1
|
Wang Y, Huang L, Cen X, Liang Y, Chen K. Canonical MAPK signaling in auditory neuropathy. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167619. [PMID: 39662753 DOI: 10.1016/j.bbadis.2024.167619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 11/27/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024]
Abstract
Auditory neuropathy (AN) is an under-recognized form of hearing loss characterized by lesions in inner hair cells (IHCs), ribbon synapses and spiral ganglion neurons (SGNs). The lack of a targeted therapy for AN has increased the need for a better understanding of the pathogenic mechanism of AN. As mitogen-activated protein kinase (MAPK) signaling is ubiquitous in many biological processes, its alteration may facilitate the pathogenesis of multiple sites in AN. Here, we summaries the characteristics of AN under different molecular bases and first explore the mechanism of MAPK at different lesion sites. Alterations of extracellular signal-regulated kinase (ERK)/MAPK occur in IHCs and SGNs, whereas modulations of p38 and c-Jun NH2-terminal kinase (JNK) were found in ribbon synapses and SGNs. In conclusion, inductive MAPK alterations in the pathogenesis and development of AN are likely to represent a potential therapeutic target to guide the development of treatments.
Collapse
Affiliation(s)
- Yueying Wang
- Otorhinolaryngology Hospital, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Institute of Otorhinolaryngology, Sun Yat-sen University, Guangzhou 510080, China
| | - Lusha Huang
- Otorhinolaryngology Hospital, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Institute of Otorhinolaryngology, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiaoqing Cen
- Otorhinolaryngology Hospital, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Institute of Otorhinolaryngology, Sun Yat-sen University, Guangzhou 510080, China
| | - Yue Liang
- Otorhinolaryngology Hospital, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Institute of Otorhinolaryngology, Sun Yat-sen University, Guangzhou 510080, China
| | - Kaitian Chen
- Otorhinolaryngology Hospital, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Institute of Otorhinolaryngology, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
2
|
Toms M, Heppell C, Owen N, Malka S, Moosajee M. A Novel De Novo Missense Variant in Netrin-1 (NTN1) Associated With Chorioretinal Coloboma, Sensorineural Hearing Loss and Polydactyly. Clin Genet 2025; 107:292-299. [PMID: 39648562 PMCID: PMC11790524 DOI: 10.1111/cge.14651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/07/2024] [Accepted: 11/10/2024] [Indexed: 12/10/2024]
Abstract
Microphthalmia, anophthalmia and coloboma (MAC) comprise a highly heterogeneous spectrum of congenital ocular malformations with an estimated incidence of 1 in 5000 to 1 in 30 000 live births. Although there is likely to be a genetic component in the majority of cases, many remain without a molecular diagnosis. Netrin-1 was previously identified as a mediator of optic fissure closure from transcriptome analyses of chick and zebrafish and was shown to cause ocular coloboma when knocked out in both mouse and zebrafish. Here, we report the first patient with chorioretinal coloboma and microphthalmia harbouring a novel heterozygous likely pathogenic NTN1 missense variant, c.1483T>A p.(Tyr495Asn), validating a conserved gene function in ocular development. In addition, the patient displayed bilateral sensorineural hearing loss which was investigated by examining the sensory hair cells of ntn1a morphant zebrafish, suggesting a role for netrin-1 in hair cell development.
Collapse
Affiliation(s)
- Maria Toms
- Development, Ageing and DiseaseUCL Institute of OphthalmologyLondonUK
- The Francis Crick InstituteLondonUK
| | - Cara Heppell
- Department of GeneticsMoorfields Eye Hospital NHS Foundation TrustLondonUK
| | - Nicholas Owen
- Development, Ageing and DiseaseUCL Institute of OphthalmologyLondonUK
| | - Samantha Malka
- Department of GeneticsMoorfields Eye Hospital NHS Foundation TrustLondonUK
| | - Mariya Moosajee
- Development, Ageing and DiseaseUCL Institute of OphthalmologyLondonUK
- The Francis Crick InstituteLondonUK
- Department of GeneticsMoorfields Eye Hospital NHS Foundation TrustLondonUK
| | | |
Collapse
|
3
|
Cui X, Huang C, Huang Y, Zhang Y, Wu J, Wang G, Zhou XZ, Zhang J, Wang L, Cheng L, Zhang KQ. Amplification of Metalloregulatory Proteins in Macrophages by Bioactive ZnMn@SF Hydrogels for Spinal Cord Injury Repair. ACS NANO 2024; 18:33614-33628. [PMID: 39579147 DOI: 10.1021/acsnano.4c12236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2024]
Abstract
Macrophages are rapidly activated and polarized toward the M1 phenotype after spinal cord injury (SCI), and inhibiting M1-like macrophages has emerged as a promising SCI treatment approach. Metalloregulatory proteins, which sense specific metal ions with high affinity and specificity, play a critical role in immune regulation. Here, we screened various bioactive metal ions associated with metalloregulatory proteins and discovered that Zn2+ and Mn2+ effectively suppressed M1 polarization. Based on these findings, mildly alkaline ZnMn-based layered double hydroxides (ZnMn-LDHs) self-assembled from Zn2+ coordinated with Mn2+ were developed to inhibit M1-like macrophages. ZnMn-LDHs effectively neutralized the acidic environment and promoted the expression of metalloregulatory proteins, including metallothionein (MT), superoxide dismutase 1 (SOD1), and superoxide dismutase 2 (SOD2), thereby eliciting robust M1-like macrophage inhibition. More importantly, nerve growth factor (NGF) released by macrophages following the regulation by ZnMn-LDHs promoted the elongation and spreading of Schwann cells. By integrating ZnMn-LDHs with silk fibroin (SF), ZnMn@SF injectable hydrogels were constructed for SCI repair. An in vivo animal model further revealed the excellent anti-inflammatory effects of the ZnMn@SF hydrogels in treating SCI, which promoted functional recovery. Our findings underscore the importance of metalloregulatory proteins regulated by metal ions in inhibiting M1-like macrophages, providing a promising therapeutic strategy for SCI treatment.
Collapse
Affiliation(s)
- Xiaoliang Cui
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| | - Cheng Huang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Yechen Huang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Yuxuan Zhang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Jie Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Gang Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Xiao-Zhong Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Jun Zhang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| | - Li Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Ke-Qin Zhang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| |
Collapse
|
4
|
Hu Y, Sun Y, Yuan H, Liu J, Chen L, Liu D, Xu Y, Zhou X, Ding L, Zhang Z, Xiong L, Xue L, Wang T. Vof16-miR-185-5p-GAP43 network improves the outcomes following spinal cord injury via enhancing self-repair and promoting axonal growth. CNS Neurosci Ther 2024; 30:e14535. [PMID: 38168094 PMCID: PMC11017428 DOI: 10.1111/cns.14535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/31/2023] [Accepted: 11/04/2023] [Indexed: 01/05/2024] Open
Abstract
INTRODUCTION Self-repair of spinal cord injury (SCI) has been found in humans and experimental animals with partial recovery of neurological functions. However, the regulatory mechanisms underlying the spontaneous locomotion recovery after SCI are elusive. AIMS This study was aimed at evaluating the pathological changes in injured spinal cord and exploring the possible mechanism related to the spontaneous recovery. RESULTS Immunofluorescence staining was performed to detect GAP43 expression in lesion site after spinal cord transection (SCT) in rats. Then RNA sequencing and gene ontology (GO) analysis were employed to predict lncRNA that correlates with GAP43. LncRNA smart-silencing was applied to verify the function of lncRNA vof16 in vitro, and knockout rats were used to evaluate its role in neurobehavioral functions after SCT. MicroRNA sequencing, target scan, and RNA22 prediction were performed to further explore the underlying regulatory mechanisms, and miR-185-5p stands out. A miR-185-5p site-regulated relationship with GAP43 and vof16 was determined by luciferase activity analysis. GAP43-silencing, miR-185-5p-mimic/inhibitor, and miR-185-5p knockout rats were also applied to elucidate their effects on spinal cord neurite growth and neurobehavioral function after SCT. We found that a time-dependent increase of GAP43 corresponded with the limited neurological recovery in rats with SCT. CRNA chip and GO analysis revealed lncRNA vof16 was the most functional in targeting GAP43 in SCT rats. Additionally, silencing vof16 suppressed neurite growth and attenuated the motor dysfunction in SCT rats. Luciferase reporter assay showed that miR-185-5p competitively bound the same regulatory region of vof16 and GAP43. CONCLUSIONS Our data indicated miR-185-5p could be a detrimental factor in SCT, and vof16 may function as a ceRNA by competitively binding miR-185-5p to modulate GAP43 in the process of self-recovery after SCT. Our study revealed a novel vof16-miR-185-5p-GAP43 regulatory network in neurological self-repair after SCT and may underlie the potential treatment target for SCI.
