1
|
Lernmark Å, Akolkar B, Hagopian W, Krischer J, McIndoe R, Rewers M, Toppari J, Vehik K, Ziegler AG. Possible heterogeneity of initial pancreatic islet beta-cell autoimmunity heralding type 1 diabetes. J Intern Med 2023; 294:145-158. [PMID: 37143363 PMCID: PMC10524683 DOI: 10.1111/joim.13648] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
The etiology of type 1 diabetes (T1D) foreshadows the pancreatic islet beta-cell autoimmune pathogenesis that heralds the clinical onset of T1D. Standardized and harmonized tests of autoantibodies against insulin (IAA), glutamic acid decarboxylase (GADA), islet antigen-2 (IA-2A), and ZnT8 transporter (ZnT8A) allowed children to be followed from birth until the appearance of a first islet autoantibody. In the Environmental Determinants of Diabetes in the Young (TEDDY) study, a multicenter (Finland, Germany, Sweden, and the United States) observational study, children were identified at birth for the T1D high-risk HLA haploid genotypes DQ2/DQ8, DQ2/DQ2, DQ8/DQ8, and DQ4/DQ8. The TEDDY study was preceded by smaller studies in Finland, Germany, Colorado, Washington, and Sweden. The aims were to follow children at increased genetic risk to identify environmental factors that trigger the first-appearing autoantibody (etiology) and progress to T1D (pathogenesis). The larger TEDDY study found that the incidence rate of the first-appearing autoantibody was split into two patterns. IAA first peaked already during the first year of life and tapered off by 3-4 years of age. GADA first appeared by 2-3 years of age to reach a plateau by about 4 years. Prior to the first-appearing autoantibody, genetic variants were either common or unique to either pattern. A split was also observed in whole blood transcriptomics, metabolomics, dietary factors, and exposures such as gestational life events and early infections associated with prolonged shedding of virus. An innate immune reaction prior to the adaptive response cannot be excluded. Clarifying the mechanisms by which autoimmunity is triggered to either insulin or GAD65 is key to uncovering the etiology of autoimmune T1D.
Collapse
Affiliation(s)
- Åke Lernmark
- Department of Clinical Sciences, Lund University CRC, Skåne University Hospital, Malmö, Sweden
| | - Beena Akolkar
- National Institute of Diabetes & Digestive & Kidney Diseases, Bethesda, MD USA
| | | | - Jeffrey Krischer
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL USA
| | - Richard McIndoe
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA USA
| | - Marian Rewers
- Barbara Davis Center for Diabetes, University of Colorado, Aurora, Colorado USA
| | - Jorma Toppari
- Department of Pediatrics, Turku University Hospital, and Institute of Biomedicine, Research Centre for Integrated Physiology and Pharmacology, University of Turku, Turku, Finland
| | - Kendra Vehik
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL USA
| | - Anette-G. Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, and Klinikum rechts der Isar, Technische Universität München, and Forschergruppe Diabetes e.V., Neuherberg, Germany
| | | |
Collapse
|
2
|
Pandey R, Bakay M, Hakonarson H. CLEC16A-An Emerging Master Regulator of Autoimmunity and Neurodegeneration. Int J Mol Sci 2023; 24:ijms24098224. [PMID: 37175930 PMCID: PMC10179542 DOI: 10.3390/ijms24098224] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
CLEC16A is emerging as an important genetic risk factor for several autoimmune disorders and for Parkinson disease (PD), opening new avenues for translational research and therapeutic development. While the exact role of CLEC16A in health and disease is still being elucidated, the gene plays a critical role in the regulation of autophagy, mitophagy, endocytosis, intracellular trafficking, immune function, and in biological processes such as insulin secretion and others that are important to cellular homeostasis. As shown in both human and animal modeling studies, CLEC16A hypofunction predisposes to both autoinflammatory phenotype and neurodegeneration. While the two are clearly related, further functional studies are needed to fully understand the mechanisms involved for optimized therapeutic interventions. Based on recent data, mitophagy-inducing drugs may be warranted, and such therapy should be tested in clinical trials as these drugs would tackle the underlying pathogenic mechanism (s) and could treat or prevent symptoms of autoimmunity and neurodegeneration in individuals with CLEC16A risk variants. Accordingly, interventions directed at reversing the dysregulated mitophagy and the consequences of loss of function of CLEC16A without activating other detrimental cellular pathways could present an effective therapy. This review presents the emerging role of CLEC16A in health and disease and provides an update on the disease processes that are attributed to variants located in the CLEC16A gene, which are responsible for autoimmune disorders and neurodegeneration with emphasis on how this information is being translated into practical and effective applications in the clinic.
Collapse
Affiliation(s)
- Rahul Pandey
- Center for Applied Genomics, Children's Hospital of Philadelphia, Abramson Research Center, 3615 Civic Center Boulevard, Philadelphia, PA 19104-4318, USA
| | - Marina Bakay
- Center for Applied Genomics, Children's Hospital of Philadelphia, Abramson Research Center, 3615 Civic Center Boulevard, Philadelphia, PA 19104-4318, USA
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Abramson Research Center, 3615 Civic Center Boulevard, Philadelphia, PA 19104-4318, USA
- Department of Pediatrics, The University of Pennsylvania School of Medicine, Philadelphia, PA 19104-4318, USA
| |
Collapse
|
3
|
Eriksson AM, Leikfoss IS, Abrahamsen G, Sundvold V, Isom MM, Keshari PK, Rognes T, Landsverk OJB, Bos SD, Harbo HF, Spurkland A, Berge T. Exploring the role of the multiple sclerosis susceptibility gene CLEC16A in T cells. Scand J Immunol 2021; 94:e13050. [PMID: 34643957 DOI: 10.1111/sji.13050] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 04/20/2021] [Accepted: 04/28/2021] [Indexed: 12/29/2022]
Abstract
C-type lectin-like domain family 16 member A (CLEC16A) is associated with autoimmune disorders, including multiple sclerosis (MS), but its functional relevance is not completely understood. CLEC16A is expressed in several immune cells, where it affects autophagic processes and receptor expression. Recently, we reported that the risk genotype of an MS-associated single nucleotide polymorphism in CLEC16A intron 19 is associated with higher expression of CLEC16A in CD4+ T cells. Here, we show that CLEC16A expression is induced in CD4+ T cells upon T cell activation. By the use of imaging flow cytometry and confocal microscopy, we demonstrate that CLEC16A is located in Rab4a-positive recycling endosomes in Jurkat TAg T cells. CLEC16A knock-down in Jurkat cells resulted in lower cell surface expression of the T cell receptor, however, this did not have a major impact on T cell activation response in vitro in Jurkat nor in human, primary CD4+ T cells.
