1
|
Yuan Z, Lv G, Liu X, Xiao Y, Tan Y, Zhu Y. Machine learning selection of basement membrane-associated genes and development of a predictive model for kidney fibrosis. Sci Rep 2025; 15:6567. [PMID: 39994219 PMCID: PMC11850825 DOI: 10.1038/s41598-025-89733-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 02/07/2025] [Indexed: 02/26/2025] Open
Abstract
This study investigates the role of basement membrane-related genes in kidney fibrosis, a significant factor in the progression of chronic kidney disease that can lead to end-stage renal failure. The authors aim to develop a predictive model using machine learning techniques due to the limitations of existing diagnostic methods, which often lack sensitivity and specificity. Utilizing gene expression data from the GEO database, the researchers applied LASSO, Random Forest, and SVM-RFE methods to identify five pivotal genes: ARID4B, EOMES, KCNJ3, LIF, and STAT1. These genes were analyzed across training and validation datasets, resulting in the development of a Nomogram prediction model. Performance metrics, including the area under the ROC curve (AUC), calibration curves, and decision curve analysis, indicated excellent predictive capabilities with an AUC of 0.923. Experimental validation through qRT-PCR in clinical samples and TGF-β-treated HK-2 cells corroborated the expression patterns identified in silico, showing upregulation of ARID4B, EOMES, LIF, and STAT1, and downregulation of KCNJ3. The findings emphasize the importance of basement membrane-related genes in kidney fibrosis and pave the way for enhanced early diagnosis and targeted therapeutic strategies.
Collapse
Affiliation(s)
- Ziwei Yuan
- Department of Laboratory Medicine, The Third People's Hospital of Ganzhou, 341000, Ganzhou, China
| | - Guangjia Lv
- College of Life Sciences, Northeast Forestry University, Harbin, 150004, China
| | - Xinyan Liu
- Zhanggong District Maternal and Child Health Hospital, Ganzhou, 341000, China.
| | - Yanyi Xiao
- Department of Thyroid and Breast Surgery, Wenzhou Central Hospital, Wenzhou, 325000, China.
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Yuanfang Tan
- Department of Laboratory Medicine, The Third People's Hospital of Ganzhou, 341000, Ganzhou, China.
| | - Youyou Zhu
- Department of pathology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, 317000, China.
| |
Collapse
|
2
|
Xing J, Huang L, Ren W, Mei X. Risk factors for rapid kidney function decline in diabetes patients. Ren Fail 2024; 46:2398188. [PMID: 39258389 PMCID: PMC11391878 DOI: 10.1080/0886022x.2024.2398188] [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: 06/10/2024] [Revised: 08/15/2024] [Accepted: 08/23/2024] [Indexed: 09/12/2024] Open
Abstract
Diabetic nephropathy, as a severe microvascular complication of diabetes, manifests in four clinical types: classic, albuminuria regression, a rapid decline in kidney function (RDKF), and non-proteinuric or non-albuminuric DKD. Rapidly progressive diabetic nephropathy advances to end-stage renal disease more swiftly than the typical form, posing significant risks. However, a comprehensive understanding of rapidly progressive diabetic nephropathy is currently lacking. This article reviewed latest developments in genetic and clinical risk factors associated with rapidly progressive diabetic nephropathy, aiming to broad perspectives concerning the diagnosis and interventions of this condition.
Collapse
Affiliation(s)
- Jixin Xing
- Department of Nephrology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Linxi Huang
- Department of Nephrology, PLA Navy No. 905 Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Weifu Ren
- Department of Nephrology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Xiaobin Mei
- Department of Nephrology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| |
Collapse
|
3
|
Zheng Q, Gu X, He JC, Xie J. Progress in therapeutic targets on podocyte for Alport syndrome. J Transl Int Med 2024; 12:129-133. [PMID: 38812923 PMCID: PMC11135632 DOI: 10.2478/jtim-2024-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024] Open
Affiliation(s)
- Qimin Zheng
- Department of Nephrology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
- Institute of Nephrology, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Xiangchen Gu
- Department of Nephrology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
- Institute of Nephrology, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - John Cijiang He
- Department of Medicine, Barbara T. Murphy Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Jingyuan Xie
- Department of Nephrology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
- Institute of Nephrology, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Gao X, Li M, Wang K, Li Z, Han C. Pregnancy in women with autosomal recessive Alport syndrome caused by novel compound heterozygous mutations of COL4A3 gene: Two cases reports. Medicine (Baltimore) 2023; 102:e36057. [PMID: 37986374 PMCID: PMC10659596 DOI: 10.1097/md.0000000000036057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 10/20/2023] [Indexed: 11/22/2023] Open
Abstract
RATIONALE Autosomal recessive Alport syndrome (ARAS) is an hereditary heterogeneous disease that poses a serious risk to pregnant women. PATIENT CONCERNS We reported 2 cases of pregnancy with progressive proteinuria. The case 1 was a 21-year-old woman with 24-h proteinuria increased from 2.03 to 11.72 g at 13 to 35 weeks of gestation, and the case 2 was a 28-year-old woman with 24-h proteinuria increased from 2.10 to 9.32 g at 8 to 36 weeks of gestation. In advanced stage of pregnancy, the fetal development was smaller than the gestational age. DIAGNOSES Sanger sequencing showed that novel compound heterozygous mutations [c.1315 G>T (p.G439C) and c.4847 G>A (p.C1616Y)] of the collagen type IV alpha 3 chain (COL4A3) gene were found in the 2 cases. Renal puncture pathology confirmed the diagnosis of ARAS. INTERVENTIONS The 2 cases were treated with albumin, compounded amino acids, calcium, vitamin D, and low molecular weight heparin in addition to conventional treatment during pregnancy. Pregnancy was terminated by cesarean section at 36 to 37 weeks of gestation. After delivery, the patients were treated with Losartan for anti-proteinuric therapy for 1 year. OUTCOMES The neonatal weights and Apgar scores were normal. The patients recovered well and 24-h proteinuria decreased to pre-pregnancy level. LESSONS When pregnant women present with a persistent increasing proteinuria, ARAS needs to be considered. Sanger sequencing is useful to assist in the diagnosis of ARAS. Multidisciplinary treatments from nephrologists and gynecologists are needed to ensure the safety of pregnancy and the fetus.
