1
|
Alotibi ANA, Aldayhani A, Almajed E, Alruwaily AF, Alzahrani A. Radical laparoscopic nephrectomy for a huge left renal mass in situs inversus totalis: A case report and video presentation. Urol Case Rep 2025; 60:103032. [PMID: 40242038 PMCID: PMC12002879 DOI: 10.1016/j.eucr.2025.103032] [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: 03/03/2025] [Revised: 03/28/2025] [Accepted: 03/30/2025] [Indexed: 04/18/2025] Open
Abstract
This case report describes the successful laparoscopic radical nephrectomy in a 69-year-old woman with situs inversus totalis and a large, hypervascular left renal mass. Despite the presence of metastatic disease, the patient underwent neoadjuvant immunotherapy followed by surgery. The successful laparoscopic approach highlights the feasibility of minimally invasive surgery even in patients with complex anatomical variations. This report provides valuable insights into the surgical management of renal tumors in individuals with situs inversus totalis.
Collapse
Affiliation(s)
| | | | - Ebtesam Almajed
- Department of Clinical Sciences, College of Medicine, Princess Nourah Bint, Abdulrahman University, Riyadh, Saudi Arabia
| | | | - Ahmed Alzahrani
- Department of Urology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| |
Collapse
|
2
|
Pîrlog LM, Pătrăşcanu AA, Kutasi E, Iordănescu I, Militaru MS. Primary ciliary dyskinesia: a case report of double DNAH11 mutant alleles. Med Pharm Rep 2025; 98:252-256. [PMID: 40371404 PMCID: PMC12070959 DOI: 10.15386/mpr-2743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/06/2024] [Accepted: 08/19/2024] [Indexed: 05/16/2025] Open
Abstract
Primary ciliary dyskinesia (PCD), a rare disorder, is genetically varied. Mutations in proteins involved in the structure, function, or assembly of cilia are known to determine situs inversus, male infertility, and chronic destructive airway disease. PCD is inherited by an autosomal recessive pattern of inheritance in most cases. Nonetheless, patterns of autosomal dominant and X-linked inheritance have been mentioned. A history of recurrent upper and lower respiratory tract infections raised clinical suspicion of primary ciliary dyskinesia in a 10-year-old patient. Genetic tests were performed using next-generation sequencing technology (Illumina NextGen) with the multiplex ligation-dependent probe amplification technique for primary ciliopathies and syndromes subject to differential diagnosis. Genetic testing identified two pathogenic variants, not previously associated with a case report in the literature, c.7727A>G (p.Asp2576Gly) and c.8578G>A (p.Gly2860Ser), within the DNAH11 gene, which is associated with autosomal recessive PCD. The result also reported mutations in other genes involved in autosomal recessive PCD (DNAH8, DNAH9 and ZMYND10), which were classified as variants with uncertain clinical significance. Transmission electron microscopy of respiratory cilia and nasal nitric oxide measurement cannot be used to diagnose PCD in patients with DNAH11 mutations because the structure of cilia is normal, and the levels of NO are not constantly low. High-speed video microscopy analysis can be helpful because DNAH11 mutations cause a distinct phenotype of PCD. Nevertheless, the mutation analysis of various PCD-causing genes remains the easiest to conduct and with good results. Genetic research on PCD has identified a number of significant ciliary genes in recent years, offering fresh perspectives on the molecular processes underlying cilia assembly and function. This facilitates the development of new methods for the diagnosis, prevention, and treatment of PCD. However, because it is a highly complex and heterogeneous disease, the field of gene diagnosis and therapy in PCD is still in its infancy.
Collapse
Affiliation(s)
- Lorin-Manuel Pîrlog
- Department of Molecular Sciences, Faculty of Medicine, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Andrada-Adelaida Pătrăşcanu
- Department of Molecular Sciences, Faculty of Medicine, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Eniko Kutasi
- Department of Molecular Sciences, Faculty of Medicine, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Irina Iordănescu
- Regional Laboratory Bucharest, Department of Medical Genetics, Regina Maria Health Network, Bucharest, Romania
| | - Mariela Sanda Militaru
- Department of Molecular Sciences, Faculty of Medicine, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
3
|
Szenker-Ravi E, Ott T, Yusof A, Chopra M, Khatoo M, Pak B, Xuan Goh W, Beckers A, Brady AF, Ewans LJ, Djaziri N, Almontashiri NAM, Alghamdi MA, Alharby E, Dasouki M, Romo L, Tan WH, Maddirevula S, Alkuraya FS, Giordano JL, Alkelai A, Wapner RJ, Stals K, Alfadhel M, Alswaid AF, Bogusch S, Schafer-Kosulya A, Vogel S, Vick P, Schweickert A, Wakeling M, Moreau de Bellaing A, Alshamsi AM, Sanlaville D, Mbarek H, Saad C, Ellard S, Eisenhaber F, Tripolszki K, Beetz C, Bauer P, Gossler A, Eisenhaber B, Blum M, Bouvagnet P, Bertoli-Avella A, Amiel J, Gordon CT, Reversade B. CIROZ is dispensable in ancestral vertebrates but essential for left-right patterning in humans. Am J Hum Genet 2025; 112:353-373. [PMID: 39753129 PMCID: PMC11866977 DOI: 10.1016/j.ajhg.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 02/09/2025] Open
Abstract
Four genes-DAND5, PKD1L1, MMP21, and CIROP-form a genetic module that has specifically evolved in vertebrate species that harbor motile cilia in their left-right organizer (LRO). We find here that CIROZ (previously known as C1orf127) is also specifically expressed in the LRO of mice, frogs, and fish, where it encodes a protein with a signal peptide followed by 3 zona pellucida N domains, consistent with extracellular localization. We report 16 individuals from 10 families with bi-allelic CIROZ inactivation variants, which cause heterotaxy with congenital heart defects. While the knockout of Ciroz in mice also leads to situs anomalies, we unexpectedly find that its targeted inactivation in zebrafish and Xenopus does not lead to observable LR anomalies. Moreover, CIROZ is absent or obsolete in select animals with motile cilia at their LRO, including Carnivora, Atherinomorpha fish, or jawless vertebrates. In summary, this evo-devo study identifies CIROZ as an essential gene for breaking bilateral embryonic symmetry in humans and mice, whereas we witness its contemporary pseudogenization in discrete vertebrate species.
Collapse
Affiliation(s)
- Emmanuelle Szenker-Ravi
- Laboratory of Human Genetics & Therapeutics, Genome Institute of Singapore (GIS), A(∗)STAR, Singapore, Singapore; Laboratory of Human Genetics & Therapeutics, BESE, KAUST, Thuwal, Saudi Arabia.
| | - Tim Ott
- Institute of Biology, University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany
| | - Amirah Yusof
- Laboratory of Human Genetics & Therapeutics, Genome Institute of Singapore (GIS), A(∗)STAR, Singapore, Singapore; Laboratory of Human Genetics & Therapeutics, BESE, KAUST, Thuwal, Saudi Arabia
| | - Maya Chopra
- Rosamund Stone Zander Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | - Muznah Khatoo
- Laboratory of Human Genetics & Therapeutics, Genome Institute of Singapore (GIS), A(∗)STAR, Singapore, Singapore
| | - Beatrice Pak
- Laboratory of Human Genetics & Therapeutics, Genome Institute of Singapore (GIS), A(∗)STAR, Singapore, Singapore
| | - Wei Xuan Goh
- Laboratory of Human Genetics & Therapeutics, Genome Institute of Singapore (GIS), A(∗)STAR, Singapore, Singapore
| | - Anja Beckers
- Institute for Molecular Biology, OE5250, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Angela F Brady
- North West Thames Regional Genetics Service, London North West University Healthcare NHS Trust, Northwick Park Hospital, Harrow HA1 3UJ, UK
| | - Lisa J Ewans
- Center for Clinical Genetics, Sydney Children's Hospitals Network Randwick, Discipline of Pediatrics and Child Health, Faculty of Medicine and Health, UNSW, Center for Community Genomics, the Garvan Institute, Sydney, NSW, Australia
| | - Nabila Djaziri
- Université de Paris, Imagine Institute, Laboratory of Embryology and Genetics of Malformations, INSERM UMR 1163, 75015 Paris, France
| | - Naif A M Almontashiri
- Center for Genetics and Inherited Diseases, Taibah University, Medina, Saudi Arabia; Faculty of Applied Medical Sciences, Taibah University, Medina, Saudi Arabia
| | - Malak Ali Alghamdi
- Medical Genetics Department, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Essa Alharby
- Center for Genetics and Inherited Diseases, Taibah University, Medina, Saudi Arabia
| | - Majed Dasouki
- AdventHealth Genomics & Personalized Health at Orlando, Department of Medical Genetics & Genomics, 601 E. Rollins St., Suite 125, Orlando, FL 32804, USA
| | - Lindsay Romo
- Boston Children's Hospital and Harvard Medical School, Division of Genetics and Genomics, Boston, MA, USA; Harvard Medical Genetics Training Program, Boston Children's Hospital, Boston, MA 02115, USA
| | - Wen-Hann Tan
- Boston Children's Hospital and Harvard Medical School, Division of Genetics and Genomics, Boston, MA, USA
| | - Sateesh Maddirevula
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Fowzan S Alkuraya
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Jessica L Giordano
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Anna Alkelai
- Institute for Genomic Medicine, Columbia University Medical Center, New York, NY, USA
| | - Ronald J Wapner
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Karen Stals
- Institute for Clinical and Biomedical Science, University of Exeter Medical School, Exeter, UK
| | - Majid Alfadhel
- Genetics and Precision Medicine Department King Abdullah Specialized Children Hospital (KASCH), King Abdulaziz Medical City (KAMC), MNG-HA, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdulaziz University for Health Sciences, MNG-HA, Riyadh, Saudi Arabia
| | - Abdulrahman Faiz Alswaid
- Genetics and Precision Medicine Department King Abdullah Specialized Children Hospital (KASCH), King Abdulaziz Medical City (KAMC), MNG-HA, Riyadh, Saudi Arabia
| | - Susanne Bogusch
- University of Hohenheim, Institute of Biology, Department of Zoology, Garbenstr. 30, 70593 Stuttgart, Germany
| | - Anna Schafer-Kosulya
- University of Hohenheim, Institute of Biology, Department of Zoology, Garbenstr. 30, 70593 Stuttgart, Germany
| | - Sebastian Vogel
- University of Hohenheim, Institute of Biology, Department of Zoology, Garbenstr. 30, 70593 Stuttgart, Germany
| | - Philipp Vick
- University of Hohenheim, Institute of Biology, Department of Zoology, Garbenstr. 30, 70593 Stuttgart, Germany
| | - Axel Schweickert
- University of Hohenheim, Institute of Biology, Department of Zoology, Garbenstr. 30, 70593 Stuttgart, Germany
| | - Matthew Wakeling
- Exeter Genomics Laboratory, Royal Devon & Exeter NHS Foundation Trust, Exeter, UK
| | - Anne Moreau de Bellaing
- Service de Génétique, Groupe Hospitalier Est, Hospices Civils de Lyon, Lyon, France; Université Lyon 1 Claude Bernard, Lyon, France
| | - Aisha M Alshamsi
- Department of Pediatrics, Tawam Hospital, Al-Ain, United Arab Emirates
| | - Damien Sanlaville
- Service de Génétique, Groupe Hospitalier Est, Hospices Civils de Lyon, Lyon, France; Université Lyon 1, CNRS, INSERM, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008 Lyon, France
| | - Hamdi Mbarek
- Qatar Genome Program, Qatar Foundation Research, Development and Innovation, Qatar Foundation, Doha, Qatar
| | - Chadi Saad
- Qatar Genome Program, Qatar Foundation Research, Development and Innovation, Qatar Foundation, Doha, Qatar
| | - Sian Ellard
- Institute for Clinical and Biomedical Science, University of Exeter Medical School, Exeter, UK; Exeter Genomics Laboratory, Royal Devon & Exeter NHS Foundation Trust, Exeter, UK
| | - Frank Eisenhaber
- Bioinformatics Institute (BII), A(∗)STAR, Singapore, Singapore; Genome Institute of Singapore (GIS), A(∗)STAR, Singapore, Singapore; School of Biological Sciences (SBS), Nanyang Technological University (NTU), Singapore, Singapore
| | | | | | | | - Achim Gossler
- Institute for Molecular Biology, OE5250, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Birgit Eisenhaber
- Bioinformatics Institute (BII), A(∗)STAR, Singapore, Singapore; Genome Institute of Singapore (GIS), A(∗)STAR, Singapore, Singapore
| | - Martin Blum
- Institute of Biology, University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany
| | - Patrice Bouvagnet
- CPDP, Hôpital MFME, CHU de Martinique, BP632, 97200 Fort de France, France
| | | | - Jeanne Amiel
- Université de Paris, Imagine Institute, Laboratory of Embryology and Genetics of Malformations, INSERM UMR 1163, 75015 Paris, France; Fédération de Génétique, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
| | - Christopher T Gordon
- Université de Paris, Imagine Institute, Laboratory of Embryology and Genetics of Malformations, INSERM UMR 1163, 75015 Paris, France
| | - Bruno Reversade
- Laboratory of Human Genetics & Therapeutics, Genome Institute of Singapore (GIS), A(∗)STAR, Singapore, Singapore; Laboratory of Human Genetics & Therapeutics, BESE, KAUST, Thuwal, Saudi Arabia; Department of Physiology, Cardiovascular Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
4
|
Vingerhoets G. The relationship between brain and visceral asymmetry: Evidence from situs inversus in humans. HANDBOOK OF CLINICAL NEUROLOGY 2025; 208:47-61. [PMID: 40074416 DOI: 10.1016/b978-0-443-15646-5.00022-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
This review examines the relationship between visceral and brain asymmetry and explores whether their alignment observed in some vertebrate species also exists in humans. While the development of visceral and brain asymmetry may have occurred for different reasons, it is possible that the basic mechanisms for left-right differentiation of the visceral system were duplicated in the brain. We describe the main phenotypical anomalies and the general mechanism of left-right differentiation in vertebrates, followed by a systematic review of available human studies on behavioral and brain asymmetry in individuals with reversed visceral organization. The available evidence shows no direct link between human visceral and brain laterality. Most individuals with situs inversus totalis (SIT) show typical population biases for handedness and brain functional asymmetry, although an increased prevalence of atypical hemispheric segregation may be present. Perisylvian brain structural asymmetries also reveal the expected population bias in participants with SIT. However, several independent studies indicate that SIT is associated with a general reversal of the gross morphologic asymmetry of brain torque. Potential differences in brain structural and functional asymmetries between subtypes of situs inversus with ciliary and nonciliary causes remain to be determined.
Collapse
Affiliation(s)
- Guy Vingerhoets
- Department of Experimental Clinical and Health Psychology, Ghent University, Ghent, Belgium.
| |
Collapse
|
5
|
Constantin AA, Bivolaru S, Matache RS, Sima R, Mihaltan FD. Pulmonary Infarction in a Young Man With Situs Inversus Totalis. Cureus 2024; 16:e71334. [PMID: 39399275 PMCID: PMC11470973 DOI: 10.7759/cureus.71334] [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] [Accepted: 10/12/2024] [Indexed: 10/15/2024] Open
Abstract
We report the case of a 45-year-old male patient, a smoker, with a known condition of situs inversus totalis (SIT), who was diagnosed with an alveolar consolidation process during a chest imaging examination. Thorough medical investigations, including a surgical lung biopsy, resulted in the diagnosis of pulmonary infarction. The patient's clinical picture began suddenly, with chest pain of a stabbing character on the left side, inspiratory dyspnea, one episode of hemoptysis, fever (40°C), chills, and profuse sweating. Pulmonary infarction can have many different causes and determining the underlying etiology is frequently a considerable challenge, particularly given the urgency imposed by the severity of the condition. The association of pulmonary infarction in a patient with SIT is particularly noteworthy, as each of these conditions represents distinct pathological entities, with their overlap addressed in only a few cases in the literature. A rare genetic predisposition, possibly a fairly ordinary pairing, or even incidental coexistence, are some of the speculations discussed in this case presentation. We emphasize that pulmonary consolidation requires a comprehensive diagnostic approach due to its broad differential diagnosis. This highlights the critical importance of surgical lung biopsy and histopathological analysis in securing a precise and accurate diagnosis.
