1
|
Ali M, Kutlowski JW, Holland JN, Riley BB. Foxm1 promotes differentiation of neural progenitors in the zebrafish inner ear. Dev Biol 2025; 520:21-30. [PMID: 39761737 DOI: 10.1016/j.ydbio.2025.01.001] [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: 08/02/2024] [Revised: 12/17/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025]
Abstract
During development of the vertebrate inner ear, sensory epithelia and neurons of the statoacoustic ganglion (SAG) arise from lineage-restricted progenitors that proliferate extensively before differentiating into mature post-mitotic cell types. Development of progenitors is regulated by Fgf, Wnt and Notch signaling, but how these pathways are coordinated to achieve an optimal balance of proliferation and differentiation is not well understood. Here we investigate the role in zebrafish of Foxm1, a transcription factor commonly associated with proliferation in developing tissues and tumors. Targeted knockout of foxm1 causes no overt defects in development. Homozygous mutants are viable and exhibit no obvious defects except male sterility. However, the mutant allele acts dominantly to reduce accumulation of SAG neurons, and maternal loss-of-function slightly enhances this deficiency. Neural progenitors are specified normally but, unexpectedly, persist in an early state of rapid proliferation and are delayed in differentiation. Progenitors eventually shift to a slower rate of proliferation similar to wild-type and differentiate to produce a normal number of SAG neurons, although the arrangement of neurons remains variably disordered. Mutant progenitors remain responsive to Fgf and Notch, as blocking these pathways partially alleviates the delay in differentiation. However, the ability of elevated Wnt/beta-catenin to block neural specification is impaired in foxm1 mutants. Modulating Wnt at later stages has no effect on progenitors in mutant or wild-type embryos. Our findings document an unusual role for foxm1 in promoting differentiation of SAG progenitors from an early, rapidly dividing phase to a more mature slower phase prior to differentiation.
Collapse
Affiliation(s)
- Maria Ali
- Biology Department, Texas A&M University, College Station, TX, 7843-3258, USA
| | - James W Kutlowski
- Biology Department, Texas A&M University, College Station, TX, 7843-3258, USA
| | - Jorden N Holland
- Biology Department, Texas A&M University, College Station, TX, 7843-3258, USA
| | - Bruce B Riley
- Biology Department, Texas A&M University, College Station, TX, 7843-3258, USA.
| |
Collapse
|
2
|
Zhang B, Hu Y, Du H, Han S, Ren L, Cheng H, Wang Y, Gao X, Zheng S, Cui Q, Tian L, Liu T, Sun J, Chai R. Tissue engineering strategies for spiral ganglion neuron protection and regeneration. J Nanobiotechnology 2024; 22:458. [PMID: 39085923 PMCID: PMC11293049 DOI: 10.1186/s12951-024-02742-8] [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: 03/08/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024] Open
Abstract
Cochlear implants can directly activate the auditory system's primary sensory neurons, the spiral ganglion neurons (SGNs), via circumvention of defective cochlear hair cells. This bypass restores auditory input to the brainstem. SGN loss etiologies are complex, with limited mammalian regeneration. Protecting and revitalizing SGN is critical. Tissue engineering offers a novel therapeutic strategy, utilizing seed cells, biomolecules, and scaffold materials to create a cellular environment and regulate molecular cues. This review encapsulates the spectrum of both human and animal research, collating the factors contributing to SGN loss, the latest advancements in the utilization of exogenous stem cells for auditory nerve repair and preservation, the taxonomy and mechanism of action of standard biomolecules, and the architectural components of scaffold materials tailored for the inner ear. Furthermore, we delineate the potential and benefits of the biohybrid neural interface, an incipient technology in the realm of implantable devices. Nonetheless, tissue engineering requires refined cell selection and differentiation protocols for consistent SGN quality. In addition, strategies to improve stem cell survival, scaffold biocompatibility, and molecular cue timing are essential for biohybrid neural interface integration.
Collapse
Affiliation(s)
- Bin Zhang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Yangnan Hu
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| | - Haoliang Du
- Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing University, Nanjing, 210008, China
| | - Shanying Han
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Lei Ren
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Hong Cheng
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Yusong Wang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Xin Gao
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Shasha Zheng
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Qingyue Cui
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Lei Tian
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
| | - Tingting Liu
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
| | - Jiaqiang Sun
- Department of Otolaryngology-Head and Neck Surgery, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230001, China.
| | - Renjie Chai
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Department of Neurology, Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Science, Beijing, China.
- Southeast University Shenzhen Research Institute, Shenzhen, 518063, China.
| |
Collapse
|
3
|
Bertagnoli LE, Seist R, Batts S, Stankovic KM. Potential Ototoxicity of Insulin-like Growth Factor 1 Receptor Signaling Inhibitors: An In Silico Drug Repurposing Study of the Regenerating Cochlear Neuron Transcriptome. J Clin Med 2023; 12:jcm12103485. [PMID: 37240591 DOI: 10.3390/jcm12103485] [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: 02/28/2023] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Spiral ganglion neurons (SGNs) connect cochlear hair cells with higher auditory pathways and their degeneration due to drug toxicity (ototoxicity) contributes to hearing loss. This study aimed to identify drug classes that are negatively correlated with the transcriptome of regenerating SGNs. Human orthologs of differentially expressed genes within the regenerating neonatal mouse SGN transcriptome were entered into CMap and the LINCS unified environment and perturbation-driven gene expression was analyzed. The CMap connectivity scores ranged from 100 (positive correlation) to -100 (negative correlation). Insulin-like growth factor 1/receptor (IGF-1/R) inhibitors were highly negatively correlated with the regenerating SGN transcriptome (connectivity score: -98.87). A systematic literature review of clinical trials and observational studies reporting otologic adverse events (AEs) with IGF-1/R inhibitors identified 108 reports (6141 treated patients). Overall, 16.9% of the treated patients experienced any otologic AE; the rate was highest for teprotumumab (42.9%). In a meta-analysis of two randomized placebo-controlled trials of teprotumumab, there was a significantly higher risk of hearing-related (pooled Peto OR [95% CI]: 7.95 [1.57, 40.17]) and of any otologic AEs (3.56 [1.35, 9.43]) with teprotumumab vs. a placebo, whether or not dizziness/vertigo AEs were included. These results call for close audiological monitoring during IGF-1-targeted treatment, with prompt referral to an otolaryngologist should otologic AEs develop.
Collapse
Affiliation(s)
- Lino E Bertagnoli
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Paracelsus Medical University, 5020 Salzburg, Austria
| | - Richard Seist
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Paracelsus Medical University, 5020 Salzburg, Austria
| | - Shelley Batts
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Konstantina M Stankovic
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Wu Tsai Neuroscience Institute, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
4
|
IGF-1 Controls Metabolic Homeostasis and Survival in HEI-OC1 Auditory Cells through AKT and mTOR Signaling. Antioxidants (Basel) 2023; 12:antiox12020233. [PMID: 36829792 PMCID: PMC9952701 DOI: 10.3390/antiox12020233] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Insulin-like growth factor 1 (IGF-1) is a trophic factor for the nervous system where it exerts pleiotropic effects, including the regulation of metabolic homeostasis. IGF-1 deficiency induces morphological alterations in the cochlea, apoptosis and hearing loss. While multiple studies have addressed the role of IGF-1 in hearing protection, its potential function in the modulation of otic metabolism remains unclear. Here, we report that "House Ear Institute-organ of Corti 1" (HEI-OC1) auditory cells express IGF-system genes that are regulated during their differentiation. Upon binding to its high-affinity receptor IGF1R, IGF-1 activates AKT and mTOR signaling to stimulate anabolism and, concomitantly, to reduce autophagic catabolism in HEI-OC1 progenitor cells. Notably, IGF-1 stimulation during HEI-OC1 differentiation to mature otic cells sustained both constructive metabolism and autophagic flux, possibly to favor cell remodeling. IGF1R engagement and downstream AKT signaling promoted HEI-OC1 cell survival by maintaining redox balance, even when cells were challenged with the ototoxic agent cisplatin. Our findings establish that IGF-1 not only serves an important function in otic metabolic homeostasis but also activates antioxidant defense mechanisms to promote hair cell survival during the stress response to insults.
Collapse
|
5
|
García-Mato Á, Cervantes B, Murillo-Cuesta S, Rodríguez-de la Rosa L, Varela-Nieto I. Insulin-like Growth Factor 1 Signaling in Mammalian Hearing. Genes (Basel) 2021; 12:genes12101553. [PMID: 34680948 PMCID: PMC8535591 DOI: 10.3390/genes12101553] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 02/06/2023] Open
Abstract
Insulin-like growth factor 1 (IGF-1) is a peptide hormone belonging to the insulin family of proteins. Almost all of the biological effects of IGF-1 are mediated through binding to its high-affinity tyrosine kinase receptor (IGF1R), a transmembrane receptor belonging to the insulin receptor family. Factors, receptors and IGF-binding proteins form the IGF system, which has multiple roles in mammalian development, adult tissue homeostasis, and aging. Consequently, mutations in genes of the IGF system, including downstream intracellular targets, underlie multiple common pathologies and are associated with multiple rare human diseases. Here we review the contribution of the IGF system to our understanding of the molecular and genetic basis of human hearing loss by describing, (i) the expression patterns of the IGF system in the mammalian inner ear; (ii) downstream signaling of IGF-1 in the hearing organ; (iii) mouse mutations in the IGF system, including upstream regulators and downstream targets of IGF-1 that inform cochlear pathophysiology; and (iv) human mutations in these genes causing hearing loss.
Collapse
Affiliation(s)
- Ángela García-Mato
- Institute for Biomedical Research “Alberto Sols” (IIBm), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; (Á.G.-M.); (B.C.); (S.M.-C.)
- Rare Diseases Networking Biomedical Research Centre (CIBERER), CIBER, Carlos III Institute of Health, 28029 Madrid, Spain
| | - Blanca Cervantes
- Institute for Biomedical Research “Alberto Sols” (IIBm), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; (Á.G.-M.); (B.C.); (S.M.-C.)
- Rare Diseases Networking Biomedical Research Centre (CIBERER), CIBER, Carlos III Institute of Health, 28029 Madrid, Spain
| | - Silvia Murillo-Cuesta
- Institute for Biomedical Research “Alberto Sols” (IIBm), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; (Á.G.-M.); (B.C.); (S.M.-C.)
- Rare Diseases Networking Biomedical Research Centre (CIBERER), CIBER, Carlos III Institute of Health, 28029 Madrid, Spain
- La Paz Hospital Institute for Health Research (IdiPAZ), 28046 Madrid, Spain
| | - Lourdes Rodríguez-de la Rosa
- Rare Diseases Networking Biomedical Research Centre (CIBERER), CIBER, Carlos III Institute of Health, 28029 Madrid, Spain
- La Paz Hospital Institute for Health Research (IdiPAZ), 28046 Madrid, Spain
- Correspondence: (L.R.-d.l.R.); (I.V.-N.)
| | - Isabel Varela-Nieto
- Institute for Biomedical Research “Alberto Sols” (IIBm), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; (Á.G.-M.); (B.C.); (S.M.-C.)
- Rare Diseases Networking Biomedical Research Centre (CIBERER), CIBER, Carlos III Institute of Health, 28029 Madrid, Spain
- La Paz Hospital Institute for Health Research (IdiPAZ), 28046 Madrid, Spain
- Correspondence: (L.R.-d.l.R.); (I.V.-N.)
| |
Collapse
|
6
|
Bermúdez-Muñoz JM, Celaya AM, García-Mato Á, Muñoz-Espín D, Rodríguez-de la Rosa L, Serrano M, Varela-Nieto I. Dual-Specificity Phosphatase 1 (DUSP1) Has a Central Role in Redox Homeostasis and Inflammation in the Mouse Cochlea. Antioxidants (Basel) 2021; 10:1351. [PMID: 34572983 PMCID: PMC8467085 DOI: 10.3390/antiox10091351] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/16/2021] [Accepted: 08/23/2021] [Indexed: 12/13/2022] Open
Abstract
Stress-activated protein kinases (SAPK) are associated with sensorineural hearing loss (SNHL) of multiple etiologies. Their activity is tightly regulated by dual-specificity phosphatase 1 (DUSP1), whose loss of function leads to sustained SAPK activation. Dusp1 gene knockout in mice accelerates SNHL progression and triggers inflammation, redox imbalance and hair cell (HC) death. To better understand the link between inflammation and redox imbalance, we analyzed the cochlear transcriptome in Dusp1-/- mice. RNA sequencing analysis (GSE176114) indicated that Dusp1-/- cochleae can be defined by a distinct profile of key cellular expression programs, including genes of the inflammatory response and glutathione (GSH) metabolism. To dissociate the two components, we treated Dusp1-/- mice with N-acetylcysteine, and hearing was followed-up longitudinally by auditory brainstem response recordings. A combination of immunofluorescence, Western blotting, enzymatic activity, GSH levels measurements and RT-qPCR techniques were used. N-acetylcysteine treatment delayed the onset of SNHL and mitigated cochlear damage, with fewer TUNEL+ HC and lower numbers of spiral ganglion neurons with p-H2AX foci. N-acetylcysteine not only improved the redox balance in Dusp1-/- mice but also inhibited cytokine production and reduced macrophage recruitment. Our data point to a critical role for DUSP1 in controlling the cross-talk between oxidative stress and inflammation.
Collapse
Affiliation(s)
- Jose M. Bermúdez-Muñoz
- Institute for Biomedical Research “Alberto Sols”, Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; (A.M.C.); (Á.G.-M.); (L.R.-d.l.R.)
- Rare Diseases Networking Biomedical Research Centre (CIBERER), CIBER, Carlos III Institute of Health, 28029 Madrid, Spain
| | - Adelaida M. Celaya
- Institute for Biomedical Research “Alberto Sols”, Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; (A.M.C.); (Á.G.-M.); (L.R.-d.l.R.)
- Rare Diseases Networking Biomedical Research Centre (CIBERER), CIBER, Carlos III Institute of Health, 28029 Madrid, Spain
| | - Ángela García-Mato
- Institute for Biomedical Research “Alberto Sols”, Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; (A.M.C.); (Á.G.-M.); (L.R.-d.l.R.)
- Rare Diseases Networking Biomedical Research Centre (CIBERER), CIBER, Carlos III Institute of Health, 28029 Madrid, Spain
| | - Daniel Muñoz-Espín
- CRUK Cambridge Centre Early Detection Programme, Department of Oncology, University of Cambridge, Hutchison/MRC Research Centre, Cambridge CB2 0XZ, UK;
| | - Lourdes Rodríguez-de la Rosa
- Institute for Biomedical Research “Alberto Sols”, Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; (A.M.C.); (Á.G.-M.); (L.R.-d.l.R.)
- Rare Diseases Networking Biomedical Research Centre (CIBERER), CIBER, Carlos III Institute of Health, 28029 Madrid, Spain
- Hospital La Paz Institute for Health Research (IdiPAZ), 28029 Madrid, Spain
| | - Manuel Serrano
- Institute for Research in Biomedicine, Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain;
- Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
| | - Isabel Varela-Nieto
- Institute for Biomedical Research “Alberto Sols”, Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; (A.M.C.); (Á.G.-M.); (L.R.-d.l.R.)
