1
|
Sou YS, Yamaguchi J, Masuda K, Uchiyama Y, Maeda Y, Koike M. Golgi pH homeostasis stabilizes the lysosomal membrane through N-glycosylation of membrane proteins. Life Sci Alliance 2024; 7:e202402677. [PMID: 39079741 PMCID: PMC11289521 DOI: 10.26508/lsa.202402677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 07/19/2024] [Accepted: 07/19/2024] [Indexed: 08/02/2024] Open
Abstract
Protein glycosylation plays a vital role in various cellular functions, many of which occur within the Golgi apparatus. The Golgi pH regulator (GPHR) is essential for the proper functioning of the Golgi apparatus. The lysosomal membrane contains highly glycosylated membrane proteins in abundance. This study investigated the role of the Golgi luminal pH in N-glycosylation of lysosomal membrane proteins and the effect of this protein modification on membrane stability using Gphr-deficient MEFs. We showed that Gphr deficiency causes an imbalance in the Golgi luminal pH, resulting in abnormal protein N-glycosylation, indicated by a reduction in sialylated glycans and markedly reduced molecular weight of glycoproteins. Further experiments using FRAP and PLA revealed that Gphr deficiency prevented the trafficking dynamics and proximity condition of glycosyltransferases in the Golgi apparatus. In addition, incomplete N-glycosylation of lysosomal membrane proteins affected lysosomal membrane stability, as demonstrated by the increased susceptibility to lysosomal damage. Thus, this study highlights the critical role of Golgi pH regulation in controlling protein glycosylation and the impact of Golgi dysfunction on lysosomal membrane stability.
Collapse
Affiliation(s)
- Yu-Shin Sou
- Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Bunkyo, Japan
| | - Junji Yamaguchi
- Laboratory of Morphology and Image Analysis, Research Support Center, Juntendo University Graduate School of Medicine, Bunkyo, Japan
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Bunkyo, Japan
| | - Keisuke Masuda
- Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Bunkyo, Japan
| | - Yasuo Uchiyama
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Bunkyo, Japan
| | - Yusuke Maeda
- https://ror.org/035t8zc32 Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Masato Koike
- Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Bunkyo, Japan
| |
Collapse
|
2
|
Jankauskas SS, Varzideh F, Kansakar U, Al Tibi G, Densu Agyapong E, Gambardella J, Santulli G. Insights into molecular and cellular functions of the Golgi calcium/manganese-proton antiporter TMEM165. J Biol Chem 2024; 300:107567. [PMID: 39002685 PMCID: PMC11345563 DOI: 10.1016/j.jbc.2024.107567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/19/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024] Open
Abstract
The Golgi compartment performs a number of crucial roles in the cell. However, the exact molecular mechanisms underlying these actions are not fully defined. Pathogenic mutations in genes encoding Golgi proteins may serve as an important source for expanding our knowledge. For instance, mutations in the gene encoding Transmembrane protein 165 (TMEM165) were discovered as a cause of a new type of congenital disorder of glycosylation (CDG). Comprehensive studies of TMEM165 in different model systems, including mammals, yeast, and fish uncovered the new realm of Mn2+ homeostasis regulation. TMEM165 was shown to act as a Ca2+/Mn2+:H+ antiporter in the medial- and trans-Golgi network, pumping the metal ions into the Golgi lumen and protons outside. Disruption of TMEM165 antiporter activity results in defects in N- and O-glycosylation of proteins and glycosylation of lipids. Impaired glycosylation of TMEM165-CDG arises from a lack of Mn2+ within the Golgi. Nevertheless, Mn2+ insufficiency in the Golgi is compensated by the activity of the ATPase SERCA2. TMEM165 turnover has also been found to be regulated by Mn2+ cytosolic concentration. Besides causing CDG, recent investigations have demonstrated the functional involvement of TMEM165 in several other pathologies including cancer and mental health disorders. This systematic review summarizes the available information on TMEM165 molecular structure, cellular function, and its roles in health and disease.
Collapse
Affiliation(s)
- Stanislovas S Jankauskas
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, New York, USA
| | - Fahimeh Varzideh
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, New York, USA
| | - Urna Kansakar
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, New York, USA
| | - Ghaith Al Tibi
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, New York, USA
| | - Esther Densu Agyapong
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, New York, USA
| | - Jessica Gambardella
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, New York, USA; Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Gaetano Santulli
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, New York, USA; Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy; International Translational Research and Medical Education (ITME) Consortium, Academic Research Unit, Naples, Italy; Department of Molecular Pharmacology, Einstein Institute for Aging Research, Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York City, New York, USA.
| |
Collapse
|
3
|
Zhang Y, Sun M, Li N, Zhao Y, Zhang F, Shu J, Liu Y, Cai C. Identification of a novel intronic variant of ATP6V0A2 in a Han-Chinese family with cutis laxa. Mol Biol Rep 2024; 51:498. [PMID: 38598037 DOI: 10.1007/s11033-024-09446-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 03/13/2024] [Indexed: 04/11/2024]
Abstract
BACKGROUND Cutis laxa is a connective tissue disease caused by abnormal synthesis or secretion of skin elastic fibers, leading to skin flabby and saggy in various body parts. It can be divided into congenital cutis laxa and acquired cutis laxa, and inherited cutis laxa syndromes is more common in clinic. METHODS In this study, we reported a case of a Han-Chinese male newborn with ATP6V0A2 gene variant leading to cutis laxa. The proband was identified by whole-exome sequencing to determine the novel variant, and their parents were verified by Sanger sequencing. Bioinformatics analysis and minigene assay were used to verify the effect of this variant on splicing function. RESULTS The main manifestations of the proband are skin laxity, abnormal facial features, and enlargement of the anterior fontanelle. Whole-exome sequencing showed that the newborn carried a non-canonical splicing-site variant c.117 + 5G > T, p. (?) in ATP6V0A2 gene. Sanger sequencing showed that both parents of the proband carried the heterozygous variant. The results of bioinformatics analysis and minigene assay displayed that the variant site affected the splicing function of pre-mRNA of the ATP6V0A2 gene. CONCLUSIONS In this study, it was identified that ATP6V0A2 gene c. 117 + 5G > T may be the cause of the disease. The non-canonical splicing variants of ATP6V0A2 gene were rarely reported in the past, and this variant expanded the variants spectrum of the gene. The functional study of minigene assay plays a certain role in improving the level of evidence for the pathogenicity of splicing variants, which lays a foundation for prenatal counseling and follow-up gene therapy.
Collapse
Affiliation(s)
- Ying Zhang
- Graduate College of Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Mei Sun
- Graduate College of Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Na Li
- Graduate College of Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, China
- Department of Neonatology, Tianjin Children's Hospital (Children's Hospital of Tianjin University, No. 238 Longyan Road, Beichen District, Tianjin, 300134, China
| | - Yiran Zhao
- Graduate College of Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, China
- Maternal and Child Health Hospital of Tangshan, No. 14 Jianshe south Road, Lu nan District, Tangshan City, Hebei Province, 063000, China
| | - Fang Zhang
- Department of Neonatology, Tianjin Children's Hospital (Children's Hospital of Tianjin University, No. 238 Longyan Road, Beichen District, Tianjin, 300134, China
| | - Jianbo Shu
- Tianjin Children's Hospital (Children's Hospital of Tianjin University), No. 238 Longyan Road, Beichen District, Tianjin, 300134, China.
- Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, No. 238 Longyan Road, Beichen District, Tianjin, 300134, China.
- Tianjin Pediatric Research Institute, Tianjin Children's Hospital, No. 238 Longyan Road, Beichen District, Tianjin, 300134, China.
| | - Yang Liu
- Department of Neonatology, Tianjin Children's Hospital (Children's Hospital of Tianjin University, No. 238 Longyan Road, Beichen District, Tianjin, 300134, China.
| | - Chunquan Cai
- Tianjin Children's Hospital (Children's Hospital of Tianjin University), No. 238 Longyan Road, Beichen District, Tianjin, 300134, China.
- Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, No. 238 Longyan Road, Beichen District, Tianjin, 300134, China.
- Tianjin Pediatric Research Institute, Tianjin Children's Hospital, No. 238 Longyan Road, Beichen District, Tianjin, 300134, China.
| |
Collapse
|
4
|
Shafagh Shishavan N, Morovvati S. A novel deletion mutation in the ATP6V0A2 gene in an Iranian patient affected by autosomal recessive cutis laxa. Ir J Med Sci 2023; 192:2279-2282. [PMID: 36520350 DOI: 10.1007/s11845-022-03246-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022]
Abstract
Cutis laxa (CL) can be caused by mutations in a number of genes. Cutis laxa with autosomal recessive inheritance due to mutations in several genes, including mutations in the ATP6V0A2 gene, causes autosomal recessive cutis laxa type 2A (ARCL2A). The ATP6V0A2 gene encodes the a2 subunit in the V-ATPases pump. The V-ATPases are located in the membrane of some organelles, including the Golgi or some vesicles, and act as ATP-dependent proton pumps to pH adjustment intracellular segments. Mutations in the ATP6V0A2 gene consist present in ARCL2A patients. We present the case of a 12-year-old girl who was referred to Rasad Laboratory (Tehran, Iran) at the age of 5 with a set of symptoms of congenital disorders. Her clinical phenotype contains distal symmetrical sensory and motor polyneuropathy, loose joints, large nasal roots, growth delay, and wrinkled skin. Also, there was a history of the parental marriage of consanguinity. A potentially pathogenic homozygous deletion mutation was detected in the ATP6V0A2 gene related to ARCL2A. This mutation has not been reported in the other patients with ARCL2A. A novel homozygous deletion mutation in ATP6V0A2 is supposed to be the reason for disease in our proband.
Collapse
Affiliation(s)
- Negar Shafagh Shishavan
- Department of Genetics, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran
| | - Saeid Morovvati
- Department of Genetics, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
5
|
Fiumara A, Sapuppo A, Ferri L, Arena A, Prato A, Garozzo D, Sturiale L, Morrone A, Barone R. Higher frequency of TMEM199-CDG in the southern mediterranean area is associated with c.92G>C (p.Arg31Pro) mutation. Eur J Med Genet 2023; 66:104709. [PMID: 36706865 DOI: 10.1016/j.ejmg.2023.104709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 09/17/2022] [Accepted: 01/22/2023] [Indexed: 01/25/2023]
Abstract
Congenital disorders of glycosylation (CDG) are genetic multisystem diseases, characterized by defective glycoconjugate synthesis. A small number of CDG with isolated liver damage have been described, such as TMEM199-CDG, a non-encephalopathic liver disorder with Wilson disease-like phenotype. Only eight patients with TMEM199-CDG have been described including seven Europeans (originating from Greece and Italy) and one Chinese. Three patients from southern Italy (Campania) shared the same known missense mutation pathogenetic variant NM_152464.3:c. 92G > C (p.Arg31Pro), also found in a Greek patient. Here we report a new patient from southern Italy (Sicily), with a homozygous c.92G > C p.(Arg31Pro) variant in TMEM199. The patient's phenotype is characterized by mild non-progressive hepatopathy with a normal hepatic echo structure. A persistent increase in serum transaminases, total and low-density lipoprotein cholesterol and low serum ceruloplasmin and copper levels and normal urinary copper excretion were observed. Matrix-assisted laser desorption/ionization mass spectrometry analyses showed abnormal N- and O- protein glycosylation, indicative of a Golgi processing defect and supporting the function of TMEM199 in maintaining Golgi homeostasis. TMEM199-CDG is an ultra-rare CDG relatively frequent in the southern Mediterranean area (7 in 9 patients, 77%). It is mainly associated with the c.92G > C (p.Arg31Pro) pathogenetic allele globally reported in 4 out of 7 families (57%), including one from Greece and three unrelated families from southern Italy. The almost uniform clinical phenotype described in patients with TMEM199-CDG appears to reflect a higher prevalence of the same variant in patients from the southern Mediterranean area.
Collapse
Affiliation(s)
- Agata Fiumara
- CRR-MET, Department of Clinical and Experimental Medicine, University of Catania, Italy
| | - Annamaria Sapuppo
- CRR-MET, Department of Clinical and Experimental Medicine, University of Catania, Italy
| | - Lorenzo Ferri
- Molecular Biology of Neurometabolic Diseases, Neuroscience Department, Meyer Children's Hospital, Florence, Italy
| | - Alessia Arena
- CRR-MET, Department of Clinical and Experimental Medicine, University of Catania, Italy
| | - Adriana Prato
- Child Neuropsychiatry- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Domenico Garozzo
- Institute of Polymers, Composites and Biomaterials - CNR, Catania, Italy
| | - Luisa Sturiale
- Institute of Polymers, Composites and Biomaterials - CNR, Catania, Italy
| | - Amelia Morrone
- Molecular Biology of Neurometabolic Diseases, Neuroscience Department, Meyer Children's Hospital, Florence, Italy; Department of NEUROFARBA, University of Florence, Florence, Italy
| | - Rita Barone
- CRR-MET, Department of Clinical and Experimental Medicine, University of Catania, Italy; Child Neuropsychiatry- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy; Institute of Polymers, Composites and Biomaterials - CNR, Catania, Italy.
| |
Collapse
|
6
|
Barua S, Berger S, Pereira EM, Jobanputra V. Expanding the phenotype of ATP6AP1 deficiency. Cold Spring Harb Mol Case Stud 2022; 8:mcs.a006195. [PMID: 35732497 PMCID: PMC9235842 DOI: 10.1101/mcs.a006195] [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: 01/13/2022] [Accepted: 04/08/2022] [Indexed: 11/24/2022] Open
Abstract
Vacuolar ATPases (V-ATPases) are large multisubunit proton pumps conserved among all eukaryotic cells that are involved in diverse functions including acidification of membrane-bound intracellular compartments. The ATP6AP1 gene encodes an accessory subunit of the vacuolar (V)-ATPase protein pump. Pathogenic variants in ATP6AP1 have been described in association with a congenital disorder of glycosylation (CDG), which are highly variable, but often characterized by immunodeficiency, hepatopathy, and neurologic manifestations. Although the most striking and common clinical feature is hepatopathy, the phenotypic and genotypic spectrum of ATP6AP1-CDG continues to expand. Here, we report identical twins who presented with acute liver failure and jaundice. Prenatal features included cystic hygroma, atrial septal defect, and ventriculomegaly. Postnatal features included pectus carinatum, connective tissue abnormalities, and hypospadias. Whole-exome sequencing (WES) revealed a novel de novo in-frame deletion in the ATP6AP1 gene (c.230_232delACT;p.Tyr77del). Although both twins have the commonly reported clinical feature of hepatopathy seen in other individuals with ATP6AP1-CDG-related disorder, they do not have neurological sequelae. This report expands the phenotypic spectrum of ATP6AP1-CDG-related disorder with both probands exhibiting unique prenatal and postnatal features, including fetal ventriculomegaly, umbilical hernia, pectus carinatum, micropenis, and hypospadias. Furthermore, this case affirms that neurological features described in the initial case series on ATP6AP1-CDG do not appear to be central, whereas the prenatal and connective tissue manifestations may be more common than previously thought. This emphasizes the importance of long-term clinical follow-up and variant interpretation using current updated recommendations.
