1
|
Guan X, Fan Y, Six R, Van Soom A, Pavani KC, Peelman L. MicroRNAs bta-novel-miR-117, bta-novel-miR-234 and bta-novel-miR-417 have adverse effects on blastocyst formation. Theriogenology 2025; 233:88-99. [PMID: 39613498 DOI: 10.1016/j.theriogenology.2024.11.018] [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: 08/09/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 12/01/2024]
Abstract
In a previous study we found that the levels of the novel microRNAs (miRNAs) bta-novel-miR-117 bta-novel-miR-234 and bta-novel-miR-417 (P < 0.001) are significantly up-regulated in extracellular vesicles (EVs) in the culture medium of degenerating embryos compared to blastocysts. Because the functions of these novel miRNAs are still unknown, we investigated their regulatory roles during bovine blastocyst development by adding their mimics and inhibitors to the culture medium. The addition of mimics for bta-novel-miR-117, bta-novel-miR-234 and bta-novel-miR-417 resulted in a decreased blastocyst rate, and supplementation of bta-novel-miR-234 inhibitors increased the cleavage rate significantly (P < 0.001). Low-input transcriptome analysis and RT-qPCR results revealed that bta-novel-miR-117, bta-novel-miR-234 and bta-novel-miR-417 co-target genes such as ANKEF1, HAND2 and SLC2A2, downregulated their expression significantly (P < 0.001). These genes associated with glucose transmembrane transport and plasma membrane raft metabolism play crucial roles in embryonic development. The results suggest that overexpressing of these three novel miRNAs impairs embryonic development, and they might serve as biomarkers to detect failing bovine embryos.
Collapse
Affiliation(s)
- Xuefeng Guan
- Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820, Merelbeke, Belgium
| | - Yuan Fan
- Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820, Merelbeke, Belgium
| | - Rani Six
- Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820, Merelbeke, Belgium
| | - Ann Van Soom
- Department of Reproduction, Obstetrics and Herd Health, Faculty of Veterinary Medicine, University of Ghent, Salisburylaan 133, B-9820, Merelbeke, Belgium
| | - Krishna Chaitanya Pavani
- Department of Reproduction, Obstetrics and Herd Health, Faculty of Veterinary Medicine, University of Ghent, Salisburylaan 133, B-9820, Merelbeke, Belgium; Department for Reproductive Medicine, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Gent, Belgium.
| | - Luc Peelman
- Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820, Merelbeke, Belgium.
| |
Collapse
|
2
|
Velde HM, Vaseghi-Shanjani M, Smits JJ, Ramakrishnan G, Oostrik J, Wesdorp M, Astuti G, Yntema HG, Hoefsloot L, Lanting CP, Huynen MA, Lehman A, Turvey SE, Pennings RJE, Kremer H. Exome variant prioritization in a large cohort of hearing-impaired individuals indicates IKZF2 to be associated with non-syndromic hearing loss and guides future research of unsolved cases. Hum Genet 2024; 143:1379-1399. [PMID: 39406892 PMCID: PMC11522133 DOI: 10.1007/s00439-024-02706-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/27/2024] [Indexed: 10/30/2024]
Abstract
Although more than 140 genes have been associated with non-syndromic hereditary hearing loss (HL), at least half of the cases remain unexplained in medical genetic testing. One reason is that pathogenic variants are located in 'novel' deafness genes. A variant prioritization approach was used to identify novel (candidate) genes for HL. Exome-wide sequencing data were assessed for subjects with presumed hereditary HL that remained unexplained in medical genetic testing by gene-panel analysis. Cases in group AD had presumed autosomal dominantly inherited HL (n = 124), and in group AR, presumed autosomal recessive HL (n = 337). Variants in known and candidate deafness genes were prioritized based on allele frequencies and predicted effects. Selected variants were tested for their co-segregation with HL. Two cases were solved by variants in recently identified deafness genes (ABHD12, TRRAP). Variant prioritization also revealed potentially causative variants in candidate genes associated with recessive and X-linked HL. Importantly, missense variants in IKZF2 were found to co-segregate with dominantly inherited non-syndromic HL in three families. These variants specifically affected Zn2+-coordinating cysteine or histidine residues of the zinc finger motifs 2 and 3 of the encoded protein Helios. This finding indicates a complex genotype-phenotype correlation for IKZF2 defects, as this gene was previously associated with non-syndromic dysfunction of the immune system and ICHAD syndrome, including HL. The designed strategy for variant prioritization revealed that IKZF2 variants can underlie non-syndromic HL. The large number of candidate genes for HL and variants therein stress the importance of inclusion of family members for variant prioritization.
Collapse
Affiliation(s)
- Hedwig M Velde
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboudumc, Nijmegen, The Netherlands
| | - Maryam Vaseghi-Shanjani
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital, Vancouver, BC, Canada
| | - Jeroen J Smits
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands
- Department of Clinical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Jaap Oostrik
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands
| | - Mieke Wesdorp
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands
| | - Galuh Astuti
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
| | - Helger G Yntema
- Donders Institute for Brain, Cognition and Behaviour, Radboudumc, Nijmegen, The Netherlands
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
| | - Lies Hoefsloot
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Cris P Lanting
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboudumc, Nijmegen, The Netherlands
| | - Martijn A Huynen
- Center for Molecular and Biomolecular Informatics, Radboudumc, Nijmegen, The Netherlands
| | - Anna Lehman
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital, Vancouver, BC, Canada
| | - Stuart E Turvey
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital, Vancouver, BC, Canada
| | - Ronald J E Pennings
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboudumc, Nijmegen, The Netherlands
| | - Hannie Kremer
- Department of Otorhinolaryngology, Radboudumc, Nijmegen, The Netherlands.
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands.
| |
Collapse
|
3
|
Jukic A, Lei Z, Cebul ER, Pinter K, Tadesse Y, Jarysta A, David S, Mosqueda N, Tarchini B, Kindt K. Presynaptic Nrxn3 is essential for ribbon-synapse maturation in hair cells. Development 2024; 151:dev202723. [PMID: 39254120 PMCID: PMC11488651 DOI: 10.1242/dev.202723] [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: 01/25/2024] [Accepted: 08/28/2024] [Indexed: 09/11/2024]
Abstract
Hair cells of the inner ear and lateral-line system rely on specialized ribbon synapses to transmit sensory information to the central nervous system. The molecules required to assemble these synapses are not fully understood. We show that Nrxn3, a presynaptic adhesion molecule, is crucial for ribbon-synapse maturation in hair cells. In both mouse and zebrafish models, the loss of Nrxn3 results in significantly fewer intact ribbon synapses. We show in zebrafish that, initially, Nrxn3 loss does not alter pre- and postsynapse numbers but, later, synapses fail to pair, leading to postsynapse loss. We also demonstrate that Nrxn3 subtly influences synapse selectivity in zebrafish lateral-line hair cells that detect anterior flow. Loss of Nrxn3 leads to a 60% loss of synapses in zebrafish, which dramatically reduces pre- and postsynaptic responses. Despite fewer synapses, auditory responses in zebrafish and mice are unaffected. This work demonstrates that Nrxn3 is a crucial and conserved molecule required for the maturation of ribbon synapses. Understanding how ribbon synapses mature is essential to generating new therapies to treat synaptopathies linked to auditory or vestibular dysfunction.
Collapse
Affiliation(s)
- Alma Jukic
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | - Zhengchang Lei
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | - Elizabeth R. Cebul
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | - Katherine Pinter
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | - Yommi Tadesse
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | | | - Sandeep David
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | - Natalie Mosqueda
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | - Basile Tarchini
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Katie Kindt
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| |
Collapse
|
4
|
Wang S, Chakraborty S, Fu Y, Lee MP, Liu J, Waldhaus J. 3D reconstruction of the mouse cochlea from scRNA-seq data suggests morphogen-based principles in apex-to-base specification. Dev Cell 2024; 59:1538-1552.e6. [PMID: 38593801 PMCID: PMC11187690 DOI: 10.1016/j.devcel.2024.03.028] [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: 09/20/2022] [Revised: 04/03/2023] [Accepted: 03/13/2024] [Indexed: 04/11/2024]
Abstract
In the mammalian auditory system, frequency discrimination depends on numerous morphological and physiological properties of the organ of Corti, which gradually change along the apex-to-base (tonotopic) axis of the organ. For example, the basilar membrane stiffness changes tonotopically, thus affecting the tuning properties of individual hair cells. At the molecular level, those frequency-specific characteristics are mirrored by gene expression gradients; however, the molecular mechanisms controlling tonotopic gene expression in the mouse cochlea remain elusive. Through analyzing single-cell RNA sequencing (scRNA-seq) data from E12.5 and E14.5 time points, we predicted that morphogens, rather than a cell division-associated mechanism, confer spatial identity in the extending cochlea. Subsequently, we reconstructed the developing cochlea in 3D space from scRNA-seq data to investigate the molecular pathways mediating positional information. The retinoic acid (RA) and hedgehog pathways were found to form opposing apex-to-base gradients, and functional interrogation using mouse cochlear explants suggested that both pathways jointly specify the longitudinal axis.
Collapse
Affiliation(s)
- Shuze Wang
- Department of Otolaryngology-Head and Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Saikat Chakraborty
- Department of Otolaryngology-Head and Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yujuan Fu
- Biomedical Informatics and Medical Education, University of Washington, Seattle, WA 98195, USA
| | - Mary P Lee
- Department of Otolaryngology-Head and Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jie Liu
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Joerg Waldhaus
- Department of Otolaryngology-Head and Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
5
|
Polesskaya O, Boussaty E, Cheng R, Lamonte O, Zhou T, Du E, Sanches TM, Nguyen KM, Okamoto M, Palmer AA, Friedman R. Genome-wide association study for age-related hearing loss in CFW mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.10.598304. [PMID: 38915500 PMCID: PMC11195089 DOI: 10.1101/2024.06.10.598304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Age-related hearing impairment is the most common cause of hearing loss and is one of the most prevalent conditions affecting the elderly globally. It is influenced by a combination of environmental and genetic factors. The mouse and human inner ears are functionally and genetically homologous. Investigating the genetic basis of age-related hearing loss (ARHL) in an outbred mouse model may lead to a better understanding of the molecular mechanisms of this condition. We used Carworth Farms White (CFW) outbred mice, because they are genetically diverse and exhibit variation in the onset and severity of ARHL. The goal of this study was to identify genetic loci involved in regulating ARHL. Hearing at a range of frequencies was measured using Auditory Brainstem Response (ABR) thresholds in 946 male and female CFW mice at the age of 1, 6, and 10 months. We obtained genotypes at 4.18 million single nucleotide polymorphisms (SNP) using low-coverage (mean coverage 0.27x) whole-genome sequencing followed by imputation using STITCH. To determine the accuracy of the genotypes we sequenced 8 samples at >30x coverage and used calls from those samples to estimate the discordance rate, which was 0.45%. We performed genetic analysis for the ABR thresholds for each frequency at each age, and for the time of onset of deafness for each frequency. The SNP heritability ranged from 0 to 42% for different traits. Genome-wide association analysis identified several regions associated with ARHL that contained potential candidate genes, including Dnah11, Rapgef5, Cpne4, Prkag2, and Nek11. We confirmed, using functional study, that Prkag2 deficiency causes age-related hearing loss at high frequency in mice; this makes Prkag2 a candidate gene for further studies. This work helps to identify genetic risk factors for ARHL and to define novel therapeutic targets for the treatment and prevention of ARHL.
Collapse
Affiliation(s)
- Oksana Polesskaya
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ely Boussaty
- Department of Otolaryngology - Head and Neck Surgery, University of California San Diego, La Jolla, CA, 92093, USA
| | - Riyan Cheng
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Olivia Lamonte
- Department of Otolaryngology - Head and Neck Surgery, University of California San Diego, La Jolla, CA, 92093, USA
| | - Thomas Zhou
- Department of Otolaryngology - Head and Neck Surgery, University of California San Diego, La Jolla, CA, 92093, USA
| | - Eric Du
- Department of Otolaryngology - Head and Neck Surgery, University of California San Diego, La Jolla, CA, 92093, USA
| | | | - Khai-Minh Nguyen
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Mika Okamoto
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Abraham A Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Rick Friedman
- Department of Otolaryngology - Head and Neck Surgery, University of California San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
6
|
Holt JR, Fettiplace R, Müller U. Sensory transduction in auditory hair cells-PIEZOs can't touch this. J Gen Physiol 2024; 156:e202413585. [PMID: 38727631 PMCID: PMC11090049 DOI: 10.1085/jgp.202413585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2024] Open
Abstract
In this Viewpoint, Holt, Fettiplace, and Müller weigh the evidence supporting a role for PIEZO and TMC channels in mechanosensory transduction in inner ear hair cells.
Collapse
Affiliation(s)
- Jeffrey R. Holt
- Departments of Otolaryngology and Neurology, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Robert Fettiplace
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Ulrich Müller
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
7
|
Gwilliam K, Sperber M, Perry K, Rose KP, Ginsberg L, Paladugu N, Song Y, Milon B, Elkon R, Hertzano R. A cell type-specific approach to elucidate the role of miR-96 in inner ear hair cells. FRONTIERS IN AUDIOLOGY AND OTOLOGY 2024; 2:1400576. [PMID: 38826689 PMCID: PMC11141775 DOI: 10.3389/fauot.2024.1400576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Introduction Mutations in microRNA-96 (miR-96), a microRNA expressed within the hair cells (HCs) of the inner ear, result in progressive hearing loss in both mouse models and humans. In this study, we present the first HC-specific RNA-sequencing (RNA-seq) dataset from newborn Mir96Dmdo heterozygous, homozygous mutant, and wildtype mice. Methods Bulk RNA-seq was performed on HCs of newborn Mir96Dmdo heterozygous, homozygous mutant, and wildtype mice. Differentially expressed gene analysis was conducted on Mir96Dmdo homozygous mutant HCs compared to wildtype littermate controls, followed by GO term and protein-protein interaction analysis on these differentially expressed genes. Results We identify 215 upregulated and 428 downregulated genes in the HCs of the Mir96Dmdo homozygous mutant mice compared to their wildtype littermate controls. Many of the significantly downregulated genes in Mir96Dmdo homozygous mutant HCs have established roles in HC development and/or known roles in deafness including Myo15a, Myo7a, Ush1c, Gfi1, and Ptprq and have enrichment in gene ontology (GO) terms with biological functions such as sensory perception of sound. Interestingly, upregulated genes in Mir96Dmdo homozygous mutants, including possible miR-96 direct targets, show higher wildtype expression in supporting cells compared to HCs. Conclusion Our data further support a role for miR-96 in HC development, possibly as a repressor of supporting cell transcriptional programs in HCs. The HC-specific Mir96Dmdo RNA-seq data set generated from this manuscript are now publicly available in a dedicated profile in the gene expression analysis resource (gEAR-https://umgear.org/p?l=miR96).
