1
|
Kebede MA, Mengistu YT, Loge BY, Eshetu MA, Shash EP, Wirtu AT, Gemechu JM. Determinants of Disease Progression in Autosomal Dominant Polycystic Kidney Disease. J Pers Med 2024; 14:936. [PMID: 39338190 PMCID: PMC11433103 DOI: 10.3390/jpm14090936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/24/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Despite its severity, there has been a lack of adequate study on autosomal dominant polycystic kidney disease (ADPKD) in Ethiopia. This study assessed the clinical profile and determinant factors contributing to renal disease progression. METHODS A retrospective study was conducted on 114 patients for 6 years in Addis Ababa. Patients with ADPKD who had follow-up visits at two health centers were included. RESULTS The mean age at diagnosis was 42.7 ± 12.7 years, with 43% reporting a positive family history of ADPKD. Approximately 22 patients (20%) developed end-stage renal disease, and 12 patients died. The mean estimated glomerular filtration rate at the initial visit was 72.4 mL/min/1.73 m2. The key risk factors associated with disease progression included younger age at diagnosis [adjusted Odds Ratio (aOR): 0.92, 95% CI: 0.87-0.98; p = 0.007], male gender (aOR: 4.5, 95% CI: 1.3-15.95, p = 0.017), higher baseline systolic blood pressure (aOR: 1.05, 95% CI: 1.01-1.10, p = 0.026), and the presence of comorbidities (aOR: 3.95, 95% CI: 1.10-14.33, p = 0.037). The progression of renal disease in ADPKD patients significantly correlates with age at diagnosis, gender, presence of comorbidities, and higher baseline systolic blood pressure. CONCLUSIONS These findings underscore the importance of early detection and management of hypertension and comorbidities in ADPKD patients to mitigate disease progression and improve treatment outcomes.
Collapse
Affiliation(s)
- Molla Asnake Kebede
- Department of Internal Medicine, School of Medicine, College of Medicine and Health Science, Mizan-Tepi University, Mizan-Aman P.O. Box 260, Ethiopia; (M.A.K.); (M.A.E.); (E.P.S.)
| | - Yewondwosen Tadesse Mengistu
- Department of Nephrology, College of Health Sciences, Addis Ababa University, Addis Ababa P.O. Box 9086, Ethiopia;
| | - Biruk Yacob Loge
- Durame General Hospital, Internal Medicine Unite, SNNPR, Durame P.O. Box 143, Ethiopia;
| | - Misikr Alemu Eshetu
- Department of Internal Medicine, School of Medicine, College of Medicine and Health Science, Mizan-Tepi University, Mizan-Aman P.O. Box 260, Ethiopia; (M.A.K.); (M.A.E.); (E.P.S.)
| | - Erkihun Pawlos Shash
- Department of Internal Medicine, School of Medicine, College of Medicine and Health Science, Mizan-Tepi University, Mizan-Aman P.O. Box 260, Ethiopia; (M.A.K.); (M.A.E.); (E.P.S.)
| | - Amenu Tolera Wirtu
- Meritus Medical Center, Meritus School of Osteopathic Medicine, Hagerstown, MD 21742, USA;
| | - Jickssa Mulissa Gemechu
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA
| |
Collapse
|
2
|
Sekar T, Sebire NJ. Renal Pathology of Ciliopathies. Pediatr Dev Pathol 2024; 27:411-425. [PMID: 38616607 DOI: 10.1177/10935266241242173] [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] [Indexed: 04/16/2024]
Abstract
Renal ciliopathies are a group of genetic disorders that affect the function of the primary cilium in the kidney, as well as other organs. Since primary cilia are important for regulation of cell signaling pathways, ciliary dysfunction results in a range of clinical manifestations, including renal failure, cyst formation, and hypertension. We summarize the current understanding of the pathophysiological and pathological features of renal ciliopathies in childhood, including autosomal dominant and recessive polycystic kidney disease, nephronophthisis, and Bardet-Biedl syndrome, as well as skeletal dysplasia associated renal ciliopathies. The genetic basis of these disorders is now well-established in many cases, with mutations in a large number of cilia-related genes such as PKD1, PKD2, BBS, MKS, and NPHP being responsible for the majority of cases. Renal ciliopathies are broadly characterized by development of interstitial fibrosis and formation of multiple renal cysts which gradually enlarge and replace normal renal tissue, with each condition demonstrating subtle differences in the degree, location, and age-related development of cysts and fibrosis. Presentation varies from prenatal diagnosis of congenital multisystem syndromes to an asymptomatic childhood with development of complications in later adulthood and therefore clinicopathological correlation is important, including increasing use of targeted genetic testing or whole genome sequencing, allowing greater understanding of genetic pathophysiological mechanisms.
Collapse
Affiliation(s)
- Thivya Sekar
- Histopathology Department, Level 3 CBL Labs, Great Ormond Street Hospital, London, UK
| | - Neil J Sebire
- Histopathology Department, Level 3 CBL Labs, Great Ormond Street Hospital, London, UK
| |
Collapse
|
3
|
Qian C, Yan J, Huang X, Wang Z, Lin F. Novel splice site and nonsense variants in PKHD1 cause autosomal recessive polycystic kidney disease in a Chinese Zhuang ethnic family. Medicine (Baltimore) 2024; 103:e39216. [PMID: 39093746 PMCID: PMC11296461 DOI: 10.1097/md.0000000000039216] [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: 12/28/2023] [Accepted: 07/17/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND This study aims to report the clinical characteristics of a child with autosomal recessive polycystic kidney disease (ARPKD) within a Chinese Zhuang ethnic family. METHODS We used whole exome sequencing (WES) in the family to examine the genetic cause of the disease. Candidate pathogenic variants were validated by Sanger sequencing. RESULTS We identified previously unreported mutations in the PKHD1 gene of the proband with ARPKD through WES: a splice site mutation c.6809-2A > T, a nonsense mutation c.4192C > T(p.Gln1398Ter), and a missense mutation c.2181T > G(p.Asn727Lys). Her mother is a heterozygous carrier of c.2181T > G(p.Asn727Lys) mutation. Her father is a carrier of c.6809-2A > T mutation and c.4192C > T(p.Gln1398Ter) mutation. CONCLUSIONS The identification of novel mutations in the PKHD1 gene through WES not only expands the spectrum of known variants but also potentially enhances genetic counseling and prenatal diagnostic approaches for families affected by ARPKD.
