1
|
Cao Y, Ren Q, Chang S, Cui W, Zhao P, Wang Y. N6-methyladenosine RNA methylation modification regulates the transcription of viral-derived E (XSR) miRNAs to promote ALV-J replication. Vet Microbiol 2024; 298:110218. [PMID: 39159504 DOI: 10.1016/j.vetmic.2024.110218] [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: 04/24/2024] [Revised: 08/07/2024] [Accepted: 08/13/2024] [Indexed: 08/21/2024]
Abstract
The E (XSR) element located in the 3'UTR of the ALV-J genome has the capability to transcribe and generate viral-derived E (XSR) miRNA. However, the biological function and transcriptional regulation mechanism of this process remain unclear. In this study, the impact of E (XSR) miRNA on ALV-J replication and the regulatory effect of N6-methyladenosine (m6A) methylation on its transcription were investigated. The results demonstrated that E (XSR) miRNA could stimulate ALV-J replication and suppress apoptosis in DF-1 cells in vitro. E (XSR) miRNA's promotion of ALV-J replication was not associated with the type I interferon pathway, but achieved by suppressing the expression of the host GPC5 gene. The transcription of E (XSR) miRNA could be promoted by m6A methylation modification, where m6A modification was found at the A6880 and A7016 sites of ALV-J gRNA. This study provides a new perspective on the transcription of ALV-J E (XSR) miRNA and its regulatory function in ALV-J replication.
Collapse
Affiliation(s)
- Yuqing Cao
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China; Shandong Provincial Key Laboratory of Zoonoses, Tai'an, China
| | - Qingling Ren
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China; Shandong Provincial Key Laboratory of Zoonoses, Tai'an, China
| | - Shuang Chang
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China; Shandong Provincial Key Laboratory of Zoonoses, Tai'an, China
| | - Wenping Cui
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China; Shandong Provincial Key Laboratory of Zoonoses, Tai'an, China
| | - Peng Zhao
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China; Shandong Provincial Key Laboratory of Zoonoses, Tai'an, China.
| | - Yixin Wang
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China; Shandong Provincial Key Laboratory of Zoonoses, Tai'an, China.
| |
Collapse
|
2
|
Iglesias Pastrana C, Navas González FJ, Macri M, Martínez Martínez MDA, Ciani E, Delgado Bermejo JV. Identification of novel genetic loci related to dromedary camel (Camelus dromedarius) morphometrics, biomechanics, and behavior by genome-wide association studies. BMC Vet Res 2024; 20:418. [PMID: 39294626 PMCID: PMC11409489 DOI: 10.1186/s12917-024-04263-w] [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: 06/24/2024] [Accepted: 09/03/2024] [Indexed: 09/21/2024] Open
Abstract
In the realm of animal breeding for sustainability, domestic camels have traditionally been valued for their milk and meat production. However, key aspects such as zoometrics, biomechanics, and behavior have often been overlooked in terms of their genetic foundations. Recognizing this gap, the present study perfomed genome-wide association analyses to identify genetic markers associated with zoometrics-, biomechanics-, and behavior-related traits in dromedary camels (Camelus dromedarius). 16 and 108 genetic markers were significantly associated (q < 0.05) at genome and chromosome-wide levels of significance, respectively, with zoometrics- (width, length, and perimeter/girth), biomechanics- (acceleration, displacement, spatial position, and velocity), and behavior-related traits (general cognition, intelligence, and Intelligence Quotient (IQ)) in dromedaries. In most association loci, the nearest protein-coding genes are linkedto neurodevelopmental and sensory disorders. This suggests that genetic variations related to neural development and sensory perception play crucial roles in shaping a dromedary camel's physical characteristics and behavior. In summary, this research advances our understanding of the genomic basis of essential traits in dromedary camels. Identifying specific genetic markers associated with zoometrics, biomechanics, and behavior provides valuable insights into camel domestication. Moreover, the links between these traits and genes related to neurodevelopmental and sensory disorders highlight the broader implications of domestication and modern selection on the health and welfare of dromedary camels. This knowledge could guide future breeding strategies, fostering a more holistic approach to camel husbandry and ensuring the sustainability of these animals in diverse agricultural contexts.
Collapse
Affiliation(s)
| | | | - Martina Macri
- Department of Genetics, Faculty of Veterinary Sciences, University of Córdoba, Córdoba, Spain
- Animal Breeding Consulting S.L, Parque Científico Tecnológico de Córdoba, Córdoba, Spain
| | | | - Elena Ciani
- Department of Biosciences, Biotechnologies and Environment, Faculty of Veterinary Sciences, University of Bari 'Aldo Moro', Bari, Italy
| | | |
Collapse
|
3
|
Yang X, Sullivan PF, Li B, Fan Z, Ding D, Shu J, Guo Y, Paschou P, Bao J, Shen L, Ritchie MD, Nave G, Platt ML, Li T, Zhu H, Zhao B. Multi-organ imaging-derived polygenic indexes for brain and body health. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.04.18.23288769. [PMID: 38883759 PMCID: PMC11177904 DOI: 10.1101/2023.04.18.23288769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
The UK Biobank (UKB) imaging project is a crucial resource for biomedical research, but is limited to 100,000 participants due to cost and accessibility barriers. Here we used genetic data to predict heritable imaging-derived phenotypes (IDPs) for a larger cohort. We developed and evaluated 4,375 IDP genetic scores (IGS) derived from UKB brain and body images. When applied to UKB participants who were not imaged, IGS revealed links to numerous phenotypes and stratified participants at increased risk for both brain and somatic diseases. For example, IGS identified individuals at higher risk for Alzheimer's disease and multiple sclerosis, offering additional insights beyond traditional polygenic risk scores of these diseases. When applied to independent external cohorts, IGS also stratified those at high disease risk in the All of Us Research Program and the Alzheimer's Disease Neuroimaging Initiative study. Our results demonstrate that, while the UKB imaging cohort is largely healthy and may not be the most enriched for disease risk management, it holds immense potential for stratifying the risk of various brain and body diseases in broader external genetic cohorts.
Collapse
Affiliation(s)
- Xiaochen Yang
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Patrick F. Sullivan
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bingxuan Li
- UCLA Samueli School of Engineering, Los Angeles, CA 90095, USA
| | - Zirui Fan
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dezheng Ding
- Department of Electrical and Systems Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Juan Shu
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Yuxin Guo
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Peristera Paschou
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Jingxuan Bao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marylyn D. Ritchie
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Biomedical Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Gideon Nave
- Marketing Department, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael L. Platt
- Marketing Department, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tengfei Li
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hongtu Zhu
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Statistics and Operations Research, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bingxin Zhao
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Biomedical Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Applied Mathematics and Computational Science Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for AI and Data Science for Integrated Diagnostics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Population Aging Research Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
4
|
Heshmatzad K, Naderi N, Maleki M, Abbasi S, Ghasemi S, Ashrafi N, Fazelifar AF, Mahdavi M, Kalayinia S. Role of non-coding variants in cardiovascular disease. J Cell Mol Med 2023; 27:1621-1636. [PMID: 37183561 PMCID: PMC10273088 DOI: 10.1111/jcmm.17762] [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: 10/31/2022] [Revised: 03/29/2023] [Accepted: 04/25/2023] [Indexed: 05/16/2023] Open
Abstract
Cardiovascular diseases (CVDs) constitute one of the significant causes of death worldwide. Different pathological states are linked to CVDs, which despite interventions and treatments, still have poor prognoses. The genetic component, as a beneficial tool in the risk stratification of CVD development, plays a role in the pathogenesis of this group of diseases. The emergence of genome-wide association studies (GWAS) have led to the identification of non-coding parts associated with cardiovascular traits and disorders. Variants located in functional non-coding regions, including promoters/enhancers, introns, miRNAs and 5'/3' UTRs, account for 90% of all identified single-nucleotide polymorphisms associated with CVDs. Here, for the first time, we conducted a comprehensive review on the reported non-coding variants for different CVDs, including hypercholesterolemia, cardiomyopathies, congenital heart diseases, thoracic aortic aneurysms/dissections and coronary artery diseases. Additionally, we present the most commonly reported genes involved in each CVD. In total, 1469 non-coding variants constitute most reports on familial hypercholesterolemia, hypertrophic cardiomyopathy and dilated cardiomyopathy. The application and identification of non-coding variants are beneficial for the genetic diagnosis and better therapeutic management of CVDs.
Collapse
Affiliation(s)
- Katayoun Heshmatzad
- Rajaie Cardiovascular Medical and Research CenterIran University of Medical SciencesTehranIran
| | - Niloofar Naderi
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research CenterIran University of Medical SciencesTehranIran
| | - Majid Maleki
- Rajaie Cardiovascular Medical and Research CenterIran University of Medical SciencesTehranIran
| | - Shiva Abbasi
- Rajaie Cardiovascular Medical and Research CenterIran University of Medical SciencesTehranIran
| | - Serwa Ghasemi
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research CenterIran University of Medical SciencesTehranIran
| | - Nooshin Ashrafi
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research CenterIran University of Medical SciencesTehranIran
| | - Amir Farjam Fazelifar
- Rajaie Cardiovascular Medical and Research CenterIran University of Medical SciencesTehranIran
| | - Mohammad Mahdavi
- Rajaie Cardiovascular Medical and Research CenterIran University of Medical SciencesTehranIran
| | - Samira Kalayinia
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research CenterIran University of Medical SciencesTehranIran
| |
Collapse
|
5
|
Li J, Gao L, Ye Y. HiSV: A control-free method for structural variation detection from Hi-C data. PLoS Comput Biol 2023; 19:e1010760. [PMID: 36608109 PMCID: PMC11960816 DOI: 10.1371/journal.pcbi.1010760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 04/01/2025] [Accepted: 11/24/2022] [Indexed: 01/07/2023] Open
Abstract
Structural variations (SVs) play an essential role in the evolution of human genomes and are associated with cancer genetics and rare disease. High-throughput chromosome capture (Hi-C) technology probed all genome-wide crosslinked chromatin to study the spatial architecture of chromosomes. Hi-C read pairs can span megabases, making the technology useful for detecting large-scale SVs. So far, the identification of SVs from Hi-C data is still in the early stages with only a few methods available. Therefore, we developed HiSV (Hi-C for Structural Variation), a control-free method for identifying large-scale SVs from a Hi-C sample. Inspired by the single image saliency detection model, HiSV constructed a saliency map of interaction frequencies and extracted saliency segments as large-scale SVs. By evaluating both simulated and real data, HiSV not only detected all variant types, but also achieved a higher level of accuracy and sensitivity than most existing methods. Moreover, our results on cancer cell lines showed that HiSV effectively detected eight complex SV events and identified two novel SVs of key factors associated with cancer development. Finally, we found that integrating the result of HiSV helped the WGS method to identify a total number of 94 novel SVs in two cancer cell lines.
Collapse
Affiliation(s)
- Junping Li
- Department of Computer Science, School of Computer Science and Technology, Xidian University, Xi’an, Shaanxi, China
| | - Lin Gao
- Department of Computer Science, School of Computer Science and Technology, Xidian University, Xi’an, Shaanxi, China
| | - Yusen Ye
- Department of Computer Science, School of Computer Science and Technology, Xidian University, Xi’an, Shaanxi, China
| |
Collapse
|
6
|
Busato D, Mossenta M, Dal Bo M, Macor P, Toffoli G. The Proteoglycan Glypican-1 as a Possible Candidate for Innovative Targeted Therapeutic Strategies for Pancreatic Ductal Adenocarcinoma. Int J Mol Sci 2022; 23:ijms231810279. [PMID: 36142190 PMCID: PMC9499405 DOI: 10.3390/ijms231810279] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/02/2022] [Accepted: 09/04/2022] [Indexed: 11/19/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) accounts for 90% of all pancreatic cancers, with a 5-year survival rate of 7% and 80% of patients diagnosed with advanced or metastatic malignancies. Despite recent advances in diagnostic testing, surgical techniques, and systemic therapies, there remain limited options for the effective treatment of PDAC. There is an urgent need to develop targeted therapies that are able to differentiate between cancerous and non-cancerous cells to reduce side effects and better inhibit tumor growth. Antibody-targeted strategies are a potentially effective option for introducing innovative therapies. Antibody-based immunotherapies and antibody-conjugated nanoparticle-based targeted therapies with antibodies targeting specific tumor-associated antigens (TAA) can be proposed. In this context, glypican-1 (GPC1), which is highly expressed in PDAC and not expressed or expressed at very low levels in non-malignant lesions and healthy pancreatic tissues, is a useful TAA that can be achieved by a specific antibody-based immunotherapy and antibody-conjugated nanoparticle-based targeted therapy. In this review, we describe the main clinical features of PDAC. We propose the proteoglycan GPC1 as a useful TAA for PDAC-targeted therapies. We also provide a digression on the main developed approaches of antibody-based immunotherapy and antibody-conjugated nanoparticle-based targeted therapy, which can be used to target GPC1.
Collapse
Affiliation(s)
- Davide Busato
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
- Correspondence: ; Tel.: +39-0434-659816
| | - Monica Mossenta
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| | - Paolo Macor
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| |
Collapse
|
7
|
Hayashida K, Aquino RS, Park PW. Coreceptor Functions of Cell Surface Heparan Sulfate Proteoglycans. Am J Physiol Cell Physiol 2022; 322:C896-C912. [PMID: 35319900 PMCID: PMC9109798 DOI: 10.1152/ajpcell.00050.2022] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Receptor-ligand interactions play an important role in many biological processes by triggering specific cellular responses. These interactions are frequently regulated by coreceptors that facilitate, alter, or inhibit signaling. Coreceptors work in parallel with other specific and accessory molecules to coordinate receptor-ligand interactions. Cell surface heparan sulfate proteoglycans (HSPGs) function as unique coreceptors because they can bind to many ligands and receptors through their HS and core protein motifs. Cell surface HSPGs are typically expressed in abundance of the signaling receptors and, thus, are capable of mediating the initial binding of ligands to the cell surface. HSPG coreceptors do not possess kinase domains or intrinsic enzyme activities and, for the most part, binding to cell surface HSPGs does not directly stimulate intracellular signaling. Because of these features, cell surface HSPGs primarily function as coreceptors for many receptor-ligand interactions. Given that cell surface HSPGs are widely conserved, they likely serve fundamental functions to preserve basic physiological processes. Indeed, cell surface HSPGs can support specific cellular interactions with growth factors, morphogens, chemokines, extracellular matrix (ECM) components, and microbial pathogens and their secreted virulence factors. Through these interactions, HSPG coreceptors regulate cell adhesion, proliferation, migration and differentiation, and impact the onset, progression, and outcome of pathophysiological processes, such as development, tissue repair, inflammation, infection, and tumorigenesis. This review seeks to provide an overview of the various mechanisms of how cell surface HSPGs function as coreceptors.