Collapse
Affiliation(s)
- Kathleen Gwilliam
- Section on Omics and Translational Science of Hearing, Neurotology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| | - Michal Sperber
- Department of Human Molecular Genetics and Biochemistry, Tel Aviv University School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Katherine Perry
- Department of Otorhinolaryngology Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Kevin P. Rose
- Section on Omics and Translational Science of Hearing, Neurotology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| | - Laura Ginsberg
- Department of Otorhinolaryngology Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Nikhil Paladugu
- Department of Otorhinolaryngology Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Yang Song
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Beatrice Milon
- Section on Omics and Translational Science of Hearing, Neurotology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| | - Ran Elkon
- Department of Human Molecular Genetics and Biochemistry, Tel Aviv University School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ronna Hertzano
- Section on Omics and Translational Science of Hearing, Neurotology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
- Department of Otorhinolaryngology Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
8
|
Zhang X, Wang S, Liu S, Du Z, Wu G, Liang Y, Huang Y, Shang X, Hu Y, Zhu Z, Sun W, Zhang X, Yu H. Epidemiologic association and shared genetic architecture between cataract and hearing difficulties among middle-aged and older adults. Hum Genomics 2024; 18:39. [PMID: 38632618 PMCID: PMC11022469 DOI: 10.1186/s40246-024-00601-z] [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: 08/06/2023] [Accepted: 03/20/2024] [Indexed: 04/19/2024] Open
Abstract
Age-related cataract and hearing difficulties are major sensory disorders that often co-exist in the global-wide elderly and have a tangible influence on the quality of life. However, the epidemiologic association between cataract and hearing difficulties remains unexplored, while little is known about whether the two share their genetic etiology. We first investigated the clinical association between cataract and hearing difficulties using the UK Biobank covering 502,543 individuals. Both unmatched analysis (adjusted for confounders) and a matched analysis (one control matched for each patient with cataract according to confounding factors) were undertaken and confirmed that cataract was associated with hearing difficulties (OR, 2.12; 95% CI, 1.98-2.27; OR, 2.03; 95% CI, 1.86-2.23, respectively). Furthermore, we explored and quantified the shared genetic architecture of these two complex sensory disorders at the common variant level using the bivariate causal mixture model (MiXeR) and conditional/conjunctional false discovery rate method based on the largest available genome-wide association studies of cataract (N = 585,243) and hearing difficulties (N = 323,978). Despite detecting only a negligible genetic correlation, we observe polygenic overlap between cataract and hearing difficulties and identify 6 shared loci with mixed directions of effects. Follow-up analysis of the shared loci implicates candidate genes QKI, STK17A, TYR, NSF, and TCF4 likely contribute to the pathophysiology of cataracts and hearing difficulties. In conclusion, this study demonstrates the presence of epidemiologic association between cataract and hearing difficulties and provides new insights into the shared genetic architecture of these two disorders at the common variant level.
Collapse
Affiliation(s)
- Xiayin Zhang
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Shan Wang
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shunming Liu
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zijing Du
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Guanrong Wu
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yingying Liang
- Department of Ophthalmology, Guangzhou First people's Hospital, Guangzhou, China
| | - Yu Huang
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xianwen Shang
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yijun Hu
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhuoting Zhu
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, VIC, East Melbourne, Australia
| | - Wei Sun
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Xueli Zhang
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Honghua Yu
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China.
| |
Collapse
|
9
|
Yang S, Min X, Hu L, Zheng M, Lu S, Zhao M, Jia S. RFX1 regulates foam cell formation and atherosclerosis by mediating CD36 expression. Int Immunopharmacol 2024; 130:111751. [PMID: 38402833 DOI: 10.1016/j.intimp.2024.111751] [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: 09/07/2023] [Revised: 02/06/2024] [Accepted: 02/21/2024] [Indexed: 02/27/2024]
Abstract
BACKGROUND AND AIMS Atherosclerosis (AS) is a continuously low-grade inflammatory disease, and monocyte-derived macrophages play a vital role in AS pathogenesis. Regulatory factor X1 (RFX1) has been reported to participate in differentiation of various cells. Our previous report showed that RFX1 expression in CD14+ monocytes from AS patients was decreased and closely related to AS development. Macrophages mostly derive from monocytes and play an important role in AS plaque formation and stability. However, the functions of RFX1 in the formation of macrophage-derived foam cells and consequent AS development are unclear. METHODS We explored the effects of RFX1 on oxidation low lipoprotein (ox-LDL)-stimulated foam cell formation and CD36 expression by increasing or silencing Rfx1 expression in mouse peritoneal macrophages (PMAs). The ApoE-/-Rfx1f/f or ApoE-/-Rfx1f/f Lyz2-Cre mice fed a high-fat diet for 24 weeks were used to further examine the effect of RFX1 on AS pathogenesis. We then performed dual luciferase reporter assays to study the regulation of RFX1 for CD36 transcription. RESULTS Our results demonstrate that RFX1 expression was significantly reduced in ox-LDL induced foam cells and negatively correlated with lipid uptake in macrophages. Besides, Rfx1 deficiency in myeloid cells aggravated atherosclerotic lesions in ApoE-/- mice. Mechanistically, RFX1 inhibited CD36 expression by directly regulating CD36 transcription in macrophages. CONCLUSIONS The reduction of RFX1 expression in macrophages is a vital determinant for foam cell formation and the initiation of AS, proving a potential novel approach for the treatment of AS disease.
Collapse
Affiliation(s)
- Shuang Yang
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha 410011, China
| | - Xiaoli Min
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
| | - Longyuan Hu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha 410011, China
| | - Meiling Zheng
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha 410011, China
| | - Shuang Lu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha 410011, China
| | - Ming Zhao
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha 410011, China; Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
| | - Sujie Jia
- Department of Pharmacy, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China.
| |
Collapse
|
10
|
Waldhaus J, Jiang L, Liu L, Liu J, Duncan RK. Mapping the developmental potential of mouse inner ear organoids at single-cell resolution. iScience 2024; 27:109069. [PMID: 38375227 PMCID: PMC10875570 DOI: 10.1016/j.isci.2024.109069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/20/2023] [Accepted: 01/25/2024] [Indexed: 02/21/2024] Open
Abstract
Inner ear organoids recapitulate development and are intended to generate cell types of the otic lineage for applications such as basic science research and cell replacement strategies. Here, we use single-cell sequencing to study the cellular heterogeneity of late-stage mouse inner ear organoid sensory epithelia, which we validated by comparison with datasets of the mouse cochlea and vestibular epithelia. We resolved supporting cell sub-types, cochlear-like hair cells, and vestibular type I and type II-like hair cells. While cochlear-like hair cells aligned best with an outer hair cell trajectory, vestibular-like hair cells followed developmental trajectories similar to in vivo programs branching into type II and then type I extrastriolar hair cells. These results highlight the transcriptional accuracy of the organoid developmental program but will also inform future strategies to improve synaptic connectivity and regional specification.
Collapse
Affiliation(s)
- Joerg Waldhaus
- Department of Otolaryngology–Head and Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI, USA
| | - Linghua Jiang
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Liqian Liu
- Department of Otolaryngology–Head and Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI, USA
| | - Jie Liu
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Robert Keith Duncan
- Department of Otolaryngology–Head and Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI, USA
- Ann Arbor Department of Veterans Affairs Medical Center, Ann Arbor, MI, USA
| |
Collapse
|
11
|
Jukic A, Lei Z, Cebul ER, Pinter K, Mosqueda N, David S, Tarchini B, Kindt K. Presynaptic Nrxn3 is essential for ribbon-synapse assembly in hair cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.14.580267. [PMID: 38410471 PMCID: PMC10896334 DOI: 10.1101/2024.02.14.580267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Hair cells of the inner ear rely on specialized ribbon synapses to transmit sensory information to the central nervous system. The molecules required to assemble these synapses are not fully understood. We show that Nrxn3, a presynaptic adhesion molecule, is critical for ribbon-synapse assembly in hair cells. In both mouse and zebrafish models, loss of Nrxn3 results in significantly fewer intact ribbon synapses. In zebrafish we demonstrate that a 60% loss of synapses in nrxn3 mutants dramatically reduces both presynaptic responses in hair cells and postsynaptic responses in afferent neurons. Despite a reduction in synapse function in this model, we find no deficits in the acoustic startle response, a behavior reliant on these synapses. Overall, this work demonstrates that Nrxn3 is a critical and conserved molecule required to assemble ribbon synapses. Understanding how ribbon synapses assemble is a key step towards generating novel therapies to treat forms of age-related and noise-induced hearing loss that occur due to loss of ribbon synapses.
Collapse
Affiliation(s)
- Alma Jukic
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD, 20892, USA
| | - Zhengchang Lei
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD, 20892, USA
| | - Elizabeth R Cebul
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD, 20892, USA
| | - Katherine Pinter
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD, 20892, USA
| | - Natalie Mosqueda
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD, 20892, USA
| | - Sandeep David
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD, 20892, USA
| | | | - Katie Kindt
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD, 20892, USA
| |
Collapse
|
12
|
Li J, Choi BY, Eltawil Y, Ismail Mohamad N, Park Y, Matthews IR, Han JH, Kim BJ, Sherr EH, Chan DK. TMTC4 is a hair cell-specific human deafness gene. JCI Insight 2023; 8:e172665. [PMID: 37943620 PMCID: PMC10807715 DOI: 10.1172/jci.insight.172665] [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: 05/30/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023] Open
Abstract
Transmembrane and tetratricopeptide repeat 4 (Tmtc4) is a deafness gene in mice. Tmtc4-KO mice have rapidly progressive postnatal hearing loss due to overactivation of the unfolded protein response (UPR); however, the cellular basis and human relevance of Tmtc4-associated hearing loss in the cochlea was not heretofore appreciated. We created a hair cell-specific conditional KO mouse that phenocopies the constitutive KO with postnatal onset deafness, demonstrating that Tmtc4 is a hair cell-specific deafness gene. Furthermore, we identified a human family in which Tmtc4 variants segregate with adult-onset progressive hearing loss. Lymphoblastoid cells derived from multiple affected and unaffected family members, as well as human embryonic kidney cells engineered to harbor each of the variants, demonstrated that the human Tmtc4 variants confer hypersensitivity of the UPR toward apoptosis. These findings provide evidence that TMTC4 is a deafness gene in humans and further implicate the UPR in progressive hearing loss.
Collapse
Affiliation(s)
- Jiang Li
- Department of Neurology and
- Department of Pediatrics, Institute of Human Genetics, Weill Institute for Neurosciences, UCSF, San Francisco, California, USA
| | - Byung Yoon Choi
- Department of Otorhinolaryngology, Seoul National University Bundang Hospital, Seoul, South Korea
| | - Yasmin Eltawil
- Department of Otolaryngology-Head and Neck Surgery, San Francisco, California, USA
| | - Noura Ismail Mohamad
- Department of Otolaryngology-Head and Neck Surgery, San Francisco, California, USA
| | - Yesai Park
- Department of Otolaryngology-Head and Neck Surgery, San Francisco, California, USA
| | - Ian R. Matthews
- Department of Otolaryngology-Head and Neck Surgery, San Francisco, California, USA
| | - Jin Hee Han
- Department of Otorhinolaryngology, Seoul National University Bundang Hospital, Seoul, South Korea
- Department of Otorhinolaryngology, Chungnam National University College of Medicine, Chungnam National University Sejong Hospital, Sejong City, South Korea
| | - Bong Jik Kim
- Department of Otorhinolaryngology, Chungnam National University College of Medicine, Chungnam National University Sejong Hospital, Sejong City, South Korea
| | - Elliott H. Sherr
- Department of Neurology and
- Department of Pediatrics, Institute of Human Genetics, Weill Institute for Neurosciences, UCSF, San Francisco, California, USA
| | - Dylan K. Chan
- Department of Otolaryngology-Head and Neck Surgery, San Francisco, California, USA
| |
Collapse
|
13
|
Lue PY, Oliver MH, Neeff M, Thorne PR, Suzuki-Kerr H. Sheep as a large animal model for hearing research: comparison to common laboratory animals and humans. Lab Anim Res 2023; 39:31. [PMID: 38012676 PMCID: PMC10680324 DOI: 10.1186/s42826-023-00182-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 11/10/2023] [Accepted: 11/15/2023] [Indexed: 11/29/2023] Open
Abstract
Sensorineural hearing loss (SNHL), caused by pathology in the cochlea, is the most common type of hearing loss in humans. It is generally irreversible with very few effective pharmacological treatments available to prevent the degenerative changes or minimise the impact. Part of this has been attributed to difficulty of translating "proof-of-concept" for novel treatments established in small animal models to human therapies. There is an increasing interest in the use of sheep as a large animal model. In this article, we review the small and large animal models used in pre-clinical hearing research such as mice, rats, chinchilla, guinea pig, rabbit, cat, monkey, dog, pig, and sheep to humans, and compare the physiology, inner ear anatomy, and some of their use as model systems for SNHL, including cochlear implantation surgeries. Sheep have similar cochlear anatomy, auditory threshold, neonatal auditory system development, adult and infant body size, and number of birth as humans. Based on these comparisons, we suggest that sheep are well-suited as a potential translational animal model that bridges the gap between rodent model research to the clinical use in humans. This is especially in areas looking at changes across the life-course or in specific areas of experimental investigation such as cochlear implantation and other surgical procedures, biomedical device development and age-related sensorineural hearing loss research. Combined use of small animals for research that require higher throughput and genetic modification and large animals for medical translation could greatly accelerate the overall translation of basic research in the field of auditory neuroscience from bench to clinic.
Collapse
Affiliation(s)
- Po-Yi Lue
- Department of Physiology, The University of Auckland, Auckland, New Zealand
- Eisdell Moore Centre, The University of Auckland, Auckland, New Zealand
| | - Mark H Oliver
- Liggins Institute, The University of Auckland, Auckland, New Zealand
- Ngapouri Research Farm Laboratory, University of Auckland, Waiotapu, New Zealand
| | - Michel Neeff
- Department of Physiology, The University of Auckland, Auckland, New Zealand
- Department of Surgery, Auckland District Health Board, Auckland, New Zealand
| | - Peter R Thorne
- Department of Physiology, The University of Auckland, Auckland, New Zealand
- Eisdell Moore Centre, The University of Auckland, Auckland, New Zealand
- Section of Audiology, The University of Auckland, Auckland, New Zealand
| | - Haruna Suzuki-Kerr
- Department of Physiology, The University of Auckland, Auckland, New Zealand.