Collapse
Affiliation(s)
- Subit Barua
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York 10032, USA
| | - Sara Berger
- Division of Clinical Genetics, Department of Pediatrics, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York 10032, USA
| | - Elaine M Pereira
- Division of Clinical Genetics, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian Morgan Stanley Children's Hospital, New York, New York 10032, USA
| | - Vaidehi Jobanputra
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York 10032, USA
| |
Collapse
|
7
|
Hellicar J, Stevenson NL, Stephens DJ, Lowe M. Supply chain logistics - the role of the Golgi complex in extracellular matrix production and maintenance. J Cell Sci 2022; 135:273996. [PMID: 35023559 PMCID: PMC8767278 DOI: 10.1242/jcs.258879] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The biomechanical and biochemical properties of connective tissues are determined by the composition and quality of their extracellular matrix. This, in turn, is highly dependent on the function and organisation of the secretory pathway. The Golgi complex plays a vital role in directing matrix output by co-ordinating the post-translational modification and proteolytic processing of matrix components prior to their secretion. These modifications have broad impacts on the secretion and subsequent assembly of matrix components, as well as their function in the extracellular environment. In this Review, we highlight the role of the Golgi in the formation of an adaptable, healthy matrix, with a focus on proteoglycan and procollagen secretion as example cargoes. We then discuss the impact of Golgi dysfunction on connective tissue in the context of human disease and ageing.
Collapse
Affiliation(s)
- John Hellicar
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK.,Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673
| | - Nicola L Stevenson
- Cell Biology Laboratories, School of Biochemistry, Faculty of Life Sciences, University Walk, University of Bristol, Bristol, BS8 1TD, UK
| | - David J Stephens
- Cell Biology Laboratories, School of Biochemistry, Faculty of Life Sciences, University Walk, University of Bristol, Bristol, BS8 1TD, UK
| | - Martin Lowe
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| |
Collapse
|
8
|
Pottie L, Van Gool W, Vanhooydonck M, Hanisch FG, Goeminne G, Rajkovic A, Coucke P, Sips P, Callewaert B. Loss of zebrafish atp6v1e1b, encoding a subunit of vacuolar ATPase, recapitulates human ARCL type 2C syndrome and identifies multiple pathobiological signatures. PLoS Genet 2021; 17:e1009603. [PMID: 34143769 PMCID: PMC8244898 DOI: 10.1371/journal.pgen.1009603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 06/30/2021] [Accepted: 05/17/2021] [Indexed: 11/27/2022] Open
Abstract
The inability to maintain a strictly regulated endo(lyso)somal acidic pH through the proton-pumping action of the vacuolar-ATPases (v-ATPases) has been associated with various human diseases including heritable connective tissue disorders. Autosomal recessive (AR) cutis laxa (CL) type 2C syndrome is associated with genetic defects in the ATP6V1E1 gene and is characterized by skin wrinkles or loose redundant skin folds with pleiotropic systemic manifestations. The underlying pathological mechanisms leading to the clinical presentations remain largely unknown. Here, we show that loss of atp6v1e1b in zebrafish leads to early mortality, associated with craniofacial dysmorphisms, vascular anomalies, cardiac dysfunction, N-glycosylation defects, hypotonia, and epidermal structural defects. These features are reminiscent of the phenotypic manifestations in ARCL type 2C patients. Our data demonstrates that loss of atp6v1e1b alters endo(lyso)somal protein levels, and interferes with non-canonical v-ATPase pathways in vivo. In order to gain further insights into the processes affected by loss of atp6v1e1b, we performed an untargeted analysis of the transcriptome, metabolome, and lipidome in early atp6v1e1b-deficient larvae. We report multiple affected pathways including but not limited to oxidative phosphorylation, sphingolipid, fatty acid, and energy metabolism together with profound defects on mitochondrial respiration. Taken together, our results identify complex pathobiological effects due to loss of atp6v1e1b in vivo. Cutis laxa syndromes are pleiotropic disorders of the connective tissue, characterized by skin redundancy and variable systemic manifestations. Cutis laxa syndromes are caused by pathogenic variants in genes encoding structural and regulatory components of the extracellular matrix or in genes encoding components of cellular trafficking, metabolism, and mitochondrial function. Pathogenic variants in genes coding for vacuolar-ATPases, a multisubunit complex responsible for the acidification of multiple intracellular vesicles, cause type 2 cutis laxa syndromes, a group of cutis laxa subtypes further characterized by neurological, skeletal, and rarely cardiopulmonary manifestations. To investigate the pathomechanisms of vacuolar-ATPase dysfunction, we generated zebrafish models that lack a crucial subunit of the vacuolar-ATPases. The mutant zebrafish models show morphological and functional features reminiscent of the phenotypic manifestations in cutis laxa patients carrying pathogenic variants in ATP6V1E1. In-depth analysis at multiple -omic levels identified biological signatures that indicate impairment of signaling pathways, lipid metabolism, and mitochondrial respiration. We anticipate that these data will contribute to a better understanding of the pathogenesis of cutis laxa syndromes and other disorders involving defective v-ATPase function, which may eventually improve patient treatment and management.
Collapse
Affiliation(s)
- Lore Pottie
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Wouter Van Gool
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Michiel Vanhooydonck
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Franz-Georg Hanisch
- Institute of Biochemistry II, Medical Faculty, University of Cologne, Cologne, Germany
| | - Geert Goeminne
- VIB Metabolomics Core Ghent, Ghent, Belgium
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
| | - Andreja Rajkovic
- Department of Food technology, Safety and Health, Faculty of Bioscience Engineering, University of Ghent, Ghent, Belgium
| | - Paul Coucke
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Patrick Sips
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Bert Callewaert
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- * E-mail:
| |
Collapse
|
9
|
ATP7A-Regulated Enzyme Metalation and Trafficking in the Menkes Disease Puzzle. Biomedicines 2021; 9:biomedicines9040391. [PMID: 33917579 PMCID: PMC8067471 DOI: 10.3390/biomedicines9040391] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/01/2021] [Accepted: 04/02/2021] [Indexed: 12/12/2022] Open
Abstract
Copper is vital for numerous cellular functions affecting all tissues and organ systems in the body. The copper pump, ATP7A is critical for whole-body, cellular, and subcellular copper homeostasis, and dysfunction due to genetic defects results in Menkes disease. ATP7A dysfunction leads to copper deficiency in nervous tissue, liver, and blood but accumulation in other tissues. Site-specific cellular deficiencies of copper lead to loss of function of copper-dependent enzymes in all tissues, and the range of Menkes disease pathologies observed can now be explained in full by lack of specific copper enzymes. New pathways involving copper activated lysosomal and steroid sulfatases link patient symptoms usually related to other inborn errors of metabolism to Menkes disease. Additionally, new roles for lysyl oxidase in activation of molecules necessary for the innate immune system, and novel adapter molecules that play roles in ERGIC trafficking of brain receptors and other proteins, are emerging. We here summarize the current knowledge of the roles of copper enzyme function in Menkes disease, with a focus on ATP7A-mediated enzyme metalation in the secretory pathway. By establishing mechanistic relationships between copper-dependent cellular processes and Menkes disease symptoms in patients will not only increase understanding of copper biology but will also allow for the identification of an expanding range of copper-dependent enzymes and pathways. This will raise awareness of rare patient symptoms, and thus aid in early diagnosis of Menkes disease patients.
Collapse
|
10
|
Morlino S, Nardella G, Castellana S, Micale L, Copetti M, Fusco C, Castori M. Review of clinical and molecular variability in autosomal recessive cutis laxa 2A. Am J Med Genet A 2020; 185:955-965. [PMID: 33369135 DOI: 10.1002/ajmg.a.62047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 11/10/2020] [Accepted: 12/13/2020] [Indexed: 11/06/2022]
Abstract
ATP6V0A2-related cutis laxa, also known as autosomal recessive cutis laxa type 2A (ARCL2A), is a subtype of hereditary cutis laxa originally characterized by skin, skeletal, and neurological involvement, and a combined defect of N-glycosylation and O-glycosylation. The associated clinical spectrum subsequently expanded to a less severe phenotype dominated by cutaneous involvement. At the moment, ARCL2A was described in a few case reports and series only. An Italian adult woman ARCL2A with a phenotype restricted to skin and the two novel c.3G>C and c.1101dup ATP6V0A2 variants has been reported. A systematic literature review allowed us to identify 69 additional individuals from 64 families. Available data were scrutinized in order to describe the clinical and molecular variability of ARCL2A. About 78.3% of known variants were predicted null alleles, while 11 were missense and 2 affected noncanonical splice sites. Age at ascertainment appeared as the unique phenotypic discriminator with earlier age more commonly associated with facial dysmorphism (p .02), high/cleft palate (p .005), intellectual disability/global developmental delay (p .013), and seizures (p .024). No specific genotype-phenotype correlations were identified. This work confirmed the existence of an attenuated phenotype associated with ATP6V0A2 biallelic variants and offers an updated critique to the clinical and molecular variability of ARCL2A.
Collapse
Affiliation(s)
- Silvia Morlino
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, Foggia, Italy
| | - Grazia Nardella
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, Foggia, Italy
| | - Stefano Castellana
- Unit of Bioinformatics, Fondazione IRCCS-Casa Sollievo della Sofferenza, Foggia, Italy
| | - Lucia Micale
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, Foggia, Italy
| | - Massimiliano Copetti
- Unit of Biostatistics, Fondazione IRCCS-Casa Sollievo della Sofferenza, Foggia, Italy
| | - Carmela Fusco
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, Foggia, Italy
| | - Marco Castori
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, Foggia, Italy
| |
Collapse
|
11
|
Mohamed M, Gardeitchik T, Balasubramaniam S, Guerrero‐Castillo S, Dalloyaux D, van Kraaij S, Venselaar H, Hoischen A, Urban Z, Brandt U, Al‐Shawi R, Simons JP, Frison M, Ngu L, Callewaert B, Spelbrink H, Kallemeijn WW, Aerts JMFG, Waugh MG, Morava E, Wevers RA. Novel defect in phosphatidylinositol 4-kinase type 2-alpha (PI4K2A) at the membrane-enzyme interface is associated with metabolic cutis laxa. J Inherit Metab Dis 2020; 43:1382-1391. [PMID: 32418222 PMCID: PMC7687218 DOI: 10.1002/jimd.12255] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 05/11/2020] [Accepted: 05/13/2020] [Indexed: 12/16/2022]
Abstract
Inherited cutis laxa, or inelastic, sagging skin is a genetic condition of premature and generalised connective tissue ageing, affecting various elastic components of the extracellular matrix. Several cutis laxa syndromes are inborn errors of metabolism and lead to severe neurological symptoms. In a patient with cutis laxa, a choreoathetoid movement disorder, dysmorphic features and intellectual disability we performed exome sequencing to elucidate the underlying genetic defect. We identified the amino acid substitution R275W in phosphatidylinositol 4-kinase type IIα, caused by a homozygous missense mutation in the PI4K2A gene. We used lipidomics, complexome profiling and functional studies to measure phosphatidylinositol 4-phosphate synthesis in the patient and evaluated PI4K2A deficient mice to define a novel metabolic disorder. The R275W residue, located on the surface of the protein, is involved in forming electrostatic interactions with the membrane. The catalytic activity of PI4K2A in patient fibroblasts was severely reduced and lipid mass spectrometry showed that particular acyl-chain pools of PI4P and PI(4,5)P2 were decreased. Phosphoinositide lipids play a major role in intracellular signalling and trafficking and regulate the balance between proliferation and apoptosis. Phosphatidylinositol 4-kinases such as PI4K2A mediate the first step in the main metabolic pathway that generates PI4P, PI(4,5)P2 and PI(3,4,5)P3 . Although neurologic involvement is common, cutis laxa has not been reported previously in metabolic defects affecting signalling. Here we describe a patient with a complex neurological phenotype, premature ageing and a mutation in PI4K2A, illustrating the importance of this enzyme in the generation of inositol lipids with particular acylation characteristics.