Collapse
Affiliation(s)
- Chen Qian
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Jie Yan
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Ximei Huang
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Zila Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Faquan Lin
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| |
Collapse
|
4
|
Alhaddad ME, Mohammad A, Dashti KM, John SE, Bahbahani Y, Abu-Farha M, Abubaker J, Thanaraj TA, Bastaki L, Al-Mulla F, Al-Ali M, Ali H. Genetic landscape and clinical outcomes of autosomal recessive polycystic kidney disease in Kuwait. Heliyon 2024; 10:e33898. [PMID: 39071699 PMCID: PMC11282974 DOI: 10.1016/j.heliyon.2024.e33898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/30/2024] Open
Abstract
Background Autosomal recessive polycystic kidney disease (ARPKD), a rare genetic disorder characterized by kidney cysts, shows complex clinical and genetic heterogeneity. This study aimed to explore the genetic landscape of ARPKD in Kuwait and examine the intricate relationship between its genes and clinical presentation to enhance our understanding and contribute towards more efficient management strategies for ARPKD. Methods This study recruited 60 individuals with suspected ARPKD from 44 different families in Kuwait. The participants were of different ethnicities and aged 0-70 years. Additionally, 33 were male, 15 were female, and 12 had indeterminant sex due to congenital anomalies. Comprehensive clinical data were collected. Mutations were identified by next-generation whole exome sequencing and confirmed using Sanger sequencing. Results Of the 60 suspected ARPKD cases, 20 (33.3 %) died within hours of birth or by the end of the first month of life and one (1.7 %) within 12 months of birth. The remaining 39 (65.0 %) cases were alive, at the time of the study, and exhibited diverse clinical features related to ARPKD, including systematic hypertension (5.0 %), pulmonary hypoplasia (11.7 %), dysmorphic features (40.0 %), cardiac problems (8.3 %), cystic liver (5.0 %), Potter syndrome (13.3 %), developmental delay (8.3 %), and enlarged cystic kidneys (100 %). Twelve mutations, including novel truncating mutations, were identified in 31/60 cases (51.7 %) from 17/44 families (38.6 %). Additionally, 8/12 (66.7 %) mutations were in the PKHD1 gene, with the remaining four in different genes: NPHP3, VPS13P, CC2D2A, and ZNF423. Conclusions This study highlights the spectrum of clinical features and genetic mutations of patients with ARPKD in Kuwait. It highlights the necessity for personalized approaches to improve ARPKD diagnosis and treatment, offering crucial insights into managing ARPKD.
Collapse
Affiliation(s)
- Mariam E. Alhaddad
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, Health Sciences Center (HSC), Kuwait University, Jabriya, Kuwait
| | - Anwar Mohammad
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute (DDI), Dasman, Kuwait
| | - Khadija M. Dashti
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, Health Sciences Center (HSC), Kuwait University, Jabriya, Kuwait
| | - Sumi Elsa John
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute (DDI), Dasman, Kuwait
| | - Yousif Bahbahani
- Division of Nephrology, Mubarak Al-Kabeer Hospital, Ministry of Health, Jabriya, Kuwait
| | - Mohamed Abu-Farha
- Next Generation Sequencing Laboratory, Kuwait Medical Genetics Center, Ministry of Health, Sulaibikhat, Kuwait
| | - Jehad Abubaker
- Next Generation Sequencing Laboratory, Kuwait Medical Genetics Center, Ministry of Health, Sulaibikhat, Kuwait
| | | | - Laila Bastaki
- Next Generation Sequencing Laboratory, Kuwait Medical Genetics Center, Ministry of Health, Sulaibikhat, Kuwait
| | - Fahd Al-Mulla
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute (DDI), Dasman, Kuwait
| | - Mohammad Al-Ali
- Next Generation Sequencing Laboratory, Kuwait Medical Genetics Center, Ministry of Health, Sulaibikhat, Kuwait
| | - Hamad Ali
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, Health Sciences Center (HSC), Kuwait University, Jabriya, Kuwait
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute (DDI), Dasman, Kuwait
| |
Collapse
|
5
|
Na DH, Cui S, Fang X, Lee H, Eum SH, Shin YJ, Lim SW, Yang CW, Chung BH. Advancements in Research on Genetic Kidney Diseases Using Human-Induced Pluripotent Stem Cell-Derived Kidney Organoids. Cells 2024; 13:1190. [PMID: 39056771 PMCID: PMC11274677 DOI: 10.3390/cells13141190] [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: 05/14/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Genetic or hereditary kidney disease stands as a pivotal cause of chronic kidney disease (CKD). The proliferation and widespread utilization of DNA testing in clinical settings have notably eased the diagnosis of genetic kidney diseases, which were once elusive but are now increasingly identified in cases previously deemed CKD of unknown etiology. However, despite these diagnostic strides, research into disease pathogenesis and novel drug development faces significant hurdles, chiefly due to the dearth of appropriate animal models and the challenges posed by limited patient cohorts in clinical studies. Conversely, the advent and utilization of human-induced pluripotent stem cells (hiPSCs) offer a promising avenue for genetic kidney disease research. Particularly, the development of hiPSC-derived kidney organoid systems presents a novel platform for investigating various forms of genetic kidney diseases. Moreover, the integration of the CRISPR/Cas9 technique into this system holds immense potential for efficient research on genetic kidney diseases. This review aims to explore the applications of in vitro kidney organoids generated from hiPSCs in the study of diverse genetic kidney diseases. Additionally, it will delve into the limitations of this research platform and outline future perspectives for advancing research in this crucial area.
