A. Carbon Black Tinted Contact Lenses for Reduction of Photophobia in Cystinosis Patients. Curr Eye Res 2019; 44:497-504. [PMID: 30624086 DOI: 10.1080/02713683.2018.1563701] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
PURPOSE To examine if current development on using contact lenses for drug delivery of cysteamine to treat ocular symptoms of cystinosis can be tinted to mitigate photophobia common in patients by reducing transmittance Methods: Commercial contact lenses were placed in a carbon black solution to examine loading after lens synthesis. Silicone hydrogel contact lenses were also synthesized with carbon black added prior to UV curing. Transmittance was measured using UV-vis spectrophotometry over the range of 190-1190 nm and compared to unmodified contact lenses. Lens parameters of refractive index, ion permeability, and Young's modulus were measured using a refractometer, release of sodium chloride, and the cantilever method. Cysteamine release was measured by loading lenses into 5% cysteamine solution and then monitoring the release of the drug using UV-vis spectrophotometry. Vitamin E diffusion barriers were also added to lenses via ethanol solution, and the release of cysteamine from these modified lenses was also examined. RESULTS No leeching of carbon black was detected during experiments. Loading of pre-made contact lenses led to uneven distribution of carbon black throughout lens. Adding 0.3% carbon black to lens monomer solution prior to UV-curing led to even distribution and a transmittance reduction of approximately 50%. Ion permeability was reduced from 6.19 ± 0.90 x 10-3 to 1.28 ± 0.06 x 10-3 mm2 min-1, and Young's modulus was decreased from 1.58 ± 0.08 to 1.29 ± 0.06 MPa. Cysteamine releases from carbon black lenses with and without vitamin E were comparable to controls, although the loading solution of vitamin E/ethanol had to be tripled to achieve a similar mass loading to control. CONCLUSIONS Carbon black increases the softness of contact lenses, but a loading of 0.3% maintains lens parameters required for wear. The release of cysteamine is also possible with carbon black lenses, albeit requiring a higher loading concentration of vitamin E to achieve similar release times.
Collapse
Affiliation(s)
- Phillip Dixon
- a Department of Chemical Engineering , University of Florida , Gainesville , FL , USA
| | - Anuj Chauhan
- a Department of Chemical Engineering , University of Florida , Gainesville , FL , USA
| |
Collapse
|
136
|
Zhang J, He J, Johnson JL, Rahman F, Gavathiotis E, Cuervo AM, Catz SD. Chaperone-Mediated Autophagy Upregulation Rescues Megalin Expression and Localization in Cystinotic Proximal Tubule Cells. Front Endocrinol (Lausanne) 2019; 10:21. [PMID: 30774622 PMCID: PMC6367655 DOI: 10.3389/fendo.2019.00021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 01/14/2019] [Indexed: 01/16/2023] Open
Abstract
Cystinosis is a lysosomal storage disorder caused by defects in CTNS, the gene that encodes the lysosomal cystine transporter cystinosin. Patients with nephropathic cystinosis are characterized by endocrine defects, defective proximal tubule cell (PTC) function, the development of Fanconi syndrome and, eventually, end-stage renal disease. Kidney disease is developed despite the use of cysteamine, a drug that decreases lysosomal cystine overload but fails to correct overload-independent defects. Chaperone-mediated autophagy (CMA), a selective form of autophagy, is defective in cystinotic mouse fibroblasts, and treatment with cysteamine is unable to correct CMA defects in vivo, but whether the vesicular trafficking mechanisms that lead to defective CMA in cystinosis are manifested in human PTCs is not currently known and whether PTC-specific mechanisms are corrected upon CMA upregulation remains to be elucidated. Here, using CRISPR-Cas9 technology, we develop a new human PTC line with defective cystinosin expression (CTNS-KO PTCs). We show that the expression and localization of the CMA receptor, LAMP2A, is defective in CTNS-KO PTCs. The expression of the lipidated form of LC3B, a marker for another form of autophagy (macroautophagy), is decreased in CTNS-KO PTCs indicating decreased autophagosome numbers under basal conditions. However, the autophagic flux is functional, as measured by induction by starvation or by blockage using the v-ATPase inhibitor bafilomycin A, and by degradation of the macroautophagy substrate SQSTM1 under starvation and proteasome-inhibited conditions. Previous studies showed that LAMP2A accumulates in Rab11-positive vesicles in cystinotic cells. Here, we show defective Rab11 expression, localization and trafficking in CTNS-KO PTCs as determined by confocal microscopy, immunoblotting and TIRFM. We also show that both Rab11 expression and trafficking in cystinotic PTCs are rescued by the upregulation of CMA using small-molecule CMA activators. Cystinotic PTCs are characterized by PTC de-differentiation accompanied by loss of the endocytic receptor megalin, and megalin recycling is regulated by Rab11. Here we show that megalin plasma membrane localization is defective in CTNS-KO PTCs and its expression is rescued by treatment with CMA activators. Altogether, our data support that CMA upregulation has the potential to improve PTC function in cystinosis.