Collapse
Affiliation(s)
- Miski Mohamed
- Department of PaediatricsRadboud University Medical CenterNijmegenThe Netherlands
| | - Thatjana Gardeitchik
- Department of PaediatricsRadboud University Medical CenterNijmegenThe Netherlands
- Department of GeneticsRadboud University Medical CenterNijmegenThe Netherlands
| | - Shanti Balasubramaniam
- Clinical Genetic DepartmentHospital Kuala Lumpur, Jalan PahangKuala LumpurMalaysia
- Discipline of Genetic Medicine, Sydney Medical SchoolUniversity of SydneySydneyNew South WalesAustralia
- Western Sydney Genetics ProgramThe Children's Hospital at WestmeadSydneyNew South WalesAustralia
| | - Sergio Guerrero‐Castillo
- Radboud Center for Mitochondrial MedicineRadboud University Medical CenterNijmegenThe Netherlands
- Translational Metabolic Laboratory, Department of Laboratory MedicineRadboud University Medical CenterNijmegenThe Netherlands
| | - Daisy Dalloyaux
- Department of PaediatricsRadboud University Medical CenterNijmegenThe Netherlands
| | - Sanne van Kraaij
- Translational Metabolic Laboratory, Department of Laboratory MedicineRadboud University Medical CenterNijmegenThe Netherlands
| | - Hanka Venselaar
- Center of Molecular and Biomolecular InformaticsRadboud University Medical CenterNijmegenThe Netherlands
| | - Alexander Hoischen
- Department of GeneticsRadboud University Medical CenterNijmegenThe Netherlands
- Department of Internal MedicineRadboud University Medical CenterNijmegenThe Netherlands
- Radboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Zsolt Urban
- Department of Human Genetics, Graduate School of Public HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Ulrich Brandt
- Radboud Center for Mitochondrial MedicineRadboud University Medical CenterNijmegenThe Netherlands
- Translational Metabolic Laboratory, Department of Laboratory MedicineRadboud University Medical CenterNijmegenThe Netherlands
| | - Raya Al‐Shawi
- Wolfson Drug Discovery Unit, Division of Medicine, Royal Free CampusUniversity College LondonLondonUK
| | - J. Paul Simons
- Wolfson Drug Discovery Unit, Division of Medicine, Royal Free CampusUniversity College LondonLondonUK
| | - Michele Frison
- Wolfson Drug Discovery Unit, Division of Medicine, Royal Free CampusUniversity College LondonLondonUK
| | - Lock‐Hock Ngu
- Clinical Genetic DepartmentHospital Kuala Lumpur, Jalan PahangKuala LumpurMalaysia
| | - Bert Callewaert
- Center for Medical GeneticsGhent University HospitalGhentBelgium
| | - Hans Spelbrink
- Department of PaediatricsRadboud University Medical CenterNijmegenThe Netherlands
| | - Wouter W. Kallemeijn
- Department of Medical Biochemistry, Leiden Institute of ChemistryLeiden UniversityLeidenThe Netherlands
- Department of ChemistryImperial College LondonLondonUK
| | - Johannes M. F. G. Aerts
- Department of Medical Biochemistry, Leiden Institute of ChemistryLeiden UniversityLeidenThe Netherlands
| | - Mark G. Waugh
- Lipid and Membrane Biology Group, Institute for Liver & Digestive HealthUniversity College LondonLondonUK
| | - Eva Morava
- Haywards Genetics CenterTulane UniversityNew OrleansLouisianaUSA
- Department of PediatricsUniversity Medical CentreLeuvenBelgium
| | - Ron A. Wevers
- Translational Metabolic Laboratory, Department of Laboratory MedicineRadboud University Medical CenterNijmegenThe Netherlands
| |
Collapse
|
12
|
González-Domínguez CA, López-Valdez J, Martínez-Duncker Ramírez I, Salinas-Marín R. Análisis de la mutación c.187 C>T en el gen ATP6V0A2 mediante PCR-ARMS. TIP REVISTA ESPECIALIZADA EN CIENCIAS QUÍMICO-BIOLÓGICAS 2020. [DOI: 10.22201/fesz.23958723e.2020.0.262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Los desórdenes congénitos de la glicosilación (CDG) son enfermedades poco frecuentes (EPOF) de tipo metabólico y hereditarias que ocurren como consecuencia de mutaciones en los genes que codifican para proteínas que participan, directa o indirectamente, en este proceso. La enfermedad clínicamente denominada Cutis Laxa Autosómica Recesiva tipo II-A (ARCL2A) es un tipo de CDG (ATP6V0A2-CDG) causado por mutaciones en ATP6V0A2, que codifica para la subunidad a2 del dominio v0 de una ATPasa vacuolar que tiene como función el transporte de iones H+ a través de las membranas celulares, regulando así el pH de los compartimentos celulares, e incluye la acidificación del aparato de Golgi. En 2014, nuestro grupo de investigación reportó por primera vez en México, la existencia de dos pacientes con ATP6V0A2-CDG. En este trabajo, se estableció una metodología para identificar a los portadores de la mutación c.187 C>T en el ATP6V0A2 mediante PCR-ARMS.
Collapse
|
13
|
Abstract
La glycosylation est un processus cellulaire complexe conduisant à des transferts successifs de monosaccharides sur une molécule acceptrice, le plus souvent une protéine ou un lipide. Ce processus est universel chez tous les organismes vivants et est très conservé au cours de l’évolution. Chez l’homme, des perturbations survenant au cours d’une ou plusieurs réactions de glycosylation sont à l’origine de glycopathologies génétiques rares, appelées anomalies congénitales de la glycosylation ou congenital disorders of glycosylation (CDG). Cette revue propose de revisiter ces CDG, de 1980 à aujourd’hui, en présentant leurs découvertes, leurs diagnostics, leurs causes biochimiques et les traitements actuellement disponibles.
Collapse
|
14
|
Song Q, Meng B, Xu H, Mao Z. The emerging roles of vacuolar-type ATPase-dependent Lysosomal acidification in neurodegenerative diseases. Transl Neurodegener 2020; 9:17. [PMID: 32393395 PMCID: PMC7212675 DOI: 10.1186/s40035-020-00196-0] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 04/23/2020] [Indexed: 12/15/2022] Open
Abstract
Background Lysosomes digest extracellular material from the endocytic pathway and intracellular material from the autophagic pathway. This process is performed by the resident hydrolytic enzymes activated by the highly acidic pH within the lysosomal lumen. Lysosome pH gradients are mainly maintained by the vacuolar (H+) ATPase (or V-ATPase), which pumps protons into lysosomal lumen by consuming ATP. Dysfunction of V-ATPase affects lysosomal acidification, which disrupts the clearance of substrates and leads to many disorders, including neurodegenerative diseases. Main body As a large multi-subunit complex, the V-ATPase is composed of an integral membrane V0 domain involved in proton translocation and a peripheral V1 domain catalyzing ATP hydrolysis. The canonical functions of V-ATPase rely on its H+-pumping ability in multiple vesicle organelles to regulate endocytic traffic, protein processing and degradation, synaptic vesicle loading, and coupled transport. The other non-canonical effects of the V-ATPase that are not readily attributable to its proton-pumping activity include membrane fusion, pH sensing, amino-acid-induced activation of mTORC1, and scaffolding for protein-protein interaction. In response to various stimuli, V-ATPase complex can reversibly dissociate into V1 and V0 domains and thus close ATP-dependent proton transport. Dysregulation of pH and lysosomal dysfunction have been linked to many human diseases, including neurodegenerative disorders such as Alzheimer disease, Parkinson’s disease, amyotrophic lateral sclerosis as well as neurodegenerative lysosomal storage disorders. Conclusion V-ATPase complex is a universal proton pump and plays an important role in lysosome acidification in all types of cells. Since V-ATPase dysfunction contributes to the pathogenesis of multiple neurodegenerative diseases, further understanding the mechanisms that regulate the canonical and non-canonical functions of V-ATPase will reveal molecular details of disease process and help assess V-ATPase or molecules related to its regulation as therapeutic targets.
Collapse
Affiliation(s)
- Qiaoyun Song
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Department of Reproductive Genetics, Hebei General Hospital, Shijiazhuang, Hebei Province, 050051, People's Republic of China.,Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Bo Meng
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Haidong Xu
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Zixu Mao
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA. .,Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
15
|
Wada Y. Matrix-Assisted Laser Desorption/Ionization Mass Spectrometry to Detect Diagnostic Glycopeptide Markers of Congenital Disorders of Glycosylation. ACTA ACUST UNITED AC 2020; 9:A0084. [PMID: 32547898 PMCID: PMC7242785 DOI: 10.5702/massspectrometry.a0084] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 03/04/2020] [Indexed: 12/04/2022]
Abstract
Congenital disorders of glycosylation (CDG), an increasingly recognized group of diseases that affect glycosylation, comprise the largest known subgroup of approximately 100 responsible genes related to N-glycosylation. This subgroup presents various molecular abnormalities, of either the CDG-I or the CDG-II type, attributable to a lack of glycans or abnormal glycoform profiles, respectively. The most effective approach to identifying these N-glycosylation disorders is mass spectrometry (MS) using either released glycans, intact glycoproteins or proteolytic peptides as analytes. Among these, MS of tryptic peptides derived from transferrin can be used to reliably identify signature peptides that are characteristic of CDG-I and II. In the present study, matrix-assisted laser desorption/ionization (MALDI) MS was applied to various N-glycosylation disorders including ALG1-CDG, B4GALT1-CDG, SLC35A2-CDG, ATP6V0A2-CDG, TRAPPC11-CDG and MAN1B1-CDG. This method does not require the prior enrichment of glycopeptides or chromatographic separation, and thus serves as a practical alternative to liquid chromatography-electrospray ionization MS. The signature peptides are biomarkers of CDG.
Collapse
Affiliation(s)
- Yoshinao Wada
- Osaka Women's and Children's Hospital (OWCH), 840 Murodo-cho, Izumi, Osaka 594-1101, Japan
| |
Collapse
|
16
|
A new pH sensor localized in the Golgi apparatus of Saccharomyces cerevisiae reveals unexpected roles of Vph1p and Stv1p isoforms. Sci Rep 2020; 10:1881. [PMID: 32024908 PMCID: PMC7002768 DOI: 10.1038/s41598-020-58795-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/21/2020] [Indexed: 12/22/2022] Open
Abstract
The gradual acidification of the secretory pathway is conserved and extremely important for eukaryotic cells, but until now there was no pH sensor available to monitor the pH of the early Golgi apparatus in Saccharomyces cerevisiae. Therefore, we developed a pHluorin-based sensor for in vivo measurements in the lumen of the Golgi. By using this new tool we show that the cis- and medial-Golgi pH is equal to 6.6–6.7 in wild type cells during exponential phase. As expected, V-ATPase inactivation results in a near neutral Golgi pH. We also uncover that surprisingly Vph1p isoform of the V-ATPase is prevalent to Stv1p for Golgi acidification. Additionally, we observe that during changes of the cytosolic pH, the Golgi pH is kept relatively stable, mainly thanks to the V-ATPase. Eventually, this new probe will allow to better understand the mechanisms involved in the acidification and the pH control within the secretory pathway.
Collapse
|
17
|
Peterson TV, Jaiswal MK, Beaman KD, Reynolds JM. Conditional Deletion of the V-ATPase a2-Subunit Disrupts Intrathymic T Cell Development. Front Immunol 2019; 10:1911. [PMID: 31456807 PMCID: PMC6700305 DOI: 10.3389/fimmu.2019.01911] [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: 01/25/2019] [Accepted: 07/29/2019] [Indexed: 11/13/2022] Open
Abstract
Proper orchestration of T lymphocyte development is critical, as T cells underlie nearly all responses of the adaptive immune system. Developing thymocytes differentiate in response to environmental cues carried from cell surface receptors to the nucleus, shaping a distinct transcriptional program that defines their developmental outcome. Our recent work has identified a previously undescribed role for the vacuolar ATPase (V-ATPase) in facilitating the development of murine thymocytes progressing toward the CD4+ and CD8+ αβ T cell lineages. Vav1Cre recombinase-mediated deletion of the a2 isoform of the V-ATPase (a2V) in mouse hematopoietic cells leads to a specific and profound loss of peripheral CD4+ and CD8+ αβ T cells. Utilizing T cell-restricted LckCre and CD4Cre strains, we further traced this deficiency to the thymus and found that a2V plays a cell-intrinsic role throughout intrathymic development. Loss of a2V manifests as a partial obstruction in the double negative stage of T cell development, and later, a near complete failure of positive selection. These data deepen our understanding of the biological mechanisms that orchestrate T cell development and lend credence to the recent focus on V-ATPase as a potential chemotherapeutic target to combat proliferative potential in T cell lymphoblastic leukemias and autoimmune disease.
Collapse
Affiliation(s)
- Theodore V Peterson
- Center for Cancer Cell Biology, Immunology and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Mukesh K Jaiswal
- Center for Cancer Cell Biology, Immunology and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Kenneth D Beaman
- Center for Cancer Cell Biology, Immunology and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Joseph M Reynolds
- Center for Cancer Cell Biology, Immunology and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States.,Edward Hines, Jr. VA Hospital, Hines, IL, United States
| |
Collapse
|
18
|
Sahoo M, Katara GK, Bilal MY, Ibrahim SA, Kulshrestha A, Fleetwood S, Suzue K, Beaman KD. Hematopoietic stem cell specific V-ATPase controls breast cancer progression and metastasis via cytotoxic T cells. Oncotarget 2018; 9:33215-33231. [PMID: 30237863 PMCID: PMC6145706 DOI: 10.18632/oncotarget.26061] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 08/13/2018] [Indexed: 01/11/2023] Open
Abstract
The interaction of recruited immune effector cells and cancer cells within tumor microenvironment (TME) shapes the fate of cancer progression and metastasis. Many cancers including breast cancer, express a specific vacuolar ATPase (a2V) on their cell surface which acidifies the extracellular milieu helping cancer cell proliferation and metastasis. To understand the role of immune cell-associated-a2V during breast tumor pathogenesis, we knocked-out a2V (KO) from the hematopoietic stem cells (HSC) and generated breast tumors in mice. The a2V-KO mice developed faster growing, larger, and metastatic breast tumors compared to control mice. Further investigation of the TME revealed a significant reduction in the presence of CD4+ and CD8+ T cells in the a2V-KO tumors. Targeted RNA-Seq of the cells of the TME demonstrated that pro-inflammatory cytokines, death receptors, death receptor ligands, and cytotoxic effectors were significantly down-regulated within the a2V-KO TME. Interestingly, analysis of immune cells in the blood, spleen, and thymus of the non-tumor bearing a2V-KO mice revealed a significant decrease in CD4+ and CD8+ T cell populations. For the first time, this study demonstrates that inhibition of V-ATPase expression in HSC leads to a decrease in CD4+ and CD8+ T cell populations and thus promotes breast tumor growth and metastasis.