Collapse
Affiliation(s)
- Kazutaka Hayashida
- Department of Medicine, Boston Children's Hospital, Boston, MA, United States
| | - Rafael S Aquino
- Department of Medicine, Boston Children's Hospital, Boston, MA, United States
| | - Pyong Woo Park
- Department of Medicine, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
8
|
Liu YC, Wierbowski BM, Salic A. Hedgehog pathway modulation by glypican 3-conjugated heparan sulfate. J Cell Sci 2022; 135:274739. [PMID: 35142364 PMCID: PMC8977055 DOI: 10.1242/jcs.259297] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 02/04/2022] [Indexed: 11/20/2022] Open
Abstract
Glypicans are a family of cell surface heparan sulfate proteoglycans that play critical roles in multiple cell signaling pathways. Glypicans consist of a globular core, an unstructured stalk modified with sulfated glycosaminoglycan chains, and a glycosylphosphatidylinositol anchor. Though these structural features are conserved, their individual contribution to glypican function remains obscure. Here, we investigate how glypican 3 (GPC3), which is mutated in Simpson-Golabi-Behmel tissue overgrowth syndrome, regulates Hedgehog signaling. We find that GPC3 is necessary for the Hedgehog response, surprisingly controlling a downstream signal transduction step. Purified GPC3 ectodomain rescues signaling when artificially recruited to the surface of GPC3-deficient cells but has dominant-negative activity when unattached. Strikingly, the purified stalk, modified with heparan sulfate but not chondroitin sulfate, is necessary and sufficient for activity. Our results demonstrate a novel function for GPC3-associated heparan sulfate and provide a framework for the functional dissection of glycosaminoglycans by in vivo biochemical complementation. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Yulu Cherry Liu
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA.,Department of Biology, Hood College, Frederick, MD 21701, USA
| | | | - Adrian Salic
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
9
|
Abstract
Glypicans are proteoglycans that are bound to the outer surface of the plasma membrane by a glycosylphosphatidylinositol anchor. The mammalian genome contains six members of the glypican family (GPC1 to GPC6). Although the degree of sequence homology within the family is rather low, the three-dimensional structure of these proteoglycans is highly conserved. Glypicans are predominantly expressed during embryonic development. Genetic and biochemical studies have shown that glypicans can stimulate or inhibit the signaling pathways triggered by Wnts, Hedgehogs, Fibroblast Growth Factors, and Bone Morphogenetic Proteins. The study of mutant mouse strains demonstrated that glypicans have important functions in the developmental morphogenesis of various organs. In addition, a role of glypicans in synapsis formation has been established. Notably, glypican loss-of-function mutations are the cause of three human inherited syndromes. Recent analysis of glypican compound mutant mice have demonstrated that members of this protein family display redundant functions during embryonic development.
Collapse
Affiliation(s)
- Jorge Filmus
- Biological Sciences, Sunnybrook Research Institute, and Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
10
|
Ray PE, Li J, Das JR, Yu J. Association of circulating fibroblast growth factor-2 with progression of HIV-chronic kidney diseases in children. Pediatr Nephrol 2021; 36:3933-3944. [PMID: 34125285 PMCID: PMC8602783 DOI: 10.1007/s00467-021-05075-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/08/2021] [Accepted: 03/26/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Children living with HIV frequently show high plasma levels of fibroblast growth factor-2 (FGF-2/bFGF). FGF-2 accelerates the progression of several experimental kidney diseases; however, the role of circulating FGF-2 in childhood HIV-chronic kidney diseases (HIV-CKDs) is unknown. We carried out this study to determine whether high plasma FGF-2 levels were associated with the development of HIV-CKDs in children. METHODS The plasma and urine FGF-2 levels were measured in 84 children (< 12 years of age) living with HIV during the pre-modern antiretroviral era, and followed for at least 3 years to determine the prevalence of proteinuria and HIV-CKDs. We also assessed the distribution of the kidney FGF-2 binding sites by autoradiography and Alcian blue staining, and explored potential mechanisms by which circulating FGF-2 may precipitate HIV-CKDs in cultured kidney epithelial and mononuclear cells derived from children with HIV-CKDs. RESULTS High plasma FGF-2 levels were associated with a high viral load. Thirteen children (~ 15%) developed HIV-CKDs and showed a large reservoir of FGF-2 low-affinity binding sites in the kidney, which can facilitate the recruitment of circulating FGF-2. Children with high plasma and urine FGF-2 levels had 73-fold increased odds (95% CI 9-791) of having HIV-CKDs relative to those with normal FGF-2 values. FGF-2 induced the proliferation and decreased the expression of APOL-1 mRNA in podocytes, and increased the attachment and survival of infected mononuclear cells cultured from children with HIV-CKDs. CONCLUSIONS High plasma FGF-2 levels appear to be an additional risk factor for developing progressive childhood HIV-CKDs.
Collapse
Affiliation(s)
- Patricio E Ray
- Child Health Research Center, Department of Pediatrics, School of Medicine, University of Virginia, Room 2120, MR4 Building, 409 Lane Road, Charlottesville, VA, 22908, USA.
| | - Jinliang Li
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC, USA
- Department of Pediatrics, The George Washington University School of Medicine, Washington, DC, USA
| | - Jharna R Das
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC, USA
- Department of Pediatrics, The George Washington University School of Medicine, Washington, DC, USA
| | - Jing Yu
- Child Health Research Center, Department of Pediatrics, School of Medicine, University of Virginia, Room 2120, MR4 Building, 409 Lane Road, Charlottesville, VA, 22908, USA
| |
Collapse
|
11
|
Tamura H. Trends in pediatric nephrotic syndrome. World J Nephrol 2021; 10:88-100. [PMID: 34631479 PMCID: PMC8477269 DOI: 10.5527/wjn.v10.i5.88] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/15/2021] [Accepted: 08/10/2021] [Indexed: 02/06/2023] Open
Abstract
Nephrotic syndrome (NS) is relatively common in children, with most of its histological types being minimal changed disease. Its etiology has long been attributed to lymphocyte (especially T-cell) dysfunction, while T-cell-mediated vascular hyperpermeability increases protein permeability in glomerular capillaries, leading to proteinuria and hypoproteinemia. Based on this etiology, steroids and immunosuppressive drugs that are effective against this disease have also been considered to correct T-cell dysfunction. However, in recent years, this has been questioned. The primary cause of NS has been considered damage to glomerular epithelial cells and podocyte-related proteins. Therefore, we first describe the changes in expression of molecules involved in NS etiology, and then describe the mechanism by which abnormal expression of these molecules induces proteinuria. Finally, we consider the mechanism by which infection causes the recurrence of NS.
Collapse
Affiliation(s)
- Hiroshi Tamura
- Department of Pediatrics, Kumamoto University, Kumamoto 8608556, Japan
| |
Collapse
|
12
|
Abstract
Diabetes is a complex disorder responsible for the mortality and morbidity of millions of individuals worldwide. Although many approaches have been used to understand and treat diabetes, the role of proteoglycans, in particular heparan sulfate proteoglycans (HSPGs), has only recently received attention. The HSPGs are heterogeneous, highly negatively charged, and are found in all cells primarily attached to the plasma membrane or present in the extracellular matrix (ECM). HSPGs are involved in development, cell migration, signal transduction, hemostasis, inflammation, and antiviral activity, and regulate cytokines, chemokines, growth factors, and enzymes. Hyperglycemia, accompanying diabetes, increases reactive oxygen species and upregulates the enzyme heparanase that degrades HSPGs or affects the synthesis of the HSPGs altering their structure. The modified HSPGs in the endothelium and ECM in the blood vessel wall contribute to the nephropathy, cardiovascular disease, and retinopathy seen in diabetes. Besides the blood vessel, other cells and tissues in the heart, kidney, and eye are affected by diabetes. Although not well understood, the adipose tissue, intestine, and brain also reveal HSPG changes associated with diabetes. Further, HSPGs are significantly involved in protecting the β cells of the pancreas from autoimmune destruction and could be a focus of prevention of type I diabetes. In some circumstances, HSPGs may contribute to the pathology of the disease. Understanding the role of HSPGs and how they are modified by diabetes may lead to new treatments as well as preventative measures to reduce the morbidity and mortality associated with this complex condition.
Collapse
Affiliation(s)
- Linda M Hiebert
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|
13
|
Hu S, Han R, Chen L, Qin W, Xu X, Shi J, Zhu X, Zhang M, Zeng C, Tang Z, Bao H, Liu Z. Upregulated LRRC55 promotes BK channel activation and aggravates cell injury in podocytes. J Exp Med 2021; 218:e20192373. [PMID: 33346797 PMCID: PMC7756252 DOI: 10.1084/jem.20192373] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 07/27/2020] [Accepted: 09/16/2020] [Indexed: 12/15/2022] Open
Abstract
Podocyte injury is a common hallmark in various glomerular diseases. The level of LRRC55 was increased in podocytes of patients with focal segmental glomerulosclerosis (FSGS), diabetic nephropathy (DN), and membranous nephropathy (MN). Upregulated LRRC55 and increased intracellular Ca2+ led to BK channel activation and the loss of intracellular potassium, resulting in apoptosome formation and caspase-3 activation in angiotensin II (Ang II)-treated podocytes. Knockout of Lrrc55 or the BK channel prevented the BK current and ameliorated podocyte injury in Ang II-treated mice. Upstream, NFATc3 regulated the expression of LRRC55. Increased LRRC55 expression in podocytes was also evident in animal models of FSGS, DN, and MN. Treatment with losartan or LRRC55 siRNA suppressed LRRC55 expression, prevented BK channel activation, and attenuated podocyte injury in animal models of FSGS, DN, and MN. In conclusion, upregulated LRRC55 promotes BK channel activation and aggravates cell injury in podocytes in FSGS, DN, and MN. LRRC55 inhibition may represent a new therapeutic approach for podocyte injury.
Collapse
Affiliation(s)
- Shuai Hu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Runhong Han
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Long Chen
- National Standard Laboratory of Pharmacology for Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Weisong Qin
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Xiaodong Xu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Jingsong Shi
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Xiaodong Zhu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Mingchao Zhang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Caihong Zeng
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Zheng Tang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Hao Bao
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Zhihong Liu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| |
Collapse
|
14
|
Agrawal S, Ransom RF, Saraswathi S, Garcia-Gonzalo E, Webb A, Fernandez-Martinez JL, Popovic M, Guess AJ, Kloczkowski A, Benndorf R, Sadee W, Smoyer WE. Sulfatase 2 Is Associated with Steroid Resistance in Childhood Nephrotic Syndrome. J Clin Med 2021; 10:523. [PMID: 33540508 PMCID: PMC7867139 DOI: 10.3390/jcm10030523] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/20/2021] [Accepted: 01/23/2021] [Indexed: 01/17/2023] Open
Abstract
Glucocorticoid (GC) resistance complicates the treatment of ~10-20% of children with nephrotic syndrome (NS), yet the molecular basis for resistance remains unclear. We used RNAseq analysis and in silico algorithm-based approaches on peripheral blood leukocytes from 12 children both at initial NS presentation and after ~7 weeks of GC therapy to identify a 12-gene panel able to differentiate steroid resistant NS (SRNS) from steroid-sensitive NS (SSNS). Among this panel, subsequent validation and analyses of one biologically relevant candidate, sulfatase 2 (SULF2), in up to a total of 66 children, revealed that both SULF2 leukocyte expression and plasma arylsulfatase activity Post/Pre therapy ratios were greater in SSNS vs. SRNS. However, neither plasma SULF2 endosulfatase activity (measured by VEGF binding activity) nor plasma VEGF levels, distinguished SSNS from SRNS, despite VEGF's reported role as a downstream mediator of SULF2's effects in glomeruli. Experimental studies of NS-related injury in both rat glomeruli and cultured podocytes also revealed decreased SULF2 expression, which were partially reversible by GC treatment of podocytes. These findings together suggest that SULF2 levels and activity are associated with GC resistance in NS, and that SULF2 may play a protective role in NS via the modulation of downstream mediators distinct from VEGF.
Collapse
Affiliation(s)
- Shipra Agrawal
- Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA; (R.F.R.); (M.P.); (A.J.G.); (R.B.)
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA;
| | - Richard F. Ransom
- Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA; (R.F.R.); (M.P.); (A.J.G.); (R.B.)
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA;
| | - Saras Saraswathi
- Battelle Center for Mathematical Medicine at Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA;
| | | | - Amy Webb
- Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH 43210, USA;
| | | | - Milan Popovic
- Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA; (R.F.R.); (M.P.); (A.J.G.); (R.B.)
| | - Adam J. Guess
- Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA; (R.F.R.); (M.P.); (A.J.G.); (R.B.)
| | - Andrzej Kloczkowski
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA;
- Battelle Center for Mathematical Medicine at Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA;
| | - Rainer Benndorf
- Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA; (R.F.R.); (M.P.); (A.J.G.); (R.B.)
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA;
| | - Wolfgang Sadee
- Department of Cancer Biology and Genetics, Center for Pharmacogenomics, The Ohio State University College of Medicine, Columbus, OH 43210, USA;
| | - William E. Smoyer
- Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA; (R.F.R.); (M.P.); (A.J.G.); (R.B.)