Collapse
Affiliation(s)
- Jinzhong Zhang
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - Jing He
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - Jennifer L. Johnson
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - Farhana Rahman
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - Evripidis Gavathiotis
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Ana Maria Cuervo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Sergio D. Catz
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
- *Correspondence: Sergio D. Catz
| |
Collapse
|
137
|
Effects of long-term cysteamine treatment in patients with cystinosis. Pediatr Nephrol 2019; 34:571-578. [PMID: 29260317 PMCID: PMC6394685 DOI: 10.1007/s00467-017-3856-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 11/05/2017] [Accepted: 11/16/2017] [Indexed: 01/10/2023]
Abstract
Cystinosis is a rare autosomal-recessive lysosomal storage disease with high morbidity and mortality. It is caused by mutations in the CTNS gene that encodes the cystine transporter, cystinosin, which leads to lysosomal cystine accumulation. Patients with infantile nephropathic cystinosis, the most common and most severe clinical form of cystinosis, commonly present with renal Fanconi syndrome by 6-12 months of age, and without specific treatment, almost all will develop end-stage renal disease (ESRD) by 10-12 years of age. Early corneal cystine crystal deposition is a hallmark of the disease. Cystinosis also presents with gastrointestinal symptoms (e.g., vomiting, decreased appetite, and feeding difficulties) and severe growth retardation and may affect several other organs over time, including the eye, thyroid gland, gonads, pancreas, muscles, bone marrow, liver, nervous system, lungs, and bones. Cystine-depleting therapy with cysteamine orally is the only specific targeted therapy available for managing cystinosis and needs to be combined with cysteamine eye drops for corneal disease involvement. In patients with early treatment initiation and good compliance to therapy, long-term cysteamine treatment delays progression to ESRD, significantly improves growth, decreases the frequency and severity of extrarenal complications, and is associated with extended life expectancy. Therefore, early diagnosis of cystinosis and adequate life-long treatment with cysteamine are essential for preventing end-organ damage and improving the overall prognosis in these patients.
Collapse
|
138
|
Shimizu Y, Yanobu-Takanashi R, Nakano K, Hamase K, Shimizu T, Okamura T. A deletion in the Ctns gene causes renal tubular dysfunction and cystine accumulation in LEA/Tohm rats. Mamm Genome 2018; 30:23-33. [PMID: 30591971 PMCID: PMC6397714 DOI: 10.1007/s00335-018-9790-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 12/19/2018] [Indexed: 11/21/2022]
Abstract
The Long-Evans Agouti (LEA/Tohm) rat has recently been established as a new rat model of type 2 diabetes. The onset of diabetes mellitus was observed only in male LEA/Tohm rats; however, urinary glucose appeared before the onset of diabetes. To clarify the genetic basis of urinary glucose, we performed genetic linkage analysis using (BN × LEA) F2 intercross progeny. A recessively acting locus responsible for urinary glucose excretion (ugl) was mapped to a 7.9 Mb region of chromosome 10, which contains the cystinosin (Ctns) gene. The Ctns gene encodes the cystine transporter, which transports cystine out of the lysosome and is responsible for nephropathic cystinosis in humans. Sequence analysis identified a 13-bp deletion in the Ctns gene, leading to a truncated and loss-of-function protein, which cause cystine accumulation in various tissues. We also developed a novel congenic rat strain harboring the Ctnsugl mutation on the F344 genetic background. Phenotypic analysis of F344-Ctnsugl rats indicated that the incidence of urinary glucose was 100% in both males and females at around 40 weeks of age, and marked cystine accumulation was observed in the tissues, as well as remarkable renal lesions and cystine crystals in the lysosomes of the renal cortex. Furthermore, treatment with cysteamine depleted the cystine contents in F344-Ctnsugl rat embryonic fibroblasts. These results indicated that the F344-Ctnsugl rat provides a novel rat model of cystinosis, which allows not only a better understanding of the pathogenesis and pathophysiology of cystinosis but will also contribute to the development of new therapies.