Collapse
Affiliation(s)
- Ancuta-Alina Constantin
- Pulmonology, Institute of Pneumology "Marius Nasta", Bucharest, ROU
- Respiratory Medicine, University of Medicine and Pharmacy "Carol Davila", Bucharest, ROU
| | - Sorin Bivolaru
- Medical Clinical, Dunarea de Jos University of Galati, Galati, ROU
| | - Radu S Matache
- Thoracic Surgery, Institute of Pneumology "Marius Nasta", Bucharest, ROU
| | - Romina Sima
- Obstetrics and Gynecology, University of Medicine and Pharmacy "Carol Davila", Bucharest, ROU
| | | |
Collapse
|
6
|
Guo S, Tang D, Chen Y, Yu H, Gu M, Geng H, Fang J, Wu B, Ruan L, Li K, Xu C, Gao Y, Tan Q, Duan Z, Wu H, Hua R, Guo R, Wei Z, Zhou P, Xu Y, Cao Y, He X, Sha Y, Lv M. Association of novel DNAH11 variants with asthenoteratozoospermia lead to male infertility. Hum Genomics 2024; 18:97. [PMID: 39256880 PMCID: PMC11389119 DOI: 10.1186/s40246-024-00658-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 08/13/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Bi-allelic variants in DNAH11 have been identified as causative factors in Primary Ciliary Dyskinesia, leading to abnormal respiratory cilia. Nonetheless, the specific impact of these variants on human sperm flagellar and their involvement in male infertility remain largely unknown. METHODS A collaborative effort involving two Chinese reproductive centers conducted a study with 975 unrelated infertile men. Whole-exome sequencing was employed for variant screening, and Sanger sequencing confirmed the identified variants. Morphological and ultrastructural analyses of sperm were conducted using Scanning Electron Microscopy and Transmission Electron Microscopy. Western Blot Analysis and Immunofluorescence Analysis were utilized to assess protein levels and localization. ICSI was performed to evaluate its efficacy in achieving favorable pregnancy outcomes for individuals with DNAH11 variants. RESULTS In this study, we identified seven novel variants in the DNAH11 gene in four asthenoteratozoospermia subjects. These variants led the absence of DNAH11 proteins and ultrastructure defects in sperm flagella, particularly affecting the outer dynein arms (ODAs) and adjacent structures. The levels of ODA protein DNAI2 and axoneme related proteins were down regulated, instead of inner dynein arms (IDA) proteins DNAH1 and DNAH6. Two out of four individuals with DNAH11 variants achieved clinical pregnancies through ICSI. The findings confirm the association between male infertility and bi-allelic deleterious variants in DNAH11, resulting in the aberrant assembly of sperm flagella and contributing to asthenoteratozoospermia. Importantly, ICSI emerges as an effective intervention for overcoming reproductive challenges caused by DNAH11 gene variants.
Collapse
Affiliation(s)
- Senzhao Guo
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Dongdong Tang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, 230032, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, 230032, China
| | - Yuge Chen
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Hui Yu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Meng Gu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Hao Geng
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Jiajun Fang
- The First Clinical Medical College of Anhui Medical University, Hefei, 230032, China
| | - Baoyan Wu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Lewen Ruan
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Kuokuo Li
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, 230032, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, 230032, China
| | - Chuan Xu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Yang Gao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Qing Tan
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Anhui Provincial Human Sperm Bank First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zongliu Duan
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Huan Wu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Rong Hua
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Rui Guo
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Zhaolian Wei
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Ping Zhou
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Yuping Xu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Yunxia Cao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, 230032, China.
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, 230032, China.
| | - Xiaojin He
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, 230032, China.
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yanwei Sha
- Department of Andrology, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China.
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, Fujian, China.
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, Fujian, China.
| | - Mingrong Lv
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, 230032, China.
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, 230032, China.
| |
Collapse
|
7
|
Yu K, Chen W, Chen Y, Shen L, Wu B, Zhang Y, Zhou X. De novo and inherited micro-CNV at 16p13.11 in 21 Chinese patients with defective cardiac left-right patterning. Front Genet 2024; 15:1458953. [PMID: 39315310 PMCID: PMC11416941 DOI: 10.3389/fgene.2024.1458953] [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: 07/03/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024] Open
Abstract
Objective Copy number changes at Chromosomal 16p13.11 have been implicated in a variety of human diseases including congenital cardiac abnormalities. The clinical correlation of copy number variants (CNVs) in this region with developmental abnormalities remains controversial as most of the patients inherit the duplication from an unaffected parent. Methods We performed CNV analysis on 164 patients with defective left-right (LR) patterning based on whole genome-exome sequencing (WG-ES) followed by multiplex ligation-dependent probe amplification (MLPA) validation. Most cases were accompanied with complex congenital heart disease (CHD). Results CNVs at 16p13.11 were identified in a total of 21 cases, accounting for 12.80% (21/164) evaluated cases. We observed a marked overrepresentation of chromosome 16p13.11 duplications in cases when compared with healthy controls according to literature reports (15/164, 9.14% versus 0.09% in controls). Notably, in two independent family trios, de novo 16p13.11 micro-duplications were identified in two patients with laterality defects and CHD. Moreover, 16p13.11 micro-duplication was segregated with the disease in a family trio containing 2 affected individuals. Notably, five coding genes, NOMO1, PKD1P3, NPIPA1, PDXDC1, and NTAN1, were potentially affected by micro-CNV at 16p13.11 in these patients. Conclusion Our study provides new family-trio based evidences to support 16p13.11 micro-duplications predispose individuals to defective cardiac left-right patterning and laterality disorder.
Collapse
Affiliation(s)
- Kun Yu
- The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Soochow, China
| | - Weicheng Chen
- Pediatric Cardiovascular Center, Children’s Hospital of Fudan University, Shanghai, China
| | - Yan Chen
- Obstetrics and Gynecology Hospital of Fudan University, Fudan University Shanghai Medical College, Shanghai, China
| | - Libing Shen
- International Human Phenome Institutes (IHPI), Shanghai, China
| | - Boxuan Wu
- Obstetrics and Gynecology Hospital of Fudan University, Fudan University Shanghai Medical College, Shanghai, China
| | - Yuan Zhang
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiangyu Zhou
- Obstetrics and Gynecology Hospital of Fudan University, Fudan University Shanghai Medical College, Shanghai, China
| |
Collapse
|
8
|
Zlotina A, Barashkova S, Zhuk S, Skitchenko R, Usoltsev D, Sokolnikova P, Artomov M, Alekseenko S, Simanova T, Goloborodko M, Berleva O, Kostareva A. Characterization of pathogenic genetic variants in Russian patients with primary ciliary dyskinesia using gene panel sequencing and transcript analysis. Orphanet J Rare Dis 2024; 19:310. [PMID: 39180133 PMCID: PMC11344339 DOI: 10.1186/s13023-024-03318-3] [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: 02/13/2024] [Accepted: 08/08/2024] [Indexed: 08/26/2024] Open
Abstract
BACKGROUND Primary ciliary dyskinesia (PCD) is a group of rare genetically heterogeneous disorders caused by defective cilia and flagella motility. The clinical phenotype of PCD patients commonly includes chronic oto-sino-pulmonary disease, infertility, and, in about half of cases, laterality defects due to randomization of left-right body asymmetry. To date, pathogenic variants in more than 50 genes responsible for motile cilia structure and assembly have been reported in such patients. While multiple population-specific mutations have been described in PCD cohorts from different countries, the data on genetic spectrum of PCD in Russian population are still extremely limited. RESULTS The present study provides a comprehensive clinical and genetic characterization of 21 Russian families with PCD living in various country regions. Anomalies of ciliary beating in patients` respiratory epithelial cells were confirmed by high-speed video microscopy. In the most cases, custom-designed panel sequencing allowed to uncover causative variants in well-known or rarely mentioned PCD-related genes, including DNAH5, DNAH11, CFAP300, LRRC6, ZMYND10, CCDC103, HYDIN, ODAD4, DNAL1, and OFD1. The variations comprised common mutations, as well as novel genetic variants, some of which probably specific for Russian patients. Additional targeted analysis of mRNA transcripts from ciliated cells enabled us to specify functional effects of newly identified genetic variants in DNAH5 (c.2052+3G>T, c.3599-2A>G), HYDIN (c.10949-2A>G, c.1797C>G), and ZMYND10 (c.510+1G>C) on splicing process. In particular, the splice site variant c.2052+3G>T, detected in four unrelated families, resulted in skipping of exon 14 in DNAH5 transcripts and, according to haplotype analysis of affected probands, was proposed as an ancestral founder mutation in Udmurt population. CONCLUSIONS The reported data provide a vital insight into genetic background of primary ciliary dyskinesia in the Russian population. The findings clearly illustrate the utility of gene panel sequencing coupled with transcriptional analysis in identification and clinical interpretation of novel genetic variants.
Collapse
Affiliation(s)
- Anna Zlotina
- Almazov National Medical Research Centre, Saint-Petersburg, Russia, 197341.
| | - Svetlana Barashkova
- Almazov National Medical Research Centre, Saint-Petersburg, Russia, 197341
- K.A. Raukhfus Children's City Multidisciplinary Clinical Center for High Medical Technologies, Saint-Petersburg, Russia, 191036
| | - Sergey Zhuk
- Almazov National Medical Research Centre, Saint-Petersburg, Russia, 197341
| | | | - Dmitrii Usoltsev
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, 43205, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, 43215, USA
| | - Polina Sokolnikova
- Almazov National Medical Research Centre, Saint-Petersburg, Russia, 197341
| | - Mykyta Artomov
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, 43205, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, 43215, USA
| | - Svetlana Alekseenko
- K.A. Raukhfus Children's City Multidisciplinary Clinical Center for High Medical Technologies, Saint-Petersburg, Russia, 191036
| | - Tatiana Simanova
- Republican Children's Clinical Hospital of the Ministry of Health of the Udmurt Republic, Izhevsk, Russia, 426009
| | - Maria Goloborodko
- K.A. Raukhfus Children's City Multidisciplinary Clinical Center for High Medical Technologies, Saint-Petersburg, Russia, 191036
| | - Olga Berleva
- K.A. Raukhfus Children's City Multidisciplinary Clinical Center for High Medical Technologies, Saint-Petersburg, Russia, 191036
| | - Anna Kostareva
- Almazov National Medical Research Centre, Saint-Petersburg, Russia, 197341
- Department of Women's and Children's Health, Center for Molecular Medicine, Karolinska Institutet, 17176, Stockholm, Sweden
| |
Collapse
|
9
|
Zhang S, Wang J, Sun L, Han J, Xiong X, Xiao D, Wu Q. Investigation of the genetic and clinical features of laterality disorders in prenatal diagnosis: discovery of a novel compound heterozygous mutation in the DNAH11 gene. Arch Gynecol Obstet 2024; 310:695-704. [PMID: 38852111 DOI: 10.1007/s00404-024-07574-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 05/27/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND Left-right laterality disorders are a heterogeneous group of disorders caused by an altered position or orientation of the thoracic and intra-abdominal organs and vasculature across the left-right axis. They mainly include situs inversus and heterotaxy. Those disorders are complicated by cardiovascular abnormalities significantly more frequently than situs solitus. METHODS In this study, 16 patients with a fetal diagnosis of laterality disorder with congenital heart defects (CHD) were evaluated with a single nucleotide polymorphism array (SNP-arry) combined with whole-exome sequencing (WES). RESULTS Although the diagnostic rate of copy number variations was 0 and the diagnostic rate of WES was 6.3% (1/16), the likely pathogenic gene DNAH11 and the candidate gene OFD1 were ultimately identified. In addition, novel compound heterozygous mutations in the DNAH11 gene and novel hemizygous variants in the OFD1 gene were found. Among the combined CHD, a single atrium/single ventricle had the highest incidence (50%, 8/16), followed by atrioventricular septal defects (37.5%, 6/16). Notably, two rare cases of common pulmonary vein atresia (CPVA) were also found on autopsy. CONCLUSION This study identified the types of CHD with a high incidence in patients with laterality disorders. It is clear that WES is an effective tool for diagnosing laterality disorders and can play an important role in future research.
Collapse
Affiliation(s)
- Simin Zhang
- Department of Ultrasound, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 251 Yaojiayuan Road, Chaoyang District, Beijing, 100026, People's Republic of China
- Department of Medical Ultrasound Center, Northwest Women's and Children's Hospital, Xi'an, Shaanxi, People's Republic of China
| | - Jingjing Wang
- Department of Ultrasound, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 251 Yaojiayuan Road, Chaoyang District, Beijing, 100026, People's Republic of China
| | - Lijuan Sun
- Department of Ultrasound, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 251 Yaojiayuan Road, Chaoyang District, Beijing, 100026, People's Republic of China
| | - Jijing Han
- Department of Ultrasound, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 251 Yaojiayuan Road, Chaoyang District, Beijing, 100026, People's Republic of China
| | - Xiaowei Xiong
- Department of Ultrasound, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 251 Yaojiayuan Road, Chaoyang District, Beijing, 100026, People's Republic of China
| | - Dan Xiao
- Center of Medical Genetics, Northwest Women's and Children's Hospital, Xi'an, Shaanxi, People's Republic of China
| | - Qingqing Wu
- Department of Ultrasound, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 251 Yaojiayuan Road, Chaoyang District, Beijing, 100026, People's Republic of China.
| |
Collapse
|
10
|
Shaikh Qureshi WM, Hentges KE. Functions of cilia in cardiac development and disease. Ann Hum Genet 2024; 88:4-26. [PMID: 37872827 PMCID: PMC10952336 DOI: 10.1111/ahg.12534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/08/2023] [Accepted: 10/02/2023] [Indexed: 10/25/2023]
Abstract
Errors in embryonic cardiac development are a leading cause of congenital heart defects (CHDs), including morphological abnormalities of the heart that are often detected after birth. In the past few decades, an emerging role for cilia in the pathogenesis of CHD has been identified, but this topic still largely remains an unexplored area. Mouse forward genetic screens and whole exome sequencing analysis of CHD patients have identified enrichment for de novo mutations in ciliary genes or non-ciliary genes, which regulate cilia-related pathways, linking cilia function to aberrant cardiac development. Key events in cardiac morphogenesis, including left-right asymmetric development of the heart, are dependent upon cilia function. Cilia dysfunction during left-right axis formation contributes to CHD as evidenced by the substantial proportion of heterotaxy patients displaying complex CHD. Cilia-transduced signaling also regulates later events during heart development such as cardiac valve formation, outflow tract septation, ventricle development, and atrioventricular septa formation. In this review, we summarize the role of motile and non-motile (primary cilia) in cardiac asymmetry establishment and later events during heart development.
Collapse
Affiliation(s)
- Wasay Mohiuddin Shaikh Qureshi
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science CentreUniversity of ManchesterManchesterUK
| | - Kathryn E. Hentges
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science CentreUniversity of ManchesterManchesterUK
| |
Collapse
|
11
|
Bu X, Li X, Peng C, Li H, Zhou S, Zhu Z, He J, Linpeng S. Case report: Paternal uniparental disomy on chromosome 7 and homozygous SUGCT mutation in a fetus with overweight after birth. Front Genet 2023; 14:1272028. [PMID: 37920852 PMCID: PMC10619901 DOI: 10.3389/fgene.2023.1272028] [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: 08/04/2023] [Accepted: 10/04/2023] [Indexed: 11/04/2023] Open
Abstract
Background: Paternal uniparental disomy (UPD) of chromosome 7 is extremely rare, and only a few postnatal cases have been reported. The effects on growth were discordant in these cases, and the relevance of paternal UPD(7) to growth caused by imprinting remains questionable. Case presentation: Here, we report a prenatal case that underwent invasive prenatal diagnosis due to the high risk of Down's syndrome and failed noninvasive prenatal screening. The fetus had a normal karyotype and no apparent copy number variation. Homozygous copy-neutral regions on chromosome 7 were identified using a single nucleotide polymorphism (SNP) array; the data for the parent-child trios showed that the fetus carried the whole paternal isodisomy of chromosome 7. Whole exome and Sanger sequencing revealed a homozygous frameshift mutation in SUGCT at 7p14.1, from the heterozygous carrier father, with no contribution from the mother. The parents decided to continue with the pregnancy after genetic counseling, and the neonate had normal physical findings at birth and showed overweight after birth during a long-term intensive follow-up. Conclusion: We report the first prenatal case who carried paternal UPD(7) and homozygous SUGCT mutation with an overweight phenotype after birth. The overweight may be caused by paternal UPD(7) or homozygous frameshift mutation of SUGCT, or both of them, but it is unclear which contributes more.