Collapse
Affiliation(s)
- Do Hyun Na
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sheng Cui
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
| | - Xianying Fang
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
| | - Hanbi Lee
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sang Hun Eum
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
- Division of Nephrology, Department of Internal Medicine, Incheon St. Mary’s Hospital, The College of Medicine, The Catholic University of Korea, Incheon 21431, Republic of Korea
| | - Yoo Jin Shin
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
| | - Sun Woo Lim
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
| | - Chul Woo Yang
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Byung Ha Chung
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
6
|
Zhang X, Wu J, Zhou J, Liang J, Han Y, Qi Y, Zhu T, Yuan D, Zhu Z, Zhai J. Pathogenic relationship between phenotypes of ARPKD and novel compound heterozygous mutations of PKHD1. Front Genet 2024; 15:1429336. [PMID: 39015774 PMCID: PMC11250243 DOI: 10.3389/fgene.2024.1429336] [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: 05/08/2024] [Accepted: 06/10/2024] [Indexed: 07/18/2024] Open
Abstract
Background To investigate whether the novel mutation of PKHD1 could cause polycystic kidney disease by affecting splicing with a recessive inheritance pattern. Methods A nonconsanguineous Chinese couple with two recurrent pregnancies showed fetal enlarged echogenic polycystic kidney and oligoamnios were recruited. Pedigree WES, minigene splicing assay experiment and following bioinformatics analysis were performed to verify the effects, and inheritance pattern of diseasing-causing mutations. Results WES revealed that both fetuses were identified as carrying the same novel mutation c.3592_3628 + 45del, p.? and c.11207 T>C, p.(Ile3736Thr) in the PKHD1 gene (NM_138694.4), which inherited from the father and mother respectively. Both bioinformatic method prediction and minigene splicing assay experience results supported the mutation c.3592_3628 + 45del, p.? affects the splicing of the PKHD1 transcript, resulting in exon 31 skipping. Another missense mutation c.11207 T>C, p.(Ile3736Thr) has a low frequency in populations and is predicted to be deleterious by bioinformatic methods. Conclusion These findings provide a direct clinical and functional evidence that the truncating mutations of the PKHD1 gene could lead to more severe phenotypes, and cause ARPKD as a homozygous or compound heterozygous pattern. Our study broadens the variant spectrum of the PKHD1 gene and provides a basis for genetic counseling and diagnosis of ARPKD.
Collapse
Affiliation(s)
- Xinrong Zhang
- Xuzhou Central Hospital, Xuzhou Clinical College of Xuzhou Medical University, Xuzhou, China
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Prenatal Diagnosis Medical Center, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
| | - Jiebin Wu
- Xuzhou Central Hospital, Xuzhou Clinical College of Xuzhou Medical University, Xuzhou, China
- Department of Prenatal Diagnosis Medical Center, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
| | - Jianteng Zhou
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jie Liang
- Xuzhou Central Hospital, Xuzhou Clinical College of Xuzhou Medical University, Xuzhou, China
- Department of Prenatal Diagnosis Medical Center, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
- Key Laboratory of Brain Diseases Bioinformation of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yu Han
- Xuzhou Central Hospital, Xuzhou Clinical College of Xuzhou Medical University, Xuzhou, China
- Department of Prenatal Diagnosis Medical Center, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
- Key Laboratory of Brain Diseases Bioinformation of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yunmeng Qi
- Xuzhou Central Hospital, Xuzhou Clinical College of Xuzhou Medical University, Xuzhou, China
- Department of Prenatal Diagnosis Medical Center, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
- Key Laboratory of Brain Diseases Bioinformation of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Tao Zhu
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dejian Yuan
- Department of Medical Genetics, Liuzhou Municipal Maternity and Child Healthcare Hospital, Liuzhou, China
| | - Zuobin Zhu
- Xuzhou Central Hospital, Xuzhou Clinical College of Xuzhou Medical University, Xuzhou, China
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jingfang Zhai
- Xuzhou Central Hospital, Xuzhou Clinical College of Xuzhou Medical University, Xuzhou, China
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Prenatal Diagnosis Medical Center, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
- Key Laboratory of Brain Diseases Bioinformation of Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
7
|
Zhou W, Du Q, Liu Q, Liu X, Li L, Zhang H. A case report of autosomal recessive polycystic kidney disease with noncompaction of ventricular myocardium: coincidence or different manifestations of ciliopathy? BMC Nephrol 2024; 25:209. [PMID: 38918687 PMCID: PMC11201303 DOI: 10.1186/s12882-024-03642-7] [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: 07/04/2023] [Accepted: 06/17/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND Autosomal recessive polycystic kidney disease (ARPKD) is a rare inherited cystic disease characterized by bilateral renal cyst formation and congenital liver fibrosis. Cardiovascular disorders such as noncompaction of ventricular myocardium (NVM) have not been reported with ARPKD. CASE PRESENTATION A 5-month-old girl was examined after presenting with a fever and turbid urine for one day and was diagnosed as urinary tract infection. Urinary ultrasound showed multiple round, small cysts varying in size in both kidneys. Genetic testing revealed two heterozygous mutations and one exon deletion in the polycystic kidney and hepatic disease 1 gene, indicating a diagnosis of ARPKD. During hospitalization, she was found to have chronic heart failure after respiratory tract infection, with an ejection fraction of 29% and fraction shortening of 13%. When the patient was 15 months old, it was found that she had prominent trabeculations and deep intertrabecular recesses with the appearance of blood flow from the ventricular cavity into the intertrabecular recesses by echocardiography. The noncompaction myocardium was 0.716 cm and compaction myocardium was 0.221 cm (N/C = 3.27), indicating a diagnosis of NVM. Liver and kidney function remained normal during four-year follow-up. CONCLUSIONS This is the first report of NVM in a patient with ARPKD. It is unsure if the coexistence of NVM and ARPKD is a coincidence or they are different manifestations of ciliary dysfunction in the heart and kidneys.