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA;
| | | |
Collapse
|
15
|
Prognostic value of Glypican family genes in early-stage pancreatic ductal adenocarcinoma after pancreaticoduodenectomy and possible mechanisms. BMC Gastroenterol 2020; 20:415. [PMID: 33302876 PMCID: PMC7731467 DOI: 10.1186/s12876-020-01560-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 11/24/2020] [Indexed: 01/05/2023] Open
Abstract
Background This study explored the prognostic significance of Glypican (GPC) family genes in patients with pancreatic ductal adenocarcinoma (PDAC) after pancreaticoduodenectomy using data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). Methods A total of 112 PDAC patients from TCGA and 48 patients from GEO were included in the analysis. The relationship between overall survival and the expression of GPC family genes as well as basic clinical characteristics was analyzed using the Kaplan-Meier method with the log-rank test. Joint effects survival analysis was performed to further examine the relationship between GPC genes and prognosis. A prognosis nomogram was established based on clinical characteristics and prognosis-related genes. Prognosis-related genes were investigated by genome-wide co-expression analysis and gene set enrichment analysis (GSEA) was carried out to identify potential mechanisms of these genes affecting prognosis. Results In TCGA database, high expression of GPC2, GPC3, and GPC5 was significantly associated with favorable survival (log-rank P = 0.031, 0.021, and 0.028, respectively; adjusted P value = 0.005, 0.022, and 0.020, respectively), and joint effects analysis of these genes was effective for prognosis prediction. The prognosis nomogram was applied to predict the survival probability using the total scores calculated. Genome-wide co-expression and GSEA analysis suggested that the GPC2 may affect prognosis through sequence-specific DNA binding, protein transport, cell differentiation and oncogenic signatures (KRAS, RAF, STK33, and VEGFA). GPC3 may be related to cell adhesion, angiogenesis, inflammatory response, signaling pathways like Ras, Rap1, PI3K-Akt, chemokine, GPCR, and signatures like cyclin D1, p53, PTEN. GPC5 may be involved in transcription factor complex, TFRC1, oncogenic signatures (HOXA9 and BMI1), gene methylation, phospholipid metabolic process, glycerophospholipid metabolism, cell cycle, and EGFR pathway. Conclusion GPC2, GPC3, and GPC5 expression may serve as prognostic indicators in PDAC, and combination of these genes showed a higher efficiency for prognosis prediction.
Collapse
|
16
|
Abstract
The glomerular filtration barrier is a highly specialized capillary wall comprising fenestrated endothelial cells, podocytes, and an intervening basement membrane. In glomerular disease, this barrier loses functional integrity, allowing the passage of macromolecules and cells, and there are associated changes in both cell morphology and the extracellular matrix. Over the past 3 decades, there has been a transformation in our understanding about glomerular disease, fueled by genetic discovery, and this is leading to exciting advances in our knowledge about glomerular biology and pathophysiology. In current clinical practice, a genetic diagnosis already has important implications for management, ranging from estimating the risk of disease recurrence post-transplant to the life-changing advances in the treatment of atypical hemolytic uremic syndrome. Improving our understanding about the mechanistic basis of glomerular disease is required for more effective and personalized therapy options. In this review, we describe genotype and phenotype correlations for genetic disorders of the glomerular filtration barrier, with a particular emphasis on how these gene defects cluster by both their ontology and patterns of glomerular pathology.
Collapse
Affiliation(s)
- Anna S. Li
- Division of Cell-Matrix Biology and Regenerative Medicine, Wellcome Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
- Department of Nephrology, Manchester University Hospitals National Health Service Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Jack F. Ingham
- Division of Cell-Matrix Biology and Regenerative Medicine, Wellcome Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
| | - Rachel Lennon
- Division of Cell-Matrix Biology and Regenerative Medicine, Wellcome Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
- Department of Paediatric Nephrology, Royal Manchester Children’s Hospital, Manchester University Hospitals National Health Service Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| |
Collapse
|
17
|
Matsuura R, Hiraishi A, Holzman LB, Hanayama H, Harano K, Nakamura E, Hamasaki Y, Doi K, Nangaku M, Noiri E. SHROOM3, the gene associated with chronic kidney disease, affects the podocyte structure. Sci Rep 2020; 10:21103. [PMID: 33273487 PMCID: PMC7713385 DOI: 10.1038/s41598-020-77952-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 11/15/2020] [Indexed: 01/11/2023] Open
Abstract
Chronic kidney disease is a public health burden and it remains unknown which genetic loci are associated with kidney function in the Japanese population, our genome-wide association study using the Biobank Japan dataset (excluding secondary kidney diseases, such as diabetes mellitus) clearly revealed that almost half of the top 50 single nucleotide polymorphisms associated with estimated glomerular filtration rate are located in the SHROOM3 gene, suggesting that SHROOM3 will be responsible for kidney function. Thus, to confirm this finding, supportive functional analyses were performed on Shroom3 in mice using fullerene-based siRNA delivery, which demonstrated that Shroom3 knockdown led to albuminuria and podocyte foot process effacement. The in vitro experiment shows that knockdown of Shroom3 caused defective formation of lamellipodia in podocyte, which would lead to the disruption of slit diaphragm. These results from the GWAS, in vivo and in vitro experiment were consistent with recent studies reporting that albuminuria leads to impairment of kidney function.
Collapse
Affiliation(s)
- Ryo Matsuura
- Department of Nephrology and Endocrinology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Atsuko Hiraishi
- Department of Nephrology and Endocrinology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
- Division of Genomic Medicine and Disease Prevention, Institute of Medical Science, The University of Tokyo, Shirokanedai, 4-6-1 Minato-ku, Tokyo, 108-8639, Japan
| | - Lawrence B Holzman
- Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, 19104, USA
| | - Hiroki Hanayama
- Department of Chemistry, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Koji Harano
- Department of Chemistry, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Eiichi Nakamura
- Department of Chemistry, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Yoshifumi Hamasaki
- Department of Hemodialysis and Apheresis, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kent Doi
- Department of Acute Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Masaomi Nangaku
- Department of Nephrology and Endocrinology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Eisei Noiri
- Department of Nephrology and Endocrinology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
- National Center Biobank Network, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku, Tokyo, 162-8655, Japan.
| |
Collapse
|
18
|
Park E, Lee C, Kim NKD, Ahn YH, Park YS, Lee JH, Kim SH, Cho MH, Cho H, Yoo KH, Shin JI, Kang HG, Ha IS, Park WY, Cheong HI. Genetic Study in Korean Pediatric Patients with Steroid-Resistant Nephrotic Syndrome or Focal Segmental Glomerulosclerosis. J Clin Med 2020; 9:jcm9062013. [PMID: 32604935 PMCID: PMC7355646 DOI: 10.3390/jcm9062013] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 06/16/2020] [Accepted: 06/19/2020] [Indexed: 11/17/2022] Open
Abstract
Steroid-resistant nephrotic syndrome (SRNS) is one of the major causes of end-stage renal disease (ESRD) in childhood and is mostly associated with focal segmental glomerulosclerosis (FSGS). More than 50 monogenic causes of SRNS or FSGS have been identified. Recently, the mutation detection rate in pediatric patients with SRNS has been reported to be approximately 30%. In this study, genotype-phenotype correlations in a cohort of 291 Korean pediatric patients with SRNS/FSGS were analyzed. The overall mutation detection rate was 43.6% (127 of 291 patients). WT1 was the most common causative gene (23.6%), followed by COQ6 (8.7%), NPHS1 (8.7%), NUP107 (7.1%), and COQ8B (6.3%). Mutations in COQ6, NUP107, and COQ8B were more frequently detected, and mutations in NPHS2 were less commonly detected in this cohort than in study cohorts from Western countries. The mutation detection rate was higher in patients with congenital onset, those who presented with proteinuria or chronic kidney disease/ESRD, and those who did not receive steroid treatment. Genetic diagnosis in patients with SRNS provides not only definitive diagnosis but also valuable information for decisions on treatment policy and prediction of prognosis. Therefore, further genotype-phenotype correlation studies are required.
Collapse
Affiliation(s)
- Eujin Park
- Department of Pediatrics, Seoul National University College of Medicine, Seoul 03080, Korea; (E.P.); (Y.H.A.); (H.G.K.); (I.-S.H.)
- Department of Pediatrics, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul 07441, Korea
| | - Chung Lee
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Korea; (C.L.); (N.K.D.K.); (W.-Y.P.)
- GENINUS Inc., Seoul 05836, Korea
| | - Nayoung K. D. Kim
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Korea; (C.L.); (N.K.D.K.); (W.-Y.P.)
- GENINUS Inc., Seoul 05836, Korea
| | - Yo Han Ahn
- Department of Pediatrics, Seoul National University College of Medicine, Seoul 03080, Korea; (E.P.); (Y.H.A.); (H.G.K.); (I.-S.H.)
| | - Young Seo Park
- Department of Pediatrics, Asan Medical Center Children’s Hospital, University of Ulsan College of Medicine, Seoul 05505, Korea; (Y.S.P.); (J.H.L.)
| | - Joo Hoon Lee
- Department of Pediatrics, Asan Medical Center Children’s Hospital, University of Ulsan College of Medicine, Seoul 05505, Korea; (Y.S.P.); (J.H.L.)
| | - Seong Heon Kim
- Department of Pediatrics, Pusan National University Children’s Hospital, Yangsan 50612, Korea;
| | - Min Hyun Cho
- Department of Pediatrics, Kyungpook National University School of Medicine, Daegu 41944, Korea;
| | - Heeyeon Cho
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea;
| | - Kee Hwan Yoo
- Department of Pediatrics, Korea University Guro Hospital, Seoul 02841, Korea;
| | - Jae Il Shin
- Department of Pediatrics, Yonsei University College of Medicine, Seoul 03722, Korea;
- Division of Pediatric Nephrology, Severance Children’s Hospital, Seoul 03722, Korea
| | - Hee Gyung Kang
- Department of Pediatrics, Seoul National University College of Medicine, Seoul 03080, Korea; (E.P.); (Y.H.A.); (H.G.K.); (I.-S.H.)
| | - Il-Soo Ha
- Department of Pediatrics, Seoul National University College of Medicine, Seoul 03080, Korea; (E.P.); (Y.H.A.); (H.G.K.); (I.-S.H.)
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Korea; (C.L.); (N.K.D.K.); (W.-Y.P.)
- GENINUS Inc., Seoul 05836, Korea
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
| | - Hae Il Cheong
- Department of Pediatrics, Seoul National University College of Medicine, Seoul 03080, Korea; (E.P.); (Y.H.A.); (H.G.K.); (I.-S.H.)
- Correspondence: ; Tel.: +82-2-2072-2810
| |
Collapse
|
19
|
Pessentheiner AR, Ducasa GM, Gordts PLSM. Proteoglycans in Obesity-Associated Metabolic Dysfunction and Meta-Inflammation. Front Immunol 2020; 11:769. [PMID: 32508807 PMCID: PMC7248225 DOI: 10.3389/fimmu.2020.00769] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 04/06/2020] [Indexed: 12/16/2022] Open
Abstract
Proteoglycans are a specific subset of glycoproteins found at the cell surface and in the extracellular matrix, where they interact with a plethora of proteins involved in metabolic homeostasis and meta-inflammation. Over the last decade, new insights have emerged on the mechanism and biological significance of these interactions in the context of diet-induced disorders such as obesity and type-2 diabetes. Complications of energy metabolism drive most diet-induced metabolic disorders, which results in low-grade chronic inflammation, thereby affecting proper function of many vital organs involved in energy homeostasis, such as the brain, liver, kidney, heart and adipose tissue. Here, we discuss how heparan, chondroitin and keratan sulfate proteoglycans modulate obesity-induced metabolic dysfunction and low-grade inflammation that impact the initiation and progression of obesity-associated morbidities.
Collapse
Affiliation(s)
- Ariane R. Pessentheiner
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA, United States
| | - G. Michelle Ducasa
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA, United States
| | - Philip L. S. M. Gordts
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA, United States
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
20
|
McPherson KC, Shields CA, Poudel B, Johnson AC, Taylor L, Stubbs C, Nichols A, Cornelius DC, Garrett MR, Williams JM. Altered renal hemodynamics is associated with glomerular lipid accumulation in obese Dahl salt-sensitive leptin receptor mutant rats. Am J Physiol Renal Physiol 2020; 318:F911-F921. [PMID: 32068459 DOI: 10.1152/ajprenal.00438.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The present study examined whether development of renal injury in the nondiabetic obese Dahl salt-sensitive leptin receptor mutant (SSLepRmutant) strain is associated with elevations in glomerular filtration rate and renal lipid accumulation. Baseline mean arterial pressure at 6 wk of age was similar between Dahl salt-sensitive wild-type (SSWT) and SSLepRmutant rats. However, by 18 wk of age, the SSLepRmutant strain developed hypertension, while the elevation in mean arterial pressure was not as severe in SSWT rats (192 ± 4 and 149 ± 6 mmHg, respectively). At baseline, proteinuria was fourfold higher in SSLepRmutant than SSWT rats and remained elevated throughout the study. The early development of progressive proteinuria was associated with renal hyperfiltration followed by a decline in renal function over the course of study in the SSLepRmutant compared with SSWT rats. Kidneys from the SSLepRmutant strain displayed more glomerulosclerosis and glomerular lipid accumulation than SSWT rats. Glomeruli were isolated from the renal cortex of both strains at 6 and 18 wk of age, and RNA sequencing was performed to identify genes and pathways driving glomerular injury. We observed significant increases in expression of the influx lipid transporters, chemokine (C-X-C motif) ligand 16 (Cxcl16) and scavenger receptor and fatty acid translocase (Cd36), respectively, and a significant decrease in expression of the efflux lipid transporter, ATP-binding cassette subfamily A member 2 (Abca2; cholesterol efflux regulatory protein 2), in SSLepRmutant compared with SSWT rats at 6 and 18 wk of age, which were validated by RT-PCR analysis. These data suggest an association between glomerular hyperfiltration and glomerular lipid accumulation during the early development of proteinuria associated with obesity.
Collapse
Affiliation(s)
- Kasi C McPherson
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Corbin A Shields
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Bibek Poudel
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Ashley C Johnson
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Lateia Taylor
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Cassandra Stubbs
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Alyssa Nichols
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Denise C Cornelius
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi.,Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Michael R Garrett
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jan M Williams
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
21
|
Hinrichs GR, Weyer K, Friis UG, Svenningsen P, Lund IK, Nielsen R, Mollet G, Antignac C, Bistrup C, Jensen BL, Birn H. Urokinase-type plasminogen activator contributes to amiloride-sensitive sodium retention in nephrotic range glomerular proteinuria in mice. Acta Physiol (Oxf) 2019; 227:e13362. [PMID: 31423748 DOI: 10.1111/apha.13362] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 08/12/2019] [Accepted: 08/15/2019] [Indexed: 12/12/2022]
Abstract
AIM Activation of sodium reabsorption by urinary proteases has been implicated in sodium retention associated with nephrotic syndrome. The study was designed to test the hypothesis that nephrotic proteinuria in mice after conditional deletion of podocin leads to urokinase-dependent, amiloride-sensitive plasmin-mediated sodium and water retention. METHODS Ten days after podocin knockout, urine and faeces were collected for 10 days in metabolic cages and analysed for electrolytes, plasminogen, protease activity and ability to activate γENaC by patch clamp and western blot. Mice were treated with amiloride (2.5 mg kg-1 for 2 days and 10 mg kg-1 for 2 days) or an anti-urokinase-type plasminogen activator (uPA) targeting antibody (120 mg kg-1 /24 h) and compared to controls. RESULTS Twelve days after deletion, podocin-deficient mice developed significant protein and albuminuria associated with increased body wt, ascites, sodium accumulation and suppressed plasma renin. This was associated with increased urinary excretion of plasmin and plasminogen that correlated with albumin excretion, urine protease activity co-migrating with active plasmin, and the ability of urine to induce an amiloride-sensitive inward current in M1 cells in vitro. Amiloride treatment in podocin-deficient mice resulted in weight loss, increased sodium excretion, normalization of sodium balance and prevention of the activation of plasminogen to plasmin in urine in a reversible way. Administration of uPA targeting antibody abolished urine activation of plasminogen, attenuated sodium accumulation and prevented cleavage of γENaC. CONCLUSIONS Nephrotic range glomerular proteinuria leads to urokinase-dependent intratubular plasminogen activation and γENaC cleavage which contribute to sodium accumulation.