Collapse
Affiliation(s)
- Anna M Eriksson
- Department of Neurology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ingvild Sørum Leikfoss
- Department of Neurology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Neuroscience Research Unit, Department of Research, Innovation and Education, Oslo University Hospital, Oslo, Norway
| | - Greger Abrahamsen
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Vibeke Sundvold
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | | | - Pankaj K Keshari
- Department of Neurology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Torbjørn Rognes
- Department of Informatics, University of Oslo, Oslo, Norway.,Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | | | - Steffan D Bos
- Department of Neurology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Hanne F Harbo
- Department of Neurology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Anne Spurkland
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Tone Berge
- Neuroscience Research Unit, Department of Research, Innovation and Education, Oslo University Hospital, Oslo, Norway.,Department of Mechanical, Electronic and Chemical Engineering, Oslo Metropolitan University, Oslo, Norway
| |
Collapse
|
4
|
Albalawi AM, Al-Barry MA. Genetic variations in autoimmune genes and VKH disease. Int Ophthalmol 2020; 40:3175-3186. [PMID: 32974831 DOI: 10.1007/s10792-020-01407-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 04/28/2020] [Indexed: 02/02/2023]
Abstract
PURPOSE Vogt-Koyanagi-Harada (VKH) disease is a rare autoimmune disease. The autoimmune response in VKH disease is against the melanin-producing cells; therefore, in affected individuals melanocyte-containing organs manifest disease symptoms including eyes, ears, skin and nervous system. VKH is a multifactorial disease, and the precise cause of the VKH disease is unknown. Studies have suggested that both environmental and genetic factors are responsible for the VKH disease. In this review, the authors have collected all the available literature on the genetics of VKH to their knowledge and discussed the role of genetic variants in causing VKH disease. METHODS An extensive literature search was performed in order to review all the published studies regarding VKH clinical phenotyping and genetic variants in VKH disease. Medline, PubMed, Cochrane library, and Scopus was searched using combination of keywords. RESULTS It was found that variants in HLA genes, IL-12b, TNFSF4, and miR-20-5p genes are significantly associated with VKH; however, variants in genes ATG10, TNIP1 and CLEC16A did not achieve significant genome-wide association threshold. Moreover, polymorphisms in TNIP1 and CLEC16A play a protective role against VKH. CONCLUSION The authors conclude that increased sample size and a more homogeneous VKH patient population may reveal a significant association of variants in ATG10, TNIP1 and CLEC16A genes with VKH disease.
Collapse
Affiliation(s)
- Alia M Albalawi
- Department of Biology, College of Science, King AbdulAziz University Jeddah, Jeddah, Saudi Arabia.,Center for Genetics and Inherited Diseases, Taibah University Almadinah Almunawwarah, Medina, Saudi Arabia
| | - Maan A Al-Barry
- Department of Ophthalmology, College of Medicine, Taibah University Almadinah Almunawwarah, Medina, Saudi Arabia.
| |
Collapse
|
5
|
Rijvers L, Melief MJ, van Langelaar J, van der Vuurst de Vries RM, Wierenga-Wolf AF, Koetzier SC, Priatel JJ, Jorritsma T, van Ham SM, Hintzen RQ, van Luijn MM. The Role of Autoimmunity-Related Gene CLEC16A in the B Cell Receptor-Mediated HLA Class II Pathway. THE JOURNAL OF IMMUNOLOGY 2020; 205:945-956. [PMID: 32641384 DOI: 10.4049/jimmunol.1901409] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 06/09/2020] [Indexed: 12/14/2022]
Abstract
C-type lectin CLEC16A is located next to CIITA, the master transcription factor of HLA class II (HLA-II), at a susceptibility locus for several autoimmune diseases, including multiple sclerosis (MS). We previously found that CLEC16A promotes the biogenesis of HLA-II peptide-loading compartments (MIICs) in myeloid cells. Given the emerging role of B cells as APCs in these diseases, in this study, we addressed whether and how CLEC16A is involved in the BCR-dependent HLA-II pathway. CLEC16A was coexpressed with surface class II-associated invariant chain peptides (CLIP) in human EBV-positive and not EBV-negative B cell lines. Stable knockdown of CLEC16A in EBV-positive Raji B cells resulted in an upregulation of surface HLA-DR and CD74 (invariant chain), whereas CLIP was slightly but significantly reduced. In addition, IgM-mediated Salmonella uptake was decreased, and MIICs were less clustered in CLEC16A-silenced Raji cells, implying that CLEC16A controls both HLA-DR/CD74 and BCR/Ag processing in MIICs. In primary B cells, CLEC16A was only induced under CLIP-stimulating conditions in vitro and was predominantly expressed in CLIPhigh naive populations. Finally, CLIP-loaded HLA-DR molecules were abnormally enriched, and coregulation with CLEC16A was abolished in blood B cells of patients who rapidly develop MS. These findings demonstrate that CLEC16A participates in the BCR-dependent HLA-II pathway in human B cells and that this regulation is impaired during MS disease onset. The abundance of CLIP already on naive B cells of MS patients may point to a chronically induced stage and a new mechanism underlying B cell-mediated autoimmune diseases such as MS.