Collapse
Affiliation(s)
- Xiaoli Gao
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Meilu Li
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Kan Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Zengyan Li
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Cha Han
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
5
|
Downie ML, Desjarlais A, Verdin N, Woodlock T, Collister D. Precision Medicine in Diabetic Kidney Disease: A Narrative Review Framed by Lived Experience. Can J Kidney Health Dis 2023; 10:20543581231209012. [PMID: 37920777 PMCID: PMC10619345 DOI: 10.1177/20543581231209012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 09/10/2023] [Indexed: 11/04/2023] Open
Abstract
Purpose of review Diabetic kidney disease (DKD) is a leading cause of chronic kidney disease (CKD) for which many treatments exist that have been shown to prevent CKD progression and kidney failure. However, DKD is a complex and heterogeneous etiology of CKD with a spectrum of phenotypes and disease trajectories. In this narrative review, we discuss precision medicine approaches to DKD, including genomics, metabolomics, proteomics, and their potential role in the management of diabetes mellitus and DKD. A patient and caregivers of patients with lived experience with CKD were involved in this review. Sources of information Original research articles were identified from MEDLINE and Google Scholar using the search terms "diabetes," "diabetic kidney disease," "diabetic nephropathy," "chronic kidney disease," "kidney failure," "dialysis," "nephrology," "genomics," "metabolomics," and "proteomics." Methods A focused review and critical appraisal of existing literature regarding the precision medicine approaches to the diagnosis, prognosis, and treatment of diabetes and DKD framed by a patient partner's/caregiver's lived experience. Key findings Distinguishing diabetic nephropathy from CKD due to other types of DKD and non-DKD is challenging and typically requires a kidney biopsy for a diagnosis. Biomarkers have been identified to assist with the prediction of the onset and progression of DKD, but they have yet to be incorporated and evaluated relative to clinical standard of care CKD and kidney failure risk prediction tools. Genomics has identified multiple causal genetic variants for neonatal diabetes mellitus and monogenic diabetes of the young that can be used for diagnostic purposes and to specify antiglycemic therapy. Genome-wide-associated studies have identified genes implicated in DKD pathophysiology in the setting of type 1 and 2 diabetes but their translational benefits are lagging beyond polygenetic risk scores. Metabolomics and proteomics have been shown to improve diagnostic accuracy in DKD, have been used to identify novel pathways involved in DKD pathogenesis, and can be used to improve the prediction of CKD progression and kidney failure as well as predict response to DKD therapy. Limitations There are a limited number of large, high-quality prospective observational studies and no randomized controlled trials that support the use of precision medicine based approaches to improve clinical outcomes in adults with or at risk of diabetes and DKD. It is unclear which patients may benefit from the clinical use of genomics, metabolomics and proteomics along the spectrum of DKD trajectory. Implications Additional research is needed to evaluate the role of the use of precision medicine for DKD management, including diagnosis, differentiation of diabetic nephropathy from other etiologies of DKD and CKD, short-term and long-term risk prognostication kidney outcomes, and the prediction of response to and safety of disease-modifying therapies.
Collapse
Affiliation(s)
- Mallory L. Downie
- McGill University Health Center Research Institute, Montreal, QC, Canada
| | - Arlene Desjarlais
- Kidney Research Scientist Core Education and National Training Program, Montreal, QC, Canada
| | - Nancy Verdin
- Kidney Research Scientist Core Education and National Training Program, Montreal, QC, Canada
| | - Tania Woodlock
- Kidney Research Scientist Core Education and National Training Program, Montreal, QC, Canada
| | - David Collister
- Department of Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| |
Collapse
|
6
|
Shi L, Li C, Wang J, Zhong H, Wei T, Fan W, Li Z. The intellectual base and global trends in inflammation of diabetic kidney disease: a bibliometric analysis. Ren Fail 2023; 45:2270061. [PMID: 37870857 PMCID: PMC11001326 DOI: 10.1080/0886022x.2023.2270061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 10/08/2023] [Indexed: 10/24/2023] Open
Abstract
Diabetic kidney disease (DKD) is a severe complication of diabetes mellitus (DM). The literature on DKD inflammation research has experienced substantial growth. However, there is a lack of bibliometric analyses. This study aimed to examine the existing research on inflammation in DKD by analyzing articles published in the Web of Science Core Collection (WOSCC) over the past 30 years. We conducted a visualization analysis using several software, including CiteSpace and VOSviewer. We found that the literature on inflammation research in DKD has experienced substantial growth, indicating a rising interest in this developing area of study. In this field, Navarro-Gonzalez, JF is the most frequently cited author, Kidney International is the most frequently cited journal, China had the highest number of publications in the field of DKD inflammation, and Monash University emerged as the institution with the most published research. The research area on inflammation in DKD primarily centers around the investigation of 'Glycation end-products', 'chronic kidney disease', and 'diabetic nephropathy'. The emerging research trends in this field will focus on the 'Gut microbiota', 'NLRP3 inflammasome', 'autophagy', 'pyroptosis', 'sglt2 inhibitor', and 'therapeutic target'. Future research on DKD may focus on further exploring the inflammatory response, identifying specific therapeutic targets, studying biomarkers, investigating stem cell therapy and tissue engineering, and exploring gene therapy and gene editing. In summary, this study examines the main areas of study, frontiers, and trends in DKD inflammation, which have significant implications for future research.
Collapse
Affiliation(s)
- LuYao Shi
- Department of Nephrology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - ChangYan Li
- Department of Nephrology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Jian Wang
- The Second People’s Hospital of Baoshan City, Baoshan, China
| | - HuaChen Zhong
- First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Tao Wei
- Kunming Medical University, Kunming, Yunnan Province, China
| | - WenXing Fan
- Department of Nephrology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Zhen Li
- Organ Transplantation Center, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| |
Collapse
|
7
|
Zhang L, Liu M, Zhang Z, Chen D, Chen G, Liu M. Machine learning based identification of hub genes in renal clear cell carcinoma using multi-omics data. Methods 2022; 207:110-117. [PMID: 36179770 DOI: 10.1016/j.ymeth.2022.09.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/12/2022] [Accepted: 09/24/2022] [Indexed: 11/18/2022] Open
Abstract
Renal cell carcinoma is one of the most universal urinary system cancers in the world. The most common renal cell carcinoma subtype is renal clear cell carcinoma. It is usually associated with high rates of metastasis and mortality. Therefore, finding effective therapeutic targets and prognostic molecular markers is of great significance to improve the early diagnosis rate and prognostic accuracy of renal clear cell carcinoma. In this work, we successfully identified six hub genes that are closely related to the occurrence, development and prognosis of renal clear cell carcinoma and proposed three new potential prognostic markers, namely ATP4B, AC144831.1 and Tfcp2l1 through differentially expressed genes (DEGs) analysis, GO functional enrichment and KEGG pathway analysis, WGCNA analysis, and survival analysis. In addition, we established machine learning models to predict the occurrence of tumors through the gene expression data of patients. It is expected that the results of this study can provide reference value for the treatment of renal clear cell carcinoma.
