1
|
Meng W, Kong L, Abulizi A, Cong J, Sun Z, Chang Y. Sex determination factor, a novel male-linked gene in the sea cucumber Apostichopus japonicus: Molecular characterization, expression patterns and effects of gene knockdown. Comp Biochem Physiol B Biochem Mol Biol 2025; 277:111071. [PMID: 39778676 DOI: 10.1016/j.cbpb.2025.111071] [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: 10/15/2024] [Revised: 01/05/2025] [Accepted: 01/05/2025] [Indexed: 01/11/2025]
Abstract
Apostichopus japonicus is a highly significant marine aquaculture species. Research findings have indicated that male sea cucumbers demonstrate a more rapid growth rate compared to females, underscoring the potential advantages of establishing an all-male population. In this study, we identified a specific protein-coding gene (ORFan) within a 4565 bp male fragment and named it sex determination factor (sdf). The sdf transcript exhibited ubiquitous expression in various adult male tissues, along with dynamic expression patterns in the testis across different developmental stages. Notably, knockdown of the sdf gene through immersion of embryos in its specific vivo-morpholino oligomers (vivo-MO) resulted in significant changes in the expression levels of several sex-related genes including piwi1, vasa, foxl2, and DNMT3. Additionally, a transcriptomic analysis showed that sdf knockdown resulted in significant alterations in multiple biological processes encompassing various sex-related gene ontology terms such as male gonad development, ovarian follicle development, and steroidogenesis. These results provide a molecular foundation for comprehending ORFans in sea cucumbers while offering a valuable method for gene knockdown studies in echinoderms.
Collapse
Affiliation(s)
- Weihan Meng
- Key Laboratory of Mariculture& Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian 116023, China
| | - Lingnan Kong
- Key Laboratory of Mariculture& Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian 116023, China
| | - Abudula Abulizi
- Key Laboratory of Mariculture& Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian 116023, China
| | - Jingjing Cong
- Key Laboratory of Mariculture& Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian 116023, China; School of Life Science, Liaoning Normal University, Dalian 116029, China
| | - Zhihui Sun
- Key Laboratory of Mariculture& Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian 116023, China.
| | - Yaqing Chang
- Key Laboratory of Mariculture& Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian 116023, China
| |
Collapse
|
2
|
Kim KS, Koo HY, Bok J. Alternative splicing in shaping the molecular landscape of the cochlea. Front Cell Dev Biol 2023; 11:1143428. [PMID: 36936679 PMCID: PMC10018040 DOI: 10.3389/fcell.2023.1143428] [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: 01/13/2023] [Accepted: 02/16/2023] [Indexed: 03/06/2023] Open
Abstract
The cochlea is a complex organ comprising diverse cell types with highly specialized morphology and function. Until now, the molecular underpinnings of its specializations have mostly been studied from a transcriptional perspective, but accumulating evidence points to post-transcriptional regulation as a major source of molecular diversity. Alternative splicing is one of the most prevalent and well-characterized post-transcriptional regulatory mechanisms. Many molecules important for hearing, such as cadherin 23 or harmonin, undergo alternative splicing to produce functionally distinct isoforms. Some isoforms are expressed specifically in the cochlea, while some show differential expression across the various cochlear cell types and anatomical regions. Clinical phenotypes that arise from mutations affecting specific splice variants testify to the functional relevance of these isoforms. All these clues point to an essential role for alternative splicing in shaping the unique molecular landscape of the cochlea. Although the regulatory mechanisms controlling alternative splicing in the cochlea are poorly characterized, there are animal models with defective splicing regulators that demonstrate the importance of RNA-binding proteins in maintaining cochlear function and cell survival. Recent technological breakthroughs offer exciting prospects for overcoming some of the long-standing hurdles that have complicated the analysis of alternative splicing in the cochlea. Efforts toward this end will help clarify how the remarkable diversity of the cochlear transcriptome is both established and maintained.
Collapse
Affiliation(s)
- Kwan Soo Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hei Yeun Koo
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jinwoong Bok
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of Korea
- *Correspondence: Jinwoong Bok,
| |
Collapse
|
3
|
A Predictive Model of Bilateral Sensorineural Hearing Loss in Meniere Disease Using Clinical Data. Ear Hear 2021; 43:1079-1085. [PMID: 34799494 DOI: 10.1097/aud.0000000000001169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
OBJECTIVES Meniere disease (MD) is defined by a clinical syndrome of recurrent attacks of spontaneous vertigo associated with tinnitus, aural fullness, and sensorineural hearing loss (SNHL). Most patients have unilateral SNHL, but some of them will develop contralateral SNHL during the course of the disease. Several studies have reported a frequency of 2 to 73% SNHL in the second ear, according to the duration of disease and the period of follow-up. We hypothesize that unilateral and bilateral MD are different conditions, the first would initially involve the apical turn of the cochlea, while bilateral MD would affect the entire length of the cochlea. The aim of the study is to search for clinical predictors of bilateral SNHL in MD to build a predictive model of bilateral involvement. DESIGN A retrospective, longitudinal study including two cohorts with a total of 400 patients with definite MD was carried out. The inception cohort consisted of 150 patients with MD and the validation cohort included 250 cases. All of the cases were diagnosed of unilateral MD according to their hearing loss thresholds. The following variables were assessed as predictors of bilateral SNHL for the two cohorts: sex, age of onset, familiar history of MD, migraine and high-frequency hearing loss (HFHL, defined if hearing threshold >20 dB in two or more consecutive frequencies from 2 to 8 KHz). A descriptive analysis was carried out according to the presence of HFHL in the first audiogram for the main variables. By using multiple logistic regression, we built-up several predictive models for the inception cohort and validated it with the replication cohort and merged dataset. RESULTS Twenty-three (19.3%) and 78 (41%) of patients with HFHL developed contralateral SNHL during the follow-up, in the inception and validation cohorts, respectively. In the inception cohort, the best predictive model included HFHL in the first audiogram (OR = 6.985, p = 0.063) and the absence of migraine (OR = 0.215, p = 0.144) as clinical predictors for bilateral SNHL [area under the curve (AUC) = 0.641, p = 0.002]. The model was validated in the second cohort (AUC = 0.621, p < 0.001). Finally, we merged both datasets to improve the precision of the model including HFHL in the first audiogram (OR = 3.168, p = 0.001), migraine (OR = 0.482, p = 0.036) and age of onset >35 years old (OR = 2.422, p = 0.006) as clinical predictors (AUC = 0.639, p < 0.001). CONCLUSIONS A predictive model including the age of onset, HFHL in the first audiogram and migraine can help to assess the risk of bilateral SNHL in MD. This model may have significant implications for clinical management of patients with MD.
Collapse
|
4
|
Koo H, Hwang JY, Jung S, Park H, Bok J, Park JW. Position Specific Alternative Splicing and Gene Expression Profiles Along the Tonotopic Axis of Chick Cochlea. Front Mol Biosci 2021; 8:726976. [PMID: 34568429 PMCID: PMC8456117 DOI: 10.3389/fmolb.2021.726976] [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: 06/17/2021] [Accepted: 08/25/2021] [Indexed: 11/17/2022] Open
Abstract
Alternative splicing (AS) refers to the production of multiple mRNA isoforms from a single gene due to alternative selection of exons or splice sites during pre-mRNA splicing. It is a primary mechanism of gene regulation in higher eukaryotes and significantly expands the functional complexity of eukaryotic organisms, contributing to animal development and disease. Recent studies have shown that AS also influences functional diversity by affecting the transcriptomic and proteomic profiles in a position-dependent manner in a single organ. The peripheral hearing organ, the cochlea, is organized to detect sounds at different frequencies depending on its location along the longitudinal axis. This unique functional configuration, the tonotopy, is known to be facilitated by differential gene expression along the cochlear duct. We profiled transcriptome-wide gene expression and AS changes that occur within the different positions of chick cochlea. These analyses revealed distinct gene expression profiles and AS, including a splicing program that is unique to tonotopy. Changes in the expression of splicing factors PTBP3, ESRP1, and ESRP2 were demonstrated to contribute to position-specific AS. RNA-binding motif enrichment analysis near alternatively spliced exons provided further insight into the combinatorial regulation of AS at different positions by different RNA-binding proteins. These data, along with gene ontology (GO) analysis, represent a comprehensive analysis of the dynamic regulation of AS at different positions in chick cochlea.