Collapse
Affiliation(s)
- Xiufen Bu
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, China
| | - Xu Li
- Department of Physiology, Changsha Health Vocational College, Changsha, China
| | - Can Peng
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, China
| | - Hongyu Li
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, China
| | - Shihao Zhou
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, China
| | - Zesen Zhu
- Technical Support Center, Zhejiang Biosan Biochemical Technologies Co., Ltd., Hangzhou, China
| | - Jun He
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, China
| | - Siyuan Linpeng
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, China
| |
Collapse
|
12
|
Raidt J, Loges NT, Olbrich H, Wallmeier J, Pennekamp P, Omran H. Primary ciliary dyskinesia. Presse Med 2023; 52:104171. [PMID: 37516247 DOI: 10.1016/j.lpm.2023.104171] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 07/24/2023] [Indexed: 07/31/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Primary ciliary dyskinesia (PCD, ORPHA:244) is a group of rare genetic disorders characterized by dysfunction of motile cilia. It is phenotypically and genetically heterogeneous, with more than 50 genes involved. Thanks to genetic, clinical, and functional characterization, immense progress has been made in the understanding and diagnosis of PCD. Nevertheless, it is underdiagnosed due to the heterogeneous phenotype and complexity of diagnosis. This review aims to help clinicians navigate this heterogeneous group of diseases. Here, we describe the broad spectrum of phenotypes associated with PCD and address pitfalls and difficult-to-interpret findings to avoid misinterpretation. METHOD Review of literature CONCLUSION: PCD diagnosis is complex and requires integration of history, clinical picture, imaging, functional and structural analysis of motile cilia and, if available, genetic analysis to make a definitive diagnosis. It is critical that we continue to expand our knowledge of this group of rare disorders to improve the identification of PCD patients and to develop evidence-based therapeutic approaches.
Collapse
Affiliation(s)
- Johanna Raidt
- Department of General Pediatrics, University Children's Hospital Muenster, Albert-Schweitzer-Campus 1, 48149 Muenster, Germany
| | - Niki Tomas Loges
- Department of General Pediatrics, University Children's Hospital Muenster, Albert-Schweitzer-Campus 1, 48149 Muenster, Germany
| | - Heike Olbrich
- Department of General Pediatrics, University Children's Hospital Muenster, Albert-Schweitzer-Campus 1, 48149 Muenster, Germany
| | - Julia Wallmeier
- Department of General Pediatrics, University Children's Hospital Muenster, Albert-Schweitzer-Campus 1, 48149 Muenster, Germany
| | - Petra Pennekamp
- Department of General Pediatrics, University Children's Hospital Muenster, Albert-Schweitzer-Campus 1, 48149 Muenster, Germany
| | - Heymut Omran
- Department of General Pediatrics, University Children's Hospital Muenster, Albert-Schweitzer-Campus 1, 48149 Muenster, Germany.
| |
Collapse
|
13
|
Hunter‐Schouela J, Geraghty MT, Hegele RA, Dyment DA, Pierre DS, Richer J, Sheffield H, Zariwala MA, Knowles MR, Lehman A, Dell S, Shapiro AJ, Kovesi TA. First reports of primary ciliary dyskinesia caused by a shared DNAH11 allele in Canadian Inuit. Pediatr Pulmonol 2023; 58:1942-1949. [PMID: 37088965 PMCID: PMC10330405 DOI: 10.1002/ppul.26414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 02/07/2023] [Accepted: 03/31/2023] [Indexed: 04/25/2023]
Abstract
BACKGROUND Primary ciliary dyskinesia (PCD) is typically an autosomal recessive disease characterized by recurrent infections of the lower respiratory tract, frequent and severe otitis media, chronic rhinosinusitis, neonatal respiratory distress, and organ laterality defects. While severe lower respiratory tract infections and bronchiectasis are common in Inuit, PCD has not been recognized in this population. METHODS We report a case series of seven Inuit patients with PCD identified by genetic testing in three Canadian PCD centers. RESULTS Patients ranged from 4 to 59 years of age (at time of last evaluation) and originated in the Qikiqtaaluk region (Baffin Island, n = 5), Nunavut, or Nunavik (northern Quebec, n = 2), Canada. They had typical features of PCD, including neonatal respiratory distress (five patients), situs inversus totalis (four patients), bronchiectasis (four patients), chronic atelectasis (six patients), and chronic otitis media (six patients). Most had chronic rhinitis. Genetic evaluation demonstrated that all had homozygous pathogenic variants in DNAH11 at NM_001277115.1:c.4095+2C>A. CONCLUSIONS The discovery of this homozygous DNAH11 variant in widely disparate parts of the Nunangat (Inuit homelands) suggests this is a founder mutation that may be widespread in Inuit. Thus, PCD may be an important cause of chronic lung, sinus, and middle ear disease in this population. Inuit with chronic lung disease, including bronchiectasis or laterality defects, should undergo genetic testing for PCD. Consideration of including PCD genetic analysis in routine newborn screening should be considered in Inuit regions.
Collapse
Affiliation(s)
- Julia Hunter‐Schouela
- Department of Pediatrics, Children’s Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Michael T. Geraghty
- Department of Pediatrics, Children’s Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Robert A. Hegele
- Department of Medicine and Robarts Research Institute, Western University, London, Ontario, Canada
| | - David A. Dyment
- Department of Genetics, Children’s Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - David St Pierre
- Respiratory Epidemiology and Clinical Research Unit, Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Julie Richer
- Department of Genetics, Children’s Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Holden Sheffield
- Department of Pediatrics, Qikiqtani General Hospital, Iqaluit, Nunavut, Canada
| | - Maimoona A. Zariwala
- Department of Pathology and Laboratory Medicine, Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Michael R. Knowles
- Department of Medicine, Marsico Lung Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Anna Lehman
- Department of Medical Genetics, University of British Columbia on behalf of the Silent Genomes Precision Medicine Consortium, Vancouver, British Columbia, Canada
| | - Sharon Dell
- Department of Pediatrics, BC Children’s Hospital, Vancouver, British Columbia, Canada
| | - Adam J. Shapiro
- Department of Pediatrics, McGill University Health Centre Research Institute, Montreal, Quebec, Canada
| | - Thomas A. Kovesi
- Department of Pediatrics, Children’s Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| |
Collapse
|
14
|
Ritter J, Lisec K, Klinner M, Heinrich M, von Schweinitz D, Kappler R, Hubertus J. Genetic Disruption of Cilia-Associated Signaling Pathways in Patients with VACTERL Association. CHILDREN (BASEL, SWITZERLAND) 2023; 10:children10050882. [PMID: 37238430 DOI: 10.3390/children10050882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/05/2023] [Accepted: 05/13/2023] [Indexed: 05/28/2023]
Abstract
VACTERL association is a rare malformation complex consisting of vertebral defects, anorectal malformation, cardiovascular defects, tracheoesophageal fistulae with esophageal atresia, renal malformation, and limb anomalies. According to current knowledge, VACTERL is based on a multifactorial pathogenesis including genomic alterations. This study aimed to improve the understanding of the genetic mechanisms in the development of VACTERL by investigating the genetic background with a focus on signaling pathways and cilia function. The study was designed as genetic association study. For this, whole-exome sequencing with subsequent functional enrichment analyses was performed for 21 patients with VACTERL or a VACTERL-like phenotype. In addition, whole-exome sequencing was performed for three pairs of parents and Sanger-sequencing was performed for ten pairs of parents. Analysis of the WES-data revealed genetic alteration in the Shh- and Wnt-signaling pathways. Additional performed functional enrichment analysis identified an overrepresentation of the cilia, including 47 affected ciliary genes with clustering in the DNAH gene family and the IFT-complex. The examination of the parents showed that most of the genetic changes were inherited. In summary, this study indicates three genetically determined damage mechanisms for VACTERL with the potential to influence each other, namely Shh- and Wnt-signaling pathway disruption, structural cilia defects and disruption of the ciliary signal transduction.
Collapse
Affiliation(s)
- Jessica Ritter
- Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, LMU Munich University, 80337 Munich, Germany
- Department of Diagnostic and Interventional Radiology, Klinikum Rechts der Isar, Technical University of Munich, Ismaninger Straße 22, 81675 Munich, Germany
| | - Kristina Lisec
- Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, LMU Munich University, 80337 Munich, Germany
| | - Marina Klinner
- Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, LMU Munich University, 80337 Munich, Germany
| | - Martina Heinrich
- Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, LMU Munich University, 80337 Munich, Germany
| | - Dietrich von Schweinitz
- Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, LMU Munich University, 80337 Munich, Germany
| | - Roland Kappler
- Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, LMU Munich University, 80337 Munich, Germany
| | - Jochen Hubertus
- Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, LMU Munich University, 80337 Munich, Germany
- Department of Pediatric Surgery, Marien Hospital Witten, Ruhr-University Bochum, 58452 Witten, Germany
| |
Collapse
|
15
|
Cai HY, Chen SR, Wang Y, Jiao JJ, Qiao J, Hölscher C, Wang ZJ, Zhang SX, Wu MN. Integrated analysis of the lncRNA-associated ceRNA network in Alzheimer's disease. Gene 2023; 876:147484. [PMID: 37187245 DOI: 10.1016/j.gene.2023.147484] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 04/07/2023] [Accepted: 05/09/2023] [Indexed: 05/17/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease that worsens with age. Long non-coding RNAs (lncRNAs) dysregulation and its associated competing endogenous RNA (ceRNA) network have a potential connection with the occurrence and development of AD. A total of 358 differentially expressed genes (DEGs) were screened via RNA sequencing, including 302 differentially expressed mRNAs (DEmRNAs) and 56 differential expressed lncRNAs (DElncRNAs). Anti-sense lncRNA is the main type of DElncRNA, which plays a major role in the cis and trans regulation. The constructed ceRNA network consisted of 4 lncRNAs (NEAT1, LINC00365, FBXL19-AS1, RAI1-AS1719) and 4 microRNAs (miRNAs) (HSA-Mir-27a-3p, HSA-Mir-20b-5p, HSA-Mir-17-5p, HSA-Mir-125b-5p), and 2 mRNAs (MKNK2, F3). Functional enrichment analysis revealed that DEmRNAs are involved in related biological functions of AD. The co-expressed DEmRNAs (DNAH11, HGFAC, TJP3, TAC1, SPTSSB, SOWAHB, RGS4, ADCYAP1) of humans and mice were screened and verified by real-time quantitative polymerase chain reaction (qRT-PCR). In this study, we analyzed the expression profile of human AD-related lncRNA genes, constructed a ceRNA network, and performed functional enrichment analysis of DEmRNAs between human and mice. The obtained gene regulatory networks and target genes can be used to further analyze AD-related pathological mechanisms to optimize AD diagnosis and treatment.
Collapse
Affiliation(s)
- Hong-Yan Cai
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, China; Key Laboratory of Cellular Physiology, Shanxi Province, China.
| | - Si-Ru Chen
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, China
| | - Yu Wang
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, China
| | - Juan-Juan Jiao
- Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Jun Qiao
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, China; Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Christian Hölscher
- Academy of Chinese Medical Science, Henan university of Chinese medicine, Zhengzhou, China
| | - Zhao-Jun Wang
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, China; Key Laboratory of Cellular Physiology, Shanxi Province, China; Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Sheng-Xiao Zhang
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, China; Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Mei-Na Wu
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, China; Key Laboratory of Cellular Physiology, Shanxi Province, China; Department of Physiology, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
16
|
Li L, Li J, Ou Y, Wu J, Li H, Wang X, Tang L, Dai X, Yang C, Wei Z, Yin Z, Shu Y. Ccdc57 is required for straightening the body axis by regulating ciliary motility in the brain ventricle of zebrafish. J Genet Genomics 2023; 50:253-263. [PMID: 36669737 DOI: 10.1016/j.jgg.2022.12.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 12/22/2022] [Accepted: 12/31/2022] [Indexed: 01/19/2023]
Abstract
Recently, cilia defects have been proposed to contribute to scoliosis. Here, we demonstrate that coiled-coil domain-containing 57 (Ccdc57) plays an essential role in straightening the body axis of zebrafish by regulating ciliary beating in the brain ventricle (BV). Zygotic ccdc57 (Zccdc57) mutant zebrafish developes scoliosis without significant changes in their bone density and calcification, and the maternal-zygotic ccdc57 (MZccdc57) mutant embryos display curved bodies since the long-pec stage. The expression of ccdc57 is enriched in ciliated tissues and immunofluorescence analysis reveals colocalization of Ccdc57-HA with acetylated α-tubulin, implicating it in having a role in ciliary function. Further examination reveals that it is the coordinated cilia beating of multiple cilia bundles (MCB) in the MZccdc57 mutant embryos that is affected at 48 hours post fertilization, when the compromised cerebrospinal fluid flow and curved body axis have already occurred. Either ccdc57 mRNA injection or epinephrine treatment reverses the spinal curvature in MZccdc57 mutant larvae from ventrally curly to straight or even dorsally curly and significantly upregulates urotensin signaling. This study reveals the role of ccdc57 in maintaining coordinated cilia beating of MCB in the BV.
Collapse
Affiliation(s)
- Lu Li
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China; College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Juan Li
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China; College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Yuan Ou
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China; College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Jiaxin Wu
- School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Huilin Li
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China; College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Xin Wang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China; College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Liying Tang
- College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Xiangyan Dai
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Southwest University, Chongqing 400715, China
| | - Conghui Yang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China; College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Zehong Wei
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China; College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Zhan Yin
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei 430072, China
| | - Yuqin Shu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China; College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China.
| |
Collapse
|
17
|
Higgins K, Moore BA, Berberovic Z, Adissu HA, Eskandarian M, Flenniken AM, Shao A, Imai DM, Clary D, Lanoue L, Newbigging S, Nutter LMJ, Adams DJ, Bosch F, Braun RE, Brown SDM, Dickinson ME, Dobbie M, Flicek P, Gao X, Galande S, Grobler A, Heaney JD, Herault Y, de Angelis MH, Chin HJG, Mammano F, Qin C, Shiroishi T, Sedlacek R, Seong JK, Xu Y, Lloyd KCK, McKerlie C, Moshiri A. Analysis of genome-wide knockout mouse database identifies candidate ciliopathy genes. Sci Rep 2022; 12:20791. [PMID: 36456625 PMCID: PMC9715561 DOI: 10.1038/s41598-022-19710-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 09/02/2022] [Indexed: 12/05/2022] Open
Abstract
We searched a database of single-gene knockout (KO) mice produced by the International Mouse Phenotyping Consortium (IMPC) to identify candidate ciliopathy genes. We first screened for phenotypes in mouse lines with both ocular and renal or reproductive trait abnormalities. The STRING protein interaction tool was used to identify interactions between known cilia gene products and those encoded by the genes in individual knockout mouse strains in order to generate a list of "candidate ciliopathy genes." From this list, 32 genes encoded proteins predicted to interact with known ciliopathy proteins. Of these, 25 had no previously described roles in ciliary pathobiology. Histological and morphological evidence of phenotypes found in ciliopathies in knockout mouse lines are presented as examples (genes Abi2, Wdr62, Ap4e1, Dync1li1, and Prkab1). Phenotyping data and descriptions generated on IMPC mouse line are useful for mechanistic studies, target discovery, rare disease diagnosis, and preclinical therapeutic development trials. Here we demonstrate the effective use of the IMPC phenotype data to uncover genes with no previous role in ciliary biology, which may be clinically relevant for identification of novel disease genes implicated in ciliopathies.