Collapse
Affiliation(s)
- Yukiko Shimizu
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine (NCGM), 1-21-1 Toyama, Shinjyuku-ku, Tokyo, 162-8655, Japan.,Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Rieko Yanobu-Takanashi
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine (NCGM), 1-21-1 Toyama, Shinjyuku-ku, Tokyo, 162-8655, Japan
| | - Kenta Nakano
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine (NCGM), 1-21-1 Toyama, Shinjyuku-ku, Tokyo, 162-8655, Japan.,Laboratory of Laboratory Animal Science and Medicine, School of Veterinary Medicine, Kitasato University, Higashi 23-35-1, Towada, Aomori, 034-8628, Japan
| | - Kenji Hamase
- Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Toshiaki Shimizu
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Tadashi Okamura
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine (NCGM), 1-21-1 Toyama, Shinjyuku-ku, Tokyo, 162-8655, Japan. .,Section of Animal Models, Department of Infections Diseases, Research Institute, National Center for Global Health and Medicine (NCGM), 1-21-1 Toyama, Shinjyuku-ku, Tokyo, 162-8655, Japan.
| |
Collapse
|
139
|
Biswas S, Sornalingam K. The Ocular Status of Cystinosis Patients Receiving a Hospital Pharmacy-Made Preparation of Cysteamine Eye Drops: A Case Series. Ophthalmol Ther 2018; 8:125-136. [PMID: 30519924 PMCID: PMC6393247 DOI: 10.1007/s40123-018-0156-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Indexed: 12/01/2022] Open
Abstract
Introduction Infantile nephropathic cystinosis (INC) is an autosomal recessive lysosomal disorder in which patients develop deposits of cystine crystals in their kidneys and corneas from a young age. Methods We conducted a retrospective analysis of children with INC seen by ophthalmologists at the Manchester Royal Eye Hospital between 2002 and 2018, to evaluate clinical findings, symptoms and treatment. Results Twenty-two children diagnosed with INC from age 0 (prenatally) to 11 years were assessed. All evaluable patients had corneal cystine crystal deposits, and 15 had mild to moderate photophobia. Ten patients had other ocular conditions including blepharitis/chalzion (n = 6), swollen optic nerve (n = 3), punctate epitheliopathy (n = 3), corneal scarring (n = 1),and elevated intraocular pressure (n = 2). Confocal imaging identified nerve abnormalities in two patients (enlarged corneal nerve + abnormal-looking tortuous nerves in one patient and beaded nerves in the sub-basal plexus in the other), both of whom had significant crystal deposition in the anterior stroma. Visual acuity was relatively unaffected. All 22 patients were receiving oral cysteamine, and 21 were applying cysteamine eye drops (galenic preparation of 0.55% concentration, compounded by a hospital pharmacy). Recommended application frequency was at least eight times per day in all patients with dosing information available. Conclusions This case series of patients with INC highlights the consistent pattern of corneal cystine crystal deposition, which is universally present from a young age in this condition, and the high incidence of photophobia even in young children. Corneal manifestations of INC persisted despite frequent administration of the hospital pharmacy-made eye drop preparation. Reasons for this lack of efficacy may include the lag period between diagnosis and first prescription of cysteamine eye drops and the difficulty in maintaining rigorous compliance with this treatment. In addition, the challenge for patients of maintaining optimal storage conditions may adversely affect the stability and efficacy of cysteamine within this preparation. Funding Editorial assistance was funded by Orphan Europe Ltd.
