1
|
Hsieh PS, Hwang SR, Hwang SW, Hwang JH. Plasma Glial Cell-Derived Neurotrophic Factor and Insulin-like Growth Factor-1 Levels Were Not Correlated with the Severity of Age-Related Hearing Impairment in Humans. ACS OMEGA 2024; 9:1757-1761. [PMID: 38222583 PMCID: PMC10785095 DOI: 10.1021/acsomega.3c08354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/24/2023] [Accepted: 11/28/2023] [Indexed: 01/16/2024]
Abstract
The relationship between plasma glial cell-derived neurotrophic factor (GDNF) or insulin-like growth factor-1 (IGF-1) levels and age-related hearing impairment (ARHI) has not been reported in humans. By cross-sectional design, 268 subjects older than 33, with normal cognitive function and normal or symmetric sensorineural hearing loss, were selected randomly. Multivariate linear regression analysis was performed to test the impact of the plasma GDNF or IGF-1 level on the pure tone threshold of low frequencies (PTA-low) and high frequencies (PTA-high), respectively. Results showed that plasma GDNF and IGF-1 levels decreased with age without statistical significance. Multivariate linear regression analysis showed that GDNF or IGF-1 levels were not significantly correlated with PTA-low or PTA-high after adjusting age, gender, body mass index, systemic diseases, habits, and noise exposure. In conclusion, plasma GDNF or IGF-1 levels were not associated with the severity of ARHI in humans. However, these findings did not support the roles of GDNF or IGF-1 genotypes on hearing.
Collapse
Affiliation(s)
- Pei-Shan Hsieh
- Department
of Medical Research, Dalin Tzu Chi Hospital,
Buddhist Tzu Chi Medical Foundation, Chiayi 622, Taiwan
| | - Shang-Rung Hwang
- Department
of Pharmacy, Chia Nan University of Pharmacy
& Science, Tainan 71710, Taiwan
| | - Sheng-Wei Hwang
- School
of Medicine, National Yang Ming Chiao Tung
University, Taipei 112, Taiwan
| | - Juen-Haur Hwang
- Department
of Otolaryngology-Head and Neck Surgery, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi 622, Taiwan
- School
of Medicine, Tzu Chi University, Hualien970,Taiwan
- Department
of Medical Research, China Medical University
Hospital, China Medical University, Taichung404,Taiwan
| |
Collapse
|
2
|
Guo D, Zhang A, Zou T, Ding R, Chen K, Pan Y, Ji P, Ye B, Xiang M. The influence of metabolic syndrome on age-related hearing loss from the perspective of mitochondrial dysfunction. Front Aging Neurosci 2022; 14:930105. [PMID: 35966796 PMCID: PMC9372463 DOI: 10.3389/fnagi.2022.930105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/12/2022] [Indexed: 12/06/2022] Open
Abstract
With the increase in life expectancy in the global population, aging societies have emerged in many countries, including China. As a common sensory defect in the elderly population, the prevalence of age-related hearing loss and its influence on society are increasing yearly. Metabolic syndrome is currently one of the main health problems in the world. Many studies have demonstrated that metabolic syndrome and its components are correlated with a variety of age-related diseases of the peripheral sensory system, including age-related hearing loss. Both age-related hearing loss and metabolic syndrome are high-prevalence chronic diseases, and many people suffer from both at the same time. In recent years, more and more studies have found that mitochondrial dysfunction occurs in both metabolic syndrome and age-related hearing loss. Therefore, to better understand the impact of metabolic syndrome on age-related hearing loss from the perspective of mitochondrial dysfunction, we reviewed the literature related to the relationship between age-related hearing loss and metabolic syndrome and their components to discern the possible role of mitochondria in both conditions.
Collapse
Affiliation(s)
- Dongye Guo
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Andi Zhang
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Tianyuan Zou
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Rui Ding
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Kaili Chen
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Yi Pan
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Peilin Ji
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Bin Ye
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
- *Correspondence: Bin Ye,
| | - Mingliang Xiang
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
- Mingliang Xiang,
| |
Collapse
|
3
|
Xu XM, Wang J, Salvi R, Liu LJ, Chen YC, Teng GJ. Altered resting-state functional connectivity of the anterior cingulate cortex in rats post noise exposure. CNS Neurosci Ther 2022; 28:1547-1556. [PMID: 35726754 PMCID: PMC9437238 DOI: 10.1111/cns.13896] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 04/29/2022] [Accepted: 06/04/2022] [Indexed: 11/30/2022] Open
Abstract
Aims We aimed to find where and how noise‐induced cochlear hearing loss affects the central nervous system during the early state and identify the neural substrate for aberrant patterns that mediating noise‐related anxiety−/depression‐ like behaviors. Methods Broad band noise with 122 dB for 2 hours was conducted to induce hearing loss. We defined 0 day (N0D) and 10 days (N10D) post noise as the acute and sub‐acute period. Behavioral tests (Open field test and light/dark test) and resting‐state fMRI were computed to evaluate emotional conditions and aberrant neural activity. Functional connectivity analysis using the anterior cingulate cortex as a seed was computed to reveal the spatial distribution beyond auditory network during both periods. Results Anxiety−/depression‐like behaviors were found in rats with noise exposure. Between‐group analysis revealed that N0D rats displayed widespread reductions in functional connectivity, spanning primary somatosensory cortex, medial geniculate body, inferior colliculus, cingulate cortex, cerebellar lobule comparing with N10D rats and a similar pattern was also occurred in comparison with the control group. Conclusion Taken together, an “acoustic‐causing” network accounting for distress and gating of noise exposure related anxiety/depression was proposed.