Collapse
Affiliation(s)
- Kendall Higgins
- The University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
| | - Bret A Moore
- Department of Small Animal Clinical Sciences, University of Florida, College of Veterinary Medicine, Gainesville, FL, 32608, USA
| | - Zorana Berberovic
- The Centre for Phenogenomics, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | | | - Mohammad Eskandarian
- The Centre for Phenogenomics, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - Ann M Flenniken
- The Centre for Phenogenomics, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - Andy Shao
- University of Reno, Nevada, School of Medicine, Reno, NV, 89557, USA
| | - Denise M Imai
- Comparative Pathology Laboratory, U.C. Davis, Davis, 95616, USA
| | - Dave Clary
- Mouse Biology Program, U.C. Davis, Davis, CA, 95618, USA
| | - Louise Lanoue
- Mouse Biology Program, U.C. Davis, Davis, CA, 95618, USA
| | - Susan Newbigging
- The Centre for Phenogenomics, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - Lauryl M J Nutter
- The Centre for Phenogenomics, Toronto, ON, Canada
- The Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
| | - David J Adams
- The Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Fatima Bosch
- Centre of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, 08193, Barcelona, Spain
| | | | - Steve D M Brown
- Medical Research Council Harwell Institute (Mammalian Genetics Unit and Mary Lyon Centre), Harwell Campus, Oxfordshire, OX11 0RD, UK
| | - Mary E Dickinson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Michael Dobbie
- Phenomics Australia, The Australian National University, 131 Garran Rd, Acton, Canberra, ACT, 2601, Australia
| | - Paul Flicek
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Xiang Gao
- SKL of Pharmaceutical Biotechnology and Model Animal Research Center, Collaborative Innovation Center for Genetics and Development, Nanjing Biomedical Research Institute, Nanjing University, Nanjing, 210061, China
| | - Sanjeev Galande
- Indian Institutes of Science Education and Research, Dr. Homi Bhabha Rd, Ward No. 8, NCL Colony, Pashan, Pune, Maharashtra, 411008, India
| | - Anne Grobler
- Faculty of Health Sciences, PCDDP North-West University, North-West University Potchefstroom Campus 11 Hoffman Street, Potchefstroom, 2531, South Africa
| | - Jason D Heaney
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yann Herault
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université de Strasbourg, 67400, Illkirch, France
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université de Strasbourg, 1 rue Laurent Fries, 67404, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France
- Université de Strasbourg, 1 rue Laurent Fries, 67404, Illkirch, France
- CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), CNRS, INSERM, Université of Strasbourg, 1 rue Laurent Fries, 67404, Illkirch-Graffenstaden, France
| | - Martin Hrabe de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Hsian-Jean Genie Chin
- National Laboratory Animal Center, National Applied Research Laboratories (NARLabs), 3F., No. 106, Sec. 2, Heping E. Rd., Da'an Dist., Taipei City, 106214, Taiwan (R.O.C.)
| | - Fabio Mammano
- Monterotondo Mouse Clinic, Italian National Research Council (CNR), Institute of Cell Biology and Neurobiology, Adriano Buzzati-Traverso Campus, Via Ramarini, 00015, Monterotondo Scalo, Italy
| | - Chuan Qin
- National Laboratory Animal Center, National Applied Research Laboratories (NARLabs), Beijing, China
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Science, 5 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | | | - Radislav Sedlacek
- Czech Center for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, IMG BIOCEV Building SO.02 Prumyslova 595, 252 50, Vestec, Czech Republic
| | - J-K Seong
- Korea Mouse Phenotyping Consortium (KMPC) and BK21 Program for Veterinary Science, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 599 Gwanangno, Gwanak-gu, Seoul, 08826, South Korea
| | - Ying Xu
- CAM-SU Genomic Resource Center, Soochow University, Organization Planning of No. 1 Shizi Street, Suzhou, 215123, China
| | - K C Kent Lloyd
- Mouse Biology Program, U.C. Davis, Davis, CA, 95618, USA
- Department of Surgery, School of Medicine, U.C. Davis, Sacramento, CA, 95817, USA
| | - Colin McKerlie
- The Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada.
- Department of Laboratory Medicine and Pathobiology, Hospital for Sick Children (SickKids), The Centre for Phenogenomics, Faculty of Medicine, University of Toronto, 25 Orde Street, Toronto, ON, M5T 3H7, USA.
| | - Ala Moshiri
- Department of Ophthalmology and Vision Science, School of Medicine, U.C. Davis Eye Center, 4860 Y. Street, Suite 2400, Sacramento, CA, 95817, USA.
| |
Collapse
|
18
|
Chen W, Wang F, Zeng W, Zhang X, Shen L, Zhang Y, Zhou X. Biallelic mutations of TTC12 and TTC21B were identified in Chinese patients with multisystem ciliopathy syndromes. Hum Genomics 2022; 16:48. [PMID: 36273201 PMCID: PMC9587637 DOI: 10.1186/s40246-022-00421-z] [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: 07/04/2022] [Accepted: 10/12/2022] [Indexed: 12/04/2022] Open
Abstract
Background Abnormalities in cilia ultrastructure and function lead to a range of human phenotypes termed ciliopathies. Many tetratricopeptide repeat domain (TTC) family members have been reported to play critical roles in cilium organization and function.
Results Here, we describe five unrelated family trios with multisystem ciliopathy syndromes, including situs abnormality, complex congenital heart disease, nephronophthisis or neonatal cholestasis. Through whole-exome sequencing and Sanger sequencing confirmation, we identified compound heterozygous mutations of TTC12 and TTC21B in six affected individuals of Chinese origin. These nonsynonymous mutations affected highly conserved residues and were consistently predicted to be pathogenic. Furthermore, ex vivo cDNA amplification demonstrated that homozygous c.1464 + 2 T > C of TTC12 would cause a whole exon 16 skipping. Both mRNA and protein levels of TTC12 were significantly downregulated in the cells derived from the patient carrying TTC12 mutation c.1464 + 2 T > C by real-time qPCR and immunofluorescence assays when compared with two healthy controls. Transmission electron microscopy analysis further identified ultrastructural defects of the inner dynein arms in this patient. Finally, the effect of TTC12 deficiency on cardiac LR patterning was recapitulated by employing a morpholino-mediated knockdown of ttc12 in zebrafish. Conclusions To the best of our knowledge, this is the first study reporting the association between TTC12 variants and ciliopathies in a Chinese population. In addition to nephronophthisis and laterality defects, our findings demonstrated that TTC21B should also be considered a candidate gene for biliary ciliopathy, such as TTC26, which further expands the phenotypic spectrum of TTC21B deficiency in humans. Supplementary Information The online version contains supplementary material available at 10.1186/s40246-022-00421-z.
Collapse
Affiliation(s)
- Weicheng Chen
- Obstetrics and Gynecology Hospital of Fudan University, Pediatric Cardiovascular Center at Children's Hospital of Fudan University, Fudan University Shanghai Medical College, Shanghai, 200011, China
| | - Feifei Wang
- Obstetrics and Gynecology Hospital of Fudan University, Pediatric Cardiovascular Center at Children's Hospital of Fudan University, Fudan University Shanghai Medical College, Shanghai, 200011, China
| | - Weijia Zeng
- State Key Lab of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Xinyan Zhang
- Obstetrics and Gynecology Hospital of Fudan University, Pediatric Cardiovascular Center at Children's Hospital of Fudan University, Fudan University Shanghai Medical College, Shanghai, 200011, China
| | - Libing Shen
- International Human Phenome Institutes (IHPI), Shanghai, 200433, China
| | - Yuan Zhang
- Department of Assisted Reproduction, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 201204, China. .,, Shanghai, China.
| | - Xiangyu Zhou
- Obstetrics and Gynecology Hospital of Fudan University, Pediatric Cardiovascular Center at Children's Hospital of Fudan University, Fudan University Shanghai Medical College, Shanghai, 200011, China. .,Department of Assisted Reproduction, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 201204, China. .,, Shanghai, China.
| |
Collapse
|
19
|
Levkova M, Radanova M, Angelova L. Potential role of dynein-related genes in the etiology of male infertility: A systematic review and a meta-analysis. Andrology 2022; 10:1484-1499. [PMID: 36057791 DOI: 10.1111/andr.13287] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/21/2022] [Accepted: 08/28/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND The dynein-related genes may have a role in the etiology of male infertility, particularly in cases of impaired sperm motility. OBJECTIVES The goal of this review is to compile a list of the most important dynein-related candidate genes that may contribute to male factor infertility. MATERIALS AND METHODS Databases were searched using the keywords "dynein", "male", "infertility" and by applying strict inclusion criteria. A meta-analysis was also performed by using the eligible case-control studies. The odd ratios (OR), the Z-test score, and the level of significance were determined using a fixed model with a p value of 0.05. Funnel plots were used to check for publication bias. RESULTS There were 35 studies that met the inclusion criteria. There were a total of fifteen genes responsible for the production of dynein structural proteins, the production of dynein assembling factors, and potentially associated with male infertility. A total of five case-control studies were eligible for inclusion in the meta-analysis. Variants in the dynein-related genes were linked to an increased the risk of male infertility (OR = 21.52, 95% Confidence Interval (CI) 8.34 - 55.50, Z test = 6.35, p < 0.05). The percentage of heterogeneity, I2 , was 47.00%. The lack of variants in the dynein genes was an advantage and this was statistically significant. DISCUSSION The results from the present review illustrate that pathogenic variants in genes both for dynein synthesis and for dynein assembly factors could be associated with isolated cases of male infertility without any other symptoms. CONCLUSIONS The genes addressed in this study, which are involved in both the production and assembly of dynein, could be used as molecular targets for future research into the etiology of sperm motility problems. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Mariya Levkova
- Department of Medical Genetics, Medical University Varna, Marin Drinov Str 55, Varna, 9000, Bulgaria.,Laboratory of Medical Genetics, St. Marina Hospital, Hristo Smirnenski Blv 1, Varna, 9000, Bulgaria
| | - Maria Radanova
- Department of Biochemistry, Molecular Medicine and Nutrigenomics, Medical University Varna, Tzar Osvoboditel Str 84b, Varna, 9000, Bulgaria
| | - Lyudmila Angelova
- Department of Medical Genetics, Medical University Varna, Marin Drinov Str 55, Varna, 9000, Bulgaria
| |
Collapse
|
20
|
Situs inversus totalis with local metastasis of gallbladder carcinoma and variation of the common hepatic artery. BMC Gastroenterol 2022; 22:357. [PMID: 35883027 PMCID: PMC9327273 DOI: 10.1186/s12876-022-02377-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 06/10/2022] [Indexed: 11/17/2022] Open
Abstract
Background Situs inversus totalis (SIT) is a rare congenital anomaly characterized by a complete transposition of all the viscera. SIT cases were usually reported because of the presence of tumors, leading to false association between them. Therefore, any research that advances our understanding on SIT is highly required. This study firstly describes a very rare case of SIT with “jumping” metastasis to pancreas of gallbladder carcinoma. Case presentation A 69-year-old female patient presented at our hospital with complaints of one month of epigastric pain was studied. She had not sought for treatment prior the visit. Imaging examinations of this patient revealed SIT and a variation of the common hepatic artery with concomitant tumors of gallbladder and pancreas. However, there was no evidence of distant metastases beyond the abdominal cavity. She underwent a combination of radical cholecystectomy, total pancreatectomy, splenectomy and hepatic artery-splenic artery reconstruction. Histological analyses revealed metastasis of the gallbladder carcinoma in to the pancreas. Although the patient opted against chemotherapy, she survived without tumor for 16 months following the surgery. A review of the current literature on association with SIT and tumor occurrence was presented. Conclusions It is a great surgical challenge for the resection of multicenter hepatobiliary and pancreatic tumors in such rare SIT anatomical abnormalities with vascular variants. A reliable surgical plan based on detailed preoperative imaging and intraoperative anatomical exploration is crucial to achieving radical resection.
Collapse
|
21
|
Li BB, Lu SL, He X, Lei B, Yao JN, Feng SC, Yu SP. Da Vinci robot-assisted pancreato-duodenectomy in a patient with situs inversus totalis: A case report and review of literature. World J Gastrointest Oncol 2022; 14:1363-1371. [PMID: 36051094 PMCID: PMC9305577 DOI: 10.4251/wjgo.v14.i7.1363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 04/30/2022] [Accepted: 06/26/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Situs inversus totalis (SIT) is an extremely rare congenital malformation characterized by mirror displacement of the thoracoabdominal organs such as the heart, liver, spleen, and stomach. Herein, we describe a patient with SIT complicated with cholangiocarcinoma who underwent successful pancreaticoduodenectomy with the assistance of a da Vinci robot.
CASE SUMMARY A 58-year-old female presented to the hospital with paroxysmal pain in her left upper abdomen, accompanied by jaundice and staining of the sclera as chief complaints. Imaging examination detected a mass at the distal end of the common bile duct, with inverted thoracic and abdominal organs. Endoscopic retrograde cholangiopancreatography forceps biopsy revealed the presence of a well-differentiated adenocarcinoma. The patient successfully underwent robotic-assisted pancreaticoduodenectomy; the operation lasted 300 min, the intraoperative blood loss was 500 mL, and there were no intraoperative and postoperative complications.
CONCLUSION SIT is not directly related to the formation of cholangiocarcinoma. Detailed preoperative imaging examination is conducive to disease diagnosis and also convenient for determining the feasibility of tumor resection. Robot-assisted pancreaticoduodenectomy for SIT complicated with cholangiocarcinoma provides a safe, feasible, minimally invasive, and complication-free alternative with adequate preoperative planning combined with meticulous intraoperative procedures.
Collapse
Affiliation(s)
- Bai-Bei Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Shi-Liu Lu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Xiang He
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Biao Lei
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Jian-Ni Yao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Si-Chen Feng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Shui-Ping Yu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
22
|
Biallelic Variants in CCDC39 Gene Lead to Primary Ciliary Dyskinesia and Kartagener Syndrome. BIOMED RESEARCH INTERNATIONAL 2022; 2022:7130555. [PMID: 35795318 PMCID: PMC9251071 DOI: 10.1155/2022/7130555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 05/23/2022] [Accepted: 06/09/2022] [Indexed: 01/02/2023]
Abstract
Background Primary ciliary dyskinesia (PCD) is a clinical syndrome characterized by cilia with an abnormal structure or function. Its main clinical manifestations comprise chronic bronchitis, cough, recurrent respiratory infections, situs inversus, and male infertility. Single-gene variants are widely assumed to be the main cause of this rare disease, and more than 40 genes have been described to be associated with its onset. CCDC39 is essential for assembling the inner dynein arms and dynein regulatory complex and is important in cilia motility. CCDC39 variants were reported as a monogenic etiology of PCD. Methods This study investigated two unrelated Chinese patients diagnosed as PCD. The chest computed tomography scan was performed to identify PCD phenotypes of the two probands. Considering the effect of PCD on male fertility, routine semen analysis, sperm morphology examination, and scanning electron microscopy were performed to assess the semen characteristics of male proband in family 2 (F2 II-1), who had a history of infertility. Subsequently, the peripheral blood samples of probands were collected to perform whole-exome sequencing (WES) to explore the possible genetic causes of this disease. Results Whole-exome sequencing revealed a homozygous CCDC39 variant in the female proband of family 1 (F1 II-1: c.286C>T:p.Arg96Ter) and two compound heterozygous CCDC39 variants in the male proband of family 2 (F2 II-1: c.732_733del: p.Ala245PhefsTer18; c.2800_2802dup:p.Val934dup). The two probands showed the typical PCD phenotypes, including chronic bronchitis, recurrent respiratory infections, and situs inversus. The male proband also showed oligoasthenoteratospermia with multiple morphological abnormalities of the sperm flagella. Additionally, CCDC39 protein level was significantly lower in the sperm of male proband than in the sperm from normal controls. Conclusion We identified a homozygous variant reported previously and two compound heterozygous variants of CCDC39 possibly responsible for PCD pathogenesis, expanding the variant spectrum of Chinese PCD, Kartagener syndrome, and morphological abnormalities of the sperm flagella involving CCDC39.
Collapse
|
23
|
Yu X, Yuan L, Deng S, Xia H, Tu X, Deng X, Huang X, Cao X, Deng H. Identification of DNAH17 Variants in Han-Chinese Patients With Left–Right Asymmetry Disorders. Front Genet 2022; 13:862292. [PMID: 35692830 PMCID: PMC9186109 DOI: 10.3389/fgene.2022.862292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/18/2022] [Indexed: 12/12/2022] Open
Abstract
The formation of left–right asymmetry of the visceral organs is a conserved feature of the human body, and the asymmetry specification of structure and function is precisely orchestrated by multiple regulatory mechanisms. The abnormal results of organ positioning situs arise from defective cilia structure or function during embryogenesis in humans. In this study, we recruited two unrelated Han-Chinese families with left–right asymmetry disorders. The combination of whole-exome sequencing and Sanger sequencing identified two compound heterozygous variants: c.4109C>T and c.9776C>T, and c.612C>G and c.8764C>T in the dynein axonemal heavy chain 17 gene (DNAH17) in two probands with left–right asymmetry disorders. We report for the first time a possible association between DNAH17 gene variants and left–right asymmetry disorders, which is known as a causal gene for asthenozoospermia. Altogether, the findings of our study may enlarge the DNAH17 gene variant spectrum in human left–right asymmetry disorders, pave a way to illustrate the potential pathogenesis of ciliary/flagellar disorders, and provide supplementary explanation for genetic counseling.
