1
|
Martinez E, Jadali A, Qiu J, Hinman AM, Ni JZ, Kim J, Kwan KY. CHD7 binds distinct regions in the Sox11 locus to regulate neuronal differentiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.02.646816. [PMID: 40236205 PMCID: PMC11996473 DOI: 10.1101/2025.04.02.646816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
The chromodomain helicase DNA binding protein 7 (CHD7) is a nucleosome repositioner implicated in multiple cellular processes, including neuronal differentiation. We identified CHD7 genome-wide binding sites that regulate neuronal differentiation in an otic stem cell line. We identified CHD7 enrichment at the Sox11 promoter and 3' untranslated region (UTR). Sox11 is a transcription factor essential for neuronal differentiation. CRISPRi of Sox11 promoter or 3'UTR displayed decreased neurite lengths and reduced neuronal marker expression TUBB3 expression. We showed that the Sox11 locus resides at TAD boundaries, and CTCF marks the 3'UTR. We propose that CHD7 modulates chromatin accessibility of the Sox11 promoter and CTCF-marked insulators in the 3'UTR to facilitate neuronal differentiation. CRISPRi of the insulator site alters 3D chromatin organization, affects gene expression and ultimately perturbs cellular processes. Our results implicate a general mechanism of CHD7 in facilitating neuronal differentiation and provide insight into CHD7 dysfunction in CHARGE syndrome, a congenital disorder associated with hearing loss.
Collapse
|
2
|
Wang M, Han Y, An W, Wang X, Chen F, Lu J, Meng Y, Li Y, Wang Y, Li J, Zhao C, Chai R, Wang H, Liu W, Xu L. Wnt signalling facilitates neuronal differentiation of cochlear Frizzled10-positive cells in mouse cochlea via glypican 6 modulation. Cell Commun Signal 2025; 23:50. [PMID: 39871249 PMCID: PMC11771042 DOI: 10.1186/s12964-025-02039-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 01/11/2025] [Indexed: 01/29/2025] Open
Abstract
Degeneration of cochlear spiral ganglion neurons (SGNs) leads to irreversible sensorineural hearing loss (SNHL), as SGNs lack regenerative capacity. Although cochlear glial cells (GCs) have some neuronal differentiation potential, their specific identities remain unclear. This study identifies a distinct subpopulation, Frizzled10 positive (FZD10+) cells, as an important type of GC responsible for neuronal differentiation in mouse cochlea. FZD10 + cells can differentiate into various SGN subtypes in vivo, adhering to natural proportions. Wnt signaling enhances the ability of FZD10 + cells to function as neural progenitors and increases the neuronal excitability of the FZD10-derived neurons. Single-cell RNA sequencing analysis characterizes FZD10-derived differentiating cell populations, while crosstalk network analysis identifies multiple signaling pathways and target genes influenced by Wnt signaling that contribute to the function of FZD10 + cells as neural progenitors. Pseudotime analysis maps the differentiation trajectory from proliferated GCs to differentiating neurons. Further experiments indicate that glypican 6 (GPC6) may regulate this neuronal lineage, while GPC6 deficiency diminishes the effects of Wnt signaling on FZD10-derived neuronal differentiation and synapse formation. These findings suggest the critical role of Wnt signaling in the neuronal differentiation derived from cochlear FZD10 + cells and provide insights into the mechanisms potentially involved in this process.
Collapse
Affiliation(s)
- Man Wang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China
- Shandong Institute of Otorhinolaryngology, Jinan, 250022, China
| | - Yuechen Han
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China
- Shandong Institute of Otorhinolaryngology, Jinan, 250022, China
| | - Weibin An
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China
- Shandong Institute of Otorhinolaryngology, Jinan, 250022, China
| | - Xue Wang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China
- Shandong Institute of Otorhinolaryngology, Jinan, 250022, China
| | - Fang Chen
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China
- Shandong Institute of Otorhinolaryngology, Jinan, 250022, China
| | - Junze Lu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China
- Shandong Institute of Otorhinolaryngology, Jinan, 250022, China
| | - Yu Meng
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China
- Shandong Institute of Otorhinolaryngology, Jinan, 250022, China
| | - Yan Li
- Translational Medical Research Centre, The First Hospital Affiliated to Shandong First Medical University& Shandong Provincial Qianfoshan Hospital, Jinan, 250013, China
| | - Yanqing Wang
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250100, China
| | - Jingxin Li
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250100, China
| | - Chunjie Zhao
- Key Laboratory of Developmental Genes and Human Diseases, School of Medicine, Ministry of Education, Southeast University, Nanjing, 210009, China
| | - Renjie Chai
- Key Laboratory of Developmental Genes and Human Diseases, School of Medicine, Ministry of Education, Southeast University, Nanjing, 210009, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| | - Haibo Wang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China.
- Shandong Institute of Otorhinolaryngology, Jinan, 250022, China.
| | - Wenwen Liu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China.
- Shandong Institute of Otorhinolaryngology, Jinan, 250022, China.
| | - Lei Xu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China.
- Shandong Institute of Otorhinolaryngology, Jinan, 250022, China.
| |
Collapse
|
3
|
Huang G, Zhou S, Zhu R, Wang Y, Chai Y. Effect of internal and external chaotic stimuli on synchronization of piezoelectric auditory neurons in coupled time-delay systems. Cogn Neurodyn 2024; 18:2111-2126. [PMID: 39104671 PMCID: PMC11297885 DOI: 10.1007/s11571-023-10042-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 10/27/2023] [Accepted: 11/09/2023] [Indexed: 08/07/2024] Open
Abstract
Hearing impairment is considered to be related to the damage of hair cells or synaptic terminals, which will cause varying degrees of hearing loss. Numerous studies have shown that cochlear implants can balance this damage. The human ear receives external acoustic signals mostly under complex conditions, and its biophysical mechanisms have important significance for reference in the design of cochlear implants. However, the relevant biophysical mechanisms have not yet been fully determined. Using the characteristics of special acoustoelectric conversion in piezoelectric ceramics, this paper integrates them into the traditional FitzHugh-Nagumo neuron circuit and proposes a comprehensive model with coupled auditory neurons. The model comprehensively considers the effects of synaptic coupling between neurons, information transmission delay, external noise stimulation, and internal chaotic current stimulation on the synchronization of membrane potential signals of two auditory neurons. The experimental results show that coupling strength, delay size, noise intensity, and chaotic current intensity all have a certain regulatory effect on synchronization stability. In particular, when auditory neurons are in a chaotic state, their impact on synchronization stability is sensitive. Numerical results provide a reference for exploring the biophysical mechanisms of auditory neurons. At the same time, we are committed to providing assistance in using sensors to monitor signals and repair hearing impairments.