Collapse
Affiliation(s)
- Heiyeun Koo
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea
- BK21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Jae Yeon Hwang
- Department of Computer Science and Engineering, University of Louisville, Louisville, KY, United States
| | - Sungbo Jung
- Department of Computer Science and Engineering, University of Louisville, Louisville, KY, United States
| | - Hyeyoung Park
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea
| | - Jinwoong Bok
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea
- BK21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, South Korea
| | - Juw Won Park
- Department of Computer Science and Engineering, University of Louisville, Louisville, KY, United States
- KY INBRE Bioinformatics Core, University of Louisville, Louisville, KY, United States
| |
Collapse
|
5
|
Bertram S, Roll L, Reinhard J, Groß K, Dazert S, Faissner A, Volkenstein S. Pleiotrophin increases neurite length and number of spiral ganglion neurons in vitro. Exp Brain Res 2019; 237:2983-2993. [PMID: 31515588 DOI: 10.1007/s00221-019-05644-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 09/03/2019] [Indexed: 11/28/2022]
Abstract
Acoustic trauma, aging, genetic defects or ototoxic drugs are causes for sensorineural hearing loss involving sensory hair cell death and secondary degeneration of spiral ganglion neurons. Auditory implants are the only available therapy for severe to profound sensorineural hearing loss when hearing aids do not provide a sufficient speech discrimination anymore. Neurotrophic factors represent potential therapeutic candidates to improve the performance of cochlear implants (CIs) by the support of spiral ganglion neurons (SGNs). Here, we investigated the effect of pleiotrophin (PTN), a well-described neurotrophic factor for different types of neurons that is expressed in the postnatal mouse cochlea. PTN knockout mice exhibit severe deficits in auditory brainstem responses, which indicates the importance of PTN in inner ear development and function and makes it a promising candidate to support SGNs. Using organotypic explants and dissociated SGN cultures, we investigated the influence of PTN on the number of neurons, neurite number and neurite length. PTN significantly increased the number and neurite length of dissociated SGNs. We further verified the expression of important PTN-associated receptors in the SG. mRNA of anaplastic lymphoma kinase, αv integrin, β3 integrin, receptor protein tyrosine phosphatase β/ζ, neuroglycan C, low-density lipoprotein receptor-related protein 1 and syndecan 3 was detected in the inner ear. These results suggest that PTN may be a novel candidate to improve sensorineural hearing loss treatment in the future.
Collapse
Affiliation(s)
- Sebastian Bertram
- Department of Otorhinolaryngology, Head and Neck Surgery, St. Elisabeth-Hospital, Ruhr-University Bochum, Bleichstr. 15, 44787, Bochum, Germany
| | - Lars Roll
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitätsstr. 150, 44801, Bochum, Germany
| | - Jacqueline Reinhard
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitätsstr. 150, 44801, Bochum, Germany
| | - Katharina Groß
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitätsstr. 150, 44801, Bochum, Germany
| | - Stefan Dazert
- Department of Otorhinolaryngology, Head and Neck Surgery, St. Elisabeth-Hospital, Ruhr-University Bochum, Bleichstr. 15, 44787, Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitätsstr. 150, 44801, Bochum, Germany
| | - Stefan Volkenstein
- Department of Otorhinolaryngology, Head and Neck Surgery, St. Elisabeth-Hospital, Ruhr-University Bochum, Bleichstr. 15, 44787, Bochum, Germany.
| |
Collapse
|
6
|
Requena T, Gallego-Martinez A, Lopez-Escamez JA. Bioinformatic Integration of Molecular Networks and Major Pathways Involved in Mice Cochlear and Vestibular Supporting Cells. Front Mol Neurosci 2018; 11:108. [PMID: 29674954 PMCID: PMC5895758 DOI: 10.3389/fnmol.2018.00108] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 03/19/2018] [Indexed: 12/18/2022] Open
Abstract
Background: Cochlear and vestibular epithelial non-hair cells (ENHCs) are the supporting elements of the cellular architecture in the organ of Corti and the vestibular neuroepithelium in the inner ear. Intercellular and cell-extracellular matrix interactions are essential to prevent an abnormal ion redistribution leading to hearing and vestibular loss. The aim of this study is to define the main pathways and molecular networks in the mouse ENHCs. Methods: We retrieved microarray and RNA-seq datasets from mouse epithelial sensory and non-sensory cells from gEAR portal (http://umgear.org/index.html) and obtained gene expression fold-change between ENHCs and non-epithelial cells (NECs) against HCs for each gene. Differentially expressed genes (DEG) with a log2 fold change between 1 and -1 were discarded. The remaining genes were selected to search for interactions using Ingenuity Pathway Analysis and STRING platform. Specific molecular networks for ENHCs in the cochlea and the vestibular organs were generated and significant pathways were identified. Results: Between 1723 and 1559 DEG were found in the mouse cochlear and vestibular tissues, respectively. Six main pathways showed enrichment in the supporting cells in both tissues: (1) "Inhibition of Matrix Metalloproteases"; (2) "Calcium Transport I"; (3) "Calcium Signaling"; (4) "Leukocyte Extravasation Signaling"; (5) "Signaling by Rho Family GTPases"; and (6) "Axonal Guidance Si". In the mouse cochlea, ENHCs showed a significant enrichment in 18 pathways highlighting "axonal guidance signaling (AGS)" (p = 4.37 × 10-8) and "RhoGDI Signaling" (p = 3.31 × 10-8). In the vestibular dataset, there were 20 enriched pathways in ENHCs, the most significant being "Leukocyte Extravasation Signaling" (p = 8.71 × 10-6), "Signaling by Rho Family GTPases" (p = 1.20 × 10-5) and "Calcium Signaling" (p = 1.20 × 10-5). Among the top ranked networks, the most biologically significant network contained the "auditory and vestibular system development and function" terms. We also found 108 genes showing tonotopic gene expression in the cochlear ENHCs. Conclusions: We have predicted the main pathways and molecular networks for ENHCs in the organ of Corti and vestibular neuroepithelium. These pathways will facilitate the design of molecular maps to select novel candidate genes for hearing or vestibular loss to conduct functional studies.