Collapse
Affiliation(s)
- Yue Hu
- Department of Anesthesiology, Institute of Neurological Disease, Translational Neuroscience Center, West China HospitalSichuan UniversityChengduChina
- Department of Anesthesia Operation, The First People's Hospital of Shuangliu DistrictWest China Airport Hospital of Sichuan UniversityChengduChina
| | - Yi‐Fei Sun
- Department of Anesthesiology, Institute of Neurological Disease, Translational Neuroscience Center, West China HospitalSichuan UniversityChengduChina
| | - Hao Yuan
- Laboratory Zoology Department, Institute of NeuroscienceKunming Medical UniversityKunmingChina
| | - Jia Liu
- Laboratory Zoology Department, Institute of NeuroscienceKunming Medical UniversityKunmingChina
| | - Li Chen
- Department of Anesthesiology, Institute of Neurological Disease, Translational Neuroscience Center, West China HospitalSichuan UniversityChengduChina
| | - Dong‐Hui Liu
- Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Yang Xu
- Department of Anesthesiology, Institute of Neurological Disease, Translational Neuroscience Center, West China HospitalSichuan UniversityChengduChina
| | - Xin‐Fu Zhou
- Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Li Ding
- Department of Anesthesiology, Institute of Neurological Disease, Translational Neuroscience Center, West China HospitalSichuan UniversityChengduChina
| | - Ze‐Tao Zhang
- Department of Anesthesiology, Institute of Neurological Disease, Translational Neuroscience Center, West China HospitalSichuan UniversityChengduChina
| | - Liu‐Lin Xiong
- Department of AnesthesiologyAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Lu‐Lu Xue
- State Key Laboratory of BiotherapySichuan UniversityChengduSichuanChina
| | - Ting‐Hua Wang
- Department of Anesthesiology, Institute of Neurological Disease, Translational Neuroscience Center, West China HospitalSichuan UniversityChengduChina
- Laboratory Zoology Department, Institute of NeuroscienceKunming Medical UniversityKunmingChina
- State Key Laboratory of BiotherapySichuan UniversityChengduSichuanChina
| |
Collapse
|
5
|
Hayashi Y. Signaling pathways regulating the immune function of cochlear supporting cells and their involvement in cochlear pathophysiology. Glia 2024; 72:665-676. [PMID: 37933494 DOI: 10.1002/glia.24476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 09/14/2023] [Accepted: 09/20/2023] [Indexed: 11/08/2023]
Abstract
The inner ear, including the cochlea, used to be regarded as an immune-privileged site because of its immunologically isolated environment caused by the blood-labyrinthine barrier. Cochlear resident macrophages, which originate from the yolk sac or fetal liver during the embryonic stage and are maintained after birth, are distributed throughout various regions of the cochlear duct. Intriguingly, these cells are absent in the organ of Corti, where hair cells (HCs) and supporting cells (SCs) are located, except for a limited number of ionized calcium-binding adapter molecule 1 (Iba1)-positive cells. Instead, SCs exert glial functions varying from a quiescent to an emergency state. Notably, SCs acquire the nature of macrophages and begin to secrete inflammatory cytokines during viral infection in the organ of Corti, which is ostensibly unprotected owing to the lack of general resident macrophages. This review provides an overview of both positive and negative functions of SCs enabled to acquire macrophage phenotypes upon viral infection focusing on the signaling pathways that regulate these functions. The former function protects HCs from viral infection by inducting type I interferons, and the latter function induces HC death by necroptosis, leading to sensorineural hearing loss. Thus, SCs play contradictory roles as immune cells with acquired macrophage phenotypes; thereby, they are favorable and unfavorable to HCs, which play a pivotal role in hearing function.
Collapse
Affiliation(s)
- Yushi Hayashi
- Department of Molecular and Medical Genetics, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
6
|
Lammers MJW, Young E, Yanai A, Viringipurampeer IA, Le TN, Straatman LV, Westerberg BD, Gregory-Evans K. IGF-1 Mediated Neuroprotective Effects of Olfactory-Derived Mesenchymal Stem Cells on Auditory Hair Cells. J Otolaryngol Head Neck Surg 2024; 53:19160216241258431. [PMID: 38888945 PMCID: PMC11177734 DOI: 10.1177/19160216241258431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 04/11/2024] [Indexed: 06/20/2024] Open
Abstract
IMPORTANCE Mesenchymal stem cells (MSCs) have the capability of providing ongoing paracrine support to degenerating tissues. Since MSCs can be extracted from a broad range of tissues, their specific surface marker profiles and growth factor secretions can be different. We hypothesized that MSCs derived from different sources might also have different neuroprotective potential. OBJECTIVE In this study, we extracted MSCs from rodent olfactory mucosa and compared their neuroprotective effects on auditory hair cell survival with MSCs extracted from rodent adipose tissue. METHODS Organ of Corti explants were dissected from 41 cochlea and incubated with olfactory mesenchymal stem cells (OMSCs) and adipose mesenchymal stem cells (AMSCs). After 72 hours, Corti explants were fixed, stained, and hair cells counted. Growth factor concentrations were determined in the supernatant and cell lysate using Enzyme-Linked Immunosorbent Assay (ELISA). RESULTS Co-culturing of organ of Corti explants with OMSCs resulted in a significant increase in inner and outer hair cell stereocilia survival, compared to control. Comparisons between both stem cell lines, showed that co-culturing with OMSCs resulted in superior inner and outer hair cell stereocilia survival rates over co-culturing with AMSCs. Assessment of growth factor secretions revealed that the OMSCs secrete significant amounts of insulin-like growth factor 1 (IGF-1). Co-culturing OMSCs with organ of Corti explants resulted in a 10-fold increase in IGF-1 level compared to control, and their secretion was 2 to 3 times higher compared to the AMSCs. CONCLUSIONS This study has shown that OMSCs may mitigate auditory hair cell stereocilia degeneration. Their neuroprotective effects may, at least partially, be ascribed to their enhanced IGF-1 secretory abilities compared to AMSCs.
Collapse
Affiliation(s)
- Marc J. W. Lammers
- BC Rotary Hearing and Balance Centre at St. Paul’s Hospital, University of British Columbia, Vancouver, BC, Canada
- Division of Otolaryngology—Head and Neck Surgery, Department of Surgery, University of British Columbia, Vancouver, BC, Canada
- Department of Otorhinolaryngology and Head and Neck Surgery, Antwerp University Hospital, Antwerp, Belgium
- Department of Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Emily Young
- BC Rotary Hearing and Balance Centre at St. Paul’s Hospital, University of British Columbia, Vancouver, BC, Canada
- Division of Otolaryngology—Head and Neck Surgery, Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Anat Yanai
- Department of Ophthalmology and Visual Science, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Ishaq A. Viringipurampeer
- Department of Ophthalmology and Visual Science, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Trung N. Le
- Sunnybrook Research Institute, Department of Otolaryngology—Head and Neck Surgery, University of Toronto, Toronto, ON, Canada
| | - Louise V. Straatman
- Department of Otolaryngology—Head and Neck Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Brian D. Westerberg
- BC Rotary Hearing and Balance Centre at St. Paul’s Hospital, University of British Columbia, Vancouver, BC, Canada
- Division of Otolaryngology—Head and Neck Surgery, Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Kevin Gregory-Evans
- Department of Ophthalmology and Visual Science, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
7
|
IGF-1 Controls Metabolic Homeostasis and Survival in HEI-OC1 Auditory Cells through AKT and mTOR Signaling. Antioxidants (Basel) 2023; 12:antiox12020233. [PMID: 36829792 PMCID: PMC9952701 DOI: 10.3390/antiox12020233] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Insulin-like growth factor 1 (IGF-1) is a trophic factor for the nervous system where it exerts pleiotropic effects, including the regulation of metabolic homeostasis. IGF-1 deficiency induces morphological alterations in the cochlea, apoptosis and hearing loss. While multiple studies have addressed the role of IGF-1 in hearing protection, its potential function in the modulation of otic metabolism remains unclear. Here, we report that "House Ear Institute-organ of Corti 1" (HEI-OC1) auditory cells express IGF-system genes that are regulated during their differentiation. Upon binding to its high-affinity receptor IGF1R, IGF-1 activates AKT and mTOR signaling to stimulate anabolism and, concomitantly, to reduce autophagic catabolism in HEI-OC1 progenitor cells. Notably, IGF-1 stimulation during HEI-OC1 differentiation to mature otic cells sustained both constructive metabolism and autophagic flux, possibly to favor cell remodeling. IGF1R engagement and downstream AKT signaling promoted HEI-OC1 cell survival by maintaining redox balance, even when cells were challenged with the ototoxic agent cisplatin. Our findings establish that IGF-1 not only serves an important function in otic metabolic homeostasis but also activates antioxidant defense mechanisms to promote hair cell survival during the stress response to insults.