- Eisdell Moore Centre, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
14
|
Zhou LY, Jin CX, Wang WX, Song L, Shin JB, Du TT, Wu H. Differential regulation of hair cell actin cytoskeleton mediated by SRF and MRTFB. eLife 2023; 12:e90155. [PMID: 37982489 PMCID: PMC10703445 DOI: 10.7554/elife.90155] [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: 06/13/2023] [Accepted: 11/17/2023] [Indexed: 11/21/2023] Open
Abstract
The MRTF-SRF pathway has been extensively studied for its crucial role in driving the expression of a large number of genes involved in actin cytoskeleton of various cell types. However, the specific contribution of MRTF-SRF in hair cells remains unknown. In this study, we showed that hair cell-specific deletion of Srf or Mrtfb, but not Mrtfa, leads to similar defects in the development of stereocilia dimensions and the maintenance of cuticular plate integrity. We used fluorescence-activated cell sorting-based hair cell RNA-Seq analysis to investigate the mechanistic underpinnings of the changes observed in Srf and Mrtfb mutants, respectively. Interestingly, the transcriptome analysis revealed distinct profiles of genes regulated by Srf and Mrtfb, suggesting different transcriptional regulation mechanisms of actin cytoskeleton activities mediated by Srf and Mrtfb. Exogenous delivery of calponin 2 using Adeno-associated virus transduction in Srf mutants partially rescued the impairments of stereocilia dimensions and the F-actin intensity of cuticular plate, suggesting the involvement of Cnn2, as an Srf downstream target, in regulating the hair bundle morphology and cuticular plate actin cytoskeleton organization. Our study uncovers, for the first time, the unexpected differential transcriptional regulation of actin cytoskeleton mediated by Srf and Mrtfb in hair cells, and also demonstrates the critical role of SRF-CNN2 in modulating actin dynamics of the stereocilia and cuticular plate, providing new insights into the molecular mechanism underlying hair cell development and maintenance.
Collapse
Affiliation(s)
- Ling-Yun Zhou
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Ear Institute, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose DiseasesShanghaiChina
| | - Chen-Xi Jin
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Ear Institute, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose DiseasesShanghaiChina
| | - Wen-Xiao Wang
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Ear Institute, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose DiseasesShanghaiChina
| | - Lei Song
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Ear Institute, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose DiseasesShanghaiChina
| | - Jung-Bum Shin
- Department of Neuroscience, University of VirginiaCharlottesvilleUnited States
| | - Ting-Ting Du
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Ear Institute, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose DiseasesShanghaiChina
| | - Hao Wu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Ear Institute, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose DiseasesShanghaiChina
| |
Collapse
|
15
|
Doda D, Alonso Jimenez S, Rehrauer H, Carreño JF, Valsamides V, Di Santo S, Widmer HR, Edge A, Locher H, van der Valk WH, Zhang J, Koehler KR, Roccio M. Human pluripotent stem cell-derived inner ear organoids recapitulate otic development in vitro. Development 2023; 150:dev201865. [PMID: 37791525 PMCID: PMC10565253 DOI: 10.1242/dev.201865] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 08/01/2023] [Indexed: 10/05/2023]
Abstract
Our molecular understanding of the early stages of human inner ear development has been limited by the difficulty in accessing fetal samples at early gestational stages. As an alternative, previous studies have shown that inner ear morphogenesis can be partially recapitulated using induced pluripotent stem cells directed to differentiate into inner ear organoids (IEOs). Once validated and benchmarked, these systems could represent unique tools to complement and refine our understanding of human otic differentiation and model developmental defects. Here, we provide the first direct comparisons of the early human embryonic otocyst and fetal sensory organs with human IEOs. We use multiplexed immunostaining and single-cell RNA-sequencing to characterize IEOs at three key developmental steps, providing a new and unique signature of in vitro-derived otic placode, epithelium, neuroblasts and sensory epithelia. In parallel, we evaluate the expression and localization of crucial markers at these equivalent stages in human embryos. Together, our data indicate that the current state-of-the-art protocol enables the specification of bona fide otic tissue, supporting the further application of IEOs to inform inner ear biology and disease.
Collapse
Affiliation(s)
- Daniela Doda
- Inner Ear Stem Cell Laboratory, Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Zurich (USZ), 8091 Zurich,Switzerland
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Zurich (UZH), 8006 Zurich, Switzerland
| | - Sara Alonso Jimenez
- Inner Ear Stem Cell Laboratory, Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Zurich (USZ), 8091 Zurich,Switzerland
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Zurich (UZH), 8006 Zurich, Switzerland
| | - Hubert Rehrauer
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Zurich (UZH), 8006 Zurich, Switzerland
- Functional Genomics Center Zurich (ETH Zurich and University of Zurich), 8092 Zurich, Switzerland
| | - Jose F. Carreño
- Inner Ear Stem Cell Laboratory, Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Zurich (USZ), 8091 Zurich,Switzerland
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Zurich (UZH), 8006 Zurich, Switzerland
- Functional Genomics Center Zurich (ETH Zurich and University of Zurich), 8092 Zurich, Switzerland
| | - Victoria Valsamides
- Inner Ear Stem Cell Laboratory, Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Zurich (USZ), 8091 Zurich,Switzerland
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Zurich (UZH), 8006 Zurich, Switzerland
| | - Stefano Di Santo
- Program for Regenerative Neuroscience, Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland
| | - Hans R. Widmer
- Program for Regenerative Neuroscience, Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland
| | - Albert Edge
- Eaton Peabody Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA
- Department of Otorhinolaryngology - Head and Neck Surgery, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Heiko Locher
- OtoBiology Leiden, Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Wouter H. van der Valk
- OtoBiology Leiden, Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Jingyuan Zhang
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA 02115, USA
- F.M. Kirby Neurobiology Center, Boston Children's Hospital,Boston, MA 02115, USA
| | - Karl R. Koehler
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA 02115, USA
- F.M. Kirby Neurobiology Center, Boston Children's Hospital,Boston, MA 02115, USA
- Department of Plastic and Oral Surgery, Boston Children's Hospital, Boston, MA 02115, USA
| | - Marta Roccio
- Inner Ear Stem Cell Laboratory, Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Zurich (USZ), 8091 Zurich,Switzerland
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Zurich (UZH), 8006 Zurich, Switzerland
| |
Collapse
|
16
|
Moore ST, Nakamura T, Nie J, Solivais AJ, Aristizábal-Ramírez I, Ueda Y, Manikandan M, Reddy VS, Romano DR, Hoffman JR, Perrin BJ, Nelson RF, Frolenkov GI, Chuva de Sousa Lopes SM, Hashino E. Generating high-fidelity cochlear organoids from human pluripotent stem cells. Cell Stem Cell 2023; 30:950-961.e7. [PMID: 37419105 PMCID: PMC10695300 DOI: 10.1016/j.stem.2023.06.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 05/15/2023] [Accepted: 06/14/2023] [Indexed: 07/09/2023]
Abstract
Mechanosensitive hair cells in the cochlea are responsible for hearing but are vulnerable to damage by genetic mutations and environmental insults. The paucity of human cochlear tissues makes it difficult to study cochlear hair cells. Organoids offer a compelling platform to study scarce tissues in vitro; however, derivation of cochlear cell types has proven non-trivial. Here, using 3D cultures of human pluripotent stem cells, we sought to replicate key differentiation cues of cochlear specification. We found that timed modulations of Sonic Hedgehog and WNT signaling promote ventral gene expression in otic progenitors. Ventralized otic progenitors subsequently give rise to elaborately patterned epithelia containing hair cells with morphology, marker expression, and functional properties consistent with both outer and inner hair cells in the cochlea. These results suggest that early morphogenic cues are sufficient to drive cochlear induction and establish an unprecedented system to model the human auditory organ.
Collapse
Affiliation(s)
- Stephen T Moore
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Takashi Nakamura
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Otolaryngology-Head & Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Jing Nie
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Alexander J Solivais
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | - Yoshitomo Ueda
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Mayakannan Manikandan
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - V Shweta Reddy
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Daniel R Romano
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - John R Hoffman
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Benjamin J Perrin
- Department of Biology, Purdue School of Science, Indianapolis, IN 46202, USA
| | - Rick F Nelson
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | - Eri Hashino
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
17
|
Erickson T, Biggers WP, Williams K, Butland SE, Venuto A. Regionalized Protein Localization Domains in the Zebrafish Hair Cell Kinocilium. J Dev Biol 2023; 11:28. [PMID: 37367482 DOI: 10.3390/jdb11020028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 05/05/2023] [Accepted: 06/02/2023] [Indexed: 06/28/2023] Open
Abstract
Sensory hair cells are the receptors for auditory, vestibular, and lateral line sensory organs in vertebrates. These cells are distinguished by "hair"-like projections from their apical surface collectively known as the hair bundle. Along with the staircase arrangement of the actin-filled stereocilia, the hair bundle features a single, non-motile, true cilium called the kinocilium. The kinocilium plays an important role in bundle development and the mechanics of sensory detection. To understand more about kinocilial development and structure, we performed a transcriptomic analysis of zebrafish hair cells to identify cilia-associated genes that have yet to be characterized in hair cells. In this study, we focused on three such genes-ankef1a, odf3l2a, and saxo2-because human or mouse orthologs are either associated with sensorineural hearing loss or are located near uncharacterized deafness loci. We made transgenic fish that express fluorescently tagged versions of their proteins, demonstrating their localization to the kinocilia of zebrafish hair cells. Furthermore, we found that Ankef1a, Odf3l2a, and Saxo2 exhibit distinct localization patterns along the length of the kinocilium and within the cell body. Lastly, we have reported a novel overexpression phenotype of Saxo2. Overall, these results suggest that the hair cell kinocilium in zebrafish is regionalized along its proximal-distal axis and set the groundwork to understand more about the roles of these kinocilial proteins in hair cells.
Collapse
Affiliation(s)
- Timothy Erickson
- Department of Biology, University of New Brunswick, Fredericton, NB E3B 5A3, Canada
| | | | - Kevin Williams
- Department of Biology, East Carolina University, Greenville, NC 27858, USA
| | - Shyanne E Butland
- Department of Biology, University of New Brunswick, Fredericton, NB E3B 5A3, Canada
| | - Alexandra Venuto
- Department of Biology, East Carolina University, Greenville, NC 27858, USA
| |
Collapse
|
18
|
Feugere L, Bates A, Emagbetere T, Chapman E, Malcolm LE, Bulmer K, Hardege J, Beltran-Alvarez P, Wollenberg Valero KC. Heat induces multiomic and phenotypic stress propagation in zebrafish embryos. PNAS NEXUS 2023; 2:pgad137. [PMID: 37228511 PMCID: PMC10205475 DOI: 10.1093/pnasnexus/pgad137] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 04/11/2023] [Indexed: 05/27/2023]
Abstract
Heat alters biology from molecular to ecological levels, but may also have unknown indirect effects. This includes the concept that animals exposed to abiotic stress can induce stress in naive receivers. Here, we provide a comprehensive picture of the molecular signatures of this process, by integrating multiomic and phenotypic data. In individual zebrafish embryos, repeated heat peaks elicited both a molecular response and a burst of accelerated growth followed by a growth slowdown in concert with reduced responses to novel stimuli. Metabolomes of the media of heat treated vs. untreated embryos revealed candidate stress metabolites including sulfur-containing compounds and lipids. These stress metabolites elicited transcriptomic changes in naive receivers related to immune response, extracellular signaling, glycosaminoglycan/keratan sulfate, and lipid metabolism. Consequently, non-heat-exposed receivers (exposed to stress metabolites only) experienced accelerated catch-up growth in concert with reduced swimming performance. The combination of heat and stress metabolites accelerated development the most, mediated by apelin signaling. Our results prove the concept of indirect heat-induced stress propagation toward naive receivers, inducing phenotypes comparable with those resulting from direct heat exposure, but utilizing distinct molecular pathways. Group-exposing a nonlaboratory zebrafish line, we independently confirm that the glycosaminoglycan biosynthesis-related gene chs1 and the mucus glycoprotein gene prg4a, functionally connected to the candidate stress metabolite classes sugars and phosphocholine, are differentially expressed in receivers. This hints at the production of Schreckstoff-like cues in receivers, leading to further stress propagation within groups, which may have ecological and animal welfare implications for aquatic populations in a changing climate.
Collapse
Affiliation(s)
- Lauric Feugere
- Department of Biological and Marine Sciences, University of Hull, Kingston upon Hull HU6 7RX, UK
| | - Adam Bates
- Department of Biological and Marine Sciences, University of Hull, Kingston upon Hull HU6 7RX, UK
- Wellcome Sanger Institute, Hinxton CB10 1SA, UK
| | - Timothy Emagbetere
- Department of Biological and Marine Sciences, University of Hull, Kingston upon Hull HU6 7RX, UK
| | - Emma Chapman
- Department of Biological and Marine Sciences, University of Hull, Kingston upon Hull HU6 7RX, UK
| | - Linsey E Malcolm
- Biomedical Institute for Multimorbidities, Centre for Biomedicine, Hull York Medical School, University of Hull, Kingston upon Hull HU6 7RX, UK
| | - Kathleen Bulmer
- Biomedical Institute for Multimorbidities, Centre for Biomedicine, Hull York Medical School, University of Hull, Kingston upon Hull HU6 7RX, UK
| | - Jörg Hardege
- Department of Biological and Marine Sciences, University of Hull, Kingston upon Hull HU6 7RX, UK
| | - Pedro Beltran-Alvarez
- Biomedical Institute for Multimorbidities, Centre for Biomedicine, Hull York Medical School, University of Hull, Kingston upon Hull HU6 7RX, UK
| | | |
Collapse
|
19
|
Doda D, Jimenez SA, Rehrauer H, Carre O JF, Valsamides V, Santo SD, Widmer HR, Edge A, Locher H, van der Valk W, Zhang J, Koehler KR, Roccio M. Human pluripotent stem cells-derived inner ear organoids recapitulate otic development in vitro. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.11.536448. [PMID: 37090562 PMCID: PMC10120641 DOI: 10.1101/2023.04.11.536448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Our molecular understanding of the early stages of human inner ear development has been limited by the difficulty in accessing fetal samples at early gestational stages. As an alternative, previous studies have shown that inner ear morphogenesis can be partially recapitulated using induced pluripotent stem cells (iPSCs) directed to differentiate into Inner Ear Organoids (IEOs). Once validated and benchmarked, these systems could represent unique tools to complement and refine our understanding of human otic differentiation and model developmental defects. Here, we provide the first direct comparisons of the early human embryonic otocyst and human iPSC-derived IEOs. We use multiplexed immunostaining, and single-cell RNA sequencing to characterize IEOs at three key developmental steps, providing a new and unique signature of in vitro derived otic -placode, -epithelium, -neuroblasts, and -sensory epithelia. In parallel, we evaluate the expression and localization of critical markers at these equivalent stages in human embryos. We show that the placode derived in vitro (days 8-12) has similar marker expression to the developing otic placode of Carnegie Stage (CS) 11 embryos and subsequently (days 20-40) this gives rise to otic epithelia and neuroblasts comparable to the CS13 embryonic stage. Differentiation of sensory epithelia, including supporting cells and hair cells starts in vitro at days 50-60 of culture. The maturity of these cells is equivalent to vestibular sensory epithelia at week 10 or cochlear tissue at week 12 of development, before functional onset. Together, our data indicate that the current state-of-the-art protocol enables the specification of bona fide otic tissue, supporting the further application of IEOs to inform inner ear biology and disease.