Collapse
Affiliation(s)
- Gitte R. Hinrichs
- Department of Molecular Medicine, Cardiovascular and Renal Research University of Southern Denmark Odense Denmark
| | - Kathrin Weyer
- Department of Biomedicine Aarhus University Aarhus Denmark
| | - Ulla G. Friis
- Department of Molecular Medicine, Cardiovascular and Renal Research University of Southern Denmark Odense Denmark
| | - Per Svenningsen
- Department of Molecular Medicine, Cardiovascular and Renal Research University of Southern Denmark Odense Denmark
| | - Ida K. Lund
- The Finsen Laboratory Rigshospitalet Copenhagen Denmark
- Biotech Research & Innovation Centre (BRIC) University of Copenhagen Copenhagen Denmark
| | - Rikke Nielsen
- Department of Biomedicine Aarhus University Aarhus Denmark
| | - Géraldine Mollet
- Laboratory of Hereditary Kidney Diseases Imagine Institute Inserm, U1163 Paris Descartes‐Sorbonne Paris Cité University Paris France
| | - Corinne Antignac
- Laboratory of Hereditary Kidney Diseases Imagine Institute Inserm, U1163 Paris Descartes‐Sorbonne Paris Cité University Paris France
- Department of Genetics Necker Hospital Assistance Publique‐Hôpitaux de Paris Paris France
| | - Claus Bistrup
- Department of Nephrology Odense University Hospital Odense Denmark
- Department of Clinical Research University of Southern Denmark Odense Denmark
| | - Boye L. Jensen
- Department of Molecular Medicine, Cardiovascular and Renal Research University of Southern Denmark Odense Denmark
| | - Henrik Birn
- Department of Biomedicine Aarhus University Aarhus Denmark
- Department of Renal Medicine Aarhus University Hospital Aarhus Denmark
| |
Collapse
|
22
|
Bensimhon AR, Williams AE, Gbadegesin RA. Treatment of steroid-resistant nephrotic syndrome in the genomic era. Pediatr Nephrol 2019; 34:2279-2293. [PMID: 30280213 PMCID: PMC6445770 DOI: 10.1007/s00467-018-4093-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 09/13/2018] [Accepted: 09/18/2018] [Indexed: 12/25/2022]
Abstract
The pathogenesis of steroid-resistant nephrotic syndrome (SRNS) is not completely known. Recent advances in genomics have elucidated some of the molecular mechanisms and pathophysiology of the disease. More than 50 monogenic causes of SRNS have been identified; however, these genes are responsible for only a small fraction of SRNS in outbred populations. There are currently no guidelines for genetic testing in SRNS, but evidence from the literature suggests that testing should be guided by the genetic architecture of the disease in the population. Notably, most genetic forms of SRNS do not respond to current immunosuppressive therapies; however, a small subset of patients with monogenic SRNS will achieve partial or complete remission with specific immunomodulatory agents, presumably due to non-immunosuppressive effects of these agents. We suggest a pragmatic approach to the therapy of genetic SRNS, as there is no evidence-based algorithm for the management of the disease.
Collapse
Affiliation(s)
- Adam R. Bensimhon
- Department of Pediatrics, Division of Nephrology, Duke University Medical Center, Durham, NC 27710, USA
| | - Anna E. Williams
- Department of Pediatrics, Division of Nephrology, Duke University Medical Center, Durham, NC 27710, USA
| | - Rasheed A. Gbadegesin
- Department of Pediatrics, Division of Nephrology, Duke University Medical Center, Durham, NC 27710, USA,Department of Medicine, Division of Nephrology, Duke University Medical Center, Durham, NC 27710, USA,Duke Molecular Physiology Institute, Durham, NC, USA
| |
Collapse
|
23
|
Amor DJ, Stephenson SE, Mustapha M, Mensah MA, Ockeloen CW, Lee WS, Tankard RM, Phelan DG, Shinawi M, de Brouwer AP, Pfundt R, Dowling C, Toler TL, Sutton VR, Agolini E, Rinelli M, Capolino R, Martinelli D, Zampino G, Dumić M, Reardon W, Shaw-Smith C, Leventer RJ, Delatycki MB, Kleefstra T, Mundlos S, Mortier G, Bahlo M, Allen NJ, Lockhart PJ. Pathogenic Variants in GPC4 Cause Keipert Syndrome. Am J Hum Genet 2019; 104:914-924. [PMID: 30982611 DOI: 10.1016/j.ajhg.2019.02.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 02/25/2019] [Indexed: 12/22/2022] Open
Abstract
Glypicans are a family of cell-surface heparan sulfate proteoglycans that regulate growth-factor signaling during development and are thought to play a role in the regulation of morphogenesis. Whole-exome sequencing of the Australian family that defined Keipert syndrome (nasodigitoacoustic syndrome) identified a hemizygous truncating variant in the gene encoding glypican 4 (GPC4). This variant, located in the final exon of GPC4, results in premature termination of the protein 51 amino acid residues prior to the stop codon, and in concomitant loss of functionally important N-linked glycosylation (Asn514) and glycosylphosphatidylinositol (GPI) anchor (Ser529) sites. We subsequently identified seven affected males from five additional kindreds with novel and predicted pathogenic variants in GPC4. Segregation analysis and X-inactivation studies in carrier females provided supportive evidence that the GPC4 variants caused the condition. Furthermore, functional studies of recombinant protein suggested that the truncated proteins p.Gln506∗ and p.Glu496∗ were less stable than the wild type. Clinical features of Keipert syndrome included a prominent forehead, a flat midface, hypertelorism, a broad nose, downturned corners of mouth, and digital abnormalities, whereas cognitive impairment and deafness were variable features. Studies of Gpc4 knockout mice showed evidence of the two primary features of Keipert syndrome: craniofacial abnormalities and digital abnormalities. Phylogenetic analysis demonstrated that GPC4 is most closely related to GPC6, which is associated with a bone dysplasia that has a phenotypic overlap with Keipert syndrome. Overall, we have shown that pathogenic variants in GPC4 cause a loss of function that results in Keipert syndrome, making GPC4 the third human glypican to be linked to a genetic syndrome.
Collapse
|
24
|
Balancing the genetic risk of APOL1 kidney disease variants. Nephrol Ther 2019; 15 Suppl 1:S79-S84. [PMID: 30981400 DOI: 10.1016/j.nephro.2019.03.007] [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: 02/05/2019] [Accepted: 03/04/2019] [Indexed: 01/03/2023]
Abstract
African-Americans exhibit an excess risk for chronic and end-stage kidney disease compared to the non-African populations. Two APOL1 genetic variants were shown to account for the majority of this racial disparity in glomerulopathies and other non-diabetic kidney disease. The high-risk genotype has only been reported in populations with recent African ancestry (14 % in African-Americans and up to more than 30 % in West Africa). In less than 10 years, the community has accumulated extensive knowledge on APOL1 and its genetic variants, from their positive selection for resistance against African trypanosomes to potential molecular mechanisms of podocyte injury. Finally, APOL1 associations with kidney transplantation outcomes and with postdonation end-stage kidney disease in living donors have paved the way for a personalized medicine implementation of APOL1 genotyping.
Collapse
|
25
|
Genetic testing in steroid-resistant nephrotic syndrome: why, who, when and how? Pediatr Nephrol 2019; 34:195-210. [PMID: 29181713 PMCID: PMC6311200 DOI: 10.1007/s00467-017-3838-6] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 10/24/2017] [Accepted: 10/25/2017] [Indexed: 12/23/2022]
Abstract
Steroid-resistant nephrotic syndrome (SRNS) is a common cause of chronic kidney disease in childhood and has a significant risk of rapid progression to end-stage renal disease. The identification of over 50 monogenic causes of SRNS has revealed dysfunction in podocyte-associated proteins in the pathogenesis of proteinuria, highlighting their essential role in glomerular function. Recent technological advances in high-throughput sequencing have enabled indication-driven genetic panel testing for patients with SRNS. The availability of genetic testing, combined with the significant phenotypic variability of monogenic SRNS, poses unique challenges for clinicians when directing genetic testing. This highlights the need for clear clinical guidelines that provide a systematic approach for mutational screening in SRNS. The likelihood of identifying a causative mutation is inversely related to age at disease onset and is increased with a positive family history or the presence of extra-renal manifestations. An unequivocal molecular diagnosis could allow for a personalised treatment approach with weaning of immunosuppressive therapy, avoidance of renal biopsy and provision of accurate, well-informed genetic counselling. Identification of novel causative mutations will continue to unravel the pathogenic mechanisms of glomerular disease and provide new insights into podocyte biology and glomerular function.
Collapse
|
26
|
Association of Exposure to Fine-Particulate Air Pollution and Acidic Gases with Incidence of Nephrotic Syndrome. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2018; 15:ijerph15122860. [PMID: 30558173 PMCID: PMC6313436 DOI: 10.3390/ijerph15122860] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 11/29/2018] [Accepted: 12/12/2018] [Indexed: 01/15/2023]
Abstract
Background: Air pollution has been associated with autoimmune diseases. Nephrotic syndrome is a clinical manifestation of immune-mediated glomerulopathy. However, the association between nephrotic syndrome and air pollution constituents remains unknown. We conducted this nationwide retrospective study to investigate the association between PM2.5 and nephrotic syndrome. Methods: We used the Longitudinal Health Insurance Database (LHID) and the Taiwan Air Quality-Monitoring Database (TAQMD). We combined and stratified the LHID and the TAQMD data by residential areas of insurants linked to nearby air quality-monitoring stations. Air pollutant concentrations were grouped into four levels based on quartile. Univariable and multivariable Cox proportional hazard regression models were applied. Findings: Relative to Q1-level SO₂, subjects exposed to the Q4 level were associated with a 2.00-fold higher risk of nephrotic syndrome (adjusted HR = 2.00, 95% CI = 1.66⁻2.41). In NOx, relative to Q1 NOx concentrations, the adjusted HRs of nephrotic syndrome risk were 1.53 (95% CI = 1.23⁻1.91), 1.30 (95% CI = 1.03⁻1.65), and 2.08 (95% CI = 1.69⁻2.56) for Q2, Q3, and Q4 levels, respectively. The results revealed an increasing trend for nephrotic syndrome risk correlating with increasing levels of NO, NO₂, and PM2.5 concentrations. Interpretation: High concentrations of PM2.5, NO, NO₂, and SO₂ are associated with increased risk of nephrotic syndrome.
Collapse
|
27
|
Zhai S, Zhao L, Zhang Y, Ma Q. Interleukin-7 stimulation inhibits nephrin activation and induces podocyte injury. Biochem Biophys Res Commun 2018; 507:100-105. [PMID: 30454893 DOI: 10.1016/j.bbrc.2018.10.173] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 10/28/2018] [Indexed: 12/26/2022]
Abstract
The glomerular podocytes control filtration barrier permeability in the kidney, and their disturbance underlies the pathogenesis of idiopathic nephrotic syndrome (INS), a kidney disease that predominantly occurs in children. In this study, we found that the interleukin-7 receptor (IL-7R) was induced in the glomeruli of adriamycin (ADR)-induced mouse nephropathy, a rodent model of nephrotic syndrome. In addition, IL-7R was also induced by ADR in mouse podocytes cultured in vitro. Functionally, we discovered that IL-7R activation through the stimulation of recombinant IL-7 induced apoptosis of podocytes, and moreover, IL-7 stimulation inhibited nephrin activation and caused actin cytoskeleton disorganization, indicating that IL-7 stimulation induces podocyte injury. Furthermore, IL-7 stimulation impaired the filtration barrier function of podocyte monolayer. Together, these results identify IL-7 and its receptor IL-7R as potential regulators of podocyte function, which might offer a novel therapeutic target in the treatment of INS.
Collapse
Affiliation(s)
- Shubo Zhai
- Department of Pediatric Nephropathy, The First Hospital of Jilin University, China
| | - Lengyue Zhao
- Department of Pediatric Nephropathy, The First Hospital of Jilin University, China
| | - Yan Zhang
- Department of Pediatric Nephropathy, The First Hospital of Jilin University, China
| | - Qingshan Ma
- Department of Pediatric Nephropathy, The First Hospital of Jilin University, China.
| |
Collapse
|
28
|
Kameyama H, Uchimura K, Yamashita T, Kuwabara K, Mizuguchi M, Hung SC, Okuhira K, Masuda T, Kosugi T, Ohgita T, Saito H, Ando Y, Nishitsuji K. The Accumulation of Heparan Sulfate S-Domains in Kidney Transthyretin Deposits Accelerates Fibril Formation and Promotes Cytotoxicity. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 189:308-319. [PMID: 30414409 DOI: 10.1016/j.ajpath.2018.09.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 09/18/2018] [Accepted: 09/26/2018] [Indexed: 10/27/2022]
Abstract
The highly sulfated domains of heparan sulfate (HS), alias HS S-domains, are made up of repeated trisulfated disaccharide units [iduronic acid (2S)-glucosamine (NS, 6S)] and are selectively remodeled by extracellular endoglucosamine 6-sulfatases (Sulfs). Although HS S-domains are critical for signal transduction of several growth factors, their roles in amyloidoses are not yet fully understood. Herein, we found HS S-domains in the kidney of a patient with transthyretin amyloidosis. In in vitro assays with cells stably expressing human Sulfs, heparin, a structural analog of HS S-domains, promoted aggregation of transthyretin in an HS S-domain-dependent manner. Interactions of cells with transthyretin fibrils and cytotoxicity of these fibrils also depended on HS S-domains at the cell surface. Furthermore, glypican-5, encoded by the susceptibility gene for nephrotic syndrome GPC5, was found to be accumulated in the transthyretin amyloidosis kidney. Our study, thus, provides a novel insight into the pathologic roles of HS S-domains in amyloidoses, and we propose that enzymatic remodeling of HS chains by Sulfs may offer an effective approach to inhibiting formation and cytotoxicity of amyloid fibrils.