Collapse
|
8
|
Malfeld K, Armbrecht N, Pich A, Volk HA, Lenarz T, Scheper V. Prevention of Noise-Induced Hearing Loss In Vivo: Continuous Application of Insulin-like Growth Factor 1 and Its Effect on Inner Ear Synapses, Auditory Function and Perilymph Proteins. Int J Mol Sci 2022; 24:ijms24010291. [PMID: 36613734 PMCID: PMC9820558 DOI: 10.3390/ijms24010291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/19/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
As noise-induced hearing loss (NIHL) is a leading cause of occupational diseases, there is an urgent need for the development of preventive and therapeutic interventions. To avoid user-compliance-based problems occurring with conventional protection devices, the pharmacological prevention is currently in the focus of hearing research. Noise exposure leads to an increase in reactive oxygen species (ROS) in the cochlea. This way antioxidant agents are a promising option for pharmacological interventions. Previous animal studies reported preventive as well as therapeutic effects of Insulin-like growth factor 1 (IGF-1) in the context of NIHL. Unfortunately, in patients the time point of the noise trauma cannot always be predicted, and additive effects may occur. Therefore, continuous prevention seems to be beneficial. The present study aimed to investigate the preventive potential of continuous administration of low concentrations of IGF-1 to the inner ear in an animal model of NIHL. Guinea pigs were unilaterally implanted with an osmotic minipump. One week after surgery they received noise trauma, inducing a temporary threshold shift. Continuous IGF-1 delivery lasted for seven more days. It did not lead to significantly improved hearing thresholds compared to control animals. Quite the contrary, there is a hint for a higher noise susceptibility. Nevertheless, changes in the perilymph proteome indicate a reduced damage and better repair mechanisms through the IGF-1 treatment. Thus, future studies should investigate delivery methods enabling continuous prevention but reducing the risk of an overdosage.
Collapse
Affiliation(s)
- Kathrin Malfeld
- Department of Otolaryngology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
- Department of Small Animal Medicine and Surgery, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany
| | - Nina Armbrecht
- Department of Otolaryngology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Andreas Pich
- Core Facility Proteomics, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Holger A. Volk
- Department of Small Animal Medicine and Surgery, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany
| | - Thomas Lenarz
- Department of Otolaryngology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
- Cluster of Excellence “Hearing4all”, German Research Foundation (DFG; “Deutsche Forschungsgemeinschaft”), Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Verena Scheper
- Department of Otolaryngology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
- Cluster of Excellence “Hearing4all”, German Research Foundation (DFG; “Deutsche Forschungsgemeinschaft”), Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
- Correspondence:
| |
Collapse
|
9
|
Peter MS, Warnecke A, Staecker H. A Window of Opportunity: Perilymph Sampling from the Round Window Membrane Can Advance Inner Ear Diagnostics and Therapeutics. J Clin Med 2022; 11:jcm11020316. [PMID: 35054010 PMCID: PMC8781055 DOI: 10.3390/jcm11020316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/29/2021] [Accepted: 01/06/2022] [Indexed: 12/12/2022] Open
Abstract
In the clinical setting, the pathophysiology of sensorineural hearing loss is poorly defined and there are currently no diagnostic tests available to differentiate between subtypes. This often leaves patients with generalized treatment options such as steroids, hearing aids, or cochlear implantation. The gold standard for localizing disease is direct biopsy or imaging of the affected tissue; however, the inaccessibility and fragility of the cochlea make these techniques difficult. Thus, the establishment of an indirect biopsy, a sampling of inner fluids, is needed to advance inner ear diagnostics and allow for the development of novel therapeutics for inner ear disease. A promising source is perilymph, an inner ear liquid that bathes multiple structures critical to sound transduction. Intraoperative perilymph sampling via the round window membrane of the cochlea has been successfully used to profile the proteome, metabolome, and transcriptome of the inner ear and is a potential source of biomarker discovery. Despite its potential to provide insight into inner ear pathologies, human perilymph sampling continues to be controversial and is currently performed only in conjunction with a planned procedure where the inner ear is opened. Here, we review the safety of procedures in which the inner ear is opened, highlight studies where perilymph analysis has advanced our knowledge of inner ear diseases, and finally propose that perilymph sampling could be done as a stand-alone procedure, thereby advancing our ability to accurately classify sensorineural hearing loss.
Collapse
Affiliation(s)
- Madeleine St. Peter
- Department of Otolaryngology-Head & Neck Surgery, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - Athanasia Warnecke
- Department of Otolaryngology Head and Neck Surgery, Hannover Medical School, D-30625 Hanover, Germany;
| | - Hinrich Staecker
- Department of Otolaryngology-Head & Neck Surgery, University of Kansas Medical Center, Kansas City, KS 66160, USA;
- Correspondence:
| |
Collapse
|
10
|
Celaya AM, Rodríguez-de la Rosa L, Bermúdez-Muñoz JM, Zubeldia JM, Romá-Mateo C, Avendaño C, Pallardó FV, Varela-Nieto I. IGF-1 Haploinsufficiency Causes Age-Related Chronic Cochlear Inflammation and Increases Noise-Induced Hearing Loss. Cells 2021; 10:cells10071686. [PMID: 34359856 PMCID: PMC8304185 DOI: 10.3390/cells10071686] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 06/28/2021] [Accepted: 06/28/2021] [Indexed: 12/18/2022] Open
Abstract
Insulin-like growth factor 1 (IGF-1) deficiency is an ultrarare syndromic human sensorineural deafness. Accordingly, IGF-1 is essential for the postnatal maturation of the cochlea and the correct wiring of hearing in mice. Less severe decreases in human IGF-1 levels have been associated with other hearing loss rare genetic syndromes, as well as with age-related hearing loss (ARHL). However, the underlying mechanisms linking IGF-1 haploinsufficiency with auditory pathology and ARHL have not been studied. Igf1-heterozygous mice express less Igf1 transcription and have 40% lower IGF-1 serum levels than wild-type mice. Along with ageing, IGF-1 levels decreased concomitantly with the increased expression of inflammatory cytokines, Tgfb1 and Il1b, but there was no associated hearing loss. However, noise exposure of these mice caused increased injury to sensory hair cells and irreversible hearing loss. Concomitantly, there was a significant alteration in the expression ratio of pro- and anti-inflammatory cytokines in Igf1+/- mice. Unbalanced inflammation led to the activation of the stress kinase JNK and the failure to activate AKT. Our data show that IGF-1 haploinsufficiency causes a chronic subclinical proinflammatory age-associated state and, consequently, greater susceptibility to stressors. This work provides the molecular bases to further understand hearing disorders linked to IGF-1 deficiency.
Collapse
Affiliation(s)
- Adelaida M. Celaya
- Institute for Biomedical Research “Alberto Sols” (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; (A.M.C.); (J.M.B.-M.); (J.M.Z.)
- Rare Diseases Biomedical Research Networking Centre (CIBERER), The Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain; (C.R.-M.); (F.V.P.)
| | - Lourdes Rodríguez-de la Rosa
- Institute for Biomedical Research “Alberto Sols” (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; (A.M.C.); (J.M.B.-M.); (J.M.Z.)
- Rare Diseases Biomedical Research Networking Centre (CIBERER), The Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain; (C.R.-M.); (F.V.P.)
- Hospital La Paz Institute for Health Research (IdiPAZ), 28029 Madrid, Spain;
- Correspondence: (L.R.-d.l.R.); (I.V.-N.)
| | - Jose M. Bermúdez-Muñoz
- Institute for Biomedical Research “Alberto Sols” (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; (A.M.C.); (J.M.B.-M.); (J.M.Z.)
