1
|
Sakamoto K, Miyajima M, Nakajima M, Ogino I, Horikoshi K, Miyahara R, Kawamura K, Karagiozov K, Kamohara C, Nakamura E, Tada N, Kondo A. Loss of Dnah5 Downregulates Dync1h1 Expression, Causing Cortical Development Disorders and Congenital Hydrocephalus. Cells 2024; 13:1882. [PMID: 39594631 PMCID: PMC11593149 DOI: 10.3390/cells13221882] [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: 10/10/2024] [Revised: 11/04/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Dnah5 is associated with primary ciliary dyskinesia in humans. Dnah5-knockout (Dnah5-/- mice develop acute hydrocephalus shortly after birth owing to impaired ciliary motility and cerebrospinal fluid (CSF) stagnation. In contrast to chronic adult-onset hydrocephalus observed in other models, this rapid ventricular enlargement indicates additional factors beyond CSF stagnation. Herein, we investigated the contributors to rapid ventricular enlargement in congenital hydrocephalus. Dnah5-/- mice were generated using CRISPR/Cas9. The expression of dynein, N-cadherin, and nestin in the cerebral cortex was assessed using microarrays and immunostaining. Real-time PCR and Western blotting were performed for gene and protein quantification, respectively. All Dnah5-/- mice developed hydrocephalus, confirmed by electron microscopy, indicating the absence of axonemal outer dynein arms. Ventricular enlargement occurred rapidly, with a 25% reduction in the number of mature neurons in the motor cortex. Dync1h1 expression was decreased, while cytoplasmic dynein levels were 56.3% lower. Levels of nestin and N-cadherin in the lateral ventricular walls decreased by 31.7% and 33.3%, respectively. Reduced cytoplasmic dynein disrupts neurogenesis and axonal growth and reduces neuron cortical density. Hydrocephalus in Dnah5-/- mice may result from cortical maldevelopment due to cytoplasmic dynein deficiency, further exacerbating ventricular enlargement due to CSF stagnation caused by impaired motile ciliary function.
Collapse
Affiliation(s)
- Koichiro Sakamoto
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (M.N.); (I.O.); (K.H.); (R.M.); (K.K.); (K.K.); (C.K.); (A.K.)
| | - Masakazu Miyajima
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (M.N.); (I.O.); (K.H.); (R.M.); (K.K.); (K.K.); (C.K.); (A.K.)
- Department of Neurosurgery, Juntendo Tokyo Koto Geriatric Medical Center, 3-3-20 Shinsuna, Koto-ku, Tokyo 136-0075, Japan
| | - Madoka Nakajima
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (M.N.); (I.O.); (K.H.); (R.M.); (K.K.); (K.K.); (C.K.); (A.K.)
| | - Ikuko Ogino
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (M.N.); (I.O.); (K.H.); (R.M.); (K.K.); (K.K.); (C.K.); (A.K.)
| | - Kou Horikoshi
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (M.N.); (I.O.); (K.H.); (R.M.); (K.K.); (K.K.); (C.K.); (A.K.)
| | - Ryo Miyahara
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (M.N.); (I.O.); (K.H.); (R.M.); (K.K.); (K.K.); (C.K.); (A.K.)
| | - Kaito Kawamura
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (M.N.); (I.O.); (K.H.); (R.M.); (K.K.); (K.K.); (C.K.); (A.K.)
| | - Kostadin Karagiozov
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (M.N.); (I.O.); (K.H.); (R.M.); (K.K.); (K.K.); (C.K.); (A.K.)
| | - Chihiro Kamohara
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (M.N.); (I.O.); (K.H.); (R.M.); (K.K.); (K.K.); (C.K.); (A.K.)
| | - Eri Nakamura
- Department of Genetic Analysis Model Laboratory, Juntendo University Graduate School of Medicine, Hongo Bunkyo-ku, Tokyo 113-8421, Japan; (E.N.); (N.T.)
| | - Nobuhiro Tada
- Department of Genetic Analysis Model Laboratory, Juntendo University Graduate School of Medicine, Hongo Bunkyo-ku, Tokyo 113-8421, Japan; (E.N.); (N.T.)
| | - Akihide Kondo
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (M.N.); (I.O.); (K.H.); (R.M.); (K.K.); (K.K.); (C.K.); (A.K.)
| |
Collapse
|
2
|
Guo Z, Luo Y, Bi Y, Liu L, Qi Y, Yan J, Cai C, Xi C, Tan Y, Yao S, Qu Y, Chen P, Chen J, Wang Y, Mao X, Ye B, Gao S, He G, Bian S. Association between situs inversus and maternal SARS-CoV-2 infection at gestational age 4-6 weeks. MED 2024; 5:1433-1441.e3. [PMID: 39094582 DOI: 10.1016/j.medj.2024.07.009] [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: 02/01/2024] [Revised: 04/11/2024] [Accepted: 07/10/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND A dramatic increase in fetal situs inversus diagnoses by ultrasound in the months following the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) surge of December 2022 in China led us to investigate whether maternal SARS-CoV-2 exposure could be associated with elevated risk of fetal situs inversus. METHODS In this multi-institutional, hospital-based, matched case-control study, we investigated pregnant women who underwent ultrasonographic fetal biometric assessment at gestational weeks 20-24 at our hospitals. Each pregnant woman carrying a situs inversus fetus was randomly matched with four controls based on the date of confinement. Relevant information, including SARS-CoV-2 infection, and other potential risk factors were collected. Conditional logistic regression was used to test possible associations between fetal situs inversus and SARS-CoV-2 infection at different gestational weeks as well as individual risk factors. FINDINGS A total of 52 pregnant women diagnosed with fetal situs inversus between January 1 and October 31, 2023 and 208 matched controls with normal fetuses were enrolled. We found no association between an increased risk of fetal situs inversus with gestational SARS-CoV-2 infection or with other risk factors. However, fetal situs inversus was significantly associated with SARS-CoV-2 infection specifically in gestational weeks 4-6 (adjusted odds ratio [aOR] 6.54 [95% confidence interval 1.76-24.34]), but not with infection at other gestational ages, after adjusting for covariates. CONCLUSIONS Increased risk of fetal situs inversus is significantly associated with maternal SARS-CoV-2 infection at gestational weeks 4-6, corresponding to the fetal developmental window for visceral lateralization in humans. FUNDING National Key R&D Program of China, etc.
Collapse
Affiliation(s)
- Zhenming Guo
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yingchun Luo
- Department of Ultrasonography, Hunan Provincial Maternal and Child Health Care Hospital, University of South China, Changsha, China
| | - Yan Bi
- Department of Prenatal Diagnosis Center, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liangjie Liu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Psychotic Disorders and Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
| | - Yuan Qi
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jin Yan
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Chunhai Cai
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Chenxiang Xi
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yihan Tan
- Department of Ultrasonography, Hunan Provincial Maternal and Child Health Care Hospital, University of South China, Changsha, China
| | - Shifa Yao
- Department of Ultrasonography, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanhui Qu
- Department of Ultrasonography, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping Chen
- Department of Ultrasonography, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiayu Chen
- Clinical and Translation Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yanlin Wang
- Department of Prenatal Diagnosis Center, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Xiao Mao
- National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, University of South China, Changsha, China.
| | - Baoying Ye
- Department of Ultrasonography, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Shaorong Gao
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China; Clinical and Translation Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China.
| | - Guang He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Psychotic Disorders and Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China.
| | - Shan Bian
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China; National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, University of South China, Changsha, China; China Regional Research Center, International Center for Genetic Engineering and Biotechnology, Taizhou, China.
| |
Collapse
|
3
|
Mascho K, Yatsenko SA, Lo CW, Xu X, Johnson J, Helvaty LR, Burns Wechsler S, Murali CN, Lalani SR, Garg V, Hodge JC, McBride KL, Ware SM, Lin JHI. Case Report: An association of left ventricular outflow tract obstruction with 5p deletions. Front Genet 2024; 15:1451746. [PMID: 39492880 PMCID: PMC11527671 DOI: 10.3389/fgene.2024.1451746] [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: 06/19/2024] [Accepted: 09/25/2024] [Indexed: 11/05/2024] Open
Abstract
Introduction 5p deletion syndrome, also called Cri-du-chat syndrome 5p is a rare genetic syndrome with reports up to 36% of patients are associated with congenital heart defects. We investigated the association between left outflow tract obstruction and Cri-du-chat syndrome. Methods A retrospective review of the abnormal microarray cases with congenital heart defects in Children's Hospital of Pittsburgh and the Cytogenomics of Cardiovascular Malformations Consortium. Results A retrospective review at nine pediatric centers identified 4 patients with 5p deletions and left outflow tract obstruction (LVOTO). Three of these patients had additional copy number variants. We present data suggesting an association of LVOTO with 5p deletion with high mortality in the presence of additional copy number variants. Conclusion A rare combination of 5p deletion and left ventricular outflow obstruction was observed in the registry of copy number variants and congenital heart defects.