Collapse
Affiliation(s)
- Manoranjan Sahoo
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Gajendra K Katara
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Mahmood Y Bilal
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Safaa A Ibrahim
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Arpita Kulshrestha
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Sara Fleetwood
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Kimiko Suzue
- Department of Pathology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Kenneth D Beaman
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| |
Collapse
|
19
|
Bruneel A, Cholet S, Drouin-Garraud V, Jacquemont ML, Cano A, Mégarbané A, Ruel C, Cheillan D, Dupré T, Vuillaumier-Barrot S, Seta N, Fenaille F. Complementarity of electrophoretic, mass spectrometric, and gene sequencing techniques for the diagnosis and characterization of congenital disorders of glycosylation. Electrophoresis 2018; 39:3123-3132. [PMID: 29869806 DOI: 10.1002/elps.201800021] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 05/09/2018] [Accepted: 05/25/2018] [Indexed: 12/25/2022]
Abstract
Congenital disorders of glycosylation (CDG) are rare autosomal genetic diseases affecting the glycosylation of proteins and lipids. Since CDG-related clinical symptoms are classically extremely variable and nonspecific, a combination of electrophoretic, mass spectrometric, and gene sequencing techniques is often mandatory for obtaining a definitive CDG diagnosis, as well as identifying causative gene mutations and deciphering the underlying biochemical mechanisms. Here, we illustrate the potential of integrating data from capillary electrophoresis of transferrin, two-dimensional electrophoresis of N- and O-glycoproteins, mass spectrometry analyses of total serum N-linked glycans and mucin core1 O-glycosylated apolipoprotein C-III for the determination of various culprit CDG gene mutations. "Step-by-step" diagnosis pathways of four particular and new CDG cases, including MGAT2-CDG, ATP6V0A2-CDG, SLC35A2-CDG, and SLC35A3-CDG, are described as illustrative examples.
Collapse
Affiliation(s)
- Arnaud Bruneel
- AP-HP, Biochimie Métabolique et Cellulaire, Hôpital Bichat-Claude Bernard, Paris, France.,INSERM UMR-1193 "Mécanismes cellulaires et moléculaires de l'adaptation au stress et cancérogenèse", Université Paris-Sud, Châtenay-Malabry, France
| | - Sophie Cholet
- Service de Pharmacologie et d'Immunoanalyse, Laboratoire d'Etude du Métabolisme des Médicaments, CEA, INRA, Université Paris Saclay, Gif-sur-Yvette, France
| | | | | | - Aline Cano
- Centre de Référence des Maladies Héréditaires du Métabolisme, CHU la Timone-Marseille, Marseille, France
| | | | - Coralie Ruel
- Service de Pharmacologie et d'Immunoanalyse, Laboratoire d'Etude du Métabolisme des Médicaments, CEA, INRA, Université Paris Saclay, Gif-sur-Yvette, France.,Proteins and Nanotechnology in Analytical Science (PNAS), CNRS, Université Paris-Sud, Châtenay-Malabry, France
| | - David Cheillan
- Service de Biochimie et Biologie Moléculaire Grand Est, UM Pathologies Métaboliques, Erythrocytaires et Dépistage Périnatal, Centre de Biologie et de Pathologie Est, Groupement Hospitalier Est-Hospices Civils de Lyon, Bron, France
| | - Thierry Dupré
- AP-HP, Biochimie Métabolique et Cellulaire, Hôpital Bichat-Claude Bernard, Paris, France
| | | | - Nathalie Seta
- AP-HP, Biochimie Métabolique et Cellulaire, Hôpital Bichat-Claude Bernard, Paris, France.,Paris Descartes University, Paris, France
| | - François Fenaille
- Service de Pharmacologie et d'Immunoanalyse, Laboratoire d'Etude du Métabolisme des Médicaments, CEA, INRA, Université Paris Saclay, Gif-sur-Yvette, France
| |
Collapse
|
20
|
Pamarthy S, Kulshrestha A, Katara GK, Beaman KD. The curious case of vacuolar ATPase: regulation of signaling pathways. Mol Cancer 2018; 17:41. [PMID: 29448933 PMCID: PMC5815226 DOI: 10.1186/s12943-018-0811-3] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 02/07/2018] [Indexed: 02/06/2023] Open
Abstract
The Vacuolar ATPase (V-ATPase) is a proton pump responsible for controlling the intracellular and extracellular pH of cells. The structure of V-ATPase has been highly conserved among all eukaryotic cells and is involved in diverse functions across species. V-ATPase is best known for its acidification of endosomes and lysosomes and is also important for luminal acidification of specialized cells. Several reports have suggested the involvement of V-ATPase in maintaining an alkaline intracellular and acidic extracellular pH thereby aiding in proliferation and metastasis of cancer cells respectively. Increased expression of V-ATPase and relocation to the plasma membrane aids in cancer modulates key tumorigenic cell processes like autophagy, Warburg effect, immunomoduation, drug resistance and most importantly cancer cell signaling. In this review, we discuss the direct role of V-ATPase in acidification and indirect regulation of signaling pathways, particularly Notch Signaling.
Collapse
Affiliation(s)
- Sahithi Pamarthy
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, 60611, USA
| | - Arpita Kulshrestha
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Gajendra K Katara
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Kenneth D Beaman
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA.
| |
Collapse
|
21
|
Vajro P, Zielinska K, Ng BG, Maccarana M, Bengtson P, Poeta M, Mandato C, D'Acunto E, Freeze HH, Eklund EA. Three unreported cases of TMEM199-CDG, a rare genetic liver disease with abnormal glycosylation. Orphanet J Rare Dis 2018; 13:4. [PMID: 29321044 PMCID: PMC5763540 DOI: 10.1186/s13023-017-0757-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Accepted: 12/29/2017] [Indexed: 01/21/2023] Open
Abstract
Background TMEM199 deficiency was recently shown in four patients to cause liver disease with steatosis, elevated serum transaminases, cholesterol and alkaline phosphatase and abnormal protein glycosylation. There is no information on the long-term outcome in this disorder. Results We here present three novel patients with TMEM199-CDG. All three patients carried the same set of mutations (c.13-14delTT (p.Ser4Serfs*30) and c.92G > C (p.Arg31Pro), despite only two were related (siblings). One mutation (c.92G > C) was described previously whereas the other was deemed pathogenic due to its early frameshift. Western Blot analysis confirmed a reduced level of TMEM199 protein in patient fibroblasts and all patients showed a similar glycosylation defect. The patients presented with a very similar clinical and biochemical phenotype to the initial publication, confirming that TMEM199-CDG is a non-encephalopathic liver disorder. Two of the patients were clinically assessed over two decades without deterioration. Conclusion A rising number of disorders affecting Golgi homeostasis have been published over the last few years. A hallmark finding is deficiency in protein glycosylation, both in N- and O-linked types. Most of these disorders have signs of both liver and brain involvement. However, the present and the four previously reported patients do not show encephalopathy but a chronic, non-progressive (over decades) liver disease with hypertransaminasemia and steatosis. This information is crucial for the patient/families and clinician at diagnosis, as it distinguishes it from other Golgi homeostasis disorders, in having a much more favorable course.
Collapse
Affiliation(s)
- Pietro Vajro
- Unit of Pediatrics, Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, Baronissi, (Sa), Italy
| | | | - Bobby G Ng
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Marco Maccarana
- Section for Matrix Biology, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Per Bengtson
- Division of Clinical Chemistry, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Marco Poeta
- Unit of Pediatrics, Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, Baronissi, (Sa), Italy
| | - Claudia Mandato
- Children's Hospital "Santobono-Pausilipon", 1st Division of Pediatrics, Naples, Italy
| | - Elisa D'Acunto
- Unit of Pediatrics, Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, Baronissi, (Sa), Italy
| | - Hudson H Freeze
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Erik A Eklund
- Division of Pediatrics, Lund University, Lund, Sweden.
| |
Collapse
|
22
|
Esmail S, Kartner N, Yao Y, Kim JW, Reithmeier RAF, Manolson MF. Molecular mechanisms of cutis laxa- and distal renal tubular acidosis-causing mutations in V-ATPase a subunits, ATP6V0A2 and ATP6V0A4. J Biol Chem 2018; 293:2787-2800. [PMID: 29311258 DOI: 10.1074/jbc.m117.818872] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 12/20/2017] [Indexed: 12/19/2022] Open
Abstract
The a subunit is the largest of 15 different subunits that make up the vacuolar H+-ATPase (V-ATPase) complex, where it functions in proton translocation. In mammals, this subunit has four paralogous isoforms, a1-a4, which may encode signals for targeting assembled V-ATPases to specific intracellular locations. Despite the functional importance of the a subunit, its structure remains controversial. By studying molecular mechanisms of human disease-causing missense mutations within a subunit isoforms, we may identify domains critical for V-ATPase targeting, activity and/or regulation. cDNA-encoded FLAG-tagged human wildtype ATP6V0A2 (a2) and ATP6V0A4 (a4) subunits and their mutants, a2P405L (causing cutis laxa), and a4R449H and a4G820R (causing renal tubular acidosis, dRTA), were transiently expressed in HEK 293 cells. N-Glycosylation was assessed using endoglycosidases, revealing that a2P405L, a4R449H, and a4G820R were fully N-glycosylated. Cycloheximide (CHX) chase assays revealed that a2P405L and a4R449H were unstable relative to wildtype. a4R449H was degraded predominantly in the proteasomal pathway, whereas a2P405L was degraded in both proteasomal and lysosomal pathways. Immunofluorescence studies disclosed retention in the endoplasmic reticulum and defective cell-surface expression of a4R449H and defective Golgi trafficking of a2P405L Co-immunoprecipitation studies revealed an increase in association of a4R449H with the V0 assembly factor VMA21, and a reduced association with the V1 sector subunit, ATP6V1B1 (B1). For a4G820R, where stability, degradation, and trafficking were relatively unaffected, 3D molecular modeling suggested that the mutation causes dRTA by blocking the proton pathway. This study provides critical information that may assist rational drug design to manage dRTA and cutis laxa.
Collapse
Affiliation(s)
- Sally Esmail
- Dental Research Institute, Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1G6
| | - Norbert Kartner
- Dental Research Institute, Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1G6
| | - Yeqi Yao
- Dental Research Institute, Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1G6
| | - Joo Wan Kim
- Dental Research Institute, Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1G6
| | | | - Morris F Manolson
- Dental Research Institute, Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1G6; Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| |
Collapse
|
23
|
Katara GK, Kulshrestha A, Mao L, Wang X, Sahoo M, Ibrahim S, Pamarthy S, Suzue K, Shekhawat GS, Gilman-Sachs A, Beaman KD. Mammary epithelium-specific inactivation of V-ATPase reduces stiffness of extracellular matrix and enhances metastasis of breast cancer. Mol Oncol 2017; 12:208-223. [PMID: 29178186 PMCID: PMC5792725 DOI: 10.1002/1878-0261.12159] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 09/25/2017] [Accepted: 11/07/2017] [Indexed: 01/01/2023] Open
Abstract
Extracellular matrix (ECM) critically impacts tumor progression and is influenced by both cancer and host tissue cells. While our understanding of cancer cell ECM remodeling is widespread, the importance of host tissue ECM, which provides initial congenial environment for primary tumor formation, is partly understood. Here, we report a novel role of epithelial cell-associated vacuolar ATPase 'a2' isoform (a2V) in regulating breast tissue ECM stiffness to control metastasis. Using a mammary gland-specific a2V-knockout model, we show that in the absence of a2V, breast tumors exhibit atypically soft tumor phenotype, less tumor rigidity, and necrotic tumor microenvironment. These tumors contain a decreased number of cancer cells at primary tumor site, but showed extensive metastases compared to control. Nanomechanical evaluation of normal breast tissues revealed a decrease in stiffness and collagen content in ECM of a2V-deleted breast tissues. Mechanistically, inhibition of a2V expression caused dispersed Golgi morphology with relocation of glycosyltransferase enzymes to early endosomes in mammary epithelial cells. This resulted in defective glycosylation of ECM proteins and production of compromised ECM that further influenced tumor metastasis. Clinically, in patients with cancer, low a2V expression levels in normal breast tissue correlated with lymph node metastasis. Thus, using a new knockout mouse model, we have identified a2V expression in epithelial cells as a key requirement for proper ECM formation in breast tissue and its expression levels can significantly modulate breast tumor dissemination. Evaluation of a2V expression in normal breast tissues can help in identifying patients with high risk of developing metastases.