- Rare Diseases Networking Biomedical Research Centre (CIBERER), CIBER, Carlos III Institute of Health, 28029 Madrid, Spain
- Hospital La Paz Institute for Health Research (IdiPAZ), 28029 Madrid, Spain
| |
Collapse
|
7
|
Celaya AM, Rodríguez-de la Rosa L, Bermúdez-Muñoz JM, Zubeldia JM, Romá-Mateo C, Avendaño C, Pallardó FV, Varela-Nieto I. IGF-1 Haploinsufficiency Causes Age-Related Chronic Cochlear Inflammation and Increases Noise-Induced Hearing Loss. Cells 2021; 10:cells10071686. [PMID: 34359856 PMCID: PMC8304185 DOI: 10.3390/cells10071686] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 06/28/2021] [Accepted: 06/28/2021] [Indexed: 12/18/2022] Open
Abstract
Insulin-like growth factor 1 (IGF-1) deficiency is an ultrarare syndromic human sensorineural deafness. Accordingly, IGF-1 is essential for the postnatal maturation of the cochlea and the correct wiring of hearing in mice. Less severe decreases in human IGF-1 levels have been associated with other hearing loss rare genetic syndromes, as well as with age-related hearing loss (ARHL). However, the underlying mechanisms linking IGF-1 haploinsufficiency with auditory pathology and ARHL have not been studied. Igf1-heterozygous mice express less Igf1 transcription and have 40% lower IGF-1 serum levels than wild-type mice. Along with ageing, IGF-1 levels decreased concomitantly with the increased expression of inflammatory cytokines, Tgfb1 and Il1b, but there was no associated hearing loss. However, noise exposure of these mice caused increased injury to sensory hair cells and irreversible hearing loss. Concomitantly, there was a significant alteration in the expression ratio of pro- and anti-inflammatory cytokines in Igf1+/- mice. Unbalanced inflammation led to the activation of the stress kinase JNK and the failure to activate AKT. Our data show that IGF-1 haploinsufficiency causes a chronic subclinical proinflammatory age-associated state and, consequently, greater susceptibility to stressors. This work provides the molecular bases to further understand hearing disorders linked to IGF-1 deficiency.
Collapse
Affiliation(s)
- Adelaida M. Celaya
- Institute for Biomedical Research “Alberto Sols” (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; (A.M.C.); (J.M.B.-M.); (J.M.Z.)
- Rare Diseases Biomedical Research Networking Centre (CIBERER), The Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain; (C.R.-M.); (F.V.P.)
| | - Lourdes Rodríguez-de la Rosa
- Institute for Biomedical Research “Alberto Sols” (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; (A.M.C.); (J.M.B.-M.); (J.M.Z.)
- Rare Diseases Biomedical Research Networking Centre (CIBERER), The Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain; (C.R.-M.); (F.V.P.)
- Hospital La Paz Institute for Health Research (IdiPAZ), 28029 Madrid, Spain;
- Correspondence: (L.R.-d.l.R.); (I.V.-N.)
| | - Jose M. Bermúdez-Muñoz
- Institute for Biomedical Research “Alberto Sols” (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; (A.M.C.); (J.M.B.-M.); (J.M.Z.)
- Rare Diseases Biomedical Research Networking Centre (CIBERER), The Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain; (C.R.-M.); (F.V.P.)
| | - José M. Zubeldia
- Institute for Biomedical Research “Alberto Sols” (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; (A.M.C.); (J.M.B.-M.); (J.M.Z.)
- Rare Diseases Biomedical Research Networking Centre (CIBERER), The Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain; (C.R.-M.); (F.V.P.)
- Allergy Service, Gregorio Marañon General University Hospital, 28009 Madrid, Spain
- Gregorio Marañon Health Research Institute (IiSGM), 28009 Madrid, Spain
| | - Carlos Romá-Mateo
- Rare Diseases Biomedical Research Networking Centre (CIBERER), The Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain; (C.R.-M.); (F.V.P.)
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Spain and FIHCUV-INCLIVA, 46010 Valencia, Spain
| | - Carlos Avendaño
- Hospital La Paz Institute for Health Research (IdiPAZ), 28029 Madrid, Spain;
- Department of Anatomy, Histology & Neuroscience, Medical School, Autonomous University of Madrid, 28029 Madrid, Spain
| | - Federico V. Pallardó
- Rare Diseases Biomedical Research Networking Centre (CIBERER), The Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain; (C.R.-M.); (F.V.P.)
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Spain and FIHCUV-INCLIVA, 46010 Valencia, Spain
| | - Isabel Varela-Nieto
- Institute for Biomedical Research “Alberto Sols” (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; (A.M.C.); (J.M.B.-M.); (J.M.Z.)
- Rare Diseases Biomedical Research Networking Centre (CIBERER), The Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain; (C.R.-M.); (F.V.P.)
- Hospital La Paz Institute for Health Research (IdiPAZ), 28029 Madrid, Spain;
- Correspondence: (L.R.-d.l.R.); (I.V.-N.)
| |
Collapse
|
8
|
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: 6] [Impact Index Per Article: 1.5] [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
|
9
|
León Y, Magariños M, Varela-Nieto I. Ceramide Kinase Inhibition Blocks IGF-1-Mediated Survival of Otic Neurosensory Progenitors by Impairing AKT Phosphorylation. Front Cell Dev Biol 2021; 9:678760. [PMID: 34179008 PMCID: PMC8220815 DOI: 10.3389/fcell.2021.678760] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 05/12/2021] [Indexed: 11/29/2022] Open
Abstract
Sphingolipids are bioactive lipid components of cell membranes with important signal transduction functions in health and disease. Ceramide is the central building block for sphingolipid biosynthesis and is processed to form structurally and functionally distinct sphingolipids. Ceramide can be phosphorylated by ceramide kinase (CERK) to generate ceramide-1-phosphate, a cytoprotective signaling molecule that has been widely studied in multiple tissues and organs, including the developing otocyst. However, little is known about ceramide kinase regulation during inner ear development. Using chicken otocysts, we show that genes for CERK and other enzymes of ceramide metabolism are expressed during the early stages of inner ear development and that CERK is developmentally regulated at the otic vesicle stage. To explore its role in inner ear morphogenesis, we blocked CERK activity in organotypic cultures of otic vesicles with a specific inhibitor. Inhibition of CERK activity impaired proliferation and promoted apoptosis of epithelial otic progenitors. CERK inhibition also compromised neurogenesis of the acoustic-vestibular ganglion. Insulin-like growth factor-1 (IGF-1) is a key factor for proliferation, survival and differentiation in the chicken otocyst. CERK inhibition decreased IGF-1-induced AKT phosphorylation and blocked IGF-1-induced cell survival. Overall, our data suggest that CERK is activated as a central element in the network of anti-apoptotic pro-survival pathways elicited by IGF-1 during early inner ear development.
Collapse
Affiliation(s)
- Yolanda León
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, Madrid, Spain.,Departamento de Biología, Universidad Autónoma de Madrid, Madrid, Spain
| | - Marta Magariños
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, Madrid, Spain.,Departamento de Biología, Universidad Autónoma de Madrid, Madrid, Spain.,CIBERER, Unit 761, CIBER, ISCIII, Madrid, Spain
| | - Isabel Varela-Nieto
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, Madrid, Spain.,CIBERER, Unit 761, CIBER, ISCIII, Madrid, Spain
| |
Collapse
|
10
|
Growth Hormone and the Auditory Pathway: Neuromodulation and Neuroregeneration. Int J Mol Sci 2021; 22:ijms22062829. [PMID: 33799503 PMCID: PMC7998811 DOI: 10.3390/ijms22062829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/26/2021] [Accepted: 03/05/2021] [Indexed: 02/06/2023] Open
Abstract
Growth hormone (GH) plays an important role in auditory development during the embryonic stage. Exogenous agents such as sound, noise, drugs or trauma, can induce the release of this hormone to perform a protective function and stimulate other mediators that protect the auditory pathway. In addition, GH deficiency conditions hearing loss or central auditory processing disorders. There are promising animal studies that reflect a possible regenerative role when exogenous GH is used in hearing impairments, demonstrated in in vivo and in vitro studies, and also, even a few studies show beneficial effects in humans presented and substantiated in the main text, although they should not exaggerate the main conclusions.
Collapse
|
11
|
Bermúdez‐Muñoz JM, Celaya AM, Hijazo‐Pechero S, Wang J, Serrano M, Varela‐Nieto I. G6PD overexpression protects from oxidative stress and age-related hearing loss. Aging Cell 2020; 19:e13275. [PMID: 33222382 PMCID: PMC7744953 DOI: 10.1111/acel.13275] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 09/03/2020] [Accepted: 10/18/2020] [Indexed: 12/17/2022] Open
Abstract
Aging of the auditory system is associated with the incremental production of reactive oxygen species (ROS) and the accumulation of oxidative damage in macromolecules, which contributes to cellular malfunction, compromises cell viability, and, ultimately, leads to functional decline. Cellular detoxification relies in part on the production of NADPH, which is an important cofactor for major cellular antioxidant systems. NADPH is produced principally by the housekeeping enzyme glucose‐6‐phosphate dehydrogenase (G6PD), which catalyzes the rate‐limiting step in the pentose phosphate pathway. We show here that G6PD transgenic mice (G6PD‐Tg), which show enhanced constitutive G6PD activity and NADPH production along life, have lower auditory thresholds than wild‐type mice during aging, together with preserved inner hair cell (IHC) and outer hair cell (OHC), OHC innervation, and a conserved number of synapses per IHC. Gene expression of antioxidant enzymes was higher in 3‐month‐old G6PD‐Tg mice than in wild‐type counterparts, whereas the levels of pro‐apoptotic proteins were lower. Consequently, nitration of proteins, mitochondrial damage, and TUNEL+ apoptotic cells were all lower in 9‐month‐old G6PD‐Tg than in wild‐type counterparts. Unexpectedly, G6PD overexpression triggered low‐grade inflammation that was effectively resolved in young mice, as shown by the absence of cochlear cellular damage and macrophage infiltration. Our results lead us to propose that NADPH overproduction from an early stage is an efficient mechanism to maintain the balance between the production of ROS and cellular detoxification power along aging and thus prevents hearing loss progression.
Collapse
Affiliation(s)
- Jose M. Bermúdez‐Muñoz
- Institute for Biomedical Research “Alberto Sols” (IIBM) Spanish National Research Council‐Autonomous University of Madrid (CSIC‐UAM Madrid Spain
- Rare Diseases Networking Biomedical Research Centre (CIBERER) CIBER Carlos III Institute of Health Madrid Spain
| | - Adelaida M. Celaya
- Institute for Biomedical Research “Alberto Sols” (IIBM) Spanish National Research Council‐Autonomous University of Madrid (CSIC‐UAM Madrid Spain
- Rare Diseases Networking Biomedical Research Centre (CIBERER) CIBER Carlos III Institute of Health Madrid Spain
| | - Sara Hijazo‐Pechero
- Institute for Biomedical Research “Alberto Sols” (IIBM) Spanish National Research Council‐Autonomous University of Madrid (CSIC‐UAM Madrid Spain
| | - Jing Wang
- INSERM ‐ UMR 1051 Institut des Neurosciences de Montpellier Montpellier France
| | - Manuel Serrano
- Institute for Research in Biomedicine (IRB) Barcelona Spain
| | - Isabel Varela‐Nieto
- Institute for Biomedical Research “Alberto Sols” (IIBM) Spanish National Research Council‐Autonomous University of Madrid (CSIC‐UAM Madrid Spain
- Rare Diseases Networking Biomedical Research Centre (CIBERER) CIBER Carlos III Institute of Health Madrid Spain
- Hospital La Paz Institute for Health Research (IdiPAZ) Madrid Spain
| |
Collapse
|
12
|
Gao L, Kita T, Katsuno T, Yamamoto N, Omori K, Nakagawa T. Insulin-Like Growth Factor 1 on the Maintenance of Ribbon Synapses in Mouse Cochlear Explant Cultures. Front Cell Neurosci 2020; 14:571155. [PMID: 33132846 PMCID: PMC7579230 DOI: 10.3389/fncel.2020.571155] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/27/2020] [Indexed: 01/31/2023] Open
Abstract
Hearing loss has become one of the most common disabilities worldwide. The synaptic connections between inner hair cells (IHCs) and spiral ganglion neurons have specialized synaptic constructions, termed ribbon synapses, which are important for auditory function. The ribbon synapses in the cochlea are quite vulnerable to various insults. As such, the maintenance of ribbon synapses is important for ensuring hearing function. Insulin-like growth factor 1 (IGF1) plays a critical role in the development and maintenance of the cochlea and has the potential to protect cochlear hair cells from various insults. In this study, we examined the role of IGF1 in the maintenance of ribbon synapses in cochlear explants of postnatal day four mice. We cultured cochlear explants with an IGF1 receptor antagonist, JB1, which is an IGF1 peptide analog. Results showed that exposure to JB1 for 24 h resulted in the loss of ribbon synapses. After an additional 24-h culture without JB1, the number of ribbon synapses spontaneously recovered. The application of exogenous IGF1 showed two different aspects of ribbon synapses. Low doses of exogenous IGF1 promoted the recovery of ribbon synapses, while it compromised the spontaneous recovery of ribbon synapses at high doses. Altogether, these results indicate that the paracrine or autocrine release of IGF1 in the cochlea plays a crucial role in the maintenance of cochlear ribbon synapses.
Collapse
Affiliation(s)
- Li Gao
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomoko Kita
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tatsuya Katsuno
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Norio Yamamoto
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koichi Omori
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takayuki Nakagawa
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
13
|
Kempfle JS, Luu NNC, Petrillo M, Al-Asad R, Zhang A, Edge ASB. Lin28 reprograms inner ear glia to a neuronal fate. Stem Cells 2020; 38:890-903. [PMID: 32246510 PMCID: PMC10908373 DOI: 10.1002/stem.3181] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 02/05/2020] [Accepted: 02/08/2020] [Indexed: 12/16/2022]
Abstract
Sensorineural hearing loss is irreversible and can be caused by loss of auditory neurons. Regeneration of neural cells from endogenous cells may offer a future tool to restore the auditory circuit and to enhance the performance of implantable hearing devices. Neurons and glial cells in the peripheral nervous system are closely related and originate from a common progenitor. Prior work in our lab indicated that in the early postnatal mouse inner ear, proteolipid protein 1 (Plp1) expressing glial cells could act as progenitor cells for neurons in vitro. Here, we used a transgenic mouse model to transiently overexpress Lin28, a neural stem cell regulator, in Plp1-positive glial cells. Lin28 promoted proliferation and conversion of auditory glial cells into neurons in vitro. To study the effects of Lin28 on endogenous glial cells after loss of auditory neurons in vivo, we produced a model of auditory neuropathy by selectively damaging auditory neurons with ouabain. After neural damage was confirmed by the auditory brainstem response, we briefly upregulated the Lin28 in Plp1-expressing inner ear glial cells. One month later, we analyzed the cochlea for neural marker expression by quantitative RT-PCR and immunohistochemistry. We found that transient Lin28 overexpression in Plp1-expressing glial cells induced expression of neural stem cell markers and subsequent conversion into neurons. This suggests the potential for inner ear glia to be converted into neurons as a regeneration therapy for neural replacement in auditory neuropathy.