Collapse
Affiliation(s)
- Xiao-Min Xu
- Jiangsu Key Laboratory of Molecular Imaging and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China.,Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jian Wang
- School of Human Communication Disorders, Dalhousie University, Halifax, Canada
| | - Richard Salvi
- Center for Hearing and Deafness, University at Buffalo, Buffalo, New York, USA
| | - Li-Jie Liu
- Department of Physiology, Southeast University, Nanjing, China
| | - Yu-Chen Chen
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Gao-Jun Teng
- Jiangsu Key Laboratory of Molecular Imaging and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| |
Collapse
|
4
|
Luo J, Lin X, Li L, Tan J, Li P. β-Cyclodextrin and Oligoarginine Peptide-Based Dendrimer-Entrapped Gold Nanoparticles for Improving Drug Delivery to the Inner Ear. Front Bioeng Biotechnol 2022; 10:844177. [PMID: 35480970 PMCID: PMC9038081 DOI: 10.3389/fbioe.2022.844177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 02/24/2022] [Indexed: 11/13/2022] Open
Abstract
Here, we developed a safe and highly effective nanocarrier using β-cyclodextrin (β-CD) and oligoarginine peptide (Arg8)-modified dendrimer-entrapped gold nanoparticles (Au@CD-PAMAM-Arg8), with a diameter of 5 nm, for improved delivery of dexamethasone (Dex) to the inner ear. The properties and in vivo distribution of the Au@CD-PAMAM-Arg8 were assessed in vitro, and a streptomycin (SM) ototoxicity model was used in vivo. Flow cytometry analysis of HEIOC1 cells treated with Au@CD-PAMAM-Arg8 and Au @CD-PAMAM at different time intervals indicated that cell uptake efficiency of the drug delivery carrier Au@CD-PAMAM-Arg8 was higher than that of Au @CD-PAMAM. Au@CD-PAMAM-Arg8 carrying Dex (Au@CD-PAMAM-Arg8/Dex) were mainly distributed in hair cells, the spiral ganglion, lateral wall, and nerve fibers and had stronger protective effects on the inner ear than Dex administration alone. In vivo tracer tests revealed that tympanic injection was significantly more effective than posterior ear injection, muscle injection, and tail vein injection, whereas clinical retro-auricular injection could not increase the efficiency of drug delivery into the ear. Electrocochleography results showed that Au@CD-PAMAM-Arg8/Dex significantly improved hearing in C57/BL6 mice after SM exposure. These findings indicate that Au@CD-PAMAM-Arg8 may be the useful drug carriers for the treatment of inner ear diseases.
Collapse
Affiliation(s)
- Jia Luo
- Department of Otolaryngology Head and Neck Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - XueXin Lin
- Department of Otolaryngology Head and Neck Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - LiLing Li
- Department of Otolaryngology Head and Neck Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - JingQian Tan
- Department of Otolaryngology Head and Neck Surgery, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Peng Li
- Department of Otolaryngology Head and Neck Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- *Correspondence: Peng Li,
| |
Collapse
|
5
|
Chern A, Gudis DA, Dagi Glass LR. Teprotumumab and hearing loss: hear the warnings. Orbit 2021; 40:355-356. [PMID: 33622158 DOI: 10.1080/01676830.2021.1886311] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 01/28/2021] [Indexed: 06/12/2023]
Affiliation(s)
- Alexander Chern
- NewYork-Presbyterian/Columbia University Irving Medical Center and Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - David A Gudis
- NewYork-Presbyterian/Columbia University Irving Medical Center and Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Lora R Dagi Glass
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
6
|
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: 11] [Impact Index Per Article: 3.7] [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
|
7
|
Ultrasound Microbubbles Enhance the Efficacy of Insulin-Like Growth Factor-1 Therapy for the Treatment of Noise-Induced Hearing Loss. Molecules 2021; 26:molecules26123626. [PMID: 34199327 PMCID: PMC8231984 DOI: 10.3390/molecules26123626] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/02/2021] [Accepted: 06/11/2021] [Indexed: 11/17/2022] Open
Abstract
The application of insulin-like growth factor 1 (IGF-1) to the round window membrane (RWM) is an emerging treatment for inner ear diseases. RWM permeability is the key factor for efficient IGF-1 delivery. Ultrasound microbubbles (USMBs) can increase drug permeation through the RWM. In the present study, the enhancing effect of USMBs on the efficacy of IGF-1 application and the treatment effect of USMB-mediated IGF-1 delivery for noise-induced hearing loss (NIHL) were investigated. Forty-seven guinea pigs were assigned to three groups: the USM group, which received local application of recombinant human IGF-1 (rhIGF-1, 10 µg/µL) following application of USMBs to the RWM; the RWS group, which received IGF-1 application alone; and the saline-treated group. The perilymphatic concentration of rhIGF-1 in the USM group was 1.95- and 1.67- fold of that in the RWS group, 2 and 24 h after treatment, respectively. After 5 h of 118 dB SPL noise exposure, the USM group had the lowest threshold shift in auditory brainstem response, least loss of cochlear outer hair cells, and least reduction in the number of synaptic ribbons on postexposure day 28 among the three groups. The combination of USMB and IGF-1 led to a better therapeutic response to NIHL. Two hours after treatment, the USM group had significantly higher levels of Akt1 and Mapk3 gene expression than the other two groups. The most intense immunostaining for phosphor-AKT and phospho-ERK1/2 was detected in the cochlea in the USM group. These results suggested that USMB can be applied to enhance the efficacy of IGF-1 therapy in the treatment of inner ear diseases.
Collapse
|
8
|
Chern A, Dagi Glass LR, Gudis DA. Thyroid Eye Disease, Teprotumumab, and Hearing Loss: An Evolving Role for Otolaryngologists. Otolaryngol Head Neck Surg 2021; 165:757-758. [PMID: 33781112 DOI: 10.1177/01945998211004240] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Teprotumumab is a human monoclonal antibody and IGF-1R (insulin-like growth factor 1 receptor) inhibitor approved for treatment of thyroid eye disease in adults. Recent clinical trials have demonstrated side effects, notably hearing loss, in the treatment cohort as compared with the placebo cohort. These unexpected otologic side effects may be understood through a mechanistic understanding of IGF-1 (insulin-like growth factor 1). As otolaryngologists who historically play a significant role in the multidisciplinary treatment of thyroid disease and its associated complications, we should be aware of and monitor the otologic side effects of teprotumumab. Clinicians who prescribe teprotumumab should strongly consider monitoring patients' hearing with an audiologist and otolaryngologist.