Collapse
Affiliation(s)
- Weiran Zhou
- Department of Pediatric Nephrology and Rheumatism and Immunology, Children's Hospital Affiliated to Shandong University, Jinan, Shandong Province, China
- Department of Pediatric Nephrology and Rheumatism and Immunology, Jinan Children's Hospital, Jinan, Shandong Province, China
| | - Qingxia Du
- Department of Cardiovascular Medicine, Children's Hospital Affiliated to Shandong University, Jinan, Shandong Province, China.
| | - Qinghua Liu
- Department of Ultrasonography, Children's Hospital Affiliated to Shandong University, Jinan, Shandong Province, China
| | - Xiaofang Liu
- Department of Ultrasonography, Children's Hospital Affiliated to Shandong University, Jinan, Shandong Province, China
| | - Lei Li
- Department of Cardiovascular Medicine, Children's Hospital Affiliated to Shandong University, Jinan, Shandong Province, China
| | - Hongxia Zhang
- Department of Pediatric Nephrology and Rheumatism and Immunology, Children's Hospital Affiliated to Shandong University, Jinan, Shandong Province, China.
- Department of Pediatric Nephrology and Rheumatism and Immunology, Jinan Children's Hospital, Jinan, Shandong Province, China.
| |
Collapse
|
8
|
Ghaedi H, Davey SK, Feilotter H. Variant Classification Discordance: Contributing Factors and Predictive Models. J Mol Diagn 2024; 26:115-126. [PMID: 38008287 DOI: 10.1016/j.jmoldx.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/04/2023] [Accepted: 11/07/2023] [Indexed: 11/28/2023] Open
Abstract
An ever-growing catalog of human variants is hosted in the ClinVar database. In this database, submissions on a variant are combined into a multisubmitter record; and in the case of discordance in variant classification between submitters, the record is labeled as conflicting. The current study used ClinVar data to identify characteristics that would make variants more likely to be associated with the conflict class of variants. Furthermore, the Extreme Gradient Boosting algorithm was used to train classifier models to provide prediction of classification discordance for single submission variants in ClinVar database. Population allele frequency, the gene harboring the variant, variant type, consequence on protein, variant deleteriousness score, first submitter identity, and submission count were associated with conflict in variant classification. Using such features, the optimized classifier showed accuracy on the test set of 88% with the weighted average of precision, recall, and f1-score of 0.84, 0.88, and 0.85, respectively. There were pronounced associations between variant classification discordance and allele frequency, gene type, and the identity of the first submitter. The study provides the predicted discordance status for single-submitter variants deposited in ClinVar. This approach can be used to assess whether single-submitter variants are likely to be supported, or in conflict with, future entries; this knowledge may help laboratories with clinical variant assessment.
Collapse
Affiliation(s)
- Hamid Ghaedi
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Scott K Davey
- Division of Cancer Biology and Genetics, Department of Pathology and Molecular Medicine, Queen's University Cancer Research Institute, Kingston, Ontario, Canada
| | - Harriet Feilotter
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
9
|
Satariano M, Ghose S, Raina R. The Pathophysiology of Inherited Renal Cystic Diseases. Genes (Basel) 2024; 15:91. [PMID: 38254980 PMCID: PMC10815569 DOI: 10.3390/genes15010091] [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: 12/06/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Renal cystic diseases (RCDs) can arise from utero to early adulthood and present with a variety of symptoms including renal, hepatic, and cardiovascular manifestations. It is well known that common RCDs such as autosomal polycystic kidney disease and autosomal recessive kidney disease are linked to genes such as PKD1 and PKHD1, respectively. However, it is important to investigate the genetic pathophysiology of how these gene mutations lead to clinical symptoms and include some of the less-studied RCDs, such as autosomal dominant tubulointerstitial kidney disease, multicystic dysplastic kidney, Zellweger syndrome, calyceal diverticula, and more. We plan to take a thorough look into the genetic involvement and clinical sequalae of a number of RCDs with the goal of helping to guide diagnosis, counseling, and treatment.