Collapse
Affiliation(s)
- Xuehui Yu
- Health Management Center, The Third Xiangya Hospital, Central South University, Changsha, China
- Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Lamei Yuan
- Health Management Center, The Third Xiangya Hospital, Central South University, Changsha, China
- Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
- Disease Genome Research Center, Central South University, Changsha, China
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Sheng Deng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Hong Xia
- Department of Emergency, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiaolong Tu
- Department of Emergency, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiong Deng
- Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiangjun Huang
- Department of General Surgery, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Xiao Cao
- Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Hao Deng
- Health Management Center, The Third Xiangya Hospital, Central South University, Changsha, China
- Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
- Disease Genome Research Center, Central South University, Changsha, China
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Hao Deng,
| |
Collapse
|
24
|
Breuer K, Riedhammer KM, Müller N, Schaidinger B, Dombrowsky G, Dittrich S, Zeidler S, Bauer UMM, Westphal DS, Meitinger T, Dakal TC, Hitz MP, Breuer J, Reutter H, Hilger AC, Hoefele J. Exome sequencing in individuals with cardiovascular laterality defects identifies potential candidate genes. Eur J Hum Genet 2022; 30:946-954. [PMID: 35474353 PMCID: PMC9349204 DOI: 10.1038/s41431-022-01100-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 02/26/2022] [Accepted: 04/04/2022] [Indexed: 12/30/2022] Open
Abstract
The birth prevalence of laterality defects is about 1.1/10,000 comprising different phenotypes ranging from situs inversus totalis to heterotaxy, mostly associated with complex congenital heart defects (CHD) and situs abnormalities such as intestinal malrotation, biliary atresia, asplenia, or polysplenia. A proportion of laterality defects arise in the context of primary ciliary dyskinesia (PCD) accompanied by respiratory symptoms or infertility. In this study, exome sequencing (ES) was performed in 14 case-parent trios/quattros with clinical exclusion of PCD prior to analysis. Moreover, all cases and parents underwent detailed clinical phenotyping including physical examination, echocardiography by a skilled paediatric cardiologist and abdominal ultrasound examinations not to miss mildly affected individuals. Subsequent survey of the exome data comprised filtering for monoallelic de novo, rare biallelic, and X-linked recessive variants. In two families, rare variants of uncertain significance (VUS) in PKD1L1 and ZIC3 were identified. Both genes have been associated with laterality defects. In two of the remaining families, biallelic variants in LMBRD1 and DNAH17, respectively, were prioritized. In another family, an ultra-rare de novo variant in WDR47 was found. Extensive exome survey of 2,109 single exomes of individuals with situs inversus totalis, heterotaxy, or isolated CHD identified two individuals with novel monoallelic variants in WDR47, but no further individuals with biallelic variants in DNAH17 or LMBRD1. Overall, ES of 14 case-parent trios/quattros with cardiovascular laterality defects identified rare VUS in two families in known disease-associated genes PKD1L1 and ZIC3 and suggests DNAH17, LMBRD1, and WDR47 as potential genes involved in laterality defects.
Collapse
Affiliation(s)
- Katinka Breuer
- Institute of Human Genetics, University Hospital of Bonn, Bonn, Germany.,Department of Pediatric Cardiology, Pediatric Heart Center, University Hospital of Bonn, Bonn, Germany
| | - Korbinian M Riedhammer
- Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany.,Department of Nephrology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Nicole Müller
- Department of Pediatric Cardiology, Pediatric Heart Center, University Hospital of Bonn, Bonn, Germany
| | - Birthe Schaidinger
- Department of Pediatric Cardiology, Pediatric Heart Center, University Hospital of Bonn, Bonn, Germany
| | - Gregor Dombrowsky
- Department of Congenital Heart Disease and Pediatric Cardiology, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Sven Dittrich
- Department of Pediatric Cardiology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Susanne Zeidler
- Pediatric Department, Asklepios clinics, Sankt Augustin, Germany
| | - Ulrike M M Bauer
- Competence Network for Congenital Heart Defects & National Register for Congenital Heart Defects, German Center for Cardiovascular Research (DZHK), Berlin, Germany
| | - Dominik S Westphal
- Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany.,DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Berlin, Germany.,Department of Internal Medicine I, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Thomas Meitinger
- Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Tikam Chand Dakal
- Department of Biotechnology, Mohanlal Sukhadia University Udaipur, Udaipur, Rajasthan, India
| | - Marc-Phillip Hitz
- Department of Congenital Heart Disease and Pediatric Cardiology, University Hospital of Schleswig-Holstein, Kiel, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site, Kiel, Germany
| | - Johannes Breuer
- Department of Pediatric Cardiology, Pediatric Heart Center, University Hospital of Bonn, Bonn, Germany
| | - Heiko Reutter
- Institute of Human Genetics, University Hospital of Bonn, Bonn, Germany.,Division of Neonatology and Pediatric Intensive Care Medicine, Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Alina C Hilger
- Institute of Human Genetics, University Hospital of Bonn, Bonn, Germany
| | - Julia Hoefele
- Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany.
| |
Collapse
|
25
|
Djenoune L, Berg K, Brueckner M, Yuan S. A change of heart: new roles for cilia in cardiac development and disease. Nat Rev Cardiol 2022; 19:211-227. [PMID: 34862511 PMCID: PMC10161238 DOI: 10.1038/s41569-021-00635-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/11/2021] [Indexed: 12/27/2022]
Abstract
Although cardiac abnormalities have been observed in a growing class of human disorders caused by defective primary cilia, the function of cilia in the heart remains an underexplored area. The primary function of cilia in the heart was long thought to be restricted to left-right axis patterning during embryogenesis. However, new findings have revealed broad roles for cilia in congenital heart disease, valvulogenesis, myocardial fibrosis and regeneration, and mechanosensation. In this Review, we describe advances in our understanding of the mechanisms by which cilia function contributes to cardiac left-right axis development and discuss the latest findings that highlight a broader role for cilia in cardiac development. Specifically, we examine the growing line of evidence connecting cilia function to the pathogenesis of congenital heart disease. Furthermore, we also highlight research from the past 10 years demonstrating the role of cilia function in common cardiac valve disorders, including mitral valve prolapse and aortic valve disease, and describe findings that implicate cardiac cilia in mechanosensation potentially linking haemodynamic and contractile forces with genetic regulation of cardiac development and function. Finally, given the presence of cilia on cardiac fibroblasts, we also explore the potential role of cilia in fibrotic growth and summarize the evidence implicating cardiac cilia in heart regeneration.
Collapse
Affiliation(s)
- Lydia Djenoune
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kathryn Berg
- Department of Paediatrics, Yale University School of Medicine, New Haven, CT, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Martina Brueckner
- Department of Paediatrics, Yale University School of Medicine, New Haven, CT, USA.
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.
| | - Shiaulou Yuan
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
26
|
Pervasive occurrence of splice-site-creating mutations and their possible involvement in genetic disorders. NPJ Genom Med 2022; 7:22. [PMID: 35304488 PMCID: PMC8933504 DOI: 10.1038/s41525-022-00294-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 02/15/2022] [Indexed: 01/06/2023] Open
Abstract
The search for causative mutations in human genetic disorders has mainly focused on mutations that disrupt coding regions or splice sites. Recently, however, it has been reported that mutations creating splice sites can also cause a range of genetic disorders. In this study, we identified 5656 candidate splice-site-creating mutations (SCMs), of which 3942 are likely to be pathogenic, in 4054 genes responsible for genetic disorders. Reanalysis of exome data obtained from ciliopathy patients led us to identify 38 SCMs as candidate causative mutations. We estimate that, by focusing on SCMs, the increase in diagnosis rate is approximately 5.9–8.5% compared to the number of already known pathogenic variants. This finding suggests that SCMs are mutations worth focusing on in the search for causative mutations of genetic disorders.
Collapse
|
27
|
Langel SN, Kelly FL, Brass DM, Nagler AE, Carmack D, Tu JJ, Travieso T, Goswami R, Permar SR, Blasi M, Palmer SM. E-cigarette and food flavoring diacetyl alters airway cell morphology, inflammatory and antiviral response, and susceptibility to SARS-CoV-2. Cell Death Dis 2022; 8:64. [PMID: 35169120 PMCID: PMC8847558 DOI: 10.1038/s41420-022-00855-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 01/18/2022] [Accepted: 01/27/2022] [Indexed: 11/09/2022]
Abstract
Diacetyl (DA) is an α-diketone that is used to flavor microwave popcorn, coffee, and e-cigarettes. Occupational exposure to high levels of DA causes impaired lung function and obstructive airway disease. Additionally, lower levels of DA exposure dampen host defenses in vitro. Understanding DA’s impact on lung epithelium is important for delineating exposure risk on lung health. In this study, we assessed the impact of DA on normal human bronchial epithelial cell (NHBEC) morphology, transcriptional profiles, and susceptibility to SARS-CoV-2 infection. Transcriptomic analysis demonstrated cilia dysregulation, an increase in hypoxia and sterile inflammation associated pathways, and decreased expression of interferon-stimulated genes after DA exposure. Additionally, DA exposure resulted in cilia loss and increased hyaluronan production. After SARS-CoV-2 infection, both genomic and subgenomic SARS-CoV-2 RNA were increased in DA vapor- compared to vehicle-exposed NHBECs. This work suggests that transcriptomic and physiologic changes induced by DA vapor exposure damage cilia and increase host susceptibility to SARS-CoV-2.
Collapse
Affiliation(s)
- Stephanie N Langel
- Duke Center for Human Systems Immunology and Department of Surgery, Durham, NC, USA
| | - Francine L Kelly
- Duke Clinical Research Institute and Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - David M Brass
- Duke Clinical Research Institute and Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Andrew E Nagler
- Duke Clinical Research Institute and Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Dylan Carmack
- Duke Clinical Research Institute and Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Joshua J Tu
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA.,Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Tatianna Travieso
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA.,Department of Medicine, Division of Infectious Diseases, Duke University Medical Center, Durham, NC, USA
| | - Ria Goswami
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Sallie R Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Maria Blasi
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA. .,Department of Medicine, Division of Infectious Diseases, Duke University Medical Center, Durham, NC, USA.
| | - Scott M Palmer
- Duke Clinical Research Institute and Department of Medicine, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
28
|
Biallelic DNAH9 mutations are identified in Chinese patients with defective left-right patterning and cilia-related complex congenital heart disease. Hum Genet 2022; 141:1339-1353. [PMID: 35050399 DOI: 10.1007/s00439-021-02426-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/21/2021] [Indexed: 12/31/2022]
Abstract
Defective left-right (LR) pattering results in a spectrum of laterality disorders including situs inversus totalis (SIT) and heterotaxy syndrome (Htx). Approximately, 50% of patients with primary ciliary dyskinesia (PCD) displayed SIT. Recessive variants in DNAH9 have recently been implicated in patients with situs inversus. Here, we describe six unrelated family trios and 2 sporadic patients with laterality defects and complex congenital heart disease (CHD). Through whole exome sequencing (WES), we identified compound heterozygous mutations in DNAH9 in the affected individuals of these family trios. Ex vivo cDNA amplification revealed that DNAH9 mRNA expression was significantly downregulated in these patients carrying biallelic DNAH9 mutations, which cause a premature stop codon or exon skipping. Transmission electron microscopy (TEM) analysis identified ultrastructural defects of the outer dynein arms in these affected individuals. dnah9 knockdown in zebrafish lead to the disturbance of cardiac left-right patterning without affecting ciliogenesis in Kupffer's vesicle (KV). By generating a Dnah9 knockout (KO) C57BL/6n mouse model, we found that Dnah9 loss leads to compromised cardiac function. In this study, we identified recessive DNAH9 mutations in Chinese patients with cardiac abnormalities and defective LR pattering.
Collapse
|
29
|
Berland S, Rustad CF, Bentsen MHL, Wollen EJ, Turowski G, Johansson S, Houge G, Haukanes BI. Double paternal uniparental isodisomy 7 and 15 presenting with Beckwith-Wiedemann spectrum features. Cold Spring Harb Mol Case Stud 2021; 7:mcs.a006113. [PMID: 34615670 PMCID: PMC8751407 DOI: 10.1101/mcs.a006113] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 08/31/2021] [Indexed: 12/16/2022] Open
Abstract
Here we describe for the first time double paternal uniparental isodisomy (iUPD) 7 and 15 in a baby boy with features in the Beckwith–Wiedemann syndrome spectrum (BWSp) (placentomegaly, hyperinsulinism, enlarged viscera, hemangiomas, and earlobe creases) in addition to conjugated hyperbilirubinemia. His phenotype was also reminiscent of genome-wide paternal uniparental isodisomy. We discuss the most likely origin of the UPDs: a maternal double monosomy 7 and 15 rescued by duplication of the paternal chromosomes after fertilization. So far, paternal UPD7 is not associated with an abnormal phenotype, whereas paternal UPD15 causes Angelman syndrome. Methylation analysis for other clinically relevant imprinting disorders, including BWSp, was normal. Therefore, we hypothesized that the double UPD affected other imprinted genes. To look for such effects, patient fibroblast RNA was isolated and analyzed for differential expression compared to six controls. We did not find apparent transcription differences in imprinted genes outside Chromosomes 7 and 15 in patient fibroblast. PEG10 (7q21.3) was the only paternally imprinted gene on these chromosomes up-regulated beyond double-dose expectation (sixfold). We speculate that a high PEG10 level could have a growth-promoting effect as his phenotype was not related to aberrations in BWS locus on 11p15.5 after DNA, RNA, and methylation testing. However, many genes in gene sets associated with growth were up-regulated. This case broadens the phenotypic spectrum of UPDs but does not show evidence of involvement of an imprinted gene network.
Collapse
Affiliation(s)
- Siren Berland
- Department of Medical Genetics, Haukeland University Hospital, 5021 Bergen, Norway
| | - Cecilie F Rustad
- Department of Medical Genetics, Oslo University Hospital, 0424 Oslo, Norway
| | - Mariann H L Bentsen
- Department of Pediatric and Adolescent Medicine, Haukeland University Hospital, 5021 Bergen, Norway
| | - Embjørg J Wollen
- Department of Pediatric Hepatology, Division of Pediatric and Adolescent Medicine, University of Oslo, Oslo University Hospital HF, 0424 Oslo, Norway
| | - Gitta Turowski
- Department of Pathology, Center for Perinatal and Pregnancy-Related Pathology, Oslo University Hospital-Ullevål, 0424 Oslo, Norway
| | - Stefan Johansson
- Department of Medical Genetics, Haukeland University Hospital, 5021 Bergen, Norway.,Department of Clinical Science, University of Bergen, 5007 Bergen, Norway
| | - Gunnar Houge
- Department of Medical Genetics, Haukeland University Hospital, 5021 Bergen, Norway
| | - Bjørn I Haukanes
- Department of Medical Genetics, Haukeland University Hospital, 5021 Bergen, Norway
| |
Collapse
|
30
|
Paff T, Omran H, Nielsen KG, Haarman EG. Current and Future Treatments in Primary Ciliary Dyskinesia. Int J Mol Sci 2021; 22:9834. [PMID: 34575997 PMCID: PMC8470068 DOI: 10.3390/ijms22189834] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/31/2021] [Accepted: 09/02/2021] [Indexed: 01/05/2023] Open
Abstract
Primary ciliary dyskinesia (PCD) is a rare genetic ciliopathy in which mucociliary clearance is disturbed by the abnormal motion of cilia or there is a severe reduction in the generation of multiple motile cilia. Lung damage ensues due to recurrent airway infections, sometimes even resulting in respiratory failure. So far, no causative treatment is available and treatment efforts are primarily aimed at improving mucociliary clearance and early treatment of bacterial airway infections. Treatment guidelines are largely based on cystic fibrosis (CF) guidelines, as few studies have been performed on PCD. In this review, we give a detailed overview of the clinical studies performed investigating PCD to date, including three trials and several case reports. In addition, we explore precision medicine approaches in PCD, including gene therapy, mRNA transcript and read-through therapy.
Collapse
Affiliation(s)
- Tamara Paff
- Department of Paediatric Pulmonology, Emma Children’s Hospital, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands;
| | - Heymut Omran
- Department of General Pediatrics, University Childrens’s Hospital Muenster, 48149 Muenster, Germany;
| | - Kim G. Nielsen
- Danish PCD Centre, Danish Paediatric Pulmonary Service, Department of Paediatrics and Adolescent Medicine, Righospitalet, Copenhagen University Hospital, DK-2100 Copenhagen, Denmark;
- Department of Clinical Medicine, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Eric G. Haarman
- Department of Paediatric Pulmonology, Emma Children’s Hospital, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands;
| |
Collapse
|
31
|
Xiong Y, Xia H, Yuan L, Deng S, Ding Z, Deng H. Identification of compound heterozygous DNAH11 variants in a Han-Chinese family with primary ciliary dyskinesia. J Cell Mol Med 2021; 25:9028-9037. [PMID: 34405951 PMCID: PMC8435457 DOI: 10.1111/jcmm.16866] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 07/24/2021] [Accepted: 08/04/2021] [Indexed: 12/13/2022] Open
Abstract
Primary ciliary dyskinesia (PCD) is a group of genetically and clinically heterogeneous disorders with motile cilia dysfunction. It is clinically characterized by oto‐sino‐pulmonary diseases and subfertility, and half of the patients have situs inversus (Kartagener syndrome). To identify the genetic cause in a Han‐Chinese pedigree, whole‐exome sequencing was conducted in the 37‐year‐old proband, and then, Sanger sequencing was performed on available family members. Minigene splicing assay was applied to verify the impact of the splice‐site variant. Compound heterozygous variants including a splice‐site variant (c.1974‐1G>C, rs1359107415) and a missense variant (c.7787G>A, p.(Arg2596Gln), rs780492669), in the dynein axonemal heavy chain 11 gene (DNAH11) were identified and confirmed as the disease‐associated variants of this lineage. The minigene expression in vitro revealed that the c.1974‐1G>C variant could cause skipping over exon 12, predicted to result in a truncated protein. This discovery may enlarge the DNAH11 variant spectrum of PCD, promote accurate genetic counselling and contribute to PCD diagnosis.