Collapse
Affiliation(s)
- Susmito Biswas
- Manchester Royal Eye Hospital, Manchester Academic Health Science Centre, Manchester, UK.
| | - Krishanthy Sornalingam
- Manchester Royal Eye Hospital, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
140
|
Sumayao R, Newsholme P, McMorrow T. The Role of Cystinosin in the Intermediary Thiol Metabolism and Redox Homeostasis in Kidney Proximal Tubular Cells. Antioxidants (Basel) 2018; 7:antiox7120179. [PMID: 30513914 PMCID: PMC6315507 DOI: 10.3390/antiox7120179] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/22/2018] [Accepted: 11/24/2018] [Indexed: 01/26/2023] Open
Abstract
Cystinosin is a lysosomal transmembrane protein which facilitates transport of the disulphide amino acid cystine (CySS) from the lysosomes of the cell. This protein is encoded by the CTNS gene which is defective in the lysosomal storage disorder, cystinosis. Because of the apparent involvement of cystinosin in the intermediary thiol metabolism, its discovery has fuelled investigations into its role in modulating cellular redox homeostasis. The kidney proximal tubular cells (PTCs) have become the focus of various studies on cystinosin since the protein is highly expressed in these cells and kidney proximal tubular transport dysfunction is the foremost clinical manifestation of cystinosis. The lysosomal CySS pool is a major source of cytosolic cysteine (Cys), the limiting amino acid for the synthesis of an important antioxidant glutathione (GSH) via the γ-glutamyl cycle. Therefore, loss of cystinosin function is presumed to lead to cytosolic deficit of Cys which may impair GSH synthesis. However, studies using in vitro models lacking cystinosin yielded inconsistent results and failed to establish the mechanistic role of cystinosin in modulating GSH synthesis and redox homeostasis. Because of the complexity of the metabolic micro- and macro-environment in vivo, using in vitro models alone may not be able to capture the complete sequence of biochemical and physiological events that occur as a consequence of loss of cystinosin function. The coexistence of pathways for the overall handling and disposition of GSH, the modulation of CTNS gene by intracellular redox status and the existence of a non-canonical isoform of cystinosin may constitute possible rescue mechanisms in vivo to remediate redox perturbations in renal PTCs. Importantly, the mitochondria seem to play a critical role in orchestrating redox imbalances initiated by cystinosin dysfunction. Non-invasive techniques such as in vivo magnetic resonance imaging with the aid of systems biology approaches may provide invaluable mechanistic insights into the role of cystinosin in the essential intermediary thiol metabolism and in the overall regulation cellular redox homeostasis.
Collapse
Affiliation(s)
- Rodolfo Sumayao
- Chemistry Department, De La Salle University, Manila 1004, Philippines.
| | - Philip Newsholme
- School of Pharmacy and Biomedical Sciences and Curtin Health Innovation Research Institute (CHIRI), Curtin University, Perth 6845, Australia.
| | - Tara McMorrow
- Conway Institute, School of Biomolecular and Biomedical Science, University College Dublin, Dublin 4, Ireland.
| |
Collapse
|
141
|
Milone M, Liewluck T. The unfolding spectrum of inherited distal myopathies. Muscle Nerve 2018; 59:283-294. [PMID: 30171629 DOI: 10.1002/mus.26332] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/26/2018] [Accepted: 08/28/2018] [Indexed: 12/30/2022]
Abstract
Distal myopathies are a group of rare muscle diseases characterized by distal weakness at onset. Although acquired myopathies can occasionally present with distal weakness, the majority of distal myopathies have a genetic etiology. Their age of onset varies from early-childhood to late-adulthood while the predominant muscle weakness can affect calf, ankle dorsiflexor, or distal upper limb muscles. A spectrum of muscle pathological changes, varying from nonspecific myopathic changes to rimmed vacuoles to myofibrillar pathology to nuclei centralization, have been noted. Likewise, the underlying molecular defect is heterogeneous. In addition, there is emerging evidence that distal myopathies can result from defective proteins encoded by genes causative of neurogenic disorders, be manifestation of multisystem proteinopathies or the result of the altered interplay between different genes. In this review, we provide an overview on the clinical, electrophysiological, pathological, and molecular aspects of distal myopathies, focusing on the most recent developments in the field. Muscle Nerve 59:283-294, 2019.