Collapse
Affiliation(s)
- Matthew Satariano
- Department of Medicine, Northeast Ohio Medical University, Rootstown, OH 44272, USA; (M.S.); (S.G.)
| | - Shaarav Ghose
- Department of Medicine, Northeast Ohio Medical University, Rootstown, OH 44272, USA; (M.S.); (S.G.)
| | - Rupesh Raina
- Akron Nephrology Associates, Cleveland Clinic Akron General Medical Center, Akron, OH 44307, USA
- Department of Nephrology, Akron Children’s Hospital, Akron, OH 44308, USA
| |
Collapse
|
10
|
Harafuji N, Yang C, Wu M, Thiruvengadam G, Gordish-Dressman H, Thompson RG, Bell PD, Rosenberg AZ, Dafinger C, Liebau MC, Bebok Z, Caldovic L, Guay-Woodford LM. Differential regulation of MYC expression by PKHD1/Pkhd1 in human and mouse kidneys: phenotypic implications for recessive polycystic kidney disease. Front Cell Dev Biol 2023; 11:1270980. [PMID: 38125876 PMCID: PMC10731465 DOI: 10.3389/fcell.2023.1270980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/30/2023] [Indexed: 12/23/2023] Open
Abstract
Autosomal recessive polycystic kidney disease (ARPKD; MIM#263200) is a severe, hereditary, hepato-renal fibrocystic disorder that leads to early childhood morbidity and mortality. Typical forms of ARPKD are caused by pathogenic variants in the PKHD1 gene, which encodes the fibrocystin/polyductin (FPC) protein. MYC overexpression has been proposed as a driver of renal cystogenesis, but little is known about MYC expression in recessive PKD. In the current study, we provide the first evidence that MYC is overexpressed in kidneys from ARPKD patients and confirm that MYC is upregulated in cystic kidneys from cpk mutant mice. In contrast, renal MYC expression levels were not altered in several Pkhd1 mutant mice that lack a significant cystic kidney phenotype. We leveraged previous observations that the carboxy-terminus of mouse FPC (FPC-CTD) is proteolytically cleaved through Notch-like processing, translocates to the nucleus, and binds to double stranded DNA, to examine whether the FPC-CTD plays a role in regulating MYC/Myc transcription. Using immunofluorescence, reporter gene assays, and ChIP, we demonstrate that both human and mouse FPC-CTD can localize to the nucleus, bind to the MYC/Myc P1 promoter, and activate MYC/Myc expression. Interestingly, we observed species-specific differences in FPC-CTD intracellular trafficking. Furthermore, our informatic analyses revealed limited sequence identity of FPC-CTD across vertebrate phyla and database queries identified temporal differences in PKHD1/Pkhd1 and CYS1/Cys1 expression patterns in mouse and human kidneys. Given that cystin, the Cys1 gene product, is a negative regulator of Myc transcription, these temporal differences in gene expression could contribute to the relative renoprotection from cystogenesis in Pkhd1-deficient mice. Taken together, our findings provide new mechanistic insights into differential mFPC-CTD and hFPC-CTD regulation of MYC expression in renal epithelial cells, which may illuminate the basis for the phenotypic disparities between human patients with PKHD1 pathogenic variants and Pkhd1-mutant mice.
Collapse
Affiliation(s)
- Naoe Harafuji
- Center for Translational Research, Children’s National Hospital, Washington, DC, United States
| | - Chaozhe Yang
- Center for Translational Research, Children’s National Hospital, Washington, DC, United States
| | - Maoqing Wu
- Center for Translational Research, Children’s National Hospital, Washington, DC, United States
| | - Girija Thiruvengadam
- Center for Translational Research, Children’s National Hospital, Washington, DC, United States
| | | | - R. Griffin Thompson
- Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - P. Darwin Bell
- Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Avi Z. Rosenberg
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Claudia Dafinger
- Department of Pediatrics and Center for Molecular Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Max C. Liebau
- Department of Pediatrics, Center for Family Health, Center for Rare Diseases and Center for Molecular Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Zsuzsanna Bebok
- Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ljubica Caldovic
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, United States
- Department of Genomics and Precision Medicine, School of Medical and Health Sciences, The George Washington University, Washington, DC, United States
| | - Lisa M. Guay-Woodford
- Center for Translational Research, Children’s National Hospital, Washington, DC, United States
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, United States
| |
Collapse
|
11
|
Kocaaga A, Atikel YÖ, Sak M, Karakaya T. The genetic spectrum of polycystic kidney disease in children. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2023; 69:e20230334. [PMID: 37909612 PMCID: PMC10610762 DOI: 10.1590/1806-9282.20230334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 08/11/2023] [Indexed: 11/03/2023]
Abstract
OBJECTIVE Autosomal dominant polycystic kidney disease is an inherited kidney disorder with mutations in polycystin-1 or polycystin-2. Autosomal recessive polycystic kidney disease is a severe form of polycystic kidney disease that is characterized by enlarged kidneys and congenital hepatic fibrosis. Mutations at PKHD1 are responsible for all typical forms of autosomal recessive polycystic kidney disease. METHODS We evaluated the children diagnosed with polycystic kidney disease between October 2020 and May 2022. The diagnosis was established by family history, ultrasound findings, and/or genetic analysis. The demographic, clinical, and laboratory findings were evaluated retrospectively. RESULTS There were 28 children (male/female: 11:17) evaluated in this study. Genetic analysis was performed in all patients (polycystin-1 variants in 13, polycystin-2 variants in 7, and no variants in 8 patients). A total of 18 variants in polycystin-1 and polycystin-2 were identified and 9 (50%) of them were not reported before. A total of eight novel variants were identified as definite pathogenic or likely pathogenic mutations. There was no variant detected in the PKDH1 gene. CONCLUSION Our results highlighted molecular features of Turkish children with polycystic kidney disease and demonstrated novel variations that can be utilized in clinical diagnosis and prognosis.