Collapse
Affiliation(s)
- Teresa Requena
- Otology & Neurotology Group CTS495, Department of Genomic Medicine-Centro de Genómica e Investigación Oncológica-Pfizer/Universidad de Granada/Junta de Andalucía (GENYO), Granada, Spain
| | - Alvaro Gallego-Martinez
- Otology & Neurotology Group CTS495, Department of Genomic Medicine-Centro de Genómica e Investigación Oncológica-Pfizer/Universidad de Granada/Junta de Andalucía (GENYO), Granada, Spain
| | - Jose A Lopez-Escamez
- Otology & Neurotology Group CTS495, Department of Genomic Medicine-Centro de Genómica e Investigación Oncológica-Pfizer/Universidad de Granada/Junta de Andalucía (GENYO), Granada, Spain.,Department of Otolaryngology, Instituto de Investigación Biosanitaria, ibs.GRANADA, Hospital Virgen de las Nieves, Universidad de Granada, Granada, Spain.,Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| |
Collapse
|
7
|
Du J, Wang X, Zhang X, Zhang X, Jiang H. DNER modulates the length, polarity and synaptogenesis of spiral ganglion neurons via the Notch signaling pathway. Mol Med Rep 2017; 17:2357-2365. [PMID: 29207144 PMCID: PMC5783477 DOI: 10.3892/mmr.2017.8115] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 02/16/2017] [Indexed: 02/05/2023] Open
Abstract
The Delta/Notch-like epidermal growth factor-related receptor (DNER) serves an important role in the developing central nervous system. However, the actions of DNER in the development of the spiral ganglion in the inner ear have yet to be elucidated. Wild-type C57BL/6 mice were housed and time-mated for use in the present study. Primary neuronal cultures were prepared using spiral ganglion progenitors isolated from the modiolus of postnatal day 1 (P1) mice. DNER recombinant lentiviral vectors were constructed and transfected into the cultured primary neurons. The relative proportion of differentiated neurons and the length of their neurites were evaluated using microscopy. The results of the present study demonstrated that DNER was expressed in spiral ganglion neurons (SGNs) that exhibited significant polarity in the early differentiation stages; DNER expression gradually decreased until the polarity was lost on week 35. The in vitro expression of DNER was revealed to be similar to that in vivo. When DNER expression was silenced using RNA interference, the polarity of the differentiated neurons was altered and they exhibited significantly reduced dendritic length. In addition, the proportion of bipolar neurons was decreased compared with the control group. Furthermore, the expression of α-synuclein and the GluR2/3 subunits of the α-amin-o-3-hydroxy-5-methyl-4-isoxazolepropionic acid glutamate receptor were also reduced in cultured neurons in which DNER was silenced. Notch1 was co-expressed with DNER in SGNs isolated from P1 mice. The indirect Notch inhibitor N-[N-(3,5-Difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester also affected the polarity and the formation of protrusions, and reduced the expression of DNER and glial fibrillary acidic protein in SGNs. In conclusion, the present study demonstrated that DNER was expressed in SGNs and appeared to be involved in the mechanisms underlying neuronal polarity and neuritogenesis, via a Notch-dependent signaling pathway.
Collapse
Affiliation(s)
- Jintao Du
- Department of Otorhinolaryngology Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xianren Wang
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Xiaobo Zhang
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Xuemei Zhang
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Hongyan Jiang
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
8
|
Keeley PW, Reese BE. DNER and NFIA are expressed by developing and mature AII amacrine cells in the mouse retina. J Comp Neurol 2017; 526:467-479. [PMID: 29071714 DOI: 10.1002/cne.24345] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 09/27/2017] [Accepted: 10/17/2017] [Indexed: 02/02/2023]
Abstract
The present study has taken advantage of publicly available cell type specific mRNA expression databases in order to identify potential genes participating in the development of retinal AII amacrine cells. We profile two such genes, Delta/Notch-like EGF repeat containing (Dner) and nuclear factor I/A (Nfia), that are each heavily expressed in AII amacrine cells in the mature mouse retina, and which conjointly identify this retinal cell population in its entirety when using antibodies to DNER and NFIA. DNER is present on the plasma membrane, while NFIA is confined to the nucleus, consistent with known functions of each of these two proteins. DNER also identifies some other subsets of retinal ganglion and amacrine cell types, along with horizontal cells, while NFIA identifies a subset of bipolar cells as well as Muller glia and astrocytes. During early postnatal development, NFIA labels astrocytes on the day of birth, AII amacrine cells at postnatal (P) day 5, and Muller glia by P10, when horizontal cells also transiently exhibit NFIA immunofluorescence. DNER, by contrast, is present in ganglion and amacrine cells on P1, also labeling the horizontal cells by P10. Developing AII amacrine cells exhibit accumulating DNER labeling at the dendritic stalk, labeling that becomes progressively conspicuous by P10, as it is in maturity. This developmental time course is consistent with a prospective role for each gene in the differentiation of AII amacrine cells.
Collapse
Affiliation(s)
- Patrick W Keeley
- Neuroscience Research Institute, University of California at Santa Barbara, Santa Barbara, California
| | - Benjamin E Reese
- Neuroscience Research Institute, University of California at Santa Barbara, Santa Barbara, California.,Department of Psychological & Brain Sciences, University of California at Santa Barbara, Santa Barbara, California
| |
Collapse
|
9
|
Kural KC, Tandon N, Skoblov M, Kel-Margoulis OV, Baranova AV. Pathways of aging: comparative analysis of gene signatures in replicative senescence and stress induced premature senescence. BMC Genomics 2016; 17:1030. [PMID: 28105936 PMCID: PMC5249001 DOI: 10.1186/s12864-016-3352-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background In culturing normal diploid cells, senescence may either happen naturally, in the form of replicative senescence, or it may be a consequence of external challenges such as oxidative stress. Here we present a comparative analysis aimed at reconstruction of molecular cascades specific for replicative (RS) and stressinduced senescence (SIPS) in human fibroblasts. Results An involvement of caspase-3/keratin-18 pathway and serine/threonine kinase Aurora A/ MDM2 pathway was shared between RS and SIPS. Moreover, stromelysin/MMP3 and N-acetylglucosaminyltransferase enzyme MGAT1, which initiates the synthesis of hybrid and complex Nglycans, were identified as key orchestrating components in RS and SIPS, respectively. In RS only, Aurora-B driven cell cycle signaling was deregulated in concert with the suppression of anabolic branches of the fatty acids and estrogen metabolism. In SIPS, Aurora-B signaling is deprioritized, and the synthetic branches of cholesterol metabolism are upregulated, rather than downregulated. Moreover, in SIPS, proteasome/ubiquitin ligase pathways of protein degradation dominate the regulatory landscape. This picture indicates that SIPS proceeds in cells that are actively fighting stress which facilitates premature senescence while failing to completely activate the orderly program of RS. The promoters of genes differentially expressed in either RS or SIPS are unusually enriched by the binding sites for homeobox family proteins, with particular emphasis on HMX1, IRX2, HDX and HOXC13. Additionally, we identified Iroquois Homeobox 2 (IRX2) as a master regulator for the secretion of SPP1-encoded osteopontin, a stromal driver for tumor growth that is overexpressed by both RS and SIPS fibroblasts. The latter supports the hypothesis that senescence-specific de-repression of SPP1 aids in SIPS-dependent stromal activation. Conclusions Reanalysis of previously published experimental data is cost-effective approach for extraction of additional insignts into the functioning of biological systems. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-3352-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kamil C Kural
- School of Systems Biology, George Mason University, Manassas, VA, 20110, USA
| | | | - Mikhail Skoblov
- Research Centre for Medical Genetics, Moscow, Russia.,Moscow Institute of Physics and Technology, Dolgoprudny, 141700, Russia
| | | | - Ancha V Baranova
- School of Systems Biology, George Mason University, Manassas, VA, 20110, USA. .,Research Centre for Medical Genetics, Moscow, Russia. .,Moscow Institute of Physics and Technology, Dolgoprudny, 141700, Russia.