Collapse
Affiliation(s)
| | - Teerin Liewluck
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
142
|
Vaisbich MH, Satiro CAF, Roz D, Nunes DDAD, Messa ACHL, Lanetzki C, Ferreira JCDOA. Multidisciplinary approach for patients with nephropathic cystinosis: model for care in a rare and chronic renal disease. J Bras Nefrol 2018; 41:131-141. [PMID: 30465592 PMCID: PMC6534023 DOI: 10.1590/2175-8239-jbn-2018-0139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 08/30/2018] [Indexed: 11/21/2022] Open
Abstract
Care for patients with chronic and rare diseases is complex, especially considering the lack of knowledge about the disease, which makes early and precise diagnosis difficult, as well as the need for specific tests, sometimes of high complexity and cost. Added to these factors are difficulties in obtaining adequate treatment when available, in raising patient and family awareness about the disease and treatment compliance. Nephropathic cystinosis is among these diseases. After more than 20 years as a care center for these patients, the authors propose a follow-up protocol, which has been used with improvement in the quality of care and consists of a multidisciplinary approach, including care provided by a physician, nurse, psychologist, nutritionist and social worker. In this paper, each field objectively exposes how to address points that involve the stages of diagnosis and its communication with the patient and their relatives or guardians, covering the particularities of the disease and the treatment, the impact on the lives of patients and families, the approach to psychological and social issues and guidelines on medications and diets. This protocol could be adapted to the follow-up of patients with other rare diseases, including those with renal involvement. This proposal is expected to reach the largest number of professionals involved in the follow-up of these patients, strengthening the bases for the creation of a national protocol, observing the particularities of each case.
Collapse
Affiliation(s)
- Maria Helena Vaisbich
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, São Paulo, SP, Brasil
| | | | - Deborah Roz
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, São Paulo, SP, Brasil
| | | | - Ana Carola H Lobo Messa
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, São Paulo, SP, Brasil
| | - Camila Lanetzki
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, São Paulo, SP, Brasil
| | | |
Collapse
|
143
|
Lyu SJ, Ren WR, Zhu HL, Liu T. [The clinical characteristics and molecular pathogenesis of a variant Glanzmann's thrombasthenia-like pedigree]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2018; 39:807-811. [PMID: 30369200 PMCID: PMC7348282 DOI: 10.3760/cma.j.issn.0253-2727.2018.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Indexed: 02/05/2023]
Abstract
Objective: To review the clinical characteristics of a pedigree with inherited hemorrhagic disease to explore its molecular pathogenesis. Methods: The clinical data of the pedigree with inherited hemorrhagic disease were collected. After extracting DNA, next generation sequencing was utilized to detect the potential gene mutation. The changes of RASGRP2 transcript of this proband and his parents were detected using RT-PCR to compare with normal control. Results: The phenotype of the proband in this pedigree with inherited platelet dysfunction and bleeding disorder was similar to variant Glanzmann's thrombasthenia, the maximum aggregations of platelet in response to the physiological agonists including ADP, epinephrine and arachidonic acid were significantly lower, leading to severe spontaneous mucosal bleeding. Integrin αIIbβ3 gene mutation was not detected, but another gene mutation RASGRP2 IVS3-1 stood out. The mutation was homozygous in the proband and heterozygosis in both of his parents. Two transcript types were detected in the proband, without transcripts coding functional RASGRP2 protein, however, his parents had functional transcripts and abnormal transcripts, with the normal transcripts in the majority. Conclusions: The RASGRP2 IVS3-1 gene mutation was responsible for the inherited hemorrhagic disease. The RASGRP2 IVS3-1 gene mutation led to abnormal alternative splicing, without formation of functional RASGRP2 protein. The RASGRP2 protein is at the nexus of calcium-dependent platelet activation and hemostasis after damage of blood vessels. Spontaneous mucosal bleeding was a result of the lack of the functional RASGRP2 protein. This was the first report of RASGRP2 gene mutation resulting in bleeding disorder in China, and also the first report of the mutation type of RASGRP2 IVS3-1.