Collapse
Affiliation(s)
- Ying Xiong
- Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Hong Xia
- Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China.,Department of Emergency, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Lamei Yuan
- Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China.,Disease Genome Research Center, Central South University, Changsha, China
| | - Sheng Deng
- Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China.,Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Zerui Ding
- Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Hao Deng
- Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China.,Disease Genome Research Center, Central South University, Changsha, China.,Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
32
|
Huang J, Yang H, Wang M, Zhao X, Shao S, Zhang F, Que R, Hu Q, Liang T. Gallbladder Adenosquamous Cancer with Situs Inversus Totalis: A Case Report and Literature Review. Onco Targets Ther 2021; 14:4299-4304. [PMID: 34349522 PMCID: PMC8327361 DOI: 10.2147/ott.s319030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 07/13/2021] [Indexed: 12/11/2022] Open
Abstract
Background Situs inversus totalis (SIT) is a rare genetic congenital disease, characterized with complete right-to-left inversion of all the internal organs. We herein describe a meaningful case which was diagnosed as gallbladder adenosquamous carcinoma, a rare histology type of gallbladder cancer, with SIT. Case Presentation A 59-year-old Chinese woman was admitted for persistent epigastric distention and intermittent abdominal pain. The abdominal CT scan revealed a huge mass at the gallbladder bottom, involving the adjacent transverse colon and liver. En-bloc radical resection of the gallbladder cancer, including partial colectomy and hepatectomy with regional node dissection, followed by colocolostomy and Roux-en-Y choledochojejunostomy, was successfully performed. Pathology analysis indicated an adenosquamous carcinoma with positive adenocarcinoma markers (CK7, CK19) and squamous carcinoma markers (CK5/6, P63). Conclusion The SIT anomaly might increase the risk of malignancies by sharing genome mutations, suggesting the importance of surveillance in the SIT settings.
Collapse
Affiliation(s)
- Junming Huang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, People's Republic of China
| | - Hanjin Yang
- Department of Pathology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, People's Republic of China
| | - Meng Wang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, People's Republic of China
| | - Xinyu Zhao
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, People's Republic of China
| | - Shiyi Shao
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, People's Republic of China
| | - Fu Zhang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, People's Republic of China
| | - Risheng Que
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, People's Republic of China
| | - Qida Hu
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, People's Republic of China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, People's Republic of China
| |
Collapse
|
33
|
Antony D, Brunner HG, Schmidts M. Ciliary Dyneins and Dynein Related Ciliopathies. Cells 2021; 10:cells10081885. [PMID: 34440654 PMCID: PMC8391580 DOI: 10.3390/cells10081885] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/19/2021] [Accepted: 07/19/2021] [Indexed: 12/22/2022] Open
Abstract
Although ubiquitously present, the relevance of cilia for vertebrate development and health has long been underrated. However, the aberration or dysfunction of ciliary structures or components results in a large heterogeneous group of disorders in mammals, termed ciliopathies. The majority of human ciliopathy cases are caused by malfunction of the ciliary dynein motor activity, powering retrograde intraflagellar transport (enabled by the cytoplasmic dynein-2 complex) or axonemal movement (axonemal dynein complexes). Despite a partially shared evolutionary developmental path and shared ciliary localization, the cytoplasmic dynein-2 and axonemal dynein functions are markedly different: while cytoplasmic dynein-2 complex dysfunction results in an ultra-rare syndromal skeleto-renal phenotype with a high lethality, axonemal dynein dysfunction is associated with a motile cilia dysfunction disorder, primary ciliary dyskinesia (PCD) or Kartagener syndrome, causing recurrent airway infection, degenerative lung disease, laterality defects, and infertility. In this review, we provide an overview of ciliary dynein complex compositions, their functions, clinical disease hallmarks of ciliary dynein disorders, presumed underlying pathomechanisms, and novel developments in the field.
Collapse
Affiliation(s)
- Dinu Antony
- Center for Pediatrics and Adolescent Medicine, University Hospital Freiburg, Freiburg University Faculty of Medicine, Mathildenstrasse 1, 79106 Freiburg, Germany;
- Genome Research Division, Human Genetics Department, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 KL Nijmegen, The Netherlands;
- Radboud Institute for Molecular Life Sciences (RIMLS), Geert Grooteplein Zuid 10, 6525 KL Nijmegen, The Netherlands
| | - Han G. Brunner
- Genome Research Division, Human Genetics Department, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 KL Nijmegen, The Netherlands;
- Radboud Institute for Molecular Life Sciences (RIMLS), Geert Grooteplein Zuid 10, 6525 KL Nijmegen, The Netherlands
| | - Miriam Schmidts
- Center for Pediatrics and Adolescent Medicine, University Hospital Freiburg, Freiburg University Faculty of Medicine, Mathildenstrasse 1, 79106 Freiburg, Germany;
- Genome Research Division, Human Genetics Department, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 KL Nijmegen, The Netherlands;
- Radboud Institute for Molecular Life Sciences (RIMLS), Geert Grooteplein Zuid 10, 6525 KL Nijmegen, The Netherlands
- Correspondence: ; Tel.: +49-761-44391; Fax: +49-761-44710
| |
Collapse
|
34
|
Sone N, Konishi S, Igura K, Tamai K, Ikeo S, Korogi Y, Kanagaki S, Namba T, Yamamoto Y, Xu Y, Takeuchi K, Adachi Y, Chen-Yoshikawa TF, Date H, Hagiwara M, Tsukita S, Hirai T, Torisawa YS, Gotoh S. Multicellular modeling of ciliopathy by combining iPS cells and microfluidic airway-on-a-chip technology. Sci Transl Med 2021; 13:13/601/eabb1298. [PMID: 34233948 DOI: 10.1126/scitranslmed.abb1298] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 12/07/2020] [Accepted: 05/19/2021] [Indexed: 12/14/2022]
Abstract
Mucociliary clearance is an essential lung function that facilitates the removal of inhaled pathogens and foreign matter unidirectionally from the airway tract and is innately achieved by coordinated ciliary beating of multiciliated cells. Should ciliary function become disturbed, mucus can accumulate in the airway causing subsequent obstruction and potentially recurrent pneumonia. However, it has been difficult to recapitulate unidirectional mucociliary flow using human-derived induced pluripotent stem cells (iPSCs) in vitro and the mechanism governing the flow has not yet been elucidated, hampering the proper humanized airway disease modeling. Here, we combine human iPSCs and airway-on-a-chip technology, to demonstrate the effectiveness of fluid shear stress (FSS) for regulating the global axis of multicellular planar cell polarity (PCP), as well as inducing ciliogenesis, thereby contributing to quantifiable unidirectional mucociliary flow. Furthermore, we applied the findings to disease modeling of primary ciliary dyskinesia (PCD), a genetic disease characterized by impaired mucociliary clearance. The application of an airway cell sheet derived from patient-derived iPSCs and their gene-edited counterparts, as well as genetic knockout iPSCs of PCD causative genes, made it possible to recapitulate the abnormal ciliary functions in organized PCP using the airway-on-a-chip. These findings suggest that the disease model of PCD developed here is a potential platform for making diagnoses and identifying therapeutic targets and that airway reconstruction therapy using mechanical stress to regulate PCP might have therapeutic value.
Collapse
Affiliation(s)
- Naoyuki Sone
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Satoshi Konishi
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan.,Laboratory of Biological Science, Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Koichi Igura
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Koji Tamai
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Satoshi Ikeo
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Yohei Korogi
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Shuhei Kanagaki
- Department of Drug Discovery for Lung Diseases, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Toshinori Namba
- Graduate School of Arts and Sciences, The University of Tokyo, Tokyo 153-8902, Japan
| | - Yuki Yamamoto
- Department of Drug Discovery for Lung Diseases, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Yifei Xu
- Department of Otorhinolaryngology, Head and Neck Surgery, Mie University Graduate School of Medicine, Tsu 514-8507, Japan
| | - Kazuhiko Takeuchi
- Department of Otorhinolaryngology, Head and Neck Surgery, Mie University Graduate School of Medicine, Tsu 514-8507, Japan
| | - Yuichi Adachi
- Department of Pediatrics, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Toyofumi F Chen-Yoshikawa
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan.,Department of Thoracic Surgery, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Hiroshi Date
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Masatoshi Hagiwara
- Department of Anatomy and Developmental Biology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Sachiko Tsukita
- Laboratory of Biological Science, Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan.,Strategic Innovation and Research Center, Teikyo University, Tokyo 173-8605, Japan
| | - Toyohiro Hirai
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Yu-Suke Torisawa
- Hakubi Center for Advanced Research, Kyoto University, Kyoto 615-8540, Japan.,Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Kyoto 615-8540, Japan
| | - Shimpei Gotoh
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan. .,Department of Drug Discovery for Lung Diseases, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
35
|
Xia H, Huang X, Deng S, Xu H, Yang Y, Liu X, Yuan L, Deng H. DNAH11 compound heterozygous variants cause heterotaxy and congenital heart disease. PLoS One 2021; 16:e0252786. [PMID: 34133440 PMCID: PMC8208527 DOI: 10.1371/journal.pone.0252786] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 05/21/2021] [Indexed: 12/16/2022] Open
Abstract
Heterotaxy (HTX), a condition characterized by internal organs not being arranged as expected relative to each other and to the left-right axis, is often accompanied with congenital heart disease (CHD). The purpose was to detect the pathogenic variants in a Chinese family with HTX and CHD. A non-consanguineous Han Chinese family with HTX and CHD, and 200 unrelated healthy subjects were enlisted. Exome sequencing and Sanger sequencing were applied to identify the genetic basis of the HTX family. Compound heterozygous variants, c.3426-1G>A and c.4306C>T (p.(Arg1436Trp)), in the dynein axonemal heavy chain 11 gene (DNAH11) were identified in the proband via exome sequencing and further confirmed by Sanger sequencing. Neither c.3426-1G>A nor c.4306C>T variant in the DNAH11 gene was detected in 200 healthy controls. The DNAH11 c.3426-1G>A variant was predicted as altering the acceptor splice site and most likely affecting splicing. The DNAH11 c.4306C>T variant was predicted to be damaging, which may reduce the phenotype severity. The compound heterozygous variants, c.3426-1G>A and c.4306C>T, in the DNAH11 gene might be the pathogenic alterations resulting in HTX and CHD in this family. These findings broaden the variant spectrum of the DNAH11 gene and increase knowledge used in genetic counseling for the HTX family.
Collapse
Affiliation(s)
- Hong Xia
- Center for Experimental Medicine, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Emergency, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiangjun Huang
- Department of General Surgery, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Sheng Deng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hongbo Xu
- Center for Experimental Medicine, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yan Yang
- Department of Neurology, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xin Liu
- Center for Experimental Medicine, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lamei Yuan
- Center for Experimental Medicine, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hao Deng
- Center for Experimental Medicine, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
36
|
Whole-exome sequencing reveals a combination of extremely rare single-nucleotide polymorphism of DNAH9 and RSPH1 genes in a Japanese fetus with situs viscerum inversus. Med Mol Morphol 2021; 54:275-280. [PMID: 34008076 DOI: 10.1007/s00795-021-00287-5] [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: 01/06/2021] [Accepted: 04/08/2021] [Indexed: 10/21/2022]
Abstract
Randomization of left-right body asymmetry, situs viscerum inversus (heterotaxy), is commonly associated with primary ciliary dyskinesia (PCD) resulting from an abnormal ciliary structure, with approximately 50% of PCD patients exhibiting organ laterality defects. I herein report an intrauterine fetal death case, in which an autopsy revealed two lobes of the bilateral lungs as well as heterotaxy of abdominal organs (right-sided spleen and inversion of the alimentary and biliary organs). Whole-exome sequencing (WES) identified a heterozygous single-nucleotide change (c.12775T>C) in exon 68 of the DNAH9 gene, which is a rare single-nucleotide polymorphism (SNP) of rs746081639 and results in the amino acid change of p.C4259R. WES also identified a rare SNP of rs763089682 (c.121G>A) in the RSPH1 gene that causes a heterozygous amino acid alteration of p.G41R. The frequencies of both SNPs, C in rs746081639 and A in rs763089682, are 0.00000824, and a polyphen-2 analysis predicted these amino acid changes to be probably damaging, with a score of 1.000. The combination of extremely rare SNPs in DNAH9 and RSPH1 genes might have been the possible mechanism underlying the development of the laterality defect in the present case.
Collapse
|
37
|
Haisma S, Weersma RK, Joosse ME, de Koning BAE, de Meij T, Koot BGP, Wolters V, Norbruis O, Daly MJ, Stevens C, Xavier RJ, Koskela J, Rivas MA, Visschedijk MC, Verkade HJ, Barbieri R, Jansen DBH, Festen EAM, van Rheenen PF, van Diemen CC. Exome sequencing in patient-parent trios suggests new candidate genes for early-onset primary sclerosing cholangitis. Liver Int 2021; 41:1044-1057. [PMID: 33590606 PMCID: PMC8252477 DOI: 10.1111/liv.14831] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 01/29/2021] [Accepted: 02/07/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND & AIMS Primary sclerosing cholangitis (PSC) is a rare bile duct disease strongly associated with inflammatory bowel disease (IBD). Whole-exome sequencing (WES) has contributed to understanding the molecular basis of very early-onset IBD, but rare protein-altering genetic variants have not been identified for early-onset PSC. We performed WES in patients diagnosed with PSC ≤ 12 years to investigate the contribution of rare genetic variants to early-onset PSC. METHODS In this multicentre study, WES was performed on 87 DNA samples from 29 patient-parent trios with early-onset PSC. We selected rare (minor allele frequency < 2%) coding and splice-site variants that matched recessive (homozygous and compound heterozygous variants) and dominant (de novo) inheritance in the index patients. Variant pathogenicity was predicted by an in-house developed algorithm (GAVIN), and PSC-relevant variants were selected using gene expression data and gene function. RESULTS In 22 of 29 trios we identified at least 1 possibly pathogenic variant. We prioritized 36 genes, harbouring a total of 54 variants with predicted pathogenic effects. In 18 genes, we identified 36 compound heterozygous variants, whereas in the other 18 genes we identified 18 de novo variants. Twelve of 36 candidate risk genes are known to play a role in transmembrane transport, adaptive and innate immunity, and epithelial barrier function. CONCLUSIONS The 36 candidate genes for early-onset PSC need further verification in other patient cohorts and evaluation of gene function before a causal role can be attributed to its variants.
Collapse
Affiliation(s)
- Sjoukje‐Marije Haisma
- Department of Paediatric Gastroenterology Hepatology and NutritionUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Rinse K. Weersma
- Department of Gastroenterology and HepatologyUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Maria E. Joosse
- Department of Paediatric GastroenterologyErasmus University Medical CenterSophia Children's HospitalRotterdamThe Netherlands
| | - Barbara A. E. de Koning
- Department of Paediatric GastroenterologyErasmus University Medical CenterSophia Children's HospitalRotterdamThe Netherlands
| | - Tim de Meij
- Department of Pediatric GastroenterologyVU University Medical CenterAmsterdamThe Netherlands
| | - Bart G. P. Koot
- Pediatric GastroenterologyEmma Children's HospitalAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Victorien Wolters
- Department of Pediatric GastroenterologyUniversity Medical Center Utrecht – Wilhelmina Children's HospitalUtrechtThe Netherlands
| | - Obbe Norbruis
- Department of PediatricsIsala HospitalZwolleThe Netherlands
| | - Mark J. Daly
- Broad Institute of Harvard and Massachusetts Institute of TechnologyBostonMAUSA
| | - Christine Stevens
- Broad Institute of Harvard and Massachusetts Institute of TechnologyBostonMAUSA
| | | | - Jukka Koskela
- Massachusetts General Hospital, GastroenterologyBostonMAUSA,Institute for Molecular Medicine Finland (FIMM)University of HelsinkiHelsinkiFinland,Clinic of Gastroenterology HelsinkiHelsinki University and Helsinki University HospitalHelsinkiFinland
| | | | - Marijn C. Visschedijk
- Department of Gastroenterology and HepatologyUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Henkjan J. Verkade
- Department of Paediatric Gastroenterology Hepatology and NutritionUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Ruggero Barbieri
- Department of Gastroenterology and HepatologyUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands,Department of GeneticsUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Dianne B. H. Jansen
- Department of Gastroenterology and HepatologyUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Eleonora A. M. Festen
- Department of Gastroenterology and HepatologyUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands,Department of GeneticsUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Patrick F. van Rheenen
- Department of Paediatric Gastroenterology Hepatology and NutritionUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Cleo C. van Diemen
- Department of GeneticsUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| |
Collapse
|
38
|
Abstract
The alignment of visceral and brain asymmetry observed in some vertebrate species raises the question of whether this association also exists in humans. While the visceral and brain systems may have developed asymmetry for different reasons, basic visceral left–right differentiation mechanisms could have been duplicated to establish brain asymmetry. We describe the main phenotypical anomalies and the general mechanism of left–right differentiation of vertebrate visceral and brain laterality. Next, we systematically review the available human studies that explored the prevalence of atypical behavioral and brain asymmetry in visceral situs anomalies, which almost exclusively involved participants with the mirrored visceral organization (situs inversus). The data show no direct link between human visceral and brain functional laterality as most participants with situs inversus show the typical population bias for handedness and brain functional asymmetry, although an increased prevalence of functional crowding may be present. At the same time, several independent studies present evidence for a possible relation between situs inversus and the gross morphological asymmetry of the brain torque with potential differences between subtypes of situs inversus with ciliary and non-ciliary etiologies.