Collapse
Affiliation(s)
- Liza Rijvers
- Department of Immunology, Erasmus MC, 3015 CN Rotterdam, the Netherlands.,MS Center ErasMS, Erasmus MC, 3015 CN Rotterdam, the Netherlands
| | - Marie-José Melief
- Department of Immunology, Erasmus MC, 3015 CN Rotterdam, the Netherlands.,MS Center ErasMS, Erasmus MC, 3015 CN Rotterdam, the Netherlands
| | - Jamie van Langelaar
- Department of Immunology, Erasmus MC, 3015 CN Rotterdam, the Netherlands.,MS Center ErasMS, Erasmus MC, 3015 CN Rotterdam, the Netherlands
| | - Roos M van der Vuurst de Vries
- MS Center ErasMS, Erasmus MC, 3015 CN Rotterdam, the Netherlands.,Department of Neurology, Erasmus MC, 3015 CN Rotterdam, the Netherlands
| | - Annet F Wierenga-Wolf
- Department of Immunology, Erasmus MC, 3015 CN Rotterdam, the Netherlands.,MS Center ErasMS, Erasmus MC, 3015 CN Rotterdam, the Netherlands
| | - Steven C Koetzier
- Department of Immunology, Erasmus MC, 3015 CN Rotterdam, the Netherlands.,MS Center ErasMS, Erasmus MC, 3015 CN Rotterdam, the Netherlands
| | - John J Priatel
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada.,BC Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada; and
| | - Tineke Jorritsma
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center, University of Amsterdam, 1066 CX Amsterdam, the Netherlands
| | - S Marieke van Ham
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center, University of Amsterdam, 1066 CX Amsterdam, the Netherlands
| | - Rogier Q Hintzen
- Department of Immunology, Erasmus MC, 3015 CN Rotterdam, the Netherlands.,MS Center ErasMS, Erasmus MC, 3015 CN Rotterdam, the Netherlands.,Department of Neurology, Erasmus MC, 3015 CN Rotterdam, the Netherlands
| | - Marvin M van Luijn
- Department of Immunology, Erasmus MC, 3015 CN Rotterdam, the Netherlands; .,MS Center ErasMS, Erasmus MC, 3015 CN Rotterdam, the Netherlands
| |
Collapse
|
6
|
Pandey R, Bakay M, Hain HS, Strenkowski B, Yermakova A, Kushner JA, Orange JS, Hakonarson H. The Autoimmune Disorder Susceptibility Gene CLEC16A Restrains NK Cell Function in YTS NK Cell Line and Clec16a Knockout Mice. Front Immunol 2019; 10:68. [PMID: 30774629 PMCID: PMC6367972 DOI: 10.3389/fimmu.2019.00068] [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: 03/23/2018] [Accepted: 01/11/2019] [Indexed: 12/21/2022] Open
Abstract
CLEC16A locus polymorphisms have been associated with several autoimmune diseases. We overexpressed CLEC16A in YTS natural killer (NK) cells and observed reduced NK cell cytotoxicity and IFN-γ release, delayed dendritic cell (DC) maturation, decreased conjugate formation, cell-surface receptor downregulation and increased autophagy. In contrast, siRNA mediated knockdown resulted in increased NK cell cytotoxicity, reversal of receptor expression and disrupted mitophagy. Subcellular localization studies demonstrated that CLEC16A is a cytosolic protein that associates with Vps16A, a subunit of class C Vps-HOPS complex, and modulates receptor expression via autophagy. Clec16a knockout (KO) in mice resulted in altered immune cell populations, increased splenic NK cell cytotoxicity, imbalance of dendritic cell subsets, altered receptor expression, upregulated cytokine and chemokine secretion. Taken together, our findings indicate that CLEC16A restrains secretory functions including cytokine release and cytotoxicity and that a delicate balance of CLEC16A is needed for NK cell function and homeostasis.
Collapse
Affiliation(s)
- Rahul Pandey
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Marina Bakay
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Heather S Hain
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Bryan Strenkowski
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Anastasiya Yermakova
- Section of Immunology, Allergy, and Rheumatology, Department of Pediatric Medicine, Texas Children's Hospital, Houston, TX, United States
| | - Jake A Kushner
- Section of Pediatric Diabetes and Endocrinology, Department of Pediatric Medicine, Endocrine-Metabolism, Texas Children's Hospital, Houston, TX, United States
| | - Jordan S Orange
- Section of Immunology, Allergy, and Rheumatology, Department of Pediatric Medicine, Texas Children's Hospital, Houston, TX, United States
| | - Hakon Hakonarson
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, United States.,Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
7
|
Tam RCY, Li MWM, Gao YP, Pang YT, Yan S, Ge W, Lau CS, Chan VSF. Human CLEC16A regulates autophagy through modulating mTOR activity. Exp Cell Res 2017; 352:304-312. [PMID: 28223137 DOI: 10.1016/j.yexcr.2017.02.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 01/17/2017] [Accepted: 02/11/2017] [Indexed: 12/28/2022]
Abstract
CLEC16A is genetically linked with multiple autoimmune disorders but its functional relevance in autoimmunity remains obscure. Recent evidence has signposted the emerging role of autophagy in autoimmune disease development. Here, by ectopic expression and siRNA silencing, we show that CLEC16A has an inhibitory role in starvation-induced autophagy in human cells. Combining quantitative proteomics and immunoblotting analyses, we found that CLEC16A likely regulates autophagy by activating mTOR pathway. Overexpression of CLEC16A was found to sensitize cells towards the availability of nutrients, resulting in a heightened mTOR activity, which in turn diminished LC3 autophagic activity following nutrient deprivation. CLEC16A deficiency, on the other hand, delayed mTOR activity in response to nutrient sensing, thereby resulted in an augmented autophagic response. CLEC16A was found residing in cytosolic vesicles and the Golgi, and nutrient removal promoted a stronger clustering within the Golgi, where it was possibly in a vantage position to activate mTOR upon nutrient replenishment. These findings suggest that Golgi-associated CLEC16A negatively regulates autophagy via modulation of mTOR activity, and may provide support for a functional link between CLEC16A and autoimmunity.