Collapse
Affiliation(s)
- Kira Mascho
- Division of Pediatric Critical Care, Rainbow Babies and Children’s Hospital, Cleveland, OH, United States
| | | | - Cecilia W. Lo
- University of Pittsburgh, Pittsburgh, PA, United States
| | - Xinxiu Xu
- University of Pittsburgh, Pittsburgh, PA, United States
| | - Jennifer Johnson
- UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Lindsey R. Helvaty
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | | | - Chaya N. Murali
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | - Seema R. Lalani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | - Vidu Garg
- Center for Cardiovascular Research and Heart Center, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Jennelle C. Hodge
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Kim L. McBride
- Center for Cardiovascular Research and Heart Center, Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Medical Genetics, University of Calgary, Calgary, AB, Canada
| | - Stephanie M. Ware
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jiuann-Huey Ivy Lin
- University of Pittsburgh, Pittsburgh, PA, United States
- UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
4
|
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
|
5
|
Geng H, Wang K, Liang D, Ni X, Yu H, Tang D, Lv M, Wu H, Li K, Shen Q, Gao Y, Xu C, Zhou P, Wei Z, Cao Y, Sha Y, Yang X, He X. Further evidence from DNAH12 supports favorable fertility outcomes of infertile males with dynein axonemal heavy chain gene family variants. iScience 2024; 27:110366. [PMID: 39071892 PMCID: PMC11278020 DOI: 10.1016/j.isci.2024.110366] [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: 03/03/2024] [Revised: 04/03/2024] [Accepted: 06/21/2024] [Indexed: 07/30/2024] Open
Abstract
Male infertility is a major concern affecting reproductive health. Biallelic deleterious variants of most DNAH gene family members have been linked to male infertility, with intracytoplasmic sperm injection (ICSI) being an efficacious way to achieve offspring. However, the association between DNAH12 and male infertility is still limited. Here, we identified one homozygous variant and two compound heterozygous variants in DNAH12 from three infertile Chinese men. Semen analysis revealed severe asthenozoospermia, abnormal morphology, and structure of sperm flagella. Furthermore, the Dnah12 knock-out mouse revealed severe spermatogenesis failure and validated the same male infertility phenotype. Favorable fertility outcomes were achieved through ICSI in three human individuals and Dnah12 knock-out mice. Collectively, our study indicated that biallelic variants of DNAH12 can induce male infertility in both human beings and mice. Notably, evidence from DNAH12 enhanced that ICSI was an optimal intervention to achieve favorable fertility outcomes for infertile males with DNAH gene family variants.
Collapse
Affiliation(s)
- Hao Geng
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
| | - Kai Wang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, Anhui, China
| | - Dan Liang
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
| | - Xiaoqing Ni
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, Anhui, China
| | - Hui Yu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, Anhui, China
| | - Dongdong Tang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
| | - Mingrong Lv
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
| | - Huan Wu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
| | - Kuokuo Li
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
| | - Qunshan Shen
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
| | - Yang Gao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, Anhui, China
| | - Chuan Xu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (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, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
| | - Zhaolian Wei
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
| | - Yunxia Cao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
| | - Yanwei Sha
- School of Public Health & Women and Children's Hospital, Xiamen University, Xiamen, Fujian, China
| | - Xiaoyu Yang
- State Key Laboratory of Reproductive Medicine and Offspring Health, The Center for Clinical Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaojin He
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, Anhui, China
| |
Collapse
|
6
|
Yao XJ, Chen Q, Yu HP, Ruan DD, Li SJ, Wu M, Liao LS, Lin XF, Fang ZT, Luo JW, Xie BS. A novel splicing mutation DNAH5 c.13,338 + 5G > C is involved in the pathogenesis of primary ciliary dyskinesia in a family with primary familial brain calcification. BMC Pulm Med 2024; 24:343. [PMID: 39014333 PMCID: PMC11251106 DOI: 10.1186/s12890-024-03164-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: 11/02/2023] [Accepted: 07/11/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND Primary ciliary dyskinesia (PCD) is an autosomal recessive hereditary disease characterized by recurrent respiratory infections. In clinical manifestations, DNAH5 (NM_001361.3) is one of the recessive pathogenic genes. Primary familial brain calcification (PFBC) is a neurodegenerative disease characterized by bilateral calcification in the basal ganglia and other brain regions. PFBC can be inherited in an autosomal dominant or recessive manner. A family with PCD caused by a DNAH5 compound heterozygous variant and PFBC caused by a MYORG homozygous variant was analyzed. METHODS In this study, we recruited three generations of Han families with primary ciliary dyskinesia combined with primary familial brain calcification. Their clinical phenotype data were collected, next-generation sequencing was performed to screen suspected pathogenic mutations in the proband and segregation analysis of families was carried out by Sanger sequencing. The mutant and wild-type plasmids were constructed and transfected into HEK293T cells instantaneously, and splicing patterns were detected by Minigene splicing assay. The structure and function of mutations were analyzed by bioinformatics analysis. RESULTS The clinical phenotypes of the proband (II10) and his sister (II8) were bronchiectasis, recurrent pulmonary infection, multiple symmetric calcifications of bilateral globus pallidus and cerebellar dentate nucleus, paranasal sinusitis in the whole group, and electron microscopy of bronchial mucosa showed that the ciliary axoneme was defective. There was also total visceral inversion in II10 but not in II8. A novel splice variant C.13,338 + 5G > C and a frameshift variant C.4314delT (p. Asn1438lysfs *10) were found in the DNAH5 gene in proband (II10) and II8. c.347_348dupCTGGCCTTCCGC homozygous insertion variation was found in the MYORG of the proband. The two pathogenic genes were co-segregated in the family. Minigene showed that DNAH5 c.13,338 + 5G > C has two abnormal splicing modes: One is that part of the intron bases where the mutation site located is translated, resulting in early translation termination of DNAH5; The other is the mutation resulting in the deletion of exon76. CONCLUSIONS The newly identified DNAH5 splicing mutation c.13,338 + 5G > C is involved in the pathogenesis of PCD in the family, and forms a compound heterozygote with the pathogenic variant DNAH5 c.4314delT lead to the pathogenesis of PCD.
Collapse
Grants
- 2021J02053, 2023J011159, 2022J01996 Natural Science Foundation of Fujian Province
- 2020-822, 2021-157, 2021-848, 2021-917, 2022-840) Fujian Provincial Finance Department
- 2020-822, 2021-157, 2021-848, 2021-917, 2022-840) Fujian Provincial Finance Department
- 2022CXA001, 2021CXB001, 2022CXB002 Medical Innovation Project of Fujian Province
- National famous and old Chinese medicine experts (Xuemei Zhang, Xiaohua Yan) inheritance studio construction project
Collapse
Affiliation(s)
- Xiu-Juan Yao
- Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, no. 134 East Street, Fuzhou, 350001, China
- Respiratory department, Fujian Provincial Hospital, Fuzhou, China
| | - Qian Chen
- Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, no. 134 East Street, Fuzhou, 350001, China
| | - Hong-Ping Yu
- Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, no. 134 East Street, Fuzhou, 350001, China
| | - Dan-Dan Ruan
- Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, no. 134 East Street, Fuzhou, 350001, China
| | - Shi-Jie Li
- Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, no. 134 East Street, Fuzhou, 350001, China
| | - Min Wu
- Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, no. 134 East Street, Fuzhou, 350001, China
| | - Li-Sheng Liao
- Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, no. 134 East Street, Fuzhou, 350001, China
- Department of Hematology, Fujian Provincial Hospital, Fuzhou, China
| | - Xin-Fu Lin
- Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, no. 134 East Street, Fuzhou, 350001, China
- Pediatrics department, Fujian Provincial Hospital, Fuzhou, China
| | - Zhu-Ting Fang
- Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, no. 134 East Street, Fuzhou, 350001, China.