Collapse
Affiliation(s)
- Lichao Zhang
- School of Intelligent Manufacturing and Equipment, Shenzhen Institute of Information Technology, Shenzhen, China
| | - Mingjun Liu
- School of Intelligent Manufacturing and Equipment, Shenzhen Institute of Information Technology, Shenzhen, China
| | - Zhenjiu Zhang
- School of Intelligent Manufacturing and Equipment, Shenzhen Institute of Information Technology, Shenzhen, China
| | | | | | - Mingyang Liu
- Beidahuang Industry Group General Hospital, Harbin, China.
| |
Collapse
|
8
|
Devarajan P, Chertow GM, Susztak K, Levin A, Agarwal R, Stenvinkel P, Chapman AB, Warady BA. Emerging Role of Clinical Genetics in CKD. Kidney Med 2022; 4:100435. [PMID: 35372818 PMCID: PMC8971313 DOI: 10.1016/j.xkme.2022.100435] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Chronic kidney disease (CKD) afflicts 15% of adults in the United States, of whom 25% have a family history. Genetic testing is supportive in identifying and possibly confirming diagnoses of CKD, thereby guiding care. Advances in the clinical genetic evaluation include next-generation sequencing with targeted gene panels, whole exome sequencing, and whole genome sequencing. These platforms provide DNA sequence reads with excellent coverage throughout the genome and have identified novel genetic causes of CKD. New pathologic genetic variants identified in previously unrecognized biological pathways have elucidated disease mechanisms underlying CKD etiologies, potentially establishing prognosis and guiding treatment selection. Molecular diagnoses using genetic sequencing can detect rare, potentially treatable mutations, avoid misdiagnoses, guide selection of optimal therapy, and decrease the risk of unnecessary and potentially harmful interventions. Genetic testing has been widely adopted in pediatric nephrology; however, it is less frequently used to date in adult nephrology. Extension of clinical genetic approaches to adult patients may achieve similar benefits in diagnostic refinement and treatment selection. This review aimed to identify clinical CKD phenotypes that may benefit the most from genetic testing, outline the commonly available platforms, and provide examples of successful deployment of these approaches in CKD.
Collapse
Affiliation(s)
- Prasad Devarajan
- Division of Nephrology and Hypertension, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH
| | | | - Katalin Susztak
- Division of Nephrology, Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Adeera Levin
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Rajiv Agarwal
- Division of Nephrology, Indiana University, Indianapolis, IN
| | - Peter Stenvinkel
- Department of Renal Medicine, Karolinska University Hospital at Huddinge, Karolinkska Institutet, Stockholm, Sweden
| | | | - Bradley A. Warady
- Division of Pediatric Nephrology, Children’s Mercy Kansas City, Kansas City, MO
| |
Collapse
|
9
|
El Jellas K, Dušátková P, Haldorsen IS, Molnes J, Tjora E, Johansson BB, Fjeld K, Johansson S, Průhová Š, Groop L, Löhr JM, Njølstad PR, Molven A. Two New Mutations in the CEL Gene Causing Diabetes and Hereditary Pancreatitis: How to Correctly Identify MODY8 Cases. J Clin Endocrinol Metab 2022; 107:e1455-e1466. [PMID: 34850019 PMCID: PMC8947231 DOI: 10.1210/clinem/dgab864] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Indexed: 11/26/2022]
Abstract
CONTEXT Maturity onset diabetes of the young, type 8 (MODY8) is associated with mutations in the CEL gene, which encodes the digestive enzyme carboxyl ester lipase. Several diabetes cases and families have in recent years been attributed to mutations in CEL without any functional or clinical evidence provided. OBJECTIVE To facilitate correct MODY8 diagnostics, we screened 2 cohorts of diabetes patients and delineated the phenotype. METHODS Young, lean Swedish and Finnish patients with a diagnosis of type 2 diabetes (352 cases, 406 controls) were screened for mutations in the CEL gene. We also screened 58 Czech MODY cases who had tested negative for common MODY genes. For CEL mutation-positive subjects, family history was recorded, and clinical investigations and pancreatic imaging performed. RESULTS Two cases (1 Swedish and 1 Czech) with germline mutation in CEL were identified. Clinical and radiological investigations of these 2 probands and their families revealed dominantly inherited insulin-dependent diabetes, pancreatic exocrine dysfunction, and atrophic pancreas with lipomatosis and cysts. Notably, hereditary pancreatitis was the predominant phenotype in 1 pedigree. Both families carried single-base pair deletions in the proximal part of the CEL variable number of tandem repeat (VNTR) region in exon 11. The mutations are predicted to lead to aberrant protein tails that make the CEL protein susceptible to aggregation. CONCLUSION The diagnosis of MODY8 requires a pancreatic exocrine phenotype and a deletion in the CEL VNTR in addition to dominantly inherited diabetes. CEL screening may be warranted also in families with hereditary pancreatitis of unknown genetic etiology.
Collapse
Affiliation(s)
- Khadija El Jellas
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, N-5020 Bergen, Norway
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway
| | - Petra Dušátková
- Department of Pediatrics, Charles University in Prague, Second Faculty of Medicine and University Hospital Motol, CZ-15006 Prague, Czech Republic
| | - Ingfrid S Haldorsen
- Mohn Medical Imaging and Visualization Centre, Department of Radiology, Haukeland University Hospital, N-5021 Bergen, Norway
- Section for Radiology, Department of Clinical Medicine, University of Bergen, N-5020 Bergen, Norway
| | - Janne Molnes
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway
- Department of Medical Genetics, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Erling Tjora
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway
- Children and Youth Clinic, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Bente B Johansson
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway
| | - Karianne Fjeld
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, N-5020 Bergen, Norway
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway
- Department of Medical Genetics, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Stefan Johansson
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway
- Department of Medical Genetics, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Štěpánka Průhová
- Department of Pediatrics, Charles University in Prague, Second Faculty of Medicine and University Hospital Motol, CZ-15006 Prague, Czech Republic
| | - Leif Groop
- Institute for Molecular Medicine Finland, Helsinki University, FI-00014 Helsinki, Finland
- Lund University Diabetes Centre, Department of Clinical Sciences, Lund University, Skåne University Hospital, SE-214 28 Malmö, Sweden
| | - J Matthias Löhr
- Department for Digestive Diseases, Karolinska University Hospital, SE-141 86 Stockholm, Sweden
- Department of Clinical Science, Intervention, and Technology (CLINTEC), Karolinska Institute, SE-141 86 Stockholm, Sweden
| | - Pål R Njølstad
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway
- Children and Youth Clinic, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Anders Molven
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, N-5020 Bergen, Norway
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway
- Department of Pathology, Haukeland University Hospital, N-5021 Bergen, Norway
| |
Collapse
|
10
|
Rafique I, Mir A, Siddiqui S, Saqib MAN, Fawwad A, Marchand L, Adnan M, Naeem M, Basit A, Polychronakos C. Comprehensive genetic screening reveals wide spectrum of genetic variants in monogenic forms of diabetes among Pakistani population. World J Diabetes 2021; 12:1957-1966. [PMID: 34888019 PMCID: PMC8613659 DOI: 10.4239/wjd.v12.i11.1957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/14/2021] [Accepted: 10/27/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Monogenic forms of diabetes (MFD) are single gene disorders. Their diagnosis is challenging, and symptoms overlap with type 1 and type 2 diabetes.
AIM To identify the genetic variants responsible for MFD in the Pakistani population and their frequencies.