| |
Collapse
|
10
|
Pyott SJ, Duncan RK. BK Channels in the Vertebrate Inner Ear. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 128:369-99. [PMID: 27238269 DOI: 10.1016/bs.irn.2016.03.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The perception of complex acoustic stimuli begins with the deconstruction of sound into its frequency components. This spectral processing occurs first and foremost in the inner ear. In vertebrates, two very different strategies of frequency analysis have evolved. In nonmammalian vertebrates, the sensory hair cells of the inner ear are intrinsically electrically tuned to a narrow band of acoustic frequencies. This electrical tuning relies on the interplay between BK channels and voltage-gated calcium channels. Systematic variations in BK channel density and kinetics establish a gradient in electrical resonance that enables the coding of a broad range of acoustic frequencies. In contrast, mammalian hair cells are extrinsically tuned by mechanical properties of the cochlear duct. Even so, mammalian hair cells also express BK channels. These BK channels play critical roles in various aspects of mammalian auditory signaling, from developmental maturation to protection against acoustic trauma. This review summarizes the anatomical localization, biophysical properties, and functional contributions of BK channels in vertebrate inner ears. Areas of future research, based on an updated understanding of the biology of both BK channels and the inner ear, are also highlighted. Investigation of BK channels in the inner ear continues to provide fertile research grounds for examining both BK channel biophysics and the molecular mechanisms underlying signal processing in the auditory periphery.
Collapse
Affiliation(s)
- S J Pyott
- University Medical Center Groningen, Groningen, The Netherlands.
| | - R K Duncan
- Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
11
|
Kwiatkowska M, Reinhard J, Roll L, Kraft N, Dazert S, Faissner A, Volkenstein S. The expression pattern and inhibitory influence of Tenascin-C on the growth of spiral ganglion neurons suggest a regulatory role as boundary formation molecule in the postnatal mouse inner ear. Neuroscience 2016; 319:46-58. [PMID: 26812032 DOI: 10.1016/j.neuroscience.2016.01.039] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 12/08/2015] [Accepted: 01/15/2016] [Indexed: 02/02/2023]
Abstract
Sensorineural hearing loss, as a consequence of acoustic trauma, aging, genetic defects or ototoxic drugs, is highly associated with irreversible damage of cochlear hair cells (HCs) and secondary degeneration of spiral ganglion (SG) cells. Cochlear implants (CIs), which bypass the lost HC function by direct electrical stimulation of the remaining auditory neurons, offer an effective therapy option. Several studies imply that components of the extracellular matrix (ECM) have a great impact on the adhesion and growth of spiral ganglion neurons (SGNs) during development. Based on these findings, ECM proteins might act as bioactive CI substrates to optimize the electrode-nerve interface and to improve efficacy of these implants. In the present study, we focused on the ECM glycoproteins Tenascin-C (TN-C), Laminin (LN), and Fibronectin (FN), which show a prominent expression along the growth route of SGNs and the niche around HCs during murine postnatal development in vivo. We compared their influence on adhesion, neurite length, and neurite number of SGNs in vitro. Moreover, we studied the expression of the chondroitin sulfate proteoglycan (CSPG) dermatan sulfate-dependent proteoglycan-1 (DSD-1-PG), an interaction partner of TN-C. In sum, our in vitro data suggest that TN-C acts as an anti-adhesive and inhibitory factor for the growth of SGNs. The DSD-1 carbohydrate epitope is specifically localized to HC stereocilia and SG fibers. Interestingly, TN-C and the DSD-1-PG exhibit a mutually exclusive expression pattern, with the exception of a very restricted region beneath the habenula perforata, where SG neurites grow through the basilar membrane (BM) toward the HCs. The complementary expression of TN-C, LN, FN, and the DSD-1 epitope suggests that TN-C may act as an important boundary formation molecule in the developing postnatal mouse inner ear, which makes it a promising candidate to regulate neurite outgrowth in the light of CIs.
Collapse
Affiliation(s)
- M Kwiatkowska
- Department of Otorhinolaryngology, Head & Neck Surgery, Ruhr-University Bochum, St. Elisabeth-Hospital, Bleichstrasse 15, 44787 Bochum, Germany
| | - J Reinhard
- Department of Cell Morphology & Molecular Neurobiology, Ruhr-University Bochum, Faculty of Biology & Biotechnology, Universitätsstrasse 150, 44801 Bochum, Germany
| | - L Roll
- Department of Cell Morphology & Molecular Neurobiology, Ruhr-University Bochum, Faculty of Biology & Biotechnology, Universitätsstrasse 150, 44801 Bochum, Germany
| | - N Kraft
- Department of Otorhinolaryngology, Head & Neck Surgery, Ruhr-University Bochum, St. Elisabeth-Hospital, Bleichstrasse 15, 44787 Bochum, Germany
| | - S Dazert
- Department of Otorhinolaryngology, Head & Neck Surgery, Ruhr-University Bochum, St. Elisabeth-Hospital, Bleichstrasse 15, 44787 Bochum, Germany
| | - A Faissner
- Department of Cell Morphology & Molecular Neurobiology, Ruhr-University Bochum, Faculty of Biology & Biotechnology, Universitätsstrasse 150, 44801 Bochum, Germany
| | - S Volkenstein
- Department of Otorhinolaryngology, Head & Neck Surgery, Ruhr-University Bochum, St. Elisabeth-Hospital, Bleichstrasse 15, 44787 Bochum, Germany.
| |
Collapse
|
12
|
Lu X, Sipe CW. Developmental regulation of planar cell polarity and hair-bundle morphogenesis in auditory hair cells: lessons from human and mouse genetics. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2015; 5:85-101. [PMID: 26265594 DOI: 10.1002/wdev.202] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 06/12/2015] [Accepted: 06/29/2015] [Indexed: 12/11/2022]
Abstract
Hearing loss is the most common and costly sensory defect in humans and genetic causes underlie a significant proportion of affected individuals. In mammals, sound is detected by hair cells (HCs) housed in the cochlea of the inner ear, whose function depends on a highly specialized mechanotransduction organelle, the hair bundle. Understanding the factors that regulate the development and functional maturation of the hair bundle is crucial for understanding the pathophysiology of human deafness. Genetic analysis of deafness genes in animal models, together with complementary forward genetic screens and conditional knock-out mutations in essential genes, have provided great insights into the molecular machinery underpinning hair-bundle development and function. In this review, we highlight recent advances in our understanding of hair-bundle morphogenesis, with an emphasis on the molecular pathways governing hair-bundle polarity and orientation. We next discuss the proteins and structural elements important for hair-cell mechanotransduction as well as hair-bundle cohesion and maintenance. In addition, developmental signals thought to regulate tonotopic features of HCs are introduced. Finally, novel approaches that complement classic genetics for studying the molecular etiology of human deafness are presented. WIREs Dev Biol 2016, 5:85-101. doi: 10.1002/wdev.202 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Xiaowei Lu
- Department of Cell Biology, University of Virginia, Charlottesville, VA, USA
| | - Conor W Sipe
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
13
|
Expression and Misexpression of the miR-183 Family in the Developing Hearing Organ of the Chicken. PLoS One 2015; 10:e0132796. [PMID: 26176784 PMCID: PMC4503353 DOI: 10.1371/journal.pone.0132796] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 06/19/2015] [Indexed: 12/31/2022] Open
Abstract
The miR-183 family consists of 3 related microRNAs (miR-183, miR-96, miR-182) that are required to complete maturation of primary sensory cells in the mammalian inner ear. Because the level of these microRNAs is not uniform across hair cell subtypes in the murine cochlea, the question arises as to whether hair cell phenotypes are influenced by microRNA expression levels. To address this, we used the chicken embryo to study expression and misexpression of this gene family. By in situ hybridization, expression of all 3 microRNAs is robust in immature hair cells of both auditory and vestibular organs and is present in the statoacoustic ganglion. The auditory organ, called the basilar papilla, shows a weak radial gradient (highest on the neural side) in prosensory cells near the base on embryonic day 7. About nine days later, the basilar papilla also displays a longitudinal gradient (highest in apical hair cells) for the 3 microRNAs. Tol2-mediated gene delivery was used to ask whether cell phenotypes are malleable when the prosensory epithelium was forced to overexpress the miR-183 family. The expression plasmid included EGFP as a reporter located upstream of an intron carrying the microRNA genes. The vectors were electroporated into the otic cup/vesicle, resulting in strong co-expression of EGFP and the miR-183 family that persisted for at least 2 weeks. This manipulation did not generate ectopic hair cells in non-sensory territories of the cochlear duct, although within the basilar papilla, hair cells were over-represented relative to supporting cells. There was no evidence for a change in hair cell phenotypes, such as short-to-tall, or basal-to-apical hair cell features. Therefore, while increasing expression of the miR-183 family was sufficient to influence cell lineage decisions, it did not redirect the differentiation of hair cells towards alternative radial or longitudinal phenotypes.