Collapse
Affiliation(s)
- S J Lyu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu 610041, China
| | | | | | | |
Collapse
|
144
|
Bellomo F, Signorile A, Tamma G, Ranieri M, Emma F, De Rasmo D. Impact of atypical mitochondrial cyclic-AMP level in nephropathic cystinosis. Cell Mol Life Sci 2018; 75:3411-3422. [PMID: 29549422 PMCID: PMC11105431 DOI: 10.1007/s00018-018-2800-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 02/20/2018] [Accepted: 03/14/2018] [Indexed: 02/06/2023]
Abstract
Nephropathic cystinosis (NC) is a rare disease caused by mutations in the CTNS gene encoding for cystinosin, a lysosomal transmembrane cystine/H+ symporter, which promotes the efflux of cystine from lysosomes to cytosol. NC is the most frequent cause of Fanconi syndrome (FS) in young children, the molecular basis of which is not well established. Proximal tubular cells have very high metabolic rate due to the active transport of many solutes. Not surprisingly, mitochondrial disorders are often characterized by FS. A similar mechanism may also apply to NC. Because cAMP has regulatory properties on mitochondrial function, we have analyzed cAMP levels and mitochondrial targets in CTNS-/- conditionally immortalized proximal tubular epithelial cells (ciPTEC) carrying the classical homozygous 57-kb deletion (delCTNS-/-) or with compound heterozygous loss-of-function mutations (mutCTNS-/-). Compared to wild-type cells, cystinotic cells had significantly lower mitochondrial cAMP levels (delCTNS-/- ciPTEC by 56% ± 10.5, P < 0.0001; mutCTNS-/- by 26% ± 4.3, P < 0.001), complex I and V activities, mitochondrial membrane potential, and SIRT3 protein levels, which were associated with increased mitochondrial fragmentation. Reduction of complex I and V activities was associated with lower expression of part of their subunits. Treatment with the non-hydrolysable cAMP analog 8-Br-cAMP restored mitochondrial potential and corrected mitochondria morphology. Treatment with cysteamine, which reduces the intra-lysosomal cystine, was able to restore mitochondrial cAMP levels, as well as most other abnormal mitochondrial findings. These observations were validated in CTNS-silenced HK-2 cells, indicating a pivotal role of mitochondrial cAMP in the proximal tubular dysfunction observed in NC.
Collapse
Affiliation(s)
- Francesco Bellomo
- Laboratory of Nephrology, Department of Rare Diseases, Bambino Gesù Children's Hospital, Viale di S. Paolo, 15, 00149, Rome, Italy.
| | - Anna Signorile
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari "Aldo Moro", Policlinico, Piazza G. Cesare, 11, 70124, Bari, Italy
| | - Grazia Tamma
- Department of Bioscience, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Bari, Italy
| | - Marianna Ranieri
- Department of Bioscience, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Bari, Italy
| | - Francesco Emma
- Laboratory of Nephrology, Department of Rare Diseases, Bambino Gesù Children's Hospital, Viale di S. Paolo, 15, 00149, Rome, Italy
- Division of Nephrology, Department of Pediatric Subspecialties, Bambino Gesù Children's Hospital, Rome, Italy
| | - Domenico De Rasmo
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari "Aldo Moro", Policlinico, Piazza G. Cesare, 11, 70124, Bari, Italy.
- Institute of Biomembrane, Bioenergetics and Molecular Biotechnology (IBIOM), National Research Council (CNR), Bari, Italy.