Collapse
Affiliation(s)
- Hirokazu Kameyama
- Department of Molecular Physical Pharmaceutics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Kenji Uchimura
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan; Unité de Glycobiologie Structurale et Fonctionnelle, UMR 8576 CNRS, Université de Lille 1, Villeneuve d'Ascq, France
| | - Taro Yamashita
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kaori Kuwabara
- Department of Molecular Physical Pharmaceutics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | | | | | - Keiichiro Okuhira
- Department of Molecular Physical Pharmaceutics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Tomohiro Masuda
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomoki Kosugi
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Ohgita
- Department of Biophysical Chemistry, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Hiroyuki Saito
- Department of Biophysical Chemistry, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Yukio Ando
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kazuchika Nishitsuji
- Department of Biochemistry, Wakayama Medical University, Wakayama, Japan; Department of Pathology and Laboratory Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.
| |
Collapse
|
29
|
Gillies CE, Putler R, Menon R, Otto E, Yasutake K, Nair V, Hoover P, Lieb D, Li S, Eddy S, Fermin D, McNulty MT, Hacohen N, Kiryluk K, Kretzler M, Wen X, Sampson MG. An eQTL Landscape of Kidney Tissue in Human Nephrotic Syndrome. Am J Hum Genet 2018; 103:232-244. [PMID: 30057032 PMCID: PMC6081280 DOI: 10.1016/j.ajhg.2018.07.004] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 06/29/2018] [Indexed: 01/14/2023] Open
Abstract
Expression quantitative trait loci (eQTL) studies illuminate the genetics of gene expression and, in disease research, can be particularly illuminating when using the tissues directly impacted by the condition. In nephrology, there is a paucity of eQTL studies of human kidney. Here, we used whole-genome sequencing (WGS) and microdissected glomerular (GLOM) and tubulointerstitial (TI) transcriptomes from 187 individuals with nephrotic syndrome (NS) to describe the eQTL landscape in these functionally distinct kidney structures. Using MatrixEQTL, we performed cis-eQTL analysis on GLOM (n = 136) and TI (n = 166). We used the Bayesian "Deterministic Approximation of Posteriors" (DAP) to fine-map these signals, eQTLBMA to discover GLOM- or TI-specific eQTLs, and single-cell RNA-seq data of control kidney tissue to identify the cell type specificity of significant eQTLs. We integrated eQTL data with an IgA Nephropathy (IgAN) GWAS to perform a transcriptome-wide association study (TWAS). We discovered 894 GLOM eQTLs and 1,767 TI eQTLs at FDR < 0.05. 14% and 19% of GLOM and TI eQTLs, respectively, had >1 independent signal associated with its expression. 12% and 26% of eQTLs were GLOM specific and TI specific, respectively. GLOM eQTLs were most significantly enriched in podocyte transcripts and TI eQTLs in proximal tubules. The IgAN TWAS identified significant GLOM and TI genes, primarily at the HLA region. In this study, we discovered GLOM and TI eQTLs, identified those that were tissue specific, deconvoluted them into cell-specific signals, and used them to characterize known GWAS alleles. These data are available for browsing and download via our eQTL browser, "nephQTL."
Collapse
Affiliation(s)
- Christopher E Gillies
- Department of Pediatrics-Nephrology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Rosemary Putler
- Department of Pediatrics-Nephrology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Rajasree Menon
- Department of Computational Medicine and Bioinformatics, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Edgar Otto
- Department of Medicine-Nephrology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Kalyn Yasutake
- Department of Pediatrics-Nephrology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Viji Nair
- Department of Medicine-Nephrology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Paul Hoover
- Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA; Broad Institute of the Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA 02142, USA
| | - David Lieb
- Broad Institute of the Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA 02142, USA
| | - Shuqiang Li
- Broad Institute of the Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA 02142, USA
| | - Sean Eddy
- Department of Medicine-Nephrology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Damian Fermin
- Department of Pediatrics-Nephrology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Michelle T McNulty
- Department of Pediatrics-Nephrology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Nir Hacohen
- Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA; Broad Institute of the Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA 02142, USA
| | - Krzysztof Kiryluk
- Department of Medicine, Division of Nephrology, College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Matthias Kretzler
- Department of Computational Medicine and Bioinformatics, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA; Department of Medicine-Nephrology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Xiaoquan Wen
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Matthew G Sampson
- Department of Pediatrics-Nephrology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA.
| |
Collapse
|
30
|
Jia X, Horinouchi T, Hitomi Y, Shono A, Khor SS, Omae Y, Kojima K, Kawai Y, Nagasaki M, Kaku Y, Okamoto T, Ohwada Y, Ohta K, Okuda Y, Fujimaru R, Hatae K, Kumagai N, Sawanobori E, Nakazato H, Ohtsuka Y, Nakanishi K, Shima Y, Tanaka R, Ashida A, Kamei K, Ishikura K, Nozu K, Tokunaga K, Iijima K. Strong Association of the HLA-DR/DQ Locus with Childhood Steroid-Sensitive Nephrotic Syndrome in the Japanese Population. J Am Soc Nephrol 2018; 29:2189-2199. [PMID: 30012571 DOI: 10.1681/asn.2017080859] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 05/11/2018] [Indexed: 12/26/2022] Open
Abstract
Background Nephrotic syndrome is the most common cause of chronic glomerular disease in children. Most of these patients develop steroid-sensitive nephrotic syndrome (SSNS), but the loci conferring susceptibility to childhood SSNS are mainly unknown.Methods We conducted a genome-wide association study (GWAS) in the Japanese population; 224 patients with childhood SSNS and 419 adult healthy controls were genotyped using the Affymetrix Japonica Array in the discovery stage. Imputation for six HLA genes (HLA-A, -C, -B, -DRB1, -DQB1, and -DPB1) was conducted on the basis of Japanese-specific references. We performed genotyping for HLA-DRB1/-DQB1 using a sequence-specific oligonucleotide-probing method on a Luminex platform. Whole-genome imputation was conducted using a phased reference panel of 2049 healthy Japanese individuals. Replication was performed in an independent Japanese sample set including 216 patients and 719 healthy controls. We genotyped candidate single-nucleotide polymorphisms using the DigiTag2 assay.Results The most significant association was detected in the HLA-DR/DQ region and replicated (rs4642516 [minor allele G], combined Pallelic=7.84×10-23; odds ratio [OR], 0.33; 95% confidence interval [95% CI], 0.26 to 0.41; rs3134996 [minor allele A], combined Pallelic=1.72×10-25; OR, 0.29; 95% CI, 0.23 to 0.37). HLA-DRB1*08:02 (Pc=1.82×10-9; OR, 2.62; 95% CI, 1.94 to 3.54) and HLA-DQB1*06:04 (Pc=2.09×10-12; OR, 0.10; 95% CI, 0.05 to 0.21) were considered primary HLA alleles associated with childhood SSNS. HLA-DRB1*08:02-DQB1*03:02 (Pc=7.01×10-11; OR, 3.60; 95% CI, 2.46 to 5.29) was identified as the most significant genetic susceptibility factor.Conclusions The most significant association with childhood SSNS was detected in the HLA-DR/DQ region. Further HLA allele/haplotype analyses should enhance our understanding of molecular mechanisms underlying SSNS.
Collapse
Affiliation(s)
- Xiaoyuan Jia
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomoko Horinouchi
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuki Hitomi
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akemi Shono
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Seik-Soon Khor
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yosuke Omae
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kaname Kojima
- Department of Integrative Genomics, Tohoku Medical Megabank Organization.,Graduate School of Medicine, and.,Graduate School of Information Sciences, Tohoku University, Sendai, Japan
| | - Yosuke Kawai
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Integrative Genomics, Tohoku Medical Megabank Organization
| | - Masao Nagasaki
- Department of Integrative Genomics, Tohoku Medical Megabank Organization.,Graduate School of Medicine, and.,Graduate School of Information Sciences, Tohoku University, Sendai, Japan
| | - Yoshitsugu Kaku
- Department of Nephrology, Fukuoka Children's Hospital, Fukuoka, Japan
| | - Takayuki Okamoto
- Department of Pediatrics, Hokkaido University Hospital, Sapporo, Japan
| | - Yoko Ohwada
- Department of Pediatrics, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | - Kazuhide Ohta
- Department of Pediatrics, Kanazawa Medical Center, Kanazawa, Japan
| | - Yusuke Okuda
- Department of Pediatrics, Shiga University of Medical Science, Shiga, Japan
| | - Rika Fujimaru
- Department of Pediatrics, Osaka City General Hospital, Osaka, Japan
| | - Ken Hatae
- Department of Pediatrics, Japanese Red Cross Fukuoka Hospital, Fukuoka, Japan
| | - Naonori Kumagai
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Emi Sawanobori
- Department of Pediatrics, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Hitoshi Nakazato
- Department of Pediatrics, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yasufumi Ohtsuka
- Department of Pediatrics, Faculty of Medicine, Saga University, Saga, Japan
| | - Koichi Nakanishi
- Department of Pediatrics, Wakayama Medical University, Wakayama, Japan
| | - Yuko Shima
- Department of Pediatrics, Wakayama Medical University, Wakayama, Japan
| | - Ryojiro Tanaka
- Department of Nephrology, Hyogo Prefectural Kobe Children's Hospital, Kobe, Japan
| | - Akira Ashida
- Department of Pediatrics, Osaka Medical College, Osaka, Japan; and
| | - Koichi Kamei
- Division of Nephrology and Rheumatology, National Center for Child Health and Development, Tokyo, Japan
| | - Kenji Ishikura
- Division of Nephrology and Rheumatology, National Center for Child Health and Development, Tokyo, Japan
| | - Kandai Nozu
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Katsushi Tokunaga
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan;
| | - Kazumoto Iijima
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan;
| | | |
Collapse
|
31
|
Limou S, Vince N, Parsa A. Lessons from CKD-Related Genetic Association Studies-Moving Forward. Clin J Am Soc Nephrol 2018; 13:140-152. [PMID: 29242368 PMCID: PMC5753320 DOI: 10.2215/cjn.09030817] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Over the past decade, genetic association studies have uncovered numerous determinants of kidney function in the general, diabetic, hypertensive, CKD, ESRD, and GN-based study populations (e.g., IgA nephropathy, membranous nephropathy, FSGS). These studies have led to numerous novel and unanticipated findings, which are helping improve our understanding of factors and pathways affecting both normal and pathologic kidney function. In this review, we report on major discoveries and advances resulting from this rapidly progressing research domain. We also predict some of the next steps the nephrology community should embrace to accelerate the identification of genetic and molecular processes leading to kidney dysfunction, pathophysiologically based disease subgroups, and specific therapeutic targets, as we attempt to transition toward a more precision-based medicine approach.
Collapse
Affiliation(s)
- Sophie Limou
- Centre de Recherche en Transplantation et Immunologie Unité Mixte de Recherche 1064, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Nantes, Nantes, France
- Institut de Transplantation Urologie et Néphrologie, Centre Hospitalier Universitaire Nantes, Nantes, France
- Ecole Centrale de Nantes, Nantes, France
- Basic Science Program, Basic Research Laboratory, National Cancer Institute/National Institutes of Health, Leidos Biomedical Research Inc., Frederick National Laboratory, Frederick, Maryland
| | - Nicolas Vince
- Centre de Recherche en Transplantation et Immunologie Unité Mixte de Recherche 1064, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Nantes, Nantes, France
- Institut de Transplantation Urologie et Néphrologie, Centre Hospitalier Universitaire Nantes, Nantes, France
| | - Afshin Parsa
- Division of Nephrology, University of Maryland School of Medicine, Baltimore, Maryland; and
- Department of Medicine, Baltimore VA Medical Center, Baltimore, Maryland
| |
Collapse
|
32
|
Tabatabaeifar M, Wlodkowski T, Simic I, Denc H, Mollet G, Weber S, Moyers JJ, Brühl B, Randles MJ, Lennon R, Antignac C, Schaefer F. An inducible mouse model of podocin-mutation-related nephrotic syndrome. PLoS One 2017; 12:e0186574. [PMID: 29049388 PMCID: PMC5648285 DOI: 10.1371/journal.pone.0186574] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 09/10/2017] [Indexed: 12/03/2022] Open
Abstract
Mutations in the NPHS2 gene, encoding podocin, cause hereditary nephrotic syndrome. The most common podocin mutation, R138Q, is associated with early disease onset and rapid progression to end-stage renal disease. Knock-in mice carrying a R140Q mutation, the mouse analogue of human R138Q, show developmental arrest of podocytes and lethal renal failure at neonatal age. Here we created a conditional podocin knock-in model named NPHS2R140Q/-, using a tamoxifen-inducible Cre recombinase, which permits to study the effects of the mutation in postnatal life. Within the first week of R140Q hemizygosity induction the animals developed proteinuria, which peaked after 4–5 weeks. Subsequently the animals developed progressive renal failure, with a median survival time of 12 (95% CI: 11–13) weeks. Foot process fusion was observed within one week, progressing to severe and global effacement in the course of the disease. The number of podocytes per glomerulus gradually diminished to 18% compared to healthy controls 12–16 weeks after induction. The fraction of segmentally sclerosed glomeruli was 25%, 85% and 97% at 2, 4 and 8 weeks, respectively. Severe tubulointerstitial fibrosis was present at later disease stage and was correlated quantitatively with the level of proteinuria at early disease stages. While R140Q podocin mRNA expression was elevated, protein abundance was reduced by more than 50% within one week following induction. Whereas miRNA21 expression persistently increased during the first 4 weeks, miRNA-193a expression peaked 2 weeks after induction. In conclusion, the inducible R140Q-podocin mouse model is an auspicious model of the most common genetic cause of human nephrotic syndrome, with a spontaneous disease course strongly reminiscent of the human disorder. This model constitutes a valuable tool to test the efficacy of novel pharmacological interventions aimed to improve podocyte function and viability and attenuate proteinuria, glomerulosclerosis and progressive renal failure.