Collapse
|
14
|
Conserved role of Sonic Hedgehog in tonotopic organization of the avian basilar papilla and mammalian cochlea. Proc Natl Acad Sci U S A 2015; 112:3746-51. [PMID: 25775517 DOI: 10.1073/pnas.1417856112] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Sound frequency discrimination begins at the organ of Corti in mammals and the basilar papilla in birds. Both of these hearing organs are tonotopically organized such that sensory hair cells at the basal (proximal) end respond to high frequency sound, whereas their counterparts at the apex (distal) respond to low frequencies. Sonic hedgehog (Shh) secreted by the developing notochord and floor plate is required for cochlear formation in both species. In mice, the apical region of the developing cochlea, closer to the ventral midline source of Shh, requires higher levels of Shh signaling than the basal cochlea farther away from the midline. Here, gain-of-function experiments using Shh-soaked beads in ovo or a mouse model expressing constitutively activated Smoothened (transducer of Shh signaling) show up-regulation of apical genes in the basal cochlea, even though these regionally expressed genes are not necessarily conserved between the two species. In chicken, these altered gene expression patterns precede morphological and physiological changes in sensory hair cells that are typically associated with tonotopy such as the total number of stereocilia per hair cell and gene expression of an inward rectifier potassium channel, IRK1, which is a bona fide feature of apical hair cells in the basilar papilla. Furthermore, our results suggest that this conserved role of Shh in establishing cochlear tonotopy is initiated early in development by Shh emanating from the notochord and floor plate.
Collapse
|
15
|
Xiong W, Wagner T, Yan L, Grillet N, Müller U. Using injectoporation to deliver genes to mechanosensory hair cells. Nat Protoc 2014; 9:2438-49. [PMID: 25232939 PMCID: PMC4241755 DOI: 10.1038/nprot.2014.168] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Mechanosensation, the transduction of mechanical force into electrochemical signals, allows organisms to detect touch and sound, to register movement and gravity, and to sense changes in cell volume and shape. The hair cells of the mammalian inner ear are the mechanosensors for the detection of sound and head movement. The analysis of gene function in hair cells has been hampered by the lack of an efficient gene transfer method. Here we describe a method termed injectoporation that combines tissue microinjection with electroporation to express cDNAs and shRNAs in mouse cochlear hair cells. Injectoporation allows for gene transfer into dozens of hair cells, and it is compatible with the analysis of hair cell function using imaging approaches and electrophysiology. Tissue dissection and injectoporation can be carried out within a few hours, and the tissue can be cultured for days for subsequent functional analyses.
Collapse
Affiliation(s)
- Wei Xiong
- 1] Department of Molecular and Cellular Neuroscience, The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California, USA. [2]
| | - Thomas Wagner
- Department of Molecular and Cellular Neuroscience, The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California, USA
| | - Linxuan Yan
- Department of Molecular and Cellular Neuroscience, The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California, USA
| | - Nicolas Grillet
- 1] Department of Molecular and Cellular Neuroscience, The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California, USA. [2]
| | - Ulrich Müller
- Department of Molecular and Cellular Neuroscience, The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California, USA
| |
Collapse
|
16
|
Ferguson DP, Dangott LJ, Lightfoot JT. Lessons learned from vivo-morpholinos: How to avoid vivo-morpholino toxicity. Biotechniques 2014; 56:251-6. [PMID: 24806225 DOI: 10.2144/000114167] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 03/26/2014] [Indexed: 11/23/2022] Open
Abstract
Vivo-morpholinos are a promising tool for gene silencing. These oligonucleotide analogs transiently silence genes by blocking either translation or pre-mRNA splicing. Little to no toxicity has been reported for vivo-morpholino treatment. However, in a recent study conducted in our lab, treatment of mice with vivo-morpholinos resulted in high mortality rates. We hypothesized that the deaths were the result of oligonucleotide hybridization, causing an increased cationic charge associated with the dendrimer delivery moiety of the vivo-morpholino. The cationic charge increased blood clot formation in whole blood treated with vivo-morpholinos, suggesting that clotting could have caused cardiac arrest in the deceased mice. Therefore, we investigate the mechanism by which some vivo-morpholinos increase mortality rates and propose techniques to alleviate vivo-morpholino toxicity.
Collapse
Affiliation(s)
- David P Ferguson
- Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX; Biology of Physical Activity Laboratory, Dept. of Health & Kinesiology, Texas A&M University, College Station, TX
| | - Lawrence J Dangott
- Protein Chemistry Laboratory, Dept. of Biochemistry/Biophysics, Texas A&M University, College Station, TX
| | - J Timothy Lightfoot
- Biology of Physical Activity Laboratory, Dept. of Health & Kinesiology, Texas A&M University, College Station, TX
| |
Collapse
|
17
|
Ferguson DP, Schmitt EE, Lightfoot JT. Vivo-morpholinos induced transient knockdown of physical activity related proteins. PLoS One 2013; 8:e61472. [PMID: 23630592 PMCID: PMC3632599 DOI: 10.1371/journal.pone.0061472] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 03/13/2013] [Indexed: 11/19/2022] Open
Abstract
Physical activity is associated with disease prevention and overall wellbeing. Additionally there has been evidence that physical activity level is a result of genetic influence. However, there has not been a reliable method to silence candidate genes in vivo to determine causal mechanisms of physical activity regulation. Vivo-morpholinos are a potential method to transiently silence specific genes. Thus, the aim of this study was to validate the use of Vivo-morpholinos in a mouse model for voluntary physical activity with several sub-objectives. We observed that Vivo-morpholinos achieved between 60–97% knockdown of Drd1-, Vmat2-, and Glut4-protein in skeletal muscle, the delivery moiety of Vivo-morpholinos (scramble) did not influence physical activity and that a cocktail of multiple Vivo-morpholinos can be given in a single treatment to achieve protein knockdown of two different targeted proteins in skeletal muscle simultaneously. Knocking down Drd1, Vmat2, or Glut4 protein in skeletal muscle did not affect physical activity. Vivo-morpholinos injected intravenously alone did not significantly knockdown Vmat2-protein expression in the brain (p = 0.28). However, the use of a bradykinin analog to increase blood-brain-barrier permeability in conjunction with the Vivo-morpholinos significantly (p = 0.0001) decreased Vmat2-protein in the brain with a corresponding later over-expression of Vmat2 coincident with a significant (p = 0.0016) increase in physical activity. We conclude that Vivo-morpholinos can be a valuable tool in determining causal gene-phenotype relationships in whole animal models.