Collapse
Affiliation(s)
- Gajendra K Katara
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Arpita Kulshrestha
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Liqun Mao
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Xin Wang
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
| | - Manoranjan Sahoo
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Safaa Ibrahim
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Sahithi Pamarthy
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Kimiko Suzue
- Department of Pathology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA.,Advocate Lutheran General Hospital, Park Ridge, IL, USA
| | - Gajendra S Shekhawat
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
| | - Alice Gilman-Sachs
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Kenneth D Beaman
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| |
Collapse
|
24
|
Van Damme T, Gardeitchik T, Mohamed M, Guerrero-Castillo S, Freisinger P, Guillemyn B, Kariminejad A, Dalloyaux D, van Kraaij S, Lefeber DJ, Syx D, Steyaert W, De Rycke R, Hoischen A, Kamsteeg EJ, Wong SY, van Scherpenzeel M, Jamali P, Brandt U, Nijtmans L, Korenke GC, Chung BHY, Mak CCY, Hausser I, Kornak U, Fischer-Zirnsak B, Strom TM, Meitinger T, Alanay Y, Utine GE, Leung PKC, Ghaderi-Sohi S, Coucke P, Symoens S, De Paepe A, Thiel C, Haack TB, Malfait F, Morava E, Callewaert B, Wevers RA. Mutations in ATP6V1E1 or ATP6V1A Cause Autosomal-Recessive Cutis Laxa. Am J Hum Genet 2017; 100:216-227. [PMID: 28065471 DOI: 10.1016/j.ajhg.2016.12.010] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 12/08/2016] [Indexed: 02/03/2023] Open
Abstract
Defects of the V-type proton (H+) ATPase (V-ATPase) impair acidification and intracellular trafficking of membrane-enclosed compartments, including secretory granules, endosomes, and lysosomes. Whole-exome sequencing in five families affected by mild to severe cutis laxa, dysmorphic facial features, and cardiopulmonary involvement identified biallelic missense mutations in ATP6V1E1 and ATP6V1A, which encode the E1 and A subunits, respectively, of the V1 domain of the heteromultimeric V-ATPase complex. Structural modeling indicated that all substitutions affect critical residues and inter- or intrasubunit interactions. Furthermore, complexome profiling, a method combining blue-native gel electrophoresis and liquid chromatography tandem mass spectrometry, showed that they disturb either the assembly or the stability of the V-ATPase complex. Protein glycosylation was variably affected. Abnormal vesicular trafficking was evidenced by delayed retrograde transport after brefeldin A treatment and abnormal swelling and fragmentation of the Golgi apparatus. In addition to showing reduced and fragmented elastic fibers, the histopathological hallmark of cutis laxa, transmission electron microscopy of the dermis also showed pronounced changes in the structure and organization of the collagen fibers. Our findings expand the clinical and molecular spectrum of metabolic cutis laxa syndromes and further link defective extracellular matrix assembly to faulty protein processing and cellular trafficking caused by genetic defects in the V-ATPase complex.
Collapse
Affiliation(s)
- Tim Van Damme
- Center for Medical Genetics, Ghent University and Ghent University Hospital, Ghent 9000, Belgium
| | - Thatjana Gardeitchik
- Department of Pediatrics, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands; Department of Human Genetics, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands
| | - Miski Mohamed
- Department of Pediatrics, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands
| | - Sergio Guerrero-Castillo
- Radboud Center for Mitochondrial Medicine, Department of Pediatrics, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands; Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands
| | - Peter Freisinger
- Childrens' Hospital, Klinikum am Steinenberg, Reutlingen 72764, Germany
| | - Brecht Guillemyn
- Center for Medical Genetics, Ghent University and Ghent University Hospital, Ghent 9000, Belgium
| | | | - Daisy Dalloyaux
- Department of Pediatrics, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands; Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands
| | - Sanne van Kraaij
- Department of Pediatrics, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands; Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands
| | - Dirk J Lefeber
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands; Department of Neurology, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands
| | - Delfien Syx
- Center for Medical Genetics, Ghent University and Ghent University Hospital, Ghent 9000, Belgium
| | - Wouter Steyaert
- Center for Medical Genetics, Ghent University and Ghent University Hospital, Ghent 9000, Belgium
| | - Riet De Rycke
- Department of Biomedical Molecular Biology, Ghent University, Ghent 9000, Belgium; Inflammation Research Center, VIB, Ghent 9000, Belgium
| | - Alexander Hoischen
- Department of Human Genetics, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands
| | - Erik-Jan Kamsteeg
- Department of Human Genetics, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands
| | - Sunnie Y Wong
- Hayward Genetics Center, Tulane University Medical School, New Orleans, LA 70112, USA
| | - Monique van Scherpenzeel
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands; Department of Neurology, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands
| | - Payman Jamali
- Shahrood Genetic Counseling Center, Semnan 36156, Iran
| | - Ulrich Brandt
- Radboud Center for Mitochondrial Medicine, Department of Pediatrics, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands; Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands
| | - Leo Nijtmans
- Radboud Center for Mitochondrial Medicine, Department of Pediatrics, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands; Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands
| | - G Christoph Korenke
- Department of Neuropediatrics, Children's Hospital Klinikum Oldenburg, Oldenburg 26133, Germany
| | - Brian H Y Chung
- Department of Paediatrics & Adolescent Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Christopher C Y Mak
- Department of Paediatrics & Adolescent Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Ingrid Hausser
- Institute of Pathology, Universitätsklinikum Heidelberg, Heidelberg 69120, Germany
| | - Uwe Kornak
- Institute of Medical Genetics and Human Genetics, Charité - Universitaetsmedizin Berlin, Berlin 13353, Germany; Max Planck Institute for Molecular Genetics, Berlin 14195, Germany
| | - Björn Fischer-Zirnsak
- Institute of Medical Genetics and Human Genetics, Charité - Universitaetsmedizin Berlin, Berlin 13353, Germany; Max Planck Institute for Molecular Genetics, Berlin 14195, Germany
| | - Tim M Strom
- Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg 85764, Germany
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg 85764, Germany
| | - Yasemin Alanay
- Pediatric Genetics Unit, Department of Pediatrics, Acibadem University School of Medicine, Istanbul 34752, Turkey
| | - Gulen E Utine
- Pediatric Genetics Unit, Department of Pediatrics, Ihsan Doğramacı Children's Hospital, Hacettepe School of Medicine, Ankara 06100, Turkey
| | - Peter K C Leung
- Department of Paediatrics & Adolescent Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | | | - Paul Coucke
- Center for Medical Genetics, Ghent University and Ghent University Hospital, Ghent 9000, Belgium
| | - Sofie Symoens
- Center for Medical Genetics, Ghent University and Ghent University Hospital, Ghent 9000, Belgium
| | - Anne De Paepe
- Center for Medical Genetics, Ghent University and Ghent University Hospital, Ghent 9000, Belgium
| | - Christian Thiel
- Center for Child and Adolescent Medicine, Klinik Kinderheilkunde I, Universitätsklinikum Heidelberg, Heidelberg 69120, Germany
| | - Tobias B Haack
- Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg 85764, Germany; Institute of Human Genetics, Technische Universität München, Munich 81675, Germany; Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen 72076, Germany
| | - Fransiska Malfait
- Center for Medical Genetics, Ghent University and Ghent University Hospital, Ghent 9000, Belgium
| | - Eva Morava
- Hayward Genetics Center, Tulane University Medical School, New Orleans, LA 70112, USA; Department of Pediatrics, University Hospital Leuven, Leuven 3000, Belgium
| | - Bert Callewaert
- Center for Medical Genetics, Ghent University and Ghent University Hospital, Ghent 9000, Belgium.
| | - Ron A Wevers
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands.
| |
Collapse
|
25
|
Colacurcio DJ, Nixon RA. Disorders of lysosomal acidification-The emerging role of v-ATPase in aging and neurodegenerative disease. Ageing Res Rev 2016; 32:75-88. [PMID: 27197071 DOI: 10.1016/j.arr.2016.05.004] [Citation(s) in RCA: 320] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/02/2016] [Accepted: 05/13/2016] [Indexed: 12/21/2022]
Abstract
Autophagy and endocytosis deliver unneeded cellular materials to lysosomes for degradation. Beyond processing cellular waste, lysosomes release metabolites and ions that serve signaling and nutrient sensing roles, linking the functions of the lysosome to various pathways for intracellular metabolism and nutrient homeostasis. Each of these lysosomal behaviors is influenced by the intraluminal pH of the lysosome, which is maintained in the low acidic range by a proton pump, the vacuolar ATPase (v-ATPase). New reports implicate altered v-ATPase activity and lysosomal pH dysregulation in cellular aging, longevity, and adult-onset neurodegenerative diseases, including forms of Parkinson disease and Alzheimer disease. Genetic defects of subunits composing the v-ATPase or v-ATPase-related proteins occur in an increasingly recognized group of familial neurodegenerative diseases. Here, we review the expanding roles of the v-ATPase complex as a platform regulating lysosomal hydrolysis and cellular homeostasis. We discuss the unique vulnerability of neurons to persistent low level lysosomal dysfunction and review recent clinical and experimental studies that link dysfunction of the v-ATPase complex to neurodegenerative diseases across the age spectrum.
Collapse
|
26
|
Jansen J, Timal S, van Scherpenzeel M, Michelakakis H, Vicogne D, Ashikov A, Moraitou M, Hoischen A, Huijben K, Steenbergen G, van den Boogert M, Porta F, Calvo P, Mavrikou M, Cenacchi G, van den Bogaart G, Salomon J, Holleboom A, Rodenburg R, Drenth J, Huynen M, Wevers R, Morava E, Foulquier F, Veltman J, Lefeber D. TMEM199 Deficiency Is a Disorder of Golgi Homeostasis Characterized by Elevated Aminotransferases, Alkaline Phosphatase, and Cholesterol and Abnormal Glycosylation. Am J Hum Genet 2016; 98:322-30. [PMID: 26833330 DOI: 10.1016/j.ajhg.2015.12.011] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 12/14/2015] [Indexed: 01/01/2023] Open
Abstract
Congenital disorders of glycosylation (CDGs) form a genetically and clinically heterogeneous group of diseases with aberrant protein glycosylation as a hallmark. A subgroup of CDGs can be attributed to disturbed Golgi homeostasis. However, identification of pathogenic variants is seriously complicated by the large number of proteins involved. As part of a strategy to identify human homologs of yeast proteins that are known to be involved in Golgi homeostasis, we identified uncharacterized transmembrane protein 199 (TMEM199, previously called C17orf32) as a human homolog of yeast V-ATPase assembly factor Vph2p (also known as Vma12p). Subsequently, we analyzed raw exome-sequencing data from families affected by genetically unsolved CDGs and identified four individuals with different mutations in TMEM199. The adolescent individuals presented with a mild phenotype of hepatic steatosis, elevated aminotransferases and alkaline phosphatase, and hypercholesterolemia, as well as low serum ceruloplasmin. Affected individuals showed abnormal N- and mucin-type O-glycosylation, and mass spectrometry indicated reduced incorporation of galactose and sialic acid, as seen in other Golgi homeostasis defects. Metabolic labeling of sialic acids in fibroblasts confirmed deficient Golgi glycosylation, which was restored by lentiviral transduction with wild-type TMEM199. V5-tagged TMEM199 localized with ERGIC and COPI markers in HeLa cells, and electron microscopy of a liver biopsy showed dilated organelles suggestive of the endoplasmic reticulum and Golgi apparatus. In conclusion, we have identified TMEM199 as a protein involved in Golgi homeostasis and show that TMEM199 deficiency results in a hepatic phenotype with abnormal glycosylation.
Collapse
|
27
|
Jansen J, Cirak S, van Scherpenzeel M, Timal S, Reunert J, Rust S, Pérez B, Vicogne D, Krawitz P, Wada Y, Ashikov A, Pérez-Cerdá C, Medrano C, Arnoldy A, Hoischen A, Huijben K, Steenbergen G, Quelhas D, Diogo L, Rymen D, Jaeken J, Guffon N, Cheillan D, van den Heuvel L, Maeda Y, Kaiser O, Schara U, Gerner P, van den Boogert M, Holleboom A, Nassogne MC, Sokal E, Salomon J, van den Bogaart G, Drenth J, Huynen M, Veltman J, Wevers R, Morava E, Matthijs G, Foulquier F, Marquardt T, Lefeber D. CCDC115 Deficiency Causes a Disorder of Golgi Homeostasis with Abnormal Protein Glycosylation. Am J Hum Genet 2016; 98:310-21. [PMID: 26833332 DOI: 10.1016/j.ajhg.2015.12.010] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 12/11/2015] [Indexed: 01/06/2023] Open
Abstract
Disorders of Golgi homeostasis form an emerging group of genetic defects. The highly heterogeneous clinical spectrum is not explained by our current understanding of the underlying cell-biological processes in the Golgi. Therefore, uncovering genetic defects and annotating gene function are challenging. Exome sequencing in a family with three siblings affected by abnormal Golgi glycosylation revealed a homozygous missense mutation, c.92T>C (p.Leu31Ser), in coiled-coil domain containing 115 (CCDC115), the function of which is unknown. The same mutation was identified in three unrelated families, and in one family it was compound heterozygous in combination with a heterozygous deletion of CCDC115. An additional homozygous missense mutation, c.31G>T (p.Asp11Tyr), was found in a family with two affected siblings. All individuals displayed a storage-disease-like phenotype involving hepatosplenomegaly, which regressed with age, highly elevated bone-derived alkaline phosphatase, elevated aminotransferases, and elevated cholesterol, in combination with abnormal copper metabolism and neurological symptoms. Two individuals died of liver failure, and one individual was successfully treated by liver transplantation. Abnormal N- and mucin type O-glycosylation was found on serum proteins, and reduced metabolic labeling of sialic acids was found in fibroblasts, which was restored after complementation with wild-type CCDC115. PSI-BLAST homology detection revealed reciprocal homology with Vma22p, the yeast V-ATPase assembly factor located in the endoplasmic reticulum (ER). Human CCDC115 mainly localized to the ERGIC and to COPI vesicles, but not to the ER. These data, in combination with the phenotypic spectrum, which is distinct from that associated with defects in V-ATPase core subunits, suggest a more general role for CCDC115 in Golgi trafficking. Our study reveals CCDC115 deficiency as a disorder of Golgi homeostasis that can be readily identified via screening for abnormal glycosylation in plasma.