| |
Collapse
|
145
|
Anastasiya KA, Elena OG, Natalia BV, Anna KY, Kirill TY, Olesya KI, Tatiana NA, Inessa FD, Ekaterina SI, Peter SA, Valery IV. Atypical onset of nephropathic infantile cystinosis in a Russian patient with rare CTNS mutation. Clin Case Rep 2018; 6:1871-1876. [PMID: 30214781 PMCID: PMC6132087 DOI: 10.1002/ccr3.1678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 06/11/2018] [Indexed: 11/10/2022] Open
Abstract
We report a Russian patient with atypical onset of infantile nephropathic cystinosis. The disease debuted with vomiting and loss of weight and motor skills. Nephropathic changes appeared 6 months after onset of disease. Exome sequencing can be useful for diagnosing cystinosis in patients with neurological abnormalities before onset of nephropathic symptoms.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Nikishina A. Tatiana
- Veltischev Research and Clinical Institute for Pediatrics of the Pirogov Russian National Research Medical UniversityMoscowRussia
| | | | | | - Shatalov A. Peter
- Genotek Ltd.MoscowRussia
- Veltischev Research and Clinical Institute for Pediatrics of the Pirogov Russian National Research Medical UniversityMoscowRussia
| | - Ilinsky V. Valery
- Institute of Biomedical ChemistryMoscowRussia
- Genotek Ltd.MoscowRussia
- Vavilov Institute of General GeneticsMoscowRussia
| |
Collapse
|
146
|
Krause A, Seymour H, Ramsay M. Common and Founder Mutations for Monogenic Traits in Sub-Saharan African Populations. Annu Rev Genomics Hum Genet 2018; 19:149-175. [DOI: 10.1146/annurev-genom-083117-021256] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
This review highlights molecular genetic studies of monogenic traits where common pathogenic mutations occur in black families from sub-Saharan Africa. Examples of founder mutations have been identified for oculocutaneous albinism, cystic fibrosis, Fanconi anemia, and Gaucher disease. Although there are few studies from Africa, some of the mutations traverse populations across the continent, and they are almost all different from the common mutations observed in non-African populations. Myotonic dystrophy is curiously absent among Africans, and nonsyndromic deafness does not arise from mutations in GJB2 and GJB7. Locus heterogeneity is present for Huntington disease, with two common triplet expansion loci in Africa, HTT and JPH3. These findings have important clinical consequences for diagnosis, treatment, and genetic counseling in affected families. We currently have just a glimpse of the molecular etiology of monogenic diseases in sub-Saharan Africa, a proverbial “ears of the hippo” situation.
Collapse
Affiliation(s)
- Amanda Krause
- Division of Human Genetics, National Health Laboratory Service, and Division of Human Genetics, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Heather Seymour
- Division of Human Genetics, National Health Laboratory Service, and Division of Human Genetics, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Michèle Ramsay
- Division of Human Genetics, National Health Laboratory Service, and Division of Human Genetics, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
147
|
Kandasamy P, Gyimesi G, Kanai Y, Hediger MA. Amino acid transporters revisited: New views in health and disease. Trends Biochem Sci 2018; 43:752-789. [PMID: 30177408 DOI: 10.1016/j.tibs.2018.05.003] [Citation(s) in RCA: 294] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 05/23/2018] [Accepted: 05/25/2018] [Indexed: 02/09/2023]
Abstract
Amino acid transporters (AATs) are membrane-bound transport proteins that mediate transfer of amino acids into and out of cells or cellular organelles. AATs have diverse functional roles ranging from neurotransmission to acid-base balance, intracellular energy metabolism, and anabolic and catabolic reactions. In cancer cells and diabetes, dysregulation of AATs leads to metabolic reprogramming, which changes intracellular amino acid levels, contributing to the pathogenesis of cancer, obesity and diabetes. Indeed, the neutral amino acid transporters (NATs) SLC7A5/LAT1 and SLC1A5/ASCT2 are likely involved in several human malignancies. However, a clinical therapy that directly targets AATs has not yet been developed. The purpose of this review is to highlight the structural and functional diversity of AATs, their diverse physiological roles in different tissues and organs, their wide-ranging implications in human diseases and the emerging strategies and tools that will be necessary to target AATs therapeutically.
Collapse
Affiliation(s)
- Palanivel Kandasamy
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, CH-3012 Bern, Switzerland
| | - Gergely Gyimesi
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, CH-3012 Bern, Switzerland
| | - Yoshikatsu Kanai
- Division of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Osaka, Japan.
| | - Matthias A Hediger
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, CH-3012 Bern, Switzerland.