Collapse
Affiliation(s)
- Guodong Huang
- School of Mathematics and Physics, Shanghai University of Electric Power, Shanghai, 201306 China
| | - Shu Zhou
- School of Mathematics and Physics, Shanghai University of Electric Power, Shanghai, 201306 China
| | - Rui Zhu
- School of Mathematics and Physics, Shanghai University of Electric Power, Shanghai, 201306 China
| | - Yunhai Wang
- School of Mathematics and Physics, Shanghai University of Electric Power, Shanghai, 201306 China
| | - Yuan Chai
- School of Mathematics and Physics, Shanghai University of Electric Power, Shanghai, 201306 China
| |
Collapse
|
4
|
Dönges L, Damle A, Mainardi A, Bock T, Schönenberger M, Martin I, Barbero A. Engineered human osteoarthritic cartilage organoids. Biomaterials 2024; 308:122549. [PMID: 38554643 DOI: 10.1016/j.biomaterials.2024.122549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 03/18/2024] [Accepted: 03/21/2024] [Indexed: 04/02/2024]
Abstract
The availability of human cell-based models capturing molecular processes of cartilage degeneration can facilitate development of disease-modifying therapies for osteoarthritis [1], a currently unmet clinical need. Here, by imposing specific inflammatory challenges upon mesenchymal stromal cells at a defined stage of chondrogenesis, we engineered a human organotypic model which recapitulates main OA pathological traits such as chondrocyte hypertrophy, cartilage matrix mineralization, enhanced catabolism and mechanical stiffening. To exemplify the utility of the model, we exposed the engineered OA cartilage organoids to factors known to attenuate pathological features, including IL-1Ra, and carried out mass spectrometry-based proteomics. We identified that IL-1Ra strongly reduced production of the transcription factor CCAAT/enhancer-binding protein beta [2] and demonstrated that inhibition of the C/EBPβ-activating kinases could revert the degradative processes. Human OA cartilage organoids thus represent a relevant tool towards the discovery of new molecular drivers of cartilage degeneration and the assessment of therapeutics targeting associated pathways.
Collapse
Affiliation(s)
- Laura Dönges
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031, Basel, Switzerland
| | - Atharva Damle
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031, Basel, Switzerland
| | - Andrea Mainardi
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031, Basel, Switzerland
| | - Thomas Bock
- Proteomics Core Facility, Biozentrum University of Basel, 4056, Basel, Switzerland
| | - Monica Schönenberger
- Nano Imaging Lab, Swiss Nanoscience Institute, University of Basel, 4056, Basel, Switzerland
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031, Basel, Switzerland.
| | - Andrea Barbero
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031, Basel, Switzerland
| |
Collapse
|
5
|
Zhao A, Xu W, Han R, Wei J, Yu Q, Wang M, Li H, Li M, Chi G. Role of histone modifications in neurogenesis and neurodegenerative disease development. Ageing Res Rev 2024; 98:102324. [PMID: 38762100 DOI: 10.1016/j.arr.2024.102324] [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: 12/10/2023] [Revised: 04/30/2024] [Accepted: 05/05/2024] [Indexed: 05/20/2024]
Abstract
Progressive neuronal dysfunction and death are key features of neurodegenerative diseases; therefore, promoting neurogenesis in neurodegenerative diseases is crucial. With advancements in proteomics and high-throughput sequencing technology, it has been demonstrated that histone post-transcriptional modifications (PTMs) are often altered during neurogenesis when the brain is affected by disease or external stimuli and that the degree of histone modification is closely associated with the development of neurodegenerative diseases. This review aimed to show the regulatory role of histone modifications in neurogenesis and neurodegenerative diseases by discussing the changing patterns and functional significance of histone modifications, including histone methylation, acetylation, ubiquitination, phosphorylation, and lactylation. Finally, we explored the control of neurogenesis and the development of neurodegenerative diseases by artificially modulating histone modifications.
Collapse
Affiliation(s)
- Anqi Zhao
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Wenhong Xu
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Rui Han
- Department of Neurovascular Surgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Junyuan Wei
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Qi Yu
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Miaomiao Wang
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Haokun Li
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Meiying Li
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| | - Guangfan Chi
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| |
Collapse
|
6
|
Tisi A, Palaniappan S, Maccarrone M. Advanced Omics Techniques for Understanding Cochlear Genome, Epigenome, and Transcriptome in Health and Disease. Biomolecules 2023; 13:1534. [PMID: 37892216 PMCID: PMC10605747 DOI: 10.3390/biom13101534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/10/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Advanced genomics, transcriptomics, and epigenomics techniques are providing unprecedented insights into the understanding of the molecular underpinnings of the central nervous system, including the neuro-sensory cochlea of the inner ear. Here, we report for the first time a comprehensive and updated overview of the most advanced omics techniques for the study of nucleic acids and their applications in cochlear research. We describe the available in vitro and in vivo models for hearing research and the principles of genomics, transcriptomics, and epigenomics, alongside their most advanced technologies (like single-cell omics and spatial omics), which allow for the investigation of the molecular events that occur at a single-cell resolution while retaining the spatial information.
Collapse
Affiliation(s)
- Annamaria Tisi
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Sakthimala Palaniappan
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Mauro Maccarrone
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
- Laboratory of Lipid Neurochemistry, European Center for Brain Research (CERC), Santa Lucia Foundation IRCCS, 00143 Rome, Italy
| |
Collapse
|
7
|
Nagamatsu ST, Pietrzak RH, Xu K, Krystal JH, Gelernter J, Montalvo-Ortiz JL. Dissecting the epigenomic differences between smoking and nicotine dependence in a veteran cohort. Addict Biol 2023; 28:e13259. [PMID: 36577721 DOI: 10.1111/adb.13259] [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: 01/14/2022] [Revised: 08/26/2022] [Accepted: 11/10/2022] [Indexed: 12/02/2022]
Abstract
Smoking is a serious public health issue linked to more than 8 million deaths per year worldwide and may lead to nicotine dependence (ND). Although the epigenomic literature on smoking is well established, studies evaluating the role of epigenetics in ND are limited. In this study, we examined the epigenomic signatures of ND and how these differ from smoking exposure to identify biomarkers specific to ND. We investigated the peripheral epigenetic profile of smoking status (SS) and ND in a US male veteran cohort. DNA from saliva was collected from 1135 European American (EA) male US military veterans. DNAm was assessed using the Illumina Infinium Human MethylationEPIC BeadChip array. SS was evaluated as current smokers (n = 137; 12.1%) and non-current smokers (never and former; n = 998; 87.9%). NDFTND was assessed as a continuous variable using the Fagerström Test for ND (FTND; n = 1135; mean = 2.54 ± 2.29). Epigenome-wide association studies (EWAS) and co-methylation analyses were conducted for SS and NDFTND . A total of 450 and 22 genome-wide significant differentially methylated sites (DMS) were associated with SS and NDFTND , respectively (15 overlapped DMS). We identified 97 DMS (43 genes) in SS-EWAS previously reported in the literature, including AHRR and F2RL3 genes (p-value: 1.95 × 10-83 to 4.55 × 10-33 ). NDFTND novel DMS mapped to NEUROG1, ANPEP, and SLC29A1. Co-methylation analysis identified 386 modules (11 SS-related and 19 NDFTND -related). SS-related modules showed enrichment for alcoholism, while NDFTND -related modules were enriched for nicotine addiction. This study confirms previous findings associated with SS and identifies novel and-potentially specific-epigenetic biomarkers of ND that may inform prognosis and novel treatment strategies.