Collapse
|
20
|
Carlton AJ, Jeng J, Grandi FC, De Faveri F, Ceriani F, De Tomasi L, Underhill A, Johnson SL, Legan KP, Kros CJ, Richardson GP, Mustapha M, Marcotti W. A critical period of prehearing spontaneous Ca 2+ spiking is required for hair-bundle maintenance in inner hair cells. EMBO J 2023; 42:e112118. [PMID: 36594367 PMCID: PMC9929643 DOI: 10.15252/embj.2022112118] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 11/22/2022] [Accepted: 11/28/2022] [Indexed: 01/04/2023] Open
Abstract
Sensory-independent Ca2+ spiking regulates the development of mammalian sensory systems. In the immature cochlea, inner hair cells (IHCs) fire spontaneous Ca2+ action potentials (APs) that are generated either intrinsically or by intercellular Ca2+ waves in the nonsensory cells. The extent to which either or both of these Ca2+ signalling mechansims are required for IHC maturation is unknown. We find that intrinsic Ca2+ APs in IHCs, but not those elicited by Ca2+ waves, regulate the maturation and maintenance of the stereociliary hair bundles. Using a mouse model in which the potassium channel Kir2.1 is reversibly overexpressed in IHCs (Kir2.1-OE), we find that IHC membrane hyperpolarization prevents IHCs from generating intrinsic Ca2+ APs but not APs induced by Ca2+ waves. Absence of intrinsic Ca2+ APs leads to the loss of mechanoelectrical transduction in IHCs prior to hearing onset due to progressive loss or fusion of stereocilia. RNA-sequencing data show that pathways involved in morphogenesis, actin filament-based processes, and Rho-GTPase signaling are upregulated in Kir2.1-OE mice. By manipulating in vivo expression of Kir2.1 channels, we identify a "critical time period" during which intrinsic Ca2+ APs in IHCs regulate hair-bundle function.
Collapse
Affiliation(s)
| | - Jing‐Yi Jeng
- School of BiosciencesUniversity of SheffieldSheffieldUK
| | | | | | | | | | | | - Stuart L Johnson
- School of BiosciencesUniversity of SheffieldSheffieldUK
- Neuroscience InstituteUniversity of SheffieldSheffieldUK
| | - Kevin P Legan
- School of Life SciencesUniversity of Sussex, FalmerBrightonUK
| | - Corné J Kros
- School of Life SciencesUniversity of Sussex, FalmerBrightonUK
| | | | - Mirna Mustapha
- School of BiosciencesUniversity of SheffieldSheffieldUK
- Neuroscience InstituteUniversity of SheffieldSheffieldUK
| | - Walter Marcotti
- School of BiosciencesUniversity of SheffieldSheffieldUK
- Neuroscience InstituteUniversity of SheffieldSheffieldUK
| |
Collapse
|
21
|
New Insights into the Identity of the DFNA58 Gene. Genes (Basel) 2022; 13:genes13122274. [PMID: 36553541 PMCID: PMC9777997 DOI: 10.3390/genes13122274] [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: 10/18/2022] [Revised: 11/21/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
Hearing loss is the most common sensory deficit, affecting 466 million people worldwide. The vast and diverse genes involved reflect the complexity of auditory physiology, which requires the use of animal models in order to gain a fuller understanding. Among the loci with a yet-to-be validated gene is the DFNA58, in which ~200 Kb genomic duplication, including three protein-coding genes (PLEK, CNRIP1, and PPP3R1's exon1), was found to segregate with autosomal dominant hearing loss. Through whole genome sequencing, the duplication was found to be in tandem and inserted in an intergenic region, without the disruption of the topological domains. Reanalysis of transcriptomes data studies (zebrafish and mouse), and RT-qPCR analysis of adult zebrafish target organs, in order to access their orthologues expression, highlighted promising results with Cnrip1a, corroborated by zebrafish in situ hybridization and immunofluorescence. Mouse data also suggested Cnrip1 as the best candidate for a relevant role in auditory physiology, and its importance in hearing seems to have remained conserved but the cell type exerting its function might have changed, from hair cells to spiral ganglion neurons.
Collapse
|
22
|
Connolly K, Gonzalez-Cordero A. Modelling inner ear development and disease using pluripotent stem cells - a pathway to new therapeutic strategies. Dis Model Mech 2022; 15:dmm049593. [PMID: 36331565 PMCID: PMC10621662 DOI: 10.1242/dmm.049593] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023] Open
Abstract
The sensory epithelia of the mammalian inner ear enable sound and movement to be perceived. Damage to these epithelia can cause irreversible sensorineural hearing loss and vestibular dysfunction because they lack regenerative capacity. The human inner ear cannot be biopsied without causing permanent damage, significantly limiting the tissue samples available for research. Investigating disease pathology and therapeutic developments have therefore traditionally relied on animal models, which often cannot completely recapitulate the human otic systems. These challenges are now being partly addressed using induced pluripotent stem cell-derived cultures, which generate the sensory epithelial-like tissues of the inner ear. Here, we review how pluripotent stem cells have been used to produce two-dimensional and three-dimensional otic cultures, the strengths and limitations of these new approaches, and how they have been employed to investigate genetic and acquired forms of audiovestibular dysfunction. This Review provides an overview of the progress in pluripotent stem cell-derived otic cultures thus far, focusing on their applications in disease modelling and therapeutic trials. We survey their current limitations and future directions, highlighting their prospective utility for high-throughput drug screening and developing personalised medicine approaches.
Collapse
Affiliation(s)
- Keeva Connolly
- Stem Cell Medicine Group, Children's Medical Research Institute, Westmead, 2145 NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Westmead, 2145 NSW, Australia
| | - Anai Gonzalez-Cordero
- Stem Cell Medicine Group, Children's Medical Research Institute, Westmead, 2145 NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Westmead, 2145 NSW, Australia
| |
Collapse
|
23
|
Gao G, Guo S, Zhang Q, Zhang H, Zhang C, Peng G. Kiaa1024L/Minar2 is essential for hearing by regulating cholesterol distribution in hair bundles. eLife 2022; 11:e80865. [PMID: 36317962 PMCID: PMC9714970 DOI: 10.7554/elife.80865] [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: 06/07/2022] [Accepted: 10/31/2022] [Indexed: 12/05/2022] Open
Abstract
Unbiased genetic screens implicated a number of uncharacterized genes in hearing loss, suggesting some biological processes required for auditory function remain unexplored. Loss of Kiaa1024L/Minar2, a previously understudied gene, caused deafness in mice, but how it functioned in the hearing was unclear. Here, we show that disruption of kiaa1024L/minar2 causes hearing loss in the zebrafish. Defects in mechanotransduction, longer and thinner hair bundles, and enlarged apical lysosomes in hair cells are observed in the kiaa1024L/minar2 mutant. In cultured cells, Kiaa1024L/Minar2 is mainly localized to lysosomes, and its overexpression recruits cholesterol and increases cholesterol labeling. Strikingly, cholesterol is highly enriched in the hair bundle membrane, and loss of kiaa1024L/minar2 reduces cholesterol localization to the hair bundles. Lowering cholesterol levels aggravates, while increasing cholesterol levels rescues the hair cell defects in the kiaa1024L/minar2 mutant. Therefore, cholesterol plays an essential role in hair bundles, and Kiaa1024L/Minar2 regulates cholesterol distribution and homeostasis to ensure normal hearing.
Collapse
Affiliation(s)
- Ge Gao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan UniversityShanghaiChina
| | - Shuyu Guo
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan UniversityShanghaiChina
| | - Quan Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan UniversityShanghaiChina
| | - Hefei Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan UniversityShanghaiChina
| | - Cuizhen Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan UniversityShanghaiChina
| | - Gang Peng
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan UniversityShanghaiChina
| |
Collapse
|
24
|
You D, Guo J, Zhang Y, Guo L, Lu X, Huang X, Sun S, Li H. The heterogeneity of mammalian utricular cells over the course of development. Clin Transl Med 2022; 12:e1052. [PMID: 36178017 PMCID: PMC9523683 DOI: 10.1002/ctm2.1052] [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: 03/21/2022] [Revised: 08/19/2022] [Accepted: 08/25/2022] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND The inner ear organ is a delicate tissue consisting of hair cells (HCs) and supporting cells (SCs).The mammalian inner ear HCs are terminally differentiated cells that cannot spontaneously regenerate in adults. Epithelial non-hair cells (ENHCs) in the utricle include HC progenitors and SCs, and the progenitors share similar characteristics with SCs in the neonatal inner ear. METHODS We applied single-cell sequencing to whole mouse utricles from the neonatal period to adulthood, including samples from postnatal day (P)2, P7 and P30 mice. Furthermore, using transgenic mice and immunostaining, we traced the source of new HC generation. RESULTS We identified several sensory epithelial cell clusters and further found that new HCs arose mainly through differentiation from Sox9+ progenitor cells and that only a few cells were produced by mitotic proliferation in both neonatal and adult mouse utricles. In addition, we identified the proliferative cells using the marker UbcH10 and demonstrated that in adulthood the mitotically generated HCs were primarily found in the extrastriola. Moreover, we observed that not only Type II, but also Type I HCs could be regenerated by either mitotic cell proliferation or progenitor cell differentiation. CONCLUSIONS Overall, our findings expand our understanding of ENHC cell fate and the characteristics of the vestibular organs in mammals over the course of development.
Collapse
Affiliation(s)
- Dan You
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina,Department of Otorhinolaryngology‐Head and Neck SurgeryZhongshan HospitalFudan UniversityShanghaiChina
| | - Jin Guo
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina
| | - Yunzhong Zhang
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina
| | - Luo Guo
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina
| | - Xiaoling Lu
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina
| | - Xinsheng Huang
- Department of Otorhinolaryngology‐Head and Neck SurgeryZhongshan HospitalFudan UniversityShanghaiChina
| | - Shan Sun
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina
| | - Huawei Li
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina,Institutes of Biomedical SciencesFudan UniversityShanghaiChina,NHC Key Laboratory of Hearing Medicine, Fudan UniversityShanghaiChina,The Institutes of Brain Science and the Collaborative Innovation Center for Brain ScienceFudan UniversityShanghaiChina
| |
Collapse
|
25
|
Abstract
Current estimates suggest that nearly half a billion people worldwide are affected by hearing loss. Because of the major psychological, social, economic, and health ramifications, considerable efforts have been invested in identifying the genes and molecular pathways involved in hearing loss, whether genetic or environmental, to promote prevention, improve rehabilitation, and develop therapeutics. Genomic sequencing technologies have led to the discovery of genes associated with hearing loss. Studies of the transcriptome and epigenome of the inner ear have characterized key regulators and pathways involved in the development of the inner ear and have paved the way for their use in regenerative medicine. In parallel, the immense preclinical success of using viral vectors for gene delivery in animal models of hearing loss has motivated the industry to work on translating such approaches into the clinic. Here, we review the recent advances in the genomics of auditory function and dysfunction, from patient diagnostics to epigenetics and gene therapy.
Collapse
Affiliation(s)
- Shahar Taiber
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; ,
| | - Kathleen Gwilliam
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA; ,
| | - Ronna Hertzano
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA; ,
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Karen B Avraham
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; ,
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
26
|
Mutations in MINAR2 encoding membrane integral NOTCH2-associated receptor 2 cause deafness in humans and mice. Proc Natl Acad Sci U S A 2022; 119:e2204084119. [PMID: 35727972 PMCID: PMC9245706 DOI: 10.1073/pnas.2204084119] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Discovery of deafness genes and elucidating their functions have substantially contributed to our understanding of hearing physiology and its pathologies. Here we report on DNA variants in MINAR2, encoding membrane integral NOTCH2-associated receptor 2, in four families underlying autosomal recessive nonsyndromic deafness. Neurologic evaluation of affected individuals at ages ranging from 4 to 80 y old does not show additional abnormalities. MINAR2 is a recently annotated gene with limited functional understanding. We detected three MINAR2 variants, c.144G > A (p.Trp48*), c.412_419delCGGTTTTG (p.Arg138Valfs*10), and c.393G > T, in 13 individuals with congenital- or prelingual-onset severe-to-profound sensorineural hearing loss (HL). The c.393G > T variant is shown to disrupt a splice donor site. We show that Minar2 is expressed in the mouse inner ear, with the protein localizing mainly in the hair cells, spiral ganglia, the spiral limbus, and the stria vascularis. Mice with loss of function of the Minar2 protein (Minar2tm1b/tm1b) present with rapidly progressive sensorineural HL associated with a reduction in outer hair cell stereocilia in the shortest row and degeneration of hair cells at a later age. We conclude that MINAR2 is essential for hearing in humans and mice and its disruption leads to sensorineural HL. Progressive HL observed in mice and in some affected individuals and as well as relative preservation of hair cells provides an opportunity to interfere with HL using genetic therapies.