- Interventional Department, Fujian Provincial Hospital, Fuzhou, China.
| | - Jie-Wei Luo
- Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, no. 134 East Street, Fuzhou, 350001, China.
- Department of Traditional Chinese Medicine, Fujian Provincial Hospital, Fuzhou, China.
| | - Bao-Song Xie
- Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, no. 134 East Street, Fuzhou, 350001, China.
- Respiratory department, Fujian Provincial Hospital, Fuzhou, China.
| |
Collapse
|
7
|
Hjeij R, Leslie J, Rizk H, Dworniczak B, Olbrich H, Raidt J, Bode SFN, Gardham A, Stals K, Al-Haggar M, Osman E, Crosby A, Eldesoky T, Baple E, Omran H. Biallelic Variants in MNS1 Are Associated with Laterality Defects and Respiratory Involvement. Cells 2024; 13:1017. [PMID: 38920647 PMCID: PMC11202006 DOI: 10.3390/cells13121017] [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: 04/30/2024] [Revised: 05/27/2024] [Accepted: 06/06/2024] [Indexed: 06/27/2024] Open
Abstract
Defects in motile cilia, termed motile ciliopathies, result in clinical manifestations affecting the respiratory and reproductive system, as well as laterality defects and hydrocephalus. We previously defined biallelic MNS1 variants causing situs inversus and male infertility, mirroring the findings in Mns1-/- mice. Here, we present clinical and genomic findings in five newly identified individuals from four unrelated families affected by MNS1-related disorder. Ciliopathy panel testing and whole exome sequencing identified one previously reported and two novel MNS1 variants extending the genotypic spectrum of disease. A broad spectrum of laterality defects including situs inversus totalis and heterotaxia was confirmed. Interestingly, a single affected six-year-old girl homozygous for an MNS1 nonsense variant presented with a history of neonatal respiratory distress syndrome, recurrent respiratory tract infections, chronic rhinitis, and wet cough. Accordingly, immunofluorescence analysis showed the absence of MNS1 from the respiratory epithelial cells of this individual. Two other individuals with hypomorphic variants showed laterality defects and mild respiratory phenotype. This study represents the first observation of heterotaxia and respiratory disease in individuals with biallelic MNS1 variants, an important extension of the phenotype associated with MNS1-related motile ciliopathy disorder.
Collapse
Grants
- HJ 7/1-1, HJ 7/1-3, OM6/7, OM6/8, OM6/10, OM6/14, OM6/16, CRU 326, OM6/11, RA3522/1-1, OL 450/1 Deutsche Forschungsgemeinschaft
- Om2/009/12, Om2/015/16, Om2/010/20 Institute for Interdisciplinary Medicine
Collapse
Affiliation(s)
- Rim Hjeij
- Department of General Pediatrics, University Hospital Muenster, 48149 Muenster, Germany; (B.D.); (H.O.); (J.R.); (H.O.)
| | - Joseph Leslie
- Institute of Biomedical and Clinical Science, RILD Wellcome Wolfson Centre, University of Exeter Medical School, Royal Devon University Healthcare NHS Foundation Trust, Exeter EX2 5DW, UK; (J.L.); (A.C.); (E.B.)
| | - Hoda Rizk
- Department of Pediatrics, Faculty of Medicine, University of Mansoura, Mansoura 35516, Egypt; (H.R.); (E.O.); (T.E.)
| | - Bernd Dworniczak
- Department of General Pediatrics, University Hospital Muenster, 48149 Muenster, Germany; (B.D.); (H.O.); (J.R.); (H.O.)
| | - Heike Olbrich
- Department of General Pediatrics, University Hospital Muenster, 48149 Muenster, Germany; (B.D.); (H.O.); (J.R.); (H.O.)
| | - Johanna Raidt
- Department of General Pediatrics, University Hospital Muenster, 48149 Muenster, Germany; (B.D.); (H.O.); (J.R.); (H.O.)
| | | | - Alice Gardham
- North West Thames Regional Genetic Service, North West London Hospitals, London HA1 2UJ, UK;
| | - Karen Stals
- Exeter Genomics Laboratory (NHS South West Genomic Laboratory Hub), Royal Devon University Healthcare NHS Foundation Trust, Exeter EX2 5DW, UK;
| | - Mohammad Al-Haggar
- Genetics Unit, Pediatrics Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Engy Osman
- Department of Pediatrics, Faculty of Medicine, University of Mansoura, Mansoura 35516, Egypt; (H.R.); (E.O.); (T.E.)
| | - Andrew Crosby
- Institute of Biomedical and Clinical Science, RILD Wellcome Wolfson Centre, University of Exeter Medical School, Royal Devon University Healthcare NHS Foundation Trust, Exeter EX2 5DW, UK; (J.L.); (A.C.); (E.B.)
| | - Tarek Eldesoky
- Department of Pediatrics, Faculty of Medicine, University of Mansoura, Mansoura 35516, Egypt; (H.R.); (E.O.); (T.E.)
| | - Emma Baple
- Institute of Biomedical and Clinical Science, RILD Wellcome Wolfson Centre, University of Exeter Medical School, Royal Devon University Healthcare NHS Foundation Trust, Exeter EX2 5DW, UK; (J.L.); (A.C.); (E.B.)
- Peninsula Clinical Genetics Service, Royal Devon & Exeter Hospital (Heavitree), Exeter EX1 2ED, UK
| | - Heymut Omran
- Department of General Pediatrics, University Hospital Muenster, 48149 Muenster, Germany; (B.D.); (H.O.); (J.R.); (H.O.)
| |
Collapse
|
8
|
Despotes KA, Zariwala MA, Davis SD, Ferkol TW. Primary Ciliary Dyskinesia: A Clinical Review. Cells 2024; 13:974. [PMID: 38891105 PMCID: PMC11171568 DOI: 10.3390/cells13110974] [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: 05/08/2024] [Revised: 05/31/2024] [Accepted: 06/01/2024] [Indexed: 06/21/2024] Open
Abstract
Primary ciliary dyskinesia (PCD) is a rare, genetically heterogeneous, motile ciliopathy, characterized by neonatal respiratory distress, recurrent upper and lower respiratory tract infections, subfertility, and laterality defects. Diagnosis relies on a combination of tests for confirmation, including nasal nitric oxide (nNO) measurements, high-speed videomicroscopy analysis (HSVMA), immunofluorescent staining, axonemal ultrastructure analysis via transmission electron microscopy (TEM), and genetic testing. Notably, there is no single gold standard confirmatory or exclusionary test. Currently, 54 causative genes involved in cilia assembly, structure, and function have been linked to PCD; this rare disease has a spectrum of clinical manifestations and emerging genotype-phenotype relationships. In this review, we provide an overview of the structure and function of motile cilia, the emerging genetics and pathophysiology of this rare disease, as well as clinical features associated with motile ciliopathies, novel diagnostic tools, and updates on genotype-phenotype relationships in PCD.