METHODS A total of 184 patients suspected of having MFD were enrolled. The inclusion criterion was diabetes with onset below 25 years of age. Brief demographic and clinical information were taken from the participants. The maturity-onset diabetes of the young (MODY) probability score was calculated, and glutamate decarboxylase ELISA was performed. Antibody negative patients and features resembling MODY were selected (n = 28) for exome sequencing to identify the pathogenic variants.
RESULTS A total of eight missense novel or very low-frequency variants were identified in 7 patients. Three variants were found in genes for MODY, i.e. HNF1A (c.169C>A, p.Leu57Met), KLF11 (c.401G>C, p.Gly134Ala), and HNF1B (c.1058C>T, p.Ser353Leu). Five variants were found in genes other than the 14 known MODY genes, i.e. RFX6 (c.919G>A, p.Glu307Lys), WFS1 (c.478G>A, p.Glu160Lys) and WFS1 (c.517G>A, p.Glu173Lys), RFX6 (c.1212T>A, p.His404Gln) and ZBTB20 (c.1049G>A, p.Arg350His).
CONCLUSION The study showed wide spectrum of genetic variants potentially causing MFD in the Pakistani population. The MODY genes prevalent in European population (GCK, HNF1A, and HNF4a) were not found to be common in our population. Identification of novel variants will further help to understand the role of different genes causing the pathogenicity in MODY patient and their proper management and diagnosis.
Collapse
Affiliation(s)
- Ibrar Rafique
- Department of Biological Sciences, International Islamic University, Islamabad 44000, Pakistan
- Departments of Pediatrics and Human Genetics, McGill University Health Centre Research Institute, Montreal H4A 3J1, Canada
- Research Development and Coordination, Pakistan Health Research Council, Islamabad 44000, Pakistan
| | - Asif Mir
- Department of Biological Sciences, International Islamic University, Islamabad 44000, Pakistan
| | - Shajee Siddiqui
- Department of Medicine, Pakistan Institute of Medical Sciences, Islamabad 44000, Pakistan, Pakistan
| | | | - Asher Fawwad
- Department of Biochemistry, Baqai Institute of Diabetology and Endocrinology, Baqai Medical University, Karachi 74600, Sindh, Pakistan
| | - Luc Marchand
- Departments of Pediatrics and Human Genetics, McGill University Health Centre Research Institute, Montreal H4A 3J1, Canada
| | - Muhammad Adnan
- PHRC Research Centre, FJMU, Pakistan Health Research Council, Lahore 54000, Pakistan
| | - Muhammad Naeem
- Department of Biotechnology, Quaid-I-Azam University, Islamabad 44000, Pakistan
| | - Abdul Basit
- Department of Medicine, Baqai Institute of Diabetology and Endocrinology, Baqai Medical University, Karachi 74600, Sindh, Pakistan
| | - Constantin Polychronakos
- Departments of Pediatrics and Human Genetics, McGill University Health Centre Research Institute, Montreal H4A 3J1, Canada
| |
Collapse
|
11
|
Cao Y, Zhan Y, Qiu S, Chen Z, Gong K, Ni S, Duan Y. Integrative analysis of genome-wide DNA methylation and single-nucleotide polymorphism identified ACSM5 as a suppressor of lumbar ligamentum flavum hypertrophy. Arthritis Res Ther 2021; 23:251. [PMID: 34593020 PMCID: PMC8482693 DOI: 10.1186/s13075-021-02625-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 09/12/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Hypertrophy of ligamentum flavum (HLF) is a common lumbar degeneration disease (LDD) with typical symptoms of low back pain and limb numbness owing to an abnormal pressure on spinal nerves. Previous studies revealed HLF might be caused by fibrosis, inflammatory, and other bio-pathways. However, a global analysis of HLF is needed severely. METHODS A genome-wide DNA methylation and single-nucleotide polymorphism analysis were performed from five LDD patients with HLF and five LDD patients without HLF. Comprehensive integrated analysis was performed using bioinformatics analysis and the validated experiments including Sanger sequencing, methylation-specific PCR, qPCR and ROC analysis. Furthermore, the function of novel genes in ligamentum flavum cells (LFCs) was detected to explore the molecular mechanism in HLF through knock down experiment, overexpression experiment, CCK8 assay, apoptosis assay, and so on. RESULTS We identified 69 SNP genes and 735 661 differentially methylated sites that were enriched in extracellular matrix, inflammatory, and cell proliferation. A comprehensive analysis demonstrated key genes in regulating the development of HLF including ACSM5. Furthermore, the hypermethylation of ACSM5 that was mediated by DNMT1 led to downregulation of ACSM5 expression, promoted the proliferation and fibrosis, and inhibited the apoptosis of LFCs. CONCLUSION This study revealed that DNMT1/ACSM5 signaling could enhance HLF properties in vitro as a potential therapeutic strategy for HLF.
Collapse
Affiliation(s)
- Yanlin Cao
- Department of Spine Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yenan Zhan
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Sujun Qiu
- Department of Spine Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Zhong Chen
- Department of Spine Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Kaiqin Gong
- Department of Spine Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Songjia Ni
- Department of Orthopaedic Trauma, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Yang Duan
- Department of Spine Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| |
Collapse
|
12
|
Daehn IS, Duffield JS. The glomerular filtration barrier: a structural target for novel kidney therapies. Nat Rev Drug Discov 2021; 20:770-788. [PMID: 34262140 PMCID: PMC8278373 DOI: 10.1038/s41573-021-00242-0] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/28/2021] [Indexed: 12/19/2022]
Abstract
Loss of normal kidney function affects more than 10% of the population and contributes to morbidity and mortality. Kidney diseases are currently treated with immunosuppressive agents, antihypertensives and diuretics with partial but limited success. Most kidney disease is characterized by breakdown of the glomerular filtration barrier (GFB). Specialized podocyte cells maintain the GFB, and structure-function experiments and studies of intercellular communication between the podocytes and other GFB cells, combined with advances from genetics and genomics, have laid the groundwork for a new generation of therapies that directly intervene at the GFB. These include inhibitors of apolipoprotein L1 (APOL1), short transient receptor potential channels (TRPCs), soluble fms-like tyrosine kinase 1 (sFLT1; also known as soluble vascular endothelial growth factor receptor 1), roundabout homologue 2 (ROBO2), endothelin receptor A, soluble urokinase plasminogen activator surface receptor (suPAR) and substrate intermediates for coenzyme Q10 (CoQ10). These molecular targets converge on two key components of GFB biology: mitochondrial function and the actin-myosin contractile machinery. This Review discusses therapies and developments focused on maintaining GFB integrity, and the emerging questions in this evolving field.