Collapse
|
28
|
Impaired ATP6V0A2 expression contributes to Golgi dispersion and glycosylation changes in senescent cells. Sci Rep 2015; 5:17342. [PMID: 26611489 PMCID: PMC4661525 DOI: 10.1038/srep17342] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 10/29/2015] [Indexed: 02/07/2023] Open
Abstract
Many genes and signaling pathways have been found to be involved in cellular senescence program. In the present study, we have identified 16 senescence-associated genes by differential proteomic analysis of the normal human diploid fibroblast cell line, TIG-1, and focused on ATP6V0A2. The aim of this study is to clarify the role of ATP6V0A2, the causal gene for ARCL2, a syndrome of abnormal glycosylation and impaired Golgi trafficking, in cellular senescence program. Here we showed that ATP6V0A2 is critical for cellular senescence; impaired expression of ATP6V0A2 disperses the Golgi structure and triggers senescence, suggesting that ATP6V0A2 mediates these processes. FITC-lectin staining and glycoblotting revealed significantly different glycosylation structures in presenescent (young) and senescent (old) TIG-1 cells; reducing ATP6V0A2 expression in young TIG-1 cells yielded structures similar to those in old TIG-1 cells. Our results suggest that senescence-associated impaired expression of ATP6V0A2 triggers changes in Golgi structure and glycosylation in old TIG-1 cells, which demonstrates a role of ATP6V0A2 in cellular senescence program.
Collapse
|
29
|
Lu X, Meima ME, Nelson JK, Sorrentino V, Loregger A, Scheij S, Dekkers DHW, Mulder MT, Demmers JAA, M-Dallinga-Thie G, Zelcer N, Danser AHJ. Identification of the (Pro)renin Receptor as a Novel Regulator of Low-Density Lipoprotein Metabolism. Circ Res 2015; 118:222-9. [PMID: 26582775 DOI: 10.1161/circresaha.115.306799] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 11/18/2015] [Indexed: 01/16/2023]
Abstract
RATIONALE The (pro)renin receptor ([P]RR) interacts with (pro)renin at concentrations that are >1000× higher than observed under (patho)physiological conditions. Recent studies have identified renin-angiotensin system-independent functions for (P)RR related to its association with the vacuolar H(+)-ATPase. OBJECTIVE To uncover renin-angiotensin system-independent functions of the (P)RR. METHODS AND RESULTS We used a proteomics-based approach to purify and identify (P)RR-interacting proteins. This resulted in identification of sortilin-1 (SORT1) as a high-confidence (P)RR-interacting protein, a finding which was confirmed by coimmunoprecipitation of endogenous (P)RR and SORT1. Functionally, silencing (P)RR expression in hepatocytes decreased SORT1 and low-density lipoprotein (LDL) receptor protein abundance and, as a consequence, resulted in severely attenuated cellular LDL uptake. In contrast to LDL, endocytosis of epidermal growth factor or transferrin remained unaffected by silencing of the (P)RR. Importantly, reduction of LDL receptor and SORT1 protein abundance occurred in the absence of changes in their corresponding transcript level. Consistent with a post-transcriptional event, degradation of the LDL receptor induced by (P)RR silencing could be reversed by lysosomotropic agents, such as bafilomycin A1. CONCLUSIONS Our study identifies a renin-angiotensin system-independent function for the (P)RR in the regulation of LDL metabolism by controlling the levels of SORT1 and LDL receptor.
Collapse
Affiliation(s)
- Xifeng Lu
- From the Astra Zeneca-Shenzhen University Joint Institute of Nephrology, Shenzhen University Medical Center, Shenzhen University, Shenzhen, China (X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine (X.L., M.E.M., M.T.M., A.H.J.D.) and Proteomics Center (D.H.W.D., J.A.A.D.), Erasmus Medical Center, Rotterdam, The Netherlands; and Department of Medical Biochemistry (X.L., J.K.N., V.S., A.L., S.S., N.Z.) and Laboratory of Experimental Vascular Medicine (G.M.D-.T.), Academic Medical Center, Amsterdam, The Netherlands
| | - Marcel E Meima
- From the Astra Zeneca-Shenzhen University Joint Institute of Nephrology, Shenzhen University Medical Center, Shenzhen University, Shenzhen, China (X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine (X.L., M.E.M., M.T.M., A.H.J.D.) and Proteomics Center (D.H.W.D., J.A.A.D.), Erasmus Medical Center, Rotterdam, The Netherlands; and Department of Medical Biochemistry (X.L., J.K.N., V.S., A.L., S.S., N.Z.) and Laboratory of Experimental Vascular Medicine (G.M.D-.T.), Academic Medical Center, Amsterdam, The Netherlands
| | - Jessica K Nelson
- From the Astra Zeneca-Shenzhen University Joint Institute of Nephrology, Shenzhen University Medical Center, Shenzhen University, Shenzhen, China (X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine (X.L., M.E.M., M.T.M., A.H.J.D.) and Proteomics Center (D.H.W.D., J.A.A.D.), Erasmus Medical Center, Rotterdam, The Netherlands; and Department of Medical Biochemistry (X.L., J.K.N., V.S., A.L., S.S., N.Z.) and Laboratory of Experimental Vascular Medicine (G.M.D-.T.), Academic Medical Center, Amsterdam, The Netherlands
| | - Vincenzo Sorrentino
- From the Astra Zeneca-Shenzhen University Joint Institute of Nephrology, Shenzhen University Medical Center, Shenzhen University, Shenzhen, China (X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine (X.L., M.E.M., M.T.M., A.H.J.D.) and Proteomics Center (D.H.W.D., J.A.A.D.), Erasmus Medical Center, Rotterdam, The Netherlands; and Department of Medical Biochemistry (X.L., J.K.N., V.S., A.L., S.S., N.Z.) and Laboratory of Experimental Vascular Medicine (G.M.D-.T.), Academic Medical Center, Amsterdam, The Netherlands
| | - Anke Loregger
- From the Astra Zeneca-Shenzhen University Joint Institute of Nephrology, Shenzhen University Medical Center, Shenzhen University, Shenzhen, China (X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine (X.L., M.E.M., M.T.M., A.H.J.D.) and Proteomics Center (D.H.W.D., J.A.A.D.), Erasmus Medical Center, Rotterdam, The Netherlands; and Department of Medical Biochemistry (X.L., J.K.N., V.S., A.L., S.S., N.Z.) and Laboratory of Experimental Vascular Medicine (G.M.D-.T.), Academic Medical Center, Amsterdam, The Netherlands
| | - Saskia Scheij
- From the Astra Zeneca-Shenzhen University Joint Institute of Nephrology, Shenzhen University Medical Center, Shenzhen University, Shenzhen, China (X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine (X.L., M.E.M., M.T.M., A.H.J.D.) and Proteomics Center (D.H.W.D., J.A.A.D.), Erasmus Medical Center, Rotterdam, The Netherlands; and Department of Medical Biochemistry (X.L., J.K.N., V.S., A.L., S.S., N.Z.) and Laboratory of Experimental Vascular Medicine (G.M.D-.T.), Academic Medical Center, Amsterdam, The Netherlands
| | - Dick H W Dekkers
- From the Astra Zeneca-Shenzhen University Joint Institute of Nephrology, Shenzhen University Medical Center, Shenzhen University, Shenzhen, China (X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine (X.L., M.E.M., M.T.M., A.H.J.D.) and Proteomics Center (D.H.W.D., J.A.A.D.), Erasmus Medical Center, Rotterdam, The Netherlands; and Department of Medical Biochemistry (X.L., J.K.N., V.S., A.L., S.S., N.Z.) and Laboratory of Experimental Vascular Medicine (G.M.D-.T.), Academic Medical Center, Amsterdam, The Netherlands
| | - Monique T Mulder
- From the Astra Zeneca-Shenzhen University Joint Institute of Nephrology, Shenzhen University Medical Center, Shenzhen University, Shenzhen, China (X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine (X.L., M.E.M., M.T.M., A.H.J.D.) and Proteomics Center (D.H.W.D., J.A.A.D.), Erasmus Medical Center, Rotterdam, The Netherlands; and Department of Medical Biochemistry (X.L., J.K.N., V.S., A.L., S.S., N.Z.) and Laboratory of Experimental Vascular Medicine (G.M.D-.T.), Academic Medical Center, Amsterdam, The Netherlands
| | - Jeroen A A Demmers
- From the Astra Zeneca-Shenzhen University Joint Institute of Nephrology, Shenzhen University Medical Center, Shenzhen University, Shenzhen, China (X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine (X.L., M.E.M., M.T.M., A.H.J.D.) and Proteomics Center (D.H.W.D., J.A.A.D.), Erasmus Medical Center, Rotterdam, The Netherlands; and Department of Medical Biochemistry (X.L., J.K.N., V.S., A.L., S.S., N.Z.) and Laboratory of Experimental Vascular Medicine (G.M.D-.T.), Academic Medical Center, Amsterdam, The Netherlands
| | - Geesje M-Dallinga-Thie
- From the Astra Zeneca-Shenzhen University Joint Institute of Nephrology, Shenzhen University Medical Center, Shenzhen University, Shenzhen, China (X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine (X.L., M.E.M., M.T.M., A.H.J.D.) and Proteomics Center (D.H.W.D., J.A.A.D.), Erasmus Medical Center, Rotterdam, The Netherlands; and Department of Medical Biochemistry (X.L., J.K.N., V.S., A.L., S.S., N.Z.) and Laboratory of Experimental Vascular Medicine (G.M.D-.T.), Academic Medical Center, Amsterdam, The Netherlands
| | - Noam Zelcer
- From the Astra Zeneca-Shenzhen University Joint Institute of Nephrology, Shenzhen University Medical Center, Shenzhen University, Shenzhen, China (X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine (X.L., M.E.M., M.T.M., A.H.J.D.) and Proteomics Center (D.H.W.D., J.A.A.D.), Erasmus Medical Center, Rotterdam, The Netherlands; and Department of Medical Biochemistry (X.L., J.K.N., V.S., A.L., S.S., N.Z.) and Laboratory of Experimental Vascular Medicine (G.M.D-.T.), Academic Medical Center, Amsterdam, The Netherlands.
| | - A H Jan Danser
- From the Astra Zeneca-Shenzhen University Joint Institute of Nephrology, Shenzhen University Medical Center, Shenzhen University, Shenzhen, China (X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine (X.L., M.E.M., M.T.M., A.H.J.D.) and Proteomics Center (D.H.W.D., J.A.A.D.), Erasmus Medical Center, Rotterdam, The Netherlands; and Department of Medical Biochemistry (X.L., J.K.N., V.S., A.L., S.S., N.Z.) and Laboratory of Experimental Vascular Medicine (G.M.D-.T.), Academic Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
30
|
Prydz K. Determinants of Glycosaminoglycan (GAG) Structure. Biomolecules 2015; 5:2003-22. [PMID: 26308067 PMCID: PMC4598785 DOI: 10.3390/biom5032003] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 08/17/2015] [Accepted: 08/18/2015] [Indexed: 01/05/2023] Open
Abstract
Proteoglycans (PGs) are glycosylated proteins of biological importance at cell surfaces, in the extracellular matrix, and in the circulation. PGs are produced and modified by glycosaminoglycan (GAG) chains in the secretory pathway of animal cells. The most common GAG attachment site is a serine residue followed by a glycine (-ser-gly-), from which a linker tetrasaccharide extends and may continue as a heparan sulfate, a heparin, a chondroitin sulfate, or a dermatan sulfate GAG chain. Which type of GAG chain becomes attached to the linker tetrasaccharide is influenced by the structure of the protein core, modifications occurring to the linker tetrasaccharide itself, and the biochemical environment of the Golgi apparatus, where GAG polymerization and modification by sulfation and epimerization take place. The same cell type may produce different GAG chains that vary, depending on the extent of epimerization and sulfation. However, it is not known to what extent these differences are caused by compartmental segregation of protein cores en route through the secretory pathway or by differential recruitment of modifying enzymes during synthesis of different PGs. The topic of this review is how different aspects of protein structure, cellular biochemistry, and compartmentalization may influence GAG synthesis.
Collapse
Affiliation(s)
- Kristian Prydz
- Department of Biosciences, University of Oslo, Box 1066, Blindern OSLO 0316, Norway.
| |
Collapse
|
31
|
Yen-Nicolaÿ S, Boursier C, Rio M, Lefeber DJ, Pilon A, Seta N, Bruneel A. MALDI-TOF MS applied to apoC-III glycoforms of patients with congenital disorders affecting O-glycosylation. Comparison with two-dimensional electrophoresis. Proteomics Clin Appl 2015; 9:787-93. [PMID: 25641685 DOI: 10.1002/prca.201400187] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 01/07/2015] [Indexed: 11/06/2022]
Abstract
PURPOSE The O-glycan abnormalities accompanying some congenital disorders of glycosylation, namely conserved oligomeric Golgi-congenital disorders of glycosylation (COG-CDGs) and ATP6V0A2-CDGs, are mainly detected using electrophoresis methods applied to circulating apolipoprotein C-III. The objective of this study was to evaluate the reliability of MALDI-TOF MS of apoC-III for the detection and characterization of CDG-associated O-glycan defects. EXPERIMENTAL DESIGN plasmas from CDG-negative, COG-CDG, and ATP6V0A2-CDG patients were analyzed and results were compared to those obtained using 2DE followed by Western blot. RESULTS MALDI-TOF of apoC-III allowed to detect various significant O-glycan abnormalities in CDG-patients with emphasis to COG-CDG. Furthermore, in CDG samples, comparison study between 2DE and MALDI-TOF showed a particular behavior of monosialylated apoC-III in the mass spectrometer that could be related to an abnormal O-glycan structure. CONCLUSIONS AND CLINICAL RELEVANCE MALDI-TOF MS appears as a powerful technique for the analysis of apoC-III glycoforms for potential routine screening of COG- and ATP6V0A2-CDGs.