Collapse
|
27
|
Oncomodulin (OCM) uniquely regulates calcium signaling in neonatal cochlear outer hair cells. Cell Calcium 2022; 105:102613. [PMID: 35797824 PMCID: PMC9297295 DOI: 10.1016/j.ceca.2022.102613] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/06/2022] [Accepted: 06/09/2022] [Indexed: 11/23/2022]
|
28
|
Induction of functional neutrophils from mouse fibroblasts by thymidine through enhancement of Tet3 activity. Cell Mol Immunol 2022; 19:619-633. [PMID: 35301470 PMCID: PMC9061759 DOI: 10.1038/s41423-022-00842-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/27/2022] [Indexed: 11/08/2022] Open
Abstract
Neutrophils are derived from bone marrow hematopoietic stem cells (HSCs) and are the largest population among circulating white blood cells in humans, acting as the first line of defense against invading pathogens. Whether neutrophils can be generated by transdifferentiation strategies is unknown. Here, we show that thymidine induces the conversion of mouse fibroblasts to neutrophils. Induced neutrophils (iNeus) showed antibacterial effects and did not undergo malignant transformation in vivo. Importantly, iNeu transplantation cured neutropenia in mice in vivo. Mechanistically, thymidine mediates iNeu conversion by enhancing Tet3 activity. Tet3 initiates the expression of the neutrophil fate decision factors Cebpδ and Rfx1 that drive the transdifferentiation of mouse fibroblasts to neutrophils. Therefore, the induction of functional neutrophils by chemicals may provide a potential therapeutic strategy for patients with neutropenia patients and infectious diseases.Fibroblasts; Neutrophils; Thymidine; Transdifferentiation; Tet3.
Collapse
|
29
|
ASK1 is a novel molecular target for preventing aminoglycoside-induced hair cell death. J Mol Med (Berl) 2022; 100:797-813. [PMID: 35471608 PMCID: PMC9110505 DOI: 10.1007/s00109-022-02188-1] [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: 06/09/2021] [Revised: 02/07/2022] [Accepted: 03/03/2022] [Indexed: 10/31/2022]
Abstract
Aminoglycoside antibiotics are lifesaving medicines, crucial for the treatment of chronic or drug resistant infections. However, aminoglycosides are toxic to the sensory hair cells in the inner ear. As a result, aminoglycoside-treated individuals can develop permanent hearing loss and vestibular impairment. There is considerable evidence that reactive oxygen species (ROS) production and the subsequent phosphorylation of c-Jun N-terminal kinase (JNK) and P38 mitogen-activated protein kinase (P38) drives apoptosis in aminoglycoside-treated hair cells. However, treatment strategies that directly inhibit ROS, JNK, or P38 are limited by the importance of these molecules for normal cellular function. Alternatively, the upstream regulator apoptosis signal-regulating kinase 1 (ASK1/MAP3K5) is a key mediator of ROS-induced JNK and P38 activation under pathologic but not homeostatic conditions. We investigated ASK1 as a mediator of drug-induced hair cell death using cochlear explants from Ask1 knockout mice, demonstrating that Ask1 deficiency attenuates neomycin-induced hair cell death. We then evaluated pharmacological inhibition of ASK1 with GS-444217 as a potential otoprotective therapy. GS-444217 significantly attenuated hair cell death in neomycin-treated explants but did not impact aminoglycoside efficacy against P. aeruginosa in the broth dilution test. Overall, we provide significant pre-clinical evidence that ASK1 inhibition represents a novel strategy for preventing aminoglycoside ototoxicity. KEY MESSAGES: • ASK1 is an upstream, redox-sensitive regulator of P38 and JNK, which are known mediators of hair cell death. • Ask1 knockout does not affect hair cell development in vivo, but significantly reduces aminoglycoside-induced hair cell death in vitro. • A small-molecule inhibitor of ASK1 attenuates neomycin-induced hair cell death, and does not impact antibiotic efficacy in vitro. • ASK1 may be a novel molecular target for preventing aminoglycoside-induced hearing loss.
Collapse
|
30
|
Oh JK, Vargas Del Valle JG, Lima de Carvalho JR, Sun YJ, Levi SR, Ryu J, Yang J, Nagasaki T, Emanuelli A, Rasool N, Allikmets R, Sparrow JR, Izquierdo NJ, Duncan JL, Mahajan VB, Tsang SH. Expanding the phenotype of TTLL5-associated retinal dystrophy: a case series. Orphanet J Rare Dis 2022; 17:146. [PMID: 35365235 PMCID: PMC8973795 DOI: 10.1186/s13023-022-02295-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 03/21/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Inherited retinal dystrophies describe a heterogeneous group of retinal diseases that lead to the irreversible degeneration of rod and cone photoreceptors and eventual blindness. Recessive loss-of-function mutations in Tubulin Tyrosine Ligase Like 5 (TTLL5) represent a recently described cause of inherited cone-rod and cone dystrophy. This study describes the unusual phenotypes of three patients with autosomal recessive mutations in TTLL5. Examination of these patients included funduscopic evaluation, spectral-domain optical coherence tomography, short-wavelength autofluorescence, and full-field electroretinography (ffERG). Genetic diagnoses were confirmed using whole exome capture. Protein modeling of the identified variants was performed to explore potential genotype-phenotype correlations. RESULTS Genetic testing revealed five novel variants in TTLL5 in three unrelated patients with retinal dystrophy. Clinical imaging demonstrated features of sectoral cone-rod dystrophy and cone dystrophy, with phenotypic variability seen across all three patients. One patient also developed high-frequency hearing loss during a similar time period as the onset of retinal disease, potentially suggestive of a syndromic disorder. Retinal structure findings were corroborated with functional measures including ffERG findings that supported these diagnoses. Modeling of the five variants suggest that they cause different effects on protein function, providing a potential reason for genotype-phenotype correlation in these patients. CONCLUSIONS The authors report retinal phenotypic findings in three unrelated patients with novel mutations causing autosomal recessive TTLL5-mediated retinal dystrophy. These findings broaden the understanding of the phenotypes associated with TTLL5-mediated retinal disease and suggest that mutations in TTLL5 should be considered as a potential cause of sectoral retinal dystrophy in addition to cone-rod and cone dystrophies.
Collapse
Affiliation(s)
- Jin Kyun Oh
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA
- State University of New York at Downstate Medical Center, Brooklyn, NY, USA
| | | | - Jose Ronaldo Lima de Carvalho
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Ophthalmology, Hospital das Clinicas de Pernambuco (HCPE) - Empresa Brasileira de Servicos Hospitalares (EBSERH), Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
- Department of Ophthalmology, Federal University of São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Young Joo Sun
- Omics Laboratory, Byers Eye Institute, Stanford University, Palo Alto, CA, USA
| | - Sarah R Levi
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA
| | - Joseph Ryu
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA
| | - Jing Yang
- Omics Laboratory, Byers Eye Institute, Stanford University, Palo Alto, CA, USA
| | - Takayuki Nagasaki
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA
| | - Andres Emanuelli
- Department of Ophthalmology, Medical Sciences Campus, University of Puerto Rico, San Juan, PR, USA
| | - Nailyn Rasool
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Rando Allikmets
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Janet R Sparrow
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Natalio J Izquierdo
- Department of Surgery, Medical Sciences Campus, University of Puerto Rico, San Juan, PR, USA
| | - Jacque L Duncan
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Vinit B Mahajan
- Omics Laboratory, Byers Eye Institute, Stanford University, Palo Alto, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Stephen H Tsang
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, NY, USA.
- Harkness Eye Institute, Columbia University Medical Center, 635 West 165th Street, Box 212, New York, NY, 10032, USA.
| |
Collapse
|
31
|
Hertzano R, Mahurkar A. Advancing discovery in hearing research via biologist-friendly access to multi-omic data. Hum Genet 2022; 141:319-322. [PMID: 35235019 PMCID: PMC9034999 DOI: 10.1007/s00439-022-02445-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 02/24/2022] [Indexed: 01/01/2023]
Abstract
High-throughput cell type-specific multi-omic analyses have advanced our understanding of inner ear biology in an unprecedented way. The full benefit of these data, however, is reached from their re-use. Successful re-use of data requires identifying the natural users and ensuring proper data democratization and federation for their seamless and meaningful access. Here we discuss universal challenges in access and re-use of multi-omic data, possible solutions, and introduce the gEAR (the gene Expression Analysis Resource, umgear.org)-a tool for multi-omic data visualization, sharing and access for the ear field.
Collapse
Affiliation(s)
- Ronna Hertzano
- Department of Otorhinolaryngology Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| | - Anup Mahurkar
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| |
Collapse
|
32
|
Lorente-Cánovas B, Eckrich S, Lewis MA, Johnson SL, Marcotti W, Steel KP. Grxcr1 regulates hair bundle morphogenesis and is required for normal mechanoelectrical transduction in mouse cochlear hair cells. PLoS One 2022; 17:e0261530. [PMID: 35235570 PMCID: PMC8890737 DOI: 10.1371/journal.pone.0261530] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 12/03/2021] [Indexed: 12/02/2022] Open
Abstract
Tasmanian devil (tde) mice are deaf and exhibit circling behaviour. Sensory hair cells of mutants show disorganised hair bundles with abnormally thin stereocilia. The origin of this mutation is the insertion of a transgene which disrupts expression of the Grxcr1 (glutaredoxin cysteine rich 1) gene. We report here that Grxcr1 exons and transcript sequences are not affected by the transgene insertion in tde homozygous (tde/tde) mice. Furthermore, 5'RACE PCR experiments showed the presence of two different transcripts of the Grxcr1 gene, expressed in both tde/tde and in wild-type controls. However, quantitative analysis of Grxcr1 transcripts revealed a significantly decreased mRNA level in tde/tde mice. The key stereociliary proteins ESPN, MYO7A, EPS8 and PTPRQ were distributed in hair bundles of homozygous tde mutants in a similar pattern compared with control mice. We found that the abnormal morphology of the stereociliary bundle was associated with a reduction in the size and Ca2+-sensitivity of the mechanoelectrical transducer (MET) current. We propose that GRXCR1 is key for the normal growth of the stereociliary bundle prior to the onset of hearing, and in its absence hair cells are unable to mature into fully functional sensory receptors.
Collapse
Affiliation(s)
- Beatriz Lorente-Cánovas
- Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom
- Wellcome Sanger Institute, Hinxton, United Kingdom
| | - Stephanie Eckrich
- School of Biosciences, University of Sheffield, Sheffield, United Kingdom
| | - Morag A. Lewis
- Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom
- Wellcome Sanger Institute, Hinxton, United Kingdom
| | - Stuart L. Johnson
- School of Biosciences, University of Sheffield, Sheffield, United Kingdom
- Neuroscience Institute, University of Sheffield, Sheffield, United Kingdom
| | - Walter Marcotti
- School of Biosciences, University of Sheffield, Sheffield, United Kingdom
- Neuroscience Institute, University of Sheffield, Sheffield, United Kingdom
| | - Karen P. Steel
- Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom
- Wellcome Sanger Institute, Hinxton, United Kingdom
| |
Collapse
|
33
|
Multiplexed Genome Editing for Efficient Phenotypic Screening in Zebrafish. Vet Sci 2022; 9:vetsci9020092. [PMID: 35202345 PMCID: PMC8879510 DOI: 10.3390/vetsci9020092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 12/30/2022] Open
Abstract
Zebrafish are widely used to investigate candidate genes for human diseases. While the emergence of CRISPR-Cas9 technology has revolutionized gene editing, the use of individual guide RNAs limits the efficiency and application of this technology in functional genetics research. Multiplexed genome editing significantly enhances the efficiency and scope of gene editing. Herein, we describe an efficient multiplexed genome editing strategy to generate zebrafish mutants. Following behavioural tests and histological examination, we identified one new candidate gene (tmem183a) for hearing loss. This study provides a robust genetic platform to quickly obtain zebrafish mutants and to identify candidate genes by phenotypic readouts.
Collapse
|
34
|
Frejo L, Lopez-Escamez JA. Cytokines and Inflammation in Meniere Disease. Clin Exp Otorhinolaryngol 2022; 15:49-59. [PMID: 35124944 PMCID: PMC8901949 DOI: 10.21053/ceo.2021.00920] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/20/2021] [Indexed: 12/05/2022] Open
Abstract
Meniere disease (MD) is a rare set of conditions associated with the accumulation of endolymph in the cochlear duct and the vestibular labyrinth with a decrease of endocochlear potential. It is considered a chronic inflammatory disorder of the inner ear with a multifactorial origin. The clinical syndrome includes several groups of patients with a core phenotype: sensorineural hearing loss, episodes of vertigo, and tinnitus with a non-predictable course. Genetic factors and the innate immune response seem to play a central role in the pathophysiology of the condition. Autoimmune MD should be diagnosed if a patient fulfills the diagnostic criteria for MD and one of the following autoimmune disorders: autoimmune thyroid disease, psoriasis, autoimmune arthritis, ankylosing spondylitis, or systemic lupus erythematosus. We summarize the evidence to support autoimmune MD as an endophenotype in bilateral MD associated with the allelic variant rs4947296 and nuclear factor-kappa B (NF-κB)-mediated inflammation, the role of cytokines (particularly interleukin-1β and tumor necrosis factor-α) in defining a subset of patients with autoinflammation, and the potential role of cytokines as biomarkers to distinguish between patients with MD and vestibular migraine. Finally, we also introduce a list of potential drugs that could regulate the immune response in MD with potential for repurposing in clinical trials.