Collapse
Affiliation(s)
- Rachel Chun Yee Tam
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR
| | - Michelle Wing Man Li
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR
| | - Yan Pan Gao
- National Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Yuen Ting Pang
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR
| | - Sheng Yan
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR
| | - Wei Ge
- National Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Chak Sing Lau
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR
| | - Vera Sau Fong Chan
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR..
| |
Collapse
|
8
|
Abstract
Type 1 diabetes is diagnosed at the end of a prodrome of β-cell autoimmunity. The disease is most likely triggered at an early age by autoantibodies primarily directed against insulin or glutamic acid decarboxylase, or both, but rarely against islet antigen-2. After the initial appearance of one of these autoantibody biomarkers, a second, third, or fourth autoantibody against either islet antigen-2 or the ZnT8 transporter might also appear. The larger the number of β-cell autoantibody types, the greater the risk of rapid progression to clinical onset of diabetes. This association does not necessarily mean that the β-cell autoantibodies are pathogenic, but rather that they represent reproducible biomarkers of the pathogenesis. The primary risk factor for β-cell autoimmunity is genetic, mainly occurring in individuals with either HLA-DR3-DQ2 or HLA-DR4-DQ8 haplotypes, or both, but a trigger from the environment is generally needed. The pathogenesis can be divided into three stages: 1, appearance of β-cell autoimmunity, normoglycaemia, and no symptoms; 2, β-cell autoimmunity, dysglycaemia, and no symptoms; and 3, β-cell autoimmunity, dysglycaemia, and symptoms of diabetes. The genetic association with each one of the three stages can differ. Type 1 diabetes could serve as a disease model for organ-specific autoimmune disorders such as coeliac disease, thyroiditis, and Addison's disease, which show similar early markers of a prolonged disease process before clinical diagnosis.
Collapse
Affiliation(s)
- Flemming Pociot
- Department of Pediatrics, Herlev and Gentofte Hospital, DK-2730 Herlev, Denmark
| | - Åke Lernmark
- Department of Clinical Sciences, Lund University, Skåne University Hospital, SE-20502 Malmö, Sweden.
| |
Collapse
|
9
|
Redmann V, Lamb CA, Hwang S, Orchard RC, Kim S, Razi M, Milam A, Park S, Yokoyama CC, Kambal A, Kreamalmeyer D, Bosch MK, Xiao M, Green K, Kim J, Pruett-Miller SM, Ornitz DM, Allen PM, Beatty WL, Schmidt RE, DiAntonio A, Tooze SA, Virgin HW. Clec16a is Critical for Autolysosome Function and Purkinje Cell Survival. Sci Rep 2016; 6:23326. [PMID: 26987296 PMCID: PMC4796910 DOI: 10.1038/srep23326] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 02/22/2016] [Indexed: 11/29/2022] Open
Abstract
CLEC16A is in a locus genetically linked to autoimmune diseases including multiple sclerosis, but the function of this gene in the nervous system is unknown. Here we show that two mouse strains carrying independent Clec16a mutations developed neurodegenerative disease characterized by motor impairments and loss of Purkinje cells. Neurons from Clec16a-mutant mice exhibited increased expression of the autophagy substrate p62, accumulation of abnormal intra-axonal membranous structures bearing the autophagy protein LC3, and abnormal Golgi morphology. Multiple aspects of endocytosis, lysosome and Golgi function were normal in Clec16a-deficient murine embryonic fibroblasts and HeLa cells. However, these cells displayed abnormal bulk autophagy despite unimpaired autophagosome formation. Cultured Clec16a-deficient cells exhibited a striking accumulation of LC3 and LAMP-1 positive autolysosomes containing undigested cytoplasmic contents. Therefore Clec16a, an autophagy protein that is critical for autolysosome function and clearance, is required for Purkinje cell survival.