Collapse
Affiliation(s)
- Sheila Tiemi Nagamatsu
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
- VA CT Healthcare Center, West Haven, Connecticut, USA
| | - Robert H Pietrzak
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
- VA CT Healthcare Center, West Haven, Connecticut, USA
- Clinical Neurosciences Division, U.S. Department of Veterans Affairs National Center of Posttraumatic Stress Disorder, West Haven, Connecticut, USA
| | - Ke Xu
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
- VA CT Healthcare Center, West Haven, Connecticut, USA
- Clinical Neurosciences Division, U.S. Department of Veterans Affairs National Center of Posttraumatic Stress Disorder, West Haven, Connecticut, USA
| | - John H Krystal
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
- VA CT Healthcare Center, West Haven, Connecticut, USA
- Clinical Neurosciences Division, U.S. Department of Veterans Affairs National Center of Posttraumatic Stress Disorder, West Haven, Connecticut, USA
| | - Joel Gelernter
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
- VA CT Healthcare Center, West Haven, Connecticut, USA
- Clinical Neurosciences Division, U.S. Department of Veterans Affairs National Center of Posttraumatic Stress Disorder, West Haven, Connecticut, USA
| | - Janitza Liz Montalvo-Ortiz
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
- VA CT Healthcare Center, West Haven, Connecticut, USA
- Clinical Neurosciences Division, U.S. Department of Veterans Affairs National Center of Posttraumatic Stress Disorder, West Haven, Connecticut, USA
| |
Collapse
|
8
|
Kim JH, Kim K, Kim I, Seong S, Koh JT, Kim N. Overexpression of Neurogenin 1 Negatively Regulates Osteoclast and Osteoblast Differentiation. Int J Mol Sci 2022; 23:ijms23126708. [PMID: 35743149 PMCID: PMC9223505 DOI: 10.3390/ijms23126708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 02/05/2023] Open
Abstract
Neurogenin 1 (Ngn1) belongs to the basic helix–loop–helix (bHLH) transcription factor family and plays important roles in specifying neuronal differentiation. The present study aimed to determine whether forced Ngn1 expression contributes to bone homeostasis. Ngn1 inhibited the p300/CREB-binding protein-associated factor (PCAF)-induced acetylation of nuclear factor of activated T cells 1 (NFATc1) and runt-related transcription factor 2 (Runx2) through binding to PCAF, which led to the inhibition of osteoclast and osteoblast differentiation, respectively. In addition, Ngn1 overexpression inhibited the TNF-α- and IL-17A-mediated enhancement of osteoclast differentiation and IL-17A-induced osteoblast differentiation. These findings indicate that Ngn1 can serve as a novel therapeutic agent for treating ankylosing spondylitis with abnormally increased bone formation and resorption.
Collapse
Affiliation(s)
- Jung Ha Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Korea; (J.H.K.); (K.K.); (I.K.); (S.S.)
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju 61186, Korea;
| | - Kabsun Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Korea; (J.H.K.); (K.K.); (I.K.); (S.S.)
| | - Inyoung Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Korea; (J.H.K.); (K.K.); (I.K.); (S.S.)
| | - Semun Seong
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Korea; (J.H.K.); (K.K.); (I.K.); (S.S.)
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju 61186, Korea;
| | - Jeong-Tae Koh
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju 61186, Korea;
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju 61186, Korea
| | - Nacksung Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Korea; (J.H.K.); (K.K.); (I.K.); (S.S.)
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju 61186, Korea;
- Correspondence: ; Tel.: +82-61-379-2835
| |
Collapse
|
9
|
Single-cell transcriptomic landscapes of the otic neuronal lineage at multiple early embryonic ages. Cell Rep 2022; 38:110542. [PMID: 35320729 DOI: 10.1016/j.celrep.2022.110542] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 12/03/2021] [Accepted: 02/25/2022] [Indexed: 11/20/2022] Open
Abstract
Inner ear vestibular and spiral ganglion neurons (VGNs and SGNs) are known to play pivotal roles in balance control and sound detection. However, the molecular mechanisms underlying otic neurogenesis at early embryonic ages have remained unclear. Here, we use single-cell RNA sequencing to reveal the transcriptomes of mouse otic tissues at three embryonic ages, embryonic day 9.5 (E9.5), E11.5, and E13.5, covering proliferating and undifferentiated otic neuroblasts and differentiating VGNs and SGNs. We validate the high quality of our studies by using multiple assays, including genetic fate mapping analysis, and we uncover several genes upregulated in neuroblasts or differentiating VGNs and SGNs, such as Shox2, Myt1, Casz1, and Sall3. Notably, our findings suggest a general cascaded differentiation trajectory during early otic neurogenesis. The comprehensive understanding of early otic neurogenesis provided by our study holds critical implications for both basic and translational research.
Collapse
|
10
|
Elliott KL, Fritzsch B, Yamoah EN, Zine A. Age-Related Hearing Loss: Sensory and Neural Etiology and Their Interdependence. Front Aging Neurosci 2022; 14:814528. [PMID: 35250542 PMCID: PMC8891613 DOI: 10.3389/fnagi.2022.814528] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 01/03/2022] [Indexed: 12/19/2022] Open
Abstract
Age-related hearing loss (ARHL) is a common, increasing problem for older adults, affecting about 1 billion people by 2050. We aim to correlate the different reductions of hearing from cochlear hair cells (HCs), spiral ganglion neurons (SGNs), cochlear nuclei (CN), and superior olivary complex (SOC) with the analysis of various reasons for each one on the sensory deficit profiles. Outer HCs show a progressive loss in a basal-to-apical gradient, and inner HCs show a loss in a apex-to-base progression that results in ARHL at high frequencies after 70 years of age. In early neonates, SGNs innervation of cochlear HCs is maintained. Loss of SGNs results in a considerable decrease (~50% or more) of cochlear nuclei in neonates, though the loss is milder in older mice and humans. The dorsal cochlear nuclei (fusiform neurons) project directly to the inferior colliculi while most anterior cochlear nuclei reach the SOC. Reducing the number of neurons in the medial nucleus of the trapezoid body (MNTB) affects the interactions with the lateral superior olive to fine-tune ipsi- and contralateral projections that may remain normal in mice, possibly humans. The inferior colliculi receive direct cochlear fibers and second-order fibers from the superior olivary complex. Loss of the second-order fibers leads to hearing loss in mice and humans. Although ARHL may arise from many complex causes, HC degeneration remains the more significant problem of hearing restoration that would replace the cochlear implant. The review presents recent findings of older humans and mice with hearing loss.