Collapse
Affiliation(s)
- David P Ferguson
- Department of Health and Kinesiology, Texas A&M University, College Station, Texas, United States of America.
| | | | | |
Collapse
|
18
|
Chen Y, Londraville R, Brickner S, El-Shaar L, Fankhauser K, Dearth C, Fulton L, Sochacka A, Bhattarai S, Marrs JA, Liu Q. Protocadherin-17 function in Zebrafish retinal development. Dev Neurobiol 2013; 73:259-73. [PMID: 22927092 DOI: 10.1002/dneu.22053] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2012] [Revised: 08/16/2012] [Accepted: 08/22/2012] [Indexed: 11/11/2022]
Abstract
Cadherin cell adhesion molecules play crucial roles in vertebrate development including the development of the retina. Most studies have focused on examining functions of classic cadherins (e.g. N-cadherin) in retinal development. There is little information on the function of protocadherins in the development of the vertebrate visual system. We previously showed that protocadherin-17 mRNA was expressed in developing zebrafish retina during critical stages of the retinal development. To gain insight into protocadherin-17 function in the formation of the retina, we analyzed eye development and differentiation of retinal cells in zebrafish embryos injected with protocadherin-17 specific antisense morpholino oligonucleotides (MOs). Protocadherin-17 knockdown embryos (pcdh17 morphants) had significantly reduced eyes due mainly to decreased cell proliferation. Differentiation of several retinal cell types (e.g. retinal ganglion cells) was also disrupted in the pcdh17 morphants. Phenotypic rescue was achieved by injection of protocadherin-17 mRNA. Injection of a vivo-protocadherin-17 MO into one eye of embryonic zebrafish resulted in similar eye defects. Our results suggest that protocadherin-17 plays an important role in the normal formation of the zebrafish retina.
Collapse
Affiliation(s)
- Yun Chen
- Department of Biology and Integrated Bioscience Program, University of Akron, Akron, Ohio 44325, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Hsieh FY, Ma TL, Shih HY, Lin SJ, Huang CW, Wang HY, Cheng YC. Dner inhibits neural progenitor proliferation and induces neuronal and glial differentiation in zebrafish. Dev Biol 2013; 375:1-12. [PMID: 23328254 DOI: 10.1016/j.ydbio.2013.01.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Revised: 12/17/2012] [Accepted: 01/02/2013] [Indexed: 11/19/2022]
Abstract
Delta/notch-like epidermal growth factor (EGF)-related receptor (DNER) is a single-pass transmembrane protein found to be a novel ligand in the Notch signaling pathway. Its function was previously characterized in the developing cerebellum and inner ear hair cells. In this study, we isolated a zebrafish homolog of DNER and showed that this gene is expressed in the developing nervous system. Overexpression of dner or the intracellular domain of dner was sufficient to inhibit the proliferation of neural progenitors and induce neuronal and glial differentiation. In contrast, the knockdown of endogenous Dner expression using antisense morpholino oligonucleotides increased the proliferation of neural progenitors and maintained neural cells in a progenitor status through inhibition of neuronal and glial differentiation. Through analysis of the antagonistic effect on the Delta ligand and the role of the potential downstream mediator Deltex1, we showed that Dner acts in Notch-dependent and Notch-independent manner. This is the first study to demonstrate a role for Dner in neural progenitors and neuronal differentiation and provides new insights into mediation of neuronal development and differentiation by the Notch signaling pathway.
Collapse
Affiliation(s)
- Fu-Yu Hsieh
- Graduate Institute of Biomedical Sciences, School of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | | | | | | | | | | | | |
Collapse
|
20
|
Hancock DB, Artigas MS, Gharib SA, Henry A, Manichaikul A, Ramasamy A, Loth DW, Imboden M, Koch B, McArdle WL, Smith AV, Smolonska J, Sood A, Tang W, Wilk JB, Zhai G, Zhao JH, Aschard H, Burkart KM, Curjuric I, Eijgelsheim M, Elliott P, Gu X, Harris TB, Janson C, Homuth G, Hysi PG, Liu JZ, Loehr LR, Lohman K, Loos RJF, Manning AK, Marciante KD, Obeidat M, Postma DS, Aldrich MC, Brusselle GG, Chen TH, Eiriksdottir G, Franceschini N, Heinrich J, Rotter JI, Wijmenga C, Williams OD, Bentley AR, Hofman A, Laurie CC, Lumley T, Morrison AC, Joubert BR, Rivadeneira F, Couper DJ, Kritchevsky SB, Liu Y, Wjst M, Wain LV, Vonk JM, Uitterlinden AG, Rochat T, Rich SS, Psaty BM, O'Connor GT, North KE, Mirel DB, Meibohm B, Launer LJ, Khaw KT, Hartikainen AL, Hammond CJ, Gläser S, Marchini J, Kraft P, Wareham NJ, Völzke H, Stricker BHC, Spector TD, Probst-Hensch NM, Jarvis D, Jarvelin MR, Heckbert SR, Gudnason V, Boezen HM, Barr RG, Cassano PA, Strachan DP, Fornage M, Hall IP, Dupuis J, Tobin MD, London SJ. Genome-wide joint meta-analysis of SNP and SNP-by-smoking interaction identifies novel loci for pulmonary function. PLoS Genet 2012; 8:e1003098. [PMID: 23284291 PMCID: PMC3527213 DOI: 10.1371/journal.pgen.1003098] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Accepted: 10/01/2012] [Indexed: 01/20/2023] Open
Abstract
Genome-wide association studies have identified numerous genetic loci for spirometic measures of pulmonary function, forced expiratory volume in one second (FEV(1)), and its ratio to forced vital capacity (FEV(1)/FVC). Given that cigarette smoking adversely affects pulmonary function, we conducted genome-wide joint meta-analyses (JMA) of single nucleotide polymorphism (SNP) and SNP-by-smoking (ever-smoking or pack-years) associations on FEV(1) and FEV(1)/FVC across 19 studies (total N = 50,047). We identified three novel loci not previously associated with pulmonary function. SNPs in or near DNER (smallest P(JMA = )5.00×10(-11)), HLA-DQB1 and HLA-DQA2 (smallest P(JMA = )4.35×10(-9)), and KCNJ2 and SOX9 (smallest P(JMA = )1.28×10(-8)) were associated with FEV(1)/FVC or FEV(1) in meta-analysis models including SNP main effects, smoking main effects, and SNP-by-smoking (ever-smoking or pack-years) interaction. The HLA region has been widely implicated for autoimmune and lung phenotypes, unlike the other novel loci, which have not been widely implicated. We evaluated DNER, KCNJ2, and SOX9 and found them to be expressed in human lung tissue. DNER and SOX9 further showed evidence of differential expression in human airway epithelium in smokers compared to non-smokers. Our findings demonstrated that joint testing of SNP and SNP-by-environment interaction identified novel loci associated with complex traits that are missed when considering only the genetic main effects.