Collapse
Affiliation(s)
- Judith S. Kempfle
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts
- University Department of Otolaryngology, Head and Neck Surgery, Tübingen, Germany
| | - Ngoc-Nhi C. Luu
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts
- University Department of Otolaryngology, Head and Neck Surgery, Zürich, Switzerland
| | - Marco Petrillo
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts
| | - Reef Al-Asad
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts
| | - Andrea Zhang
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts
| | - Albert S. B. Edge
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts
- Program in Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, Massachusetts
- Harvard Stem Cell Institute, Cambridge, Massachusetts
| |
Collapse
|
14
|
Forouzanfar F, Asgharzade S. MicroRNAs in Noise-Induced Hearing Loss and their Regulation by Oxidative Stress and Inflammation. Curr Drug Targets 2020; 21:1216-1224. [PMID: 32538724 DOI: 10.2174/1389450121666200615145552] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 01/17/2020] [Accepted: 01/20/2020] [Indexed: 12/20/2022]
Abstract
Noise exposure (NE) has been recognized as one of the causes of sensorineural hearing loss (SNHL), which can bring about irreversible damage to sensory hair cells in the cochlea, through the launch of oxidative stress pathways and inflammation. Accordingly, determining the molecular mechanism involved in regulating hair cell apoptosis via NE is essential to prevent hair cell damage. However, the role of microRNAs (miRNAs) in the degeneration of sensory cells of the cochlea during NE has not been so far uncovered. Thus, the main purpose of this study was to demonstrate the regulatory role of miRNAs in the oxidative stress pathway and inflammation induced by NE. In this respect, articles related to noise-induced hearing loss (NIHL), oxidative stress, inflammation, and miRNA from various databases of Directory of Open Access Journals (DOAJ), Google Scholar, PubMed; Library, Information Science & Technology Abstracts (LISTA), and Web of Science were searched and retrieved. The findings revealed that several studies had suggested that up-regulation of miR-1229-5p, miR-451a, 185-5p, 186 and down-regulation of miRNA-96/182/183 and miR-30b were involved in oxidative stress and inflammation which could be used as biomarkers for NIHL. There was also a close relationship between NIHL and miRNAs, but further research is required to prove a causal association between miRNA alterations and NE, and also to determine miRNAs as biomarkers indicating responses to NE.
Collapse
Affiliation(s)
- Fatemeh Forouzanfar
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samira Asgharzade
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
15
|
Partearroyo T, Murillo-Cuesta S, Vallecillo N, Bermúdez-Muñoz JM, Rodríguez-de la Rosa L, Mandruzzato G, Celaya AM, Zeisel SH, Pajares MA, Varela-Moreiras G, Varela-Nieto I. Betaine-homocysteine S-methyltransferase deficiency causes increased susceptibility to noise-induced hearing loss associated with plasma hyperhomocysteinemia. FASEB J 2019; 33:5942-5956. [PMID: 30753104 PMCID: PMC6463923 DOI: 10.1096/fj.201801533r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 01/15/2019] [Indexed: 12/16/2022]
Abstract
Betaine-homocysteine S-methyltransferases (BHMTs) are methionine cycle enzymes that remethylate homocysteine; hence, their malfunction leads to hyperhomocysteinemia. Epidemiologic and experimental studies have revealed a correlation between hyperhomocysteinemia and hearing loss. Here, we have studied the expression of methionine cycle genes in the mouse cochlea and the impact of knocking out the Bhmt gene in the auditory receptor. We evaluated age-related changes in mouse hearing by recording auditory brainstem responses before and following exposure to noise. Also, we measured cochlear cytoarchitecture, gene expression by RNA-arrays and quantitative RT-PCR, and metabolite levels in liver and plasma by HPLC. Our results indicate that there is an age-dependent strain-specific expression of methionine cycle genes in the mouse cochlea and a further regulation during the response to noise damage. Loss of Bhmt did not cause an evident impact in the hearing acuity of young mice, but it produced higher threshold shifts and poorer recovery following noise challenge. Hearing loss was associated with increased cochlear injury, outer hair cell loss, altered expression of cochlear methionine cycle genes, and hyperhomocysteinemia. Our results suggest that BHMT plays a central role in the homeostasis of cochlear methionine metabolism and that Bhmt2 up-regulation could carry out a compensatory role in cochlear protection against noise injury in the absence of BHMT.-Partearroyo, T., Murillo-Cuesta, S., Vallecillo, N., Bermúdez-Muñoz, J. M., Rodríguez-de la Rosa, L., Mandruzzato, G., Celaya, A. M., Zeisel, S. H., Pajares, M. A., Varela-Moreiras, G., Varela-Nieto, I. Betaine-homocysteine S-methyltransferase deficiency causes increased susceptibility to noise-induced hearing loss associated with plasma hyperhomocysteinemia.
Collapse
Affiliation(s)
- Teresa Partearroyo
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad Centro de Estudios Universitarios CEU San Pablo, Madrid, Spain
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas–Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | - Silvia Murillo-Cuesta
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas–Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Sanitaria La Paz (IdiPAZ), Madrid, Spain
| | - Néstor Vallecillo
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas–Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | - Jose M. Bermúdez-Muñoz
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas–Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Lourdes Rodríguez-de la Rosa
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas–Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Sanitaria La Paz (IdiPAZ), Madrid, Spain
| | | | - Adelaida M. Celaya
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas–Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Steven H. Zeisel
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina, USA; and
| | - María A. Pajares
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas–Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Instituto de Investigación Sanitaria La Paz (IdiPAZ), Madrid, Spain
- Centro de Investigaciones Biológicas, (CSIC) Madrid, Spain
| | - Gregorio Varela-Moreiras
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad Centro de Estudios Universitarios CEU San Pablo, Madrid, Spain
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas–Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | - Isabel Varela-Nieto
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas–Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Sanitaria La Paz (IdiPAZ), Madrid, Spain
| |
Collapse
|
16
|
Gibaja A, Aburto MR, Pulido S, Collado M, Hurle JM, Varela-Nieto I, Magariños M. TGFβ2-induced senescence during early inner ear development. Sci Rep 2019; 9:5912. [PMID: 30976015 PMCID: PMC6459823 DOI: 10.1038/s41598-019-42040-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 03/21/2019] [Indexed: 01/16/2023] Open
Abstract
Embryonic development requires the coordinated regulation of apoptosis, survival, autophagy, proliferation and differentiation programs. Senescence has recently joined the cellular processes required to master development, in addition to its well-described roles in cancer and ageing. Here, we show that senescent cells are present in a highly regulated temporal pattern in the developing vertebrate inner ear, first, surrounding the otic pore and, later, in the otocyst at the endolymphatic duct. Cellular senescence is associated with areas of increased apoptosis and reduced proliferation consistent with the induction of the process when the endolymphatic duct is being formed. Modulation of senescence disrupts otic vesicle morphology. Transforming growth factor beta (TGFβ) signaling interacts with signaling pathways elicited by insulin-like growth factor type 1 (IGF-1) to jointly coordinate cellular dynamics required for morphogenesis and differentiation. Taken together, these results show that senescence is a natural occurring process essential for early inner ear development.
Collapse
Affiliation(s)
- Alejandro Gibaja
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid, Spain
| | - María R Aburto
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid, Spain
| | - Sara Pulido
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid, Spain.,CIBERER, Instituto de Salud Carlos III, Madrid, Spain
| | - Manuel Collado
- Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Juan M Hurle
- Departamento de Anatomía y Biología Celular and IDIVAL, Universidad de Cantabria, Santander, Spain
| | - Isabel Varela-Nieto
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid, Spain.,CIBERER, Instituto de Salud Carlos III, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Marta Magariños
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid, Spain. .,CIBERER, Instituto de Salud Carlos III, Madrid, Spain. .,Departamento de Biología, Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|
17
|
Celaya AM, Sánchez-Pérez I, Bermúdez-Muñoz JM, Rodríguez-de la Rosa L, Pintado-Berninches L, Perona R, Murillo-Cuesta S, Varela-Nieto I. Deficit of mitogen-activated protein kinase phosphatase 1 (DUSP1) accelerates progressive hearing loss. eLife 2019; 8:39159. [PMID: 30938680 PMCID: PMC6464786 DOI: 10.7554/elife.39159] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 04/01/2019] [Indexed: 12/12/2022] Open
Abstract
Mitogen-activated protein kinases (MAPK) such as p38 and the c-Jun N-terminal kinases (JNKs) are activated during the cellular response to stress signals. Their activity is regulated by the MAPK-phosphatase 1 (DUSP1), a key component of the anti-inflammatory response. Stress kinases are well-described elements of the response to otic injury and the otoprotective potential of JNK inhibitors is being tested in clinical trials. By contrast, there are no studies exploring the role of DUSP1 in hearing and hearing loss. Here we show that Dusp1 expression is age-regulated in the mouse cochlea. Dusp1 gene knock-out caused premature progressive hearing loss, as confirmed by auditory evoked responses in Dusp1-/- mice. Hearing loss correlated with cell death in hair cells, degeneration of spiral neurons and increased macrophage infiltration. Dusp1-/- mouse cochleae showed imbalanced redox status and dysregulated expression of cytokines. These data suggest that DUSP1 is essential for cochlear homeostasis in the response to stress during ageing.
Collapse
Affiliation(s)
- Adelaida M Celaya
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain.,Centre for Biomedical Network Research on Rare Diseases (CIBERER), CIBER, ISCIII, Madrid, Spain
| | - Isabel Sánchez-Pérez
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain.,Centre for Biomedical Network Research on Rare Diseases (CIBERER), CIBER, ISCIII, Madrid, Spain.,Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain.,Biochemistry Department, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain.,Biomedicine Unit UCLM-CSIC, Madrid, Spain
| | - Jose M Bermúdez-Muñoz
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain.,Centre for Biomedical Network Research on Rare Diseases (CIBERER), CIBER, ISCIII, Madrid, Spain
| | - Lourdes Rodríguez-de la Rosa
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain.,Centre for Biomedical Network Research on Rare Diseases (CIBERER), CIBER, ISCIII, Madrid, Spain.,Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Laura Pintado-Berninches
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain.,Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Rosario Perona
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain.,Centre for Biomedical Network Research on Rare Diseases (CIBERER), CIBER, ISCIII, Madrid, Spain.,Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Silvia Murillo-Cuesta
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain.,Centre for Biomedical Network Research on Rare Diseases (CIBERER), CIBER, ISCIII, Madrid, Spain
| | - Isabel Varela-Nieto
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain.,Centre for Biomedical Network Research on Rare Diseases (CIBERER), CIBER, ISCIII, Madrid, Spain
| |
Collapse
|
18
|
Fuentes-Santamaría V, Alvarado JC, Rodríguez-de la Rosa L, Juiz JM, Varela-Nieto I. Neuroglial Involvement in Abnormal Glutamate Transport in the Cochlear Nuclei of the Igf1 -/- Mouse. Front Cell Neurosci 2019; 13:67. [PMID: 30881288 PMCID: PMC6405628 DOI: 10.3389/fncel.2019.00067] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 02/11/2019] [Indexed: 12/25/2022] Open
Abstract
Insulin-like growth factor 1 (IGF-1) is a powerful regulator of synaptic activity and a deficit in this protein has a profound impact on neurotransmission, mostly on excitatory synapses in both the developing and mature auditory system. Adult Igf1−/− mice are animal models for the study of human syndromic deafness; they show altered cochlear projection patterns into abnormally developed auditory neurons along with impaired glutamate uptake in the cochlear nuclei, phenomena that probably reflect disruptions in neuronal circuits. To determine the cellular mechanisms that might be involved in regulating excitatory synaptic plasticity in 4-month-old Igf1−/− mice, modifications to neuroglia, astroglial glutamate transporters (GLTs) and metabotropic glutamate receptors (mGluRs) were assessed in the cochlear nuclei. The Igf1−/− mice show significant decreases in IBA1 (an ionized calcium-binding adapter) and glial fibrillary acidic protein (GFAP) mRNA expression and protein accumulation, as well as dampened mGluR expression in conjunction with enhanced glutamate transporter 1 (GLT1) expression. By contrast, no differences were observed in the expression of glutamate aspartate transporter (GLAST) between these Igf1−/− mice and their heterozygous or wildtype littermates. These observations suggest that congenital IGF-1 deficiency may lead to alterations in microglia and astrocytes, an upregulation of GLT1, and the downregulation of groups I, II and III mGluRs. Understanding the molecular, biochemical and morphological mechanisms underlying neuronal plasticity in a mouse model of hearing deficits will give us insight into new therapeutic strategies that could help to maintain or even improve residual hearing when human deafness is related to IGF-1 deficiency.
Collapse
Affiliation(s)
- Veronica Fuentes-Santamaría
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Juan C Alvarado
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Lourdes Rodríguez-de la Rosa
- Grupo de Neurobiología de la Audición, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), CIBER MP, Instituto de Salud Carlos III, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - José M Juiz
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Isabel Varela-Nieto
- Grupo de Neurobiología de la Audición, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), CIBER MP, Instituto de Salud Carlos III, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| |
Collapse
|
19
|
Nonomura Y, Sawamura S, Hanzawa K, Nishikaze T, Sekiya S, Higuchi T, Nin F, Uetsuka S, Inohara H, Okuda S, Miyoshi E, Horii A, Takahashi S, Natsuka S, Hibino H. Characterisation of N-glycans in the epithelial-like tissue of the rat cochlea. Sci Rep 2019; 9:1551. [PMID: 30733536 PMCID: PMC6367448 DOI: 10.1038/s41598-018-38079-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 12/17/2018] [Indexed: 01/08/2023] Open
Abstract
Membrane proteins (such as ion channels, transporters, and receptors) and secreted proteins are essential for cellular activities. N-linked glycosylation is involved in stability and function of these proteins and occurs at Asn residues. In several organs, profiles of N-glycans have been determined by comprehensive analyses. Nevertheless, the cochlea of the mammalian inner ear, a tiny organ mediating hearing, has yet to be examined. Here, we focused on the stria vascularis, an epithelial-like tissue in the cochlea, and characterised N-glycans by liquid chromatography with mass spectrometry. This hypervascular tissue not only expresses several ion transporters and channels to control the electrochemical balance in the cochlea but also harbours different transporters and receptors that maintain structure and activity of the organ. Seventy-nine N-linked glycans were identified in the rat stria vascularis. Among these, in 55 glycans, the complete structures were determined; in the other 24 species, partial glycosidic linkage patterns and full profiles of the monosaccharide composition were identified. In the process of characterisation, several sialylated glycans were subjected sequentially to two different alkylamidation reactions; this derivatisation helped to distinguish α2,3-linkage and α2,6-linkage sialyl isomers with mass spectrometry. These data should accelerate elucidation of the molecular architecture of the cochlea.
Collapse
Affiliation(s)
- Yoriko Nonomura
- Department of Molecular Physiology, Niigata University School of Medicine, Niigata, Japan
- Department of Otorhinolaryngology-Head and Neck Surgery, Niigata University School of Medicine, Niigata, Japan
| | - Seishiro Sawamura
- Department of Molecular Physiology, Niigata University School of Medicine, Niigata, Japan
| | - Ken Hanzawa
- Department of Biology, Faculty of Science, Niigata University, Niigata, Japan
| | - Takashi Nishikaze
- Koichi Tanaka Mass Spectrometry Research Laboratory, Shimadzu Corporation, Kyoto, Japan
| | - Sadanori Sekiya
- Koichi Tanaka Mass Spectrometry Research Laboratory, Shimadzu Corporation, Kyoto, Japan
| | - Taiga Higuchi
- Department of Molecular Physiology, Niigata University School of Medicine, Niigata, Japan
| | - Fumiaki Nin
- Department of Molecular Physiology, Niigata University School of Medicine, Niigata, Japan
- Center for Transdisciplinary Research, Niigata University, Niigata, Japan
| | - Satoru Uetsuka
- Department of Otorhinolaryngology-Head and Neck Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hidenori Inohara
- Department of Otorhinolaryngology-Head and Neck Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shujiro Okuda
- Bioinformatics Laboratory, Niigata University School of Medicine, Niigata, Japan
| | - Eiji Miyoshi
- Division of Health Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Arata Horii
- Department of Otorhinolaryngology-Head and Neck Surgery, Niigata University School of Medicine, Niigata, Japan
| | - Sugata Takahashi
- Department of Otorhinolaryngology-Head and Neck Surgery, Niigata University School of Medicine, Niigata, Japan
| | - Shunji Natsuka
- Department of Biology, Faculty of Science, Niigata University, Niigata, Japan
| | - Hiroshi Hibino
- Department of Molecular Physiology, Niigata University School of Medicine, Niigata, Japan.
- Center for Transdisciplinary Research, Niigata University, Niigata, Japan.