Collapse
Affiliation(s)
- Karen L. Elliott
- Department of Biology, University of Iowa, Iowa City, IA, United States
| | - Bernd Fritzsch
- Department of Biology, University of Iowa, Iowa City, IA, United States
- *Correspondence: Bernd Fritzsch
| | - Ebenezer N. Yamoah
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, United States
| | - Azel Zine
- LBN, Laboratory of Bioengineering and Nanoscience, University of Montpellier, Montpellier, France
| |
Collapse
|
11
|
Zebrafish foxc1a controls ventricular chamber maturation by directly regulating wwtr1 and nkx2.5 expression. J Genet Genomics 2021; 49:559-568. [PMID: 34923164 DOI: 10.1016/j.jgg.2021.12.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 12/06/2021] [Accepted: 12/06/2021] [Indexed: 11/22/2022]
Abstract
Chamber maturation is a significant process in cardiac development. Disorders of this crucial process lead to a range of congenital heart defects. Foxc1a is a critical transcription factor reported to regulate the specification of cardiac progenitor cells. However, little is known about the role of Foxc1a in modulating chamber maturation. Previously, we reported that foxc1a-null zebrafish embryos exhibit disrupted heart structures and functions. In this study, we observed that ventricle structure and cardiomyocyte proliferation were abolished during chamber maturation in foxc1a-null zebrafish embryos. To observe the endogenous localization of Foxc1a in the hearts of living embryos, we inserted eyfp at the foxc1a genomic locus using TALEN. Analysis of the knockin zebrafish showed that foxc1a was widely expressed in ventricular cardiomyocytes during chamber development. Cardiac RNA sequencing analysis revealed downregulated expression of the Hippo signaling effector wwtr1. Dual-luciferase and chromatin immunoprecipitation assays revealed that Foxc1a could bind directly to three sites in the wwtr1 promoter region. Furthermore, wwtr1 mRNA overexpression was sufficient to reverse the ventricle defects during chamber maturation. Conditional overexpression of nkx2.5 also partially rescued the ventricular defects during chamber development. These findings demonstrate that wwtr1 and nkx2.5 are direct targets of Foxc1a during ventricular chamber maturation.
Collapse
|
12
|
Aviña-Padilla K, Ramírez-Rafael JA, Herrera-Oropeza GE, Muley VY, Valdivia DI, Díaz-Valenzuela E, García-García A, Varela-Echavarría A, Hernández-Rosales M. Evolutionary Perspective and Expression Analysis of Intronless Genes Highlight the Conservation of Their Regulatory Role. Front Genet 2021; 12:654256. [PMID: 34306008 PMCID: PMC8302217 DOI: 10.3389/fgene.2021.654256] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 06/01/2021] [Indexed: 11/13/2022] Open
Abstract
The structure of eukaryotic genes is generally a combination of exons interrupted by intragenic non-coding DNA regions (introns) removed by RNA splicing to generate the mature mRNA. A fraction of genes, however, comprise a single coding exon with introns in their untranslated regions or are intronless genes (IGs), lacking introns entirely. The latter code for essential proteins involved in development, growth, and cell proliferation and their expression has been proposed to be highly specialized for neuro-specific functions and linked to cancer, neuropathies, and developmental disorders. The abundant presence of introns in eukaryotic genomes is pivotal for the precise control of gene expression. Notwithstanding, IGs exempting splicing events entail a higher transcriptional fidelity, making them even more valuable for regulatory roles. This work aimed to infer the functional role and evolutionary history of IGs centered on the mouse genome. IGs consist of a subgroup of genes with one exon including coding genes, non-coding genes, and pseudogenes, which conform approximately 6% of a total of 21,527 genes. To understand their prevalence, biological relevance, and evolution, we identified and studied 1,116 IG functional proteins validating their differential expression in transcriptomic data of embryonic mouse telencephalon. Our results showed that overall expression levels of IGs are lower than those of MEGs. However, strongly up-regulated IGs include transcription factors (TFs) such as the class 3 of POU (HMG Box), Neurog1, Olig1, and BHLHe22, BHLHe23, among other essential genes including the β-cluster of protocadherins. Most striking was the finding that IG-encoded BHLH TFs fit the criteria to be classified as microproteins. Finally, predicted protein orthologs in other six genomes confirmed high conservation of IGs associated with regulating neural processes and with chromatin organization and epigenetic regulation in Vertebrata. Moreover, this study highlights that IGs are essential modulators of regulatory processes, such as the Wnt signaling pathway and biological processes as pivotal as sensory organ developing at a transcriptional and post-translational level. Overall, our results suggest that IG proteins have specialized, prevalent, and unique biological roles and that functional divergence between IGs and MEGs is likely to be the result of specific evolutionary constraints.
Collapse
Affiliation(s)
- Katia Aviña-Padilla
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
- Centro de Investigacioìn y de Estudios Avanzados del IPN, Unidad Irapuato, Guanajuato, Mexico
| | | | - Gabriel Emilio Herrera-Oropeza
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, United Kingdom
| | | | - Dulce I. Valdivia
- Centro de Investigacioìn y de Estudios Avanzados del IPN, Unidad Irapuato, Guanajuato, Mexico
| | - Erik Díaz-Valenzuela
- Centro de Investigacioìn y de Estudios Avanzados del IPN, Unidad Irapuato, Guanajuato, Mexico
| | - Andrés García-García
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | | | | |
Collapse
|
13
|
Elliott KL, Pavlinkova G, Chizhikov VV, Yamoah EN, Fritzsch B. Neurog1, Neurod1, and Atoh1 are essential for spiral ganglia, cochlear nuclei, and cochlear hair cell development. Fac Rev 2021; 10:47. [PMID: 34131657 PMCID: PMC8170689 DOI: 10.12703/r/10-47] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
We review the molecular basis of three related basic helix–loop–helix (bHLH) genes (Neurog1, Neurod1, and Atoh1) and upstream regulators Eya1/Six1, Sox2, Pax2, Gata3, Fgfr2b, Foxg1, and Lmx1a/b during the development of spiral ganglia, cochlear nuclei, and cochlear hair cells. Neuronal development requires early expression of Neurog1, followed by its downstream target Neurod1, which downregulates Atoh1 expression. In contrast, hair cells and cochlear nuclei critically depend on Atoh1 and require Neurod1 and Neurog1 expression for various aspects of development. Several experiments show a partial uncoupling of Atoh1/Neurod1 (spiral ganglia and cochlea) and Atoh1/Neurog1/Neurod1 (cochlear nuclei). In this review, we integrate the cellular and molecular mechanisms that regulate the development of auditory system and provide novel insights into the restoration of hearing loss, beyond the limited generation of lost sensory neurons and hair cells.