Collapse
Affiliation(s)
- Alexander Chern
- Department of Otolaryngology-Head and Neck Surgery, NewYork-Presbyterian/Columbia University Irving Medical Center, and Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Lora R Dagi Glass
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, New York, New York, USA
| | - David A Gudis
- Department of Otolaryngology-Head and Neck Surgery, NewYork-Presbyterian/Columbia University Irving Medical Center, and Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| |
Collapse
|
9
|
Morris KY, Bowman J, Schulte‐Hostedde A, Wilson PJ. Functional genetic diversity of domestic and wild American mink ( Neovison vison). Evol Appl 2020; 13:2610-2629. [PMID: 33294012 PMCID: PMC7691469 DOI: 10.1111/eva.13061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/24/2020] [Accepted: 06/26/2020] [Indexed: 11/30/2022] Open
Abstract
The release of domestic organisms to the wild threatens biodiversity because the introduction of domestic genes through interbreeding can negatively impact wild conspecifics via outbreeding depression. In North America, farmed American mink (Neovison vison) frequently escape captivity, yet the impact of these events on functional genetic diversity of wild mink populations is unclear. We characterized domestic and wild mink in Ontario at 17 trinucleotide microsatellites located in functional genes thought to be associated with traits affected by domestication. We found low functional genetic diversity in both mink types, as only four of 17 genes were variable, yet allele frequencies varied widely between captive and wild populations. To determine whether allele frequencies of wild populations were affected by geographic location, we performed redundancy analysis and spatial analysis of principal components on three polymorphic loci (AR, ATN1 and IGF-1). We found evidence to suggest domestic release events are affecting the functional genetic diversity of wild mink, as sPCA showed clear distinctions between wild individuals near mink farms and those located in areas without mink farms. This is further substantiated through RDA, where spatial location was associated with genetic variation of AR, ATN1 and IGF1.
Collapse
Affiliation(s)
- Kimberley Y. Morris
- Environmental and Life Sciences Graduate ProgramTrent UniversityPeterboroughONCanada
| | - Jeff Bowman
- Environmental and Life Sciences Graduate ProgramTrent UniversityPeterboroughONCanada
- Wildlife Research and Monitoring SectionOntario Ministry of Natural Resources and ForestryPeterboroughONCanada
| | | | - Paul J. Wilson
- Environmental and Life Sciences Graduate ProgramTrent UniversityPeterboroughONCanada
- Department of BiologyTrent UniversityPeterboroughONCanada
| |
Collapse
|
10
|
Frisina RD, Bazard P, Bauer M, Pineros J, Zhu X, Ding B. Translational implications of the interactions between hormones and age-related hearing loss. Hear Res 2020; 402:108093. [PMID: 33097316 DOI: 10.1016/j.heares.2020.108093] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/05/2020] [Accepted: 10/12/2020] [Indexed: 11/30/2022]
Abstract
Provocative research has revealed both positive and negative effects of hormones on hearing as we age; with in some cases, mis-regulation of hormonal levels in instances of medical comorbidities linked to aging, lying at the heart of the problem. Animal model studies have discovered that hormonal fluctuations can sharpen hearing for improved communication and processing of mating calls during reproductive seasons. Sex hormones sometimes have positive effects on auditory processing, as is often the case with estrogen, whereas combinations of estrogen and progesterone, and testosterone, can have negative effects on hearing abilities, particularly in aging subjects. Too much or too little of some hormones can be detrimental, as is the case for aldosterone and thyroid hormones, which generally decline in older individuals. Too little insulin, as in Type 1 diabetics, or poor regulation of insulin, as in Type 2 diabetics, is also harmful to hearing in our aged population. In terms of clinical translational possibilities, hormone therapies can be problematic due to systemic side effects, as has happened for estrogen/progestin combination hormone replacement therapy (HRT) in older women, where the HRT induces a hearing loss. As hormone therapy approaches are further developed, it may be possible to lower needed doses of hormones by combining them with supplements, such as antioxidants. Another option will be to take advantage of emerging technologies for local drug delivery to the inner ear, including biodegradeable, sustained-release hydrogels and micro-pumps which can be implanted in the middle ear near the round window. In closing, exciting research completed to date, summarized in the present report bodes well for emerging biomedical therapies to prevent or treat age-related hearing loss utilizing hormonal strategies.
Collapse
Affiliation(s)
- R D Frisina
- Dept. Medical Engineering, Global Center for Hearing & Speech Research, University of South Florida, Tampa FL, USA; Dept. Communication Sciences & Disorders, Global Center for Hearing & Speech Research, University of South Florida, Tampa FL, USA.
| | - P Bazard
- Dept. Medical Engineering, Global Center for Hearing & Speech Research, University of South Florida, Tampa FL, USA
| | - M Bauer
- Dept. Medical Engineering, Global Center for Hearing & Speech Research, University of South Florida, Tampa FL, USA
| | - J Pineros
- Dept. Medical Engineering, Global Center for Hearing & Speech Research, University of South Florida, Tampa FL, USA
| | - X Zhu
- Dept. Medical Engineering, Global Center for Hearing & Speech Research, University of South Florida, Tampa FL, USA
| | - B Ding
- Dept. Medical Engineering, Global Center for Hearing & Speech Research, University of South Florida, Tampa FL, USA
| |
Collapse
|
11
|
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: 3.0] [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
|
12
|
Alassaf M, Daykin EC, Mathiaparanam J, Wolman MA. Pregnancy-associated plasma protein-aa supports hair cell survival by regulating mitochondrial function. eLife 2019; 8:47061. [PMID: 31205004 PMCID: PMC6594750 DOI: 10.7554/elife.47061] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 06/14/2019] [Indexed: 12/18/2022] Open
Abstract
To support cell survival, mitochondria must balance energy production with oxidative stress. Inner ear hair cells are particularly vulnerable to oxidative stress; thus require tight mitochondrial regulation. We identified a novel molecular regulator of the hair cells’ mitochondria and survival: Pregnancy-associated plasma protein-aa (Pappaa). Hair cells in zebrafish pappaa mutants exhibit mitochondrial defects, including elevated mitochondrial calcium, transmembrane potential, and reactive oxygen species (ROS) production and reduced antioxidant expression. In pappaa mutants, hair cell death is enhanced by stimulation of mitochondrial calcium or ROS production and suppressed by a mitochondrial ROS scavenger. As a secreted metalloprotease, Pappaa stimulates extracellular insulin-like growth factor 1 (IGF1) bioavailability. We found that the pappaa mutants’ enhanced hair cell loss can be suppressed by stimulation of IGF1 availability and that Pappaa-IGF1 signaling acts post-developmentally to support hair cell survival. These results reveal Pappaa as an extracellular regulator of hair cell survival and essential mitochondrial function.