Collapse
Affiliation(s)
- Mansoureh Tabatabaeifar
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Tanja Wlodkowski
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Ivana Simic
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Helga Denc
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Geraldine Mollet
- INSERM, U1163, Imagine Institute, Laboratory of Hereditary Kidney Diseases, Paris, France
- Paris Descartes-Sorbonne Paris Cité University, Paris, France
| | - Stefanie Weber
- Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, Philipps-University Marburg, Marburg, Germany
| | | | - Barbara Brühl
- Institute for Anatomy and Cell Biology, University of Heidelberg, Heidelberg, Germany
| | - Michael Joseph Randles
- Wellcome Trust Centre for Cell Matrix Research, University of Manchester, Manchester, United Kingdom
- Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Rachel Lennon
- Wellcome Trust Centre for Cell Matrix Research, University of Manchester, Manchester, United Kingdom
- Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Corinne Antignac
- INSERM, U1163, Imagine Institute, Laboratory of Hereditary Kidney Diseases, Paris, France
- Paris Descartes-Sorbonne Paris Cité University, Paris, France
- Department of Genetics, Necker Hospital, Assistance Publique—Hôpitaux de Paris, Paris, France
| | - Franz Schaefer
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
- * E-mail:
| |
Collapse
|
33
|
Fu Y, Zhu JY, Richman A, Zhao Z, Zhang F, Ray PE, Han Z. A Drosophila model system to assess the function of human monogenic podocyte mutations that cause nephrotic syndrome. Hum Mol Genet 2017; 26:768-780. [PMID: 28164240 DOI: 10.1093/hmg/ddw428] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Accepted: 12/16/2016] [Indexed: 12/15/2022] Open
Abstract
Many genetic mutations have been identified as monogenic causes of nephrotic syndrome (NS), but important knowledge gaps exist in the roles of these genes in kidney cell biology and renal diseases. More animal models are needed to assess the functions of these genes in vivo, and to determine how they cause NS in a timely manner. Drosophila nephrocytes and human podocytes share striking similarities, but to what degree these known NS genes play conserved roles in nephrocytes remains unknown. Here we systematically studied 40 genes associated with NS, including 7 that have not previously been analysed for renal function in an animal model. We found that 85% of these genes are required for nephrocyte functions, suggesting that a majority of human genes known to be associated with NS play conserved roles in renal function from flies to humans. To investigate functional conservation in more detail, we focused on Cindr, the fly homolog of the human NS gene CD2AP. Silencing Cindr in nephrocytes led to dramatic nephrocyte functional impairment and shortened life span, as well as collapse of nephrocyte lacunar channels and effacement of nephrocyte slit diaphragms. These phenotypes could be rescued by expression of a wild-type human CD2AP gene, but not a mutant allele derived from a patient with CD2AP-associated NS. We conclude that the Drosophila nephrocyte can be used to elucidate clinically relevant molecular mechanisms underlying the pathogenesis of most monogenic forms of NS, and to efficiently generate personalized in vivo models of genetic renal diseases bearing patient-specific mutations.
Collapse
Affiliation(s)
- Yulong Fu
- Center for Cancer and Immunology Research, Children's National Health Systems, 111 Michigan Ave. NW, Washington, DC, USA
| | - Jun-Yi Zhu
- Center for Cancer and Immunology Research, Children's National Health Systems, 111 Michigan Ave. NW, Washington, DC, USA
| | - Adam Richman
- Center for Cancer and Immunology Research, Children's National Health Systems, 111 Michigan Ave. NW, Washington, DC, USA
| | - Zhanzheng Zhao
- Department of Nephrology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Fujian Zhang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Patricio E Ray
- Center for Genetic Medicine Research, Children's National Health Systems, 111 Michigan Ave. NW, Washington, DC, USA.,Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Zhe Han
- Center for Cancer and Immunology Research, Children's National Health Systems, 111 Michigan Ave. NW, Washington, DC, USA.,Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| |
Collapse
|
34
|
Hirata M, Nagai A, Kamatani Y, Ninomiya T, Tamakoshi A, Yamagata Z, Kubo M, Muto K, Kiyohara Y, Mushiroda T, Murakami Y, Yuji K, Furukawa Y, Zembutsu H, Tanaka T, Ohnishi Y, Nakamura Y, Matsuda K. Overview of BioBank Japan follow-up data in 32 diseases. J Epidemiol 2017; 27:S22-S28. [PMID: 28190660 PMCID: PMC5363789 DOI: 10.1016/j.je.2016.12.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Accepted: 12/15/2016] [Indexed: 01/23/2023] Open
Abstract
Background We established a patient-oriented biobank, BioBank Japan, with information on approximately 200,000 patients, suffering from any of 47 common diseases. This follow-up survey focused on 32 diseases, potentially associated with poor vital prognosis, and collected patient survival information, including cause of death. We performed a survival analysis for all subjects to get an overview of BioBank Japan follow-up data. Methods A total of 141,612 participants were included. The survival data were last updated in 2014. Kaplan–Meier survival analysis was performed after categorizing subjects according to sex, age group, and disease status. Relative survival rates were estimated using a survival-rate table of the Japanese general population. Results Of 141,612 subjects (56.48% male) with 1,087,434 person-years and a 97.0% follow-up rate, 35,482 patients died during follow-up. Mean age at enrollment was 64.24 years for male subjects and 63.98 years for female subjects. The 5-year and 10-year relative survival rates for all subjects were 0.944 and 0.911, respectively, with a median follow-up duration of 8.40 years. Patients with pancreatic cancer had the least favorable prognosis (10-year relative survival: 0.184) and patients with dyslipidemia had the most favorable prognosis (1.013). The most common cause of death was malignant neoplasms. A number of subjects died from diseases other than their registered disease(s). Conclusions This is the first report to perform follow-up survival analysis across various common diseases. Further studies should use detailed clinical and genomic information to identify predictors of mortality in patients with common diseases, contributing to the implementation of personalized medicine. 141,612 participants with any of 32 diseases were included in the follow-up survey. Subject characteristics at enrollment for the follow-up survey were identified. The relative survival analysis showed the worst prognosis in pancreatic cancer. The most common cause of death in all subjects was malignant neoplasms.
Collapse
Affiliation(s)
- Makoto Hirata
- Laboratory of Genome Technology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Akiko Nagai
- Department of Public Policy, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yoichiro Kamatani
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Toshiharu Ninomiya
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akiko Tamakoshi
- Department of Public Health, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Zentaro Yamagata
- Department of Health Sciences, University of Yamanashi, Yamanashi, Japan
| | - Michiaki Kubo
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Kaori Muto
- Department of Public Policy, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yutaka Kiyohara
- Hisayama Research Institute for Lifestyle Diseases, Fukuoka, Japan
| | - Taisei Mushiroda
- Laboratory for Pharmacogenomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yoshinori Murakami
- Division of Molecular Pathology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Koichiro Yuji
- Project Division of International Advanced Medical Research, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yoichi Furukawa
- Division of Clinical Genome Research, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hitoshi Zembutsu
- Laboratory of Molecular Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Division of Genetics, National Cancer Center Research Institute, Tokyo, Japan
| | - Toshihiro Tanaka
- SNP Research Center, RIKEN Yokohama Institute, Yokohama, Japan; Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Bioresource Research Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yozo Ohnishi
- SNP Research Center, RIKEN Yokohama Institute, Yokohama, Japan; Shinko Clinic, Medical Corporation Shinkokai, Tokyo, Japan
| | - Yusuke Nakamura
- Laboratory of Molecular Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, USA
| | | | - Koichi Matsuda
- Laboratory of Molecular Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Laboratory of Clinical Genome Sequencing, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
35
|
Ebana Y, Ozaki K, Liu L, Hachiya H, Hirao K, Isobe M, Kubo M, Tanaka T, Furukawa T. Clinical utility and functional analysis of variants in atrial fibrillation-associated locus 4q25. J Cardiol 2017; 70:366-373. [PMID: 28087289 DOI: 10.1016/j.jjcc.2016.11.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 10/31/2016] [Accepted: 11/16/2016] [Indexed: 01/16/2023]
Abstract
BACKGROUND Chromosome 4q25 has been repeatedly identified as atrial fibrillation (AF)-sensitive locus in multiple genome-wide association studies (GWAS) and is considered to hold some clues to AF pathogenesis. We aimed to investigate the clinical utilities in Japanese and to unveil the function of the 4q25 locus in affecting transcription of adjacent genes. METHODS We conducted AF GWAS in Japanese population (1382 AF cases and 1478 controls) and the replication panel (1666 AF cases and 1229 controls) with detailed clinical information which showed the acceleration of AF onset. Stepwise investigations with linkage disequilibrium analysis, histone code patterns, and reporter assay in the 4q25 locus were performed. RESULTS The AF GWAS confirmed a significant association of rs4611994 and rs1906617 in chromosome 4q25 with AF. In the clinical analysis, AF onset of the individuals with risk allele accelerated 2.5 years compared with those with protective allele (p=0.00012). Next, in the functional analysis, three single nucleotide polymorphisms (SNPs) in the variant group selected by linkage disequilibrium analysis were identified as candidates for the cis-regulatory element toward adjacent genes in chromatin immunoprecipitation assay. Among them, rs4611994 and rs72900144 regions showed higher effects on the transcriptional activity of luciferase gene in the risk alleles than those in the protective alleles (p<0.0001, p<0.005, respectively). CONCLUSIONS AF GWAS in Japanese confirmed the association with 4q25 locus and indicated that its SNP affected the acceleration of AF onset. The candidate regions of the causative SNPs, rs4611994 and rs72900144, could alter the adjacent gene expression level.
Collapse
Affiliation(s)
- Yusuke Ebana
- Department of Bio-informational Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan; Life Science and Bioethics Research Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kouichi Ozaki
- Laboratory for Cardiovascular Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Lian Liu
- Department of Bio-informational Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hitoshi Hachiya
- Department of Cardiovascular Medicine, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan; Heart Rhythm Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kenzo Hirao
- Department of Cardiovascular Medicine, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan; Heart Rhythm Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mitsuaki Isobe
- Department of Cardiovascular Medicine, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Michiaki Kubo
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Toshihiro Tanaka
- Laboratory for Cardiovascular Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Department of Human Genetics and Disease Diversity, Graduate School of Medicine and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tetsushi Furukawa
- Department of Bio-informational Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan.
| |
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW In this review, we take a combined membrane biologist's and geneticist's view of the podocyte, to examine how genetics have informed our understanding of membrane receptors, channels, and other signaling molecules affecting podocyte health and disease. RECENT FINDINGS An integral part of the kidney, the glomerulus, is responsible for the kidney's filter function. Within the glomerulus, the podocyte is a unique cell serving a critically important role: it is exposed to signals from the urinary space in Bowman's capsule, it receives and transmits signals to/from the basement membrane upon which it elaborates, and it receives signals from the vascular space with which it also communicates, thus exposed to toxins, viruses, chemicals, proteins, and cellular components or debris that flow in the blood stream. Our understanding of how podocytes perform their important role has been largely informed by human genetics, and the recent revolution afforded by exome sequencing has brought a tremendous wealth of new genetic data to light. SUMMARY Genetically defined, rare/orphan podocytopathies, as reviewed here, are critically important to study as they may reveal the next generation targets for precision medicine in nephrology.
Collapse
|
37
|
Niu D, Ren Y, Xie L, Sun J, Lu W, Hao Y, Zhang Y, Yin A, Li H, Lv J, Li S. Association between CCDC132, FDX1 and TNFSF13 gene polymorphisms and the risk of IgA nephropathy. Nephrology (Carlton) 2016; 20:908-15. [PMID: 26370181 DOI: 10.1111/nep.12611] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 08/15/2015] [Accepted: 09/15/2015] [Indexed: 01/28/2023]
Abstract
AIM Previous genome-wide association studies have identified multiple susceptibility loci for IgA nephropathy (IgAN); however, validation of these findings is still needed. METHODS We performed a case-control study among 347 Chinese Han IgAN patients and 310 ethnicity-matched controls. Twenty-two single nucleotide polymorphisms (SNPs) were genotyped and association analysis was performed. RESULTS We found three alleles for IgAN in patients: the allele "C" of rs2188404 in the CCDC132 gene by recessive model (odds ratio (OR), 1.65; 95% confidence interval (CI), 1.10-2.48; P = 0.014) and additive model (OR, 1.29; 95% CI, 1.03-1.61; P = 0.024) analysis, respectively, the allele "A" of rs10488764 in FDX1 gene by additive model (OR, 1.27; 95% CI, 1.00-1.61; P = 0.048) analysis, the allele "A" of rs3803800 in TNFSF13 gene by recessive model (OR, 2.05; 95% CI, 1.16-3.62; P = 0.010) and additive model (OR, 1.35; 95% CI, 1.06-1.72; P = 0.013) analysis, respectively. However, the associations between these SNPs and the risk of IgAN were not significant when adjusted for age and sex. Additionally, we found polymorphisms of rs2188404, rs10488764 and rs3803800 were correlated with urine protein (UPRO), human serum albumin (HSA), total cholesterol (TC) and Lee's pathological grades. CONCLUSION We did not find any positive association between these SNPs and the risk of IgAN after adjustment by age and sex, but did find a significant and strong correlation with relevant clinical pathological parameters. Our study may provide a new perspective to understanding the aetiology of IgAN.
Collapse
Affiliation(s)
- Dan Niu
- Department of Nephrology, First Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, China.,Forensic Medicine College, Xi'an Jiaotong University, Xi'an, China
| | - Yongchao Ren
- National Engineering Research Center for Miniaturized Detection Systems, Xi'an, Shaanxi, China.,School of Life Sciences, Northwest University, Xi'an, China
| | - Liyi Xie
- Department of Nephrology, First Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Jiping Sun
- Department of Nephrology, First Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Wanhong Lu
- Department of Nephrology, First Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Yaning Hao
- Department of Nephrology, First Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Yali Zhang
- Department of Nephrology, First Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Aiping Yin
- Department of Nephrology, First Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Huixian Li
- Department of Nephrology, First Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Jia Lv
- Department of Nephrology, First Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Shengbin Li
- Forensic Medicine College, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
38
|
Single Nucleotide Polymorphisms in Pediatric Idiopathic Nephrotic Syndrome. Int J Nephrol 2016; 2016:1417456. [PMID: 27247801 PMCID: PMC4876225 DOI: 10.1155/2016/1417456] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 04/05/2016] [Indexed: 11/24/2022] Open
Abstract
Polymorphic variants in several molecules involved in the glomerular function and drug metabolism have been implicated in the pathophysiology of pediatric idiopathic nephrotic syndrome (INS), but the results remain inconsistent. We analyzed the association of eleven allelic variants in eight genes (angiopoietin-like 4 (ANGPTL4), glypican 5 (GPC5), interleukin-13 (IL-13), macrophage migration inhibitory factor (MIF), neural nitric oxide synthetase (nNOS), multidrug resistance-1 (MDR1), glucocorticoid-induced transcript-1 (GLCCI1), and nuclear receptor subfamily-3 (NR3C1)) in 100 INS patients followed up till adulthood. We genotyped variants using PCR and direct sequencing and evaluated estimated haplotypes of MDR1 variants. The analysis revealed few differences in SNP genotype frequencies between patients and controls, or in clinical parameters among the patients. Genotype distribution of MDR1 SNPs rs1236, rs2677, and rs3435 showed significant (p < 0.05) association with different medication regimes (glucocorticoids only versus glucocorticoids plus additional immunosuppressives). Some marginal association was detected between ANGPTL4, GPC5, GLCCI1, and NR3C1 variants and different medication regimes, number of relapses, and age of onset. Conclusion. While MDR1 variant genotype distribution associated with different medication regimes, the other analyzed gene variants showed only little or marginal clinical relevance in INS.