Collapse
|
39
|
Understanding Primary Ciliary Dyskinesia and Other Ciliopathies. J Pediatr 2021; 230:15-22.e1. [PMID: 33242470 PMCID: PMC8690631 DOI: 10.1016/j.jpeds.2020.11.040] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 12/12/2022]
Abstract
Ciliopathies are a collection of disorders related to cilia dysfunction. Cilia are specialized organelles that project from the surface of most cells. Motile and primary (sensory) cilia are essential structures and have wide ranging functions. Our understanding of the genetics, pathophysiology, and clinical manifestations of motile ciliopathies, including primary ciliary dyskinesia (PCD), has rapidly advanced since the disease was linked to ciliary ultrastructural defects nearly five decades ago. We will provide an overview of different types of cilia, their role in child health and disease, focusing on motile ciliopathies, and describe recent advances that have led to improved diagnostics and may yield therapeutic targets to restore ciliary structure and function.
Collapse
|
40
|
Abstract
Situs inversus totalis (SIT) is a rare condition of complete inversion and mirroring of normal human anatomy. The incidence is approximately 1 in 8,000 to 1 in 25,000 live births. SIT is inherited in an autosomal recessive pattern and is associated with multiple gene mutations. It is also commonly seen in a condition known as primary ciliary dyskinesia. A 39-year-old pregnant woman presented to the Labor and Delivery unit to rule out pre-eclampsia due to high blood pressure recordings in the office setting. The infant was delivered preterm at 36 weeks gestation via spontaneous vaginal delivery. The infant presented with symptoms of respiratory distress. The newborn was transferred to the neonatal intensive care unit (NICU) for further work-up and to rule in/rule out an etiology known as Wet Lung. Upon retrieving a chest X-ray for the newborn, the results demonstrated situs inversus totalis. The newborn was transferred to a level III NICU for further management and work-up for other potential etiologies. Situs inversus totalis was not seen on prenatal work-up. In summary, situs inversus totalis is a rare condition which can be associated with other detrimental conditions. In the future, if situs inversus totalis is detected in utero, patients should be instructed to deliver in a setting in which any possible etiology can be accommodated. Pediatricians should follow these infants closely and with caution as common presentations may be obscured due to complete inversion of normal human anatomy. It is also important to screen these infants for other etiologies which may present in later developmental stages such as bronchiectasis and respiratory infections.
Collapse
Affiliation(s)
- Jordan Devera
- Obstetrics and Gynecology, Pediatrics, and Medicine, University of Medicine and Health Sciences, Camps, Basseterre, KNA
| | - Francesca Licandro
- Obstetrics and Gynecology, Pediatrics, and Medicine, University of Medicine and Health Sciences, Camps, Basseterre, KNA
| | - Jean Ramos
- Obstetrics and Gynecology, Pediatrics, and Medicine, University of Medicine and Health Sciences, Camps, Basseterre, KNA
| | | | - Laurel G Yap
- Neonatology, MedStar Harbor Hospital, Baltimore, USA
| |
Collapse
|
41
|
Legendre M, Zaragosi LE, Mitchison HM. Motile cilia and airway disease. Semin Cell Dev Biol 2020; 110:19-33. [PMID: 33279404 DOI: 10.1016/j.semcdb.2020.11.007] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/10/2020] [Accepted: 11/14/2020] [Indexed: 01/10/2023]
Abstract
A finely regulated system of airway epithelial development governs the differentiation of motile ciliated cells of the human respiratory tract, conferring the body's mucociliary clearance defence system. Human cilia dysfunction can arise through genetic mutations and this is a cause of debilitating disease morbidities that confer a greatly reduced quality of life. The inherited human motile ciliopathy disorder, primary ciliary dyskinesia (PCD), can arise from mutations in genes affecting various aspects of motile cilia structure and function through deficient production, transport and assembly of cilia motility components or through defective multiciliogenesis. Our understanding about the development of the respiratory epithelium, motile cilia biology and the implications for human pathology has expanded greatly over the past 20 years since isolation of the first PCD gene, rising to now nearly 50 genes. Systems level insights about cilia motility in health and disease have been made possible through intensive molecular and omics (genomics, transcriptomics, proteomics) research, applied in ciliate organisms and in animal and human disease modelling. Here, we review ciliated airway development and the genetic stratification that underlies PCD, for which the underlying genotype can increasingly be connected to biological mechanism and disease prognostics. Progress in this field can facilitate clinical translation of research advances, with potential for great medical impact, e.g. through improvements in ciliopathy disease diagnosis, management, family counselling and by enhancing the potential for future genetically tailored approaches to disease therapeutics.
Collapse
Affiliation(s)
- Marie Legendre
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Childhood Genetic Disorders, Département de Génétique Médicale, Hôpital Armand-Trousseau, Assistance Publique-Hôpitaux de Paris, Paris 75012, France
| | | | - Hannah M Mitchison
- Genetics and Genomic Medicine, University College London, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK; NIHR Biomedical Research Centre at Great Ormond Street Hospital, London, UK.
| |
Collapse
|
42
|
Zhang Y, Chen W, Zeng W, Lu Z, Zhou X. Biallelic loss of function NEK3 mutations deacetylate α-tubulin and downregulate NUP205 that predispose individuals to cilia-related abnormal cardiac left-right patterning. Cell Death Dis 2020; 11:1005. [PMID: 33230144 PMCID: PMC7684299 DOI: 10.1038/s41419-020-03214-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 11/05/2020] [Indexed: 12/18/2022]
Abstract
Defective left–right (LR) organization involving abnormalities in cilia ultrastructure causes laterality disorders including situs inversus (SI) and heterotaxy (Htx) with the prevalence approximately 1/10,000 births. In this study, we describe two unrelated family trios with abnormal cardiac LR patterning. Through whole-exome sequencing (WES), we identified compound heterozygous mutations (c.805-1G >C; p. Ile269GlnfsTer8/c.1117dupA; p.Thr373AsnfsTer19) (c.29T>C; p.Ile10Thr/c.356A>G; p.His119Arg) of NEK3, encoding a NIMA (never in mitosis A)-related kinase, in two affected individuals, respectively. Protein levels of NEK3 were abrogated in Patient-1 with biallelic loss-of function (LoF) NEK3 mutations that causes premature stop codon. Subsequence transcriptome analysis revealed that NNMT (nicotinamide N-methyltransferase) and SIRT2 (sirtuin2) was upregulated by NEK3 knockdown in human retinal pigment epithelial (RPE) cells in vitro, which associates α-tubulin deacetylation by western blot and immunofluorescence. Transmission electron microscopy (TEM) analysis further identified defective ciliary ultrastructure in Patient-1. Furthermore, inner ring components of nuclear pore complex (NPC) including nucleoporin (NUP)205, NUP188, and NUP155 were significantly downregulated in NEK3-silenced cells. In conclusion, we identified biallelic mutations of NEK3 predispose individual to abnormal cardiac left–right patterning via SIRT2-mediated α-tubulin deacetylation and downregulation of inner ring nucleoporins. Our study suggested that NEK3 could be a candidate gene for human ciliopathies.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Assisted Reproduction, and Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, 201204, Shanghai, China
| | - Weicheng Chen
- Pediatric Cardiovascular Center, Children's Hospital of Fudan University, 201102, Shanghai, China
| | - Weijia Zeng
- School of Life Sciences, Fudan University, 200433, Shanghai, China
| | - Zhouping Lu
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, 201204, Shanghai, China
| | - Xiangyu Zhou
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, 201204, Shanghai, China.
| |
Collapse
|
43
|
Abdelhamed Z, Lukacs M, Cindric S, Ali S, Omran H, Stottmann RW. A novel hypomorphic allele of Spag17 causes primary ciliary dyskinesia phenotypes in mice. Dis Model Mech 2020; 13:dmm045344. [PMID: 32988999 PMCID: PMC7648611 DOI: 10.1242/dmm.045344] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 08/24/2020] [Indexed: 12/22/2022] Open
Abstract
Primary ciliary dyskinesia (PCD) is a human condition of dysfunctional motile cilia characterized by recurrent lung infection, infertility, organ laterality defects and partially penetrant hydrocephalus. We recovered a mouse mutant from a forward genetic screen that developed many of the hallmark phenotypes of PCD. Whole-exome sequencing identified this primary ciliary dyskinesia only (Pcdo) allele to be a nonsense mutation (c.5236A>T) in the Spag17 coding sequence creating a premature stop codon (K1746*). The Pcdo variant abolished several isoforms of SPAG17 in the Pcdo mutant testis but not in the brain. Our data indicate differential requirements for SPAG17 in different types of motile cilia. SPAG17 is essential for proper development of the sperm flagellum and is required for either development or stability of the C1 microtubule structure within the central pair apparatus of the respiratory motile cilia, but not the brain ependymal cilia. We identified changes in ependymal ciliary beating frequency, but these did not appear to alter lateral ventricle cerebrospinal fluid flow. Aqueductal stenosis resulted in significantly slower and abnormally directed cerebrospinal fluid flow, and we suggest that this is the root cause of the hydrocephalus. The Spag17Pcdo homozygous mutant mice are generally viable to adulthood but have a significantly shortened lifespan, with chronic morbidity. Our data indicate that the c.5236A>T Pcdo variant is a hypomorphic allele of Spag17 that causes phenotypes related to motile, but not primary, cilia. Spag17Pcdo is a useful new model for elucidating the molecular mechanisms underlying central pair PCD pathogenesis in the mouse.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Zakia Abdelhamed
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Anatomy and Embryology, Faculty of Medicine (Girl's Section), Al-Azhar University, Cairo 11651, Egypt
| | - Marshall Lukacs
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Medical Scientist Training Program, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Sandra Cindric
- Department of General Pediatrics, University Children's Hospital Münster, 48149 Münster, Germany
| | - Saima Ali
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Heymut Omran
- Department of General Pediatrics, University Children's Hospital Münster, 48149 Münster, Germany
| | - Rolf W Stottmann
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Medical Scientist Training Program, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH 45229, USA
| |
Collapse
|
44
|
So J, Ningappa M, Glessner J, Min J, Ashokkumar C, Ranganathan S, Higgs BW, Li D, Sun Q, Schmitt L, Biery AC, Dobrowolski S, Trautz C, Fuhrman L, Schwartz MC, Klena NT, Fusco J, Prasadan K, Adenuga M, Mohamed N, Yan Q, Chen W, Horne W, Dhawan A, Sharif K, Kelly D, Squires RH, Gittes GK, Hakonarson H, Morell V, Lo C, Subramaniam S, Shin D, Sindhi R. Biliary-Atresia-Associated Mannosidase-1-Alpha-2 Gene Regulates Biliary and Ciliary Morphogenesis and Laterality. Front Physiol 2020; 11:538701. [PMID: 33192543 PMCID: PMC7662016 DOI: 10.3389/fphys.2020.538701] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/23/2020] [Indexed: 01/10/2023] Open
Abstract
Background/Aims Infectious and genetic factors are invoked, respectively in isolated biliary atresia (BA), or syndromic BA, with major extrahepatic anomalies. However, isolated BA is also associated with minor extrahepatic gut and cardiovascular anomalies and multiple susceptibility genes, suggesting common origins. Methods We investigated novel susceptibility genes with genome-wide association, targeted sequencing and tissue staining in BA requiring liver transplantation, independent of BA subtype. Candidate gene effects on morphogenesis, developmental pathways, and ciliogenesis, which regulates left-right patterning were investigated with zebrafish knockdown and mouse knockout models, mouse airway cell cultures, and liver transcriptome analysis. Results Single nucleotide polymorphisms in Mannosidase-1-α-2 (MAN1A2) were significantly associated with BA and with other polymorphisms known to affect MAN1A2 expression but were not differentially enriched in either BA subtype. In zebrafish embryos, man1a2 knockdown caused poor biliary network formation, ciliary dysgenesis in Kupffer’s vesicle, cardiac and liver heterotaxy, and dysregulated egfra and other developmental genes. Suboptimal man1a2 knockdown synergized with suboptimal EGFR signaling or suboptimal knockdown of the EGFR pathway gene, adenosine-ribosylation-factor-6, which had minimal effects individually, to reproduce biliary defects but not heterotaxy. In cultured mouse airway epithelium, Man1a2 knockdown arrested ciliary development and motility. Man1a2–/– mice, which experience respiratory failure, also demonstrated portal and bile ductular inflammation. Human BA liver and Man1a2–/– liver exhibited reduced Man1a2 expression and dysregulated ciliary genes, known to cause multisystem human laterality defects. Conclusion BA requiring transplantation associates with sequence variants in MAN1A2. man1a2 regulates laterality, in addition to hepatobiliary morphogenesis, by regulating ciliogenesis in zebrafish and mice, providing a novel developmental basis for multisystem defects in BA.
Collapse
Affiliation(s)
- Juhoon So
- Department of Developmental Biology, McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Mylarappa Ningappa
- Hillman Center for Pediatric Transplantation of the Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
| | - Joseph Glessner
- Center for Applied Genomics of the Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Jun Min
- Departments of Bioengineering, Cellular and Molecular Medicine, and Computer Science and Engineering, University of California San Diego, La Jolla, CA, United States
| | - Chethan Ashokkumar
- Hillman Center for Pediatric Transplantation of the Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
| | - Sarangarajan Ranganathan
- Division of Pediatric Pathology, Department of Pathology, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Brandon W Higgs
- Hillman Center for Pediatric Transplantation of the Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
| | - Dong Li
- Center for Applied Genomics of the Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Qing Sun
- Hillman Center for Pediatric Transplantation of the Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
| | - Lori Schmitt
- Histology Core Laboratory, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Amy C Biery
- Division of Pediatric Pathology, Department of Pathology, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Steven Dobrowolski
- Division of Pediatric Pathology, Department of Pathology, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Christine Trautz
- Hillman Center for Pediatric Transplantation of the Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
| | - Leah Fuhrman
- Hillman Center for Pediatric Transplantation of the Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
| | | | - Nikolai Thomas Klena
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Joseph Fusco
- Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Krishna Prasadan
- Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Morayooluwa Adenuga
- Hillman Center for Pediatric Transplantation of the Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
| | - Nada Mohamed
- Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Qi Yan
- Departments of Human Genetics and Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Wei Chen
- Departments of Human Genetics and Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - William Horne
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Anil Dhawan
- Paediatric Liver, GI, and Nutrition, King's College Hospital, London, United Kingdom
| | - Khalid Sharif
- Children's Hospital of Birmingham, Birmingham, United Kingdom
| | - Deirdre Kelly
- Children's Hospital of Birmingham, Birmingham, United Kingdom
| | - Robert H Squires
- Pediatric Gastroenterology, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - George K Gittes
- Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Hakon Hakonarson
- Center for Applied Genomics of the Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Victor Morell
- Division of Cardiac Surgery, Department of Cardiothoracic Surgery, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Cecilia Lo
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Shankar Subramaniam
- Departments of Bioengineering, Cellular and Molecular Medicine, and Computer Science and Engineering, University of California San Diego, La Jolla, CA, United States
| | - Donghun Shin
- Department of Developmental Biology, McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Rakesh Sindhi
- Hillman Center for Pediatric Transplantation of the Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
| |
Collapse
|
45
|
Abstract
Motile cilia are highly complex hair-like organelles of epithelial cells lining the surface of various organ systems. Genetic mutations (usually with autosomal recessive inheritance) that impair ciliary beating cause a variety of motile ciliopathies, a heterogeneous group of rare disorders. The pathogenetic mechanisms, clinical symptoms and severity of the disease depend on the specific affected genes and the tissues in which they are expressed. Defects in the ependymal cilia can result in hydrocephalus, defects in the cilia in the fallopian tubes or in sperm flagella can cause female and male subfertility, respectively, and malfunctional motile monocilia of the left-right organizer during early embryonic development can lead to laterality defects such as situs inversus and heterotaxy. If mucociliary clearance in the respiratory epithelium is severely impaired, the disorder is referred to as primary ciliary dyskinesia, the most common motile ciliopathy. No single test can confirm a diagnosis of motile ciliopathy, which is based on a combination of tests including nasal nitric oxide measurement, transmission electron microscopy, immunofluorescence and genetic analyses, and high-speed video microscopy. With the exception of azithromycin, there is no evidence-based treatment for primary ciliary dyskinesia; therapies aim at relieving symptoms and reducing the effects of reduced ciliary motility.