Collapse
Affiliation(s)
- Mroj Alassaf
- Department of Integrative Biology, University of Wisconsin, Madison, United States.,Neuroscience Training Program, University of Wisconsin, Madison, United States
| | - Emily C Daykin
- Department of Integrative Biology, University of Wisconsin, Madison, United States
| | - Jaffna Mathiaparanam
- Department of Integrative Biology, University of Wisconsin, Madison, United States
| | - Marc A Wolman
- Department of Integrative Biology, University of Wisconsin, Madison, United States
| |
Collapse
|
13
|
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: 16] [Impact Index Per Article: 3.2] [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
|
14
|
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: 10] [Impact Index Per Article: 2.0] [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
|
15
|
Paulsen AJ, Cruickshanks KJ, Pinto A, Schubert CR, Dalton DS, Fischer ME, Klein BEK, Klein R, Tsai MY, Tweed TS. Neuroprotective factors and incident hearing impairment in the epidemiology of hearing loss study. Laryngoscope 2019; 129:2178-2183. [PMID: 30698838 DOI: 10.1002/lary.27847] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Hearing impairment (HI) is common in aging adults. Aldosterone, insulin-like growth factor (IGF1), and brain-derived neurotrophic factor (BDNF) have been identified as potentially protective of hearing. The present study aims to investigate these relationships. METHODS The Epidemiology of Hearing Loss Study is a longitudinal population-based study of aging in Beaver Dam, Wisconsin, that began in 1993. Baseline for the present investigation is the 1998 to 2000 phase. Follow-up exams occurred approximately every 5 years, with the most recent occurring from 2013 to 2016. Hearing was measured by pure-tone audiometry. HI was defined as a pure tone average (PTA) > 25 decibels hearing level in either ear. Change in PTA was the difference between follow-up examinations and baseline. Baseline serum samples were used to measure biomarkers in 2017. Hazard ratios (HRs) and 95% confidence intervals (CIs) were estimated to assess the effect of biomarker levels in the lowest quintile (Q1) versus the highest (Q5) on incident HI and PTA change. RESULTS There were 1,088 participants (69.3% women) at risk of HI included in analyses. The mean baseline age was 63.8 years (standard deviation = 7.0). The 16-year incidence of HI was 54.9% and was higher in men (61.1%) than women (52.1%). In age- and sex-adjusted models, aldosterone (HR = 1.06, 95% CI = 0.82-1.37), IGF1 (HR = 0.92, 95% CI = 0.71-1.19), and BDNF (HR = 0.86, 95% CI = 0.66-1.12) levels were not associated with risk of HI. PTA change was similarly not affected by biomarker levels. CONCLUSION Aldosterone, IGF1, and BDNF were not associated with decreased risk of age-related hearing loss in this study. LEVEL OF EVIDENCE 2b Laryngoscope, 129:2178-2183, 2019.
Collapse
Affiliation(s)
- Adam J Paulsen
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Karen J Cruickshanks
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin.,Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Alex Pinto
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Carla R Schubert
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Dayna S Dalton
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Mary E Fischer
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Barbara E K Klein
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Ronald Klein
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Michael Y Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School-Minneapolis, Minneapolis, Minnesota, U.S.A
| | - Ted S Tweed
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
16
|
Insulin-like growth factor 1 modulates bioengineered tooth morphogenesis. Sci Rep 2019; 9:368. [PMID: 30675004 PMCID: PMC6344556 DOI: 10.1038/s41598-018-36863-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 11/21/2018] [Indexed: 11/08/2022] Open
Abstract
Regenerative therapy to replace missing teeth is a critical area of research. Functional bioengineered teeth have been produced by the organ germ method using mouse tooth germ cells. However, these bioengineered teeth are significantly smaller in size and exhibit an abnormal crown shape when compared with natural teeth. The proper sizes and shapes of teeth contribute to their normal function. Therefore, a method is needed to control the morphology of bioengineered teeth. Here, we investigated whether insulin-like growth factor 1 (IGF1) can regulate the sizes and shapes of bioengineered teeth, and assessed underlying mechanisms of such regulation. IGF1 treatment significantly increased the size of bioengineered tooth germs, while preserving normal tooth histology. IGF1-treated bioengineered teeth, which were developed from bioengineered tooth germs in subrenal capsules and jawbones, showed increased sizes and cusp numbers. IGF1 increased the number of fibroblast growth factor (Fgf4)-expressing enamel knots in bioengineered tooth germs and enhanced the proliferation and differentiation of dental epithelial and mesenchymal cells. This study is the first to reveal that IGF1 increases the sizes and cusp numbers of bioengineered teeth via the induction of enamel knot formation, as well as the proliferation and differentiation of dental epithelial and mesenchymal cells.