Collapse
Affiliation(s)
- Veronika Redmann
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Christopher A. Lamb
- The Francis Crick Institute, Lincoln’s Inn Fields Laboratory, London, WC2A 3LY, UK
| | - Seungmin Hwang
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | - Robert C. Orchard
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sungsu Kim
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Minoo Razi
- The Francis Crick Institute, Lincoln’s Inn Fields Laboratory, London, WC2A 3LY, UK
| | - Ashley Milam
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sunmin Park
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Christine C. Yokoyama
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Amal Kambal
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Darren Kreamalmeyer
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Marie K. Bosch
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Maolei Xiao
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Karen Green
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jungsu Kim
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | - Shondra M. Pruett-Miller
- Genome Engineering and iPSC Center, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David M. Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Paul M. Allen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Wandy L. Beatty
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Robert E. Schmidt
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Aaron DiAntonio
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sharon A. Tooze
- The Francis Crick Institute, Lincoln’s Inn Fields Laboratory, London, WC2A 3LY, UK
| | - Herbert W. Virgin
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
10
|
Proust C, Empana JP, Boutouyrie P, Alivon M, Challande P, Danchin N, Escriou G, Esslinger U, Laurent S, Li Z, Pannier B, Regnault V, Thomas F, Jouven X, Cambien F, Lacolley P. Contribution of Rare and Common Genetic Variants to Plasma Lipid Levels and Carotid Stiffness and Geometry: A Substudy of the Paris Prospective Study 3. ACTA ACUST UNITED AC 2015; 8:628-36. [PMID: 26160806 DOI: 10.1161/circgenetics.114.000979] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 06/24/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND We assess the contribution of common and rare putatively functional genetic variants (most of them coding) present on the Illumina exome Beadchip to the variability of plasma lipids and stiffness of the common carotid artery. METHODS AND RESULTS Measurements were obtained from 2283 men and 1398 women, and after filtering and exclusion of monomorphic variants, 32,827 common (minor allele frequency >0.01) and 68,770 rare variants were analyzed. A large fraction of the heritability of plasma lipids is attributable to variants present on the array, especially for triglycerides (fraction of variance attributable to measured genotypes: V(G)/V(p)=31.4%, P<3.1×10(-11)) and high-density lipoprotein cholesterol (V(G)/V(p)=26.4%, P<4.2×10(-12)). Plasma lipids were associated with common variants located in known candidate genes, but no implication of rare variants could be established. Gene sets for plasma lipids, blood pressure, and coronary artery disease were defined on the basis of recent meta-analyses of genome-wide association studies. We observed a strong association between the plasma lipids gene set and plasma lipid variables, but none of the 3 genome-wide association studies gene sets was associated with the carotid parameters. Significant V(G)/V(p) ratios were observed for external (14.5%, P<2.7×10(-5)) and internal diameter (13.4%, P<4.3×10(-4)), stiffness (12.5%, P<8.0×10(-4)), intima-media thickness (10.6%, P<7.9×10(-4)), and wall cross-sectional area (13.2%, P<2.4×10(-5)). A significant association was observed between the common rs2903692 polymorphism of the CLEC16A gene and the internal diameter (P<4.3×10(-7)). CONCLUSIONS These results suggest an involvement of CLEC16A, a gene that has been reported to be associated with immune disorders, in the modulation of carotid vasodilatation.
Collapse
Affiliation(s)
- Carole Proust
- From the Inserm, UMR_S 1166 (C.P., G.E., U.E., F.C.), CNRS UMR 7190 (P.C.), and CNRS, UMR 8256 (Z.L.), Sorbonne Universités, UPMC Univ Paris 06; Inserm, UMR_S 970, Sorbonne Paris Cité (J.-P.E., P.B., M.A., G.E., S.L., X.J.); Department of Cardiology, European Hospital of Georges Pompidou, Université Paris Descartes (N.D.); Centre d'Investigations Préventives et Cliniques (IPC Center), Paris, France (B.P., F.T.); and Inserm, UMR_S 1116; Université de Lorraine, Nancy, France (V.R., P.L.)
| | - Jean-Philippe Empana
- From the Inserm, UMR_S 1166 (C.P., G.E., U.E., F.C.), CNRS UMR 7190 (P.C.), and CNRS, UMR 8256 (Z.L.), Sorbonne Universités, UPMC Univ Paris 06; Inserm, UMR_S 970, Sorbonne Paris Cité (J.-P.E., P.B., M.A., G.E., S.L., X.J.); Department of Cardiology, European Hospital of Georges Pompidou, Université Paris Descartes (N.D.); Centre d'Investigations Préventives et Cliniques (IPC Center), Paris, France (B.P., F.T.); and Inserm, UMR_S 1116; Université de Lorraine, Nancy, France (V.R., P.L.)
| | - Pierre Boutouyrie
- From the Inserm, UMR_S 1166 (C.P., G.E., U.E., F.C.), CNRS UMR 7190 (P.C.), and CNRS, UMR 8256 (Z.L.), Sorbonne Universités, UPMC Univ Paris 06; Inserm, UMR_S 970, Sorbonne Paris Cité (J.-P.E., P.B., M.A., G.E., S.L., X.J.); Department of Cardiology, European Hospital of Georges Pompidou, Université Paris Descartes (N.D.); Centre d'Investigations Préventives et Cliniques (IPC Center), Paris, France (B.P., F.T.); and Inserm, UMR_S 1116; Université de Lorraine, Nancy, France (V.R., P.L.)
| | - Maureen Alivon
- From the Inserm, UMR_S 1166 (C.P., G.E., U.E., F.C.), CNRS UMR 7190 (P.C.), and CNRS, UMR 8256 (Z.L.), Sorbonne Universités, UPMC Univ Paris 06; Inserm, UMR_S 970, Sorbonne Paris Cité (J.-P.E., P.B., M.A., G.E., S.L., X.J.); Department of Cardiology, European Hospital of Georges Pompidou, Université Paris Descartes (N.D.); Centre d'Investigations Préventives et Cliniques (IPC Center), Paris, France (B.P., F.T.); and Inserm, UMR_S 1116; Université de Lorraine, Nancy, France (V.R., P.L.)
| | - Pascal Challande
- From the Inserm, UMR_S 1166 (C.P., G.E., U.E., F.C.), CNRS UMR 7190 (P.C.), and CNRS, UMR 8256 (Z.L.), Sorbonne Universités, UPMC Univ Paris 06; Inserm, UMR_S 970, Sorbonne Paris Cité (J.-P.E., P.B., M.A., G.E., S.L., X.J.); Department of Cardiology, European Hospital of Georges Pompidou, Université Paris Descartes (N.D.); Centre d'Investigations Préventives et Cliniques (IPC Center), Paris, France (B.P., F.T.); and Inserm, UMR_S 1116; Université de Lorraine, Nancy, France (V.R., P.L.)