- Rare Diseases Biomedical Research Networking Centre (CIBERER), The Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain; (C.R.-M.); (F.V.P.)
| | - José M. Zubeldia
- Institute for Biomedical Research “Alberto Sols” (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; (A.M.C.); (J.M.B.-M.); (J.M.Z.)
- Rare Diseases Biomedical Research Networking Centre (CIBERER), The Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain; (C.R.-M.); (F.V.P.)
- Allergy Service, Gregorio Marañon General University Hospital, 28009 Madrid, Spain
- Gregorio Marañon Health Research Institute (IiSGM), 28009 Madrid, Spain
| | - Carlos Romá-Mateo
- Rare Diseases Biomedical Research Networking Centre (CIBERER), The Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain; (C.R.-M.); (F.V.P.)
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Spain and FIHCUV-INCLIVA, 46010 Valencia, Spain
| | - Carlos Avendaño
- Hospital La Paz Institute for Health Research (IdiPAZ), 28029 Madrid, Spain;
- Department of Anatomy, Histology & Neuroscience, Medical School, Autonomous University of Madrid, 28029 Madrid, Spain
| | - Federico V. Pallardó
- Rare Diseases Biomedical Research Networking Centre (CIBERER), The Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain; (C.R.-M.); (F.V.P.)
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Spain and FIHCUV-INCLIVA, 46010 Valencia, Spain
| | - Isabel Varela-Nieto
- Institute for Biomedical Research “Alberto Sols” (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; (A.M.C.); (J.M.B.-M.); (J.M.Z.)
- Rare Diseases Biomedical Research Networking Centre (CIBERER), The Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain; (C.R.-M.); (F.V.P.)
- Hospital La Paz Institute for Health Research (IdiPAZ), 28029 Madrid, Spain;
- Correspondence: (L.R.-d.l.R.); (I.V.-N.)
| |
Collapse
|
11
|
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: 0.5] [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
|
12
|
Eshraghi AA, Wolfovitz A, Yilmazer R, Garnham C, Yilmazer AB, Bas E, Ashman P, Roell J, Bohorquez J, Mittal R, Hessler R, Sieber D, Mittal J. Otoprotection to Implanted Cochlea Exposed to Noise Trauma With Dexamethasone Eluting Electrode. Front Cell Neurosci 2019; 13:492. [PMID: 31824265 PMCID: PMC6882736 DOI: 10.3389/fncel.2019.00492] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 10/21/2019] [Indexed: 12/23/2022] Open
Abstract
Cochlear implantation (CI) is now widely used to provide auditory rehabilitation to individuals having severe to profound sensorineural hearing loss (SNHL). However, CI can lead to electrode insertion trauma (EIT) that can cause damage to sensory cells in the inner ear resulting in loss of residual hearing. Even with soft surgical techniques where there is minimal macroscopic damage, we can still observe the generation of molecular events that may initiate programmed cell death via various mechanisms such as oxidative stress, the release of pro-inflammatory cytokines, and activation of the caspase pathway. In addition, individuals with CI may be exposed to noise trauma (NT) due to occupation and leisure activities that may affect their hearing ability. Recently, there has been an increased interest in the auditory community to determine the efficacy of drug-eluting electrodes for the protection of residual hearing. The objective of this study is to determine the effect of NT on implanted cochlea as well as the otoprotective efficacy of dexamethasone eluting electrode to implanted cochlea exposed to NT in a guinea pig model of CI. Animals were divided into five groups: EIT with dexamethasone eluting electrode exposed to NT; EIT exposed to NT; NT only; EIT only and naïve animals (control group). The hearing thresholds were determined by auditory brainstem recordings (ABRs). The cochlea was harvested and analyzed for transcript levels of inflammation, apoptosis and fibrosis genes. We observed that threshold shifts were significantly higher in EIT, NT or EIT + NT groups compared to naive animals at all the tested frequencies. The dexamethasone eluting electrode led to a significant decrease in hearing threshold shifts in implanted animals exposed to NT. Proapoptotic tumor necrosis factor-α [TNF-α, TNF-α receptor 1a (TNFαR1a)] and pro-fibrotic transforming growth factor β1 (TGFβ) genes were more than two-fold up-regulated following EIT and EIT + NT compared to the control group. The use of dexamethasone releasing electrode significantly decreased the transcript levels of pro-apoptotic and pro-fibrotic genes. The dexamethasone releasing electrode has shown promising results for hearing protection in implanted animals exposed to NT. The results of this study suggest that dexamethasone releasing electrode holds great potential in developing effective treatment modalities for NT in the implanted cochlea.
Collapse
Affiliation(s)
- Adrien A Eshraghi
- Department of Otolaryngology, University of Miami Hearing Research Laboratory, Miller School of Medicine, Miami, FL, United States.,Department of Neurological Surgery, Miller School of Medicine, Miami, FL, United States.,Department of Biomedical Engineering, University of Miami, Miami, Coral Gables, FL, United States
| | - Amit Wolfovitz
- Department of Otolaryngology, University of Miami Hearing Research Laboratory, Miller School of Medicine, Miami, FL, United States
| | - Rasim Yilmazer
- Department of Otolaryngology, University of Miami Hearing Research Laboratory, Miller School of Medicine, Miami, FL, United States
| | | | - Ayca Baskadem Yilmazer
- Department of Otolaryngology, University of Miami Hearing Research Laboratory, Miller School of Medicine, Miami, FL, United States
| | - Esperanza Bas
- Department of Otolaryngology, University of Miami Hearing Research Laboratory, Miller School of Medicine, Miami, FL, United States
| | - Peter Ashman
- Department of Otolaryngology, University of Miami Hearing Research Laboratory, Miller School of Medicine, Miami, FL, United States
| | - Jonathan Roell
- Department of Otolaryngology, University of Miami Hearing Research Laboratory, Miller School of Medicine, Miami, FL, United States
| | - Jorge Bohorquez
- Department of Biomedical Engineering, University of Miami, Miami, Coral Gables, FL, United States
| | - Rahul Mittal
- Department of Otolaryngology, University of Miami Hearing Research Laboratory, Miller School of Medicine, Miami, FL, United States
| | | | | | - Jeenu Mittal
- Department of Otolaryngology, University of Miami Hearing Research Laboratory, Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
13
|
Warnecke A, Prenzler NK, Schmitt H, Daemen K, Keil J, Dursin M, Lenarz T, Falk CS. Defining the Inflammatory Microenvironment in the Human Cochlea by Perilymph Analysis: Toward Liquid Biopsy of the Cochlea. Front Neurol 2019; 10:665. [PMID: 31293504 PMCID: PMC6603180 DOI: 10.3389/fneur.2019.00665] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 06/06/2019] [Indexed: 12/20/2022] Open
Abstract
The molecular pathomechanisms in the majority of patients suffering from acute or progressive sensorineural hearing loss cannot be determined yet. The size and the complex architecture of the cochlea make biopsy and in-depth histological analyses impossible without severe damage of the organ. Thus, histopathology correlated to inner disease is only possible after death. The establishment of a technique for perilymph sampling during cochlear implantation may enable a liquid biopsy and characterization of the cochlear microenvironment. Inflammatory processes may not only participate in disease onset and progression in the inner ear, but may also control performance of the implant. However, little is known about cytokines and chemokines in the human inner ear as predictive markers for cochlear implant performance. First attempts to use multiplex protein arrays for inflammatory markers were successful for the identification of cytokines, chemokines, and endothelial markers present in the human perilymph. Moreover, unsupervised cluster and principal component analyses were used to group patients by lead cytokines and to correlate certain proteins to clinical data. Endothelial and epithelial factors were detected at higher concentrations than typical pro-inflammatory cytokines such as TNF-a or IL-6. Significant differences in VEGF family members have been observed comparing patients with deafness to patients with residual hearing with significantly reduced VEGF-D levels in patients with deafness. In addition, there is a trend toward higher IGFBP-1 levels in these patients. Hence, endothelial and epithelial factors in combination with cytokines may present robust biomarker candidates and will be investigated in future studies in more detail. Thus, multiplex protein arrays are feasible in very small perilymph samples allowing a qualitative and quantitative analysis of inflammatory markers. More results are required to advance this method for elucidating the development and course of specific inner ear diseases or for perioperative characterization of cochlear implant patients.