Collapse
Affiliation(s)
- Ilse S Daehn
- Department of Medicine, Division of Nephrology, The Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Jeremy S Duffield
- Research and Development, Prime Medicine, Cambridge, MA, USA. .,Department of Medicine, University of Washington, Seattle, WA, USA. .,Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
13
|
Quinlan C, Rheault MN. Genetic Basis of Type IV Collagen Disorders of the Kidney. Clin J Am Soc Nephrol 2021; 16:1101-1109. [PMID: 33849932 PMCID: PMC8425620 DOI: 10.2215/cjn.19171220] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The glomerular basement membrane is a vital component of the filtration barrier of the kidney and is primarily composed of a highly structured matrix of type IV collagen. Specific isoforms of type IV collagen, the α3(IV), α4(IV), and α5(IV) isoforms, assemble into trimers that are required for normal glomerular basement membrane function. Disruption or alteration in these isoforms leads to breakdown of the glomerular basement membrane structure and function and can lead to progressive CKD known as Alport syndrome. However, there is wide variability in phenotype among patients with mutations affecting type IV collagen that depends on a complex interplay of sex, genotype, and X-chromosome inactivation. This article reviews the genetic basis of collagen disorders of the kidney as well as potential treatments for these conditions, including direct alteration of the DNA, RNA therapies, and manipulation of collagen proteins.
Collapse
Affiliation(s)
- Catherine Quinlan
- Department of Nephrology, Royal Children’s Hospital, Melbourne, Victoria, Australia,Department of Kidney Regeneration, Murdoch Children’s Research Institute, Melbourne, Victoria, Australia,Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Michelle N. Rheault
- Division of Pediatric Nephrology, Department of Pediatrics, University of Minnesota Masonic Children’s Hospital, Minneapolis, Minnesota
| |
Collapse
|
14
|
Cognitive analysis of metabolomics data for systems biology. Nat Protoc 2021; 16:1376-1418. [PMID: 33483720 DOI: 10.1038/s41596-020-00455-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 10/27/2020] [Indexed: 01/30/2023]
Abstract
Cognitive computing is revolutionizing the way big data are processed and integrated, with artificial intelligence (AI) natural language processing (NLP) platforms helping researchers to efficiently search and digest the vast scientific literature. Most available platforms have been developed for biomedical researchers, but new NLP tools are emerging for biologists in other fields and an important example is metabolomics. NLP provides literature-based contextualization of metabolic features that decreases the time and expert-level subject knowledge required during the prioritization, identification and interpretation steps in the metabolomics data analysis pipeline. Here, we describe and demonstrate four workflows that combine metabolomics data with NLP-based literature searches of scientific databases to aid in the analysis of metabolomics data and their biological interpretation. The four procedures can be used in isolation or consecutively, depending on the research questions. The first, used for initial metabolite annotation and prioritization, creates a list of metabolites that would be interesting for follow-up. The second workflow finds literature evidence of the activity of metabolites and metabolic pathways in governing the biological condition on a systems biology level. The third is used to identify candidate biomarkers, and the fourth looks for metabolic conditions or drug-repurposing targets that the two diseases have in common. The protocol can take 1-4 h or more to complete, depending on the processing time of the various software used.
Collapse
|
15
|
Aarthy R, Aston-Mourney K, Mikocka-Walus A, Radha V, Amutha A, Anjana RM, Unnikrishnan R, Mohan V. Clinical features, complications and treatment of rarer forms of maturity-onset diabetes of the young (MODY) - A review. J Diabetes Complications 2021; 35:107640. [PMID: 32763092 DOI: 10.1016/j.jdiacomp.2020.107640] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/21/2020] [Accepted: 05/21/2020] [Indexed: 12/15/2022]
Abstract
Maturity onset diabetes of the young (MODY) is the most common form of monogenic diabetes and is currently believed to have 14 subtypes. While much is known about the common subtypes of MODY (MODY-1, 2, 3 and 5) little is known about its rare subtypes (MODY4, 6-14). With the advent of next-generation sequencing (NGS) there are several reports of the rarer subtypes of MODY emerging from across the world. Therefore, a greater understanding on these rarer subtypes is needed. A search strategy was created, and common databases were searched, and 51 articles finally selected. INS-(MODY10) and ABCC8-(MODY12) mutations were reported in relatively large numbers compared to the other rare subtypes. The clinical characteristics of the rare MODY subtypes exhibited heterogeneity between families reported with the same mutation. Obesity and diabetic ketoacidosis (DKA) were also reported among rarer MODY subtypes which presents as a challenge as these are not part of the original description of MODY by Tattersal and Fajans. The treatment modalities of the rarer subtypes included oral drugs, predominantly sulfonylureas, insulin but also diet alone. Newer drugs like DPP-4 and SGLT2 inhibitors have also been tried as new modes of treatment. The microvascular and macrovascular complications among the patients with various MODY subtypes are less commonly reported. Recently, there is a view that not all the 14 forms of 'MODY' are true MODY and the very existence of some of these rarer subtypes as MODY has been questioned. This scoping review aims to report on the clinical characteristics, treatment and complications of the rarer MODY subtypes published in the literature.
Collapse
Affiliation(s)
- Ramasamy Aarthy
- School of Medicine, Deakin University, Australia; Madras Diabetes Research Foundation, Chennai, India
| | | | | | | | | | - Ranjit Mohan Anjana
- Dr Mohan's Diabetes Specialities Centre, Madras Diabetes Research Foundation, Chennai, India
| | - Ranjit Unnikrishnan
- Dr Mohan's Diabetes Specialities Centre, Madras Diabetes Research Foundation, Chennai, India
| | - Viswanathan Mohan
- Dr Mohan's Diabetes Specialities Centre, Madras Diabetes Research Foundation, Chennai, India.
| |
Collapse
|
16
|
SGLT2 inhibitors - a potential treatment for Alport syndrome. Clin Sci (Lond) 2020; 134:379-388. [PMID: 32064497 DOI: 10.1042/cs20191276] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/30/2020] [Accepted: 02/05/2020] [Indexed: 12/11/2022]
Abstract
Alport syndrome is a rare genetic disease that results in disordered basement membrane type IV collagen resulting in occular and auditory defects as well of progressive kidney disease. Although no 'cure' currently exists, therapeutic blockade of the renin-angiotensin-aldosterone system can slow the progression to end-stage kidney disease (ESKD). Clinical trials for treatments in preventing chronic kidney disease have largely been negative over the last two decades until recent trials have shown positive cardiovascular and renal outcomes of sodium-glucose co-transporter-2 (SGLT2) inhibitors in patients with diabetes mellitus. Although marketed as medications for Type 2 diabetes, SGLT2 inhibitors have been found to have additional properties that are nephroprotective which makes them a potential candidate for treatment for those with other forms of progressive kidney disease. This review discusses the evidence for the use of SGLT2 inhibitors as a potential treatment in Alport syndrome that may slow the progression of chronic kidney disease and prevent patients reaching ESKD.