- AMED-CREST, AMED, Niigata, Japan.
| |
Collapse
|
20
|
Puga AM, Pajares MA, Varela-Moreiras G, Partearroyo T. Interplay between Nutrition and Hearing Loss: State of Art. Nutrients 2018; 11:35. [PMID: 30586880 PMCID: PMC6356655 DOI: 10.3390/nu11010035] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/17/2018] [Accepted: 12/21/2018] [Indexed: 12/14/2022] Open
Abstract
Hearing loss has been recently ranked as the fifth leading cause of years lived with disability, ahead of many other chronic diseases such as diabetes, dementia, or chronic obstructive pulmonary disease. Moreover, according to the World Health Organization, moderate-to-profound hearing loss affects about 466 million people worldwide. Its incidence varies in each population segment, affecting approximately 10% of children and increasing to 30% of the population over 65 years. However, hearing loss receives still very limited research funding and public awareness. This sensory impairment is caused by genetic and environmental factors, and among the latter, the nutritional status has acquired relevance due its association to hearing loss detected in recent epidemiological studies. Several experimental models have proved that the onset and progression of hearing loss are closely linked to the availability of nutrients and their metabolism. Here, we have reviewed studies focused on nutrient effects on auditory function. These studies support the potential of nutritional therapy for the protection against hearing loss progression, which is especially relevant to the aging process and related quality of life.
Collapse
Affiliation(s)
- Ana M Puga
- Department of Pharmaceutical and Health Sciences, Faculty of Pharmacy, CEU San Pablo University, 28668 Madrid, Spain.
| | - María A Pajares
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas (CSIC), 28040 Madrid, Spain.
- Molecular Hepatology Group, Hospital La Paz Institute for Health Research (IdiPAZ), 28046 Madrid, Spain.
| | - Gregorio Varela-Moreiras
- Department of Pharmaceutical and Health Sciences, Faculty of Pharmacy, CEU San Pablo University, 28668 Madrid, Spain.
| | - Teresa Partearroyo
- Department of Pharmaceutical and Health Sciences, Faculty of Pharmacy, CEU San Pablo University, 28668 Madrid, Spain.
| |
Collapse
|
21
|
Booth KT, Azaiez H, Jahan I, Smith RJH, Fritzsch B. Intracellular Regulome Variability Along the Organ of Corti: Evidence, Approaches, Challenges, and Perspective. Front Genet 2018; 9:156. [PMID: 29868110 PMCID: PMC5951964 DOI: 10.3389/fgene.2018.00156] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/13/2018] [Indexed: 12/13/2022] Open
Abstract
The mammalian hearing organ is a regular array of two types of hair cells (HCs) surrounded by six types of supporting cells. Along the tonotopic axis, this conserved radial array of cell types shows longitudinal variations to enhance the tuning properties of basilar membrane. We present the current evidence supporting the hypothesis that quantitative local variations in gene expression profiles are responsible for local cell responses to global gene manipulations. With the advent of next generation sequencing and the unprecedented array of technologies offering high throughput analyses at the single cell level, transcriptomics will become a common tool to enhance our understanding of the inner ear. We provide an overview of the approaches and landmark studies undertaken to date to analyze single cell variations in the organ of Corti and discuss the current limitations. We next provide an overview of the complexity of known regulatory mechanisms in the inner ear. These mechanisms are tightly regulated temporally and spatially at the transcription, RNA-splicing, mRNA-regulation, and translation levels. Understanding the intricacies of regulatory mechanisms at play in the inner ear will require the use of complementary approaches, and most probably, a combinatorial strategy coupling transcriptomics, proteomics, and epigenomics technologies. We highlight how these data, in conjunction with recent insights into molecular cell transformation, can advance attempts to restore lost hair cells.
Collapse
Affiliation(s)
- Kevin T Booth
- Molecular Otolaryngology and Renal Research Laboratories, Department of Otolaryngology, University of Iowa, Iowa City, IA, United States.,Interdisciplinary Graduate Program in Molecular Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Hela Azaiez
- Molecular Otolaryngology and Renal Research Laboratories, Department of Otolaryngology, University of Iowa, Iowa City, IA, United States
| | - Israt Jahan
- Department of Biology, University of Iowa, Iowa City, IA, United States
| | - Richard J H Smith
- Molecular Otolaryngology and Renal Research Laboratories, Department of Otolaryngology, University of Iowa, Iowa City, IA, United States
| | - Bernd Fritzsch
- Molecular Otolaryngology and Renal Research Laboratories, Department of Otolaryngology, University of Iowa, Iowa City, IA, United States.,Department of Biology, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
22
|
Saavedra-García P, Nichols K, Mahmud Z, Fan LYN, Lam EWF. Unravelling the role of fatty acid metabolism in cancer through the FOXO3-FOXM1 axis. Mol Cell Endocrinol 2018; 462:82-92. [PMID: 28087388 DOI: 10.1016/j.mce.2017.01.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 12/06/2016] [Accepted: 01/09/2017] [Indexed: 02/07/2023]
Abstract
Obesity and cachexia represent divergent states of nutritional and metabolic imbalance but both are intimately linked to cancer. There is an extensive overlap in their signalling pathways and molecular components involved such as fatty acids (FAs), which likely play a crucial role in cancer. Forkhead box (FOX) proteins are responsible of a wide range of transcriptional programmes during normal development, and the FOXO3-FOXM1 axis is associated with cancer initiation, progression and drug resistance. Free fatty acids (FFAs), FA synthesis and β-oxidation are associated with cancer development and progression. Meanwhile, insulin and some adipokines, that are up-regulated by FAs, are also involved in cancer development and poor prognosis. In this review, we discuss the role of FA metabolism in cancer and how FA metabolism integrates with the FOXO3-FOXM1 axis. These new insights may provide leads to better cancer diagnostics as well as strategies for tackling cancer development, progression and drug resistance.
Collapse
Affiliation(s)
- Paula Saavedra-García
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK
| | - Katie Nichols
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK
| | - Zimam Mahmud
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK
| | - Lavender Yuen-Nam Fan
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK
| | - Eric W-F Lam
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK.
| |
Collapse
|
23
|
Hu W, Wu J, Jiang W, Tang J. MicroRNAs and Presbycusis. Aging Dis 2018; 9:133-142. [PMID: 29392088 PMCID: PMC5772851 DOI: 10.14336/ad.2017.0119] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/19/2017] [Indexed: 12/19/2022] Open
Abstract
Presbycusis (age-related hearing loss) is the most universal sensory degenerative disease in elderly people caused by the degeneration of cochlear cells. Non-coding microRNAs (miRNAs) play a fundamental role in gene regulation in almost every multicellular organism, and control the aging processes. It has been identified that various miRNAs are up- or down-regulated during mammalian aging processes in tissue-specific manners. Most miRNAs bind to specific sites on their target messenger-RNAs (mRNAs) and decrease their expression. Germline mutation may lead to dysregulation of potential miRNAs expression, causing progressive hair cell degeneration and age-related hearing loss. Therapeutic innovations could emerge from a better understanding of diverse function of miRNAs in presbycusis. This review summarizes the relationship between miRNAs and presbycusis, and presents novel miRNAs-targeted strategies against presbycusis.
Collapse
Affiliation(s)
- Weiming Hu
- 1Department of Otolaryngology-Head and Neck Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Junwu Wu
- 2Department of Otolaryngology-Head and Neck Surgery, Yiwu traditional Chinese Medicine Hospital, Yiwu 322000, China.,3Department of Otolaryngology-Head and Neck Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou 310016, China
| | - Wenjing Jiang
- 1Department of Otolaryngology-Head and Neck Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Jianguo Tang
- 3Department of Otolaryngology-Head and Neck Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou 310016, China
| |
Collapse
|
24
|
Espino Guarch M, Font-Llitjós M, Murillo-Cuesta S, Errasti-Murugarren E, Celaya AM, Girotto G, Vuckovic D, Mezzavilla M, Vilches C, Bodoy S, Sahún I, González L, Prat E, Zorzano A, Dierssen M, Varela-Nieto I, Gasparini P, Palacín M, Nunes V. Mutations in L-type amino acid transporter-2 support SLC7A8 as a novel gene involved in age-related hearing loss. eLife 2018; 7:31511. [PMID: 29355479 PMCID: PMC5811215 DOI: 10.7554/elife.31511] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 01/18/2018] [Indexed: 12/19/2022] Open
Abstract
Age-related hearing loss (ARHL) is the most common sensory deficit in the elderly. The disease has a multifactorial etiology with both environmental and genetic factors involved being largely unknown. SLC7A8/SLC3A2 heterodimer is a neutral amino acid exchanger. Here, we demonstrated that SLC7A8 is expressed in the mouse inner ear and that its ablation resulted in ARHL, due to the damage of different cochlear structures. These findings make SLC7A8 transporter a strong candidate for ARHL in humans. Thus, a screening of a cohort of ARHL patients and controls was carried out revealing several variants in SLC7A8, whose role was further investigated by in vitro functional studies. Significant decreases in SLC7A8 transport activity was detected for patient’s variants (p.Val302Ile, p.Arg418His, p.Thr402Met and p.Val460Glu) further supporting a causative role for SLC7A8 in ARHL. Moreover, our preliminary data suggest that a relevant proportion of ARHL cases could be explained by SLC7A8 mutations. Age-related hearing loss affects about one in three individuals between the ages of 65 and 74. The first symptom is difficulty hearing high-pitched sounds like children’s voices. The disease starts gradually and worsens over time. Changes in the ear, the nerve that connects it to the brain, or the brain itself can cause hearing loss. Sometimes all three play a role. Genetics, exposure to noise, disease, and aging may all contribute. The condition is so complex it is difficult for scientists to pinpoint a primary suspect or develop treatments. Now, Guarch, Font-Llitjós et al. show that errors in a protein called SLC7A8 cause age-related hearing loss in mice and humans. The SLC7A8 protein acts like a door that allows amino acids – the building blocks of proteins – to enter or leave a cell. This door is blocked in mice lacking SLC7A8 and damage occurs in the part of their inner ear responsible for hearing. As a result, the animals lose their hearing. Next, Guarch, Font-Llitjós et al. scanned the genomes of 147 people from isolated villages in Italy for mutations in the gene for SLC7A8. The people also underwent hearing tests. Mutations in the gene for SLC7A8 that partially block the door and prevent the flow of amino acids were found in people with hearing loss. Some mutations in SLC7A8 that allow the door to stay open where found in people who could hear. The experiments suggest that certain mutations in the gene for SLC7A8 are likely an inherited cause of age-related hearing loss. It is possible that other proteins that control the flow of amino acids into or out of cells also may play a role in hearing. More studies are needed to see if it is possible to fix errors in the SLC7A8 protein to delay or restore the hearing loss.
Collapse
Affiliation(s)
- Meritxell Espino Guarch
- Experimental Genetics, Sidra Medical and Research Center, Doha, Qatar.,Genes, Disease and Therapy Program, Molecular Genetics Laboratory - IDIBELL, Barcelona, Spain.,Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Mariona Font-Llitjós
- Genes, Disease and Therapy Program, Molecular Genetics Laboratory - IDIBELL, Barcelona, Spain.,Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain
| | - Silvia Murillo-Cuesta
- Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain.,Alberto Sols Biomedical Research Institute (CSIC/UAM), Madrid, Spain.,Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Ekaitz Errasti-Murugarren
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain
| | - Adelaida M Celaya
- Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain.,Alberto Sols Biomedical Research Institute (CSIC/UAM), Madrid, Spain
| | - Giorgia Girotto
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy.,Medical Genetics, Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| | - Dragana Vuckovic
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy.,Medical Genetics, Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| | | | - Clara Vilches
- Genes, Disease and Therapy Program, Molecular Genetics Laboratory - IDIBELL, Barcelona, Spain
| | - Susanna Bodoy
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain
| | - Ignasi Sahún
- Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain.,Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Laura González
- Genes, Disease and Therapy Program, Molecular Genetics Laboratory - IDIBELL, Barcelona, Spain.,Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain
| | - Esther Prat
- Genes, Disease and Therapy Program, Molecular Genetics Laboratory - IDIBELL, Barcelona, Spain.,Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain.,Genetics Section, Physiological Sciences Department, Health Sciences and Medicine Faculty, University of Barcelona, Barcelona, Spain
| | - Antonio Zorzano
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Biochemistry and Molecular Biomedicine Department, Faculty of Biology, University of Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre on Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
| | - Mara Dierssen
- Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain.,Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Isabel Varela-Nieto
- Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain.,Alberto Sols Biomedical Research Institute (CSIC/UAM), Madrid, Spain.,Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Paolo Gasparini
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy.,Medical Genetics, Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| | - Manuel Palacín
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain.,Biochemistry and Molecular Biomedicine Department, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Virginia Nunes
- Genes, Disease and Therapy Program, Molecular Genetics Laboratory - IDIBELL, Barcelona, Spain.,Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain.,Genetics Section, Physiological Sciences Department, Health Sciences and Medicine Faculty, University of Barcelona, Barcelona, Spain
| |
Collapse
|
25
|
García-Alcántara F, Murillo-Cuesta S, Pulido S, Bermúdez-Muñoz JM, Martínez-Vega R, Milo M, Varela-Nieto I, Rivera T. The expression of oxidative stress response genes is modulated by a combination of resveratrol and N-acetylcysteine to ameliorate ototoxicity in the rat cochlea. Hear Res 2017; 358:10-21. [PMID: 29304389 DOI: 10.1016/j.heares.2017.12.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 09/06/2017] [Accepted: 12/10/2017] [Indexed: 11/19/2022]
Abstract
Aminoglycoside antibiotics are used widely in medicine despite their ototoxic side-effects. Oxidative stress and inflammation are key mechanisms determining the extent and severity of the damage. Here we evaluate the protective effect of a treatment with resveratrol plus N-acetylcysteine on the ototoxic actions of kanamycin and furosemide in the rat. Resveratrol (10 mg/kg) and N-acetylcysteine (400 mg/kg) were administered together to Wistar rats on 5 consecutive days. The second day, a concentrated solution of kanamycin and furosemide was placed on the round window to induce ototoxicity. Hearing was assessed by recording auditory brainstem responses before and 5, 16 and 23 days after the beginning of the treatment. Cochlear samples were taken at day 5 (end of the treatment) and at day 23, and targeted PCR arrays or RT-qPCR were performed to analyze oxidative balance and inflammation related genes, respectively. In addition, the cytoarchitecture and the presence of apoptosis, oxidative stress and inflammation markers were evaluated in cochlear sections. Results indicate that administration of resveratrol plus N-acetylcysteine reduced the threshold shifts induced by ototoxic drugs at high frequencies (≈10 dB), although this protective effect fades after the cessation of the treatment. Gene expression analysis showed that the treatment modulated the expression of genes involved in the cellular oxidative (Gpx1, Sod1, Ccs and Noxa1) and inflammatory (Il1b, Il4, Mpo and Ncf) responses to injury. Thus, co-administration of resveratrol and NAC, routinely used individually in patients, could reduce the ototoxic secondary effects of aminoglycosides.