Collapse
|
46
|
Lee L, Ostrowski LE. Motile cilia genetics and cell biology: big results from little mice. Cell Mol Life Sci 2020; 78:769-797. [PMID: 32915243 DOI: 10.1007/s00018-020-03633-5] [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: 04/29/2020] [Revised: 08/11/2020] [Accepted: 09/03/2020] [Indexed: 12/13/2022]
Abstract
Our understanding of motile cilia and their role in disease has increased tremendously over the last two decades, with critical information and insight coming from the analysis of mouse models. Motile cilia form on specific epithelial cell types and typically beat in a coordinated, whip-like manner to facilitate the flow and clearance of fluids along the cell surface. Defects in formation and function of motile cilia result in primary ciliary dyskinesia (PCD), a genetically heterogeneous disorder with a well-characterized phenotype but no effective treatment. A number of model systems, ranging from unicellular eukaryotes to mammals, have provided information about the genetics, biochemistry, and structure of motile cilia. However, with remarkable resources available for genetic manipulation and developmental, pathological, and physiological analysis of phenotype, the mouse has risen to the forefront of understanding mammalian motile cilia and modeling PCD. This is evidenced by a large number of relevant mouse lines and an extensive body of genetic and phenotypic data. More recently, application of innovative cell biological techniques to these models has enabled substantial advancement in elucidating the molecular and cellular mechanisms underlying the biogenesis and function of mammalian motile cilia. In this article, we will review genetic and cell biological studies of motile cilia in mouse models and their contributions to our understanding of motile cilia and PCD pathogenesis.
Collapse
Affiliation(s)
- Lance Lee
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA. .,Department of Pediatrics, Sanford School of Medicine of the University of South Dakota, Sioux Falls, SD, USA.
| | - Lawrence E Ostrowski
- Marsico Lung Institute/Cystic Fibrosis Center and Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
47
|
Zhang B, Ma H, Khan T, Ma A, Li T, Zhang H, Gao J, Zhou J, Li Y, Yu C, Bao J, Ali A, Murtaza G, Yin H, Gao Q, Jiang X, Zhang F, Liu C, Khan I, Zubair M, Hussain HMJ, Khan R, Yousaf A, Yuan L, Lu Y, Xu X, Wang Y, Tao Q, Hao Q, Fang H, Cheng H, Zhang Y, Shi Q. A DNAH17 missense variant causes flagella destabilization and asthenozoospermia. J Exp Med 2020; 217:jem.20182365. [PMID: 31658987 PMCID: PMC7041708 DOI: 10.1084/jem.20182365] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 06/10/2019] [Accepted: 10/03/2019] [Indexed: 12/20/2022] Open
Abstract
Using mice modelling patients’ variant, this study demonstrates that a homozygous DNAH17 missense variant causes asthenozoospermia and specifically destabilizes microtubule doublets 4–7 in flagella, which could be largely due to the storage of sperm in epididymis. Asthenozoospermia is a common cause of male infertility, but its etiology remains incompletely understood. We recruited three Pakistani infertile brothers, born to first-cousin parents, displaying idiopathic asthenozoospermia but no ciliary-related symptoms. Whole-exome sequencing identified a missense variant (c.G5408A, p.C1803Y) in DNAH17, a functionally uncharacterized gene, recessively cosegregating with asthenozoospermia in the family. DNAH17, specifically expressed in testes, was localized to sperm flagella, and the mutation did not alter its localization. However, spermatozoa of all three patients showed higher frequencies of microtubule doublet(s) 4–7 missing at principal piece and end piece than in controls. Mice carrying a homozygous mutation (Dnah17M/M) equivalent to that in patients recapitulated the defects in patients’ sperm tails. Further examinations revealed that the doublets 4–7 were destabilized largely due to the storage of sperm in epididymis. Altogether, we first report that a homozygous DNAH17 missense variant specifically induces doublets 4–7 destabilization and consequently causes asthenozoospermia, providing a novel marker for genetic counseling and diagnosis of male infertility.
Collapse
Affiliation(s)
- Beibei Zhang
- The First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China-Shenyang Jinghua Hospital Joint Center for Human Reproduction and Genetics, Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, China
| | - Hui Ma
- The First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China-Shenyang Jinghua Hospital Joint Center for Human Reproduction and Genetics, Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, China
| | - Teka Khan
- The First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China-Shenyang Jinghua Hospital Joint Center for Human Reproduction and Genetics, Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, China
| | - Ao Ma
- The First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China-Shenyang Jinghua Hospital Joint Center for Human Reproduction and Genetics, Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, China
| | - Tao Li
- The First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China-Shenyang Jinghua Hospital Joint Center for Human Reproduction and Genetics, Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, China
| | - Huan Zhang
- The First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China-Shenyang Jinghua Hospital Joint Center for Human Reproduction and Genetics, Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, China
| | - Jianing Gao
- The First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China-Shenyang Jinghua Hospital Joint Center for Human Reproduction and Genetics, Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, China
| | - Jianteng Zhou
- The First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China-Shenyang Jinghua Hospital Joint Center for Human Reproduction and Genetics, Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, China
| | - Yang Li
- The First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China-Shenyang Jinghua Hospital Joint Center for Human Reproduction and Genetics, Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, China
| | - Changping Yu
- The First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China-Shenyang Jinghua Hospital Joint Center for Human Reproduction and Genetics, Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, China
| | - Jianqiang Bao
- The First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China-Shenyang Jinghua Hospital Joint Center for Human Reproduction and Genetics, Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, China
| | - Asim Ali
- The First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China-Shenyang Jinghua Hospital Joint Center for Human Reproduction and Genetics, Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, China
| | - Ghulam Murtaza
- The First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China-Shenyang Jinghua Hospital Joint Center for Human Reproduction and Genetics, Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, China
| | - Hao Yin
- The First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China-Shenyang Jinghua Hospital Joint Center for Human Reproduction and Genetics, Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, China
| | - Qian Gao
- The First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China-Shenyang Jinghua Hospital Joint Center for Human Reproduction and Genetics, Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, China
| | - Xiaohua Jiang
- The First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China-Shenyang Jinghua Hospital Joint Center for Human Reproduction and Genetics, Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, China
| | - Feng Zhang
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering at School of Life Sciences, Institute of Reproduction and Development, Fudan University, Shanghai, China.,Key Laboratory of Reproduction Regulation of National Population and Family Planning Commission, Collaborative Innovation Center of Genetics and Development, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Chunyu Liu
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering at School of Life Sciences, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Ihsan Khan
- The First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China-Shenyang Jinghua Hospital Joint Center for Human Reproduction and Genetics, Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, China
| | - Muhammad Zubair
- The First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China-Shenyang Jinghua Hospital Joint Center for Human Reproduction and Genetics, Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, China
| | - Hafiz Muhammad Jafar Hussain
- The First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China-Shenyang Jinghua Hospital Joint Center for Human Reproduction and Genetics, Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, China
| | - Ranjha Khan
- The First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China-Shenyang Jinghua Hospital Joint Center for Human Reproduction and Genetics, Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, China
| | - Ayesha Yousaf
- The First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China-Shenyang Jinghua Hospital Joint Center for Human Reproduction and Genetics, Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, China
| | - Limin Yuan
- Analysis and test center, Co-Innovation Center for Modern Production Technology of Grain Crops, Yangzhou University, Yangzhou, China
| | - Yan Lu
- Analysis and test center, Co-Innovation Center for Modern Production Technology of Grain Crops, Yangzhou University, Yangzhou, China
| | - Xiaoling Xu
- Department of Respiration, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yun Wang
- Department of Respiration, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qizhao Tao
- The First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China-Shenyang Jinghua Hospital Joint Center for Human Reproduction and Genetics, Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, China
| | - Qiaomei Hao
- The First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China-Shenyang Jinghua Hospital Joint Center for Human Reproduction and Genetics, Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, China
| | - Hui Fang
- The First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China-Shenyang Jinghua Hospital Joint Center for Human Reproduction and Genetics, Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, China
| | - Hongtao Cheng
- The First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China-Shenyang Jinghua Hospital Joint Center for Human Reproduction and Genetics, Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, China
| | - Yuanwei Zhang
- The First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China-Shenyang Jinghua Hospital Joint Center for Human Reproduction and Genetics, Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, China
| | - Qinghua Shi
- The First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China-Shenyang Jinghua Hospital Joint Center for Human Reproduction and Genetics, Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, China
| |
Collapse
|
48
|
Sun M, Zhang Y, JiyunYang, Wang Y, Tan H, Wang H, Lei T, Li X, Zhang X, Xiong W, Dou K, Ma Y. Novel compound heterozygous DNAAF2 mutations cause primary ciliary dyskinesia in a Han Chinese family. J Assist Reprod Genet 2020; 37:2159-2170. [PMID: 32638265 PMCID: PMC7492306 DOI: 10.1007/s10815-020-01859-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 06/10/2020] [Indexed: 02/05/2023] Open
Abstract
PURPOSE Primary ciliary dyskinesia (PCD), which commonly causes male infertility, is an inherited autosomal recessive disorder. This study aimed to investigate the clinical manifestations and screen mutations associated with the dynein axonemal assembly factor 2 (DNAAF2) gene in a Han Chinese family with PCD. METHODS A three-generation family with PCD was recruited in this study. Eight family members underwent comprehensive medical examinations. Genomic DNA was extracted from the participants' peripheral blood, and targeted next-generation sequencing technology was used to perform the mutation screening. The DNAAF2 expression was analyzed by immunostaining and Western blot. RESULTS The proband exhibited the typical clinical features of PCD. Spermatozoa from the proband showed complete immotility but relatively high viability. Two novel compound heterozygous mutations in the DNAAF2 gene, c.C156A [p.Y52X] and c.C26A [p.S9X], were identified. Both nonsense mutations were detected in the proband, whereas the other unaffected family members carried either none or only one of the two mutations. The two nonsense heterozygous mutations were not detected in the 600 ethnically matched normal controls or in the Genome Aggregation Database. The defect of the DNAAF2 and the outer dynein arms and inner dynein arms were notably observed in the spermatozoa from the proband by immunostaining. CONCLUSION This study identified two novel compound heterozygous mutations of DNAAF2 leading to male infertility as a result of PCD in a Han Chinese family. The findings may enhance the understanding of the pathogenesis of PCD and improve reproductive genetic counseling in China.
Collapse
Affiliation(s)
- Minghan Sun
- Department of Medical Genetics and Division of Human Morbid Genomics, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, 610041, Sichuan, China
- Central of Reproductive Medicine, Department of Obstetrics and Gynecology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, 32 Road West 2, the First Ring, Chengdu, 640072, Sichuan, China
| | - Yi Zhang
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - JiyunYang
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yi Wang
- Central of Reproductive Medicine, Department of Obstetrics and Gynecology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, 32 Road West 2, the First Ring, Chengdu, 640072, Sichuan, China
| | - Hao Tan
- Department of Medical Genetics and Division of Human Morbid Genomics, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, 610041, Sichuan, China
| | - Hailian Wang
- Central of Reproductive Medicine, Department of Obstetrics and Gynecology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, 32 Road West 2, the First Ring, Chengdu, 640072, Sichuan, China
| | - Tiantian Lei
- Central of Reproductive Medicine, Department of Obstetrics and Gynecology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, 32 Road West 2, the First Ring, Chengdu, 640072, Sichuan, China
| | - Xiaojie Li
- Central of Reproductive Medicine, Department of Obstetrics and Gynecology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, 32 Road West 2, the First Ring, Chengdu, 640072, Sichuan, China
| | - Xiaojian Zhang
- Central of Reproductive Medicine, Department of Obstetrics and Gynecology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, 32 Road West 2, the First Ring, Chengdu, 640072, Sichuan, China
| | - Wen Xiong
- Central of Reproductive Medicine, Department of Obstetrics and Gynecology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, 32 Road West 2, the First Ring, Chengdu, 640072, Sichuan, China
| | - Ke Dou
- Central of Reproductive Medicine, Department of Obstetrics and Gynecology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, 32 Road West 2, the First Ring, Chengdu, 640072, Sichuan, China.
| | - Yongxin Ma
- Department of Medical Genetics and Division of Human Morbid Genomics, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
49
|
Yang Y, Jiang C, Zhang X, Liu X, Li J, Qiao X, Liu H, Shen Y. Loss-of-function mutation in DNAH8 induces asthenoteratospermia associated with multiple morphological abnormalities of the sperm flagella. Clin Genet 2020; 98:396-401. [PMID: 32681648 DOI: 10.1111/cge.13815] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/10/2020] [Accepted: 07/13/2020] [Indexed: 11/28/2022]
Abstract
Asthenozoospermia is a common cause of male infertility associated with the reduced motility and/or abnormal morphology of spermatozoa, although its etiology remains incompletely understood. Multiple morphological abnormalities of the sperm flagella (MMAF) is one of the main causes of asthenozoospermia. However, the MMAF-associated genes identified to date cannot explain all the human MMAF cases. Herein, a loss-of-function mutation of DNAH8 was identified in an asthenozoospermia patient with MMAF. Moreover, the negative effect of this mutation on DNAH8 expression was confirmed by immunofluorescence staining and western blotting. Remarkably, it is the first time that DNAH8 is suggested to be associated with human MMAF. Our findings provide strong evidence that a loss-of-function mutation in DNAH8 can cause male infertility with MMAF and that DNAH8 is essential for sperm flagellar formation.
Collapse
Affiliation(s)
- Yihong Yang
- Reproduction Medical Center of West China Second University Hospital, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| | - Chuan Jiang
- Department of Obstetrics/Gynecology, Joint Laboratory of Reproductive Medicine (SCU-CUHK), Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xueguang Zhang
- Department of Obstetrics/Gynecology, Joint Laboratory of Reproductive Medicine (SCU-CUHK), Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xue Liu
- Radiology Department of West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jinghong Li
- Reproduction Medical Center of West China Second University Hospital, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| | - Xiaoyong Qiao
- Reproduction Medical Center of West China Second University Hospital, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| | - Hongqian Liu
- Department of Obstetrics and Gynecology, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Ying Shen
- Department of Obstetrics/Gynecology, Joint Laboratory of Reproductive Medicine (SCU-CUHK), Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
50
|
Namavarian A, Eid A, Goh ESY, Thakur V. A novel DNAH11 variant segregating in a sibship with heterotaxy and implications for genetic counseling. Mol Genet Genomic Med 2020; 8:e1358. [PMID: 32633470 PMCID: PMC7507105 DOI: 10.1002/mgg3.1358] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/19/2020] [Accepted: 05/21/2020] [Indexed: 01/28/2023] Open
Abstract
Background Isomerism or heterotaxy syndrome is the loss of normal asymmetry of the internal thoraco‐abdominal organs in the left‐right axis and is associated with cardiovascular malformations. Mutations within DNAH11 can be associated with primary ciliary dyskinesia and heterotaxy syndromes. Methods We report a family of healthy, nonconsanguinous parents with subsequent pregnancies demonstrating a novel likely pathogenic variant in DNAH11 segregating in a sibship with varied presentations. Result The first affected pregnancy presented with right atrial isomerism. Further DNA testing identified three variants in DNAH11 related to primary ciliary dyskinesia: a maternally inherited heterozygous variant of unknown significance (VUS) c.2772G>A (p.Met924Ile), a maternally inherited novel likely pathogenic variant c.11662C>T (p.Arg3888Cys) as well as a paternally inherited pathogenic c.1648delA variant (p.Arg550GlyfsX16). The second pregnancy inherited the same variants including the pathogenic and likely pathogenic DNAH11 variants and presented with left isomerism and extracardiac abnormalities. Conclusion We present a novel likely pathogenic variant (c.11662C>T) in DNAH11 that has manifested in heterotaxy with variability in phenotypes for subsequent pregnancies of common parents. This report demonstrates that sibship illustrates potential variability in phenotypes associated with the same pathogenic variants within a family and highlights the difficulty in genetic counseling due to the variation in clinical presentation.
Collapse
Affiliation(s)
| | - Anas Eid
- Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Elaine Suk-Ying Goh
- Laboratory Medicine and Genetics, Trillium Health Partners, Mississauga, ON, Canada
| | - Varsha Thakur
- Division of Cardiology, Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|