Collapse
Affiliation(s)
- Katherine A. Despotes
- Department of Pediatrics, UNC School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Maimoona A. Zariwala
- Department of Pediatrics, UNC School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Marsico Lung Institute, UNC School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Stephanie D. Davis
- Department of Pediatrics, UNC School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Thomas W. Ferkol
- Department of Pediatrics, UNC School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Marsico Lung Institute, UNC School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
9
|
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
|
10
|
Dong L, Zhang L, Li X, Mei S, Shen Y, Fu L, Zhao S, Tang X, Tang Y. Clinical and genetic analysis of two patients with primary ciliary dyskinesia caused by a novel variant of DNAAF2. BMC Pediatr 2023; 23:616. [PMID: 38053031 PMCID: PMC10696777 DOI: 10.1186/s12887-023-04185-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 07/08/2023] [Indexed: 12/07/2023] Open
Abstract
BACKGROUND The study describes the clinical manifestations and variant screening of two Chinese siblings with primary ciliary dyskinesia (PCD). They carry the same DNAAF2 genotype, which is an extremely rare PCD genotype in the Chinese population. In addition, the study illustrated an overview of published variants on DNAAF2 to date. METHODS A two-child family was recruited for the study. Clinical manifestations, laboratory tests, bronchoscopic and otoscopic images, and radiographic data were collected. Whole blood was collected from siblings and their parents for whole-exome sequencing (WES) and Sanger sequencing to screen variants. RESULTS The two siblings exhibited typical clinical manifestations of PCD. Two compound heterozygous variants in DNAAF2 were detected in both by WES. Nonsense variant c.156 C>A and frameshift variant c.177_178insA, which was a novel variant. CONCLUSION The study identified a novel variant of DNAAF2 in Chinese children with a typical phenotype of PCD, which may enrich our knowledge of the clinical, diagnostic and genetic information of DNAAF2-induced PCD in children.
Collapse
Affiliation(s)
- Lili Dong
- Department of Respiratory Medicine, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Lei Zhang
- Department of Respiratory Medicine, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Xiao Li
- Department of Respiratory Medicine, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Shiyue Mei
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Yuelin Shen
- Department of Respiratory Medicine, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
- Department of Respiratory Medicine, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Libing Fu
- Department of Pathology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Shunying Zhao
- Department of Respiratory Medicine, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Xiaolei Tang
- Department of Respiratory Medicine, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China.
- Department of Respiratory Medicine, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China.
| | - Yu Tang
- Department of Respiratory Medicine, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China.
| |
Collapse
|
11
|
Giriyappagoudar M, Vastrad B, Horakeri R, Vastrad C. Study on Potential Differentially Expressed Genes in Idiopathic Pulmonary Fibrosis by Bioinformatics and Next-Generation Sequencing Data Analysis. Biomedicines 2023; 11:3109. [PMID: 38137330 PMCID: PMC10740779 DOI: 10.3390/biomedicines11123109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 10/31/2023] [Accepted: 11/02/2023] [Indexed: 12/24/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic progressive lung disease with reduced quality of life and earlier mortality, but its pathogenesis and key genes are still unclear. In this investigation, bioinformatics was used to deeply analyze the pathogenesis of IPF and related key genes, so as to investigate the potential molecular pathogenesis of IPF and provide guidance for clinical treatment. Next-generation sequencing dataset GSE213001 was obtained from Gene Expression Omnibus (GEO), and the differentially expressed genes (DEGs) were identified between IPF and normal control group. The DEGs between IPF and normal control group were screened with the DESeq2 package of R language. The Gene Ontology (GO) and REACTOME pathway enrichment analyses of the DEGs were performed. Using the g:Profiler, the function and pathway enrichment analyses of DEGs were performed. Then, a protein-protein interaction (PPI) network was constructed via the Integrated Interactions Database (IID) database. Cytoscape with Network Analyzer was used to identify the hub genes. miRNet and NetworkAnalyst databaseswereused to construct the targeted microRNAs (miRNAs), transcription factors (TFs), and small drug molecules. Finally, receiver operating characteristic (ROC) curve analysis was used to validate the hub genes. A total of 958 DEGs were screened out in this study, including 479 up regulated genes and 479 down regulated genes. Most of the DEGs were significantly enriched in response to stimulus, GPCR ligand binding, microtubule-based process, and defective GALNT3 causes HFTC. In combination with the results of the PPI network, miRNA-hub gene regulatory network and TF-hub gene regulatory network, hub genes including LRRK2, BMI1, EBP, MNDA, KBTBD7, KRT15, OTX1, TEKT4, SPAG8, and EFHC2 were selected. Cyclothiazide and rotigotinethe are predicted small drug molecules for IPF treatment. Our findings will contribute to identification of potential biomarkers and novel strategies for the treatment of IPF, and provide a novel strategy for clinical therapy.
Collapse
Affiliation(s)
- Muttanagouda Giriyappagoudar
- Department of Radiation Oncology, Karnataka Institute of Medical Sciences (KIMS), Hubballi 580022, Karnataka, India;
| | - Basavaraj Vastrad
- Department of Pharmaceutical Chemistry, K.L.E. Socitey’s College of Pharmacy, Gadag 582101, Karnataka, India;
| | - Rajeshwari Horakeri
- Department of Computer Science, Govt First Grade College, Hubballi 580032, Karnataka, India;
| | - Chanabasayya Vastrad
- Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karnataka, India
| |
Collapse
|
12
|
Wang Y, Guo Z, Ye B, Liu L, Mao X, Luo Y, Gao S, He G, Bian S. Association of SARS-CoV-2 Infection during Early Weeks of Gestation with Situs Inversus. N Engl J Med 2023; 389:1722-1724. [PMID: 37913512 PMCID: PMC10755830 DOI: 10.1056/nejmc2309215] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Affiliation(s)
- Yanlin Wang
- International Peace Maternity and Child Health Hospital of China Welfare Institute, Shanghai, China
| | | | - Baoying Ye
- International Peace Maternity and Child Health Hospital of China Welfare Institute, Shanghai, China
| | | | - Xiao Mao
- Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
| | - Yingchun Luo
- Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
| | | | - Guang He
- Shanghai Jiao Tong University, Shanghai, China
| | | |
Collapse
|
13
|
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: 5] [Impact Index Per Article: 2.5] [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
|
14
|
Hjeij R, Aprea I, Poeta M, Nöthe-Menchen T, Bracht D, Raidt J, Honecker BI, Dougherty GW, Olbrich H, Schwartz O, Keller U, Nüsse H, Diderich KEM, Vogelberg C, Santamaria F, Omran H. Pathogenic variants in CLXN encoding the outer dynein arm docking-associated calcium-binding protein calaxin cause primary ciliary dyskinesia. Genet Med 2023; 25:100798. [PMID: 36727596 DOI: 10.1016/j.gim.2023.100798] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 01/24/2023] [Accepted: 01/24/2023] [Indexed: 02/03/2023] Open
Abstract
PURPOSE Primary ciliary dyskinesia (PCD) is a heterogeneous disorder that includes respiratory symptoms, laterality defects, and infertility caused by dysfunction of motile cilia. Most PCD-causing variants result in abnormal outer dynein arms (ODAs), which provide the generative force for respiratory ciliary beating and proper mucociliary clearance. METHODS In addition to studies in mouse and planaria, clinical exome sequencing and functional analyses in human were performed. RESULTS In this study, we identified homozygous pathogenic variants in CLXN (EFCAB1/ODAD5) in 3 individuals with laterality defects and respiratory symptoms. Consistently, we found that Clxn is expressed in mice left-right organizer. Transmission electron microscopy depicted ODA defects in distal ciliary axonemes. Immunofluorescence microscopy revealed absence of CLXN from the ciliary axonemes, absence of the ODA components DNAH5, DNAI1, and DNAI2 from the distal axonemes, and mislocalization or absence of DNAH9. In addition, CLXN was undetectable in ciliary axonemes of individuals with defects in the ODA-docking machinery: ODAD1, ODAD2, ODAD3, and ODAD4. Furthermore, SMED-EFCAB1-deficient planaria displayed ciliary dysmotility. CONCLUSION Our results revealed that pathogenic variants in CLXN cause PCD with defects in the assembly of distal ODAs in the respiratory cilia. CLXN should be referred to as ODA-docking complex-associated protein ODAD5.