Collapse
Affiliation(s)
- Karen L Elliott
- Department of Biology, University of Iowa, Iowa City, IA, USA
| | - Gabriela Pavlinkova
- Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czechia
| | - Victor V Chizhikov
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Ebenezer N Yamoah
- Department of Physiology and Cell Biology, University of Nevada, Reno, NV, USA
| | - Bernd Fritzsch
- Department of Biology, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
14
|
Elliott KL, Pavlínková G, Chizhikov VV, Yamoah EN, Fritzsch B. Development in the Mammalian Auditory System Depends on Transcription Factors. Int J Mol Sci 2021; 22:ijms22084189. [PMID: 33919542 PMCID: PMC8074135 DOI: 10.3390/ijms22084189] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/16/2022] Open
Abstract
We review the molecular basis of several transcription factors (Eya1, Sox2), including the three related genes coding basic helix–loop–helix (bHLH; see abbreviations) proteins (Neurog1, Neurod1, Atoh1) during the development of spiral ganglia, cochlear nuclei, and cochlear hair cells. Neuronal development requires Neurog1, followed by its downstream target Neurod1, to cross-regulate Atoh1 expression. In contrast, hair cells and cochlear nuclei critically depend on Atoh1 and require Neurod1 expression for interactions with Atoh1. Upregulation of Atoh1 following Neurod1 loss changes some vestibular neurons’ fate into “hair cells”, highlighting the significant interplay between the bHLH genes. Further work showed that replacing Atoh1 by Neurog1 rescues some hair cells from complete absence observed in Atoh1 null mutants, suggesting that bHLH genes can partially replace one another. The inhibition of Atoh1 by Neurod1 is essential for proper neuronal cell fate, and in the absence of Neurod1, Atoh1 is upregulated, resulting in the formation of “intraganglionic” HCs. Additional genes, such as Eya1/Six1, Sox2, Pax2, Gata3, Fgfr2b, Foxg1, and Lmx1a/b, play a role in the auditory system. Finally, both Lmx1a and Lmx1b genes are essential for the cochlear organ of Corti, spiral ganglion neuron, and cochlear nuclei formation. We integrate the mammalian auditory system development to provide comprehensive insights beyond the limited perception driven by singular investigations of cochlear neurons, cochlear hair cells, and cochlear nuclei. A detailed analysis of gene expression is needed to understand better how upstream regulators facilitate gene interactions and mammalian auditory system development.
Collapse
Affiliation(s)
- Karen L. Elliott
- Department of Biology, University of Iowa, Iowa City, IA 52242, USA;
| | - Gabriela Pavlínková
- Institute of Biotechnology of the Czech Academy of Sciences, 25250 Vestec, Czechia;
| | - Victor V. Chizhikov
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN 38163, USA;
| | - Ebenezer N. Yamoah
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV 89557, USA;
| | - Bernd Fritzsch
- Department of Biology, University of Iowa, Iowa City, IA 52242, USA;
- Correspondence:
| |
Collapse
|
15
|
Chen H, Li S, Xu W, Hong Y, Dou R, Shen H, Liu X, Wu T, He JC. Interleukin-17A promotes the differentiation of bone marrow mesenchymal stem cells into neuronal cells. Tissue Cell 2021; 69:101482. [PMID: 33418236 DOI: 10.1016/j.tice.2020.101482] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/17/2020] [Accepted: 12/21/2020] [Indexed: 11/25/2022]
Abstract
Ischemia or hemorrhagic stroke is one of the leading causes of death and permanent disability in the worldwide population. As a consequence of the potential increasing in stroke, stem cell therapy is currently an area of intense focus. However, there are less data available regarding the promotion of healing efficacy after stroke. The present study aimed to investigate whether the cytokine interleukin-17A (IL-17A) could have a role in promoting the neuronal differentiation of mesenchymal stem cells (MSCs) and to investigate the associated molecular mechanism. Firstly, different concentration of IL-17A at range from 5-40 ng/mL was applied to stimulate bone marrow MSCs (BMSCs) during the course of neurogenic differentiation. Then reverse transcription-PCR, histological analyses and immunofluorescence assays were used to determine the optimum concentration of IL-17A in promoting the neuronal differentiation of BMSCs, which was 20 ng/mL. Mechanistically, Wnt signaling pathway was activated and Notch signaling pathway was suppressed. In addition, there were antergic effect of these two signaling pathways modulating the neurogenic differentiation of BMSCs induced by IL-17A. The present study demonstrated the potential role of IL-17A-based BMSCs strategy for promoting neuronal differentiation in vitro. However, the treatment efficacy could be considerably confirmed in animals with ischemia stroke. Therefore, a more sophisticated strategy that addresses the complicated treatment associated with stroke is needed.
Collapse
Affiliation(s)
- Hanlin Chen
- Stomatologic Hospital & College, Anhui Medical University, Key Lab of Oral Diseases Research of Anhui Province, 69 Meishan Road, Shushan District, Hefei, Anhui, 230001, China
| | - Shasha Li
- Stomatologic Hospital & College, Anhui Medical University, Key Lab of Oral Diseases Research of Anhui Province, 69 Meishan Road, Shushan District, Hefei, Anhui, 230001, China
| | - Wanting Xu
- Stomatologic Hospital & College, Anhui Medical University, Key Lab of Oral Diseases Research of Anhui Province, 69 Meishan Road, Shushan District, Hefei, Anhui, 230001, China
| | - Yongfeng Hong
- Department of Rehabilitation Medicine, Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Shushan District, Hefei, Anhui, 230061, China
| | - Rengang Dou
- Department of Rehabilitation Medicine, Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Shushan District, Hefei, Anhui, 230061, China
| | - Hongtao Shen
- Department of Rehabilitation Medicine, Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Shushan District, Hefei, Anhui, 230061, China
| | - Xue Liu
- Department of Rehabilitation Medicine, Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Shushan District, Hefei, Anhui, 230061, China
| | - Tingting Wu
- Stomatologic Hospital & College, Anhui Medical University, Key Lab of Oral Diseases Research of Anhui Province, 69 Meishan Road, Shushan District, Hefei, Anhui, 230001, China.
| | - Jia Cai He
- Stomatologic Hospital & College, Anhui Medical University, Key Lab of Oral Diseases Research of Anhui Province, 69 Meishan Road, Shushan District, Hefei, Anhui, 230001, China.