Collapse
|
17
|
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: 5.8] [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
|
18
|
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: 22] [Impact Index Per Article: 3.1] [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
|
19
|
Lassale C, Batty GD, Steptoe A, Zaninotto P. Insulin-like Growth Factor 1 in relation to future hearing impairment: findings from the English Longitudinal Study of Ageing. Sci Rep 2017. [PMID: 28646237 PMCID: PMC5482884 DOI: 10.1038/s41598-017-04526-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Insulin-like Growth Factor 1 (IGF-1) is associated with cardiovascular disease, itself a risk factor for hearing impairment, and, in animal studies, molecular evidence suggests a role for IGF-1 in hearing function. However, the link between IGF-1 and the occurrence of hearing impairment is untested in population-based studies of humans. A total of 4390 participants aged ≥50 y (mean [SD] age 64.2 [8.0] years at baseline, 55% women) from the English Longitudinal Study of Ageing provided serum levels of IGF-1 in 2008 and again in 2012. Hearing acuity was assessed by an objective hearing test (HearCheck handheld device) in 2014 when the prevalence was 38.2%. In the full cohort, IGF-1 was not associated with subsequent hearing impairment (OR5nmol/L increase; 95% CI: 1.01; 0.94, 1.09). However, this relationship appeared to differ by age (p-value for interaction = 0.03). Thus, in younger participants (aged 50–60 y, n = 1400), IGF-1 was associated with lower odds of hearing impairment (0.86; 0.73, 1.00) after adjustment for a range of potential confounders. Among people ≥60 y (n = 2990) there was a non-significant ‘J’-shaped association. Our observational evidence that higher levels of IGF-1 appeared to confer some protection against hearing impairment in some older adults warrants replication in other prospective cohort studies.
Collapse
Affiliation(s)
- Camille Lassale
- Department of Epidemiology & Public Health, University College London, London, WC1E 6BT, United Kingdom.
| | - G David Batty
- Department of Epidemiology & Public Health, University College London, London, WC1E 6BT, United Kingdom
| | - Andrew Steptoe
- Department of Epidemiology & Public Health, University College London, London, WC1E 6BT, United Kingdom
| | - Paola Zaninotto
- Department of Epidemiology & Public Health, University College London, London, WC1E 6BT, United Kingdom
| |
Collapse
|
20
|
Magariños M, Pulido S, Aburto MR, de Iriarte Rodríguez R, Varela-Nieto I. Autophagy in the Vertebrate Inner Ear. Front Cell Dev Biol 2017; 5:56. [PMID: 28603711 PMCID: PMC5445191 DOI: 10.3389/fcell.2017.00056] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 05/09/2017] [Indexed: 12/13/2022] Open
Abstract
Autophagy is a conserved catabolic process that results in the lysosomal degradation of cell components. During development, autophagy is associated with tissue and organ remodeling, and under physiological conditions it is tightly regulated as it plays a housekeeping role in removing misfolded proteins and damaged organelles. The vertebrate inner ear is a complex sensory organ responsible for the perception of sound and for balance. Cell survival, death and proliferation, as well as cell fate specification and differentiation, are processes that are strictly coordinated during the development of the inner ear in order to generate the more than a dozen specialized cell types that constitute this structure. Here, we review the existing evidence that implicates autophagy in the generation of the vertebrate inner ear. At early stages of chicken otic development, inhibiting autophagy impairs neurogenesis and causes aberrant otocyst morphogenesis. Autophagy provides energy for the clearing of dying cells and it favors neuronal differentiation. Moreover, autophagy is required for proper vestibular development in the mouse inner ear. The autophagy-related genes Becn1, Atg4g, Atg5, and Atg9, are expressed in the inner ear from late developmental stages to adulthood, and Atg4b mutants show impaired vestibular behavior associated to defects in otoconial biogenesis that are also common to Atg5 mutants. Autophagic flux appears to be age-regulated, augmenting from perinatal stages to young adulthood in mice. This up-regulation is concomitant with the functional maturation of the hearing receptor. Hence, autophagy can be considered an intracellular pathway fundamental for in vertebrate inner ear development and maturation.
Collapse
Affiliation(s)
- Marta Magariños
- Department of Endocrine and Nervous Systems Pathophysiology, Instituto de Investigaciones Biomédicas "Alberto Sols," CSIC-UAMMadrid, Spain.,CIBERER, Unit 761, Instituto de Salud Carlos IIIMadrid, Spain.,Departamento de Biología, Universidad Autónoma de MadridMadrid, Spain
| | - Sara Pulido
- Department of Endocrine and Nervous Systems Pathophysiology, Instituto de Investigaciones Biomédicas "Alberto Sols," CSIC-UAMMadrid, Spain.,CIBERER, Unit 761, Instituto de Salud Carlos IIIMadrid, Spain
| | - María R Aburto
- Department of Endocrine and Nervous Systems Pathophysiology, Instituto de Investigaciones Biomédicas "Alberto Sols," CSIC-UAMMadrid, Spain
| | - Rocío de Iriarte Rodríguez
- Department of Endocrine and Nervous Systems Pathophysiology, Instituto de Investigaciones Biomédicas "Alberto Sols," CSIC-UAMMadrid, Spain
| | - Isabel Varela-Nieto
- Department of Endocrine and Nervous Systems Pathophysiology, Instituto de Investigaciones Biomédicas "Alberto Sols," CSIC-UAMMadrid, Spain.,CIBERER, Unit 761, Instituto de Salud Carlos IIIMadrid, Spain.,Instituto de Investigación Hospital Universitario La Paz (IdiPAZ)Madrid, Spain
| |
Collapse
|
21
|
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: 1.6] [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
|
22
|
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: 1.9] [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
|
23
|
Quantitative map of multiple auditory cortical regions with a stereotaxic fine-scale atlas of the mouse brain. Sci Rep 2016; 6:22315. [PMID: 26924462 PMCID: PMC4770424 DOI: 10.1038/srep22315] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 02/12/2016] [Indexed: 11/08/2022] Open
Abstract
Optical imaging studies have recently revealed the presence of multiple auditory cortical regions in the mouse brain. We have previously demonstrated, using flavoprotein fluorescence imaging, at least six regions in the mouse auditory cortex, including the anterior auditory field (AAF), primary auditory cortex (AI), the secondary auditory field (AII), dorsoanterior field (DA), dorsomedial field (DM), and dorsoposterior field (DP). While multiple regions in the visual cortex and somatosensory cortex have been annotated and consolidated in recent brain atlases, the multiple auditory cortical regions have not yet been presented from a coronal view. In the current study, we obtained regional coordinates of the six auditory cortical regions of the C57BL/6 mouse brain and illustrated these regions on template coronal brain slices. These results should reinforce the existing mouse brain atlases and support future studies in the auditory cortex.