Collapse
Affiliation(s)
| | - Céline Boursier
- Trans-Prot, Proteomic facility, Université Paris-Sud, Châtenay-Malabry, France
| | - Marlène Rio
- Département de Génétique, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Dirk J Lefeber
- Laboratory of Genetics, Endocrine and Metabolic Diseases, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Antoine Pilon
- EA4530, Dynamique des microtubules en physiopathologie, Faculté de Pharmacie, Université Paris-Sud, Châtenay-Malabry, France.,APHP, Unité d'Hormonologie et Immunoanalyse, Pôle de Biologie Médicale et Pathologie, Hôpitaux Universitaires Est Parisien, Paris, France
| | - Nathalie Seta
- AP-HP, Biochimie métabolique et cellulaire, hôpital Bichat, Paris, France
| | - Arnaud Bruneel
- EA4530, Dynamique des microtubules en physiopathologie, Faculté de Pharmacie, Université Paris-Sud, Châtenay-Malabry, France.,AP-HP, Biochimie métabolique et cellulaire, hôpital Bichat, Paris, France
| |
Collapse
|
32
|
Bammens R, Mehta N, Race V, Foulquier F, Jaeken J, Tiemeyer M, Steet R, Matthijs G, Flanagan-Steet H. Abnormal cartilage development and altered N-glycosylation in Tmem165-deficient zebrafish mirrors the phenotypes associated with TMEM165-CDG. Glycobiology 2015; 25:669-82. [PMID: 25609749 DOI: 10.1093/glycob/cwv009] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 01/13/2015] [Indexed: 11/13/2022] Open
Abstract
The congenital disorders of glycosylation (CDG), a group of inherited diseases characterized by aberrant glycosylation, encompass a wide range of defects, including glycosyltransferases, glycosidases, nucleotide-sugar transporters as well as proteins involved in maintaining Golgi architecture, pH and vesicular trafficking. Mutations in a previously undescribed protein, TMEM165, were recently shown to cause a new form of CDG, termed TMEM165-CDG. TMEM165-CDG patients exhibit cartilage and bone dysplasia and altered glycosylation of serum glycoproteins. We utilized a morpholino knockdown strategy in zebrafish to investigate the physiologic and pathogenic functions of TMEM165. Inhibition of tmem165 expression in developing zebrafish embryos caused craniofacial abnormalities, largely attributable to fewer chondrocytes. Decreased expression of several markers of cartilage and bone development suggests that Tmem165 deficiency alters both chondrocyte and osteoblast differentiation. Glycomic analysis of tmem165 morphants also revealed altered initiation, processing and extension of N-glycans, paralleling some of the glycosylation changes noted in human patients. Collectively, these findings highlight the utility of zebrafish to elucidate pathogenic mechanisms associated with glycosylation disorders and suggest that the cartilage and bone dysplasia manifested in TMEM165-CDG patients may stem from abnormal development of chondrocytes and osteoblasts.
Collapse
Affiliation(s)
- Riet Bammens
- Center for Human Genetics, University of Leuven, 3000 Leuven, Belgium
| | - Nickita Mehta
- Complex Carbohydrate Research Center, Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Valérie Race
- Center for Human Genetics, University of Leuven, 3000 Leuven, Belgium
| | - François Foulquier
- CNRS-UMR 8576, Structural and Functional Glycobiology Unit, IFR 147, University of Lille 1, 59655 Villeneuve d'Ascq, France
| | - Jaak Jaeken
- Center for Metabolic Disease, University Hospital Gasthuisberg, 3000 Leuven, Belgium
| | - Michael Tiemeyer
- Complex Carbohydrate Research Center, Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Richard Steet
- Complex Carbohydrate Research Center, Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Gert Matthijs
- Center for Human Genetics, University of Leuven, 3000 Leuven, Belgium
| | - Heather Flanagan-Steet
- Complex Carbohydrate Research Center, Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
33
|
Ong YS, Tran THT, Gounko NV, Hong W. TMEM115 is an integral membrane protein of the Golgi complex involved in retrograde transport. J Cell Sci 2014; 127:2825-39. [PMID: 24806965 PMCID: PMC4077589 DOI: 10.1242/jcs.136754] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Searching and evaluating the Human Protein Atlas for transmembrane proteins enabled us to identify an integral membrane protein, TMEM115, that is enriched in the Golgi complex. Biochemical and cell biological analysis suggested that TMEM115 has four candidate transmembrane domains located in the N-terminal region. Both the N- and C-terminal domains are oriented towards the cytoplasm. Immunofluorescence analysis supports that TMEM115 is enriched in the Golgi cisternae. Functionally, TMEM115 knockdown or overexpression delays Brefeldin-A-induced Golgi-to-ER retrograde transport, phenocopying cells with mutations or silencing of the conserved oligomeric Golgi (COG) complex. Co-immunoprecipitation and in vitro binding experiments reveals that TMEM115 interacts with the COG complex, and might self-interact to form dimers or oligomers. A short region (residues 206–229) immediately to the C-terminal side of the fourth transmembrane domain is both necessary and sufficient for Golgi targeting. Knockdown of TMEM115 also reduces the binding of the lectins peanut agglutinin (PNA) and Helix pomatia agglutinin (HPA), suggesting an altered O-linked glycosylation profile. These results establish that TMEM115 is an integral membrane protein of the Golgi stack regulating Golgi-to-ER retrograde transport and is likely to be part of the machinery of the COG complex.
Collapse
Affiliation(s)
- Yan Shan Ong
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Ton Hoai Thi Tran
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Natalia V Gounko
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Singapore 138673, Singapore IMB-IMCB Joint Electron Microscopy Suite, 20 Biopolis Street, Singapore 138671, Singapore
| | - Wanjin Hong
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Singapore 138673, Singapore Department of Biochemistry, National University of Singapore, Singapore 117599, Singapore
| |
Collapse
|
34
|
ATP6V0A2 mutations present in two Mexican Mestizo children with an autosomal recessive cutis laxa syndrome type IIA. Mol Genet Metab Rep 2014; 1:203-212. [PMID: 27896089 PMCID: PMC5121299 DOI: 10.1016/j.ymgmr.2014.04.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 04/02/2014] [Accepted: 04/02/2014] [Indexed: 11/30/2022] Open
Abstract
Patients with ARCL-IIA harbor mutations in ATP6V0A2 that codes for an organelle proton pump. The ARCL-IIA syndrome characteristically presents a combined glycosylation defect affecting N-linked and O-linked glycosylations, differentiating it from other cutis laxa syndromes and classifying it as a Congenital Disorder of Glycosylation (ATP6V0A2-CDG). We studied two Mexican Mestizo patients with a clinical phenotype corresponding to an ARCL-IIA syndrome. Both patients presented abnormal transferrin (N-linked) glycosylation but Patient 1 had a normal ApoCIII (O-linked) glycosylation profile. Mutational screening of ATP6V0A2 using cDNA and genomic DNA revealed in Patient 1 a previously reported homozygous nonsense mutation c.187C>T (p.R63X) associated with a novel clinical finding of a VSD. In Patient 2 we found a homozygous c.2293C>T (p.Q765X) mutation that had been previously reported but found that it also altered RNA processing generating a novel transcript not previously identified (r.2176_2293del; p.F726Sfs*10). This is the first report to describe Mestizo patients with molecular diagnosis of ARCL-IIA/ATP6V0A2-CDG and to establish that their mutations are the first to be found in patients from different regions of the world and with different genetic backgrounds.
Collapse
|
35
|
Marshansky V, Rubinstein JL, Grüber G. Eukaryotic V-ATPase: novel structural findings and functional insights. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2014; 1837:857-79. [PMID: 24508215 DOI: 10.1016/j.bbabio.2014.01.018] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Revised: 12/25/2013] [Accepted: 01/27/2014] [Indexed: 02/06/2023]
Abstract
The eukaryotic V-type adenosine triphosphatase (V-ATPase) is a multi-subunit membrane protein complex that is evolutionarily related to F-type adenosine triphosphate (ATP) synthases and A-ATP synthases. These ATPases/ATP synthases are functionally conserved and operate as rotary proton-pumping nano-motors, invented by Nature billions of years ago. In the first part of this review we will focus on recent structural findings of eukaryotic V-ATPases and discuss the role of different subunits in the function of the V-ATPase holocomplex. Despite structural and functional similarities between rotary ATPases, the eukaryotic V-ATPases are the most complex enzymes that have acquired some unconventional cellular functions during evolution. In particular, the novel roles of V-ATPases in the regulation of cellular receptors and their trafficking via endocytotic and exocytotic pathways were recently uncovered. In the second part of this review we will discuss these unique roles of V-ATPases in modulation of function of cellular receptors, involved in the development and progression of diseases such as cancer and diabetes as well as neurodegenerative and kidney disorders. Moreover, it was recently revealed that the V-ATPase itself functions as an evolutionarily conserved pH sensor and receptor for cytohesin-2/Arf-family GTP-binding proteins. Thus, in the third part of the review we will evaluate the structural basis for and functional insights into this novel concept, followed by the analysis of the potentially essential role of V-ATPase in the regulation of this signaling pathway in health and disease. Finally, future prospects for structural and functional studies of the eukaryotic V-ATPase will be discussed.
Collapse
Affiliation(s)
- Vladimir Marshansky
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Simches Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA; Kadmon Pharmaceuticals Corporation, Alexandria Center for Life Science, 450 East 29th Street, New York, NY 10016, USA.
| | - John L Rubinstein
- Molecular Structure and Function Program, The Hospital for Sick Children Research Institute, University of Toronto, Toronto, ON M5G 1X8, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5G 1X8, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Gerhard Grüber
- Nanyang Technological University, Division of Structural Biology and Biochemistry, School of Biological Sciences, Singapore 637551, Republic of Singapore; Bioinformatics Institute, A(⁎)STAR, 60 Nanyang Drive, Singapore 637551, Republic of Singapore
| |
Collapse
|
36
|
Vacuolar H+-ATPase: An Essential Multitasking Enzyme in Physiology and Pathophysiology. ACTA ACUST UNITED AC 2014. [DOI: 10.1155/2014/675430] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Vacuolar H+-ATPases (V-ATPases) are large multisubunit proton pumps that are required for housekeeping acidification of membrane-bound compartments in eukaryotic cells. Mammalian V-ATPases are composed of 13 different subunits. Their housekeeping functions include acidifying endosomes, lysosomes, phagosomes, compartments for uncoupling receptors and ligands, autophagosomes, and elements of the Golgi apparatus. Specialized cells, including osteoclasts, intercalated cells in the kidney and pancreatic beta cells, contain both the housekeeping V-ATPases and an additional subset of V-ATPases, which plays a cell type specific role. The specialized V-ATPases are typically marked by the inclusion of cell type specific isoforms of one or more of the subunits. Three human diseases caused by mutations of isoforms of subunits have been identified. Cancer cells utilize V-ATPases in unusual ways; characterization of V-ATPases may lead to new therapeutic modalities for the treatment of cancer. Two accessory proteins to the V-ATPase have been identified that regulate the proton pump. One is the (pro)renin receptor and data is emerging that indicates that V-ATPase may be intimately linked to renin/angiotensin signaling both systemically and locally. In summary, V-ATPases play vital housekeeping roles in eukaryotic cells. Specialized versions of the pump are required by specific organ systems and are involved in diseases.
Collapse
|
37
|
|
38
|
Newly characterized Golgi-localized family of proteins is involved in calcium and pH homeostasis in yeast and human cells. Proc Natl Acad Sci U S A 2013; 110:6859-64. [PMID: 23569283 DOI: 10.1073/pnas.1219871110] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Defects in the human protein TMEM165 are known to cause a subtype of Congenital Disorders of Glycosylation. Transmembrane protein 165 (TMEM165) belongs to an uncharacterized family of membrane proteins called Uncharacterized Protein Family 0016, which are well conserved throughout evolution and share characteristics reminiscent of the cation/Ca(2+) exchanger superfamily. Gcr1 dependent translation factor 1 (Gdt1p), the budding yeast member of this family, contributes to Ca(2+) homeostasis via an uncharacterized Ca(2+) transport pathway localized in the Golgi apparatus. The gdt1Δ mutant was found to be sensitive to high concentrations of Ca(2+), and interestingly, this sensitivity was suppressed by expression of TMEM165, the human ortholog of Gdt1p, indicating conservation of function among the members of this family. Patch-clamp analyses on human cells indicated that TMEM165 expression is linked to Ca(2+) ion transport. Furthermore, defects in TMEM165 affected both Ca(2+) and pH homeostasis. Based on these results, we propose that Gdt1p and TMEM165 could be members of a unique family of Golgi-localized Ca(2+)/H(+) antiporters and that modification of the Golgi Ca(2+) and pH balance could explain the glycosylation defects observed in TMEM165-deficient patients.
Collapse
|
39
|
Zeevaert R, de Zegher F, Sturiale L, Garozzo D, Smet M, Moens M, Matthijs G, Jaeken J. Bone Dysplasia as a Key Feature in Three Patients with a Novel Congenital Disorder of Glycosylation (CDG) Type II Due to a Deep Intronic Splice Mutation in TMEM165. JIMD Rep 2012; 8:145-52. [PMID: 23430531 DOI: 10.1007/8904_2012_172] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Revised: 07/21/2012] [Accepted: 07/25/2012] [Indexed: 01/05/2023] Open
Abstract
Three patients belonging to two families presented with a psychomotor-dysmorphism syndrome including postnatal growth deficiency and major spondylo-, epi-, and metaphyseal skeletal involvement. Other features were muscular hypotrophy, fat excess, partial growth hormone deficiency, and, in two of the three patients, episodes of unexplained fever. Additional investigations showed mild to moderate increases of serum transaminases (particularly of aspartate transaminase (AST)), creatine kinase (CK), and lactate dehydrogenase (LDH), as well as decreased coagulation factors VIII, IX, XI, and protein C. Diagnostic work-up revealed a type 2 serum transferrin isoelectrofocusing (IEF) pattern and a cathodal shift on apolipoprotein C-III IEF pointing to a combined N- and O-glycosylation defect. Known glycosylation disorders with similar N-glycan structures lacking galactose and sialic acid were excluded. Through a combination of homozygosity mapping and expression profiling, a deep intronic homozygous mutation (c.792 + 182G>A) was found in TMEM165 (TPARL) in the three patients. TMEM165 is a gene of unknown function, possibly involved in Golgi proton/calcium transport. Here we present a detailed clinical description of the three patients with this mutation. The TMEM165 deficiency represents a novel type of CDG (TMEM165-CDG). This disorder enlarges the group of CDG caused by deficiencies in proteins that are not specifically involved in glycosylation but that have functions in the organization and homeostasis of the intracellular compartments and the secretory pathway, like COG-CDG and ATP6V0A2-CDG.