Collapse
Affiliation(s)
- Rim Hjeij
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Isabella Aprea
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Marco Poeta
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Tabea Nöthe-Menchen
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Diana Bracht
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Johanna Raidt
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Barbara I Honecker
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Gerard W Dougherty
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Heike Olbrich
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Oliver Schwartz
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Ulrike Keller
- Institute of Medical Physics and Biophysics, University of Muenster, Muenster, Germany
| | - Harald Nüsse
- Institute of Medical Physics and Biophysics, University of Muenster, Muenster, Germany
| | | | - Christian Vogelberg
- Pediatric Department, University Hospital Carl Gustav Carus Dresden, Technical University Dresden, Dresden, Germany
| | - Francesca Santamaria
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Heymut Omran
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany.
| |
Collapse
|
15
|
Barber AT, Shapiro AJ, Davis SD, Ferkol TW, Atkinson JJ, Sagel SD, Dell SD, Olivier KN, Milla CE, Rosenfeld M, Li L, Lin FC, Sullivan KM, Capps NA, Zariwala MA, Knowles MR, Leigh MW. Laterality Defects in Primary Ciliary Dyskinesia: Relationship to Ultrastructural Defect or Genotype. Ann Am Thorac Soc 2023; 20:397-405. [PMID: 36342963 PMCID: PMC9993158 DOI: 10.1513/annalsats.202206-487oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 11/07/2022] [Indexed: 11/09/2022] Open
Abstract
Rationale: The association between organ laterality abnormalities and ciliary ultrastructural defect or genotype in primary ciliary dyskinesia is poorly understood. Objectives: To determine if there is an association between presence and/or type of laterality abnormality and ciliary ultrastructural defect or genotype. Methods: Participants with primary ciliary dyskinesia in a multicenter, prospective study were grouped based on ciliary ultrastructural defect or genotype. In a retrospective analysis of these data, the association of ciliary ultrastructural defect or genotype and likelihood of a laterality abnormality was evaluated by logistic regression adjusted for presence of two loss-of-function versus one or more not-loss-of-function variants. Results: Of 559 participants, 286 (51.2%), 215 (38.5%), and 58 (10.4%) were identified as having situs solitus, situs inversustotalis, and situs ambiguus, respectively; heterotaxy, defined as situs ambiguus with complex cardiovascular defects, was present in 14 (2.5%). Compared with the group with inner dynein arm defects with microtubular disorganization, laterality defects were more likely in the outer dynein arm defects group (odds ratio [OR], 2.07; 95% confidence interval [CI], 1.21-3.54; P < 0.01) and less likely in the normal/near normal ultrastructure group (OR, 0.04; 95% CI, 0.013-0.151; P < 0.01). Heterotaxy was present in 11 of 242 (4.5%) in the outer dynein arm defects group but 0 of 96 in the inner dynein arm defects with microtubular disorganization group (P = 0.038). Conclusion: In primary ciliary dyskinesia, risk of a laterality abnormality differs by ciliary ultrastructural defect. Pathophysiologic mechanisms underlying these differences require further exploration.
Collapse
Affiliation(s)
| | - Adam J. Shapiro
- Department of Pediatrics, McGill University Health Centre Research Institute, Montreal, Quebec, Canada
| | | | | | - Jeffrey J. Atkinson
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Scott D. Sagel
- Department of Pediatrics, Children’s Hospital Colorado, University of Colorado School of Medicine, Aurora, Colorado
| | - Sharon D. Dell
- Department of Pediatrics, BC Children’s Hospital, Vancouver, British Columbia, Canada
| | - Kenneth N. Olivier
- Pulmonary Branch, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Carlos E. Milla
- Department of Pediatrics, Stanford University, Palo Alto, California
| | - Margaret Rosenfeld
- Department of Pediatrics, Seattle Children’s Hospital, University of Washington School of Medicine, Seattle, Washington; and
| | - Lang Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Feng-Chang Lin
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | | | | | | | | - Margaret W. Leigh
- Marsico Lung Institute
- Department of Pediatrics, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
16
|
Forrest K, Barricella AC, Pohar SA, Hinman AM, Amack JD. Understanding laterality disorders and the left-right organizer: Insights from zebrafish. Front Cell Dev Biol 2022; 10:1035513. [PMID: 36619867 PMCID: PMC9816872 DOI: 10.3389/fcell.2022.1035513] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Vital internal organs display a left-right (LR) asymmetric arrangement that is established during embryonic development. Disruption of this LR asymmetry-or laterality-can result in congenital organ malformations. Situs inversus totalis (SIT) is a complete concordant reversal of internal organs that results in a low occurrence of clinical consequences. Situs ambiguous, which gives rise to Heterotaxy syndrome (HTX), is characterized by discordant development and arrangement of organs that is associated with a wide range of birth defects. The leading cause of health problems in HTX patients is a congenital heart malformation. Mutations identified in patients with laterality disorders implicate motile cilia in establishing LR asymmetry. However, the cellular and molecular mechanisms underlying SIT and HTX are not fully understood. In several vertebrates, including mouse, frog and zebrafish, motile cilia located in a "left-right organizer" (LRO) trigger conserved signaling pathways that guide asymmetric organ development. Perturbation of LRO formation and/or function in animal models recapitulates organ malformations observed in SIT and HTX patients. This provides an opportunity to use these models to investigate the embryological origins of laterality disorders. The zebrafish embryo has emerged as an important model for investigating the earliest steps of LRO development. Here, we discuss clinical characteristics of human laterality disorders, and highlight experimental results from zebrafish that provide insights into LRO biology and advance our understanding of human laterality disorders.
Collapse
Affiliation(s)
- Kadeen Forrest
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Alexandria C. Barricella
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Sonny A. Pohar
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Anna Maria Hinman
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Jeffrey D. Amack
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
- BioInspired Syracuse: Institute for Material and Living Systems, Syracuse, NY, United States
| |
Collapse
|
17
|
Yang W, Chen L, Guo J, Shi F, Yang Q, Xie L, Lu D, Li Y, Luo J, Wang L, Qiu L, Chen T, Li Y, Zhang R, Chen L, Xu W, Liu H. Multiomics Analysis of a DNAH5-Mutated PCD Organoid Model Revealed the Key Role of the TGF-β/BMP and Notch Pathways in Epithelial Differentiation and the Immune Response in DNAH5-Mutated Patients. Cells 2022; 11:cells11244013. [PMID: 36552777 PMCID: PMC9776854 DOI: 10.3390/cells11244013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/08/2022] [Accepted: 12/10/2022] [Indexed: 12/14/2022] Open
Abstract
Dynein axonemal heavy chain 5 (DNAH5) is the most mutated gene in primary ciliary dyskinesia (PCD), leading to abnormal cilia ultrastructure and function. Few studies have revealed the genetic characteristics and pathogenetic mechanisms of PCD caused by DNAH5 mutation. Here, we established a child PCD airway organoid directly from the bronchoscopic biopsy of a patient with the DNAH5 mutation. The motile cilia in the organoid were observed and could be stably maintained for an extended time. We further found abnormal ciliary function and a decreased immune response caused by the DNAH5 mutation through single-cell RNA sequencing (scRNA-Seq) and proteomic analyses. Additionally, the directed induction of the ciliated cells, regulated by TGF-β/BMP and the Notch pathway, also increased the expression of inflammatory cytokines. Taken together, these results demonstrated that the combination of multiomics analysis and organoid modelling could reveal the close connection between the immune response and the DNAH5 gene.