Collapse
|
24
|
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: 1.6] [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
|
25
|
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: 20] [Impact Index Per Article: 2.2] [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
|
26
|
Sanz L, Murillo-Cuesta S, Cobo P, Cediel-Algovia R, Contreras J, Rivera T, Varela-Nieto I, Avendaño C. Swept-sine noise-induced damage as a hearing loss model for preclinical assays. Front Aging Neurosci 2015; 7:7. [PMID: 25762930 PMCID: PMC4329813 DOI: 10.3389/fnagi.2015.00007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 01/19/2015] [Indexed: 11/27/2022] Open
Abstract
Mouse models are key tools for studying cochlear alterations in noise-induced hearing loss (NIHL) and for evaluating new therapies. Stimuli used to induce deafness in mice are usually white and octave band noises that include very low frequencies, considering the large mouse auditory range. We designed different sound stimuli, enriched in frequencies up to 20 kHz (“violet” noises) to examine their impact on hearing thresholds and cochlear cytoarchitecture after short exposure. In addition, we developed a cytocochleogram to quantitatively assess the ensuing structural degeneration and its functional correlation. Finally, we used this mouse model and cochleogram procedure to evaluate the potential therapeutic effect of transforming growth factor β1 (TGF-β1) inhibitors P17 and P144 on NIHL. CBA mice were exposed to violet swept-sine noise (VS) with different frequency ranges (2–20 or 9–13 kHz) and levels (105 or 120 dB SPL) for 30 min. Mice were evaluated by auditory brainstem response (ABR) and otoacoustic emission tests prior to and 2, 14 and 28 days after noise exposure. Cochlear pathology was assessed with gross histology; hair cell number was estimated by a stereological counting method. Our results indicate that functional and morphological changes induced by VS depend on the sound level and frequency composition. Partial hearing recovery followed the exposure to 105 dB SPL, whereas permanent cochlear damage resulted from the exposure to 120 dB SPL. Exposure to 9–13 kHz noise caused an auditory threshold shift (TS) in those frequencies that correlated with hair cell loss in the corresponding areas of the cochlea that were spotted on the cytocochleogram. In summary, we present mouse models of NIHL, which depending on the sound properties of the noise, cause different degrees of cochlear damage, and could therefore be used to study molecules which are potential players in hearing loss protection and repair.
Collapse
Affiliation(s)
- Lorena Sanz
- 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
| | - 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
| | - Pedro Cobo
- Institute for Physical and Information Technologies (ITEFI), Spanish National Research Council (CSIC) Madrid, Spain
| | - Rafael Cediel-Algovia
- 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
| | - 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
| | - 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
| | - Carlos Avendaño
- Hospital La Paz Institute for Health Research (IdiPAZ) Madrid, Spain ; Department of Anatomy, Histology and Neuroscience, Medical School, Autónoma University of Madrid Madrid, Spain
| |
Collapse
|
27
|
Milde S, Adalbert R, Elaman MH, Coleman MP. Axonal transport declines with age in two distinct phases separated by a period of relative stability. Neurobiol Aging 2014; 36:971-81. [PMID: 25443288 PMCID: PMC4321880 DOI: 10.1016/j.neurobiolaging.2014.09.018] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 09/12/2014] [Accepted: 09/17/2014] [Indexed: 01/20/2023]
Abstract
Axonal transport is critical for supplying newly synthesized proteins, organelles, mRNAs, and other cargoes from neuronal cell bodies into axons. Its impairment in many neurodegenerative conditions appears likely to contribute to pathogenesis. Axonal transport also declines during normal aging, but little is known about the timing of these changes, or about the effect of aging on specific cargoes in individual axons. This is important for understanding mechanisms of age-related axon loss and age-related axonal disorders. Here we use fluorescence live imaging of peripheral nerve and central nervous system tissue explants to investigate vesicular and mitochondrial axonal transport. Interestingly, we identify 2 distinct periods of change, 1 period during young adulthood and the other in old age, separated by a relatively stable plateau during most of adult life. We also find that after tibial nerve regeneration, even in old animals, neurons are able to support higher transport rates of each cargo for a prolonged period. Thus, the age-related decline in axonal transport is not an inevitable consequence of either aging neurons or an aging systemic milieu.
Collapse
Affiliation(s)
- Stefan Milde
- Signalling ISP, The Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Robert Adalbert
- Signalling ISP, The Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - M Handan Elaman
- Signalling ISP, The Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Michael P Coleman
- Signalling ISP, The Babraham Institute, Babraham Research Campus, Cambridge, UK.
| |
Collapse
|
28
|
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: 3.2] [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
|
29
|
Sensational placodes: neurogenesis in the otic and olfactory systems. Dev Biol 2014; 389:50-67. [PMID: 24508480 PMCID: PMC3988839 DOI: 10.1016/j.ydbio.2014.01.023] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 01/27/2014] [Accepted: 01/28/2014] [Indexed: 11/22/2022]
Abstract
For both the intricate morphogenetic layout of the sensory cells in the ear and the elegantly radial arrangement of the sensory neurons in the nose, numerous signaling molecules and genetic determinants are required in concert to generate these specialized neuronal populations that help connect us to our environment. In this review, we outline many of the proteins and pathways that play essential roles in the differentiation of otic and olfactory neurons and their integration into their non-neuronal support structures. In both cases, well-known signaling pathways together with region-specific factors transform thickened ectodermal placodes into complex sense organs containing numerous, diverse neuronal subtypes. Olfactory and otic placodes, in combination with migratory neural crest stem cells, generate highly specialized subtypes of neuronal cells that sense sound, position and movement in space, odors and pheromones throughout our lives.