Collapse
Affiliation(s)
- Fernando García-Alcántara
- Príncipe de Asturias University Hospital, Universidad de Alcalá, Carretera Alcalá-Meco s/n, 28805, Alcalá de Henares, Madrid, Spain; Institute of Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain; Centre for Biomedical Network Research in Rare Diseases (CIBERER), Institute of Health Carlos III (ISCIII), Monforte de Lemos, 3-5, 28029, Madrid, Spain.
| | - Silvia Murillo-Cuesta
- Institute of Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain; Centre for Biomedical Network Research in Rare Diseases (CIBERER), Institute of Health Carlos III (ISCIII), Monforte de Lemos, 3-5, 28029, Madrid, Spain; Hospital La Paz Institute for Health Research (IdiPAZ), Pedro Rico 6, 28029, Madrid, Spain.
| | - Sara Pulido
- Institute of Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain; Centre for Biomedical Network Research in Rare Diseases (CIBERER), Institute of Health Carlos III (ISCIII), Monforte de Lemos, 3-5, 28029, Madrid, Spain; Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S3 7HF, United Kingdom.
| | - Jose M Bermúdez-Muñoz
- Institute of Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain; Centre for Biomedical Network Research in Rare Diseases (CIBERER), Institute of Health Carlos III (ISCIII), Monforte de Lemos, 3-5, 28029, Madrid, Spain.
| | - Raquel Martínez-Vega
- Institute of Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain.
| | - Marta Milo
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S3 7HF, United Kingdom.
| | - Isabel Varela-Nieto
- Institute of Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain; Centre for Biomedical Network Research in Rare Diseases (CIBERER), Institute of Health Carlos III (ISCIII), Monforte de Lemos, 3-5, 28029, Madrid, Spain; Hospital La Paz Institute for Health Research (IdiPAZ), Pedro Rico 6, 28029, Madrid, Spain.
| | - Teresa Rivera
- Príncipe de Asturias University Hospital, Universidad de Alcalá, Carretera Alcalá-Meco s/n, 28805, Alcalá de Henares, Madrid, Spain; Institute of Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain; Centre for Biomedical Network Research in Rare Diseases (CIBERER), Institute of Health Carlos III (ISCIII), Monforte de Lemos, 3-5, 28029, Madrid, Spain.
| |
Collapse
|
26
|
Rodríguez-de la Rosa L, Lassaletta L, Calvino M, Murillo-Cuesta S, Varela-Nieto I. The Role of Insulin-Like Growth Factor 1 in the Progression of Age-Related Hearing Loss. Front Aging Neurosci 2017; 9:411. [PMID: 29311900 PMCID: PMC5733003 DOI: 10.3389/fnagi.2017.00411] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 11/27/2017] [Indexed: 12/21/2022] Open
Abstract
Aging is associated with impairment of sensorial functions and with the onset of neurodegenerative diseases. As pari passu circulating insulin-like growth factor 1 (IGF-1) bioavailability progressively decreases, we see a direct correlation with sensory impairment and cognitive performance in older humans. Age-related sensory loss is typically caused by the irreversible death of highly differentiated neurons and sensory receptor cells. Among sensory deficits, age-related hearing loss (ARHL), also named presbycusis, affects one third of the population over 65 years of age and is a major factor in the progression of cognitive problems in the elderly. The genetic and molecular bases of ARHL are largely unknown and only a few genes related to susceptibility to oxidative stress, excitotoxicity, and cell death have been identified. IGF-1 is known to be a neuroprotective agent that maintains cellular metabolism, activates growth, proliferation and differentiation, and limits cell death. Inborn IGF-1 deficiency leads to profound sensorineural hearing loss both in humans and mice. IGF-1 haploinsufficiency has also been shown to correlate with ARHL. There is not much information available on the effect of IGF-1 deficiency on other human sensory systems, but experimental models show a long-term impact on the retina. A secondary action of IGF-1 is the control of oxidative stress and inflammation, thus helping to resolve damage situations, acute or made chronic by aging. Here we will review the primary actions of IGF-1 in the auditory system and the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Lourdes Rodríguez-de la Rosa
- “Alberto Sols” Biomedical Research Institute CSIC-UAM, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Luis Lassaletta
- “Alberto Sols” Biomedical Research Institute CSIC-UAM, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
- Otorhinolaryngology Department, Hospital La Paz, Madrid, Spain
| | - Miryam Calvino
- Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
- Otorhinolaryngology Department, Hospital La Paz, Madrid, Spain
| | - Silvia Murillo-Cuesta
- “Alberto Sols” Biomedical Research Institute CSIC-UAM, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Isabel Varela-Nieto
- “Alberto Sols” Biomedical Research Institute CSIC-UAM, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| |
Collapse
|
27
|
Mao B, Moss CF, Wilkinson GS. Age-dependent gene expression in the inner ear of big brown bats (Eptesicus fuscus). PLoS One 2017; 12:e0186667. [PMID: 29073148 PMCID: PMC5658057 DOI: 10.1371/journal.pone.0186667] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Accepted: 10/05/2017] [Indexed: 12/25/2022] Open
Abstract
For echolocating bats, hearing is essential for survival. Specializations for detecting and processing high frequency sounds are apparent throughout their auditory systems. Recent studies on echolocating mammals have reported evidence of parallel evolution in some hearing-related genes in which distantly related groups of echolocating animals (bats and toothed whales), cluster together in gene trees due to apparent amino acid convergence. However, molecular adaptations can occur not only in coding sequences, but also in the regulation of gene expression. The aim of this study was to examine the expression of hearing-related genes in the inner ear of developing big brown bats, Eptesicus fuscus, during the period in which echolocation vocalizations increase dramatically in frequency. We found that seven genes were significantly upregulated in juveniles relative to adults, and that the expression of four genes through development correlated with estimated age. Compared to available data for mice, it appears that expression of some hearing genes is extended in juvenile bats. These results are consistent with a prolonged growth period required to develop larger cochlea relative to body size, a later maturation of high frequency hearing, and a greater dependence on high frequency hearing in echolocating bats.
Collapse
Affiliation(s)
- Beatrice Mao
- Department of Biology, College of Computer, Mathematical, and Natural Sciences, University of Maryland, College Park, Maryland, United States of America
| | - Cynthia F. Moss
- Department of Psychological and Brain Sciences, Zanvyl Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, Maryland, United States of America
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Gerald S. Wilkinson
- Department of Biology, College of Computer, Mathematical, and Natural Sciences, University of Maryland, College Park, Maryland, United States of America
| |
Collapse
|
28
|
Han N, Noyes HA, Brass A. TIGERi: modeling and visualizing the responses to perturbation of a transcription factor network. BMC Bioinformatics 2017; 18:260. [PMID: 28617232 PMCID: PMC5471961 DOI: 10.1186/s12859-017-1636-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Transcription factor (TF) networks play a key role in controlling the transfer of genetic information from gene to mRNA. Much progress has been made on understanding and reverse-engineering TF network topologies using a range of experimental and theoretical methodologies. Less work has focused on using these models to examine how TF networks respond to changes in the cellular environment. METHODS In this paper, we have developed a simple, pragmatic methodology, TIGERi (Transcription-factor-activity Illustrator for Global Explanation of Regulatory interaction), to model the response of an inferred TF network to changes in cellular environment. The methodology was tested using publicly available data comparing gene expression profiles of a mouse p38α (Mapk14) knock-out line to the original wild-type. RESULTS Using the model, we have examined changes in the TF network resulting from the presence or absence of p38α. A part of this network was confirmed by experimental work in the original paper. Additional relationships were identified by our analysis, for example between p38α and HNF3, and between p38α and SOX9, and these are strongly supported by published evidence. FXR and MYC were also discovered in our analysis as two novel links of p38α. To provide a computational methodology to the biomedical communities that has more user-friendly interface, we also developed a standalone GUI (graphical user interface) software for TIGERi and it is freely available at https://github.com/namshik/tigeri/ . CONCLUSIONS We therefore believe that our computational approach can identify new members of networks and new interactions between members that are supported by published data but have not been integrated into the existing network models. Moreover, ones who want to analyze their own data with TIGERi could use the software without any command line experience. This work could therefore accelerate researches in transcriptional gene regulation in higher eukaryotes.
Collapse
Affiliation(s)
- Namshik Han
- Gurdon Institute, University of Cambridge, Cambridge, UK. .,School of Computer Science and School of Health Sciences, University of Manchester, Manchester, UK.
| | - Harry A Noyes
- School of Biological Sciences, University of Liverpool, Liverpool, UK
| | - Andy Brass
- School of Computer Science and School of Health Sciences, University of Manchester, Manchester, UK.
| |
Collapse
|
29
|
Partearroyo T, Vallecillo N, Pajares MA, Varela-Moreiras G, Varela-Nieto I. Cochlear Homocysteine Metabolism at the Crossroad of Nutrition and Sensorineural Hearing Loss. Front Mol Neurosci 2017; 10:107. [PMID: 28487633 PMCID: PMC5403919 DOI: 10.3389/fnmol.2017.00107] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 03/30/2017] [Indexed: 12/27/2022] Open
Abstract
Hearing loss (HL) is one of the most common causes of disability, affecting 360 million people according to the World Health Organization (WHO). HL is most frequently of sensorineural origin, being caused by the irreversible loss of hair cells and/or spiral ganglion neurons. The etiology of sensorineural HL (SNHL) is multifactorial, with genetic and environmental factors such as noise, ototoxic substances and aging playing a role. The nutritional status is central in aging disability, but the interplay between nutrition and SNHL has only recently gained attention. Dietary supplementation could therefore constitute the first step for the prevention and potential repair of hearing damage before it reaches irreversibility. In this context, different epidemiological studies have shown correlations among the nutritional condition, increased total plasma homocysteine (tHcy) and SNHL. Several human genetic rare diseases are also associated with homocysteine (Hcy) metabolism and SNHL confirming this potential link. Accordingly, rodent experimental models have provided the molecular basis to understand the observed effects. Thus, increased tHcy levels and vitamin deficiencies, such as folic acid (FA), have been linked with SNHL, whereas long-term dietary supplementation with omega-3 fatty acids improved Hcy metabolism, cell survival and hearing acuity. Furthermore, pharmacological supplementations with the anti-oxidant fumaric acid that targets Hcy metabolism also improved SNHL. Overall these results strongly suggest that cochlear Hcy metabolism is a key player in the onset and progression of SNHL, opening the way for the design of prospective nutritional therapies.
Collapse
Affiliation(s)
- Teresa Partearroyo
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU San PabloMadrid, Spain
| | - Néstor Vallecillo
- Departamento de Fisiopatología y del Sistema Nervios, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC-UAM)Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos IIIMadrid, Spain
| | - María A Pajares
- Departamento de Fisiopatología y del Sistema Nervios, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC-UAM)Madrid, Spain.,Investigación en Otoneurocirugía, Instituto de Investigación Sanitaria La Paz (IdiPAZ)Madrid, Spain
| | - Gregorio Varela-Moreiras
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU San PabloMadrid, Spain
| | - Isabel Varela-Nieto
- Departamento de Fisiopatología y del Sistema Nervios, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC-UAM)Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos IIIMadrid, Spain.,Investigación en Otoneurocirugía, Instituto de Investigación Sanitaria La Paz (IdiPAZ)Madrid, Spain
| |
Collapse
|
30
|
Yamahara K, Nakagawa T, Ito J, Kinoshita K, Omori K, Yamamoto N. Netrin 1 mediates protective effects exerted by insulin-like growth factor 1 on cochlear hair cells. Neuropharmacology 2017; 119:26-39. [PMID: 28373074 DOI: 10.1016/j.neuropharm.2017.03.032] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Revised: 03/15/2017] [Accepted: 03/29/2017] [Indexed: 01/17/2023]
Abstract
Sensorineural hearing loss (SNHL) is mainly caused by the damage of cochlear hair cells (HCs). As HCs and supporting cells (SCs) do not proliferate in postnatal mammals, the loss of HCs and SCs is irreversible, emphasizing the importance of preserving their numbers to prevent SNHL. It is known that insulin-like growth factor 1 (IGF1) is instrumental in the treatment of SNHL. Our previous study indicates that IGF1 protects HCs against aminoglycoside by activating IGF1 receptor and its two major downstream pathways, PI3K/AKT and MEK/ERK, in SCs, which results in the upregulation of the expression of the Netrin1-encoding gene (Ntn1). However, the mechanisms underlying IGF1-induced protection of HCs via SC activation as well as the role of NTN1 in this process have not been elucidated. Here, we demonstrated that NTN1, similar to IGF1, promoted HC survival. NTN1 blocking antibody attenuated IGF1-induced HC protection from aminoglycoside, indicating that NTN1 is the effector molecule of IGF1 signaling during HC protection. In situ hybridization demonstrated that IGF1 potently induced Ntn1 expression in SCs. NTN1 receptors were abundantly expressed in the cochlea; among them, UNC5B mediated IGF1 protective effects on HCs, as NTN1 binding to UNC5B inhibited HC apoptosis. These results provide new insights into the mechanisms underlying IGF1 protection of cochlear HCs, suggesting a possibility of using NTN1 as a new treatment for SNHL.
Collapse
Affiliation(s)
- Kohei Yamahara
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto 606-8507, Japan
| | - Takayuki Nakagawa
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto 606-8507, Japan
| | - Juichi Ito
- Shiga Medical Center Research Institute, Moriyama, Shiga 524-8523, Japan
| | - Kazuo Kinoshita
- Shiga Medical Center Research Institute, Moriyama, Shiga 524-8523, Japan
| | - Koichi Omori
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto 606-8507, Japan
| | - Norio Yamamoto
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto 606-8507, Japan.
| |
Collapse
|
31
|
Murillo-Cuesta S, Vallecillo N, Cediel R, Celaya AM, Lassaletta L, Varela-Nieto I, Contreras J. A Comparative Study of Drug Delivery Methods Targeted to the Mouse Inner Ear: Bullostomy Versus Transtympanic Injection. J Vis Exp 2017. [PMID: 28362376 PMCID: PMC5407703 DOI: 10.3791/54951] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
We present two minimally invasive microsurgical techniques in rodents for specific drug delivery into the middle ear so that it may reach the inner ear. The first procedure consists of perforation of the tympanic bulla, termed bullostomy; the second one is a transtympanic injection. Both emulate human clinical intratympanic procedures. Chitosan-glycerophosphate (CGP) and Ringer´s Lactate buffer (RL) were used as biocompatible vehicles for local drug delivery. CGP is a nontoxic biodegradable polymer widely used in pharmaceutical applications. It is a viscous liquid at RT but it congeals to a semi solid phase at body temperature. RL is an isotonic solution used for intravenous administrations in humans. A small volume of this vehicle is precisely placed on the Round Window (RW) niche by means of a bullostomy. A transtympanic injection fills the middle ear and allows less control but broader access to the inner ear. The safety profiles of both techniques were studied and compared by using functional and morphological tests. Hearing was evaluated by registering the Auditory Brainstem Response (ABR) before and several times after microsurgery. The cytoarchitecture and preservation level of cochlear structures were studied by conventional histological techniques in paraformaldehyde-fixed and decalcified cochlear samples. In parallel, unfixed cochlear samples were taken and immediately frozen to analyze gene expression profiles of inflammatory markers by quantitative Reverse Transcriptase Polymerase Chain Reaction (qRT-PCR). Both procedures are suitable as drug delivery methods into the mouse middle ear, although transtympanic injection proved to be less invasive compared to bullostomy.