Collapse
Affiliation(s)
- Athanasia Warnecke
- Department of Otolaryngology, Hannover Medical School, Hanover, Germany.,Cluster of Excellence of the German Research Foundation (DFG; "Deutsche Forschungsgemeinschaft") "Hearing4all", Oldenburg, Germany
| | - Nils K Prenzler
- Department of Otolaryngology, Hannover Medical School, Hanover, Germany
| | - Heike Schmitt
- Department of Otolaryngology, Hannover Medical School, Hanover, Germany.,Cluster of Excellence of the German Research Foundation (DFG; "Deutsche Forschungsgemeinschaft") "Hearing4all", Oldenburg, Germany
| | - Kerstin Daemen
- Hannover Medical School, Institute of Transplant Immunology, Hanover, Germany
| | - Jana Keil
- Hannover Medical School, Institute of Transplant Immunology, Hanover, Germany
| | - Martin Dursin
- Department of Otolaryngology, Hannover Medical School, Hanover, Germany
| | - Thomas Lenarz
- Department of Otolaryngology, Hannover Medical School, Hanover, Germany.,Cluster of Excellence of the German Research Foundation (DFG; "Deutsche Forschungsgemeinschaft") "Hearing4all", Oldenburg, Germany
| | - Christine S Falk
- Hannover Medical School, Institute of Transplant Immunology, Hanover, Germany
| |
Collapse
|
14
|
Pollard KJ, Daniel JM. Nuclear estrogen receptor activation by insulin-like growth factor-1 in Neuro-2A neuroblastoma cells requires endogenous estrogen synthesis and is mediated by mutually repressive MAPK and PI3K cascades. Mol Cell Endocrinol 2019; 490:68-79. [PMID: 30986444 PMCID: PMC6520186 DOI: 10.1016/j.mce.2019.04.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 04/10/2019] [Accepted: 04/10/2019] [Indexed: 02/01/2023]
Abstract
Non-canonical mechanisms of estrogen receptor activation may continue to support women's cognitive health long after cessation of ovarian function. These mechanisms of estrogen receptor activation may include ligand-dependent actions via locally synthesized neuroestrogens and ligand-independent actions via growth factor-dependent activation of intracellular kinase cascades. We tested the hypothesis that ligand-dependent and ligand-independent mechanisms interact to activate nuclear estrogen receptors in the Neuro-2A neuroblastoma cell line in the absence of exogenous estrogens. Transcriptional output of estrogen receptors was measured following treatment with insulin-like growth factor-1 (IGF-1) in the presence of specific inhibitors for mitogen-activated protein kinase (MAPK), phosphoinositde-3 kinase (PI3K), and neuroestrogen synthesis. Results indicate that IGF-1-dependent activation of nuclear estrogen receptors is mediated by MAPK, is opposed PI3K, and requires concomitant endogenous neuroestrogen synthesis. We conclude that both cellular signaling context and endogenous ligand availability are important modulators of ligand-independent nuclear estrogen receptor activation.
Collapse
Affiliation(s)
- Kevin J Pollard
- Tulane Brain Institute, Tulane University, 200 Flower Hall, New Orleans, LA, 70118, USA; Neuroscience Program, Tulane University, 200 Flower Hall, New Orleans, LA, 70118, USA.
| | - Jill M Daniel
- Tulane Brain Institute, Tulane University, 200 Flower Hall, New Orleans, LA, 70118, USA; Neuroscience Program, Tulane University, 200 Flower Hall, New Orleans, LA, 70118, USA; Department of Psychology, Tulane University, 2007 Percival Stern Hall, New Orleans, LA, 70118, USA
| |
Collapse
|
15
|
Activation of IGF1 Signaling in the Cochlea Induces the Transcription of Its Mediators During the Protection of Cochlear Hair Cells Against Aminoglycoside. Otol Neurotol 2017; 38:278-282. [PMID: 27846039 DOI: 10.1097/mao.0000000000001276] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
HYPOTHESIS Transcription of the Erk and Akt genes and phosphorylation of their products are promoted by insulin-like growth factor 1 (IGF1) during hair cell protection. BACKGROUND IGF1 protects mammalian hair cells in animal models from various types of damage, including aminoglycoside. Moreover, clinical trials have revealed that IGF1 was effective for idiopathic sudden sensorineural hearing loss. In this process, activation of the downstream of IGF1 signaling, including the phosphorylation of extracellular signal-regulated kinase (ERK) and AKT proteins, is involved. However, the regulation of IGF1 signaling mediators at the transcriptional level has not been studied. METHODS We used a neomycin damage model on neonatal mouse cochlear explant culture. Explants established from neonatal mice were treated with either neomycin alone or neomycin and IGF1. The expression levels of IGF1 signaling mediator genes, Akt1, Mapk3, and Mapk1, in the explants were compared using quantitative reverse transcriptase-polymerase chain reaction at several time points. Inhibitors of IGF1 signaling were added to confirm that this observation was dependent on IGF1 signaling. RESULTS The expression levels of all genes tested were significantly upregulated in neomycin+IGF1 treatment samples (p < 0.0001, analysis of variance [ANOVA]). Addition of inhibitors of IGF1 signaling significantly attenuated the upregulation of expression (p < 0.0001, ANOVA). CONCLUSIONS IGF1 treatment upregulates the expression of its mediator genes during the protection of hair cells against aminoglycoside. The regulation of mediator gene expression may serve as a novel treatment for sensorineural hearing loss.
Collapse
|
16
|
Bodmer D. An update on drug design strategies to prevent acquired sensorineural hearing loss. Expert Opin Drug Discov 2017; 12:1161-1167. [PMID: 28838250 DOI: 10.1080/17460441.2017.1372744] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Acute sensorineural hearing loss is a dramatic event for the patient. Different pathologies might result in acute sensorineural hearing loss, such as sudden hearing loss, exposure to medications/drugs or loud sound. Current therapeutic approaches include steroids and hyperbaric oxygen in addition to other methods. Research activities of the past have shed light on the molecular mechanisms involved in damage to hair cells, the synapses at the hair cell spiral ganglion junction and the stria vascularis. Molecular events and signaling pathways which underlie damage to these structures have been discovered. Areas covered: This paper summarizes current research efforts involved in investigating the molecular mechanisms involved in acute sensorineural hearing loss. Expert opinion: While progress has been made in unraveling basic mechanisms involved in acute sensorineural hearing loss, it is difficult to translate basic concepts to the clinic. There are often conflicting data in animal and human studies on the effect of a given intervention. There is also a lack of high quality clinical trials (double blind, placebo controlled and high powered). However, this author is confident that research efforts will pay out and that some of these efforts will translate into new therapeutic options for patients with acute hearing loss.
Collapse
Affiliation(s)
- Daniel Bodmer
- a Department of Biomedicine, Head and Neck Surgery , University of Basel Hospital , Basel , Switzerland.,b Department of Otolaryngology, Head and Neck Surgery , University of Basel Hospital , Basel , Switzerland
| |
Collapse
|
17
|
Stolle M, Schulze J, Roemer A, Lenarz T, Durisin M, Warnecke A. Human Plasma Rich in Growth Factors Improves Survival and Neurite Outgrowth of Spiral Ganglion Neurons In Vitro. Tissue Eng Part A 2017; 24:493-501. [PMID: 28610547 DOI: 10.1089/ten.tea.2017.0120] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Platelet-rich and platelet-poor plasma (PRP and PPP) are autologous preparations from peripheral blood and contain several growth factors and cytokines involved in tissue repair. Although their neuroprotective and neuroregenerative properties have been already described, little is known about their effects in the inner ear. We, therefore, examined the effects of PRP and PPP on spiral ganglion neurons (SGN) in vitro. RESULTS For all experiments, spiral ganglia were isolated from neonatal rats and were cultured in serum-free medium. PRP from human venous blood was added to dissociated SGN. Treatment with PRP (1:10, 1:50) significantly increased the neuronal survival and the neuronal outgrowth of SGN. This effect was completely reversed by the addition of Bay 11 (nuclear factor kappa B-inhibitor) and SB203580 (p38 mitogen-activated protein kinase [p38MAPK]-inhibitor). Furthermore, PPP was used as a cell-free matrix for the attachment of spiral ganglion explants. Coating with activated PPP improved the adhesion and neurite outgrowth of spiral ganglia explants. Therefore, activated PPP is a promising alternative for poly d/l-ornithine and laminin coating due to the gelatinous composition through the activation of PPP with calcium gluconate. PRP promotes neuroprotective and neuroregenerative effects on SGN when administered in adequate concentrations. These beneficial effects seem to be depending on NF-κB and the p38MAPK pathways. CONCLUSION Preparations from autologous whole blood (PRP and PPP, respectively) present an interesting alternative for pharmacological intervention to the inner ear since they contain a balanced and natural composition of trophic factors.
