1
|
Nouvian R. [Two is better than one: CaBP1 and CaBP2 control calcium signaling in auditory inner hair cells]. Med Sci (Paris) 2024; 40:971-972. [PMID: 39705569 DOI: 10.1051/medsci/2024165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2024] Open
Affiliation(s)
- Régis Nouvian
- Institut des neurosciences de Montpellier, Université de Montpellier, Inserm, CNRS, Montpellier, France
| |
Collapse
|
2
|
Peng H, Wang L, Gao Y, Liu H, Lin G, Kong Y, Xu P, Liu H, Yuan Q, Liu H, Song L, Yang T, Wu H. DMXL2 Is Required for Endocytosis and Recycling of Synaptic Vesicles in Auditory Hair Cells. J Neurosci 2024; 44:e1405232024. [PMID: 39147590 PMCID: PMC11411588 DOI: 10.1523/jneurosci.1405-23.2024] [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: 07/24/2023] [Revised: 07/10/2024] [Accepted: 08/04/2024] [Indexed: 08/17/2024] Open
Abstract
Ribbon synapses of inner hair cells (IHCs) are uniquely designed for ultrafast and indefatigable neurotransmission of the sound. The molecular machinery ensuring the efficient, compensatory recycling of the synaptic vesicles (SVs), however, remains elusive. This study showed that hair cell knock-out of murine Dmxl2, whose human homolog is responsible for nonsyndromic sensorineural hearing loss DFNA71, resulted in auditory synaptopathy by impairing synaptic endocytosis and recycling. The mutant mice in the C57BL/6J background of either sex had mild hearing loss with severely diminished wave I amplitude of the auditory brainstem response. Membrane capacitance measurements of the IHCs revealed deficiency in sustained synaptic exocytosis and endocytic membrane retrieval. Consistent with the electrophysiological findings, 3D electron microscopy reconstruction showed reduced reserve pool of SVs and endocytic compartments, while the membrane-proximal and ribbon-associated vesicles remain intact. Our results propose an important role of DMXL2 in hair cell endocytosis and recycling of the SVs.
Collapse
Affiliation(s)
- Hu Peng
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200023, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai 200125, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai 200125, China
- Department of Otolaryngology-Head and Neck Surgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Longhao Wang
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200023, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai 200125, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai 200125, China
| | - Yunge Gao
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200023, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai 200125, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai 200125, China
| | - Huihui Liu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200023, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai 200125, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai 200125, China
| | - Guotong Lin
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200023, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai 200125, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai 200125, China
| | - Yu Kong
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Pengcheng Xu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200023, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai 200125, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai 200125, China
| | - Hongchao Liu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200023, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai 200125, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai 200125, China
| | - Qingyue Yuan
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200023, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai 200125, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai 200125, China
| | - Huanhai Liu
- Department of Otolaryngology-Head and Neck Surgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Lei Song
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200023, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai 200125, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai 200125, China
| | - Tao Yang
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200023, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai 200125, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai 200125, China
| | - Hao Wu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200023, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai 200125, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai 200125, China
| |
Collapse
|
3
|
Liu JB, Yuan HL, Zhang G, Ke JB. Comprehensive Characterization of a Subfamily of Ca 2+-Binding Proteins in Mouse and Human Retinal Neurons at Single-Cell Resolution. eNeuro 2024; 11:ENEURO.0145-24.2024. [PMID: 39260891 PMCID: PMC11419601 DOI: 10.1523/eneuro.0145-24.2024] [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: 04/02/2024] [Revised: 08/20/2024] [Accepted: 09/03/2024] [Indexed: 09/13/2024] Open
Abstract
Ca2+-binding proteins (CaBPs; CaBP1-5) are a subfamily of neuronal Ca2+ sensors with high homology to calmodulin. Notably, CaBP4, which is exclusively expressed in rod and cone photoreceptors, is crucial for maintaining normal retinal functions. However, the functional roles of CaBP1, CaBP2, and CaBP5 in the retina remain elusive, primarily due to limited understanding of their expression patterns within inner retinal neurons. In this study, we conducted a comprehensive transcript analysis using single-cell RNA sequencing datasets to investigate the gene expression profiles of CaBPs in mouse and human retinal neurons. Our findings revealed notable similarities in the overall expression patterns of CaBPs across both species. Specifically, nearly all amacrine cell, ganglion cell, and horizontal cell types exclusively expressed CaBP1. In contrast, the majority of bipolar cell types, including rod bipolar (RB) cells, expressed distinct combinations of CaBP1, CaBP2, and CaBP5, rather than a single CaBP as previously hypothesized. Remarkably, mouse rods and human cones exclusively expressed CaBP4, whereas mouse cones and human rods coexpressed both CaBP4 and CaBP5. Our single-cell reverse transcription polymerase chain reaction analysis confirmed the coexpression CaBP1 and CaBP5 in individual RBs from mice of either sex. Additionally, all three splice variants of CaBP1, primarily L-CaBP1, were detected in mouse RBs. Taken together, our study offers a comprehensive overview of the distribution of CaBPs in mouse and human retinal neurons, providing valuable insights into their roles in visual functions.
Collapse
Affiliation(s)
- Jun-Bin Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - He-Lan Yuan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Gong Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Jiang-Bin Ke
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou 325000, China
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| |
Collapse
|
4
|
Nouvian R. Letting the calcium flow. eLife 2024; 13:e96139. [PMID: 38334748 PMCID: PMC10857785 DOI: 10.7554/elife.96139] [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] [Indexed: 02/10/2024] Open
Abstract
Two calcium-binding proteins, CaBP1 and CaBP2, cooperate to keep calcium channels in the hair cells of the inner ear open.