Collapse
|
39
|
Sampson MG, Pollak MR. Opportunities and Challenges of Genotyping Patients With Nephrotic Syndrome in the Genomic Era. Semin Nephrol 2016. [PMID: 26215859 DOI: 10.1016/j.semnephrol.2015.04.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Both targeted and genome-wide linkage and association studies have identified a number of genes and genetic variants associated with nephrotic syndrome (NS). Genotype-phenotype studies of patients with these variants have identified correlations of clear clinical significance. Combined with improved genomic technologies, this has resulted in increasing, and justifiable, enthusiasm for incorporating our patients' genomic information into our clinical management decisions. Here, we summarize our understanding of NS-associated genetic factors, namely rare causal mutations or common risk alleles in apolipoprotein L1. We discuss the complexities inherent in trying to ascribe risk or causality to these variants, particularly as we seek to extend genetic testing to a broader group of patients, including many with sporadic disease. Overall, the thoughtful application and interpretation of these genetic tests will maximize the benefits to our patients with NS in the form of more precise clinical care.
Collapse
Affiliation(s)
- Matthew G Sampson
- Department of Pediatrics, Division of Nephrology, University of Michigan School of Medicine, Ann Arbor, MI.
| | - Martin R Pollak
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| |
Collapse
|
40
|
Abstract
Podocytes are highly specialized cells of the kidney glomerulus that wrap around capillaries and that neighbor cells of the Bowman’s capsule. When it comes to glomerular filtration, podocytes play an active role in preventing plasma proteins from entering the urinary ultrafiltrate by providing a barrier comprising filtration slits between foot processes, which in aggregate represent a dynamic network of cellular extensions. Foot processes interdigitate with foot processes from adjacent podocytes and form a network of narrow and rather uniform gaps. The fenestrated endothelial cells retain blood cells but permit passage of small solutes and an overlying basement membrane less permeable to macromolecules, in particular to albumin. The cytoskeletal dynamics and structural plasticity of podocytes as well as the signaling between each of these distinct layers are essential for an efficient glomerular filtration and thus for proper renal function. The genetic or acquired impairment of podocytes may lead to foot process effacement (podocyte fusion or retraction), a morphological hallmark of proteinuric renal diseases. Here, we briefly discuss aspects of a contemporary view of podocytes in glomerular filtration, the patterns of structural changes in podocytes associated with common glomerular diseases, and the current state of basic and clinical research.
Collapse
Affiliation(s)
- Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Mehmet M Altintas
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
41
|
Verrier ER, Colpitts CC, Bach C, Heydmann L, Weiss A, Renaud M, Durand SC, Habersetzer F, Durantel D, Abou-Jaoudé G, López Ledesma MM, Felmlee DJ, Soumillon M, Croonenborghs T, Pochet N, Nassal M, Schuster C, Brino L, Sureau C, Zeisel MB, Baumert TF. A targeted functional RNA interference screen uncovers glypican 5 as an entry factor for hepatitis B and D viruses. Hepatology 2016. [PMID: 26224662 DOI: 10.1002/hep.28013] [Citation(s) in RCA: 258] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
UNLABELLED Chronic hepatitis B and D infections are major causes of liver disease and hepatocellular carcinoma worldwide. Efficient therapeutic approaches for cure are absent. Sharing the same envelope proteins, hepatitis B virus and hepatitis delta virus use the sodium/taurocholate cotransporting polypeptide (a bile acid transporter) as a receptor to enter hepatocytes. However, the detailed mechanisms of the viral entry process are still poorly understood. Here, we established a high-throughput infectious cell culture model enabling functional genomics of hepatitis delta virus entry and infection. Using a targeted RNA interference entry screen, we identified glypican 5 as a common host cell entry factor for hepatitis B and delta viruses. CONCLUSION These findings advance our understanding of virus cell entry and open new avenues for curative therapies. As glypicans have been shown to play a role in the control of cell division and growth regulation, virus-glypican 5 interactions may also play a role in the pathogenesis of virus-induced liver disease and cancer.
Collapse
Affiliation(s)
- Eloi R Verrier
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France.,Université de Strasbourg, Strasbourg, France
| | - Che C Colpitts
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France.,Université de Strasbourg, Strasbourg, France
| | - Charlotte Bach
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France.,Université de Strasbourg, Strasbourg, France
| | - Laura Heydmann
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France.,Université de Strasbourg, Strasbourg, France
| | - Amélie Weiss
- IGBMC, Plateforme de Criblage Haut-débit, Illkirch, France
| | - Mickaël Renaud
- IGBMC, Plateforme de Criblage Haut-débit, Illkirch, France
| | - Sarah C Durand
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France.,Université de Strasbourg, Strasbourg, France
| | - François Habersetzer
- Institut Hospitalo-Universitaire, Pôle Hépato-digestif, Nouvel Hôpital Civil, Strasbourg, France
| | - David Durantel
- Inserm, U1052, CNRS UMR 5286, Cancer Research Center of Lyon, Université de Lyon, Lyon, France
| | | | - Maria M López Ledesma
- Cátedra de Virología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Daniel J Felmlee
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France.,Université de Strasbourg, Strasbourg, France
| | - Magali Soumillon
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
| | - Tom Croonenborghs
- Program in Translational NeuroPsychiatric Genomics, Brigham and Women's Hospital, Harvard Medical School, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA.,KU Leuven Technology Campus Geel, AdvISe, Geel, Belgium
| | - Nathalie Pochet
- Program in Translational NeuroPsychiatric Genomics, Brigham and Women's Hospital, Harvard Medical School, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
| | - Michael Nassal
- Department of Internal Medicine II/Molecular Biology, University Hospital Freiburg, Freiburg, Germany
| | - Catherine Schuster
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France.,Université de Strasbourg, Strasbourg, France
| | - Laurent Brino
- IGBMC, Plateforme de Criblage Haut-débit, Illkirch, France
| | - Camille Sureau
- INTS, Laboratoire de Virologie Moléculaire, Paris, France
| | - Mirjam B Zeisel
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France.,Université de Strasbourg, Strasbourg, France
| | - Thomas F Baumert
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,Institut Hospitalo-Universitaire, Pôle Hépato-digestif, Nouvel Hôpital Civil, Strasbourg, France
| |
Collapse
|
42
|
Sampson MG. Actualizing the Benefits of Genomic Discovery in Pediatric Nephrology. J Pediatr Genet 2015; 5:69-75. [PMID: 27617144 DOI: 10.1055/s-0035-1557113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 01/19/2015] [Indexed: 01/13/2023]
Abstract
The discovery of genetic variation associated with pediatric kidney disease has shed light on the biology underlying these conditions and, in some cases, has improved our clinical management of patients. We are challenged to continue the momentum of the genomic era in pediatric nephrology by identifying novel disease-associated genetic variation and translating these discoveries into clinical applications. This article reviews the diverse forms of genetic architecture that have been found to be associated with kidney diseases and traits. These include rare, fully penetrant variants responsible for Mendelian forms of disease, copy number variants, and more common variants associated with increased risk of disease. These discoveries have provided us with a greater understanding of the molecular mechanisms underlying these conditions and highlighted key pathways for potential intervention. In a number of areas, the identification of rare, fully penetrant variants is immediately clinically relevant, whether in regard to diagnostic testing, prediction of outcomes, or choice of therapies and interventions. This article discusses limitations in the deterministic view of rare, putatively causal mutations, a challenge increasing in importance as sequencing expands to many more genes and patients. This article also focusses on common genetic variants, using those found to be associated with focal segmental glomerulosclerosis in African-Americans, IgA nephropathy, chronic kidney disease (CKD), and estimated glomerular filtration rate (eGFR) as examples. Identifying common genetic variants associated with disease will complement other areas of genomic inquiry, lead to a greater biological understanding of disease, and will benefit pediatric nephrology patients.
Collapse
Affiliation(s)
- Matthew G Sampson
- Division of Pediatric Nephrology, Department of Pediatrics and Communicable Diseases, University of Michigan School of Medicine, Ann Arbor, Michigan, United States
| |
Collapse
|
43
|
Rheault MN, Gbadegesin RA. The Genetics of Nephrotic Syndrome. J Pediatr Genet 2015; 5:15-24. [PMID: 27617138 DOI: 10.1055/s-0035-1557109] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 01/21/2015] [Indexed: 12/26/2022]
Abstract
Nephrotic syndrome (NS) is a common pediatric kidney disease and is defined as massive proteinuria, hypoalbuminemia, and edema. Dysfunction of the glomerular filtration barrier, which is made up of endothelial cells, glomerular basement membrane, and visceral epithelial cells known as podocytes, is evident in children with NS. While most children have steroid-responsive nephrotic syndrome (SSNS), approximately 20% have steroid-resistant nephrotic syndrome (SRNS) and are at risk for progressive kidney dysfunction. While the cause of SSNS is still not well understood, there has been an explosion of research into the genetic causes of SRNS in the past 15 years. More than 30 proteins regulating the function of the glomerular filtration barrier have been associated with SRNS including podocyte slit diaphragm proteins, podocyte actin cytoskeletal proteins, mitochondrial proteins, adhesion and glomerular basement membrane proteins, transcription factors, and others. A genetic cause of SRNS can be found in approximately 70% of infants presenting in the first 3 months of life and 50% of infants presenting between 4 and 12 months, with much lower likelihood for older patients. Identification of the underlying genetic etiology of SRNS is important in children because it allows for counseling of other family members who may be at risk, predicts risk of recurrent disease after kidney transplant, and predicts response to immunosuppressive therapy. Correlations between genetic mutation and clinical phenotype as well as genetic risk factors for SSNS and SRNS are reviewed in this article.
Collapse
Affiliation(s)
- Michelle N Rheault
- Division of Nephrology, University of Minnesota Masonic Children's Hospital, Minneapolis, Minnesota, United States
| | - Rasheed A Gbadegesin
- Division of Nephrology and Center for Human Genetics, Duke University Medical Center, Durham, North Carolina, United States
| |
Collapse
|
44
|
Glypican-5 Increases Susceptibility to Nephrotic Damage in Diabetic Kidney. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:1889-98. [PMID: 25987249 DOI: 10.1016/j.ajpath.2015.03.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 02/21/2015] [Accepted: 03/09/2015] [Indexed: 11/22/2022]
Abstract
Type 2 diabetes mellitus is a leading health issue worldwide. Among cases of diabetes mellitus nephropathy (DN), the major complication of type 2 diabetes mellitus, the nephrotic phenotype is often intractable to clinical intervention and demonstrates the rapid decline of renal function to end-stage renal disease. We recently identified the gene for glypican-5 (GPC5), a cell-surface heparan sulfate proteoglycan, as conferring susceptibility for acquired nephrotic syndrome and additionally identified an association through a genome-wide association study between a variant in GPC5 and DN of type 2 diabetes mellitus. In vivo and in vitro data showed a progressive increase of GPC5 in type 2 DN along with severity; the excess was derived from glomerular mesangial cells. In this study, diabetic kidney showed that accumulation of fibroblast growth factor (Fgf)2 strikingly induced progressive proteinuria that was avoided in Gpc5 knockdown mice. The efficacy of Gpc5 inhibition was exerted through expression of the Fgf receptors 3 and 4 provoked in the diabetic kidney attributively. Extraglomerular Fgf2 was pathogenic in DN, and the deterrence of Gpc5 effectively inhibited the glomerular accumulation of Fgf2, the subsequent increase of mesangial extracellular matrix, and the podocytes' small GTPase activity. These findings elucidate the pivotal role of GPC5, identified as a susceptible gene in the genome-wide association study, in hyperglycemia-induced glomerulopathy.
Collapse
|
45
|
Theocharis AD, Skandalis SS, Neill T, Multhaupt HAB, Hubo M, Frey H, Gopal S, Gomes A, Afratis N, Lim HC, Couchman JR, Filmus J, Sanderson RD, Schaefer L, Iozzo RV, Karamanos NK. Insights into the key roles of proteoglycans in breast cancer biology and translational medicine. Biochim Biophys Acta Rev Cancer 2015; 1855:276-300. [PMID: 25829250 DOI: 10.1016/j.bbcan.2015.03.006] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 02/27/2015] [Accepted: 03/24/2015] [Indexed: 12/18/2022]
Abstract
Proteoglycans control numerous normal and pathological processes, among which are morphogenesis, tissue repair, inflammation, vascularization and cancer metastasis. During tumor development and growth, proteoglycan expression is markedly modified in the tumor microenvironment. Altered expression of proteoglycans on tumor and stromal cell membranes affects cancer cell signaling, growth and survival, cell adhesion, migration and angiogenesis. Despite the high complexity and heterogeneity of breast cancer, the rapid evolution in our knowledge that proteoglycans are among the key players in the breast tumor microenvironment suggests their potential as pharmacological targets in this type of cancer. It has been recently suggested that pharmacological treatment may target proteoglycan metabolism, their utilization as targets for immunotherapy or their direct use as therapeutic agents. The diversity inherent in the proteoglycans that will be presented herein provides the potential for multiple layers of regulation of breast tumor behavior. This review summarizes recent developments concerning the biology of selected proteoglycans in breast cancer, and presents potential targeted therapeutic approaches based on their novel key roles in breast cancer.