| |
Collapse
|
148
|
Cysteamine, an Endogenous Aminothiol, and Cystamine, the Disulfide Product of Oxidation, Increase Pseudomonas aeruginosa Sensitivity to Reactive Oxygen and Nitrogen Species and Potentiate Therapeutic Antibiotics against Bacterial Infection. Infect Immun 2018; 86:IAI.00947-17. [PMID: 29581193 PMCID: PMC5964511 DOI: 10.1128/iai.00947-17] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 03/16/2018] [Indexed: 12/14/2022] Open
Abstract
Cysteamine is an endogenous aminothiol produced in mammalian cells as a consequence of coenzyme A metabolism through the activity of the vanin family of pantetheinase ectoenzymes. It is known to have a biological role in oxidative stress, inflammation, and cell migration. There have been several reports demonstrating anti-infective properties targeting viruses, bacteria, and even the malarial parasite. We and others have previously described broad-spectrum antimicrobial and antibiofilm activities of cysteamine. Here, we go further to demonstrate redox-dependent mechanisms of action for the compound and how its antimicrobial effects are, at least in part, due to undermining bacterial defenses against oxidative and nitrosative challenges. We demonstrate the therapeutic potentiation of antibiotic therapy against Pseudomonas aeruginosa in mouse models of infection. We also demonstrate potentiation of many different classes of antibiotics against a selection of priority antibiotic-resistant pathogens, including colistin (often considered an antibiotic of last resort), and we discuss how this endogenous antimicrobial component of innate immunity has a role in infectious disease that is beginning to be explored and is not yet fully understood.
Collapse
|
149
|
Jamalpoor A, Sparidans RW, Pou Casellas C, Rood JJM, Joshi M, Masereeuw R, Janssen MJ. Quantification of cystine in human renal proximal tubule cells using liquid chromatography-tandem mass spectrometry. Biomed Chromatogr 2018. [PMID: 29517154 PMCID: PMC6055858 DOI: 10.1002/bmc.4238] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Nephropathic cystinosis is characterized by abnormal intralysosomal accumulation of cystine throughout the body, causing irreversible damage to various organs, particularly the kidneys. Cysteamine, the currently available treatment, can reduce lysosomal cystine and postpone disease progression. However, cysteamine poses serious side effects and does not address all of the symptoms of cystinosis. To screen for new treatment options, a rapid and reliable high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS) method was developed to quantify cystine in conditionally immortalized human proximal tubular epithelial cells (ciPTEC). The ciPTEC were treated with N-ethylmaleimide, lysed and deproteinized with 15% (w/v) sulfosalicylic acid. Subsequently, cystine was measured using deuterium-labeled cystine-D4, as the internal standard. The assay developed demonstrated linearity to at least 20 μmol/L with a good precision. Accuracies were between 97.3 and 102.9% for both cell extracts and whole cell samples. Cystine was sufficiently stable under all relevant analytical conditions. The assay was successfully applied to determine cystine levels in both healthy and cystinotic ciPTEC. Control cells showed clearly distinguishable cystine levels compared with cystinotic cells treated with or without cysteamine. The method developed provides a fast and reliable quantification of cystine, and is applicable to screen for potential drugs that could reverse cystinotic symptoms in human kidney cells.
Collapse
Affiliation(s)
- Amer Jamalpoor
- Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacology, Utrecht University, Utrecht, The Netherlands
| | - Rolf W Sparidans
- Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht University, Utrecht, The Netherlands
| | - Carla Pou Casellas
- Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacology, Utrecht University, Utrecht, The Netherlands
| | - Johannes J M Rood
- Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht University, Utrecht, The Netherlands
| | - Mansi Joshi
- Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacology, Utrecht University, Utrecht, The Netherlands
| | - Rosalinde Masereeuw
- Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacology, Utrecht University, Utrecht, The Netherlands
| | - Manoe J Janssen
- Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacology, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
150
|
Abstract
Besides its essential role in protein synthesis, cysteine plays vital roles in redox homeostasis, being a component of the major antioxidant glutathione (GSH) and a potent antioxidant by itself. In addition, cysteine undergoes a variety of post-translational modifications that modulate several physiological processes. It is becoming increasingly clear that redox-modulated events play important roles not only in peripheral tissues but also in the brain where cysteine disposition is central to these pathways. Dysregulated cysteine metabolism is associated with several neurodegenerative disorders. Accordingly, restoration of cysteine balance has therapeutic benefits. This review discusses metabolic signaling pathways pertaining to cysteine disposition in the brain under normal and pathological conditions, highlighting recent findings on cysteine metabolism during aging and in neurodegenerative conditions such as Huntington's disease (HD) and molybdenum cofactor (MoCo) deficiency (MoCD) among others.
Collapse
Affiliation(s)
- Bindu D Paul
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Juan I Sbodio
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Solomon H Snyder
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|