| | - Nicolas Danchin
- From the Inserm, UMR_S 1166 (C.P., G.E., U.E., F.C.), CNRS UMR 7190 (P.C.), and CNRS, UMR 8256 (Z.L.), Sorbonne Universités, UPMC Univ Paris 06; Inserm, UMR_S 970, Sorbonne Paris Cité (J.-P.E., P.B., M.A., G.E., S.L., X.J.); Department of Cardiology, European Hospital of Georges Pompidou, Université Paris Descartes (N.D.); Centre d'Investigations Préventives et Cliniques (IPC Center), Paris, France (B.P., F.T.); and Inserm, UMR_S 1116; Université de Lorraine, Nancy, France (V.R., P.L.)
| | - Guillaume Escriou
- From the Inserm, UMR_S 1166 (C.P., G.E., U.E., F.C.), CNRS UMR 7190 (P.C.), and CNRS, UMR 8256 (Z.L.), Sorbonne Universités, UPMC Univ Paris 06; Inserm, UMR_S 970, Sorbonne Paris Cité (J.-P.E., P.B., M.A., G.E., S.L., X.J.); Department of Cardiology, European Hospital of Georges Pompidou, Université Paris Descartes (N.D.); Centre d'Investigations Préventives et Cliniques (IPC Center), Paris, France (B.P., F.T.); and Inserm, UMR_S 1116; Université de Lorraine, Nancy, France (V.R., P.L.)
| | - Ulrike Esslinger
- From the Inserm, UMR_S 1166 (C.P., G.E., U.E., F.C.), CNRS UMR 7190 (P.C.), and CNRS, UMR 8256 (Z.L.), Sorbonne Universités, UPMC Univ Paris 06; Inserm, UMR_S 970, Sorbonne Paris Cité (J.-P.E., P.B., M.A., G.E., S.L., X.J.); Department of Cardiology, European Hospital of Georges Pompidou, Université Paris Descartes (N.D.); Centre d'Investigations Préventives et Cliniques (IPC Center), Paris, France (B.P., F.T.); and Inserm, UMR_S 1116; Université de Lorraine, Nancy, France (V.R., P.L.)
| | - Stéphane Laurent
- From the Inserm, UMR_S 1166 (C.P., G.E., U.E., F.C.), CNRS UMR 7190 (P.C.), and CNRS, UMR 8256 (Z.L.), Sorbonne Universités, UPMC Univ Paris 06; Inserm, UMR_S 970, Sorbonne Paris Cité (J.-P.E., P.B., M.A., G.E., S.L., X.J.); Department of Cardiology, European Hospital of Georges Pompidou, Université Paris Descartes (N.D.); Centre d'Investigations Préventives et Cliniques (IPC Center), Paris, France (B.P., F.T.); and Inserm, UMR_S 1116; Université de Lorraine, Nancy, France (V.R., P.L.)
| | - Zhenlin Li
- From the Inserm, UMR_S 1166 (C.P., G.E., U.E., F.C.), CNRS UMR 7190 (P.C.), and CNRS, UMR 8256 (Z.L.), Sorbonne Universités, UPMC Univ Paris 06; Inserm, UMR_S 970, Sorbonne Paris Cité (J.-P.E., P.B., M.A., G.E., S.L., X.J.); Department of Cardiology, European Hospital of Georges Pompidou, Université Paris Descartes (N.D.); Centre d'Investigations Préventives et Cliniques (IPC Center), Paris, France (B.P., F.T.); and Inserm, UMR_S 1116; Université de Lorraine, Nancy, France (V.R., P.L.)
| | - Bruno Pannier
- From the Inserm, UMR_S 1166 (C.P., G.E., U.E., F.C.), CNRS UMR 7190 (P.C.), and CNRS, UMR 8256 (Z.L.), Sorbonne Universités, UPMC Univ Paris 06; Inserm, UMR_S 970, Sorbonne Paris Cité (J.-P.E., P.B., M.A., G.E., S.L., X.J.); Department of Cardiology, European Hospital of Georges Pompidou, Université Paris Descartes (N.D.); Centre d'Investigations Préventives et Cliniques (IPC Center), Paris, France (B.P., F.T.); and Inserm, UMR_S 1116; Université de Lorraine, Nancy, France (V.R., P.L.)
| | - Veronique Regnault
- From the Inserm, UMR_S 1166 (C.P., G.E., U.E., F.C.), CNRS UMR 7190 (P.C.), and CNRS, UMR 8256 (Z.L.), Sorbonne Universités, UPMC Univ Paris 06; Inserm, UMR_S 970, Sorbonne Paris Cité (J.-P.E., P.B., M.A., G.E., S.L., X.J.); Department of Cardiology, European Hospital of Georges Pompidou, Université Paris Descartes (N.D.); Centre d'Investigations Préventives et Cliniques (IPC Center), Paris, France (B.P., F.T.); and Inserm, UMR_S 1116; Université de Lorraine, Nancy, France (V.R., P.L.)
| | - Frederique Thomas
- From the Inserm, UMR_S 1166 (C.P., G.E., U.E., F.C.), CNRS UMR 7190 (P.C.), and CNRS, UMR 8256 (Z.L.), Sorbonne Universités, UPMC Univ Paris 06; Inserm, UMR_S 970, Sorbonne Paris Cité (J.-P.E., P.B., M.A., G.E., S.L., X.J.); Department of Cardiology, European Hospital of Georges Pompidou, Université Paris Descartes (N.D.); Centre d'Investigations Préventives et Cliniques (IPC Center), Paris, France (B.P., F.T.); and Inserm, UMR_S 1116; Université de Lorraine, Nancy, France (V.R., P.L.)