Collapse
Affiliation(s)
- Ayca Kocaaga
- Eskisehir City Hospital, Department of Medical Genetics – Eskişehir, Turkey
| | | | - Mehtap Sak
- Isparta City Hospital, Department of Pediatric Nephrology – Isparta, Turkey
| | - Taner Karakaya
- Isparta City Hospital, Department of Medical Genetics, – Isparta, Turkey
| |
Collapse
|
12
|
De Groof J, Dachy A, Breysem L, Mekahli D. Cystic kidney diseases in children. Arch Pediatr 2023; 30:240-246. [PMID: 37062654 DOI: 10.1016/j.arcped.2023.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/24/2022] [Accepted: 02/12/2023] [Indexed: 04/18/2023]
Abstract
Cystic kidney disease comprises a broad group of heterogeneous diseases, which differ greatly in age at onset, disease manifestation, systemic involvement, disease progression, and long-term prognosis. As our understanding of these diseases continues to evolve and new treatment strategies continue to emerge, correctly differentiating and diagnosing these diseases becomes increasingly important. In this review, we aim to highlight the key features of the most relevant cystic kidney diseases, underscore important diagnostic characteristics of each disease, and present specific management options if applicable.
Collapse
Affiliation(s)
- J De Groof
- Department of Pediatric Nephrology, University Hospitals Leuven, Leuven, Belgium
| | - A Dachy
- PKD Research Group, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; Department of Pediatrics, ULiège Academic Hospital, Liège, Belgium
| | - L Breysem
- Department of Pediatric Radiology, University Hospitals Leuven, Leuven, Belgium
| | - D Mekahli
- Department of Pediatric Nephrology, University Hospitals Leuven, Leuven, Belgium; PKD Research Group, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.
| |
Collapse
|
13
|
Yoo JJ, Jo HI, Jung EA, Lee JS, Kim SG, Kim YS, Kim BK. Evidence of nonsurgical treatment for polycystic liver disease. Ther Adv Chronic Dis 2022; 13:20406223221112563. [PMID: 35898920 PMCID: PMC9310217 DOI: 10.1177/20406223221112563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 06/21/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Polycystic liver disease (PCLD) is the most common extrarenal manifestation
of polycystic kidney disease. There is an urgent need to assess the efficacy
and safety of nonsurgical modalities to relieve symptoms and decrease the
severity of PCLD. Herein, we aimed to evaluate the efficacy of the
nonsurgical treatment of PCLD and the quality of life of affected
patients. Methods: PubMed, Ovid, MEDLINE, EMBASE, and the Cochrane Library were searched for
studies on the nonsurgical modalities, either medications or radiological
intervention to manage PCLD. Treatment efficacy, adverse events (AEs), and
patient quality of life were evaluated. Results: In total, 27 studies involving 1037 patients were selected. After nonsurgical
treatment, liver volume decreased by 259 ml/m [mean change (Δ) of 6.22%] and
the effect was higher in the radiological intervention group [−1617 ml/m
(−15.49%)] than in the medication group [−151 ml/m (−3.78%)]. The AEs and
serious AEs rates after overall nonsurgical treatment were 0.50 [95%
confidence interval (CI): 0.33–0.67] and 0.04 (95% CI: 0.01–0.07),
respectively. The results of the SF-36 questionnaire showed that PCLD
treatment improved physical function [physical component summary score of
4.18 (95% CI: 1.54–6.83)] but did not significantly improve mental function
[mental component summary score of 0.91 (95% CI: −1.20 to 3.03)]. Conclusion: Nonsurgical treatment was effective and safe for PCLD, but did not improve
the quality of life in terms of mental health. Radiological intervention
directly reduces hepatic cysts, and thus they should be considered for
immediate symptom relief in patients with severe symptoms, whereas
medication might be considered for maintenance treatment. Registration number: PROSPERO (International Prospective Register of Systematic Reviews)
CRD42021279597
Collapse
Affiliation(s)
- Jeong-Ju Yoo
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, College of Medicine, Soonchunhyang University, Bucheon, Republic of Korea
| | - Hye In Jo
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, College of Medicine, Soonchunhyang University, Bucheon, Republic of Korea
| | - Eun-Ae Jung
- Medical Library, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | - Jae Seung Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sang Gyune Kim
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, College of Medicine, Soonchunhyang University, Bucheon, Republic of Korea
| | - Young Seok Kim
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, College of Medicine, Soonchunhyang University, Bucheon, Republic of Korea
| | - Beom Kyung Kim
- Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| |
Collapse
|
14
|
Rare Monogenic Diseases: Molecular Pathophysiology and Novel Therapies. Int J Mol Sci 2022; 23:ijms23126525. [PMID: 35742964 PMCID: PMC9223693 DOI: 10.3390/ijms23126525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/07/2022] [Indexed: 12/10/2022] Open
Abstract
A rare disease is defined by its low prevalence in the general population [...].