| |
Collapse
|
16
|
Peng T, Peng JJ, Miao GY, Tan ZQ, Liu B, Zhou E. miR‑125/CDK2 axis in cochlear progenitor cell proliferation. Mol Med Rep 2020; 23:102. [PMID: 33300064 PMCID: PMC7723065 DOI: 10.3892/mmr.2020.11741] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/16/2020] [Indexed: 12/25/2022] Open
Abstract
Hearing loss ranks fourth among the principal causes of disability worldwide, and manipulation of progenitor cells may be a key strategy for hair cell regeneration. The present study investigated the role and mechanism of miR‑125 on the proliferation of cochlear progenitor cells (CPCs). CPCs were isolated from the cochleae of neonatal rats, and their morphology was observed. Furthermore, the differentiation ability of CPCs was determined by assessing the expression of 5‑bromodeoxyuridine (BrdU), nestin and myosin VII by immunofluorescence. The expression levels of miR‑125 and cyclin‑dependent kinase 2 (CDK2) as well as the cell proliferation of CPCs were assessed. In addition, following gain‑ and loss‑of‑function assays, the cell cycle was examined by flow cytometry, and the expression levels of miR‑125, CDK2, proliferating cell nuclear antigen (PCNA) and nestin were determined by reverse transcription‑quantitative PCR and western blotting. The binding sites between miR‑125 and CDK2 were predicted by TargetScan and identified by the dual luciferase reporter assay. The results demonstrated that different types of progenitor spheres were observed from CPCs with positive expression of BrdU, nestin and myosin VII. Following in vitro incubation for 2, 4 and 7 days, the spheres were enlarged, and CPC proliferation gradually increased and reached a plateau after further incubation for 3 days. Furthermore, the expression levels of nestin and PCNA in CPCs increased and then decreased during in vitro incubation for 2, 4 and 7 days. Following this incubation, the expression levels of miR‑125 in CPCs decreased; thereafter, its expression increased, and the expression pattern was different from that of CDK2. In addition, miR‑125 overexpression in CPCs decreased the expression of CDK2 and the number of cells in the S phase. Different expression patterns were found in CPCs in response to the miR‑125 knockdown. In addition, miR‑125 directly targeted CDK2. Simultaneous knockdown of miR‑125 and CDK2 enhanced CPC proliferation compared with CDK2 knockdown alone. Taken together, the findings from the present study suggested that miR‑125 may inhibit CPC proliferation by downregulating CDK2. The present study may provide a novel therapeutic direction for treatment of hearing loss.
Collapse
Affiliation(s)
- Tao Peng
- Department of Otolaryngology and Head and Neck Surgery, Hunan Provincial People's Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410007, P.R. China
| | - Jing-Jing Peng
- Department of Obstetrics and Gynecology, Changsha Maternal and Child Health Care Hospital, Changsha, Hunan 410005, P.R. China
| | - Gang-Yong Miao
- Department of Otolaryngology and Head and Neck Surgery, Hunan Provincial People's Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410007, P.R. China
| | - Zhi-Qiang Tan
- Department of Otolaryngology and Head and Neck Surgery, Hunan Provincial People's Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410007, P.R. China
| | - Bin Liu
- Department of Otolaryngology and Head and Neck Surgery, Hunan Provincial People's Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410007, P.R. China
| | - En Zhou
- Department of Otolaryngology and Head and Neck Surgery, Hunan Provincial People's Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410007, P.R. China
| |
Collapse
|
17
|
Lai R, Cai C, Wu W, Hu P, Wang Q. Exosomes derived from mouse inner ear stem cells attenuate gentamicin-induced ototoxicity in vitro through the miR-182-5p/FOXO3 axis. J Tissue Eng Regen Med 2020; 14:1149-1156. [PMID: 32593214 DOI: 10.1002/term.3089] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/13/2020] [Accepted: 06/22/2020] [Indexed: 01/06/2023]
Abstract
Gentamicin-induced cochlear hair cell ototoxicity, such as oxidative stress and apoptosis, could be attenuated by mouse inner ear stem cells (IESCs). However, it is still unclear whether such protective effects could be mediated by exosomes derived from IESCs (IESCs-ex). In the present study, HEI-OC1 cells were exposed to gentamicin (2 mM) to establish an ototoxicity model and further treated with exosomes isolated from miR-182-5p transferred or non-transferred IESCs. IESCs-ex improved HEI-OC1 cell viability, as assayed by the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyltetrazolium bromide method, and alleviated the oxidative stress response induced by the gentamicin treatment, as confirmed by measuring the malondialdehyde, superoxide dismutase, catalase, and glutathione peroxidase levels. IESCs-ex increased relative miR-182-5p expression and decreased FOXO3 expression in the gentamicin-exposed HEI-OC1 cells. Furthermore, exosomes derived from miR-182-5p mimics that were pre-treated with IESCs could increase miR-182-5p and Bcl-2 expressions and decrease FOXO3 and Bax expressions in gentamicin-exposed HEI-OC1 cells. All of these results indicate that IESCs-ex could attenuate gentamicin-induced HEI-OC1 cell apoptosis and oxidative stress through the miR-182-5p/FOXO3 axis.
Collapse
Affiliation(s)
- Ruosha Lai
- Department of Otolaryngology and Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Cuiyun Cai
- Department of Otolaryngology and Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Weijing Wu
- Department of Otolaryngology and Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Peng Hu
- Department of Otolaryngology and Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qin Wang
- Department of Otolaryngology and Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
18
|
Flitsch LJ, Laupman KE, Brüstle O. Transcription Factor-Based Fate Specification and Forward Programming for Neural Regeneration. Front Cell Neurosci 2020; 14:121. [PMID: 32508594 PMCID: PMC7251072 DOI: 10.3389/fncel.2020.00121] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 04/14/2020] [Indexed: 12/11/2022] Open
Abstract
Traditionally, in vitro generation of donor cells for brain repair has been dominated by the application of extrinsic growth factors and morphogens. Recent advances in cell engineering strategies such as reprogramming of somatic cells into induced pluripotent stem cells and direct cell fate conversion have impressively demonstrated the feasibility to manipulate cell identities by the overexpression of cell fate-determining transcription factors. These strategies are now increasingly implemented for transcription factor-guided differentiation of neural precursors and forward programming of pluripotent stem cells toward specific neural subtypes. This review covers major achievements, pros and cons, as well as future prospects of transcription factor-based cell fate specification and the applicability of these approaches for the generation of donor cells for brain repair.