Collapse
Affiliation(s)
- Dana B. Hancock
- Behavioral Health Epidemiology Program, Research Triangle Institute International, Research Triangle Park, North Carolina, United States of America
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, U.S. Department of Health and Human Services, Research Triangle Park, North Carolina, United States of America
| | - María Soler Artigas
- Departments of Health Sciences and Genetics, University of Leicester, Leicester, United Kingdom
| | - Sina A. Gharib
- Center for Lung Biology, University of Washington, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Amanda Henry
- Division of Therapeutics and Molecular Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom
| | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Public Health Sciences, Division of Biostatistics and Epidemiology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Adaikalavan Ramasamy
- Respiratory Epidemiology and Public Health, Imperial College London, London, United Kingdom
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- Department of Medical and Molecular Genetics, King's College London, Guy's Hospital, London, United Kingdom
| | - Daan W. Loth
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Inspectorate of Healthcare, The Hague, The Netherlands
| | - Medea Imboden
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Beate Koch
- Department of Internal Medicine B, University Hospital Greifswald, Greifswald, Germany
| | - Wendy L. McArdle
- ALSPAC, School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Albert V. Smith
- Icelandic Heart Association, Kopavogur, Iceland
- University of Iceland, Reykjavik, Iceland
| | - Joanna Smolonska
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Akshay Sood
- Department of Medicine, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Wenbo Tang
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, United States of America
| | - Jemma B. Wilk
- Division of Aging, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - Guangju Zhai
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
- Discipline of Genetics, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Jing Hua Zhao
- MRC Epidemiology Unit, Institute of Metabolic Science, Cambridge, United Kingdom
| | - Hugues Aschard
- Program in Molecular and Genetic Epidemiology, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Kristin M. Burkart
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
| | - Ivan Curjuric
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Mark Eijgelsheim
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- MRC-HPA Centre for Environment and Health, Imperial College London, London, United Kingdom
| | - Xiangjun Gu
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Tamara B. Harris
- National Institute on Aging, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Christer Janson
- Department of Medical Sciences, Respiratory Medicine, Uppsala University, Uppsala, Sweden
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Pirro G. Hysi
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Jason Z. Liu
- Department of Statistics, University of Oxford, United Kingdom
| | - Laura R. Loehr
- Gillings School of Global Public Health, Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Kurt Lohman
- Department of Biostatistical Sciences, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Ruth J. F. Loos
- MRC Epidemiology Unit, Institute of Metabolic Science, Cambridge, United Kingdom
| | - Alisa K. Manning
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, United States of America
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Kristin D. Marciante
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Ma'en Obeidat
- Division of Therapeutics and Molecular Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom
| | - Dirkje S. Postma
- Department of Pulmonology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- GRIAC Research Institute, University Medical Center Groningen, Groningen, The Netherlands
| | - Melinda C. Aldrich
- Department of Thoracic Surgery and Division of Epidemiology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Guy G. Brusselle
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Ting-hsu Chen
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, Veterans Administration Boston Healthcare System, Boston, Massachusetts, United States of America
- The Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | | | - Nora Franceschini
- Gillings School of Global Public Health, Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Joachim Heinrich
- Institute of Epidemiology I, Helmholtz Zentrum München, Munich, Germany
| | - Jerome I. Rotter
- Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Cisca Wijmenga
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - O. Dale Williams
- Department of Biostatistics, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, United States of America
| | - Amy R. Bentley
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Cathy C. Laurie
- Department of Biostatistics, University of Washington, Seattle, Washington, United States of America
| | - Thomas Lumley
- Department of Statistics, University of Auckland, Auckland, New Zealand
| | - Alanna C. Morrison
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Bonnie R. Joubert
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, U.S. Department of Health and Human Services, Research Triangle Park, North Carolina, United States of America
| | - Fernando Rivadeneira
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)–sponsored Netherlands Consortium for Healthy Aging (NCHA), Leiden, The Netherlands
| | - David J. Couper
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Stephen B. Kritchevsky
- Sticht Center on Aging, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Yongmei Liu
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Matthias Wjst
- Institute of Lung Biology and Disease, Comprehensive Pneumology Center, Helmholtz Zentrum München, Neuherberg, Germany
- Institute for Medical Statistics and Epidemiology (IMSE), Technical University Munich, Munich, Germany
| | - Louise V. Wain
- Departments of Health Sciences and Genetics, University of Leicester, Leicester, United Kingdom
| | - Judith M. Vonk
- GRIAC Research Institute, University Medical Center Groningen, Groningen, The Netherlands
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- The LifeLines Cohort Study, Groningen, The Netherlands
| | - André G. Uitterlinden
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)–sponsored Netherlands Consortium for Healthy Aging (NCHA), Leiden, The Netherlands
| | - Thierry Rochat
- Division of Pulmonary Medicine, University Hospitals of Geneva, Geneva, Switzerland
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit and Department of Epidemiology, University of Washington, Seattle, Washington, United States of America
- Departments of Medicine and Health Services, University of Washington, Seattle, United States of America
- Group Health Research Institute, Group Health Cooperative, Seattle, Washington
| | - George T. O'Connor
- The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts, United States of America
- The Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Kari E. North
- Gillings School of Global Public Health, Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Daniel B. Mirel
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Bernd Meibohm
- College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Lenore J. Launer
- National Institute on Aging, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Kay-Tee Khaw
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Anna-Liisa Hartikainen
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Oulu, Oulu, Finland
| | - Christopher J. Hammond
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Sven Gläser
- Department of Internal Medicine B, University Hospital Greifswald, Greifswald, Germany
| | | | - Peter Kraft
- Departments of Epidemiology and Biostatistics, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Nicholas J. Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, Cambridge, United Kingdom
| | - Henry Völzke
- Institute for Community Medicine, Study of Health In Pomerania (SHIP)/Clinical Epidemiological Research, University of Greifswald, Greifswald, Germany
| | - Bruno H. C. Stricker
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Inspectorate of Healthcare, The Hague, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Medical Informatics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Timothy D. Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Nicole M. Probst-Hensch
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Deborah Jarvis
- Respiratory Epidemiology and Public Health, Imperial College London, London, United Kingdom
- MRC-HPA Centre for Environment and Health, Imperial College London, London, United Kingdom
| | - Marjo-Riitta Jarvelin
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- MRC-HPA Centre for Environment and Health, Imperial College London, London, United Kingdom
- Department of Children, Young People, and Families, National Institute for Health and Welfare, Oulu, Finland
- Institute of Health Sciences, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Susan R. Heckbert
- Cardiovascular Health Research Unit and Department of Epidemiology, University of Washington, Seattle, Washington, United States of America
- Group Health Research Institute, Group Health Cooperative, Seattle, Washington
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- University of Iceland, Reykjavik, Iceland
| | - H. Marike Boezen
- GRIAC Research Institute, University Medical Center Groningen, Groningen, The Netherlands
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- The LifeLines Cohort Study, Groningen, The Netherlands
| | - R. Graham Barr
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York, United States of America
| | - Patricia A. Cassano
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, United States of America
- Department of Public Health, Weill Cornell Medical College, New York, New York, United States of America
| | - David P. Strachan
- Division of Population Health Sciences and Education, St. George's University of London, London, United Kingdom
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Ian P. Hall
- Division of Therapeutics and Molecular Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom
| | - Josée Dupuis
- The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts, United States of America
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - Martin D. Tobin
- Departments of Health Sciences and Genetics, University of Leicester, Leicester, United Kingdom
| | - Stephanie J. London
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, U.S. Department of Health and Human Services, Research Triangle Park, North Carolina, United States of America
- Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences, National Institutes of Health, U.S. Department of Health and Human Services, Research Triangle Park, North Carolina, United States of America
| |
Collapse
|
21
|
Abstract
The very large G protein coupled receptor (Vlgr1) is a member of adhesion receptors or large N-terminal family B-7 transmembrane helixes (LNB7TM) receptors within the seven trans-membrane receptor superfamily. Vlgr1 is the largest GPCR identified to date; its mRNA spans 19 kb and encodes 6,300 amino acids. Vlgr1 is a core component of ankle-link complex in inner ear hair cells. Knock-out and mutation mouse models show that loss of Vlgr1 function leads to abnormal stereociliary development and hearing loss, indicating crucial roles of Vlgr1 in hearing transduction or auditory system development. Over the past 10 or so years, human genetics data suggested that Vlgr1 mutations cause Usher syndromes and seizures. Although significant progresses have been made, the details of Vlgr1's function in hair cells, its signaling cascade, and the mechanisms underlying causative effects of Vlgr1 mutations in human diseases remain elusive and ask for further investigation.