Collapse
Affiliation(s)
- R Zeevaert
- Center for Metabolic Disease, Department of Pediatrics, Katholieke Universiteit Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Fischer B, Dimopoulou A, Egerer J, Gardeitchik T, Kidd A, Jost D, Kayserili H, Alanay Y, Tantcheva-Poor I, Mangold E, Daumer-Haas C, Phadke S, Peirano RI, Heusel J, Desphande C, Gupta N, Nanda A, Felix E, Berry-Kravis E, Kabra M, Wevers RA, van Maldergem L, Mundlos S, Morava E, Kornak U. Further characterization of ATP6V0A2-related autosomal recessive cutis laxa. Hum Genet 2012; 131:1761-73. [PMID: 22773132 DOI: 10.1007/s00439-012-1197-8] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Accepted: 06/21/2012] [Indexed: 12/17/2022]
Abstract
Autosomal recessive cutis laxa (ARCL) syndromes are phenotypically overlapping, but genetically heterogeneous disorders. Mutations in the ATP6V0A2 gene were found to underlie both, autosomal recessive cutis laxa type 2 (ARCL2), Debré type, and wrinkly skin syndrome (WSS). The ATP6V0A2 gene encodes the a2 subunit of the V-type H(+)-ATPase, playing a role in proton translocation, and possibly also in membrane fusion. Here, we describe a highly variable phenotype in 13 patients with ARCL2, including the oldest affected individual described so far, who showed strikingly progressive dysmorphic features and heterotopic calcifications. In these individuals we identified 17 ATP6V0A2 mutations, 14 of which are novel. Furthermore, we demonstrate a localization of ATP6V0A2 at the Golgi-apparatus and a loss of the mutated ATP6V0A2 protein in patients' dermal fibroblasts. Investigation of brefeldin A-induced Golgi collapse in dermal fibroblasts as well as in HeLa cells deficient for ATP6V0A2 revealed a delay, which was absent in cells deficient for the ARCL-associated proteins GORAB or PYCR1. Furthermore, fibroblasts from patients with ATP6V0A2 mutations displayed elevated TGF-β signalling and increased TGF-β1 levels in the supernatant. Our current findings expand the genetic and phenotypic spectrum and suggest that, besides the known glycosylation defect, alterations in trafficking and signalling processes are potential key events in the pathogenesis of ATP6V0A2-related ARCL.
Collapse
Affiliation(s)
- Björn Fischer
- Institut fuer Medizinische Genetik und Humangenetik, Charité-Universitaetsmedizin Berlin, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
COG5-CDG with a Mild Neurohepatic Presentation. JIMD Rep 2011; 3:67-70. [PMID: 23430875 DOI: 10.1007/8904_2011_61] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Revised: 02/23/2011] [Accepted: 04/04/2011] [Indexed: 12/03/2022] Open
Abstract
The conserved oligomeric Golgi (COG) complex is an eight subunit protein involved in the retrograde transport of Golgi components. It affects the localization of several Golgi glycosyltransferases and hence is involved in N- and O-glycosylation. Genetic defects in this complex belong to the rapidly expanding family of congenital disorders of glycosylation (CDG). Patients have been reported with defects of subunit 1 (CDG1-CDG), subunit 4 (CDG4-CDG), subunit 5 (CDG5-CDG), subunit 6 (CDG6-CDG), subunit 7 (CDG7-CDG), and subunit 8 (CDG8-CDG). This paper is on the second reported patient with COG5-CDG. She showed a mild neurohepatic disease with central as well as peripheral neurological involvement while in the first reported patient (with a different mutation) only mild central neurological involvement was reported.
Collapse
|
42
|
Freeze HH, Ng BG. Golgi glycosylation and human inherited diseases. Cold Spring Harb Perspect Biol 2011; 3:a005371. [PMID: 21709180 DOI: 10.1101/cshperspect.a005371] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The Golgi factory receives custom glycosylates and dispatches its cargo to the correct cellular locations. The process requires importing donor substrates, moving the cargo, and recycling machinery. Correctly glycosylated cargo reflects the Golgi's quality and efficiency. Genetic disorders in the specific equipment (enzymes), donors (nucleotide sugar transporters), or equipment recycling/reorganization components (COG, SEC, golgins) can all affect glycosylation. Dozens of human glycosylation disorders fit these categories. Many other genes, with or without familiar names, well-annotated pedigrees, or likely homologies will join the ranks of glycosylation disorders. Their broad and unpredictable case-by-case phenotypes cross the traditional medical specialty boundaries. The gene functions in patients may be elusive, but their common feature may include altered glycosylation that provide clues to Golgi function. This article focuses on a group of human disorders that affect protein or lipid glycosylation. Readers may find it useful to generalize some of these patient-based, translational observations to their own research.
Collapse
Affiliation(s)
- Hudson H Freeze
- Genetic Disease Program, Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, California 92037, USA.
| | | |
Collapse
|
43
|
Jaeken J. Congenital disorders of glycosylation (CDG): it's (nearly) all in it! J Inherit Metab Dis 2011; 34:853-8. [PMID: 21384229 DOI: 10.1007/s10545-011-9299-3] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2010] [Revised: 02/08/2011] [Accepted: 02/10/2011] [Indexed: 12/28/2022]
Abstract
Congenital disorders of glycosylation (CDG) is a booming class of metabolic diseases. Its number has increased nearly fourfold (to 45) since 2003, the year of the Komrower lecture, entitled 'Congenital disorders of glycosylation CDG): It's all in it!'. This paper presents an overview of recently discovered CDG and CDG phenotypes, of a diagnostic approach, of (the lack of) treatment, of CDG genetics, of a novel CDG nomenclature and classification, and of some future directions in the CDG field.
Collapse
Affiliation(s)
- Jaak Jaeken
- Universitair Ziekenhuis Gasthuisberg, Herestraat 49, 3000, Leuven, Belgium.
| |
Collapse
|
44
|
Mohamed M, Kouwenberg D, Gardeitchik T, Kornak U, Wevers RA, Morava E. Metabolic cutis laxa syndromes. J Inherit Metab Dis 2011; 34:907-16. [PMID: 21431621 PMCID: PMC3137780 DOI: 10.1007/s10545-011-9305-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Revised: 02/11/2011] [Accepted: 02/17/2011] [Indexed: 01/09/2023]
Abstract
Cutis laxa is a rare skin disorder characterized by wrinkled, redundant, inelastic and sagging skin due to defective synthesis of elastic fibers and other proteins of the extracellular matrix. Wrinkled, inelastic skin occurs in many cases as an acquired condition. Syndromic forms of cutis laxa, however, are caused by diverse genetic defects, mostly coding for structural extracellular matrix proteins. Surprisingly a number of metabolic disorders have been also found to be associated with inherited cutis laxa. Menkes disease was the first metabolic disease reported with old-looking, wrinkled skin. Cutis laxa has recently been found in patients with abnormal glycosylation. The discovery of the COG7 defect in patients with wrinkled, inelastic skin was the first genetic link with the Congenital Disorders of Glycosylation (CDG). Since then several inborn errors of metabolism with cutis laxa have been described with variable severity. These include P5CS, ATP6V0A2-CDG and PYCR1 defects. In spite of the evolving number of cutis laxa-related diseases a large part of the cases remain genetically unsolved. In metabolic cutis laxa syndromes the clinical and laboratory features might partially overlap, however there are some distinct, discriminative features. In this review on metabolic diseases causing cutis laxa we offer a practical approach for the differential diagnosis of metabolic cutis laxa syndromes.
Collapse
Affiliation(s)
- Miski Mohamed
- Institute for Genetic and Metabolic Disease, Radboud University Medical Centre Nijmegen, P.O Box 9101, 6500 HB Nijmegen, The Netherlands
- Department of Pediatrics, Radboud University Medical Centre Nijmegen, P.O Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Dorus Kouwenberg
- Institute for Genetic and Metabolic Disease, Radboud University Medical Centre Nijmegen, P.O Box 9101, 6500 HB Nijmegen, The Netherlands
- Department of Pediatrics, Radboud University Medical Centre Nijmegen, P.O Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Thatjana Gardeitchik
- Institute for Genetic and Metabolic Disease, Radboud University Medical Centre Nijmegen, P.O Box 9101, 6500 HB Nijmegen, The Netherlands
- Department of Pediatrics, Radboud University Medical Centre Nijmegen, P.O Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Uwe Kornak
- Max Planck Institute for Molecular genetics, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ron A. Wevers
- Laboratory of Genetic, Endocrine and Metabolic Diseases, Radboud University Medical Centre Nijmegen, Nijmegen, The Netherlands
| | - Eva Morava
- Institute for Genetic and Metabolic Disease, Radboud University Medical Centre Nijmegen, P.O Box 9101, 6500 HB Nijmegen, The Netherlands
- Department of Pediatrics, Radboud University Medical Centre Nijmegen, P.O Box 9101, 6500 HB Nijmegen, The Netherlands
| |
Collapse
|
45
|
Mohamed M, Guillard M, Wortmann S, Cirak S, Marklova E, Michelakakis H, Korsch E, Adamowicz M, Koletzko B, van Spronsen F, Niezen-Koning K, Matthijs G, Gardeitchik T, Kouwenberg D, Lim BC, Zeevaert R, Wevers R, Lefeber D, Morava E. Clinical and diagnostic approach in unsolved CDG patients with a type 2 transferrin pattern. Biochim Biophys Acta Mol Basis Dis 2011; 1812:691-8. [DOI: 10.1016/j.bbadis.2011.02.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Revised: 02/18/2011] [Accepted: 02/22/2011] [Indexed: 11/29/2022]
|
46
|
Guillard M, Morava E, van Delft FL, Hague R, Körner C, Adamowicz M, Wevers RA, Lefeber DJ. Plasma N-glycan profiling by mass spectrometry for congenital disorders of glycosylation type II. Clin Chem 2011; 57:593-602. [PMID: 21273509 DOI: 10.1373/clinchem.2010.153635] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Determination of the genetic defect in patients with a congenital disorder of glycosylation (CDG) is challenging because of the wide clinical presentation, the large number of gene products involved, and the occurrence of secondary causes of underglycosylation. Transferrin isoelectric focusing has been the method of choice for CDG screening; however, improved methods are required for the molecular diagnosis of patients with CDG type II. METHODS Plasma samples with a typical transferrin isofocusing profile were analyzed. N-glycans were released from these samples by PNGase F [peptide-N4-(acetyl-β-glucosaminyl)-asparagine amidase] digestion, permethylated and purified, and measured on a MALDI linear ion trap mass spectrometer. A set of 38 glycans was used for quantitative comparison and to establish reference intervals for such glycan features as the number of antennae, the level of truncation, and fucosylation. Plasma N-glycans from control individuals, patients with known CDG type II defects, and patients with a secondary cause of underglycosylation were analyzed. RESULTS CDGs due to mannosyl (α-1,6-)-glycoprotein β-1,2-N-acetylglucosaminyltransferase (MGAT2), β-1,4-galactosyltransferase 1 (B4GALT1), and SLC35C1 (a GDP-fucose transporter) defects could be diagnosed directly from the N-glycan profile. CDGs due to defects in proteins involved in Golgi trafficking, such as subunit 7 of the conserved oligomeric Golgi complex (COG7) and subunit V0 a2 of the lysosomal H(+)-transporting ATPase (ATP6V0A2) caused a loss of triantennary N-glycans and an increase of truncated structures. Secondary causes with liver involvement were characterized by increased fucosylation, whereas the presence of plasma sialidase produced isolated undersialylation. CONCLUSIONS MALDI ion trap analysis of plasma N-glycans documents features that discriminate between primary and secondary causes of underglycosylation and should be applied as the first step in the diagnostic track of all patients with an unsolved CDG type II.
Collapse
Affiliation(s)
- Maïlys Guillard
- Department of Laboratory Medicine, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
47
|
|
48
|
Morava E, Guillard M, Lefeber DJ, Wevers RA. Autosomal recessive cutis laxa syndrome revisited. Eur J Hum Genet 2009; 17:1099-110. [PMID: 19401719 DOI: 10.1038/ejhg.2009.22] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The clinical spectrum of the autosomal recessive cutis laxa syndromes is highly heterogeneous with respect to organ involvement and severity. One of the major diagnostic criteria is to detect abnormal elastin fibers. In several other clinically similar autosomal recessive syndromes, however, the classic histological anomalies are absent, and the definite diagnosis remains uncertain. In cutis laxa patients mutations have been demonstrated in elastin or fibulin genes, but in the majority of patients the underlying genetic etiology remains unknown. Recently, we found mutations in the ATP6V0A2 gene in families with autosomal recessive cutis laxa. This genetic defect is associated with abnormal glycosylation leading to a distinct combined disorder of the biosynthesis of N- and O-linked glycans. Interestingly, similar mutations have been found in patients with wrinkly skin syndrome, without the presence of severe skin symptoms of elastin deficiency. These findings suggest that the cutis laxa and wrinkly skin syndromes are phenotypic variants of the same disorder. Interestingly many phenotypically similar patients carry no mutations in the ATP6V0A2 gene. The variable presence of protein glycosylation abnormalities in the diverse clinical forms of the wrinkled skin-cutis laxa syndrome spectrum necessitates revisiting the diagnostic criteria to be able to offer adequate prognosis assessment and counseling. This paper aims at describing the spectrum of clinical features of the various forms of autosomal recessive cutis laxa syndromes. Based on the recently unraveled novel genetic entity we also review the genetic aspects in cutis laxa syndromes including genotype-phenotype correlations and suggest a practical diagnostic approach.
Collapse
Affiliation(s)
- Eva Morava
- Department of Paediatrics, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
| | | | | | | |
Collapse
|