Collapse
Affiliation(s)
- Lea J Flitsch
- Institute of Reconstructive Neurobiology, Life & Brain Center, University of Bonn Medical Faculty and University Hospital Bonn, Bonn, Germany
| | - Karen E Laupman
- Institute of Reconstructive Neurobiology, Life & Brain Center, University of Bonn Medical Faculty and University Hospital Bonn, Bonn, Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, Life & Brain Center, University of Bonn Medical Faculty and University Hospital Bonn, Bonn, Germany
| |
Collapse
|
19
|
Li X, Bi Z, Sun Y, Li C, Li Y, Liu Z. In vivo ectopic Ngn1 and Neurod1 convert neonatal cochlear glial cells into spiral ganglion neurons. FASEB J 2020; 34:4764-4782. [PMID: 32027432 DOI: 10.1096/fj.201902118r] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 01/14/2020] [Accepted: 01/22/2020] [Indexed: 12/22/2022]
Abstract
Damage or degeneration of inner ear spiral ganglion neurons (SGNs) causes hearing impairment. Previous in vitro studies indicate that cochlear glial cells can be reprogrammed into SGNs, however, it remains unknown whether this can occur in vivo. Here, we show that neonatal glial cells can be converted, in vivo, into SGNs (defined as new SGNs) by simultaneous induction of Neurog1 (Ngn1) and Neurod1. New SGNs express SGN markers, Tuj1, Map2, Prox1, Mafb and Gata3, and reduce glial cell marker Sox10 and Scn7a. The heterogeneity within new SGNs is illustrated by immunostaining and transcriptomic assays. Transcriptomes analysis indicates that well reprogrammed SGNs are similar to type I SGNs. In addition, reprogramming efficiency is positively correlated with the dosage of Ngn1 and Neurod1, but declined with aging. Taken together, our in vivo data demonstrates the plasticity of cochlear neonatal glial cells and the capacity of Ngn1 and Neurod1 to reprogram glial cells into SGNs. Looking ahead, we expect that combination of Neurog1 and Neurod1 along with other factors will further boost the percentage of fully converted (Mafb+/Gata3+) new SGNs.
Collapse
Affiliation(s)
- Xiang Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Zhenghong Bi
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yidi Sun
- CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Shanghai, China
| | - Chao Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yixue Li
- University of Chinese Academy of Sciences, Shanghai, China.,Bio-Med Big Data Center, Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhiyong Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| |
Collapse
|
20
|
Song Z, Laureano AS, Patel K, Yip S, Jadali A, Kwan KY. Single-Cell Fluorescence Analysis of Pseudotemporal Ordered Cells Provides Protein Expression Dynamics for Neuronal Differentiation. Front Cell Dev Biol 2019; 7:87. [PMID: 31192206 PMCID: PMC6549217 DOI: 10.3389/fcell.2019.00087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 05/08/2019] [Indexed: 12/31/2022] Open
Abstract
Stem cell replacement therapy is a potential method for repopulating lost spiral ganglion neurons (SGNs) in the inner ear. Efficacy of cell replacement relies on proper differentiation. Defining the dynamic expression of different transcription factors essential for neuronal differentiation allows us to monitor the progress and determine when the protein functions in differentiating stem cell cultures. Using immortalized multipotent otic progenitors (iMOPs) as a cellular system for SGN differentiation, a method for determining dynamic protein expression from heterogeneous cultures was developed. iMOP-derived neurons were identified and ordered by increasing neurite lengths to create a pseudotime course that reflects the differentiation trajectory. The fluorescence intensities of transcription factors SOX2 and NEUROD1 from individual pseudotemporally ordered cells were measured. Individual cells were grouped by K-means clustering and the mean fluorescence intensity for each cluster determined. Curve fit of the mean fluorescence represented the protein expression dynamics in differentiating cells. The method provides information about protein expression dynamics in differentiating stem cell cultures.
Collapse
Affiliation(s)
- Zhichao Song
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States.,Stem Cell Research Center and Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ, United States
| | - Alejandra S Laureano
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States.,Stem Cell Research Center and Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ, United States
| | - Kishan Patel
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
| | - Sylvia Yip
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
| | - Azadeh Jadali
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States.,Stem Cell Research Center and Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ, United States
| | - Kelvin Y Kwan
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States.,Stem Cell Research Center and Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ, United States
| |
Collapse
|
21
|
Walters BJ, Cox BC. Approaches for the study of epigenetic modifications in the inner ear and related tissues. Hear Res 2019; 376:69-85. [PMID: 30679030 PMCID: PMC6456365 DOI: 10.1016/j.heares.2019.01.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/12/2018] [Accepted: 01/11/2019] [Indexed: 12/12/2022]
Abstract
DNA methylation and histone modifications such as methylation, acetylation, and phosphorylation, are two types of epigenetic modifications that alter gene expression. These additions to DNA regulatory elements or to the tails of histones can be inherited or can also occur de novo. Since epigenetic modifications can have significant effects on various processes at both the cellular and organismal level, there has been a rapid increase in research on this topic throughout all fields of biology in recent years. However, epigenetic research is relativity new for the inner ear field, likely due to the limited number of cells present and their quiescent nature. Here, we provide an overview of methods used to detect DNA methylation and histone modifications with a focus on those that have been validated for use with limited cell numbers and a discussion of the strengths and limitations for each. We also provide examples for how these methods have been used to investigate the epigenetic landscape in the inner ear and related tissues.
Collapse
Affiliation(s)
- Bradley J Walters
- Departments of Neurobiology and Anatomical Sciences, and of Otolaryngology and Communicative Sciences, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Brandon C Cox
- Departments of Pharmacology and Surgery, Division of Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL 62711, USA.
| |
Collapse
|
22
|
Shin JO, Lee JJ, Kim M, Chung YW, Min H, Kim JY, Kim HP, Bok J. CTCF Regulates Otic Neurogenesis via Histone Modification in the Neurog1 Locus. Mol Cells 2018; 41:695-702. [PMID: 30008200 PMCID: PMC6078853 DOI: 10.14348/molcells.2018.0230] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 06/04/2018] [Indexed: 11/27/2022] Open
Abstract
The inner ear is a complex sensory organ responsible for hearing and balance. Formation of the inner ear is dependent on tight regulation of spatial and temporal expression of genes that direct a series of developmental processes. Recently, epigenetic regulation has emerged as a crucial regulator of the development of various organs. However, what roles higher-order chromatin organization and its regulator molecules play in inner ear development are unclear. CCCTC-binding factor (CTCF) is a highly conserved 11-zinc finger protein that regulates the three-dimensional architecture of chromatin, and is involved in various gene regulation processes. To delineate the role of CTCF in inner ear development, the present study investigated inner ear-specific Ctcf knockout mouse embryos (Pax2-Cre; Ctcffl/fl ). The loss of Ctcf resulted in multiple defects of inner ear development and severely compromised otic neurogenesis, which was partly due to a loss of Neurog1 expression. Furthermore, reduced Neurog1 gene expression by CTCF knockdown was found to be associated with changes in histone modification at the gene's promoter, as well as its upstream enhancer. The results of the present study demonstrate that CTCF plays an essential role in otic neurogenesis by modulating histone modification in the Neurog1 locus.