Collapse
Affiliation(s)
- Wenhao Yang
- Department of Paediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu 610000, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610000, China
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu 610000, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu 610000, China
- Sichuan Birth Defects Clinical Research Centre, West China Second University Hospital, Sichuan University, Chengdu 610000, China
| | - Lina Chen
- Department of Paediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu 610000, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610000, China
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu 610000, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu 610000, China
- Sichuan Birth Defects Clinical Research Centre, West China Second University Hospital, Sichuan University, Chengdu 610000, China
| | - Juncen Guo
- Department of Obstetrics/Gynaecology, Joint Laboratory of Reproductive Medicine (SCU-CUHK), Key Laboratory of Obstetric, Gynaecologic, and Paediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610000, China
| | - Fang Shi
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610000, China
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu 610000, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu 610000, China
- Sichuan Birth Defects Clinical Research Centre, West China Second University Hospital, Sichuan University, Chengdu 610000, China
| | - Qingxin Yang
- Sichuan Birth Defects Clinical Research Centre, West China Second University Hospital, Sichuan University, Chengdu 610000, China
| | - Liang Xie
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610000, China
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu 610000, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu 610000, China
- Sichuan Birth Defects Clinical Research Centre, West China Second University Hospital, Sichuan University, Chengdu 610000, China
| | - Danli Lu
- Department of Paediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu 610000, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610000, China
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu 610000, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu 610000, China
- Sichuan Birth Defects Clinical Research Centre, West China Second University Hospital, Sichuan University, Chengdu 610000, China
| | - Yingna Li
- Sichuan Birth Defects Clinical Research Centre, West China Second University Hospital, Sichuan University, Chengdu 610000, China
| | - Jiaxin Luo
- Department of Paediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu 610000, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610000, China
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu 610000, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu 610000, China
- Sichuan Birth Defects Clinical Research Centre, West China Second University Hospital, Sichuan University, Chengdu 610000, China
| | - Li Wang
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu 610000, China
| | - Li Qiu
- Department of Paediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu 610000, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610000, China
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu 610000, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu 610000, China
- Sichuan Birth Defects Clinical Research Centre, West China Second University Hospital, Sichuan University, Chengdu 610000, China
| | - Ting Chen
- Department of Paediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu 610000, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610000, China
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu 610000, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu 610000, China
- Sichuan Birth Defects Clinical Research Centre, West China Second University Hospital, Sichuan University, Chengdu 610000, China
| | - Yan Li
- Department of Paediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu 610000, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610000, China
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu 610000, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu 610000, China
- Sichuan Birth Defects Clinical Research Centre, West China Second University Hospital, Sichuan University, Chengdu 610000, China
| | - Rui Zhang
- Department of Paediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu 610000, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610000, China
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu 610000, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu 610000, China
- Sichuan Birth Defects Clinical Research Centre, West China Second University Hospital, Sichuan University, Chengdu 610000, China
| | - Lu Chen
- Sichuan Birth Defects Clinical Research Centre, West China Second University Hospital, Sichuan University, Chengdu 610000, China
| | - Wenming Xu
- Department of Obstetrics/Gynaecology, Joint Laboratory of Reproductive Medicine (SCU-CUHK), Key Laboratory of Obstetric, Gynaecologic, and Paediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610000, China
- Correspondence: (W.X.); (H.L.)
| | - Hanmin Liu
- Department of Paediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu 610000, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610000, China
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu 610000, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu 610000, China
- Sichuan Birth Defects Clinical Research Centre, West China Second University Hospital, Sichuan University, Chengdu 610000, China
- Correspondence: (W.X.); (H.L.)
| |
Collapse
|
18
|
Dnah9 mutant mice and organoid models recapitulate the clinical features of patients with PCD and provide an excellent platform for drug screening. Cell Death Dis 2022; 13:559. [PMID: 35729109 PMCID: PMC9210797 DOI: 10.1038/s41419-022-05010-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 05/30/2022] [Accepted: 06/08/2022] [Indexed: 01/21/2023]
Abstract
Primary cilia dyskinesia (PCD) is a rare genetic disease caused by ciliary structural or functional defects. It causes severe outcomes in patients, including recurrent upper and lower airway infections, progressive lung failure, and randomization of heterotaxy. To date, although 50 genes have been shown to be responsible for PCD, the etiology remains elusive. Meanwhile, owing to the lack of a model mimicking the pathogenesis that can be used as a drug screening platform, thereby slowing the development of related therapies. In the current study, we identified compound mutation of DNAH9 in a patient with PCD with the following clinical features: recurrent respiratory tract infections, low lung function, and ultrastructural defects of the outer dynein arms (ODAs). Bioinformatic analysis, structure simulation assay, and western blot analysis showed that the mutations affected the structure and expression of DNAH9 protein. Dnah9 knock-down (KD) mice recapitulated the patient phenotypes, including low lung function, mucin accumulation, and increased immune cell infiltration. Immunostaining, western blot, and co-immunoprecipitation analyses were performed to clarify that DNAH9 interacted with CCDC114/GAS8 and diminished their protein levels. Furthermore, we constructed an airway organoid of Dnah9 KD mice and discovered that it could mimic the key features of the PCD phenotypes. We then used organoid as a drug screening model to identify mitochondrial-targeting drugs that can partially elevate cilia beating in Dnah9 KD organoid. Collectively, our results demonstrated that Dnah9 KD mice and an organoid model can recapture the clinical features of patients with PCD and provide an excellent drug screening platform for human ciliopathies.
Collapse
|
19
|
Braschi B, Omran H, Witman GB, Pazour GJ, Pfister KK, Bruford EA, King SM. Consensus nomenclature for dyneins and associated assembly factors. J Cell Biol 2022; 221:e202109014. [PMID: 35006274 PMCID: PMC8754002 DOI: 10.1083/jcb.202109014] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 09/10/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022] Open
Abstract
Dyneins are highly complex, multicomponent, microtubule-based molecular motors. These enzymes are responsible for numerous motile behaviors in cytoplasm, mediate retrograde intraflagellar transport (IFT), and power ciliary and flagellar motility. Variants in multiple genes encoding dyneins, outer dynein arm (ODA) docking complex subunits, and cytoplasmic factors involved in axonemal dynein preassembly (DNAAFs) are associated with human ciliopathies and are of clinical interest. Therefore, clear communication within this field is particularly important. Standardizing gene nomenclature, and basing it on orthology where possible, facilitates discussion and genetic comparison across species. Here, we discuss how the human gene nomenclature for dyneins, ODA docking complex subunits, and DNAAFs has been updated to be more functionally informative and consistent with that of the unicellular green alga Chlamydomonas reinhardtii, a key model organism for studying dyneins and ciliary function. We also detail additional nomenclature updates for vertebrate-specific genes that encode dynein chains and other proteins involved in dynein complex assembly.
Collapse
Affiliation(s)
- Bryony Braschi
- HUGO Gene Nomenclature Committee, European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, Cambridgeshire, UK
| | - Heymut Omran
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - George B. Witman
- Division of Cell Biology and Imaging, Department of Radiology, University of Massachusetts Medical School, Worcester, MA
| | - Gregory J. Pazour
- Program in Molecular Medicine, University of Massachusetts Medical School, Biotech II, Worcester, MA
| | - K. Kevin Pfister
- Cell Biology Department, School of Medicine University of Virginia, Charlottesville, VA
| | - Elspeth A. Bruford
- HUGO Gene Nomenclature Committee, European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, Cambridgeshire, UK
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge, Cambridgeshire, UK
| | - Stephen M. King
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT
| |
Collapse
|
20
|
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
|
21
|
Zhang T, Chen J, Zhang J, Guo YT, Zhou X, Li MW, Zheng ZZ, Zhang TZ, Murphy RW, Nevo E, Shi P. Phenotypic and genomic adaptations to the extremely high elevation in plateau zokor (Myospalax baileyi). Mol Ecol 2021; 30:5765-5779. [PMID: 34510615 DOI: 10.1111/mec.16174] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 08/07/2021] [Accepted: 08/31/2021] [Indexed: 12/31/2022]
Abstract
The evolutionary outcomes of high elevation adaptation have been extensively described. However, whether widely distributed high elevation endemic animals adopt uniform mechanisms during adaptation to different elevational environments remains unknown, especially with respect to extreme high elevation environments. To explore this, we analysed the phenotypic and genomic data of seven populations of plateau zokor (Myospalax baileyi) along elevations ranging from 2,700 to 4,300 m. Based on whole-genome sequencing data and demographic reconstruction of the evolutionary history, we show that two populations of plateau zokor living at elevations exceeding 3,700 m diverged from other populations nearly 10,000 years ago. Further, phenotypic comparisons reveal stress-dependent adaptation, as two populations living at elevations exceeding 3,700 m have elevated ratios of heart mass to body mass relative to other populations, and the highest population (4,300 m) displays alterations in erythrocytes. Correspondingly, genomic analysis of selective sweeps indicates that positive selection might contribute to the observed phenotypic alterations in these two extremely high elevation populations, with the adaptive cardiovascular phenotypes of both populations possibly evolving under the functional constrains of their common ancestral population. Taken together, phenotypic and genomic evidence demonstrates that heterogeneous stressors impact adaptations to extreme elevations and reveals stress-dependent and genetically constrained adaptation to hypoxia, collectively providing new insights into the high elevation adaptation.