Collapse
|
17
|
Jia X, Yamamura T, Gbadegesin R, McNulty MT, Song K, Nagano C, Hitomi Y, Lee D, Aiba Y, Khor SS, Ueno K, Kawai Y, Nagasaki M, Noiri E, Horinouchi T, Kaito H, Hamada R, Okamoto T, Kamei K, Kaku Y, Fujimaru R, Tanaka R, Shima Y, Baek J, Kang HG, Ha IS, Han KH, Yang EM, Abeyagunawardena A, Lane B, Chryst-Stangl M, Esezobor C, Solarin A, Dossier C, Deschênes G, Vivarelli M, Debiec H, Ishikura K, Matsuo M, Nozu K, Ronco P, Cheong HI, Sampson MG, Tokunaga K, Iijima K. Common risk variants in NPHS1 and TNFSF15 are associated with childhood steroid-sensitive nephrotic syndrome. Kidney Int 2020; 98:1308-1322. [PMID: 32554042 DOI: 10.1016/j.kint.2020.05.029] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 04/23/2020] [Accepted: 05/01/2020] [Indexed: 12/16/2022]
Abstract
To understand the genetics of steroid-sensitive nephrotic syndrome (SSNS), we conducted a genome-wide association study in 987 childhood SSNS patients and 3,206 healthy controls with Japanese ancestry. Beyond known associations in the HLA-DR/DQ region, common variants in NPHS1-KIRREL2 (rs56117924, P=4.94E-20, odds ratio (OR) =1.90) and TNFSF15 (rs6478109, P=2.54E-8, OR=0.72) regions achieved genome-wide significance and were replicated in Korean, South Asian and African populations. Trans-ethnic meta-analyses including Japanese, Korean, South Asian, African, European, Hispanic and Maghrebian populations confirmed the significant associations of variants in NPHS1-KIRREL2 (Pmeta=6.71E-28, OR=1.88) and TNFSF15 (Pmeta=5.40E-11, OR=1.33) loci. Analysis of the NPHS1 risk alleles with glomerular NPHS1 mRNA expression from the same person revealed allele specific expression with significantly lower expression of the transcript derived from the risk haplotype (Wilcox test p=9.3E-4). Because rare pathogenic variants in NPHS1 cause congenital nephrotic syndrome of the Finnish type (CNSF), the present study provides further evidence that variation along the allele frequency spectrum in the same gene can cause or contribute to both a rare monogenic disease (CNSF) and a more complex, polygenic disease (SSNS).
Collapse
Affiliation(s)
- Xiaoyuan Jia
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomohiko Yamamura
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Rasheed Gbadegesin
- Department of Pediatrics, Division of Nephrology, Duke University Medical Center, Durham, North Carolina, USA
| | - Michelle T McNulty
- Department of Medicine-Nephrology, Boston Children's Hospital, Boston, Massachussetts, USA; Medical and Population Genetics, Broad Institute, Cambridge, Massachussetts, USA
| | - Kyuyong Song
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, Korea
| | - China Nagano
- 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
| | - Dongwon Lee
- Department of Medicine-Nephrology, Boston Children's Hospital, Boston, Massachussetts, USA; Medical and Population Genetics, Broad Institute, Cambridge, Massachussetts, USA; Harvard Medical School, Boston, Massachussetts, USA
| | - Yoshihiro Aiba
- Clinical Research Center, National Hospital Organization Nagasaki Medical Center, Omura, Japan
| | - Seik-Soon Khor
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazuko Ueno
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yosuke Kawai
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masao Nagasaki
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Eisei Noiri
- Department of Hemodialysis and Apheresis, The University of Tokyo Hospital, Tokyo, Japan
| | - Tomoko Horinouchi
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiroshi Kaito
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan; Department of Nephrology, Hyogo Prefectural Kobe Children's Hospital, Kobe, Japan
| | - Riku Hamada
- Department of Nephrology, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Takayuki Okamoto
- Department of Pediatrics, Hokkaido University Hospital, Sapporo, Japan
| | - Koichi Kamei
- Division of Nephrology and Rheumatology, National Center for Child Health and Development, Tokyo, Japan
| | - Yoshitsugu Kaku
- Department of Nephrology, Fukuoka Children's Hospital, Fukuoka, Japan
| | - Rika Fujimaru
- Department of Pediatrics, Osaka City General Hospital, Osaka, Japan
| | - Ryojiro Tanaka
- Department of Nephrology, Hyogo Prefectural Kobe Children's Hospital, Kobe, Japan
| | - Yuko Shima
- Department of Pediatrics, Wakayama Medical University, Wakayama, Japan
| | | | - Jiwon Baek
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, Korea
| | - Hee Gyung Kang
- Department of Pediatrics, Seoul National University Children's Hospital, Seoul, Korea
| | - Il-Soo Ha
- Department of Pediatrics, Seoul National University Children's Hospital, Seoul, Korea
| | - Kyoung Hee Han
- Department of Pediatrics, Jeju National University School of Medicine, Jeju, Korea
| | - Eun Mi Yang
- Department of Pediatrics, Chonnam National University Children's Hospital, Gwangju, Korea
| | | | - Asiri Abeyagunawardena
- Department of Pediatrics, Division of Nephrology, Duke University Medical Center, Durham, North Carolina, USA
| | - Brandon Lane
- Department of Pediatrics, Division of Nephrology, Duke University Medical Center, Durham, North Carolina, USA
| | - Megan Chryst-Stangl
- Department of Pediatrics, Division of Nephrology, Duke University Medical Center, Durham, North Carolina, USA
| | - Christopher Esezobor
- Department of Paediatrics, College of Medicine, University of Lagos, Lagos, Nigeria
| | - Adaobi Solarin
- Department of Pediatrics, Lagos State University Teaching Hospital, Ikeja, Nigeria
| | | | - Claire Dossier
- Department of Paediatric Nephrology, Public Assistance Hospital of Paris, Robert-Debré Hospital, Paris, France
| | - Georges Deschênes
- Center of Research on Inflammation, Institut National de la Santé et de la Recherche Médicale UMR 1149, University Sorbonne-Paris, Paris, France
| | | | - Marina Vivarelli
- Division of Nephrology and Dialysis, Bambino Gesù Children's Hospital and Research Institute, Rome, Italy
| | - Hanna Debiec
- Sorbonne University, INSERM UMR_S1155, and Nephrology Day Hospital, Department of Nephrology, Hôpital Tenon, Paris France
| | - Kenji Ishikura
- Division of Nephrology and Rheumatology, National Center for Child Health and Development, Tokyo, Japan
| | - Masafumi Matsuo
- Research Center for Locomotion Biology, Kobe Gakuin University, Kobe, Japan; KNC Department of Nucleic Acid Drug Discovery, Faculty of Rehabilitation, Kobe Gakuin University, Kobe, Japan
| | - Kandai Nozu
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Pierre Ronco
- Sorbonne University, INSERM UMR_S1155, and Nephrology Day Hospital, Department of Nephrology, Hôpital Tenon, Paris France
| | - Hae Il Cheong
- Department of Pediatrics, Seoul National University Children's Hospital, Seoul, Korea
| | - Matthew G Sampson
- Department of Medicine-Nephrology, Boston Children's Hospital, Boston, Massachussetts, USA; Medical and Population Genetics, Broad Institute, Cambridge, Massachussetts, USA; Harvard Medical School, Boston, Massachussetts, USA
| | - 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
|
18
|