| | - Xavier Jouven
- From the Inserm, UMR_S 1166 (C.P., G.E., U.E., F.C.), CNRS UMR 7190 (P.C.), and CNRS, UMR 8256 (Z.L.), Sorbonne Universités, UPMC Univ Paris 06; Inserm, UMR_S 970, Sorbonne Paris Cité (J.-P.E., P.B., M.A., G.E., S.L., X.J.); Department of Cardiology, European Hospital of Georges Pompidou, Université Paris Descartes (N.D.); Centre d'Investigations Préventives et Cliniques (IPC Center), Paris, France (B.P., F.T.); and Inserm, UMR_S 1116; Université de Lorraine, Nancy, France (V.R., P.L.)
| | - François Cambien
- From the Inserm, UMR_S 1166 (C.P., G.E., U.E., F.C.), CNRS UMR 7190 (P.C.), and CNRS, UMR 8256 (Z.L.), Sorbonne Universités, UPMC Univ Paris 06; Inserm, UMR_S 970, Sorbonne Paris Cité (J.-P.E., P.B., M.A., G.E., S.L., X.J.); Department of Cardiology, European Hospital of Georges Pompidou, Université Paris Descartes (N.D.); Centre d'Investigations Préventives et Cliniques (IPC Center), Paris, France (B.P., F.T.); and Inserm, UMR_S 1116; Université de Lorraine, Nancy, France (V.R., P.L.)
| | - Patrick Lacolley
- From the Inserm, UMR_S 1166 (C.P., G.E., U.E., F.C.), CNRS UMR 7190 (P.C.), and CNRS, UMR 8256 (Z.L.), Sorbonne Universités, UPMC Univ Paris 06; Inserm, UMR_S 970, Sorbonne Paris Cité (J.-P.E., P.B., M.A., G.E., S.L., X.J.); Department of Cardiology, European Hospital of Georges Pompidou, Université Paris Descartes (N.D.); Centre d'Investigations Préventives et Cliniques (IPC Center), Paris, France (B.P., F.T.); and Inserm, UMR_S 1116; Université de Lorraine, Nancy, France (V.R., P.L.).
| |
Collapse
|
11
|
Tam RCY, Lee ALH, Yang W, Lau CS, Chan VSF. Systemic Lupus Erythematosus Patients Exhibit Reduced Expression of CLEC16A Isoforms in Peripheral Leukocytes. Int J Mol Sci 2015; 16:14428-40. [PMID: 26121298 PMCID: PMC4519850 DOI: 10.3390/ijms160714428] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 05/28/2015] [Accepted: 06/15/2015] [Indexed: 02/06/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a prototypic autoimmune disease with multiple etiological factors. The SLE susceptibility locus on chromosome 16p13 encodes a novel gene CLEC16A and its functional relationship with SLE is unclear. This study aimed to investigate the expression correlation of the two major CLEC16A spliced transcripts with SLE development. Expressions of the long (V1) and short (V2) CLEC16A isoforms in the peripheral blood mononuclear cells (PBMCs) were assayed by quantitative real time PCR and compared between healthy individuals and SLE patients. Correlation of CLEC16A isoform expression levels with SLE susceptibility, disease severity and twelve clinical parameters were also evaluated. Full length transcripts of CLEC16A V1 and V2 isoforms were readily amplified from PBMCs of healthy controls and patients at varying abundance. Compared with healthy controls (n = 86), expression levels of V1 and V2 were significantly reduced by ~two- and four-fold respectively in SLE patients (n = 181). The relative V2/V1 ratio was also significantly reduced by approximately two-fold. With regard to SLE disease parameters, only a weak positive correlation was found between CLEC16A V1 expression levels and SLE disease activity index (SLEDAI) score. Taken together, CLEC16A was found to be a susceptibility factor for SLE, with possible contribution to the development of the disease.
Collapse
Affiliation(s)
- Rachel C Y Tam
- Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China.
| | - Alfred L H Lee
- Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China.
| | - Wanling Yang
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China.
| | - Chak Sing Lau
- Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China.
| | - Vera S F Chan
- Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China.
| |
Collapse
|
12
|
van Luijn MM, Kreft KL, Jongsma ML, Mes SW, Wierenga-Wolf AF, van Meurs M, Melief MJ, der Kant RV, Janssen L, Janssen H, Tan R, Priatel JJ, Neefjes J, Laman JD, Hintzen RQ. Multiple sclerosis-associated CLEC16A controls HLA class II expression via late endosome biogenesis. Brain 2015; 138:1531-47. [PMID: 25823473 DOI: 10.1093/brain/awv080] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 01/26/2015] [Indexed: 01/20/2023] Open
Abstract
C-type lectins are key players in immune regulation by driving distinct functions of antigen-presenting cells. The C-type lectin CLEC16A gene is located at 16p13, a susceptibility locus for several autoimmune diseases, including multiple sclerosis. However, the function of this gene and its potential contribution to these diseases in humans are poorly understood. In this study, we found a strong upregulation of CLEC16A expression in the white matter of multiple sclerosis patients (n = 14) compared to non-demented controls (n = 11), mainly in perivascular leukocyte infiltrates. Moreover, CLEC16A levels were significantly enhanced in peripheral blood mononuclear cells of multiple sclerosis patients (n = 69) versus healthy controls (n = 46). In peripheral blood mononuclear cells, CLEC16A was most abundant in monocyte-derived dendritic cells, in which it strongly co-localized with human leukocyte antigen class II. Treatment of these professional antigen-presenting cells with vitamin D, a key protective environmental factor in multiple sclerosis, downmodulated CLEC16A in parallel with human leukocyte antigen class II. Knockdown of CLEC16A in distinct types of model and primary antigen-presenting cells resulted in severely impaired cytoplasmic distribution and formation of human leucocyte antigen class II-positive late endosomes, as determined by immunofluorescence and electron microscopy. Mechanistically, CLEC16A participated in the molecular machinery of human leukocyte antigen class II-positive late endosome formation and trafficking to perinuclear regions, involving the dynein motor complex. By performing co-immunoprecipitations, we found that CLEC16A directly binds to two critical members of this complex, RILP and the HOPS complex. CLEC16A silencing in antigen-presenting cells disturbed RILP-mediated recruitment of human leukocyte antigen class II-positive late endosomes to perinuclear regions. Together, we identify CLEC16A as a pivotal gene in multiple sclerosis that serves as a direct regulator of the human leukocyte antigen class II pathway in antigen-presenting cells. These findings are a first step in coupling multiple sclerosis-associated genes to the regulation of the strongest genetic factor in multiple sclerosis, human leukocyte antigen class II.