Collapse
Affiliation(s)
- Tao Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Jie Chen
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Jia Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yuan-Ting Guo
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Xin Zhou
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Meng-Wen Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Zhi-Zhong Zheng
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Tong-Zuo Zhang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China
| | - Robert W Murphy
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Centre for Biodiversity and Conservation Biology, Royal Ontario Museum, Toronto, ON, Canada
| | - Eviatar Nevo
- Institute of Evolution, University of Haifa, Haifa, Israel
| | - Peng Shi
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,School of Future Technology, University of Chinese Academy of Sciences, Beijing, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
22
|
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: 39] [Impact Index Per Article: 9.8] [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
|
23
|
Wang L, Zhao X, Liang H, Zhang L, Li C, Li D, Meng X, Meng F, Gao M. Novel compound heterozygous mutations of DNAH5 identified in a pediatric patient with Kartagener syndrome: case report and literature review. BMC Pulm Med 2021; 21:263. [PMID: 34391405 PMCID: PMC8364053 DOI: 10.1186/s12890-021-01586-4] [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: 08/16/2020] [Accepted: 07/06/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Kartagener syndrome is a subtype of primary ciliary dyskinesia that may exhibit various symptoms including neonatal respiratory distress and frequent infections of the lung, sinus and middle ear because of the impaired function of motile cilia. In addition to typical symptoms of primary ciliary dyskinesia, patients with Kartagener syndrome also show situs inversus. It is an autosomal recessive disorder which is mostly caused by mutations in DNAH5. Kartagener syndrome is often underdiagnosed due to challenges in the diagnosis process. As next-generation sequencing becomes widely used in clinical laboratories, genetic testing provides an accurate approach to the diagnosis of Kartagener syndrome. CASE PRESENTATION A 7-year-old female patient presented with runny nose of 6 years duration and recurrent cough with phlegm of 2 years duration. Kartagener syndrome was diagnosed through diagnostic tests such as nasal nitric oxide (NO) concentration and transmission electron microscopy, and after performing other exams that corroborated the diagnosis, such as computed tomography, bronchoscopy and hearing test. Whole-exome sequencing was performed for the patient and both parents. The pediatric patient was diagnosed as Kartagener syndrome with the typical symptoms of ciliary dyskinesia including bronchiectasis, sinusitis, conductive hearing loss and situs inversus along with a reduced nasal NO concentration and ciliary abnormalities. The patient carried two novel compound heterozygous mutations in DNAH5, NM_001369:c.12813G > A (p. Trp4271Term) and NM_001369:c.9365delT (p. Leu3122Term). Both mutations lead to premature stop codons and thus are pathogenic. The p. Trp4271Term and p. Leu3122Term mutations were inherited from the father and the mother of the patient individually. A literature review was also conducted to summarize DNAH5 mutations in pediatric patients with Kartagener syndrome across different ethnic groups. CONCLUSIONS Our study provides a good example of the diagnosis of Kartagener syndrome in pediatric patients using a series of diagnostic tests combined with genetic testing. Two novel loss-of-function mutations in DNAH5 were identified and validated in a pediatric patient with Kartagener syndrome.
Collapse
Affiliation(s)
- Lina Wang
- Pediatric Department of Respiration II, The First Hospital of Jilin University, No.71 Xinmin Street, Changchun, 130000, China
| | - Xin Zhao
- Pediatric Department of Respiration II, The First Hospital of Jilin University, No.71 Xinmin Street, Changchun, 130000, China
| | - Hang Liang
- Pediatric Department of Respiration II, The First Hospital of Jilin University, No.71 Xinmin Street, Changchun, 130000, China
| | - Li Zhang
- Pediatric Department of Respiration II, The First Hospital of Jilin University, No.71 Xinmin Street, Changchun, 130000, China
| | - Chunyan Li
- Pediatric Department of Respiration II, The First Hospital of Jilin University, No.71 Xinmin Street, Changchun, 130000, China
| | - Deli Li
- Pediatric Department of Respiration II, The First Hospital of Jilin University, No.71 Xinmin Street, Changchun, 130000, China
| | | | - Fanzheng Meng
- Pediatric Department of Respiration II, The First Hospital of Jilin University, No.71 Xinmin Street, Changchun, 130000, China.
| | - Mao Gao
- Pediatric Department of Respiration II, The First Hospital of Jilin University, No.71 Xinmin Street, Changchun, 130000, China.
| |
Collapse
|
24
|
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
|
25
|
Aprea I, Nöthe-Menchen T, Dougherty GW, Raidt J, Loges NT, Kaiser T, Wallmeier J, Olbrich H, Strünker T, Kliesch S, Pennekamp P, Omran H. Motility of efferent duct cilia aids passage of sperm cells through the male reproductive system. Mol Hum Reprod 2021; 27:gaab009. [PMID: 33561200 PMCID: PMC7936721 DOI: 10.1093/molehr/gaab009] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 01/06/2021] [Indexed: 02/06/2023] Open
Abstract
Motile cilia line the efferent ducts of the mammalian male reproductive tract. Several recent mouse studies have demonstrated that a reduced generation of multiple motile cilia in efferent ducts is associated with obstructive oligozoospermia and fertility issues. However, the sole impact of efferent duct cilia dysmotility on male infertility has not been studied so far either in mice or human. Using video microscopy, histological- and ultrastructural analyses, we examined male reproductive tracts of mice deficient for the axonemal motor protein DNAH5: this defect exclusively disrupts the outer dynein arm (ODA) composition of motile cilia but not the ODA composition and motility of sperm flagella. These mice have immotile efferent duct cilia that lack ODAs, which are essential for ciliary beat generation. Furthermore, they show accumulation of sperm in the efferent duct. Notably, the ultrastructure and motility of sperm from these males are unaffected. Likewise, human individuals with loss-of-function DNAH5 mutations present with reduced sperm count in the ejaculate (oligozoospermia) and dilatations of the epididymal head but normal sperm motility, similar to DNAH5 deficient mice. The findings of this translational study demonstrate, in both mice and men, that efferent duct ciliary motility is important for male reproductive fitness and uncovers a novel pathomechanism distinct from primary defects of sperm motility (asthenozoospermia). If future work can identify environmental factors or defects in genes other than DNAH5 that cause efferent duct cilia dysmotility, this will help unravel other causes of oligozoospermia and may influence future practices in genetic and fertility counseling as well as ART.