Zhao L, Ren H, Zhang J, Cao Y, Wang Y, Meng D, Wu Y, Zhang R, Zou Y, Xu H, Li L, Zhang J, Cooper ME, Tong N, Liu F. DIABETIC RETINOPATHY, CLASSIFIED USING THE LESION-AWARE DEEP LEARNING SYSTEM, PREDICTS DIABETIC END-STAGE RENAL DISEASE IN CHINESE PATIENTS. Endocr Pract 2020; 26:429-443. [PMID: 31968187 DOI: 10.4158/ep-2019-0512] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Objective: To characterize the relationship between diabetic retinopathy (DR) and diabetic nephropathy (DN) in Chinese patients and to determine whether the severity of DR predicts end-stage renal disease (ESRD). Methods: Bilateral fundic photographs of 91 Chinese type 2 diabetic patients with biopsy-confirmed DN, not in ESRD stage, were obtained at the time of renal biopsy in this longitudinal study. The baseline severity of DR was determined using the Lesion-aware Deep Learning System (RetinalNET) in an open framework for deep learning and was graded using the Early Treatment Diabetic Retinopathy Study severity scale. Cox proportional hazard models were used to estimate the hazard ratio (HR) for the effect of the severity of diabetic retinopathy on ESRD. Results: During a median follow-up of 15 months, 25 patients progressed to ESRD. The severity of retinopathy at the time of biopsy was a prognostic factor for progression to ESRD (HR 2.18, 95% confidence interval 1.05 to 4.53, P = .04). At baseline, more severe retinopathy was associated with poor renal function, and more severe glomerular lesions. However, 30% of patients with mild retinopathy and severe glomerular lesions had higher low-density lipo-protein-cholesterol and more severe proteinuria than those with mild glomerular lesions. Additionally, 3% of patients with severe retinopathy and mild glomerular changes were more likely to have had diabetes a long time than those with severe glomerular lesions. Conclusion: Although the severity of DR predicted diabetic ESRD in patients with type 2 diabetes mellitus and DN, the severities of DR and DN were not always consistent, especially in patients with mild retinopathy or microalbuminuria. Abbreviations: CI = confidence interval; DM = diabetic mellitus; DN = diabetic nephropathy; DR = diabetic retinopathy; eGFR = estimated glomerular filtration rate; ESRD = end-stage renal disease; HbA1c = hemoglobin A1c; HR = hazard ratio; NPDR = nonproliferative diabetic retinopathy; PDR = proliferative diabetic retinopathy; SBP = systolic blood pressure; T2DM = type 2 diabetes mellitus; VEGF = vascular endothelial growth factor.
Collapse
|
19
|
Zhao L, Zou Y, Zhang J, Zhang R, Ren H, Li L, Guo R, Zhang J, Liu F. Serum transferrin predicts end-stage Renal Disease in Type 2 Diabetes Mellitus patients. Int J Med Sci 2020; 17:2113-2124. [PMID: 32922172 PMCID: PMC7484672 DOI: 10.7150/ijms.46259] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 07/16/2020] [Indexed: 02/05/2023] Open
Abstract
Background: To investigate the relationship between serum iron status and renal outcome in patients with type 2 diabetes mellitus (T2DM). Methods: Chinese patients (n=111) with T2DM and biopsy-proven diabetic nephropathy (DN) were surveyed in a longitudinal, retrospective study. Serum iron, total iron-binding capacity, ferritin, and transferrin were measured at the time of renal biopsy. Iron deposition and transferrin staining were performed with renal biopsy specimens of DN patients and potential kidney donors. End-stage renal disease (ESRD) was the end-point. ESRD was defined as an estimated glomerular filtration rate <15 mL/min/1.73 m2 or the need for chronic renal replacement therapy. Cox proportional hazard models were used to estimate the hazard ratios (HRs) for the influence of serum iron metabolism on ESRD. Results: During a median follow up of 30.9 months, 66 (59.5%) patients progressed to ESRD. After adjusting for age, sex, baseline systolic blood pressure, renal functions, hemoglobin, HbA1c, and pathological findings, lower serum transferrin concentrations were significantly associated with higher ESRD in multivariate models. Compared with patients in the highest transferrin quartile (≥1.65 g/L), patients in the lowest quartile (≤1.15 g/L) had multivariable-adjusted HR (95% confidence interval) of 7.36 (1.40-38.65) for ESRD. Moreover, tubular epithelial cells in DN exhibited a higher deposition of iron and transferrin expression compared with healthy controls. Conclusions: Low serum transferrin concentration was associated with diabetic ESRD in patients with T2DM. Free iron nephrotoxicity and poor nutritional status with accumulated iron or transferrin deposition might contribute to ESRD.
Collapse
Affiliation(s)
- Lijun Zhao
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, Sichuan, China.,Division of General Practice, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yutong Zou
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Junlin Zhang
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Rui Zhang
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Honghong Ren
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Lin Li
- Division of Pathology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Ruikun Guo
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Jie Zhang
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health, Regenerative Medicine Research Center, Chengdu China
| | - Fang Liu
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
20
|
Minakawa A, Fukuda A, Sato Y, Kikuchi M, Kitamura K, Wiggins RC, Fujimoto S. Podocyte hypertrophic stress and detachment precedes hyperglycemia or albuminuria in a rat model of obesity and type2 diabetes-associated nephropathy. Sci Rep 2019; 9:18485. [PMID: 31811176 PMCID: PMC6898392 DOI: 10.1038/s41598-019-54692-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 10/16/2019] [Indexed: 02/06/2023] Open
Abstract
Type2 diabetes-associated nephropathy is the commonest cause of renal failure. Mechanisms responsible are controversial. Leptin-deficient hyperphagic Zucker (fa/fa) rats were modeled to test the hypothesis that glomerular enlargement drives podocyte hypertrophic stress leading to accelerated podocyte detachment, podocyte depletion, albuminuria and progression. By 6weeks, prior to development of either hyperglycemia or albuminuria, fa/fa rats were hyperinsulinemic with high urinary IGF1/2 excretion, gaining weight rapidly, and had 1.6-fold greater glomerular volume than controls (P < 0.01). At this time the podocyte number per glomerulus was not yet reduced although podocytes were already hypertrophically stressed as shown by high podocyte phosphor-ribosomal S6 (a marker of mTORC1 activation), high urinary pellet podocin:nephrin mRNA ratio and accelerated podocyte detachment (high urinary pellet podocin:aquaporin2 mRNA ratio). Subsequently, fa/fa rats became both hyperglycemic and albuminuric. 24 hr urine albumin excretion correlated highly with decreasing podocyte density (R2 = 0.86), as a consequence of both increasing glomerular volume (R2 = 0.70) and decreasing podocyte number (R2 = 0.63). Glomerular podocyte loss rate was quantitatively related to podocyte detachment rate measured by urine pellet mRNAs. Glomerulosclerosis occurred when podocyte density reached <50/106um3. Reducing food intake by 40% to slow growth reduced podocyte hypertrophic stress and "froze" all elements of the progression process in place, but had small effect on hyperglycemia. Glomerular enlargement caused by high growth factor milieu starting in pre-diabetic kidneys appears to be a primary driver of albuminuria in fa/fa rats and thereby an under-recognized target for progression prevention. Progression risk could be identified prior to onset of hyperglycemia or albuminuria, and monitored non-invasively by urinary pellet podocyte mRNA markers.