Collapse
Affiliation(s)
- Marvin M van Luijn
- 1 Department of Immunology and MS Center ErasMS, Erasmus MC, University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Karim L Kreft
- 2 Department of Neurology and MS Center ErasMS, Erasmus MC, University Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands
| | - Marlieke L Jongsma
- 3 Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Steven W Mes
- 1 Department of Immunology and MS Center ErasMS, Erasmus MC, University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Annet F Wierenga-Wolf
- 1 Department of Immunology and MS Center ErasMS, Erasmus MC, University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Marjan van Meurs
- 1 Department of Immunology and MS Center ErasMS, Erasmus MC, University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Marie-José Melief
- 1 Department of Immunology and MS Center ErasMS, Erasmus MC, University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Rik van der Kant
- 3 Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Lennert Janssen
- 3 Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Hans Janssen
- 3 Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Rusung Tan
- 4 Department of Pathology, Sidra Medical and Research Center, Doha, Qatar 5 BC Children's Hospital and Department of Pathology and Laboratory Medicine, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - John J Priatel
- 5 BC Children's Hospital and Department of Pathology and Laboratory Medicine, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Jacques Neefjes
- 3 Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Jon D Laman
- 1 Department of Immunology and MS Center ErasMS, Erasmus MC, University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Rogier Q Hintzen
- 2 Department of Neurology and MS Center ErasMS, Erasmus MC, University Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands
| |
Collapse
|
13
|
A variant of CLEC16A gene confers protection for Vogt–Koyanagi–Harada syndrome but not for Behcet's disease in a Chinese Han population. Exp Eye Res 2015; 132:225-30. [DOI: 10.1016/j.exer.2015.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 12/13/2014] [Accepted: 01/06/2015] [Indexed: 11/21/2022]
|
14
|
Soleimanpour SA, Gupta A, Bakay M, Ferrari AM, Groff DN, Fadista J, Spruce LA, Kushner JA, Groop L, Seeholzer SH, Kaufman BA, Hakonarson H, Stoffers DA. The diabetes susceptibility gene Clec16a regulates mitophagy. Cell 2014; 157:1577-90. [PMID: 24949970 DOI: 10.1016/j.cell.2014.05.016] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 02/24/2014] [Accepted: 05/09/2014] [Indexed: 12/22/2022]
Abstract
Clec16a has been identified as a disease susceptibility gene for type 1 diabetes, multiple sclerosis, and adrenal dysfunction, but its function is unknown. Here we report that Clec16a is a membrane-associated endosomal protein that interacts with E3 ubiquitin ligase Nrdp1. Loss of Clec16a leads to an increase in the Nrdp1 target Parkin, a master regulator of mitophagy. Islets from mice with pancreas-specific deletion of Clec16a have abnormal mitochondria with reduced oxygen consumption and ATP concentration, both of which are required for normal β cell function. Indeed, pancreatic Clec16a is required for normal glucose-stimulated insulin release. Moreover, patients harboring a diabetogenic SNP in the Clec16a gene have reduced islet Clec16a expression and reduced insulin secretion. Thus, Clec16a controls β cell function and prevents diabetes by controlling mitophagy. This pathway could be targeted for prevention and control of diabetes and may extend to the pathogenesis of other Clec16a- and Parkin-associated diseases.
Collapse
Affiliation(s)
- Scott A Soleimanpour
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine and the Institute for Diabetes, Obesity and Metabolism of the University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Division of Metabolism, Endocrinology & Diabetes and Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Aditi Gupta
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine and the Institute for Diabetes, Obesity and Metabolism of the University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Marina Bakay
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Alana M Ferrari
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine and the Institute for Diabetes, Obesity and Metabolism of the University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - David N Groff
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine and the Institute for Diabetes, Obesity and Metabolism of the University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - João Fadista
- Lund University Diabetes Center, Department of Clinical Sciences, Diabetes & Endocrinology, Skåne University Hospital, Lund University, SE-205 02 Malmö, Sweden
| | - Lynn A Spruce
- Children's Hospital of Philadelphia Research Institute, Philadelphia, PA 19104, USA
| | - Jake A Kushner
- McNair Medical Institute, Pediatric Diabetes and Endocrinology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Leif Groop
- Lund University Diabetes Center, Department of Clinical Sciences, Diabetes & Endocrinology, Skåne University Hospital, Lund University, SE-205 02 Malmö, Sweden
| | - Steven H Seeholzer
- Children's Hospital of Philadelphia Research Institute, Philadelphia, PA 19104, USA
| | - Brett A Kaufman
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hakon Hakonarson
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Doris A Stoffers
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine and the Institute for Diabetes, Obesity and Metabolism of the University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|