Collapse
Affiliation(s)
- Isabella Aprea
- Department of General Pediatrics, University Hospital Muenster, Muenster 48149, Germany
| | - Tabea Nöthe-Menchen
- Department of General Pediatrics, University Hospital Muenster, Muenster 48149, Germany
| | - Gerard W Dougherty
- Department of General Pediatrics, University Hospital Muenster, Muenster 48149, Germany
| | - Johanna Raidt
- Department of General Pediatrics, University Hospital Muenster, Muenster 48149, Germany
| | - Niki T Loges
- Department of General Pediatrics, University Hospital Muenster, Muenster 48149, Germany
| | - Thomas Kaiser
- Department of General Pediatrics, University Hospital Muenster, Muenster 48149, Germany
| | - Julia Wallmeier
- Department of General Pediatrics, University Hospital Muenster, Muenster 48149, Germany
| | - Heike Olbrich
- Department of General Pediatrics, University Hospital Muenster, Muenster 48149, Germany
| | - Timo Strünker
- Department of Clinical Andrology, Centre of Reproductive Medicine and Andrology, University Hospital Muenster, Muenster 48149, Germany
| | - Sabine Kliesch
- Department of Clinical Andrology, Centre of Reproductive Medicine and Andrology, University Hospital Muenster, Muenster 48149, Germany
| | - Petra Pennekamp
- Department of General Pediatrics, University Hospital Muenster, Muenster 48149, Germany
| | - Heymut Omran
- Department of General Pediatrics, University Hospital Muenster, Muenster 48149, Germany
| |
Collapse
|
26
|
Devarbhavi P, Telang L, Vastrad B, Tengli A, Vastrad C, Kotturshetti I. Identification of key pathways and genes in polycystic ovary syndrome via integrated bioinformatics analysis and prediction of small therapeutic molecules. Reprod Biol Endocrinol 2021; 19:31. [PMID: 33622336 PMCID: PMC7901211 DOI: 10.1186/s12958-021-00706-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/28/2021] [Indexed: 02/06/2023] Open
Abstract
To enhance understanding of polycystic ovary syndrome (PCOS) at the molecular level; this investigation intends to examine the genes and pathways associated with PCOS by using an integrated bioinformatics analysis. Based on the expression profiling by high throughput sequencing data GSE84958 derived from the Gene Expression Omnibus (GEO) database, the differentially expressed genes (DEGs) between PCOS samples and normal controls were identified. We performed a functional enrichment analysis. A protein-protein interaction (PPI) network, miRNA- target genes and TF - target gene networks, were constructed and visualized, with which the hub gene nodes were identified. Validation of hub genes was performed by using receiver operating characteristic (ROC) and RT-PCR. Small drug molecules were predicted by using molecular docking. A total of 739 DEGs were identified, of which 360 genes were up regulated and 379 genes were down regulated. GO enrichment analysis revealed that up regulated genes were mainly involved in peptide metabolic process, organelle envelope and RNA binding and the down regulated genes were significantly enriched in plasma membrane bounded cell projection organization, neuron projection and DNA-binding transcription factor activity, RNA polymerase II-specific. REACTOME pathway enrichment analysis revealed that the up regulated genes were mainly enriched in translation and respiratory electron transport and the down regulated genes were mainly enriched in generic transcription pathway and transmembrane transport of small molecules. The top 10 hub genes (SAA1, ADCY6, POLR2K, RPS15, RPS15A, CTNND1, ESR1, NEDD4L, KNTC1 and NGFR) were identified from PPI network, miRNA - target gene network and TF - target gene network. The modules analysis showed that genes in modules were mainly associated with the transport of respiratory electrons and signaling NGF, respectively. We find a series of crucial genes along with the pathways that were most closely related with PCOS initiation and advancement. Our investigations provide a more detailed molecular mechanism for the progression of PCOS, detail information on the potential biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Praveenkumar Devarbhavi
- Department of Endocrinology and Metabolism, Subbaiah Institute of Medical Sciences and Research Centre, Shimoga, Karnataka, 577201, India
| | - Lata Telang
- Department of Gynaecology and Obstetrics, Subbaiah Institute of Medical Sciences and Research Centre, Shimoga, Karnataka, 577201, India
| | - Basavaraj Vastrad
- Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka, 582103, India
| | - Anandkumar Tengli
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Mysuru and JSS Academy of Higher Education & Research, Mysuru, Karnataka, 570015, India
| | - Chanabasayya Vastrad
- Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad, Karanataka, 580001, India.
| | - Iranna Kotturshetti
- Department of Ayurveda, Rajiv Gandhi Education Society's Ayurvedic Medical College, Ron, Karanataka, 562209, India
| |
Collapse
|
27
|
CFAP53 regulates mammalian cilia-type motility patterns through differential localization and recruitment of axonemal dynein components. PLoS Genet 2020; 16:e1009232. [PMID: 33347437 PMCID: PMC7817014 DOI: 10.1371/journal.pgen.1009232] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 01/20/2021] [Accepted: 10/29/2020] [Indexed: 12/12/2022] Open
Abstract
Motile cilia can beat with distinct patterns, but how motility variations are regulated remain obscure. Here, we have studied the role of the coiled-coil protein CFAP53 in the motility of different cilia-types in the mouse. While node (9+0) cilia of Cfap53 mutants were immotile, tracheal and ependymal (9+2) cilia retained motility, albeit with an altered beat pattern. In node cilia, CFAP53 mainly localized at the base (centriolar satellites), whereas it was also present along the entire axoneme in tracheal cilia. CFAP53 associated tightly with microtubules and interacted with axonemal dyneins and TTC25, a dynein docking complex component. TTC25 and outer dynein arms (ODAs) were lost from node cilia, but were largely maintained in tracheal cilia of Cfap53-/- mice. Thus, CFAP53 at the base of node cilia facilitates axonemal transport of TTC25 and dyneins, while axonemal CFAP53 in 9+2 cilia stabilizes dynein binding to microtubules. Our study establishes how differential localization and function of CFAP53 contributes to the unique motion patterns of two important mammalian cilia-types. Motile cilia in various kinds of tissues and cell-types drive fluid flow over epithelia or facilitate cellular locomotion. There are two types of motile cilia. Motile cilia with a 9+2 configuration of microtubules are found on tracheal epithelial cells and brain ependymal cells, and exhibit planar beating with effective and recovery strokes. On the other hand, 9+0 motile cilia are found in the embryonic node, show rotational movement and are involved in establishing left-right asymmetry of visceral organs. However, it is not well understood how these two types of motile cilia exhibit their characteristic motion patterns. We have uncovered distinct roles and subcellular localization of the CFAP53 protein in 9+0 versus the 9+2 motile cilia of the mouse. Our data provide novel insights into the molecular basis of motility differences that characterize these two types of mammalian motile cilia.
Collapse
|
28
|
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
|
29
|
Peng Y, Pang J, Hu J, Jia Z, Xi H, Ma N, Yang S, Liu J, Huang X, Tang C, Wang H. Clinical and molecular characterization of 12 prenatal cases of Cri-du-chat syndrome. Mol Genet Genomic Med 2020; 8:e1312. [PMID: 32500674 PMCID: PMC7434726 DOI: 10.1002/mgg3.1312] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/20/2020] [Accepted: 04/27/2020] [Indexed: 12/12/2022] Open
Abstract
Background This study aimed to define the molecular basis for 12 prenatal cases of Cri‐du‐chat syndrome (CdCS) and the potential genotyping‐phenotyping association. Methods Karyotyping and single nucleotide polymorphism array analyses for copy number variants were performed. Results Nine cases had 5p terminal deletions and three had 5p interstitial deletions, and these cases had variable deletion sizes with partial overlapping. Phenotypically, besides intrauterine growth restriction (IUGR) and brain as well as heart abnormalities, hypospadias, and lung dysplasia were observed. Potential genetic causes for specific phenotypes in these cases were identified. Conclusion This study defined the molecular bases for the patients of CdCS, which is important for genetic counseling for these families. The findings of present study expand the clinical features of CdCS in the fetal period, and provided important information for further refining the genotypic–phenotypic correlations for this syndrome.
Collapse
Affiliation(s)
- Ying Peng
- Department of Medical Genetics, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China.,National Health Commission Key Laboratory of Birth Defects Research, Prevention and Treatment, Changsha, Hunan, China
| | - Jialun Pang
- Department of Medical Genetics, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China.,National Health Commission Key Laboratory of Birth Defects Research, Prevention and Treatment, Changsha, Hunan, China
| | - Jiancheng Hu
- Department of Medical Genetics, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China.,National Health Commission Key Laboratory of Birth Defects Research, Prevention and Treatment, Changsha, Hunan, China
| | - Zhengjun Jia
- Department of Medical Genetics, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China.,National Health Commission Key Laboratory of Birth Defects Research, Prevention and Treatment, Changsha, Hunan, China
| | - Hui Xi
- Department of Medical Genetics, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China.,National Health Commission Key Laboratory of Birth Defects Research, Prevention and Treatment, Changsha, Hunan, China
| | - Na Ma
- Department of Medical Genetics, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China.,National Health Commission Key Laboratory of Birth Defects Research, Prevention and Treatment, Changsha, Hunan, China
| | - Shuting Yang
- Department of Medical Genetics, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China.,National Health Commission Key Laboratory of Birth Defects Research, Prevention and Treatment, Changsha, Hunan, China
| | - Jing Liu
- Department of Medical Genetics, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China.,National Health Commission Key Laboratory of Birth Defects Research, Prevention and Treatment, Changsha, Hunan, China
| | - Xiaoliang Huang
- Department of Medical Genetics, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China.,National Health Commission Key Laboratory of Birth Defects Research, Prevention and Treatment, Changsha, Hunan, China
| | - Chengyuan Tang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hua Wang
- Department of Medical Genetics, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China.,National Health Commission Key Laboratory of Birth Defects Research, Prevention and Treatment, Changsha, Hunan, China
| |
Collapse
|