Collapse
Affiliation(s)
- Silvia Murillo-Cuesta
- Instituto de Investigaciones Biomédicas (IIBm) Alberto Sols CSIC-UAM; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII); Instituto de Investigación Sanitaria La Paz (IdiPAZ);
| | - Néstor Vallecillo
- Instituto de Investigaciones Biomédicas (IIBm) Alberto Sols CSIC-UAM
| | - Rafael Cediel
- Instituto de Investigaciones Biomédicas (IIBm) Alberto Sols CSIC-UAM; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII); Facultad de Veterinaria, Universidad Complutense de Madrid
| | - Adelaida M Celaya
- Instituto de Investigaciones Biomédicas (IIBm) Alberto Sols CSIC-UAM; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII)
| | - Luis Lassaletta
- Instituto de Investigación Sanitaria La Paz (IdiPAZ); Departmento de Otorrino laringología, Hospital Universitario La Paz
| | - Isabel Varela-Nieto
- Instituto de Investigaciones Biomédicas (IIBm) Alberto Sols CSIC-UAM; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII); Instituto de Investigación Sanitaria La Paz (IdiPAZ)
| | - Julio Contreras
- Instituto de Investigaciones Biomédicas (IIBm) Alberto Sols CSIC-UAM; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII); Facultad de Veterinaria, Universidad Complutense de Madrid
| |
Collapse
|
32
|
Martínez-Vega R, Murillo-Cuesta S, Partearroyo T, Varela-Moreiras G, Varela-Nieto I, Pajares MA. Long-Term Dietary Folate Deficiency Accelerates Progressive Hearing Loss on CBA/Ca Mice. Front Aging Neurosci 2016; 8:209. [PMID: 27630560 PMCID: PMC5006747 DOI: 10.3389/fnagi.2016.00209] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 08/18/2016] [Indexed: 01/10/2023] Open
Abstract
Dietary folic acid deficiency induced early hearing loss in C57BL/6J mice after 2-months, corroborates the epidemiological association previously described between vitamin deficiency and this sensory impairment. However, this strain is prone to early hearing loss, and hence we decided to analyze whether the effects exerted by folate deprivation follow the same pattern in a mouse strain such as CBA/Ca, which is resistant to hearing impairment. Here, we show results of a long-term study on hearing carried out on CBA/Ca mice subjected to dietary folate deprivation. Systemic changes included decreased serum folate levels, hyperhomocysteinemia and signs of anemia in the group fed with folate-deficient (FD) diet. Initial signs of hearing loss were detected in this strain after 8-months of vitamin deficiency, and correlated with histological damage in the cochleae. In conclusion, the data presented reinforce the importance of adequate folic acid levels for the auditory system and suggest that the impact of dietary deficiencies may depend on the genetic background.
Collapse
Affiliation(s)
| | - Silvia Murillo-Cuesta
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM)Madrid, Spain; Unidad 761, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos IIIMadrid, Spain; Instituto de Investigación Hospital Universitario La Paz (IdiPAZ)Madrid, Spain
| | - Teresa Partearroyo
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU San Pablo Madrid, Spain
| | - Gregorio Varela-Moreiras
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU San Pablo Madrid, Spain
| | - Isabel Varela-Nieto
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM)Madrid, Spain; Unidad 761, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos IIIMadrid, Spain; Instituto de Investigación Hospital Universitario La Paz (IdiPAZ)Madrid, Spain
| | - María A Pajares
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM)Madrid, Spain; Instituto de Investigación Hospital Universitario La Paz (IdiPAZ)Madrid, Spain
| |
Collapse
|
33
|
Arroba AI, Rodríguez-de la Rosa L, Murillo-Cuesta S, Vaquero-Villanueva L, Hurlé JM, Varela-Nieto I, Valverde ÁM. Autophagy resolves early retinal inflammation in Igf1-deficient mice. Dis Model Mech 2016; 9:965-74. [PMID: 27483352 PMCID: PMC5047685 DOI: 10.1242/dmm.026344] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 07/04/2016] [Indexed: 01/20/2023] Open
Abstract
Insulin-like growth factor-1 (IGF-1) is a growth factor with differentiating, anti-apoptotic and metabolic functions in the periphery, and anti-inflammatory properties in the nervous system. Mice that have mutations in the Igf1 gene, rendering the gene product inactive (Igf1−/−), present with age-related visual loss accompanied by structural alterations in the first synapses of the retinal pathway. Recent advances have revealed a crucial role of autophagy in immunity and inflammation. Keeping in mind this close relationship, we aimed to decipher these processes in the context of the defects that occur during ageing in the retina of Igf1−/− mice. Tnfa and Il1b mRNAs, and phosphorylation of JNK and p38 MAPK were elevated in the retinas of 6- and 12-month old Igf1−/− mice compared to those in age-matched Igf1+/+ controls. In 6-month-old Igf1−/− retinas, increased mRNA levels of the autophagy mediators Becn1, Atg9, Atg5 and Atg4, decreased p62 (also known as SQSTM1) protein expression together with an increased LC3-II:LC3-I ratio reflected active autophagic flux. However, in retinas from 12-month-old Igf1−/− mice, Nlrp3 mRNA, processing of the IL1β pro-form and immunostaining of active caspase-1 were elevated compared to those in age-matched Igf1+/+ controls, suggesting activation of the inflammasome. This effect concurred with accumulation of autophagosomes and decreased autophagic flux in the retina. Microglia localization and status of activation in the retinas of 12-month-old Igf1+/+ and Igf1−/− mice, analyzed by immunostaining of Cd11b and Iba-1, showed a specific distribution pattern in the outer plexiform layer (OPL), inner plexiform layer (IPL) and inner nuclear layer (INL), and revealed an increased number of activated microglia cells in the retina of 12-month-old blind Igf1−/− mice. Moreover, reactive gliosis was exclusively detected in the retinas from 12-month-old blind Igf1−/− mice. In conclusion, this study provides new evidence in a mouse model of IGF-1 deficiency that autophagy is an adaptive response that might confer protection against persistent inflammation in the retina during ageing. Summary:Igf1-deficient mice show chronic inflammation in the retina, and we reveal that controlling inflammation through autophagy in young mice could prevent loss of retinal function.
Collapse
Affiliation(s)
- Ana I Arroba
- Alberto Sols Biomedical Research Institute (IIBm) (CSIC/UAM), 28029, Madrid, Spain Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERdem), ISCIII, 28029, Madrid, Spain
| | - Lourdes Rodríguez-de la Rosa
- Alberto Sols Biomedical Research Institute (IIBm) (CSIC/UAM), 28029, Madrid, Spain Biomedical Research Networking Centre on Rare Diseases (CIBERER), ISCIII, 28029, Madrid, Spain IdiPAZ Institute for Health Research, Madrid 28029, Spain
| | - Silvia Murillo-Cuesta
- Alberto Sols Biomedical Research Institute (IIBm) (CSIC/UAM), 28029, Madrid, Spain Biomedical Research Networking Centre on Rare Diseases (CIBERER), ISCIII, 28029, Madrid, Spain IdiPAZ Institute for Health Research, Madrid 28029, Spain
| | | | - Juan M Hurlé
- Departamento de Anatomía y Biología Celular and IDIVAL, Universidad de Cantabria, 39011, Santander, Spain
| | - Isabel Varela-Nieto
- Alberto Sols Biomedical Research Institute (IIBm) (CSIC/UAM), 28029, Madrid, Spain Biomedical Research Networking Centre on Rare Diseases (CIBERER), ISCIII, 28029, Madrid, Spain IdiPAZ Institute for Health Research, Madrid 28029, Spain
| | - Ángela M Valverde
- Alberto Sols Biomedical Research Institute (IIBm) (CSIC/UAM), 28029, Madrid, Spain Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERdem), ISCIII, 28029, Madrid, Spain IdiPAZ Institute for Health Research, Madrid 28029, Spain
| |
Collapse
|
34
|
Qiu Y, Yang J, Bian S, Chen G, Yu J. PPARγ suppresses the proliferation of cardiac myxoma cells through downregulation of MEF2D in a miR-122-dependent manner. Biochem Biophys Res Commun 2016; 474:560-565. [PMID: 27109478 DOI: 10.1016/j.bbrc.2016.04.112] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 04/20/2016] [Indexed: 12/26/2022]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ), a multiple functional transcription factor, has been reported to have anti-tumor effects through inhibition of cells proliferation. However, its effects on cardiac myxoma (CM) cells and the underlying signaling mechanism is unclear. In the present study, we demonstrated that the level of PPARγ is inversely correlated with that of myocyte enhancer factor 2D (MEF2D), a biomarker of CM. We found that activation of PPARγ inhibit MEF2D expression via upregulation of miR-122, which can target the 3'-UTR of MEF2D and inhibit MEF2D expression, by directly binding to the PPRE in the miR-122 promoter region. Functional experiments further showed that miR-122-dependent downregulation of MEF2D by PPARγ suppress the proliferation of CM cells. These results suggest that PPARγ may exert its antiproliferative effects by negatively regulating the MEF2D in CM cells, which through upregulation of miR-122, and PPARγ/miR-122/MEF2D signaling pathway may be a novel target for treatment of CM.
Collapse
Affiliation(s)
- Youzhu Qiu
- Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Jie Yang
- Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Shizhu Bian
- Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Guozhu Chen
- Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Jie Yu
- Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
35
|
Low JKK, Im H, Erce MA, Hart-Smith G, Snyder MP, Wilkins MR. Protein substrates of the arginine methyltransferase Hmt1 identified by proteome arrays. Proteomics 2016; 16:465-76. [DOI: 10.1002/pmic.201400564] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 09/27/2015] [Accepted: 11/10/2015] [Indexed: 01/22/2023]
Affiliation(s)
- Jason K. K. Low
- Systems Biology Initiative; School of Biotechnology and Biomolecular Sciences; The University of New South Wales; Sydney Australia
| | - Hogune Im
- Department of Genetics; Stanford University School of Medicine; Palo Alto CA USA
| | - Melissa A. Erce
- Systems Biology Initiative; School of Biotechnology and Biomolecular Sciences; The University of New South Wales; Sydney Australia
| | - Gene Hart-Smith
- Systems Biology Initiative; School of Biotechnology and Biomolecular Sciences; The University of New South Wales; Sydney Australia
| | - Michael P. Snyder
- Department of Genetics; Stanford University School of Medicine; Palo Alto CA USA
| | - Marc R. Wilkins
- Systems Biology Initiative; School of Biotechnology and Biomolecular Sciences; The University of New South Wales; Sydney Australia
| |
Collapse
|
36
|
Abstract
The identification of transcriptional differences has served as an important starting point in understanding the molecular mechanisms behind biological processes and systems. The developmental biology of the inner ear, the biology of hearing and of course the pathology of deafness are all processes that warrant a molecular description if we are to improve human health. To this end, technological innovation has meant that larger scale analysis of gene transcription has been possible for a number of years now, extending our molecular analysis of genes to beyond those that are currently in vogue for a given system. In this review, some of the contributions gene profiling has made to understanding developmental, pathological and physiological processes in the inner ear are highlighted.
Collapse
Affiliation(s)
- Thomas Schimmang
- Instituto de Biología y Genética MolecularUniversidad de Valladolid y Consejo Superior de Investigaciones CientíficasValladolidSpain
| | - Mark Maconochie
- School of Biological and Chemical SciencesQueen Mary University of LondonLondonUK
| |
Collapse
|
37
|
Yamahara K, Yamamoto N, Nakagawa T, Ito J. Insulin-like growth factor 1: A novel treatment for the protection or regeneration of cochlear hair cells. Hear Res 2015; 330:2-9. [DOI: 10.1016/j.heares.2015.04.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 04/19/2015] [Accepted: 04/24/2015] [Indexed: 11/15/2022]
|
38
|
Rodríguez-de la Rosa L, Sánchez-Calderón H, Contreras J, Murillo-Cuesta S, Falagan S, Avendaño C, Dopazo J, Varela-Nieto I, Milo M. Comparative gene expression study of the vestibular organ of the Igf1 deficient mouse using whole-transcript arrays. Hear Res 2015; 330:62-77. [PMID: 26341476 DOI: 10.1016/j.heares.2015.08.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 08/27/2015] [Accepted: 08/28/2015] [Indexed: 10/23/2022]
Abstract
The auditory and vestibular organs form the inner ear and have a common developmental origin. Insulin like growth factor 1 (IGF-1) has a central role in the development of the cochlea and maintenance of hearing. Its deficiency causes sensorineural hearing loss in man and mice. During chicken early development, IGF-1 modulates neurogenesis of the cochleovestibular ganglion but no further studies have been conducted to explore the potential role of IGF-1 in the vestibular system. In this study we have compared the whole transcriptome of the vestibular organ from wild type and Igf1(-/-) mice at different developmental and postnatal times. RNA was prepared from E18.5, P15 and P90 vestibular organs of Igf1(-/-) and Igf1(+/+) mice and the transcriptome analysed in triplicates using Affymetrix(®) Mouse Gene 1.1 ST Array Plates. These plates are whole-transcript arrays that include probes to measure both messenger (mRNA) and long intergenic non-coding RNA transcripts (lincRNA), with a coverage of over 28 thousand coding transcripts and over 7 thousands non-coding transcripts. Given the complexity of the data we used two different methods VSN-RMA and mmBGX to analyse and compare the data. This is to better evaluate the number of false positives and to quantify uncertainty of low signals. We identified a number of differentially expressed genes that we described using functional analysis and validated using RT-qPCR. The morphology of the vestibular organ did not show differences between genotypes and no evident alterations were observed in the vestibular sensory areas of the null mice. However, well-defined cellular alterations were found in the vestibular neurons with respect their number and size. Although these mice did not show a dramatic vestibular phenotype, we conducted a functional analysis on differentially expressed genes between genotypes and across time. This was with the aim to identify new pathways that are involved in the development of the vestibular organ as well as pathways that maybe affected by the lack of IGF-1 and be associated to the morphological changes of the vestibular neurons that we observed in the Igf1(-/-) mice.
Collapse
Affiliation(s)
- Lourdes Rodríguez-de la Rosa
- Neurobiology of Hearing, Department of Endocrine and Nervous System Pathophysiology, Alberto Sols Biomedical Research Institute (IIBM), CSIC-UAM, Madrid, Spain; Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III (ISCIII), Madrid, Spain; IdiPAZ Institute for Health Research, Madrid, Spain
| | - Hortensia Sánchez-Calderón
- Neurobiology of Hearing, Department of Endocrine and Nervous System Pathophysiology, Alberto Sols Biomedical Research Institute (IIBM), CSIC-UAM, Madrid, Spain
| | - Julio Contreras
- Neurobiology of Hearing, Department of Endocrine and Nervous System Pathophysiology, Alberto Sols Biomedical Research Institute (IIBM), CSIC-UAM, Madrid, Spain; Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III (ISCIII), Madrid, Spain; Department of Anatomy, Faculty of Veterinary, Complutense University, Madrid, Spain
| | - Silvia Murillo-Cuesta
- Neurobiology of Hearing, Department of Endocrine and Nervous System Pathophysiology, Alberto Sols Biomedical Research Institute (IIBM), CSIC-UAM, Madrid, Spain; Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III (ISCIII), Madrid, Spain; IdiPAZ Institute for Health Research, Madrid, Spain
| | - Sandra Falagan
- Department of Anatomy, Faculty of Medicine, Autonomous University, Madrid, Spain
| | - Carlos Avendaño
- IdiPAZ Institute for Health Research, Madrid, Spain; Department of Anatomy, Faculty of Medicine, Autonomous University, Madrid, Spain
| | - Joaquín Dopazo
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III (ISCIII), Madrid, Spain; Department of Computational Genomics, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Isabel Varela-Nieto
- Neurobiology of Hearing, Department of Endocrine and Nervous System Pathophysiology, Alberto Sols Biomedical Research Institute (IIBM), CSIC-UAM, Madrid, Spain; Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III (ISCIII), Madrid, Spain; IdiPAZ Institute for Health Research, Madrid, Spain
| | - Marta Milo
- Department of Biomedical Science, University of Sheffield, Sheffield, UK.
| |
Collapse
|
39
|
de Iriarte Rodríguez R, Pulido S, Rodríguez-de la Rosa L, Magariños M, Varela-Nieto I. Age-regulated function of autophagy in the mouse inner ear. Hear Res 2015; 330:39-50. [PMID: 26235979 DOI: 10.1016/j.heares.2015.07.020] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 07/22/2015] [Accepted: 07/29/2015] [Indexed: 12/13/2022]
Abstract
Autophagy is a highly conserved catabolic process essential for embryonic development and adult homeostasis. The autophagic machinery supplies energy by recycling intracellular components and facilitates the removal of apoptotic cells. In the inner ear, autophagy has been reported to play roles during early development in the chicken embryo and in the response to otic injury in the adult mouse. However, there are no studies on the expression of the autophagy machinery in the postnatal and adult inner ear. Insulin-like growth factor 1 (IGF-1) is one of the factors that regulate both otic development and cochlear postnatal maturation and function. Here, we hypothesised that autophagy could be one of the processes involved in the cochlear development and functional maturation. We report that autophagy-related genes (ATG) Becn1, Atg4g and Atg5 are expressed in the mouse cochlea, vestibular system and brainstem cochlear nuclei from late developmental stages to adulthood. Atg9 was studied in the mouse cochlea and showed a similar pattern. The presence of autophagic flux was confirmed by decreased sequestosome 1 (SQSTM1/p62) and increased relative levels of microtubule-associated protein light chain 3-II (LC3-II). Inner ear autophagy flux is developmentally regulated and is lower at perinatal stages than in the adult mouse, where an expression plateau is reached at the age of two-months, coinciding with the age at which full functional activity is reached. Expression is maintained in adult mice and declines after the age of twelve months. LC3B labelling showed that autophagy was primarily associated with spiral ganglion neurons. Over time, Igf1 wild type mice showed lower expression of genes coding for IGF-1 high affinity receptor and the family factor IGF-2 than null mice. Parallel analysis of autophagy machinery gene expression showed no significant differences between the genotypes over the lifespan of the null mice. Taken together, these results show that the autophagy machinery expression in the inner ear is regulated with age but is not compromised by the chronic absence of IGF-1. Our data also strongly support that the up-regulation of autophagy machinery genes is concomitant with the functional maturation of the inner ear.