Collapse
|
22
|
Luo YJ, Su YH. Opposing nodal and BMP signals regulate left-right asymmetry in the sea urchin larva. PLoS Biol 2012; 10:e1001402. [PMID: 23055827 PMCID: PMC3467216 DOI: 10.1371/journal.pbio.1001402] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 08/28/2012] [Indexed: 12/28/2022] Open
Abstract
Nodal and BMP signals are important for establishing left-right (LR) asymmetry in vertebrates. In sea urchins, Nodal signaling prevents the formation of the rudiment on the right side. However, the opposing pathway to Nodal signaling during LR axis establishment is not clear. Here, we revealed that BMP signaling is activated in the left coelomic pouch, specifically in the veg2 lineage, but not in the small micromeres. By perturbing BMP activities, we demonstrated that BMP signaling is required for activating the expression of the left-sided genes and the formation of the left-sided structures. On the other hand, Nodal signals on the right side inhibit BMP signaling and control LR asymmetric separation and apoptosis of the small micromeres. Our findings show that BMP signaling is the positive signal for left-sided development in sea urchins, suggesting that the opposing roles of Nodal and BMP signals in establishing LR asymmetry are conserved in deuterostomes.
Collapse
Affiliation(s)
| | - Yi-Hsien Su
- Institute of Cellular and Organismic Biology, Academia Sinica, Nankang, Taipei, Taiwan
| |
Collapse
|
23
|
Son EJ, Wu L, Yoon H, Kim S, Choi JY, Bok J. Developmental gene expression profiling along the tonotopic axis of the mouse cochlea. PLoS One 2012; 7:e40735. [PMID: 22808246 PMCID: PMC3395647 DOI: 10.1371/journal.pone.0040735] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 06/12/2012] [Indexed: 12/13/2022] Open
Abstract
The mammalian cochlear duct is tonotopically organized such that the basal cochlea is tuned to high frequency sounds and the apical cochlea to low frequency sounds. In an effort to understand how this tonotopic organization is established, we searched for genes that are differentially expressed along the tonotopic axis during neonatal development. Cochlear tissues dissected from P0 and P8 mice were divided into three equal pieces, representing the base, middle and apex, and gene expression profiles were determined using the microarray technique. The gene expression profiles were grouped according to changes in expression levels along the tonotopic axis as well as changes during neonatal development. The classified groups were further analyzed by functional annotation clustering analysis to determine whether genes associated with specific biological function or processes are particularly enriched in each group. These analyses identified several candidate genes that may be involved in cochlear development and acquisition of tonotopy. We examined the expression domains for a few candidate genes in the developing mouse cochlea. Tnc (tenacin C) and Nov (nephroblastoma overexpressed gene) are expressed in the basilar membrane, with increased expression toward the apex, which may contribute to graded changes in the structure of the basilar membrane along the tonotopic axis. In addition, Fst (Follistatin), an antagonist of TGF-β/BMP signaling, is expressed in the lesser epithelial ridge and at gradually higher levels towards the apex. The graded expression pattern of Fst is established at the time of cochlear specification and maintained throughout embryonic and postnatal development, suggesting its possible role in the organization of tonotopy. Our data will provide a good resource for investigating the developmental mechanisms of the mammalian cochlea including the acquisition of tonotopy.
Collapse
Affiliation(s)
- Eun Jin Son
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea
- * E-mail: (JB); (EJS)
| | - Ling Wu
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea
- BK21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Heejei Yoon
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea
| | - Sunhee Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Young Choi
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea
- BK21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Jinwoong Bok
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea
- BK21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
- * E-mail: (JB); (EJS)
| |
Collapse
|
24
|
Heterogeneous nuclear ribonucleoprotein K, an RNA-binding protein, is required for optic axon regeneration in Xenopus laevis. J Neurosci 2012; 32:3563-74. [PMID: 22399778 DOI: 10.1523/jneurosci.5197-11.2012] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Axotomized optic axons of Xenopus laevis, in contrast to those of mammals, retain their ability to regenerate throughout life. To better understand the molecular basis for this successful regeneration, we focused on the role of an RNA-binding protein, heterogeneous nuclear ribonucleoprotein (hnRNP) K, because it is required for axonogenesis during development and because several of its RNA targets are under strong post-transcriptional control during regeneration. At 11 d after optic nerve crush, hnRNP K underwent significant translocation into the nucleus of retinal ganglion cells (RGCs), indicating that the protein became activated during regeneration. To suppress its expression, we intravitreously injected an antisense Vivo-Morpholino oligonucleotide targeting hnRNP K. In uninjured eyes, it efficiently knocked down hnRNP K expression in only the RGCs, without inducing either an axotomy response or axon degeneration. After optic nerve crush, staining for multiple markers of regenerating axons showed no regrowth of axons beyond the lesion site with hnRNP K knockdown. RGCs nonetheless responded to the injury by increasing expression of multiple growth-associated RNAs and experienced no additional neurodegeneration above that normally seen with optic nerve injury. At the molecular level, hnRNP K knockdown during regeneration inhibited protein, but not mRNA, expression of several known hnRNP K RNA targets (NF-M, GAP-43) by compromising their efficient nuclear transport and disrupting their loading onto polysomes for translation. Our study therefore provides evidence of a novel post-transcriptional regulatory pathway orchestrated by hnRNP K that is essential for successful CNS axon regeneration.
Collapse
|
25
|
Ohler A, Becker-Pauly C. Morpholino knockdown of the ubiquitously expressed transmembrane serine protease TMPRSS4a in zebrafish embryos exhibits severe defects in organogenesis and cell adhesion. Biol Chem 2011; 392:653-64. [PMID: 21657981 DOI: 10.1515/bc.2011.070] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract Over the past years the members of the type II transmembrane serine protease (TTSP) family have emerged as new players in mammalian biology. TMPRSS4 (transmembrane protease/serine) is overexpressed in several human cancer tissues, promoting invasion, migration, and metastasis. However, the physiological function has not yet been elucidated. Here, we present morpholino knockdown studies targeting TMPRSS4a, a homolog of human TMPRSS4 in zebrafish embryos. By RT-PCR, we could demonstrate an expression of this protease already 5 h post-fertilization, suggesting important functions in the early stages of embryonic development. Indeed, in vivo gene silencing caused severe defects in tissue development and cell differentiation including a disturbed skeletal muscle formation, a decelerated heartbeat, and a degenerated vascular system. Scanning electron microscopy revealed strong defects in epidermal skin organization, with clearly altered cell-cell contacts, resulting in the detachment of keratinocytes from the underneath tissue. The disturbed organogenesis in general is consistent with RT-PCR results which exhibited a ubiquitous expression of TMPRSS4a, predominantly in kidney, skin, heart, and gills. Our results demonstrate the importance of TMPRSS4a in tissue development and cell differentiation. Whether its proteolytic activity is directed towards adhesion molecules or leads to the activation of other proteases needs to be investigated further.
Collapse
Affiliation(s)
- Anke Ohler
- Institute of Zoology, Cell and Matrix Biology, Johannes Gutenberg University, Germany
| | | |
Collapse
|