Collapse
Affiliation(s)
- Lidia Frejo
- Otology and Neurotology Group CTS495, Department of Genomic Medicine, Centre for Genomics and Oncological Research (GENYO), Pfizer-University of Granada-Junta de Andalucía, PTS, Granada, Spain.,Department of Otolaryngology, Instituto de Investigación Biosanitaria ibs. Granada, Hospital Universitario Virgen de las Nieves, Granada, Spain.,Sensorineural Pathology Programme, Centro de Investigación Biomédica en Red en Enfermedades Raras, CIBERER, Madrid, Spain
| | - Jose Antonio Lopez-Escamez
- Otology and Neurotology Group CTS495, Department of Genomic Medicine, Centre for Genomics and Oncological Research (GENYO), Pfizer-University of Granada-Junta de Andalucía, PTS, Granada, Spain.,Department of Otolaryngology, Instituto de Investigación Biosanitaria ibs. Granada, Hospital Universitario Virgen de las Nieves, Granada, Spain.,Sensorineural Pathology Programme, Centro de Investigación Biomédica en Red en Enfermedades Raras, CIBERER, Madrid, Spain.,Department of Surgery, Division of Otolaryngology, Universidad de Granada, Granada, Spain
| |
Collapse
|
35
|
Herranz-Pérez V, Nakatani J, Ishii M, Katada T, García-Verdugo JM, Ohata S. Ependymoma associated protein Zfta is expressed in immature ependymal cells but is not essential for ependymal development in mice. Sci Rep 2022; 12:1493. [PMID: 35087169 PMCID: PMC8795269 DOI: 10.1038/s41598-022-05526-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/14/2022] [Indexed: 12/14/2022] Open
Abstract
The fusion protein of uncharacterised zinc finger translocation associated (ZFTA) and effector transcription factor of tumorigenic NF-κB signalling, RELA (ZFTA-RELA), is expressed in more than two-thirds of supratentorial ependymoma (ST-EPN-RELA), but ZFTA's expression profile and functional analysis in multiciliated ependymal (E1) cells have not been examined. Here, we showed the mRNA expression of mouse Zfta peaks on embryonic day (E) 17.5 in the wholemount of the lateral walls of the lateral ventricle. Zfta was expressed in the nuclei of FoxJ1-positive immature E1 (pre-E1) cells in E18.5 mouse embryonic brain. Interestingly, the transcription factors promoting ciliogenesis (ciliary TFs) (e.g., multicilin) and ZFTA-RELA upregulated luciferase activity using a 5' upstream sequence of ZFTA in cultured cells. Zftatm1/tm1 knock-in mice did not show developmental defects or abnormal fertility. In the Zftatm1/tm1 E1 cells, morphology, gene expression, ciliary beating frequency and ependymal flow were unaffected. These results suggest that Zfta is expressed in pre-E1 cells, possibly under the control of ciliary TFs, but is not essential for ependymal development or flow. This study sheds light on the mechanism of the ZFTA-RELA expression in the pathogenesis of ST-EPN-RELA: Ciliary TFs initiate ZFTA-RELA expression in pre-E1 cells, and ZFTA-RELA enhances its own expression using positive feedback.
Collapse
Affiliation(s)
- Vicente Herranz-Pérez
- Laboratory of Comparative Neurobiology, Institute Cavanilles of Biodiversity and Evolutionary Biology, CIBERNED, University of Valencia, 46980, Paterna, Spain
- Department of Cell Biology, Functional Biology and Physical Anthropology, University of Valencia, 46100, Burjassot, Spain
| | - Jin Nakatani
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Shiga, 525-8577, Japan
| | - Masaki Ishii
- Molecular Cell Biology Laboratory, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo, 202-8585, Japan
| | - Toshiaki Katada
- Molecular Cell Biology Laboratory, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo, 202-8585, Japan
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, 113-0033, Japan
| | - Jose Manuel García-Verdugo
- Laboratory of Comparative Neurobiology, Institute Cavanilles of Biodiversity and Evolutionary Biology, CIBERNED, University of Valencia, 46980, Paterna, Spain
- Department of Cell Biology, Functional Biology and Physical Anthropology, University of Valencia, 46100, Burjassot, Spain
| | - Shinya Ohata
- Molecular Cell Biology Laboratory, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo, 202-8585, Japan.
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, 113-0033, Japan.
| |
Collapse
|
36
|
Khan SA, Khan S, Muhammad N, Rehman ZU, Khan MA, Nasir A, Kalsoom UE, Khan AK, Khan H, Wasif N. The First Report of a Missense Variant in RFX2 Causing Non-Syndromic Tooth Agenesis in a Consanguineous Pakistani Family. Front Genet 2022; 12:782653. [PMID: 35145545 PMCID: PMC8822170 DOI: 10.3389/fgene.2021.782653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 12/07/2021] [Indexed: 11/13/2022] Open
Abstract
Background: The syndromic and non-syndromic congenital missing teeth phenotype is termed tooth agenesis. Since tooth agenesis is a heterogeneous disorder hence, the patients show diverse absent teeth phenotypes. Thus identifying novel genes involved in the morphogenesis of ectodermal appendages, including teeth, paves the way for establishing signaling pathways.Methods and Results: We have recruited an autosomal recessive non-syndromic tooth agenesis family with two affected members. The exome sequencing technology identified a novel missense sequence variant c.1421T > C; p.(Ile474Thr) in a regulatory factor X (RFX) family member (RFX2, OMIM: 142,765). During the data analysis eight rare variants on various chromosomal locations were identified, but the co-segregation analysis using Sanger sequencing confirmed the segregation of only two variants RFX2: c.1421T > C; p.(Ile474Thr), DOHH: c.109C > G; p.(Pro37Ala) lying in a common 7.1 MB region of homozygosity on chromosome 19p13.3. Furthermore, the online protein prediction algorithms and protein modeling analysis verified the RFX2 variant as a damaging genetic alteration and ACMG pathogenicity criteria classified it as likely pathogenic. On the other hand, the DOHH variant showed benign outcomes.Conclusion:RFX2 regulates the Hedgehog and fibroblast growth factor signaling pathways, which are involved in the epithelial and mesenchymal interactions during tooth development. Prior animal model studies have confirmed the expression of rfx2 at a developmental stage governing mouth formation. Moreover, its regulatory role and close association with ciliary and non-ciliary genes causing various dental malformations makes it a potential candidate gene for tooth agenesis phenotype. Further studies will contribute to exploring the direct role of RFX2 in human tooth development.
Collapse
Affiliation(s)
- Sher Alam Khan
- Department of Biotechnology and Genetic Engineering, Kohat University of Science and Technology (KUST), Kohat, Pakistan
| | - Saadullah Khan
- Department of Biotechnology and Genetic Engineering, Kohat University of Science and Technology (KUST), Kohat, Pakistan
- *Correspondence: Saadullah Khan, ; Naveed Wasif,
| | - Noor Muhammad
- Department of Biotechnology and Genetic Engineering, Kohat University of Science and Technology (KUST), Kohat, Pakistan
| | - Zia Ur Rehman
- Department of Biotechnology and Genetic Engineering, Kohat University of Science and Technology (KUST), Kohat, Pakistan
| | - Muhammad Adnan Khan
- Dental Material, Institute of Basic Medical Sciences, Khyber Medical University Peshawar, Peshawar, Pakistan
| | - Abdul Nasir
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Umm-e- Kalsoom
- Department of Biochemistry, Hazara University, Mansehra, Pakistan
| | - Anwar Kamal Khan
- Department of Biotechnology and Genetic Engineering, Kohat University of Science and Technology (KUST), Kohat, Pakistan
| | - Hassan Khan
- Department of Biotechnology and Genetic Engineering, Kohat University of Science and Technology (KUST), Kohat, Pakistan
| | - Naveed Wasif
- Institute of Human Genetics, University of Ulm, Ulm, Germany
- Institute of Human Genetics, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
- *Correspondence: Saadullah Khan, ; Naveed Wasif,
| |
Collapse
|
37
|
Lu L, Li X, Zhong Z, Zhou W, Zhou D, Zhu M, Miao C. KMT5A downregulation participated in High Glucose-mediated EndMT via Upregulation of ENO1 Expression in Diabetic Nephropathy. Int J Biol Sci 2021; 17:4093-4107. [PMID: 34803485 PMCID: PMC8579450 DOI: 10.7150/ijbs.62867] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 09/26/2021] [Indexed: 12/17/2022] Open
Abstract
Diabetic nephropathy (DN) has become the common and principal microvascular complication of diabetes that could lead to end-stage renal disease. It was reported endothelial-to-mesenchymal transition (EndMT) in glomeruli plays an important role in DN. Enolase1 (ENO1) and Lysine Methyltransferase 5A (KMT5A) were found to modulate epithelial-to-mesenchymal transition in some situations. In the present study, we speculated KMT5A regulates ENO1 transcript, thus participating in hyperglycemia-induced EndMT in glomeruli of DN. Our study represented vimentin, αSMA and ENO1 expression elevated, and CD31 expression decreased in glomeruli of DN participants and rats. In vitro, high glucose induced EndMT by increase of ENO1 levels. Moreover, high glucose downregulated KMT5A levels and increased regulatory factor X1 (RFX1) levels. KMT5A upregulation or si-RFX1 decreased high glucose-induced ENO1 expression and EndMT. RFX1 overexpression- or sh-KMT5A-induced EndMT was attenuated by si-ENO1. Further, the association between KMT5A and RFX1 was verified. Furthermore, histone H4 lysine20 methylation (the direct target of KMT5A) and RFX1 positioned on ENO1 promoter region. sh-KMT5A enhanced positive action of RFX1 on ENO1 promoter activity. KMT5A reduction and RFX1 upregulation were verified in glomeruli of DN patients and rats. KMT5A associated with RFX1 to modulate ENO1, thus involved in hyperglycemia-mediated EndMT in glomeruli of DN.
Collapse
Affiliation(s)
- Lihong Lu
- Department of Anesthesiology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xue Li
- Department of Anesthesiology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ziwen Zhong
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.,Department of Anesthesiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Wenchang Zhou
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Di Zhou
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Minmin Zhu
- Department of Anesthesiology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, People's Republic of China
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| |
Collapse
|
38
|
Arora D, Park JE, Lim D, Choi BH, Cho IC, Srikanth K, Kim J, Park W. Comparative methylation and RNA-seq expression analysis in CpG context to identify genes involved in Backfat vs. Liver diversification in Nanchukmacdon Pig. BMC Genomics 2021; 22:801. [PMID: 34743693 PMCID: PMC8573883 DOI: 10.1186/s12864-021-08123-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 10/25/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND DNA methylation and demethylation at CpG islands is one of the main regulatory factors that allow cells to respond to different stimuli. These regulatory mechanisms help in developing tissue without affecting the genomic composition or undergoing selection. Liver and backfat play important roles in regulating lipid metabolism and control various pathways involved in reproductive performance, meat quality, and immunity. Genes inside these tissue store a plethora of information and an understanding of these genes is required to enhance tissue characteristics in the future generation. RESULTS A total of 16 CpG islands were identified, and they were involved in differentially methylation regions (DMRs) as well as differentially expressed genes (DEGs) of liver and backfat tissue samples. The genes C7orf50, ACTB and MLC1 in backfat and TNNT3, SIX2, SDK1, CLSTN3, LTBP4, CFAP74, SLC22A23, FOXC1, GMDS, GSC, GATA4, SEMA5A and HOXA5 in the liver, were categorized as differentially-methylated. Subsequently, Motif analysis for DMRs was performed to understand the role of the methylated motif for tissue-specific differentiation. Gene ontology studies revealed association with collagen fibril organization, the Bone Morphogenetic Proteins (BMP) signaling pathway in backfat and cholesterol biosynthesis, bile acid and bile salt transport, and immunity-related pathways in methylated genes expressed in the liver. CONCLUSIONS In this study, to understand the role of genes in the differentiation process, we have performed whole-genome bisulfite sequencing (WGBS) and RNA-seq analysis of Nanchukmacdon pigs. Methylation and motif analysis reveals the critical role of CpG islands and transcriptional factors binding site (TFBS) in guiding the differential patterns. Our findings could help in understanding how methylation of certain genes plays an important role and can be used as biomarkers to study tissue specific characteristics.
Collapse
Affiliation(s)
- Devender Arora
- Animal Genomics and Bioinformatics Division, National Institute of Animal Science, RDA, 55365, Wanju, Republic of Korea
| | - Jong-Eun Park
- Animal Genomics and Bioinformatics Division, National Institute of Animal Science, RDA, 55365, Wanju, Republic of Korea
| | - Dajeong Lim
- Animal Genomics and Bioinformatics Division, National Institute of Animal Science, RDA, 55365, Wanju, Republic of Korea
| | - Bong-Hwan Choi
- Animal Genomics and Bioinformatics Division, National Institute of Animal Science, RDA, 55365, Wanju, Republic of Korea
| | - In-Cheol Cho
- Subtropical Livestock Research Institute, National Institute of Animal Science, RDA, 63242, Jeju, Korea
| | - Krishnamoorthy Srikanth
- Animal Genomics and Bioinformatics Division, National Institute of Animal Science, RDA, 55365, Wanju, Republic of Korea
- Department of Animal Science, Cornell University, NY, 14853, Ithaca, USA
| | - Jaebum Kim
- Department of Biomedical Science and Engineering, Konkuk University, 05029, Seoul, Republic of Korea
| | - Woncheoul Park
- Animal Genomics and Bioinformatics Division, National Institute of Animal Science, RDA, 55365, Wanju, Republic of Korea.
| |
Collapse
|
39
|
Mizukawa T, Nishida T, Akashi S, Kawata K, Kikuchi S, Kawaki H, Takigawa M, Kamioka H, Kubota S. RFX1-mediated CCN3 induction that may support chondrocyte survival under starved conditions. J Cell Physiol 2021; 236:6884-6896. [PMID: 33655492 DOI: 10.1002/jcp.30348] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/12/2021] [Accepted: 02/16/2021] [Indexed: 02/06/2023]
Abstract
Cellular communication network factor (CCN) family members are multifunctional matricellular proteins that manipulate and integrate extracellular signals. In our previous studies investigating the role of CCN family members in cellular metabolism, we found three members that might be under the regulation of energy metabolism. In this study, we confirmed that CCN2 and CCN3 are the only members that are tightly regulated by glycolysis in human chondrocytic cells. Interestingly, CCN3 was induced under a variety of impaired glycolytic conditions. This CCN3 induction was also observed in two breast cancer cell lines with a distinct phenotype, suggesting a basic role of CCN3 in cellular metabolism. Reporter gene assays indicated a transcriptional regulation mediated by an enhancer in the proximal promoter region. As a result of analyses in silico, we specified regulatory factor binding to the X-box 1 (RFX1) as a candidate that mediated the transcriptional activation by impaired glycolysis. Indeed, the inhibition of glycolysis induced the expression of RFX1, and RFX1 silencing nullified the CCN3 induction by impaired glycolysis. Subsequent experiments with an anti-CCN3 antibody indicated that CCN3 supported the survival of chondrocytes under impaired glycolysis. Consistent with these findings in vitro, abundant CCN3 production by chondrocytes in the deep zones of developing epiphysial cartilage, which are located far away from the synovial fluid, was confirmed in vivo. Our present study uncovered that RFX1 is the mediator that enables CCN3 induction upon cellular starvation, which may eventually assist chondrocytes in retaining their viability, even when there is an energy supply shortage.