Collapse
Affiliation(s)
- Rocío de Iriarte Rodríguez
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, Madrid, Spain; CIBERER, Unit 761, Instituto de Salud Carlos III, Madrid, Spain
| | - Sara Pulido
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, Madrid, Spain
| | - Lourdes Rodríguez-de la Rosa
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, Madrid, Spain; CIBERER, Unit 761, Instituto de Salud Carlos III, Madrid, Spain; IdiPAZ, Instituto de Investigación Sanitaria, Madrid, Spain
| | - Marta Magariños
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, Madrid, Spain; CIBERER, Unit 761, Instituto de Salud Carlos III, Madrid, Spain; Departamento de Biología, Universidad Autónoma de Madrid, Madrid, Spain.
| | - Isabel Varela-Nieto
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, Madrid, Spain; CIBERER, Unit 761, Instituto de Salud Carlos III, Madrid, Spain; IdiPAZ, Instituto de Investigación Sanitaria, Madrid, Spain
| |
Collapse
|
40
|
de Iriarte Rodríguez R, Magariños M, Pfeiffer V, Rapp UR, Varela-Nieto I. C-Raf deficiency leads to hearing loss and increased noise susceptibility. Cell Mol Life Sci 2015; 72:3983-98. [PMID: 25975225 PMCID: PMC4575698 DOI: 10.1007/s00018-015-1919-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 04/21/2015] [Accepted: 04/27/2015] [Indexed: 12/20/2022]
Abstract
The family of RAF kinases transduces extracellular information to the nucleus, and their activation is crucial for cellular regulation on many levels, ranging from embryonic development to carcinogenesis. B-RAF and C-RAF modulate neurogenesis and neuritogenesis during chicken inner ear development. C-RAF deficiency in humans is associated with deafness in the rare genetic insulin-like growth factor 1 (IGF-1), Noonan and Leopard syndromes. In this study, we show that RAF kinases are expressed in the developing inner ear and in adult mouse cochlea. A homozygous C-Raf deletion in mice caused profound deafness with no evident cellular aberrations except for a remarkable reduction of the K+ channel Kir4.1 expression, a trait that suffices as a cause of deafness. To explore the role of C-Raf in cellular protection and repair, heterozygous C-Raf+/− mice were exposed to noise. A reduced C-RAF level negatively affected hearing preservation in response to noise through mechanisms involving the activation of JNK and an exacerbated apoptotic response. Taken together, these results strongly support a role for C-RAF in hearing protection.
Collapse
Affiliation(s)
- Rocío de Iriarte Rodríguez
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, Arturo Duperier 4, 28029, Madrid, Spain.,Centre for Biomedical Network Research (CIBERER), Institute of Health Carlos III (ISCIII), Madrid, Spain
| | - Marta Magariños
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, Arturo Duperier 4, 28029, Madrid, Spain. .,Centre for Biomedical Network Research (CIBERER), Institute of Health Carlos III (ISCIII), Madrid, Spain. .,Departamento de Biología, Universidad Autónoma de Madrid, Darwin 2, 28049, Madrid, Spain.
| | - Verena Pfeiffer
- Institute for Medical Radiation and Cell Research (MSZ), University of Würzburg, Versbacher Strasse 5, 97078, Würzburg, Germany.,Institute for Anatomy and Cell Biology, University of Würzburg, Koellikerstraße 6, 97070, Würzburg, Germany
| | - Ulf R Rapp
- Institute for Medical Radiation and Cell Research (MSZ), University of Würzburg, Versbacher Strasse 5, 97078, Würzburg, Germany.,Molecular Mechanisms of Lung Cancer, Max Planck Institute for Heart and Lung Research, Parkstr. 1, 61231, Bad Nauheim, Germany
| | - Isabel Varela-Nieto
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, Arturo Duperier 4, 28029, Madrid, Spain.,Centre for Biomedical Network Research (CIBERER), Institute of Health Carlos III (ISCIII), Madrid, Spain.,Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| |
Collapse
|
41
|
Murillo-Cuesta S, Rodríguez-de la Rosa L, Contreras J, Celaya AM, Camarero G, Rivera T, Varela-Nieto I. Transforming growth factor β1 inhibition protects from noise-induced hearing loss. Front Aging Neurosci 2015; 7:32. [PMID: 25852546 PMCID: PMC4367183 DOI: 10.3389/fnagi.2015.00032] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 02/28/2015] [Indexed: 12/20/2022] Open
Abstract
Excessive exposure to noise damages the principal cochlear structures leading to hearing impairment. Inflammatory and immune responses are central mechanisms in cochlear defensive response to noise but, if unregulated, they contribute to inner ear damage and hearing loss. Transforming growth factor β (TGF-β) is a key regulator of both responses and high levels of this factor have been associated with cochlear injury in hearing loss animal models. To evaluate the potential of targeting TGF-β as a therapeutic strategy for preventing or ameliorating noise-induced hearing loss (NIHL), we studied the auditory function, cochlear morphology, gene expression and oxidative stress markers in mice exposed to noise and treated with TGF-β1 peptidic inhibitors P17 and P144, just before or immediately after noise insult. Our results indicate that systemic administration of both peptides significantly improved both the evolution of hearing thresholds and the degenerative changes induced by noise-exposure in lateral wall structures. Moreover, treatments ameliorated the inflammatory state and redox balance. These therapeutic effects were dose-dependent and more effective if the TGF-β1 inhibitors were administered prior to inducing the injury. In conclusion, inhibition of TGF-β1 actions with antagonistic peptides represents a new, promising therapeutic strategy for the prevention and repair of noise-induced cochlear damage.
Collapse
Affiliation(s)
- Silvia Murillo-Cuesta
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM) Madrid, Spain ; Centre for Biomedical Network Research (CIBER), Institute of Health Carlos III (ISCIII) Madrid, Spain ; Hospital La Paz Institute for Health Research (IdiPAZ) Madrid, Spain
| | - Lourdes Rodríguez-de la Rosa
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM) Madrid, Spain ; Centre for Biomedical Network Research (CIBER), Institute of Health Carlos III (ISCIII) Madrid, Spain ; Hospital La Paz Institute for Health Research (IdiPAZ) Madrid, Spain
| | - Julio Contreras
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM) Madrid, Spain ; Centre for Biomedical Network Research (CIBER), Institute of Health Carlos III (ISCIII) Madrid, Spain ; Veterinary Faculty, Complutense University of Madrid Madrid, Spain
| | - Adelaida M Celaya
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM) Madrid, Spain ; Centre for Biomedical Network Research (CIBER), Institute of Health Carlos III (ISCIII) Madrid, Spain
| | - Guadalupe Camarero
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM) Madrid, Spain ; Centre for Biomedical Network Research (CIBER), Institute of Health Carlos III (ISCIII) Madrid, Spain ; Hospital La Paz Institute for Health Research (IdiPAZ) Madrid, Spain
| | - Teresa Rivera
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM) Madrid, Spain ; Centre for Biomedical Network Research (CIBER), Institute of Health Carlos III (ISCIII) Madrid, Spain ; Príncipe de Asturias University Hospital, University of Alcalá, Alcalá de Henares Madrid, Spain
| | - Isabel Varela-Nieto
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM) Madrid, Spain ; Centre for Biomedical Network Research (CIBER), Institute of Health Carlos III (ISCIII) Madrid, Spain ; Hospital La Paz Institute for Health Research (IdiPAZ) Madrid, Spain
| |
Collapse
|
42
|
Martínez-Vega R, Garrido F, Partearroyo T, Cediel R, Zeisel SH, Martínez-Álvarez C, Varela-Moreiras G, Varela-Nieto I, Pajares MA. Folic acid deficiency induces premature hearing loss through mechanisms involving cochlear oxidative stress and impairment of homocysteine metabolism. FASEB J 2014; 29:418-32. [PMID: 25384423 DOI: 10.1096/fj.14-259283] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Nutritional imbalance is emerging as a causative factor of hearing loss. Epidemiologic studies have linked hearing loss to elevated plasma total homocysteine (tHcy) and folate deficiency, and have shown that folate supplementation lowers tHcy levels potentially ameliorating age-related hearing loss. The purpose of this study was to address the impact of folate deficiency on hearing loss and to examine the underlying mechanisms. For this purpose, 2-mo-old C57BL/6J mice (Animalia Chordata Mus musculus) were randomly divided into 2 groups (n = 65 each) that were fed folate-deficient (FD) or standard diets for 8 wk. HPLC analysis demonstrated a 7-fold decline in serum folate and a 3-fold increase in tHcy levels. FD mice exhibited severe hearing loss measured by auditory brainstem recordings and TUNEL-positive-apoptotic cochlear cells. RT-quantitative PCR and Western blotting showed reduced levels of enzymes catalyzing homocysteine (Hcy) production and recycling, together with a 30% increase in protein homocysteinylation. Redox stress was demonstrated by decreased expression of catalase, glutathione peroxidase 4, and glutathione synthetase genes, increased levels of manganese superoxide dismutase, and NADPH oxidase-complex adaptor cytochrome b-245, α-polypeptide (p22phox) proteins, and elevated concentrations of glutathione species. Altogether, our findings demonstrate, for the first time, that the relationship between hyperhomocysteinemia induced by folate deficiency and premature hearing loss involves impairment of cochlear Hcy metabolism and associated oxidative stress.
Collapse
Affiliation(s)
- Raquel Martínez-Vega
- *Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain; Unidad 761, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain; Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU San Pablo, Boadilla del Monte, Madrid, Spain; Hospital Clínico Veterinario, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain; University of North Carolina at Chapel Hill, Nutrition Research Institute, Kannapolis, North Carolina, USA; Departamento de Anatomía y Embriología Humana I, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain; and Instituto de Investigación Sanitaria La Paz, Madrid, Spain
| | - Francisco Garrido
- *Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain; Unidad 761, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain; Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU San Pablo, Boadilla del Monte, Madrid, Spain; Hospital Clínico Veterinario, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain; University of North Carolina at Chapel Hill, Nutrition Research Institute, Kannapolis, North Carolina, USA; Departamento de Anatomía y Embriología Humana I, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain; and Instituto de Investigación Sanitaria La Paz, Madrid, Spain
| | - Teresa Partearroyo
- *Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain; Unidad 761, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain; Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU San Pablo, Boadilla del Monte, Madrid, Spain; Hospital Clínico Veterinario, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain; University of North Carolina at Chapel Hill, Nutrition Research Institute, Kannapolis, North Carolina, USA; Departamento de Anatomía y Embriología Humana I, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain; and Instituto de Investigación Sanitaria La Paz, Madrid, Spain
| | - Rafael Cediel
- *Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain; Unidad 761, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain; Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU San Pablo, Boadilla del Monte, Madrid, Spain; Hospital Clínico Veterinario, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain; University of North Carolina at Chapel Hill, Nutrition Research Institute, Kannapolis, North Carolina, USA; Departamento de Anatomía y Embriología Humana I, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain; and Instituto de Investigación Sanitaria La Paz, Madrid, Spain
| | - Steven H Zeisel
- *Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain; Unidad 761, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain; Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU San Pablo, Boadilla del Monte, Madrid, Spain; Hospital Clínico Veterinario, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain; University of North Carolina at Chapel Hill, Nutrition Research Institute, Kannapolis, North Carolina, USA; Departamento de Anatomía y Embriología Humana I, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain; and Instituto de Investigación Sanitaria La Paz, Madrid, Spain
| | - Concepción Martínez-Álvarez
- *Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain; Unidad 761, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain; Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU San Pablo, Boadilla del Monte, Madrid, Spain; Hospital Clínico Veterinario, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain; University of North Carolina at Chapel Hill, Nutrition Research Institute, Kannapolis, North Carolina, USA; Departamento de Anatomía y Embriología Humana I, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain; and Instituto de Investigación Sanitaria La Paz, Madrid, Spain
| | - Gregorio Varela-Moreiras
- *Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain; Unidad 761, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain; Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU San Pablo, Boadilla del Monte, Madrid, Spain; Hospital Clínico Veterinario, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain; University of North Carolina at Chapel Hill, Nutrition Research Institute, Kannapolis, North Carolina, USA; Departamento de Anatomía y Embriología Humana I, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain; and Instituto de Investigación Sanitaria La Paz, Madrid, Spain
| | - Isabel Varela-Nieto
- *Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain; Unidad 761, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain; Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU San Pablo, Boadilla del Monte, Madrid, Spain; Hospital Clínico Veterinario, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain; University of North Carolina at Chapel Hill, Nutrition Research Institute, Kannapolis, North Carolina, USA; Departamento de Anatomía y Embriología Humana I, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain; and Instituto de Investigación Sanitaria La Paz, Madrid, Spain
| | - María A Pajares
- *Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain; Unidad 761, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain; Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU San Pablo, Boadilla del Monte, Madrid, Spain; Hospital Clínico Veterinario, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain; University of North Carolina at Chapel Hill, Nutrition Research Institute, Kannapolis, North Carolina, USA; Departamento de Anatomía y Embriología Humana I, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain; and Instituto de Investigación Sanitaria La Paz, Madrid, Spain
| |
Collapse
|
43
|
Fuentes-Santamaría V, Alvarado JC, Rodríguez-de la Rosa L, Murillo-Cuesta S, Contreras J, Juiz JM, Varela-Nieto I. IGF-1 deficiency causes atrophic changes associated with upregulation of VGluT1 and downregulation of MEF2 transcription factors in the mouse cochlear nuclei. Brain Struct Funct 2014; 221:709-34. [PMID: 25378055 DOI: 10.1007/s00429-014-0934-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Accepted: 10/28/2014] [Indexed: 02/08/2023]
Abstract
Insulin-like growth factor 1 (IGF-1) is a neurotrophic protein that plays a crucial role in modulating neuronal function and synaptic plasticity in the adult brain. Mice lacking the Igf1 gene exhibit profound deafness and multiple anomalies in the inner ear and spiral ganglion. An issue that remains unknown is whether, in addition to these peripheral abnormalities, IGF-1 deficiency also results in structural changes along the central auditory pathway that may contribute to an imbalance between excitation and inhibition, which might be reflected in abnormal auditory brainstem responses (ABR). To assess such a possibility, we evaluated the morphological and physiological alterations in the cochlear nucleus complex of the adult mouse. The expression and distribution of the vesicular glutamate transporter 1 (VGluT1) and the vesicular inhibitory transporter (VGAT), which were used as specific markers for labeling excitatory and inhibitory terminals, and the involvement of the activity-dependent myocyte enhancer factor 2 (MEF2) transcription factors in regulating excitatory synapses were assessed in a 4-month-old mouse model of IGF-1 deficiency and neurosensorial deafness (Igf1 (-/-) homozygous null mice). The results demonstrate decreases in the cochlear nucleus area and cell size along with cell loss in the cochlear nuclei of the deficient mouse. Additionally, our results demonstrate that there is upregulation of VGluT1, but not VGAT, immunostaining and downregulation of MEF2 transcription factors together with increased wave II amplitude in the ABR recording. Our observations provide evidence of an abnormal neuronal cytoarchitecture in the cochlear nuclei of Igf1 (-/-) null mice and suggest that the increased efficacy of glutamatergic synapses might be mediated by MEF2 transcription factors.