Collapse
Affiliation(s)
- Michael Stolle
- 1 Department of Otolaryngology, Hannover Medical School , Hannover, Germany
| | - Jennifer Schulze
- 1 Department of Otolaryngology, Hannover Medical School , Hannover, Germany .,2 Cluster of Excellence of the German Research Foundation (DFG; "Deutsche Forschungsgemeinschaft") "Hearing4all", Hannover, Germany
| | - Ariane Roemer
- 1 Department of Otolaryngology, Hannover Medical School , Hannover, Germany
| | - Thomas Lenarz
- 1 Department of Otolaryngology, Hannover Medical School , Hannover, Germany .,2 Cluster of Excellence of the German Research Foundation (DFG; "Deutsche Forschungsgemeinschaft") "Hearing4all", Hannover, Germany
| | - Martin Durisin
- 1 Department of Otolaryngology, Hannover Medical School , Hannover, Germany
| | - Athanasia Warnecke
- 1 Department of Otolaryngology, Hannover Medical School , Hannover, Germany .,2 Cluster of Excellence of the German Research Foundation (DFG; "Deutsche Forschungsgemeinschaft") "Hearing4all", Hannover, Germany
| |
Collapse
|
18
|
Kalinec GM, Lomberk G, Urrutia RA, Kalinec F. Resolution of Cochlear Inflammation: Novel Target for Preventing or Ameliorating Drug-, Noise- and Age-related Hearing Loss. Front Cell Neurosci 2017; 11:192. [PMID: 28736517 PMCID: PMC5500902 DOI: 10.3389/fncel.2017.00192] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Accepted: 06/20/2017] [Indexed: 12/11/2022] Open
Abstract
A significant number of studies support the idea that inflammatory responses are intimately associated with drug-, noise- and age-related hearing loss (DRHL, NRHL and ARHL). Consequently, several clinical strategies aimed at reducing auditory dysfunction by preventing inflammation are currently under intense scrutiny. Inflammation, however, is a normal adaptive response aimed at restoring tissue functionality and homeostasis after infection, tissue injury and even stress under sterile conditions, and suppressing it could have unintended negative consequences. Therefore, an appropriate approach to prevent or ameliorate DRHL, NRHL and ARHL should involve improving the resolution of the inflammatory process in the cochlea rather than inhibiting this phenomenon. The resolution of inflammation is not a passive response but rather an active, highly controlled and coordinated process. Inflammation by itself produces specialized pro-resolving mediators with critical functions, including essential fatty acid derivatives (lipoxins, resolvins, protectins and maresins), proteins and peptides such as annexin A1 and galectins, purines (adenosine), gaseous mediators (NO, H2S and CO), as well as neuromodulators like acetylcholine and netrin-1. In this review article, we describe recent advances in the understanding of the resolution phase of inflammation and highlight therapeutic strategies that might be useful in preventing inflammation-induced cochlear damage. In particular, we emphasize beneficial approaches that have been tested in pre-clinical models of inflammatory responses induced by recognized ototoxic drugs such as cisplatin and aminoglycoside antibiotics. Since these studies suggest that improving the resolution process could be useful for the prevention of inflammation-associated diseases in humans, we discuss the potential application of similar strategies to prevent or mitigate DRHL, NRHL and ARHL.
Collapse
Affiliation(s)
- Gilda M Kalinec
- Laboratory of Auditory Cell Biology, Department of Head and Neck Surgery, David Geffen School of Medicine, University of CaliforniaLos Angeles, Los Angeles, CA, United States
| | - Gwen Lomberk
- Epigenetics and Chromatin Dynamics Laboratory, Translational Epigenomic Program, Center for Individualized Medicine (CIM) Mayo ClinicRochester, MN, United States
| | - Raul A Urrutia
- Epigenetics and Chromatin Dynamics Laboratory, Translational Epigenomic Program, Center for Individualized Medicine (CIM) Mayo ClinicRochester, MN, United States
| | - Federico Kalinec
- Laboratory of Auditory Cell Biology, Department of Head and Neck Surgery, David Geffen School of Medicine, University of CaliforniaLos Angeles, Los Angeles, CA, United States
| |
Collapse
|
19
|
Yamahara K, Nakagawa T, Ito J, Kinoshita K, Omori K, Yamamoto N. Netrin 1 mediates protective effects exerted by insulin-like growth factor 1 on cochlear hair cells. Neuropharmacology 2017; 119:26-39. [PMID: 28373074 DOI: 10.1016/j.neuropharm.2017.03.032] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Revised: 03/15/2017] [Accepted: 03/29/2017] [Indexed: 01/17/2023]
Abstract
Sensorineural hearing loss (SNHL) is mainly caused by the damage of cochlear hair cells (HCs). As HCs and supporting cells (SCs) do not proliferate in postnatal mammals, the loss of HCs and SCs is irreversible, emphasizing the importance of preserving their numbers to prevent SNHL. It is known that insulin-like growth factor 1 (IGF1) is instrumental in the treatment of SNHL. Our previous study indicates that IGF1 protects HCs against aminoglycoside by activating IGF1 receptor and its two major downstream pathways, PI3K/AKT and MEK/ERK, in SCs, which results in the upregulation of the expression of the Netrin1-encoding gene (Ntn1). However, the mechanisms underlying IGF1-induced protection of HCs via SC activation as well as the role of NTN1 in this process have not been elucidated. Here, we demonstrated that NTN1, similar to IGF1, promoted HC survival. NTN1 blocking antibody attenuated IGF1-induced HC protection from aminoglycoside, indicating that NTN1 is the effector molecule of IGF1 signaling during HC protection. In situ hybridization demonstrated that IGF1 potently induced Ntn1 expression in SCs. NTN1 receptors were abundantly expressed in the cochlea; among them, UNC5B mediated IGF1 protective effects on HCs, as NTN1 binding to UNC5B inhibited HC apoptosis. These results provide new insights into the mechanisms underlying IGF1 protection of cochlear HCs, suggesting a possibility of using NTN1 as a new treatment for SNHL.
Collapse
Affiliation(s)
- Kohei Yamahara
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto 606-8507, Japan
| | - Takayuki Nakagawa
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto 606-8507, Japan
| | - Juichi Ito
- Shiga Medical Center Research Institute, Moriyama, Shiga 524-8523, Japan
| | - Kazuo Kinoshita
- Shiga Medical Center Research Institute, Moriyama, Shiga 524-8523, Japan
| | - Koichi Omori
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto 606-8507, Japan
| | - Norio Yamamoto
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto 606-8507, Japan.
| |
Collapse
|
20
|
Liu L, Chen Y, Qi J, Zhang Y, He Y, Ni W, Li W, Zhang S, Sun S, Taketo MM, Wang L, Chai R, Li H. Wnt activation protects against neomycin-induced hair cell damage in the mouse cochlea. Cell Death Dis 2016; 7:e2136. [PMID: 26962686 PMCID: PMC4823936 DOI: 10.1038/cddis.2016.35] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 01/11/2016] [Accepted: 01/25/2016] [Indexed: 12/17/2022]
Abstract
Recent studies have reported the role of Wnt/β-catenin signaling in hair cell (HC) development, regeneration, and differentiation in the mouse cochlea; however, the role of Wnt/β-catenin signaling in HC protection remains unknown. In this study, we took advantage of transgenic mice to specifically knockout or overactivate the canonical Wnt signaling mediator β-catenin in HCs, which allowed us to investigate the role of Wnt/β-catenin signaling in protecting HCs against neomycin-induced damage. We first showed that loss of β-catenin in HCs made them more vulnerable to neomycin-induced injury, while constitutive activation of β-catenin in HCs reduced HC loss both in vivo and in vitro. We then showed that loss of β-catenin in HCs increased caspase-mediated apoptosis induced by neomycin injury, while β-catenin overexpression inhibited caspase-mediated apoptosis. Finally, we demonstrated that loss of β-catenin in HCs led to increased expression of forkhead box O3 transcription factor (Foxo3) and Bim along with decreased expression of antioxidant enzymes; thus, there were increased levels of reactive oxygen species (ROS) after neomycin treatment that might be responsible for the increased aminoglycoside sensitivity of HCs. In contrast, β-catenin overexpression reduced Foxo3 and Bim expression and ROS levels, suggesting that β-catenin is protective against neomycin-induced HC loss. Our findings demonstrate that Wnt/β-catenin signaling has an important role in protecting HCs against neomycin-induced HC loss and thus might be a new therapeutic target for the prevention of HC death.