Collapse
Affiliation(s)
- Tomomi Mizukawa
- Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Department of Orthodontics, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takashi Nishida
- Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Advanced Research Center for Oral and Craniofacial Sciences, Okayama University Dental School, Okayama, Japan
| | - Sho Akashi
- Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Department of Oral and Maxillofacial Reconstructive Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kazumi Kawata
- Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Sumire Kikuchi
- Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Harumi Kawaki
- Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Department of Oral Biochemistry, Asahi University School of Dentistry, Mizuho, Japan
| | - Masaharu Takigawa
- Advanced Research Center for Oral and Craniofacial Sciences, Okayama University Dental School, Okayama, Japan
| | - Hiroshi Kamioka
- Department of Orthodontics, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Satoshi Kubota
- Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
40
|
Munnamalai V, Sammudin NH, Young CA, Thawani A, Kuhn RJ, Fekete DM. Embryonic and Neonatal Mouse Cochleae Are Susceptible to Zika Virus Infection. Viruses 2021; 13:v13091823. [PMID: 34578404 PMCID: PMC8472928 DOI: 10.3390/v13091823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/09/2021] [Accepted: 09/09/2021] [Indexed: 11/16/2022] Open
Abstract
Congenital Zika Syndrome (CZS) is caused by vertical transmission of Zika virus (ZIKV) to the gestating human fetus. A subset of CZS microcephalic infants present with reduced otoacoustic emissions; this test screens for hearing loss originating in the cochlea. This observation leads to the question of whether mammalian cochlear tissues are susceptible to infection by ZIKV during development. To address this question using a mouse model, the sensory cochlea was explanted at proliferative, newly post-mitotic or maturing stages. ZIKV was added for the first 24 h and organs cultured for up to 6 days to allow for cell differentiation. Results showed that ZIKV can robustly infect proliferating sensory progenitors, as well as post-mitotic hair cells and supporting cells. Virus neutralization using ZIKV-117 antibody blocked cochlear infection. AXL is a cell surface molecule known to enhance the attachment of flavivirus to host cells. While Axl mRNA is widely expressed in embryonic cochlear tissues susceptible to ZIKV infection, it is selectively downregulated in the post-mitotic sensory organ by E15.5, even though these cells remain infectible. These findings may offer insights into which target cells could potentially contribute to hearing loss resulting from fetal exposure to ZIKV in humans.
Collapse
Affiliation(s)
- Vidhya Munnamalai
- The Jackson Laboratory, Bar Harbor, ME 04609, USA; (V.M.); (C.A.Y.)
- Graduate School of Biomedical Sciences and Engineering, University of Main, Orono, ME 04469, USA
| | - Nabilah H. Sammudin
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; (N.H.S.); (A.T.); (R.J.K.)
| | - Caryl A. Young
- The Jackson Laboratory, Bar Harbor, ME 04609, USA; (V.M.); (C.A.Y.)
| | - Ankita Thawani
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; (N.H.S.); (A.T.); (R.J.K.)
| | - Richard J. Kuhn
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; (N.H.S.); (A.T.); (R.J.K.)
- Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA
| | - Donna M. Fekete
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; (N.H.S.); (A.T.); (R.J.K.)
- Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA
- Correspondence:
| |
Collapse
|
41
|
Hertzano R, Gwilliam K, Rose K, Milon B, Matern MS. Cell Type-Specific Expression Analysis of the Inner Ear: A Technical Report. Laryngoscope 2021; 131 Suppl 5:S1-S16. [PMID: 32579737 PMCID: PMC8996438 DOI: 10.1002/lary.28765] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/21/2020] [Accepted: 05/01/2020] [Indexed: 01/11/2023]
Abstract
OBJECTIVE The cellular diversity of the inner ear has presented a technical challenge in obtaining molecular insight into its development and function. The application of technological advancements in cell type-specific expression enable clinicians and researchers to leap forward from classic genetics to obtaining mechanistic understanding of congenital and acquired hearing loss. This understanding is essential for development of therapeutics to prevent and reverse diseases of the inner ear, including hearing loss. The objective of this study is to describe and compare the available tools for cell type-specific analysis of the ear, as a means to support decision making in study design. STUDY DESIGN Three major approaches for cell type-specific analysis of the ear including fluorescence-activated cell sorting (FACS), ribosomal and RNA pulldown techniques, and single cell RNA-seq (scRNA-seq) are compared and contrasted using both published and original data. RESULTS We demonstrate the strength and weaknesses of these approaches leading to the inevitable conclusion that to maximize the utility of these approaches, it is important to match the experimental approach with the tissue of origin, cell type of interest, and the biological question. Often, a combined approach (eg, cell sorting and scRNA-seq or expression analysis using 2 separate approaches) is required. Finally, new tools for visualization and analysis of complex expression data, such as the gEAR platform (umgear.org), collate cell type-specific gene expression from the ear field and provide unprecedented access to both clinicians and researchers. LEVEL OF EVIDENCE N/A Laryngoscope, 131:S1-S16, 2021.
Collapse
Affiliation(s)
- Ronna Hertzano
- Department of Otorhinolaryngology Head and Neck Surgery University of Maryland School of Medicine 16 S Eutaw St. Suite 500 Baltimore Maryland 21201 U.S.A
- Institute for Genome Sciences, University of Maryland School of Medicine Baltimore Maryland U.S.A
- Department of Anatomy and Neurobiology University of Maryland School of Medicine Baltimore Maryland U.S.A
| | - Kathleen Gwilliam
- Department of Otorhinolaryngology Head and Neck Surgery University of Maryland School of Medicine 16 S Eutaw St. Suite 500 Baltimore Maryland 21201 U.S.A
| | - Kevin Rose
- Department of Otorhinolaryngology Head and Neck Surgery University of Maryland School of Medicine 16 S Eutaw St. Suite 500 Baltimore Maryland 21201 U.S.A
| | - Beatrice Milon
- Department of Otorhinolaryngology Head and Neck Surgery University of Maryland School of Medicine 16 S Eutaw St. Suite 500 Baltimore Maryland 21201 U.S.A
| | - Maggie S. Matern
- Department of Otorhinolaryngology Head and Neck Surgery University of Maryland School of Medicine 16 S Eutaw St. Suite 500 Baltimore Maryland 21201 U.S.A
| |
Collapse
|
42
|
Hearing Function: Identification of New Candidate Genes Further Explaining the Complexity of This Sensory Ability. Genes (Basel) 2021; 12:genes12081228. [PMID: 34440402 PMCID: PMC8394865 DOI: 10.3390/genes12081228] [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: 07/15/2021] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 11/16/2022] Open
Abstract
To date, the knowledge of the genetic determinants behind the modulation of hearing ability is relatively limited. To investigate this trait, we performed Genome-Wide Association Study (GWAS) meta-analysis using genotype and audiometric data (hearing thresholds at 0.25, 0.5, 1, 2, 4, and 8 kHz, and pure-tone averages of thresholds at low, medium, and high frequencies) collected in nine cohorts from Europe, South-Eastern USA, Caucasus, and Central Asia, for an overall number of ~9000 subjects. Three hundred seventy-five genes across all nine analyses were tagged by single nucleotide polymorphisms (SNPs) reaching a suggestive p-value (p < 10−5). Amongst these, 15 were successfully replicated using a gene-based approach in the independent Italian Salus in the Apulia cohort (n = 1774) at the nominal significance threshold (p < 0.05). In addition, the expression level of the replicated genes was assessed in published human and mouse inner ear datasets. Considering expression patterns in humans and mice, eleven genes were considered particularly promising candidates for the hearing function: BNIP3L, ELP5, MAP3K20, MATN2, MTMR7, MYO1E, PCNT, R3HDM1, SLC9A9, TGFB2, and YTHDC2. These findings represent a further contribution to our understanding of the genetic basis of hearing function and its related diseases.
Collapse
|
43
|
Mughal MN, Ye Q, Zhao L, Grevelding CG, Li Y, Di W, He X, Li X, Gasser RB, Hu M. First Evidence of Function for Schistosoma japonicumriok-1 and RIOK-1. Pathogens 2021; 10:862. [PMID: 34358012 PMCID: PMC8308690 DOI: 10.3390/pathogens10070862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 11/16/2022] Open
Abstract
Protein kinases are known as key molecules that regulate many biological processes in animals. The right open reading frame protein kinase (riok) genes are known to be essential regulators in model organisms such as the free-living nematode Caenorhabditis elegans. However, very little is known about their function in parasitic trematodes (flukes). In the present study, we characterized the riok-1 gene (Sj-riok-1) and the inferred protein (Sj-RIOK-1) in the parasitic blood fluke, Schistosoma japonicum. We gained a first insight into function of this gene/protein through double-stranded RNA interference (RNAi) and chemical inhibition. RNAi significantly reduced Sj-riok-1 transcription in both female and male worms compared with untreated control worms, and subtle morphological alterations were detected in the ovaries of female worms. Chemical knockdown of Sj-RIOK-1 with toyocamycin (a specific RIOK-1 inhibitor/probe) caused a substantial reduction in worm viability and a major accumulation of mature oocytes in the seminal receptacle (female worms), and of spermatozoa in the sperm vesicle (male worms). These phenotypic alterations indicate that the function of Sj-riok-1 is linked to developmental and/or reproductive processes in S. japonicum.
Collapse
Affiliation(s)
- Mudassar N. Mughal
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (M.N.M.); (Q.Y.); (L.Z.); (Y.L.); (X.H.); (X.L.)
- Biomedical Research Center Seltersberg, Institute of Parasitology, Justus Liebig University Giessen, D-35392 Giessen, Germany;
| | - Qing Ye
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (M.N.M.); (Q.Y.); (L.Z.); (Y.L.); (X.H.); (X.L.)
| | - Lu Zhao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (M.N.M.); (Q.Y.); (L.Z.); (Y.L.); (X.H.); (X.L.)
| | - Christoph G. Grevelding
- Biomedical Research Center Seltersberg, Institute of Parasitology, Justus Liebig University Giessen, D-35392 Giessen, Germany;
| | - Ying Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (M.N.M.); (Q.Y.); (L.Z.); (Y.L.); (X.H.); (X.L.)
| | - Wenda Di
- College of Animal Science and Technology, Guangxi University, Nanning 530005, China;
| | - Xin He
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (M.N.M.); (Q.Y.); (L.Z.); (Y.L.); (X.H.); (X.L.)
| | - Xuesong Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (M.N.M.); (Q.Y.); (L.Z.); (Y.L.); (X.H.); (X.L.)
| | - Robin B. Gasser
- Department of Veterinary Biosciences, Faculty of Veterinary and Agricultural Sciences, Melbourne Veterinary School, The University of Melbourne, Parkville, VIC 3010, Australia;
| | - Min Hu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (M.N.M.); (Q.Y.); (L.Z.); (Y.L.); (X.H.); (X.L.)
| |
Collapse
|
44
|
Jan TA, Eltawil Y, Ling AH, Chen L, Ellwanger DC, Heller S, Cheng AG. Spatiotemporal dynamics of inner ear sensory and non-sensory cells revealed by single-cell transcriptomics. Cell Rep 2021; 36:109358. [PMID: 34260939 PMCID: PMC8378666 DOI: 10.1016/j.celrep.2021.109358] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 12/25/2020] [Accepted: 06/17/2021] [Indexed: 11/28/2022] Open
Abstract
The utricle is a vestibular sensory organ that requires mechanosensitive hair cells to detect linear acceleration. In neonatal mice, new hair cells are derived from non-sensory supporting cells, yet cell type diversity and mechanisms of cell addition remain poorly characterized. Here, we perform computational analyses on single-cell transcriptomes to categorize cell types and resolve 14 individual sensory and non-sensory subtypes. Along the periphery of the sensory epithelium, we uncover distinct groups of transitional epithelial cells, marked by Islr, Cnmd, and Enpep expression. By reconstructing de novo trajectories and gene dynamics, we show that as the utricle expands, Islr+ transitional epithelial cells exhibit a dynamic and proliferative phase to generate new supporting cells, followed by coordinated differentiation into hair cells. Taken together, our study reveals a sequential and coordinated process by which non-sensory epithelial cells contribute to growth of the postnatal mouse sensory epithelium. The postnatal mouse utricle expands by more than 35% and doubles its number of hair cells during the first 8 days. Using single-cell transcriptomics, Jan et al. show that the surrounding transitional epithelial cells proliferate and contribute to the expansion of the sensory epithelium through a stepwise differentiation mechanism.
Collapse
Affiliation(s)
- Taha A Jan
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA; Department of Otolaryngology-Head and Neck Surgery, University of California San Francisco, San Francisco, CA 94115, USA
| | - Yasmin Eltawil
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Angela H Ling
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA; Department of Otolaryngology-Head and Neck Surgery, University of California San Francisco, San Francisco, CA 94115, USA
| | - Leon Chen
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Daniel C Ellwanger
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA; Genome Analysis Unit, Amgen Research, Amgen Inc., South San Francisco, CA 94080, USA
| | - Stefan Heller
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA.
| | - Alan G Cheng
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA.
| |
Collapse
|
45
|
Kohrman D, Borges BC, Cassinotti L, Ji L, Corfas G. Axon-glia interactions in the ascending auditory system. Dev Neurobiol 2021; 81:546-567. [PMID: 33561889 PMCID: PMC9004231 DOI: 10.1002/dneu.22813] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 11/25/2020] [Accepted: 02/05/2021] [Indexed: 11/09/2022]
Abstract
The auditory system detects and encodes sound information with high precision to provide a high-fidelity representation of the environment and communication. In mammals, detection occurs in the peripheral sensory organ (the cochlea) containing specialized mechanosensory cells (hair cells) that initiate the conversion of sound-generated vibrations into action potentials in the auditory nerve. Neural activity in the auditory nerve encodes information regarding the intensity and frequency of sound stimuli, which is transmitted to the auditory cortex through the ascending neural pathways. Glial cells are critical for precise control of neural conduction and synaptic transmission throughout the pathway, allowing for the precise detection of the timing, frequency, and intensity of sound signals, including the sub-millisecond temporal fidelity is necessary for tasks such as sound localization, and in humans, for processing complex sounds including speech and music. In this review, we focus on glia and glia-like cells that interact with hair cells and neurons in the ascending auditory pathway and contribute to the development, maintenance, and modulation of neural circuits and transmission in the auditory system. We also discuss the molecular mechanisms of these interactions, their impact on hearing and on auditory dysfunction associated with pathologies of each cell type.