Collapse
Affiliation(s)
- Akihiro Minakawa
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
- First Department of Internal Medicine, University of Miyazaki, Miyazaki, Japan
| | - Akihiro Fukuda
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan.
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, Yufu, Japan.
| | - Yuji Sato
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Masao Kikuchi
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
- First Department of Internal Medicine, University of Miyazaki, Miyazaki, Japan
| | - Kazuo Kitamura
- First Department of Internal Medicine, University of Miyazaki, Miyazaki, Japan
| | - Roger C Wiggins
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Shouichi Fujimoto
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
- Department of Hemovascular Medicine and Artificial Organs, University of Miyazaki, Miyazaki, Japan
| |
Collapse
|
21
|
Xia L, Cao Y, Guo Y, Ba G, Luo Q, Shi H, Feng Y, Yin S. A Novel Heterozygous Mutation of the COL4A3 Gene Causes a Peculiar Phenotype without Hematuria and Renal Function Impairment in a Chinese Family. DISEASE MARKERS 2019; 2019:8705989. [PMID: 30881523 PMCID: PMC6387717 DOI: 10.1155/2019/8705989] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 01/16/2019] [Accepted: 01/17/2019] [Indexed: 01/15/2023]
Abstract
Mutations in the COL4A3 gene are frequently reported to be associated with various types of hereditary nephropathy. COL4A3 encodes the α3 chain of type IV collagen, which is the main structural protein in the basement membrane. Mutations in this gene are always related to kidney performance, and deafness and ocular lesion have also been reported. In this study, using next-generation sequencing, we investigated the DNA of a family visiting a clinic for hearing loss. A new missense mutation was found in COL4A3 of 5 patients, c.3227C>T (p.P1076L). Based on these results, we predict that the mutation is pathogenic and leads to abnormal collagen IV. Here, we report for the first time on this autosomal dominant syndrome, characterized by hearing loss and eye abnormalities, but without renal damage, in all carriers. Since the oldest patient in the trial was less than 50 years old, however, we recommend that renal examination be reviewed regularly. Our results reveal expansion in the mutation spectrum of the COL4A3 gene and phenotypic spectrum of collagen IV disease. Our study suggests that next-generation sequencing is an economical and effective method and may help in the accurate diagnosis and treatment of these patients.
Collapse
Affiliation(s)
- Liang Xia
- Department of Otolaryngology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yishan Road, Xuhui District, 200233 Shanghai, China
| | - Yangjia Cao
- Metabolic Bone Disease and Genetic Research Unit, Department of Osteoporosis and Bone Diseases, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yi-Shan Rd, Shanghai 200233, China
| | - Yang Guo
- Department of Otolaryngology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yishan Road, Xuhui District, 200233 Shanghai, China
| | - Guangyi Ba
- Department of Otolaryngology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yishan Road, Xuhui District, 200233 Shanghai, China
| | - Qiong Luo
- Department of Otolaryngology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yishan Road, Xuhui District, 200233 Shanghai, China
| | - Haibo Shi
- Department of Otolaryngology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yishan Road, Xuhui District, 200233 Shanghai, China
| | - Yanmei Feng
- Department of Otolaryngology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yishan Road, Xuhui District, 200233 Shanghai, China
| | - Shankai Yin
- Department of Otolaryngology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yishan Road, Xuhui District, 200233 Shanghai, China
| |
Collapse
|
22
|
Wang Y, Zhao L, Zhang J, Wu Y, Zhang R, Li H, Guo R, Han Q, Wang T, Li L, Wang S, Liu F. Implications of a Family History of Diabetes and Rapid eGFR Decline in Patients With Type 2 Diabetes and Biopsy-Proven Diabetic Kidney Disease. Front Endocrinol (Lausanne) 2019; 10:855. [PMID: 31920969 PMCID: PMC6923196 DOI: 10.3389/fendo.2019.00855] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 11/21/2019] [Indexed: 02/05/2023] Open
Abstract
Objective: This study aimed to identify the risk factors for a rapid decline in the estimated glomerular filtration rate (eGFR) in Chinese patients with type 2 diabetes and biopsy-proven diabetic kidney disease (DKD). Method: This was a retrospective cohort study. Patients with biopsy-proven DKD who had been followed-up for at least 1 year were enrolled. Baseline clinicopathological data and serum creatinine levels that had been measured at least three times during follow-up in our hospital were collected. Patients were allocated to two groups of rapid decliners and slow decliners according to the median eGFR slope. The associations between potential risk factors and rapid eGFR decline were analyzed using logistic regression. Results: A total of 128 eligible patients were enrolled and they had a mean age of 51.5 ± 10.7 years. During a median follow-up of 2 years, the median eGFR slope was -8.1 ± 14.4 mL/min/1.73 m2/year. The eGFR decline was significantly faster in patients with a family history of diabetes in first-degree relatives, nephrotic-range proteinuria, higher grades of glomerular pathology, and interstitial inflammation. No differences in the rate of the eGFR decline were observed in subgroups created according to sex, age, hypertension, glycosylated hemoglobin, diabetic retinopathy, interstitial fibrosis, and tubular atrophy. Logistic regression indicated that a family history of diabetes was independently associated with a rapid decline in eGFR, even after adjustment for factors including baseline eGFR and proteinuria. Conclusion: A family history of diabetes in first-degree relatives is independently associated with a rapid decline in eGFR in the current relatively young studied patients. Our findings suggested that early diagnosis and treatment is important for these patients.
Collapse
Affiliation(s)
- Yiting Wang
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, China
| | - Lijun Zhao
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, China
| | - Junlin Zhang
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, China
| | - Yucheng Wu
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, China
| | - Rui Zhang
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, China
| | - Hanyu Li
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, China
| | - Ruikun Guo
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, China
| | - Qianqian Han
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, China
| | - Tingli Wang
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, China
| | - Lin Li
- Division of Pathology, West China Hospital of Sichuan University, Chengdu, China
| | - Shanshan Wang
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, China
| | - Fang Liu
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, China
- *Correspondence: Fang Liu
| |
Collapse
|
23
|
Affiliation(s)
- Stephen S Rich
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia
| |
Collapse
|