Collapse
|
30
|
Rodríguez-de la Rosa L, López-Herradón A, Portal-Núñez S, Murillo-Cuesta S, Lozano D, Cediel R, Varela-Nieto I, Esbrit P. Treatment with N- and C-terminal peptides of parathyroid hormone-related protein partly compensate the skeletal abnormalities in IGF-I deficient mice. PLoS One 2014; 9:e87536. [PMID: 24503961 PMCID: PMC3913635 DOI: 10.1371/journal.pone.0087536] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 12/31/2013] [Indexed: 11/18/2022] Open
Abstract
Insulin-like growth factor-I (IGF-I) deficiency causes growth delay, and IGF-I has been shown to partially mediate bone anabolism by parathyroid hormone (PTH). PTH-related protein (PTHrP) is abundant in bone, and has osteogenic features by poorly defined mechanisms. We here examined the capacity of PTHrP (1-36) and PTHrP (107-111) (osteostatin) to reverse the skeletal alterations associated with IGF-I deficiency. Igf1-null mice and their wild type littermates were treated with each PTHrP peptide (80 µg/Kg/every other day/2 weeks; 2 males and 4 females for each genotype) or saline vehicle (3 males and 3 females for each genotype). We found that treatment with either PTHrP peptide ameliorated trabecular structure in the femur in both genotypes. However, these peptides were ineffective in normalizing the altered cortical structure at this bone site in Igf1-null mice. An aberrant gene expression of factors associated with osteoblast differentiation and function, namely runx2, osteoprotegerin/receptor activator of NF-κB ligand ratio, Wnt3a , cyclin D1, connexin 43, catalase and Gadd45, as well as in osteocyte sclerostin, was found in the long bones of Igf1-null mice. These mice also displayed a lower amount of trabecular osteoblasts and osteoclasts in the tibial metaphysis than those in wild type mice. These alterations in Igf1-null mice were only partially corrected by each PTHrP peptide treatment. The skeletal expression of Igf2, Igf1 receptor and Irs2 was increased in Igf1-null mice, and this compensatory profile was further improved by treatment with each PTHrP peptide related to ERK1/2 and FoxM1 activation. In vitro, PTHrP (1-36) and osteostatin were effective in promoting bone marrow stromal cell mineralization in normal mice but not in IGF-I-deficient mice. Collectively, these findings indicate that PTHrP (1-36) and osteostatin can exert several osteogenic actions even in the absence of IGF-I in the mouse bone.
Collapse
Affiliation(s)
- Lourdes Rodríguez-de la Rosa
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Centro Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
- Unidad 761, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Hospital Universitario La Paz, Madrid, Spain
| | - Ana López-Herradón
- Laboratorio de Metabolismo Mineral y Óseo, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Madrid, Spain
- Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad, Instituto de Salud Carlos III, Madrid, Spain
| | - Sergio Portal-Núñez
- Laboratorio de Metabolismo Mineral y Óseo, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Madrid, Spain
- Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad, Instituto de Salud Carlos III, Madrid, Spain
| | - Silvia Murillo-Cuesta
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Centro Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
- Unidad 761, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Hospital Universitario La Paz, Madrid, Spain
| | - Daniel Lozano
- Instituto de Investigación Hospital Universitario La Paz, Madrid, Spain
- Laboratorio de Metabolismo Mineral y Óseo, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Madrid, Spain
- Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad, Instituto de Salud Carlos III, Madrid, Spain
| | - Rafael Cediel
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Centro Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
- Unidad 761, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Isabel Varela-Nieto
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Centro Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
- Unidad 761, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Hospital Universitario La Paz, Madrid, Spain
| | - Pedro Esbrit
- Laboratorio de Metabolismo Mineral y Óseo, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Madrid, Spain
- Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad, Instituto de Salud Carlos III, Madrid, Spain
- * E-mail:
| |
Collapse
|
31
|
Merchán MA, Saldaña E, Oliver DL. Auditory neuroanatomy: a sound foundation for sound processing. Front Neuroanat 2012; 6:48. [PMID: 23230396 PMCID: PMC3515782 DOI: 10.3389/fnana.2012.00048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 11/21/2012] [Indexed: 11/30/2022] Open
Affiliation(s)
- Miguel A Merchán
- Laboratory 6, Neuroscience Institute of Castilla y León, Universidad de Salamanca Salamanca, Spain
| | | | | |
Collapse
|
32
|
Murillo-Cuesta S, Camarero G, González-Rodríguez A, De La Rosa LR, Burks DJ, Avendaño C, Valverde AM, Varela-Nieto I. Insulin receptor substrate 2 (IRS2)-deficient mice show sensorineural hearing loss that is delayed by concomitant protein tyrosine phosphatase 1B (PTP1B) loss of function. Mol Med 2012; 18:260-9. [PMID: 22160220 DOI: 10.2119/molmed.2011.00328] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2011] [Accepted: 11/29/2011] [Indexed: 01/28/2023] Open
Abstract
The insulin receptor substrate (IRS) proteins are key mediators of insulin and insulinlike growth factor 1 (IGF-1) signaling. Protein tyrosine phosphatase (PTP)-1B dephosphorylates and inactivates both insulin and IGF-1 receptors. IRS2-deficient mice present altered hepatic insulin signaling and β-cell failure and develop type 2-like diabetes. In addition, IRS2 deficiency leads to developmental defects in the nervous system. IGF1 gene mutations cause syndromic sensorineural hearing loss in humans and mice. However, the involvement of IRS2 and PTP1B, two IGF-1 downstream signaling mediators, in hearing onset and loss has not been studied. Our objective was to study the hearing function and cochlear morphology of Irs2-null mice and the impact of PTP1B deficiency. We have studied the auditory brainstem responses and the cochlear morphology of systemic Irs2⁻/⁻Ptpn1⁺/⁺, Irs2⁺/⁺Ptpn1⁻/⁻ and Irs2⁻/⁻Ptpn1⁻/⁻ mice at different postnatal ages. The results indicated that Irs2⁻/⁻Ptpn1⁺/⁺ mice present a profound congenital sensorineural deafness before the onset of diabetes and altered cochlear morphology with hypoinnervation of the cochlear ganglion and aberrant stria vascularis, compared with wild-type mice. Simultaneous PTP1B deficiency in Irs2⁻/⁻Ptpn1⁻/⁻ mice delays the onset of deafness. We show for the first time that IRS2 is essential for hearing and that PTP1B inhibition may be useful for treating deafness associated with hyperglycemia and type 2 diabetes.