Collapse
Affiliation(s)
- David Kohrman
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| | - Beatriz C. Borges
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| | - Luis Cassinotti
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| | - Lingchao Ji
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| | - Gabriel Corfas
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| |
Collapse
|
46
|
Ghilardi A, Diana A, Bacchetta R, Santo N, Ascagni M, Prosperi L, Del Giacco L. Inner Ear and Muscle Developmental Defects in Smpx-Deficient Zebrafish Embryos. Int J Mol Sci 2021; 22:ijms22126497. [PMID: 34204426 PMCID: PMC8235540 DOI: 10.3390/ijms22126497] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/09/2021] [Accepted: 06/12/2021] [Indexed: 12/20/2022] Open
Abstract
The last decade has witnessed the identification of several families affected by hereditary non-syndromic hearing loss (NSHL) caused by mutations in the SMPX gene and the loss of function has been suggested as the underlying mechanism. In the attempt to confirm this hypothesis we generated an Smpx-deficient zebrafish model, pointing out its crucial role in proper inner ear development. Indeed, a marked decrease in the number of kinocilia together with structural alterations of the stereocilia and the kinocilium itself in the hair cells of the inner ear were observed. We also report the impairment of the mechanotransduction by the hair cells, making SMPX a potential key player in the construction of the machinery necessary for sound detection. This wealth of evidence provides the first possible explanation for hearing loss in SMPX-mutated patients. Additionally, we observed a clear muscular phenotype consisting of the defective organization and functioning of muscle fibers, strongly suggesting a potential role for the protein in the development of muscle fibers. This piece of evidence highlights the need for more in-depth analyses in search for possible correlations between SMPX mutations and muscular disorders in humans, thus potentially turning this non-syndromic hearing loss-associated gene into the genetic cause of dysfunctions characterized by more than one symptom, making SMPX a novel syndromic gene.
Collapse
Affiliation(s)
- Anna Ghilardi
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy; (A.G.); (A.D.); (L.P.)
| | - Alberto Diana
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy; (A.G.); (A.D.); (L.P.)
| | - Renato Bacchetta
- Department of Environmental Science and Policy, Università degli Studi di Milano, 20133 Milan, Italy;
| | - Nadia Santo
- Unitech NOLIMITS, Università degli Studi di Milano, 20133 Milan, Italy; (N.S.); (M.A.)
| | - Miriam Ascagni
- Unitech NOLIMITS, Università degli Studi di Milano, 20133 Milan, Italy; (N.S.); (M.A.)
| | - Laura Prosperi
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy; (A.G.); (A.D.); (L.P.)
| | - Luca Del Giacco
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy; (A.G.); (A.D.); (L.P.)
- Correspondence:
| |
Collapse
|
47
|
Yu Y, Yang J, Luan F, Gu G, Zhao R, Wang Q, Dong Z, Tang J, Wang W, Sun J, Lv P, Zhang H, Wang C. Sensorineural Hearing Loss and Mitochondrial Apoptosis of Cochlear Spiral Ganglion Neurons in Fibroblast Growth Factor 13 Knockout Mice. Front Cell Neurosci 2021; 15:658586. [PMID: 34220452 PMCID: PMC8242186 DOI: 10.3389/fncel.2021.658586] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/26/2021] [Indexed: 12/17/2022] Open
Abstract
Deafness is known to occur in more than 400 syndromes and accounts for almost 30% of hereditary hearing loss. The molecular mechanisms underlying such syndromic deafness remain unclear. Furthermore, deafness has been a common feature in patients with three main syndromes, the BÖrjeson-Forssman-Lehmann syndrome, Wildervanck syndrome, and Congenital Generalized Hirsutism, all of which are characterized by loss-of-function mutations in the Fgf13 gene. Whether the pathogenesis of deafness in these syndromes is associated with the Fgf13 mutation is not known. To elucidate its role in auditory function, we generated a mouse line with conditional knockout of the Fgf13 gene in the inner ear (Fgf13 cKO). FGF13 is expressed predominantly in the organ of Corti, spiral ganglion neurons (SGNs), stria vascularis, and the supporting cells. Conditional knockout of the gene in the inner ear led to sensorineural deafness with low amplitude and increased latency of wave I in the auditory brainstem response test but had a normal distortion product otoacoustic emission threshold. Fgf13 deficiency resulted in decreased SGN density from the apical to the basal region without significant morphological changes and those in the number of hair cells. TUNEL and caspase-3 immunocytochemistry assays showed that apoptotic cell death mediated the loss of SGNs. Further detection of apoptotic factors through qRT-PCR suggested the activation of the mitochondrial apoptotic pathway in SGNs. Together, this study reveals a novel role for Fgf13 in auditory function, and indicates that the gene could be a potential candidate for understanding deafness. These findings may provide new perspectives on the molecular mechanisms and novel therapeutic targets for treatment deafness.
Collapse
Affiliation(s)
- Yulou Yu
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Jing Yang
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
| | - Feng Luan
- Department of Otolaryngology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Guoqiang Gu
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ran Zhao
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Qiong Wang
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Zishan Dong
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Junming Tang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| | - Wei Wang
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
| | - Jinpeng Sun
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ping Lv
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Hailin Zhang
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Chuan Wang
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
48
|
Bowles B, Ferrer A, Nishimura CJ, Pinto E Vairo F, Rey T, Leheup B, Sullivan J, Schoch K, Stong N, Agolini E, Cocciadiferro D, Williams A, Cummings A, Loddo S, Genovese S, Roadhouse C, McWalter K, Wentzensen IM, Li C, Babovic-Vuksanovic D, Lanpher BC, Dentici ML, Ankala A, Hamm JA, Dallapiccola B, Radio FC, Shashi V, Gérard B, Bloch-Zupan A, Smith RJ, Klee EW. TSPEAR variants are primarily associated with ectodermal dysplasia and tooth agenesis but not hearing loss: A novel cohort study. Am J Med Genet A 2021; 185:2417-2433. [PMID: 34042254 PMCID: PMC8361973 DOI: 10.1002/ajmg.a.62347] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 04/12/2021] [Accepted: 04/22/2021] [Indexed: 12/30/2022]
Abstract
Biallelic loss‐of‐function variants in the thrombospondin‐type laminin G domain and epilepsy‐associated repeats (TSPEAR) gene have recently been associated with ectodermal dysplasia and hearing loss. The first reports describing a TSPEAR disease association identified this gene is a cause of nonsyndromic hearing loss, but subsequent reports involving additional affected families have questioned this evidence and suggested a stronger association with ectodermal dysplasia. To clarify genotype–phenotype associations for TSPEAR variants, we characterized 13 individuals with biallelic TSPEAR variants. Individuals underwent either exome sequencing or panel‐based genetic testing. Nearly all of these newly reported individuals (11/13) have phenotypes that include tooth agenesis or ectodermal dysplasia, while three newly reported individuals have hearing loss. Of the individuals displaying hearing loss, all have additional variants in other hearing‐loss‐associated genes, specifically TMPRSS3, GJB2, and GJB6, that present competing candidates for their hearing loss phenotype. When presented alongside previous reports, the overall evidence supports the association of TSPEAR variants with ectodermal dysplasia and tooth agenesis features but creates significant doubt as to whether TSPEAR variants are a monogenic cause of hearing loss. Further functional evidence is needed to evaluate this phenotypic association.
Collapse
Affiliation(s)
- Bradley Bowles
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Alejandro Ferrer
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Carla J Nishimura
- Molecular Otolaryngology and Renal Research Laboratories, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Filippo Pinto E Vairo
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota, USA
| | - Tristan Rey
- Faculté de Chirurgie Dentaire, Université de Strasbourg, Strasbourg, France.,Laboratoires de Diagnostic génétique, Pôle de Biologie, Hôpitaux Universitaires de Strasbourg, Institut de Génétique Médicale d'Alsace, Strasbourg, France.,Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258, CNRS-UMR7104, Université de Strasbourg, Illkirch, France
| | - Bruno Leheup
- Département de Médecine Infantile, CHRU de Nancy, Nancy, France
| | - Jennifer Sullivan
- Department of Pediatrics, Duke University, Durham, North Carolina, USA
| | - Kelly Schoch
- Department of Pediatrics, Duke University, Durham, North Carolina, USA
| | - Nicholas Stong
- Institute for Genomic Medicine, Columbia University, New York, New York, USA.,Brystol Myers Squibb, New York, New York, USA
| | - Emanuele Agolini
- Laboratory of Medical Genetics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Dario Cocciadiferro
- Laboratory of Medical Genetics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Abigail Williams
- Department of Pediatrics, East Tennessee Children's Hospital, Knoxville, Tennessee, USA
| | - Alex Cummings
- Department of Pediatrics, East Tennessee Children's Hospital, Knoxville, Tennessee, USA.,University of Wisconsin Hospitals and Clinics, Madison, Wisconsin, USA
| | - Sara Loddo
- Laboratory of Medical Genetics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Silvia Genovese
- Laboratory of Medical Genetics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Chelsea Roadhouse
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | - Chumei Li
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Dusica Babovic-Vuksanovic
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota, USA
| | - Brendan C Lanpher
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota, USA
| | - Maria Lisa Dentici
- Genetics and Rare Diseases Research Division, Molecular Genetics and Functional Genomics Research Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Arun Ankala
- EGL Genetics LLC, Tucker, Georgia, USA.,Emory University School of Medicine, Atlanta, Georgia, USA
| | - J Austin Hamm
- Department of Pediatrics, East Tennessee Children's Hospital, Knoxville, Tennessee, USA
| | - Bruno Dallapiccola
- Genetics and Rare Diseases Research Division, Molecular Genetics and Functional Genomics Research Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Francesca Clementina Radio
- Genetics and Rare Diseases Research Division, Molecular Genetics and Functional Genomics Research Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Vandana Shashi
- Department of Pediatrics, Duke University, Durham, North Carolina, USA
| | - Benedicte Gérard
- Laboratoires de Diagnostic génétique, Pôle de Biologie, Hôpitaux Universitaires de Strasbourg, Institut de Génétique Médicale d'Alsace, Strasbourg, France
| | - Agnes Bloch-Zupan
- Faculté de Chirurgie Dentaire, Université de Strasbourg, Strasbourg, France.,Centre de référence des maladies rares orales et dentaires O-Rares, Filière Santé Maladies rares TETE COU, European Reference Network CRANIO, Pôle de Médecine et Chirurgie Bucco-dentaires, Hôpital Civil, Hôpitaux Universitaires de Strasbourg (HUS), Strasbourg, France.,Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258, CNRS-UMR7104, Université de Strasbourg, Illkirch, France
| | - Richard J Smith
- Molecular Otolaryngology and Renal Research Laboratories, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Eric W Klee
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
49
|
Issac J, Raveendran PS, Das AV. RFX1: a promising therapeutic arsenal against cancer. Cancer Cell Int 2021; 21:253. [PMID: 33964962 PMCID: PMC8106159 DOI: 10.1186/s12935-021-01952-6] [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: 12/19/2020] [Accepted: 04/26/2021] [Indexed: 02/08/2023] Open
Abstract
Regulatory factor X1 (RFX1) is an evolutionary conserved transcriptional factor that influences a wide range of cellular processes such as cell cycle, cell proliferation, differentiation, and apoptosis, by regulating a number of target genes that are involved in such processes. On a closer look, these target genes also play a key role in tumorigenesis and associated events. Such observations paved the way for further studies evaluating the role of RFX1 in cancer. These studies were indispensable due to the failure of conventional chemotherapeutic drugs to target key cellular hallmarks such as cancer stemness, cellular plasticity, enhanced drug efflux, de-regulated DNA repair machinery, and altered pathways evading apoptosis. In this review, we compile significant evidence for the tumor-suppressive activities of RFX1 while also analyzing its oncogenic potential in some cancers. RFX1 induction decreased cellular proliferation, modulated the immune system, induced apoptosis, reduced chemoresistance, and sensitized cancer stem cells for chemotherapy. Thus, our review discusses the pleiotropic function of RFX1 in multitudinous gene regulations, decisive protein–protein interactions, and also its role in regulating key cell signaling events in cancer. Elucidation of these regulatory mechanisms can be further utilized for RFX1 targeted therapy.
Collapse
Affiliation(s)
- Joby Issac
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thycaud.P.O, Thiruvananthapuram, 695014, Kerala, India
| | - Pooja S Raveendran
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thycaud.P.O, Thiruvananthapuram, 695014, Kerala, India
| | - Ani V Das
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thycaud.P.O, Thiruvananthapuram, 695014, Kerala, India.
| |
Collapse
|
50
|
Daly AZ, Dudley LA, Peel MT, Liebhaber SA, Parker SCJ, Camper SA. Multi-omic profiling of pituitary thyrotropic cells and progenitors. BMC Biol 2021; 19:76. [PMID: 33858413 PMCID: PMC8051135 DOI: 10.1186/s12915-021-01009-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 03/23/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The pituitary gland is a neuroendocrine organ containing diverse cell types specialized in secreting hormones that regulate physiology. Pituitary thyrotropes produce thyroid-stimulating hormone (TSH), a critical factor for growth and maintenance of metabolism. The transcription factors POU1F1 and GATA2 have been implicated in thyrotrope fate, but the transcriptomic and epigenomic landscapes of these neuroendocrine cells have not been characterized. The goal of this work was to discover transcriptional regulatory elements that drive thyrotrope fate. RESULTS We identified the transcription factors and epigenomic changes in chromatin that are associated with differentiation of POU1F1-expressing progenitors into thyrotropes using cell lines that represent an undifferentiated Pou1f1 lineage progenitor (GHF-T1) and a committed thyrotrope line that produces TSH (TαT1). We compared RNA-seq, ATAC-seq, histone modification (H3K27Ac, H3K4Me1, and H3K27Me3), and POU1F1 binding in these cell lines. POU1F1 binding sites are commonly associated with bZIP transcription factor consensus binding sites in GHF-T1 cells and Helix-Turn-Helix (HTH) or basic Helix-Loop-Helix (bHLH) factors in TαT1 cells, suggesting that these classes of transcription factors may recruit or cooperate with POU1F1 binding at unique sites. We validated enhancer function of novel elements we mapped near Cga, Pitx1, Gata2, and Tshb by transfection in TαT1 cells. Finally, we confirmed that an enhancer element near Tshb can drive expression in thyrotropes of transgenic mice, and we demonstrate that GATA2 enhances Tshb expression through this element. CONCLUSION These results extend the ENCODE multi-omic profiling approach to the pituitary gland, which should be valuable for understanding pituitary development and disease pathogenesis.
Collapse
Affiliation(s)
- Alexandre Z Daly
- Department Human Genetics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Lindsey A Dudley
- Department Human Genetics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Michael T Peel
- Department Genetics, University of Pennsylvania Perelman School of Medicine, Ann Arbor, MI, 48109, USA.,Incyte, Wilmington, DE, 19803, USA
| | - Stephen A Liebhaber
- Department Human Genetics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.,Department Genetics, University of Pennsylvania Perelman School of Medicine, Ann Arbor, MI, 48109, USA
| | - Stephen C J Parker
- Department Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Sally A Camper
- Department Human Genetics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.
| |
Collapse
|