Collapse
Affiliation(s)
- L Liu
- Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, PR China.,Institutes of Biomedical Sciences, Fudan University, Shanghai, PR China
| | - Y Chen
- Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, PR China.,Laboratory Center, Affiliated Eye and ENT Hospital of Fudan University, Shanghai, PR China.,Key Laboratory of Hearing Medicine of National Health and Family Planning Commission, Shanghai, PR China
| | - J Qi
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Y Zhang
- Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, PR China.,Laboratory Center, Affiliated Eye and ENT Hospital of Fudan University, Shanghai, PR China.,Key Laboratory of Hearing Medicine of National Health and Family Planning Commission, Shanghai, PR China
| | - Y He
- Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, PR China.,Laboratory Center, Affiliated Eye and ENT Hospital of Fudan University, Shanghai, PR China.,Key Laboratory of Hearing Medicine of National Health and Family Planning Commission, Shanghai, PR China
| | - W Ni
- Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, PR China.,Key Laboratory of Hearing Medicine of National Health and Family Planning Commission, Shanghai, PR China
| | - W Li
- Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, PR China.,Laboratory Center, Affiliated Eye and ENT Hospital of Fudan University, Shanghai, PR China.,Key Laboratory of Hearing Medicine of National Health and Family Planning Commission, Shanghai, PR China
| | - S Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - S Sun
- Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, PR China.,Laboratory Center, Affiliated Eye and ENT Hospital of Fudan University, Shanghai, PR China.,Key Laboratory of Hearing Medicine of National Health and Family Planning Commission, Shanghai, PR China
| | - M M Taketo
- Department of Pharmacology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - L Wang
- Institutes of Biomedical Sciences, Fudan University, Shanghai, PR China
| | - R Chai
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - H Li
- Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, PR China.,Institutes of Biomedical Sciences, Fudan University, Shanghai, PR China.,Key Laboratory of Hearing Medicine of National Health and Family Planning Commission, Shanghai, PR China
| |
Collapse
|
21
|
Han XF, Zhang Y, Xiong LL, Xu Y, Zhang P, Xia QJ, Wang TH, Ba YC. Lentiviral-Mediated Netrin-1 Overexpression Improves Motor and Sensory Functions in SCT Rats Associated with SYP and GAP-43 Expressions. Mol Neurobiol 2016; 54:1684-1697. [PMID: 26873853 DOI: 10.1007/s12035-016-9723-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 01/13/2016] [Indexed: 02/05/2023]
Abstract
Spinal cord injury (SCI), as a major cause of disability, usually causes serious loss of motor and sensory functions. As a bifunctional axonal guidance cue, netrin-1 can attract axons via the deleted in colorectal cancer (DCC) receptors and repelling others via Unc5 receptors, but its exact role in the recovery of motor and sensory function has not well been studied, and the mechanisms remains elusive. The aim of this experiment is to determine whether lentiviral (LV)-mediated overexpression of netrin-1 or RNA interference (RNAi) can regulate the functional recovery in rats subjected to spinal cord transection (SCT). Firstly, two lentiviral vectors including Lv-exNtn-1 (netrin-1 open reading frame (ORF)) and Lv-shNtn-1 (netrin-1 sh) were constructed and injected into spinal cords rostral and caudal to the transected lesion site. Overexpressing netrin-1 enhanced significantly locomotor function, and reduced thermal and mechanical stimuli in vivo, compared with the control, while silencing netrin-1 did not significantly change the situation. Western blot and immunostaining analysis confirmed that netrin-1 ORF treatment not only effectively increased the expression level of netrin-1, also up-regulated the level of synaptophysin (SYP) in spinal cord rostral to the lesion, but also enhanced growth-associated protein-43 (GAP-43) expression in spinal cord caudal to the lesion site. Comparatively, knockdown of netrin-1 did not give rise to positive findings in our experimental condition. These findings therefore pointed that Lv-mediated netrin-1 overexpression could promote motor and sensory functional recoveries following SCT, and the underlying mechanisms were associated with SYP and GAP-43 expressions. The present study therefore provided a novel strategy for the treatment of SCI and explained the possible mechanisms for the functional improvement.
Collapse
Affiliation(s)
- Xue Fei Han
- Institute of Neuroscience and Department of Anatomy, Kunming Medical University, Kunming, 650000, China
| | - Yuan Zhang
- Institute of Neuroscience and Department of Anatomy, Kunming Medical University, Kunming, 650000, China
| | - Liu Lin Xiong
- Department of Anesthesia, Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Yang Xu
- Department of Anesthesia, Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Piao Zhang
- Institute of Neuroscience and Department of Anatomy, Kunming Medical University, Kunming, 650000, China
| | - Qing Jie Xia
- Department of Anesthesia, Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China.
| | - Ting Hua Wang
- Department of Anesthesia, Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China.
| | - Ying Chun Ba
- Institute of Neuroscience and Department of Anatomy, Kunming Medical University, Kunming, 650000, China.
| |
Collapse
|
22
|
Abstract
The identification of transcriptional differences has served as an important starting point in understanding the molecular mechanisms behind biological processes and systems. The developmental biology of the inner ear, the biology of hearing and of course the pathology of deafness are all processes that warrant a molecular description if we are to improve human health. To this end, technological innovation has meant that larger scale analysis of gene transcription has been possible for a number of years now, extending our molecular analysis of genes to beyond those that are currently in vogue for a given system. In this review, some of the contributions gene profiling has made to understanding developmental, pathological and physiological processes in the inner ear are highlighted.
Collapse
Affiliation(s)
- Thomas Schimmang
- Instituto de Biología y Genética MolecularUniversidad de Valladolid y Consejo Superior de Investigaciones CientíficasValladolidSpain
| | - Mark Maconochie
- School of Biological and Chemical SciencesQueen Mary University of LondonLondonUK
| |
Collapse
|
23
|
Yamahara K, Yamamoto N, Nakagawa T, Ito J. Insulin-like growth factor 1: A novel treatment for the protection or regeneration of cochlear hair cells. Hear Res 2015; 330:2-9. [DOI: 10.1016/j.heares.2015.04.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 04/19/2015] [Accepted: 04/24/2015] [Indexed: 11/15/2022]
|
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: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 04/21/2015] [Accepted: 04/27/2015] [Indexed: 12/20/2022]
Abstract
The family of RAF kinases transduces extracellular information to the nucleus, and their activation is crucial for cellular regulation on many levels, ranging from embryonic development to carcinogenesis. B-RAF and C-RAF modulate neurogenesis and neuritogenesis during chicken inner ear development. C-RAF deficiency in humans is associated with deafness in the rare genetic insulin-like growth factor 1 (IGF-1), Noonan and Leopard syndromes. In this study, we show that RAF kinases are expressed in the developing inner ear and in adult mouse cochlea. A homozygous C-Raf deletion in mice caused profound deafness with no evident cellular aberrations except for a remarkable reduction of the K+ channel Kir4.1 expression, a trait that suffices as a cause of deafness. To explore the role of C-Raf in cellular protection and repair, heterozygous C-Raf+/− mice were exposed to noise. A reduced C-RAF level negatively affected hearing preservation in response to noise through mechanisms involving the activation of JNK and an exacerbated apoptotic response. Taken together, these results strongly support a role for C-RAF in hearing protection.