Collapse
Affiliation(s)
- Silvia Murillo-Cuesta
- Institute of Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid-CSIC-UAM, Madrid, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Aburto MR, Magariños M, Leon Y, Varela-Nieto I, Sanchez-Calderon H. AKT signaling mediates IGF-I survival actions on otic neural progenitors. PLoS One 2012; 7:e30790. [PMID: 22292041 PMCID: PMC3264639 DOI: 10.1371/journal.pone.0030790] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 12/29/2011] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Otic neurons and sensory cells derive from common progenitors whose transition into mature cells requires the coordination of cell survival, proliferation and differentiation programmes. Neurotrophic support and survival of post-mitotic otic neurons have been intensively studied, but the bases underlying the regulation of programmed cell death in immature proliferative otic neuroblasts remains poorly understood. The protein kinase AKT acts as a node, playing a critical role in controlling cell survival and cell cycle progression. AKT is activated by trophic factors, including insulin-like growth factor I (IGF-I), through the generation of the lipidic second messenger phosphatidylinositol 3-phosphate by phosphatidylinositol 3-kinase (PI3K). Here we have investigated the role of IGF-dependent activation of the PI3K-AKT pathway in maintenance of otic neuroblasts. METHODOLOGY/PRINCIPAL FINDINGS By using a combination of organotypic cultures of chicken (Gallus gallus) otic vesicles and acoustic-vestibular ganglia, Western blotting, immunohistochemistry and in situ hybridization, we show that IGF-I-activation of AKT protects neural progenitors from programmed cell death. IGF-I maintains otic neuroblasts in an undifferentiated and proliferative state, which is characterised by the upregulation of the forkhead box M1 (FoxM1) transcription factor. By contrast, our results indicate that post-mitotic p27(Kip)-positive neurons become IGF-I independent as they extend their neuronal processes. Neurons gradually reduce their expression of the Igf1r, while they increase that of the neurotrophin receptor, TrkC. CONCLUSIONS/SIGNIFICANCE Proliferative otic neuroblasts are dependent on the activation of the PI3K-AKT pathway by IGF-I for survival during the otic neuronal progenitor phase of early inner ear development.
Collapse
Affiliation(s)
- Maria R. Aburto
- Instituto de Investigaciones Biomedicas “Alberto Sols”, CSIC-UAM, Madrid, Spain
- CIBERER, Unit 761, ISCIII, Madrid, Spain
| | - Marta Magariños
- Instituto de Investigaciones Biomedicas “Alberto Sols”, CSIC-UAM, Madrid, Spain
- CIBERER, Unit 761, ISCIII, Madrid, Spain
- Departamento de Biologia, Universidad Autonoma de Madrid, Madrid, Spain
| | - Yolanda Leon
- Instituto de Investigaciones Biomedicas “Alberto Sols”, CSIC-UAM, Madrid, Spain
- Departamento de Biologia, Universidad Autonoma de Madrid, Madrid, Spain
| | - Isabel Varela-Nieto
- Instituto de Investigaciones Biomedicas “Alberto Sols”, CSIC-UAM, Madrid, Spain
- CIBERER, Unit 761, ISCIII, Madrid, Spain
| | - Hortensia Sanchez-Calderon
- Instituto de Investigaciones Biomedicas “Alberto Sols”, CSIC-UAM, Madrid, Spain
- CIBERER, Unit 761, ISCIII, Madrid, Spain
| |
Collapse
|
34
|
Murillo-Cuesta S, Rodríguez-de la Rosa L, Cediel R, Lassaletta L, Varela-Nieto I. The role of insulin-like growth factor-I in the physiopathology of hearing. Front Mol Neurosci 2011; 4:11. [PMID: 21845174 PMCID: PMC3146045 DOI: 10.3389/fnmol.2011.00011] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Accepted: 07/11/2011] [Indexed: 01/19/2023] Open
Abstract
Insulin-like growth factor-I (IGF-I) belongs to the family of polypeptides of insulin, which play a central role in embryonic development and adult nervous system homeostasis by endocrine, autocrine, and paracrine mechanisms. IGF-I is fundamental for the regulation of cochlear development, growth, and differentiation, and its mutations are associated with hearing loss in mice and men. Low levels of IGF-I have been shown to correlate with different human syndromes showing hearing loss and with presbyacusis. Animal models are fundamental to understand the genetic, epigenetic, and environmental factors that contribute to human hearing loss. In the mouse, IGF-I serum levels decrease with aging and there is a concomitant hearing loss and retinal degeneration. In the Igf1(-/-) null mouse, hearing loss is due to neuronal loss, poor innervation of the sensory hair cells, and age-related stria vascularis alterations. In the inner ear, IGF-I actions are mediated by intracellular signaling networks, RAF, AKT, and p38 MAPK protein kinases modulate the expression and activity of transcription factors, as AP1, MEF2, FoxM1, and FoxP3, leading to the regulation of cell cycle and metabolism. Therapy with rhIGF-I has been approved in humans for the treatment of poor linear growth and certain neurodegenerative diseases. This review will discuss these findings and their implications in new IGF-I-based treatments for the protection or repair of hearing loss.
Collapse
Affiliation(s)
- Silvia Murillo-Cuesta
- Servicio de Evaluación Neurofuncional no Invasiva, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid Madrid, Spain
| | | | | | | | | |
Collapse
|