Collapse
Affiliation(s)
- Achilleas D Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece
| | - Spyros S Skandalis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece
| | - Thomas Neill
- Department of Pathology, Anatomy and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Hinke A B Multhaupt
- Department of Biomedical Sciences and Biotech Research & Innovation Center, University of Copenhagen, Denmark
| | - Mario Hubo
- University of Frankfurt, Institute of Pharmacology and Toxicology, Theodor-Stern Kai 7, Frankfurt 60590, Germany
| | - Helena Frey
- University of Frankfurt, Institute of Pharmacology and Toxicology, Theodor-Stern Kai 7, Frankfurt 60590, Germany
| | - Sandeep Gopal
- Department of Biomedical Sciences and Biotech Research & Innovation Center, University of Copenhagen, Denmark
| | - Angélica Gomes
- Department of Biomedical Sciences and Biotech Research & Innovation Center, University of Copenhagen, Denmark
| | - Nikos Afratis
- Department of Biomedical Sciences and Biotech Research & Innovation Center, University of Copenhagen, Denmark
| | - Hooi Ching Lim
- Department of Biomedical Sciences and Biotech Research & Innovation Center, University of Copenhagen, Denmark
| | - John R Couchman
- Department of Biomedical Sciences and Biotech Research & Innovation Center, University of Copenhagen, Denmark
| | - Jorge Filmus
- Department of Biological Sciences, Sunnybrook Research Institute and Department of Medical Biophysics, University of Toronto, Canada
| | - Ralph D Sanderson
- University of Alabama at Birmingham, Department of Pathology, UAB Comprehensive Cancer Center, 1720 2nd Ave. S, WTI 602B, Birmingham, AL 35294, USA
| | - Liliana Schaefer
- University of Frankfurt, Institute of Pharmacology and Toxicology, Theodor-Stern Kai 7, Frankfurt 60590, Germany
| | - Renato V Iozzo
- Department of Pathology, Anatomy and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece.
| |
Collapse
|
46
|
Sampson MG, Hodgin JB, Kretzler M. Defining nephrotic syndrome from an integrative genomics perspective. Pediatr Nephrol 2015; 30:51-63; quiz 59. [PMID: 24890338 PMCID: PMC4241380 DOI: 10.1007/s00467-014-2857-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 05/06/2014] [Accepted: 05/14/2014] [Indexed: 12/15/2022]
Abstract
Nephrotic syndrome (NS) is a clinical condition with a high degree of morbidity and mortality, caused by failure of the glomerular filtration barrier, resulting in massive proteinuria. Our current diagnostic, prognostic and therapeutic decisions in NS are largely based upon clinical or histological patterns such as "focal segmental glomerulosclerosis" or "steroid sensitive". Yet these descriptive classifications lack the precision to explain the physiologic origins and clinical heterogeneity observed in this syndrome. A more precise definition of NS is required to identify mechanisms of disease and capture various clinical trajectories. An integrative genomics approach to NS applies bioinformatics and computational methods to comprehensive experimental, molecular and clinical data for holistic disease definition. A unique aspect is analysis of data together to discover NS-associated molecules, pathways, and networks. Integrating multidimensional datasets from the outset highlights how molecular lesions impact the entire individual. Data sets integrated range from genetic variation to gene expression, to histologic changes, to progression of chronic kidney disease (CKD). This review will introduce the tenets of integrative genomics and suggest how it can increase our understanding of NS from molecular and pathophysiological perspectives. A diverse group of genome-scale experiments are presented that have sought to define molecular signatures of NS. Finally, the Nephrotic Syndrome Study Network (NEPTUNE) will be introduced as an international, prospective cohort study of patients with NS that utilizes an integrated systems genomics approach from the outset. A major NEPTUNE goal is to achieve comprehensive disease definition from a genomics perspective and identify shared molecular drivers of disease.
Collapse
Affiliation(s)
- Matthew G. Sampson
- Division of Nephrology, Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA,to whom correspondence should be addressed: Matthew Sampson, Division of Nephrology, University of Michigan School of Medicine, 8220D MSRB III, West Medical Center Drive, Ann Arbor, MI 48109, kidneyomics.org, , Telephone and Fax: 734-647-9361. Matthias Kretzler, Medicine/Nephrology and Computational Medicine and Bioinformatics, University of Michigan, 1560 MSRB II, 1150 W. Medical Center Dr.-SPC5676, Ann Arbor, MI 48109-5676, 734-615-5757, fax: 734-763-0982,
| | - Jeffrey B. Hodgin
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Matthias Kretzler
- Division of Nephrology, Department of Internal Medicine and Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA,to whom correspondence should be addressed: Matthew Sampson, Division of Nephrology, University of Michigan School of Medicine, 8220D MSRB III, West Medical Center Drive, Ann Arbor, MI 48109, kidneyomics.org, , Telephone and Fax: 734-647-9361. Matthias Kretzler, Medicine/Nephrology and Computational Medicine and Bioinformatics, University of Michigan, 1560 MSRB II, 1150 W. Medical Center Dr.-SPC5676, Ann Arbor, MI 48109-5676, 734-615-5757, fax: 734-763-0982,
| |
Collapse
|
47
|
Sabourin J, Nobel AB, Valdar W. Fine-mapping additive and dominant SNP effects using group-LASSO and fractional resample model averaging. Genet Epidemiol 2014; 39:77-88. [PMID: 25417853 DOI: 10.1002/gepi.21869] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 09/25/2014] [Accepted: 09/30/2014] [Indexed: 12/28/2022]
Abstract
Genomewide association studies (GWAS) sometimes identify loci at which both the number and identities of the underlying causal variants are ambiguous. In such cases, statistical methods that model effects of multiple single-nucleotide polymorphisms (SNPs) simultaneously can help disentangle the observed patterns of association and provide information about how those SNPs could be prioritized for follow-up studies. Current multi-SNP methods, however, tend to assume that SNP effects are well captured by additive genetics; yet when genetic dominance is present, this assumption translates to reduced power and faulty prioritizations. We describe a statistical procedure for prioritizing SNPs at GWAS loci that efficiently models both additive and dominance effects. Our method, LLARRMA-dawg, combines a group LASSO procedure for sparse modeling of multiple SNP effects with a resampling procedure based on fractional observation weights. It estimates for each SNP the robustness of association with the phenotype both to sampling variation and to competing explanations from other SNPs. In producing an SNP prioritization that best identifies underlying true signals, we show the following: our method easily outperforms a single-marker analysis; when additive-only signals are present, our joint model for additive and dominance is equivalent to or only slightly less powerful than modeling additive-only effects; and when dominance signals are present, even in combination with substantial additive effects, our joint model is unequivocally more powerful than a model assuming additivity. We also describe how performance can be improved through calibrated randomized penalization, and discuss how dominance in ungenotyped SNPs can be incorporated through either heterozygote dosage or multiple imputation.
Collapse
Affiliation(s)
- Jeremy Sabourin
- Department of Genetics, University of North Carolina at Chapel Hill, North Carolina, United States of America; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, North Carolina, United States of America
| | | | | |
Collapse
|
48
|
Gbadegesin RA, Adeyemo A, Webb NJA, Greenbaum LA, Abeyagunawardena A, Thalgahagoda S, Kale A, Gipson D, Srivastava T, Lin JJ, Chand D, Hunley TE, Brophy PD, Bagga A, Sinha A, Rheault MN, Ghali J, Nicholls K, Abraham E, Janjua HS, Omoloja A, Barletta GM, Cai Y, Milford DD, O'Brien C, Awan A, Belostotsky V, Smoyer WE, Homstad A, Hall G, Wu G, Nagaraj S, Wigfall D, Foreman J, Winn MP. HLA-DQA1 and PLCG2 Are Candidate Risk Loci for Childhood-Onset Steroid-Sensitive Nephrotic Syndrome. J Am Soc Nephrol 2014; 26:1701-10. [PMID: 25349203 DOI: 10.1681/asn.2014030247] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 09/09/2014] [Indexed: 12/11/2022] Open
Abstract
Steroid-sensitive nephrotic syndrome (SSNS) accounts for >80% of cases of nephrotic syndrome in childhood. However, the etiology and pathogenesis of SSNS remain obscure. Hypothesizing that coding variation may underlie SSNS risk, we conducted an exome array association study of SSNS. We enrolled a discovery set of 363 persons (214 South Asian children with SSNS and 149 controls) and genotyped them using the Illumina HumanExome Beadchip. Four common single nucleotide polymorphisms (SNPs) in HLA-DQA1 and HLA-DQB1 (rs1129740, rs9273349, rs1071630, and rs1140343) were significantly associated with SSNS at or near the Bonferroni-adjusted P value for the number of single variants that were tested (odds ratio, 2.11; 95% confidence interval, 1.56 to 2.86; P=1.68×10(-6) (Fisher exact test). Two of these SNPs-the missense variants C34Y (rs1129740) and F41S (rs1071630) in HLA-DQA1-were replicated in an independent cohort of children of white European ancestry with SSNS (100 cases and ≤589 controls; P=1.42×10(-17)). In the rare variant gene set-based analysis, the best signal was found in PLCG2 (P=7.825×10(-5)). In conclusion, this exome array study identified HLA-DQA1 and PLCG2 missense coding variants as candidate loci for SSNS. The finding of a MHC class II locus underlying SSNS risk suggests a major role for immune response in the pathogenesis of SSNS.
Collapse
Affiliation(s)
- Rasheed A Gbadegesin
- Department of Pediatrics, Division of Nephrology and Center for Human Genetics, Duke University Medical Center, Durham, North Carolina
| | - Adebowale Adeyemo
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland;
| | - Nicholas J A Webb
- Department of Pediatric Nephrology and NIHR/Wellcome Trust Children's Clinical Research Facility, The University of Manchester, Manchester Academic Health Science Centre, Royal Manchester Children's Hospital, Manchester, United Kingdom
| | - Larry A Greenbaum
- Division of Pediatric Nephrology, Emory University School of Medicine and Children's Healthcare of Atlanta, Georgia
| | | | | | - Arundhati Kale
- Division of Nephrology, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas
| | - Debbie Gipson
- Department of Pediatrics, Division of Nephrology, University of Michigan, Ann Arbor, Michigan
| | - Tarak Srivastava
- Division of Nephrology, Children's Mercy Hospital, Kansas City, Missouri
| | - Jen-Jar Lin
- Department of Pediatrics, Division of Nephrology, Wake Forest University Baptist Medical Center, Winston-Salem, North Carolina
| | - Deepa Chand
- Department of Pediatrics, Division of Nephrology, Rush University, Chicago, Illinois
| | - Tracy E Hunley
- Department of Pediatrics, Division of Nephrology, Vanderbilt University, Nashville, Tennessee
| | - Patrick D Brophy
- Department of Pediatrics, Division of Nephrology, University of Iowa, Iowa City, Iowa
| | - Arvind Bagga
- Department of Pediatrics, Division of Nephrology, All India Institute of Medical Science, Ansari Nagar, New Delhi, India
| | - Aditi Sinha
- Department of Pediatrics, Division of Nephrology, All India Institute of Medical Science, Ansari Nagar, New Delhi, India
| | - Michelle N Rheault
- Department of Pediatrics, Division of Nephrology, University of Minnesota Amplatz Children's Hospital, Minneapolis, Minnesota
| | - Joanna Ghali
- Department of Nephrology, Royal Melbourne Hospital, Parkville, Australia
| | - Kathy Nicholls
- Department of Nephrology, Royal Melbourne Hospital, Parkville, Australia
| | - Elizabeth Abraham
- Department of Pediatrics, Division of Nephrology, St. Louis University, St. Louis, Missouri
| | - Halima S Janjua
- Pediatric Institute, Center for Pediatric Nephrology, Cleveland Clinic, Cleveland, Ohio
| | - Abiodun Omoloja
- Division of Nephrology, Dayton Children's Hospital, Dayton, Ohio
| | | | - Yi Cai
- Division of Nephrology, Helen Devos Children's Hospital, Grand Rapids, Michigan
| | | | | | - Atif Awan
- Division of Nephrology, The Children's University Hospital, Dublin, Ireland
| | - Vladimir Belostotsky
- Department of Pediatrics, Division of Nephrology, Leeds Teaching Hospital, Leeds, United Kingdom
| | - William E Smoyer
- Center for Clinical and Translational Research, Research Institute at Nationwide Children's Hospital, Columbus, Ohio; and
| | - Alison Homstad
- Department of Pediatrics, Division of Nephrology and Center for Human Genetics, Duke University Medical Center, Durham, North Carolina
| | - Gentzon Hall
- Department of Medicine, Division of Nephrology and Center for Human Genetics, Duke University Medical Center, Durham, North Carolina
| | - Guanghong Wu
- Department of Medicine, Division of Nephrology and Center for Human Genetics, Duke University Medical Center, Durham, North Carolina
| | - Shashi Nagaraj
- Department of Pediatrics, Division of Nephrology and Center for Human Genetics, Duke University Medical Center, Durham, North Carolina
| | - Delbert Wigfall
- Department of Pediatrics, Division of Nephrology and Center for Human Genetics, Duke University Medical Center, Durham, North Carolina
| | - John Foreman
- Department of Pediatrics, Division of Nephrology and Center for Human Genetics, Duke University Medical Center, Durham, North Carolina
| | - Michelle P Winn
- Department of Medicine, Division of Nephrology and Center for Human Genetics, Duke University Medical Center, Durham, North Carolina
| | | |
Collapse
|
49
|
Zimmer V, Lammert F. Role of genetics in diagnosis and therapy of acquired liver disease. Mol Aspects Med 2014; 37:15-34. [DOI: 10.1016/j.mam.2013.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 10/07/2013] [Accepted: 10/15/2013] [Indexed: 02/08/2023]
|
50
|
Lessons from Genome-Wide Search for Disease-Related Genes with Special Reference to HLA-Disease Associations. Genes (Basel) 2014; 5:84-96. [PMID: 24705288 PMCID: PMC3978513 DOI: 10.3390/genes5010084] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 02/11/2014] [Accepted: 02/12/2014] [Indexed: 01/11/2023] Open
Abstract
The relationships between diseases and genetic factors are by no means uniform. Single-gene diseases are caused primarily by rare mutations of specific genes. Although each single-gene disease has a low prevalence, there are an estimated 5000 or more such diseases in the world. In contrast, multifactorial diseases are diseases in which both genetic and environmental factors are involved in onset. These include a variety of diseases, such as diabetes and autoimmune diseases, and onset is caused by a range of various environmental factors together with a number of genetic factors. With the astonishing advances in genome analysis technology in recent years and the accumulation of data on human genome variation, there has been a rapid progress in research involving genome-wide searches for genes related to diseases. Many of these studies have led to the recognition of the importance of the human leucocyte antigen (HLA) gene complex. Here, the current state and future challenges of genome-wide exploratory research into variations that are associated with disease susceptibilities and drug/therapy responses are described, mainly with reference to our own experience in this field.
Collapse
|