Collapse
Affiliation(s)
- Rocío de Iriarte Rodríguez
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, Arturo Duperier 4, 28029, Madrid, Spain.,Centre for Biomedical Network Research (CIBERER), Institute of Health Carlos III (ISCIII), Madrid, Spain
| | - Marta Magariños
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, Arturo Duperier 4, 28029, Madrid, Spain. .,Centre for Biomedical Network Research (CIBERER), Institute of Health Carlos III (ISCIII), Madrid, Spain. .,Departamento de Biología, Universidad Autónoma de Madrid, Darwin 2, 28049, Madrid, Spain.
| | - Verena Pfeiffer
- Institute for Medical Radiation and Cell Research (MSZ), University of Würzburg, Versbacher Strasse 5, 97078, Würzburg, Germany.,Institute for Anatomy and Cell Biology, University of Würzburg, Koellikerstraße 6, 97070, Würzburg, Germany
| | - Ulf R Rapp
- Institute for Medical Radiation and Cell Research (MSZ), University of Würzburg, Versbacher Strasse 5, 97078, Würzburg, Germany.,Molecular Mechanisms of Lung Cancer, Max Planck Institute for Heart and Lung Research, Parkstr. 1, 61231, Bad Nauheim, Germany
| | - Isabel Varela-Nieto
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, Arturo Duperier 4, 28029, Madrid, Spain.,Centre for Biomedical Network Research (CIBERER), Institute of Health Carlos III (ISCIII), Madrid, Spain.,Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| |
Collapse
|
25
|
Wong ACY, Ryan AF. Mechanisms of sensorineural cell damage, death and survival in the cochlea. Front Aging Neurosci 2015; 7:58. [PMID: 25954196 PMCID: PMC4404918 DOI: 10.3389/fnagi.2015.00058] [Citation(s) in RCA: 190] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 04/05/2015] [Indexed: 12/20/2022] Open
Abstract
The majority of acquired hearing loss, including presbycusis, is caused by irreversible damage to the sensorineural tissues of the cochlea. This article reviews the intracellular mechanisms that contribute to sensorineural damage in the cochlea, as well as the survival signaling pathways that can provide endogenous protection and tissue rescue. These data have primarily been generated in hearing loss not directly related to age. However, there is evidence that similar mechanisms operate in presbycusis. Moreover, accumulation of damage from other causes can contribute to age-related hearing loss (ARHL). Potential therapeutic interventions to balance opposing but interconnected cell damage and survival pathways, such as antioxidants, anti-apoptotics, and pro-inflammatory cytokine inhibitors, are also discussed.
Collapse
Affiliation(s)
- Ann C Y Wong
- Department of Surgery/Division of Otolaryngology, University of California, San Diego School of Medicine La Jolla, CA, USA ; Department of Physiology and Translational Neuroscience Facility, School of Medical Sciences, University of New South Wales Sydney, NSW, Australia
| | - Allen F Ryan
- Department of Surgery/Division of Otolaryngology, University of California, San Diego School of Medicine La Jolla, CA, USA ; Veterans Administration Medical Center La Jolla, CA, USA ; Department of Neurosciences, University of California, San Diego School of Medicine La Jolla, CA, USA
| |
Collapse
|
26
|
Fuentes-Santamaría V, Alvarado JC, Rodríguez-de la Rosa L, Murillo-Cuesta S, Contreras J, Juiz JM, Varela-Nieto I. IGF-1 deficiency causes atrophic changes associated with upregulation of VGluT1 and downregulation of MEF2 transcription factors in the mouse cochlear nuclei. Brain Struct Funct 2014; 221:709-34. [PMID: 25378055 DOI: 10.1007/s00429-014-0934-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Accepted: 10/28/2014] [Indexed: 02/08/2023]
Abstract
Insulin-like growth factor 1 (IGF-1) is a neurotrophic protein that plays a crucial role in modulating neuronal function and synaptic plasticity in the adult brain. Mice lacking the Igf1 gene exhibit profound deafness and multiple anomalies in the inner ear and spiral ganglion. An issue that remains unknown is whether, in addition to these peripheral abnormalities, IGF-1 deficiency also results in structural changes along the central auditory pathway that may contribute to an imbalance between excitation and inhibition, which might be reflected in abnormal auditory brainstem responses (ABR). To assess such a possibility, we evaluated the morphological and physiological alterations in the cochlear nucleus complex of the adult mouse. The expression and distribution of the vesicular glutamate transporter 1 (VGluT1) and the vesicular inhibitory transporter (VGAT), which were used as specific markers for labeling excitatory and inhibitory terminals, and the involvement of the activity-dependent myocyte enhancer factor 2 (MEF2) transcription factors in regulating excitatory synapses were assessed in a 4-month-old mouse model of IGF-1 deficiency and neurosensorial deafness (Igf1 (-/-) homozygous null mice). The results demonstrate decreases in the cochlear nucleus area and cell size along with cell loss in the cochlear nuclei of the deficient mouse. Additionally, our results demonstrate that there is upregulation of VGluT1, but not VGAT, immunostaining and downregulation of MEF2 transcription factors together with increased wave II amplitude in the ABR recording. Our observations provide evidence of an abnormal neuronal cytoarchitecture in the cochlear nuclei of Igf1 (-/-) null mice and suggest that the increased efficacy of glutamatergic synapses might be mediated by MEF2 transcription factors.
Collapse
Affiliation(s)
- V Fuentes-Santamaría
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Albacete, Spain.
| | - J C Alvarado
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Albacete, Spain
| | - L Rodríguez-de la Rosa
- Facultad de Medicina, Universidad de Castilla-La Mancha, Campus de Albacete. C/Almansa, 14, 02006, Albacete, Spain
| | - S Murillo-Cuesta
- Grupo de Neurobiología de la Audición, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain.,Centro Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain.,IdiPAZ Instituto de Investigación en Salud, Madrid, Spain
| | - J Contreras
- Grupo de Neurobiología de la Audición, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain.,Centro Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain.,IdiPAZ Instituto de Investigación en Salud, Madrid, Spain.,Facultad de Veterinaria, Universidad Complutense, Madrid, Spain
| | - J M Juiz
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Albacete, Spain
| | - I Varela-Nieto
- Grupo de Neurobiología de la Audición, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain.,Centro Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain.,IdiPAZ Instituto de Investigación en Salud, Madrid, Spain
| |
Collapse
|
27
|
Nakagawa T. Strategies for developing novel therapeutics for sensorineural hearing loss. Front Pharmacol 2014; 5:206. [PMID: 25278894 PMCID: PMC4165348 DOI: 10.3389/fphar.2014.00206] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 08/20/2014] [Indexed: 01/01/2023] Open
Abstract
Sensorineural hearing loss (SNHL) is a common disability in the world; however, at present, options for the pharmacological treatment of SNHL are very limited. Previous studies involving human temporal bone analyses have revealed that the degeneration of the cochlea is a common mechanism of SNHL. A major problem for the development of novel pharmacotherapy for SNHL has been the limited regeneration capacity in mammalian cochlear cells. However, recent progress in basic studies has led to several effective strategies for the induction of regeneration in the mammalian cochlea, in accordance with the stage of degeneration. In addition, recent advances in the identification of human deafness genes and their characterization in mouse models have elucidated cellular and/or molecular mechanisms of SNHL, which will contribute to clarify molecular targets of pharmacotherapy for treatment of SNHL.
Collapse
Affiliation(s)
- Takayuki Nakagawa
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University Kyoto, Japan
| |
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: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 08/22/2014] [Indexed: 01/10/2023] Open
Abstract
Sensorineural hearing loss (SNHL) is considered an intractable disease, given that hair and supporting cells (HCs and SCs) of the postnatal mammalian cochlea are unable to regenerate. However, with progress in regenerative medicine in the 21st century, several innovative approaches for achieving regeneration of inner ear HCs and SCs have become available. These methods include stem cell transplantation, overexpression of specific genes, and treatment with growth factors. Insulin-like growth factor-1 (IGF-1) is one of the growth factors that are involved in the development of the inner ear. Treatment with IGF-1 maintains HC numbers in the postnatal mammalian cochlea after various types of HC injuries, with activation of two major pathways downstream of IGF-1 signaling. In the aminoglycoside-treated neonatal mouse cochlear explant culture, promotion of the cell-cycle in SCs as well as inhibition of HC apoptosis was observed in the IGF-1-treated group. Activation of downstream molecules was observed in SCs and, in turn, SCs contribute to the maintenance of HC numbers. Using comprehensive analysis of the gene expression, the candidate effector molecules of the IGF-1 signaling pathway in the protection of HCs were identified as Netrin1 and Gap43. Based on these studies, a clinical trial has sought to investigate the effects of IGF-1 on SNHL. Sudden SNHL (SSHL) that was refractory to systemic steroids was treated with IGF-1 in a gelatin hydrogel and the outcome was compared with a historical control of hyperbaric oxygen therapy. The proportion of patients showing hearing improvement was significantly higher in the IGF-1-treatment group at 24 weeks after treatment than in the control group. A randomized clinical trial is ongoing to compare the effect of IGF-1 treatment with that of intra-tympanic steroids for SSHL that is refractory to systemic steroids.
Collapse
Affiliation(s)
- Norio Yamamoto
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University Kyoto Japan
| | - Takayuki Nakagawa
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University Kyoto Japan
| | - Juichi Ito
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University Kyoto Japan
| |
Collapse
|