Collapse
|
15
|
Kumar P, Zadjali F, Yao Y, Köttgen M, Hofherr A, Gross KW, Mehta D, Bissler JJ. Single Gene Mutations in Pkd1 or Tsc2 Alter Extracellular Vesicle Production and Trafficking. BIOLOGY 2022; 11:biology11050709. [PMID: 35625437 PMCID: PMC9139108 DOI: 10.3390/biology11050709] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/20/2022] [Accepted: 04/30/2022] [Indexed: 12/17/2022]
Abstract
Simple Summary Extracellular vesicles shed from primary cilia may be involved in renal cystogenesis. The disruption of the Pkd1 gene in our cell culture system increased the production of EVs in a similar way that occurs when the Tsc2 gene is disrupted. Disruption of the primary cilia depresses EV production, and this may be the reason that the combined Kif3A/Pkd1 mutant mouse has a less severe phenotype than the Pkd1 mutant alone. We initiated studies aimed at understanding the renal trafficking of renally-derived EVs and found that single gene disruptions can alter the EV kinetics based on dye tracking studies. These results raise the possibility that EV features, such as cargo, dose, tissue half-life, and targeting, may be involved in the disease process, and these features may also be fertile targets for diagnostic, prognostic, and therapeutic investigation. Abstract Patients with autosomal dominant polycystic kidney disease (ADPKD) and tuberous sclerosis complex (TSC) are born with normal or near-normal kidneys that later develop cysts and prematurely lose function. Both renal cystic diseases appear to be mediated, at least in part, by disease-promoting extracellular vesicles (EVs) that induce genetically intact cells to participate in the renal disease process. We used centrifugation and size exclusion chromatography to isolate the EVs for study. We characterized the EVs using tunable resistive pulse sensing, dynamic light scattering, transmission electron microscopy, and Western blot analysis. We performed EV trafficking studies using a dye approach in both tissue culture and in vivo studies. We have previously reported that loss of the Tsc2 gene significantly increased EV production and here demonstrate that the loss of the Pkd1 gene also significantly increases EV production. Using a cell culture system, we also show that loss of either the Tsc2 or Pkd1 gene results in EVs that exhibit an enhanced uptake by renal epithelial cells and a prolonged half-life. Loss of the primary cilia significantly reduces EV production in renal collecting duct cells. Cells that have a disrupted Pkd1 gene produce EVs that have altered kinetics and a prolonged half-life, possibly impacting the duration of the EV cargo effect on the recipient cell. These results demonstrate the interplay between primary cilia and EVs and support a role for EVs in polycystic kidney disease pathogenesis.
Collapse
Affiliation(s)
- Prashant Kumar
- Department of Pediatrics, Le Bonheur Children’s Hospital, University of Tennessee Health Science Center, Memphis, TN 38103, USA; (P.K.); (F.Z.); (Y.Y.)
- Children’s Foundation Research Institute (CFRI), Le Bonheur Children’s Hospital, Memphis, TN 38103, USA
- US FDA National Center for Toxicological Research, Jefferson, AR 72079, USA;
| | - Fahad Zadjali
- Department of Pediatrics, Le Bonheur Children’s Hospital, University of Tennessee Health Science Center, Memphis, TN 38103, USA; (P.K.); (F.Z.); (Y.Y.)
- Children’s Foundation Research Institute (CFRI), Le Bonheur Children’s Hospital, Memphis, TN 38103, USA
- Department of Clinical Biochemistry, College of Medicine & Health Sciences, Sultan Qaboos University, Muscat 123, Oman
| | - Ying Yao
- Department of Pediatrics, Le Bonheur Children’s Hospital, University of Tennessee Health Science Center, Memphis, TN 38103, USA; (P.K.); (F.Z.); (Y.Y.)
- Children’s Foundation Research Institute (CFRI), Le Bonheur Children’s Hospital, Memphis, TN 38103, USA
| | - Michael Köttgen
- Renal Division, Department of Medicine, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (M.K.); (A.H.)
- CIBSS—Centre for Integrative Biological Signaling Studies, 79104 Freiburg, Germany
| | - Alexis Hofherr
- Renal Division, Department of Medicine, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (M.K.); (A.H.)
| | - Kenneth W. Gross
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| | - Darshan Mehta
- US FDA National Center for Toxicological Research, Jefferson, AR 72079, USA;
| | - John J. Bissler
- Department of Pediatrics, Le Bonheur Children’s Hospital, University of Tennessee Health Science Center, Memphis, TN 38103, USA; (P.K.); (F.Z.); (Y.Y.)
- Children’s Foundation Research Institute (CFRI), Le Bonheur Children’s Hospital, Memphis, TN 38103, USA
- Pediatric Medicine Department, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Correspondence:
| |
Collapse
|
16
|
Genetic Kidney Diseases (GKDs) Modeling Using Genome Editing Technologies. Cells 2022; 11:cells11091571. [PMID: 35563876 PMCID: PMC9105797 DOI: 10.3390/cells11091571] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/29/2022] [Accepted: 05/04/2022] [Indexed: 02/05/2023] Open
Abstract
Genetic kidney diseases (GKDs) are a group of rare diseases, affecting approximately about 60 to 80 per 100,000 individuals, for which there is currently no treatment that can cure them (in many cases). GKDs usually leads to early-onset chronic kidney disease, which results in patients having to undergo dialysis or kidney transplant. Here, we briefly describe genetic causes and phenotypic effects of six GKDs representative of different ranges of prevalence and renal involvement (ciliopathy, glomerulopathy, and tubulopathy). One of the shared characteristics of GKDs is that most of them are monogenic. This characteristic makes it possible to use site-specific nuclease systems to edit the genes that cause GKDs and generate in vitro and in vivo models that reflect the genetic abnormalities of GKDs. We describe and compare these site-specific nuclease systems (zinc finger nucleases (ZFNs), transcription activator-like effect nucleases (TALENs) and regularly clustered short palindromic repeat-associated protein (CRISPR-Cas9)) and review how these systems have allowed the generation of cellular and animal GKDs models and how they have contributed to shed light on many still unknown fields in GKDs. We also indicate the main obstacles limiting the application of these systems in a more efficient way. The information provided here will be useful to gain an accurate understanding of the technological advances in the field of genome editing for GKDs, as well as to serve as a guide for the selection of both the genome editing tool and the gene delivery method most suitable for the successful development of GKDs models.