Collapse
Affiliation(s)
- V Fuentes-Santamaría
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Albacete, Spain.
| | - J C Alvarado
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Albacete, Spain
| | - L Rodríguez-de la Rosa
- Facultad de Medicina, Universidad de Castilla-La Mancha, Campus de Albacete. C/Almansa, 14, 02006, Albacete, Spain
| | - S Murillo-Cuesta
- Grupo de Neurobiología de la Audición, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain.,Centro Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain.,IdiPAZ Instituto de Investigación en Salud, Madrid, Spain
| | - J Contreras
- Grupo de Neurobiología de la Audición, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain.,Centro Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain.,IdiPAZ Instituto de Investigación en Salud, Madrid, Spain.,Facultad de Veterinaria, Universidad Complutense, Madrid, Spain
| | - J M Juiz
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Albacete, Spain
| | - I Varela-Nieto
- Grupo de Neurobiología de la Audición, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain.,Centro Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain.,IdiPAZ Instituto de Investigación en Salud, Madrid, Spain
| |
Collapse
|
44
|
Loddo M, Andryszkiewicz J, Rodriguez-Acebes S, Stoeber K, Jones A, Dafou D, Apostolidou S, Wollenschlaeger A, Widschwendter M, Sainsbury R, Tudzarova S, Williams GH. Pregnancy-associated plasma protein A regulates mitosis and is epigenetically silenced in breast cancer. J Pathol 2014; 233:344-56. [PMID: 24931331 DOI: 10.1002/path.4393] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 06/06/2014] [Accepted: 06/10/2014] [Indexed: 01/23/2023]
Abstract
Aberrant mitosis is a common feature of cancer, yet little is known about the altered genes causing mitotic defects. We screened human tumours for cells with morphological signatures of highly specific mitotic defects previously assigned to candidate genes in a genome-wide RNA interference screen carried out in HeLa cells (www.mitocheck.org). We discovered a striking enrichment of early mitotic configurations indicative of prophase/prometaphase delay in breast cancer. Promoter methylation analysis of MitoCheck candidate genes assigned to the corresponding 'mitotic delay' class linked this defect to epigenetic silencing of the gene encoding pregnancy-associated plasma protein-A (PAPPA), a secreted protease. PAPPA silencing was highly prevalent in precursor lesions and invasive breast cancer. Experimental manipulation of PAPPA protein levels in human mammary epithelial cells and in breast cancer cell lines demonstrates that progression through early mitosis is dependent on PAPPA function, and that breast cancer cells become more invasive after down-regulation of this protease. PAPPA regulates mitotic progression through modulating the IGF-1 signalling pathway resulting in activation of the forkhead transcription factor FoxM1, which drives a transcriptional cluster of essential mitotic genes. Our results show that PAPPA has a critical function in normal cell division and is targeted early in breast cancer development.
Collapse
Affiliation(s)
- Marco Loddo
- UCL Cancer Institute, University College London, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Differential organ phenotypes after postnatal Igf1r gene conditional deletion induced by tamoxifen in UBC-CreERT2; Igf1r fl/fl double transgenic mice. Transgenic Res 2014; 24:279-94. [DOI: 10.1007/s11248-014-9837-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 09/09/2014] [Indexed: 11/25/2022]
|
46
|
Yamamoto N, Nakagawa T, Ito J. Application of insulin-like growth factor-1 in the treatment of inner ear disorders. Front Pharmacol 2014; 5:208. [PMID: 25309440 PMCID: PMC4159992 DOI: 10.3389/fphar.2014.00208] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 08/22/2014] [Indexed: 01/10/2023] Open
Abstract
Sensorineural hearing loss (SNHL) is considered an intractable disease, given that hair and supporting cells (HCs and SCs) of the postnatal mammalian cochlea are unable to regenerate. However, with progress in regenerative medicine in the 21st century, several innovative approaches for achieving regeneration of inner ear HCs and SCs have become available. These methods include stem cell transplantation, overexpression of specific genes, and treatment with growth factors. Insulin-like growth factor-1 (IGF-1) is one of the growth factors that are involved in the development of the inner ear. Treatment with IGF-1 maintains HC numbers in the postnatal mammalian cochlea after various types of HC injuries, with activation of two major pathways downstream of IGF-1 signaling. In the aminoglycoside-treated neonatal mouse cochlear explant culture, promotion of the cell-cycle in SCs as well as inhibition of HC apoptosis was observed in the IGF-1-treated group. Activation of downstream molecules was observed in SCs and, in turn, SCs contribute to the maintenance of HC numbers. Using comprehensive analysis of the gene expression, the candidate effector molecules of the IGF-1 signaling pathway in the protection of HCs were identified as Netrin1 and Gap43. Based on these studies, a clinical trial has sought to investigate the effects of IGF-1 on SNHL. Sudden SNHL (SSHL) that was refractory to systemic steroids was treated with IGF-1 in a gelatin hydrogel and the outcome was compared with a historical control of hyperbaric oxygen therapy. The proportion of patients showing hearing improvement was significantly higher in the IGF-1-treatment group at 24 weeks after treatment than in the control group. A randomized clinical trial is ongoing to compare the effect of IGF-1 treatment with that of intra-tympanic steroids for SSHL that is refractory to systemic steroids.
Collapse
Affiliation(s)
- Norio Yamamoto
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University Kyoto Japan
| | - Takayuki Nakagawa
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University Kyoto Japan
| | - Juichi Ito
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University Kyoto Japan
| |
Collapse
|
47
|
Wolber LE, Steves CJ, Tsai PC, Deloukas P, Spector TD, Bell JT, Williams FMK. Epigenome-wide DNA methylation in hearing ability: new mechanisms for an old problem. PLoS One 2014; 9:e105729. [PMID: 25184702 PMCID: PMC4153547 DOI: 10.1371/journal.pone.0105729] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 07/28/2014] [Indexed: 11/24/2022] Open
Abstract
Epigenetic regulation of gene expression has been shown to change over time and may be associated with environmental exposures in common complex traits. Age-related hearing impairment is a complex disorder, known to be heritable, with heritability estimates of 57–70%. Epigenetic regulation might explain the observed difference in age of onset and magnitude of hearing impairment with age. Epigenetic epidemiology studies using unrelated samples can be limited in their ability to detect small effects, and recent epigenetic findings in twins underscore the power of this well matched study design. We investigated the association between venous blood DNA methylation epigenome-wide and hearing ability. Pure-tone audiometry (PTA) and Illumina HumanMethylation array data were obtained from female twin volunteers enrolled in the TwinsUK register. Two study groups were explored: first, an epigenome-wide association scan (EWAS) was performed in a discovery sample (n = 115 subjects, age range: 47–83 years, Illumina 27 k array), then replication of the top ten associated probes from the discovery EWAS was attempted in a second unrelated sample (n = 203, age range: 41–86 years, Illumina 450 k array). Finally, a set of monozygotic (MZ) twin pairs (n = 21 pairs) within the discovery sample (Illumina 27 k array) was investigated in more detail in an MZ discordance analysis. Hearing ability was strongly associated with DNA methylation levels in the promoter regions of several genes, including TCF25 (cg01161216, p = 6.6×10−6), FGFR1 (cg15791248, p = 5.7×10−5) and POLE (cg18877514, p = 6.3×10−5). Replication of these results in a second sample confirmed the presence of differential methylation at TCF25 (p(replication) = 6×10−5) and POLE (p(replication) = 0.016). In the MZ discordance analysis, twins' intrapair difference in hearing ability correlated with DNA methylation differences at ACP6 (cg01377755, r = −0.75, p = 1.2×10−4) and MEF2D (cg08156349, r = −0.75, p = 1.4×10−4). Examination of gene expression in skin, suggests an influence of differential methylation on expression, which may account for the variation in hearing ability with age.
Collapse
MESH Headings
- Acid Phosphatase/blood
- Acid Phosphatase/genetics
- Aged
- Aged, 80 and over
- Aging/blood
- Aging/genetics
- Aging/pathology
- Audiometry, Pure-Tone
- Basic Helix-Loop-Helix Transcription Factors/blood
- Basic Helix-Loop-Helix Transcription Factors/genetics
- DNA Methylation
- DNA Polymerase II/blood
- DNA Polymerase II/genetics
- Epigenesis, Genetic
- Female
- Genome, Human
- Hearing Loss, Functional/blood
- Hearing Loss, Functional/genetics
- Hearing Loss, Functional/physiopathology
- Humans
- MEF2 Transcription Factors/blood
- MEF2 Transcription Factors/genetics
- Middle Aged
- Poly-ADP-Ribose Binding Proteins
- Promoter Regions, Genetic
- Quantitative Trait, Heritable
- Receptor, Fibroblast Growth Factor, Type 1/blood
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Repressor Proteins/blood
- Repressor Proteins/genetics
- Twins, Monozygotic/genetics
Collapse
Affiliation(s)
- Lisa E. Wolber
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Claire J. Steves
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Pei-Chien Tsai
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Panos Deloukas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- King Abdulaziz University, Jeddah, Saudi Arabia
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom
| | - Tim D. Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Jordana T. Bell
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Frances M. K. Williams
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
48
|
Kim HJ, Ryu J, Woo HM, Cho SS, Sung MK, Kim SC, Park MH, Park T, Koo SK. Patterns of gene expression associated with Pten deficiency in the developing inner ear. PLoS One 2014; 9:e97544. [PMID: 24893171 PMCID: PMC4043736 DOI: 10.1371/journal.pone.0097544] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 04/19/2014] [Indexed: 12/26/2022] Open
Abstract
In inner ear development, phosphatase and tensin homolog (PTEN) is necessary for neuronal maintenance, such as neuronal survival and accurate nerve innervations of hair cells. We previously reported that Pten conditional knockout (cKO) mice exhibited disorganized fasciculus with neuronal apoptosis in spiral ganglion neurons (SGNs). To better understand the genes and signaling networks related to auditory neuron maintenance, we compared the profiles of differentially expressed genes (DEGs) using microarray analysis of the inner ear in E14.5 Pten cKO and wild-type mice. We identified 46 statistically significant transcripts using significance analysis of microarrays, with the false-discovery rate set at 0%. Among the DEGs, expression levels of candidate genes and expression domains were validated by quantitative real-time RT-PCR and in situ hybridization, respectively. Ingenuity pathway analysis using DEGs identified significant signaling networks associated with apoptosis, cellular movement, and axon guidance (i.e., secreted phosphoprotein 1 (Spp1)-mediated cellular movement and regulator of G-protein signaling 4 (Rgs4)-mediated axon guidance). This result was consistent with the phenotypic defects of SGNs in Pten cKO mice (e.g., neuronal apoptosis, abnormal migration, and irregular nerve fiber patterns of SGNs). From this study, we suggest two key regulatory signaling networks mediated by Spp1 and Rgs4, which may play potential roles in neuronal differentiation of developing auditory neurons.
Collapse
Affiliation(s)
- Hyung Jin Kim
- Division of Intractable Diseases, Center for Biomedical Sciences, National Institute of Health, Chungcheongbuk-do, South Korea
| | - Jihee Ryu
- Division of Intractable Diseases, Center for Biomedical Sciences, National Institute of Health, Chungcheongbuk-do, South Korea
| | - Hae-Mi Woo
- Division of Intractable Diseases, Center for Biomedical Sciences, National Institute of Health, Chungcheongbuk-do, South Korea
| | - Samuel Sunghwan Cho
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, South Korea
| | - Min Kyung Sung
- Korean BioInformation Center (KOBIC), Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Sang Cheol Kim
- Korean BioInformation Center (KOBIC), Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Mi-Hyun Park
- Division of Intractable Diseases, Center for Biomedical Sciences, National Institute of Health, Chungcheongbuk-do, South Korea
| | - Taesung Park
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, South Korea
- Department of Statistics, Seoul National University, Seoul, South Korea
| | - Soo Kyung Koo
- Division of Intractable Diseases, Center for Biomedical Sciences, National Institute of Health, Chungcheongbuk-do, South Korea
- * E-mail:
| |
Collapse
|
49
|
Effect of maternal care on hearing onset induced by developmental changes in the auditory periphery. J Neurosci 2014; 34:4528-33. [PMID: 24671998 DOI: 10.1523/jneurosci.4188-13.2014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Handling (H) and cross-fostering (CF) rodent pups during postnatal development triggers changes in maternal behavior which in turn trigger long-term physiological changes in the offspring. However, less is known about the short-term effects of H and CF on infant development. In this study we hypothesized that manipulations of maternal care affect the onset of hearing in Wistar rats. To test this hypothesis we obtained auditory brainstem responses (ABRs) and micro-CT x-ray scans to measure changes in the development of the auditory periphery in H and CF pups manipulated at postnatal day (P)1, P5, or P9. We found evidence of changes in hearing development in H and CF pups compared with naive pups, including changes in the percentage of animals with ABRs during development, a decrease in ABR thresholds between P13 and P15, and anatomical results consistent with an accelerated formation of the middle ear cavity and opening of the ear canal. Biochemical measurements showed elevated levels of thyroid hormone in plasma from naive and CF pups. These results provide evidence that manipulations of maternal care accelerate hearing onset in Wistar rats. Understanding the mechanisms by which maternal care affects hearing onset opens new opportunities to study experience-dependent development of mammalian hearing.
Collapse
|
50
|
Killeen AP, Morris DG, Kenny DA, Mullen MP, Diskin MG, Waters SM. Global gene expression in endometrium of high and low fertility heifers during the mid-luteal phase of the estrous cycle. BMC Genomics 2014; 15:234. [PMID: 24669966 PMCID: PMC3986929 DOI: 10.1186/1471-2164-15-234] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 03/14/2014] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND In both beef and dairy cattle, the majority of early embryo loss occurs within the first 14 days following insemination. During this time-period, embryos are completely dependent on their maternal uterine environment for development, growth and ultimately survival, therefore an optimum uterine environment is critical to their survival. The objective of this study was to investigate whether differences in endometrial gene expression during the mid-luteal phase of the estrous cycle exist between crossbred beef heifers ranked as either high (HF) or low fertility (LF) (following four rounds of artificial insemination (AI)) using the Affymetrix® 23 K Bovine Gene Chip. RESULTS Conception rates for each of the four rounds of AI were within a normal range: 70-73.3%. Microarray analysis of endometrial tissue collected on day 7 of the estrous cycle detected 419 differentially expressed genes (DEG) between HF (n = 6) and LF (n = 6) animals. The main gene pathways affected were, cellular growth and proliferation, angiogenesis, lipid metabolism, cellular and tissue morphology and development, inflammation and metabolic exchange. DEG included, FST, SLC45A2, MMP19, FADS1 and GALNT6. CONCLUSIONS This study highlights, some of the molecular mechanisms potentially controlling uterine endometrial function during the mid-luteal phase of the estrous cycle, which may contribute to uterine endometrial mediated impaired fertility in cattle. Differentially expressed genes are potential candidate genes for the identification of genetic variation influencing cow fertility, which may be incorporated into future breeding programmes.
Collapse
Affiliation(s)
| | | | | | | | | | - Sinéad M Waters
- Teagasc, Animal and Bioscience Research Department, Animal and Grassland Research and Innovation Centre, Grange, Dunsany, County Meath, Ireland.
| |
Collapse
|