Collapse
Affiliation(s)
- Jeong-Oh Shin
- Department of Anatomy, Yonsei University College of Medicine, Seoul 03722,
Korea
| | - Jong-Joo Lee
- Department of Environmental Medical Biology, Yonsei University College of Medicine, Seoul 03722,
Korea
- BK21 PLUS project for Medical Science, Yonsei University College of Medicine, Seoul 03722,
Korea
| | - Mikyoung Kim
- Department of Environmental Medical Biology, Yonsei University College of Medicine, Seoul 03722,
Korea
| | - Youn Wook Chung
- Department of Environmental Medical Biology, Yonsei University College of Medicine, Seoul 03722,
Korea
| | - Hyehyun Min
- Department of Anatomy, Yonsei University College of Medicine, Seoul 03722,
Korea
| | - Jae-Yoon Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul 03722,
Korea
- BK21 PLUS project for Medical Science, Yonsei University College of Medicine, Seoul 03722,
Korea
| | - Hyoung-Pyo Kim
- Department of Environmental Medical Biology, Yonsei University College of Medicine, Seoul 03722,
Korea
- BK21 PLUS project for Medical Science, Yonsei University College of Medicine, Seoul 03722,
Korea
| | - Jinwoong Bok
- Department of Anatomy, Yonsei University College of Medicine, Seoul 03722,
Korea
- BK21 PLUS project for Medical Science, Yonsei University College of Medicine, Seoul 03722,
Korea
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722,
Korea
| |
Collapse
|
23
|
Meas SJ, Zhang CL, Dabdoub A. Reprogramming Glia Into Neurons in the Peripheral Auditory System as a Solution for Sensorineural Hearing Loss: Lessons From the Central Nervous System. Front Mol Neurosci 2018; 11:77. [PMID: 29593497 PMCID: PMC5861218 DOI: 10.3389/fnmol.2018.00077] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 02/26/2018] [Indexed: 12/12/2022] Open
Abstract
Disabling hearing loss affects over 5% of the world’s population and impacts the lives of individuals from all age groups. Within the next three decades, the worldwide incidence of hearing impairment is expected to double. Since a leading cause of hearing loss is the degeneration of primary auditory neurons (PANs), the sensory neurons of the auditory system that receive input from mechanosensory hair cells in the cochlea, it may be possible to restore hearing by regenerating PANs. A direct reprogramming approach can be used to convert the resident spiral ganglion glial cells into induced neurons to restore hearing. This review summarizes recent advances in reprogramming glia in the CNS to suggest future steps for regenerating the peripheral auditory system. In the coming years, direct reprogramming of spiral ganglion glial cells has the potential to become one of the leading biological strategies to treat hearing impairment.
Collapse
Affiliation(s)
- Steven J Meas
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Chun-Li Zhang
- Department of Molecular Biology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Alain Dabdoub
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada.,Department of Otolaryngology - Head & Neck Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
24
|
Perny M, Ting CC, Kleinlogel S, Senn P, Roccio M. Generation of Otic Sensory Neurons from Mouse Embryonic Stem Cells in 3D Culture. Front Cell Neurosci 2017; 11:409. [PMID: 29311837 PMCID: PMC5742223 DOI: 10.3389/fncel.2017.00409] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 12/05/2017] [Indexed: 12/29/2022] Open
Abstract
The peripheral hearing process taking place in the cochlea mainly depends on two distinct sensory cell types: the mechanosensitive hair cells and the spiral ganglion neurons (SGNs). The first respond to the mechanical stimulation exerted by sound pressure waves on their hair bundles by releasing neurotransmitters and thereby activating the latter. Loss of these sensorineural cells is associated with permanent hearing loss. Stem cell-based approaches aiming at cell replacement or in vitro drug testing to identify potential ototoxic, otoprotective, or regenerative compounds have lately gained attention as putative therapeutic strategies for hearing loss. Nevertheless, they rely on efficient and reliable protocols for the in vitro generation of cochlear sensory cells for their implementation. To this end, we have developed a differentiation protocol based on organoid culture systems, which mimics the most important steps of in vivo otic development, robustly guiding mouse embryonic stem cells (mESCs) toward otic sensory neurons (OSNs). The stepwise differentiation of mESCs toward ectoderm was initiated using a quick aggregation method in presence of Matrigel in serum-free conditions. Non-neural ectoderm was induced via activation of bone morphogenetic protein (BMP) signaling and concomitant inhibition of transforming growth factor beta (TGFβ) signaling to prevent mesendoderm induction. Preplacodal and otic placode ectoderm was further induced by inhibition of BMP signaling and addition of fibroblast growth factor 2 (FGF2). Delamination and differentiation of SGNs was initiated by plating of the organoids on a 2D Matrigel-coated substrate. Supplementation with brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3) was used for further maturation until 15 days of in vitro differentiation. A large population of neurons with a clear bipolar morphology and functional excitability was derived from these cultures. Immunostaining and gene expression analysis performed at different time points confirmed the transition trough the otic lineage and final expression of the key OSN markers. Moreover, the stem cell-derived OSNs exhibited functional electrophysiological properties of native SGNs. Our established in vitro model of OSNs development can be used for basic developmental studies, for drug screening or for the exploration of their regenerative potential.
Collapse
Affiliation(s)
- Michael Perny
- Neuroinfection Laboratory, Institute for Infectious Diseases, University of Bern, Bern, Switzerland.,Laboratory of Inner Ear Research, Department for BioMedical Research, University of Bern, Bern, Switzerland.,Department of Otorhinolaryngology, Head and Neck Surgery, Inselspital, University of Bern, Bern, Switzerland.,Cluster for Regenerative Neuroscience, Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Ching-Chia Ting
- Laboratory of Inner Ear Research, Department for BioMedical Research, University of Bern, Bern, Switzerland.,Department of Otorhinolaryngology, Head and Neck Surgery, Inselspital, University of Bern, Bern, Switzerland.,Cluster for Regenerative Neuroscience, Department of Biomedical Research, University of Bern, Bern, Switzerland
| | | | - Pascal Senn
- Laboratory of Inner Ear Research, Department for BioMedical Research, University of Bern, Bern, Switzerland.,Department of Otorhinolaryngology, Head and Neck Surgery, Inselspital, University of Bern, Bern, Switzerland.,Cluster for Regenerative Neuroscience, Department of Biomedical Research, University of Bern, Bern, Switzerland.,Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Geneva (HUG), Geneva, Switzerland
| | - Marta Roccio
- Laboratory of Inner Ear Research, Department for BioMedical Research, University of Bern, Bern, Switzerland.,Department of Otorhinolaryngology, Head and Neck Surgery, Inselspital, University of Bern, Bern, Switzerland.,Cluster for Regenerative Neuroscience, Department of Biomedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|