Collapse
|
17
|
Cordido A, Vizoso-Gonzalez M, Nuñez-Gonzalez L, Molares-Vila A, Chantada-Vazquez MDP, Bravo SB, Garcia-Gonzalez MA. Quantitative Proteomic Study Unmasks Fibrinogen Pathway in Polycystic Liver Disease. Biomedicines 2022; 10:290. [PMID: 35203500 PMCID: PMC8869147 DOI: 10.3390/biomedicines10020290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 02/05/2023] Open
Abstract
(1) Background: Polycystic liver disease (PLD) is a heterogeneous group of congenital disorders characterized by bile duct dilatation and cyst development derived from cholangiocytes. Nevertheless, the cystogenesis mechanism is currently unknown and the PLD treatment is limited to liver transplantation. Novel and efficient therapeutic approaches are th6us needed. In this context, the present work has a principal aim to find novel molecular pathways, as well as new therapeutic targets, involved in the hepatic cystogenesis process. (2) Methods: Quantitative proteomics based on SWATH-MS technology were performed comparing hepatic proteomes of Wild Type and mutant/polycystic livers in a polycystic kidney disease (PKD) murine model (Pkd1cond/cond;Tam-Cre-/+). (3) Results: We identified several proteins altered in abundance, with two-fold cut-off up-regulation or down-regulation and an adjusted p-value significantly related to hepatic cystogenesis. Then, we performed enrichment and a protein-protein analysis identifying a cluster focused on hepatic fibrinogens. Finally, we validated a selection of targets by RT-qPCR, Western blotting and immunohistochemistry, finding a high correlation with quantitative proteomics data and validating the fibrinogen complex. (4) Conclusions: This work identified a novel molecular pathway in cystic liver disease, highlighting the fibrinogen complex as a possible new therapeutic target for PLD.
Collapse
Affiliation(s)
- Adrian Cordido
- Group of Genetics and Developmental Biology of Renal Diseases, Nephrology Laboratory (N°11), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela Clinical Hospital Complex (CHUS), 15706 Santiago de Compostela, Spain; (A.C.); (M.V.-G.); (L.N.-G.)
- Genomic Medicine Group, Santiago de Compostela Clinical Hospital Complex (CHUS), 15706 Santiago de Compostela, Spain
| | - Marta Vizoso-Gonzalez
- Group of Genetics and Developmental Biology of Renal Diseases, Nephrology Laboratory (N°11), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela Clinical Hospital Complex (CHUS), 15706 Santiago de Compostela, Spain; (A.C.); (M.V.-G.); (L.N.-G.)
- Genomic Medicine Group, Santiago de Compostela Clinical Hospital Complex (CHUS), 15706 Santiago de Compostela, Spain
| | - Laura Nuñez-Gonzalez
- Group of Genetics and Developmental Biology of Renal Diseases, Nephrology Laboratory (N°11), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela Clinical Hospital Complex (CHUS), 15706 Santiago de Compostela, Spain; (A.C.); (M.V.-G.); (L.N.-G.)
- Genomic Medicine Group, Santiago de Compostela Clinical Hospital Complex (CHUS), 15706 Santiago de Compostela, Spain
| | - Alberto Molares-Vila
- Biostatistics Platform, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela Clinical Hospital Complex (CHUS), 15706 Santiago de Compostela, Spain;
| | - Maria del Pilar Chantada-Vazquez
- Proteomic Platform, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela Clinical Hospital Complex (CHUS), 15706 Santiago de Compostela, Spain;
| | - Susana B. Bravo
- Proteomic Platform, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela Clinical Hospital Complex (CHUS), 15706 Santiago de Compostela, Spain;
| | - Miguel A. Garcia-Gonzalez
- Group of Genetics and Developmental Biology of Renal Diseases, Nephrology Laboratory (N°11), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela Clinical Hospital Complex (CHUS), 15706 Santiago de Compostela, Spain; (A.C.); (M.V.-G.); (L.N.-G.)
- Genomic Medicine Group, Santiago de Compostela Clinical Hospital Complex (CHUS), 15706 Santiago de Compostela, Spain
- Galician Public Foundation of Genomic Medicine, Santiago de Compostela Clinical Hospital Complex (CHUS), 15706 Santiago de Compostela, Spain
| |
Collapse
|
18
|
Goggolidou P, Richards T. The genetics of Autosomal Recessive Polycystic Kidney Disease (ARPKD). Biochim Biophys Acta Mol Basis Dis 2022; 1868:166348. [PMID: 35032595 DOI: 10.1016/j.bbadis.2022.166348] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/27/2021] [Accepted: 01/06/2022] [Indexed: 12/21/2022]
Abstract
ARPKD is a genetically inherited kidney disease that manifests by bilateral enlargement of cystic kidneys and liver fibrosis. It shows a range of severity, with 30% of individuals dying early on and the majority having good prognosis if they survive the first year of life. The reasons for this variability remain unclear. Two genes have been shown to cause ARPKD when mutated, PKHD1, mutations in which lead to most of ARPKD cases and DZIP1L, which is associated with moderate ARPKD. This mini review will explore the genetics of ARPKD and discuss potential genetic modifiers and phenocopies that could affect diagnosis.
Collapse
Affiliation(s)
- Paraskevi Goggolidou
- Faculty of Science and Engineering, University of Wolverhampton, Wulfruna Street, Wolverhampton WV1 1LY, UK.
| | - Taylor Richards
- Faculty of Science and Engineering, University of Wolverhampton, Wulfruna Street, Wolverhampton WV1 1LY, UK
| |
Collapse
|
19
|
Liebau MC. Is There a Functional Role of Mitochondrial Dysfunction in the Pathogenesis of ARPKD? Front Med (Lausanne) 2021; 8:739534. [PMID: 34676227 PMCID: PMC8523777 DOI: 10.3389/fmed.2021.739534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 09/03/2021] [Indexed: 12/02/2022] Open
Affiliation(s)
- Max Christoph Liebau
- Department of Pediatrics, Center for Molecular Medicine, and Center for Rare Diseases, University Hospital Cologne and Medical Faculty, University of Cologne, Cologne, Germany
| |
Collapse
|