Collapse
Affiliation(s)
- Régis Nouvian
- Institute for Neurosciences of Montpellier, Univ Montpellier, Inserm, CNRS, Montpellier, France
| |
Collapse
|
5
|
Nawaz H, Parveen A, Khan SA, Zalan AK, Khan MA, Muhammad N, Hassib NF, Mostafa MI, Elhossini RM, Roshdy NN, Ullah A, Arif A, Khan S, Ammerpohl O, Wasif N. Brachyolmia, dental anomalies and short stature (DASS): Phenotype and genotype analyses of Egyptian and Pakistani patients. Heliyon 2024; 10:e23688. [PMID: 38192829 PMCID: PMC10772639 DOI: 10.1016/j.heliyon.2023.e23688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 11/29/2023] [Accepted: 12/09/2023] [Indexed: 01/10/2024] Open
Abstract
Brachyolmia is a heterogeneous group of developmental disorders characterized by a short trunk, short stature, scoliosis, and generalized platyspondyly without significant deformities in the long bones. DASS (Dental Abnormalities and Short Stature), caused by alterations in the LTBP3 gene, was previously considered as a subtype of brachyolmia. The present study investigated three unrelated consanguineous families (A, B, C) with Brachyolmia and DASS from Egypt and Pakistan. In our Egyptian patients, we also observed hearing impairment. Exome sequencing was performed to determine the genetic causes of the diverse clinical conditions in the patients. Exome sequencing identified a novel homozygous splice acceptor site variant (LTBP3:c.3629-1G > T; p. ?) responsible for DASS phenotypes and a known homozygous missense variant (CABP2: c.590T > C; p.Ile197Thr) causing hearing impairment in the Egyptian patients. In addition, two previously reported homozygous frameshift variants (LTBP3:c.132delG; p.Pro45Argfs*25) and (LTBP3:c.2216delG; p.Gly739Alafs*7) were identified in Pakistani patients. This study emphasizes the vital role of LTBP3 in the axial skeleton and tooth morphogenesis and expands the mutational spectrum of LTBP3. We are reporting LTBP3 variants in seven patients of three families, majorly causing brachyolmia with dental and cardiac anomalies. Skeletal assessment documented short webbed neck, broad chest, evidences of mild long bones involvement, short distal phalanges, pes planus and osteopenic bone texture as additional associated findings expanding the clinical phenotype of DASS. The current study reveals that the hearing impairment phenotype in Egyptian patients of family A has a separate transmission mechanism independent of LTBP3.
Collapse
Affiliation(s)
- Hamed Nawaz
- Department of Biotechnology and Genetic Engineering, Kohat University of Science and Technology (KUST), Kohat, Pakistan
| | - Asia Parveen
- Department of Biochemistry, Faculty of Life Sciences, Gulab Devi Educational Complex, Gulab Devi Hospital, 54000, Lahore, Pakistan
- Faculty of Science and Technology, University of Central Punjab (UCP), Lahore, Pakistan
| | - Sher Alam Khan
- Department of Biotechnology and Genetic Engineering, Kohat University of Science and Technology (KUST), Kohat, Pakistan
- Department of Computer Science and Bioinformatics, Khushal Khan Khatak University, Karak, Pakistan
| | - Abul Khair Zalan
- BDS, MDS Registrar Pediatric Dentistry, Department of Pediatric Dentistry, School of Dentistry, PIMS, Islamabad, Pakistan
| | - Muhammad Adnan Khan
- Dental Material, Institute of Basic Medical Sciences, Khyber Medical University Peshawar, Peshawar, Pakistan
| | - Noor Muhammad
- Department of Biotechnology and Genetic Engineering, Kohat University of Science and Technology (KUST), Kohat, Pakistan
| | - Nehal F. Hassib
- Orodental Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, 12622, Egypt
- School of Dentistry, New Giza University, Giza, Egypt
| | - Mostafa I. Mostafa
- Orodental Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, 12622, Egypt
| | - Rasha M. Elhossini
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, 12622, Egypt
| | - Nehal Nabil Roshdy
- Endodontics, Faculty of Dentistry, Cairo University, Cairo, 11553, Egypt
| | - Asmat Ullah
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Amina Arif
- Faculty of Science and Technology, University of Central Punjab (UCP), Lahore, Pakistan
| | - Saadullah Khan
- Department of Biotechnology and Genetic Engineering, Kohat University of Science and Technology (KUST), Kohat, Pakistan
| | - Ole Ammerpohl
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, 89081, Ulm, Germany
| | - Naveed Wasif
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, 89081, Ulm, Germany
- Institute of Human Genetics, University Hospital Schleswig-Holstein, Campus Kiel, D-24105, Kiel, Germany
| |
Collapse
|
6
|
Yang Y, Murtha K, Climer LK, Ceriani F, Thompson P, Hornak AJ, Marcotti W, Simmons DD. Oncomodulin regulates spontaneous calcium signalling and maturation of afferent innervation in cochlear outer hair cells. J Physiol 2023; 601:4291-4308. [PMID: 37642186 PMCID: PMC10621907 DOI: 10.1113/jp284690] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 08/08/2023] [Indexed: 08/31/2023] Open
Abstract
Cochlear outer hair cells (OHCs) are responsible for the exquisite frequency selectivity and sensitivity of mammalian hearing. During development, the maturation of OHC afferent connectivity is refined by coordinated spontaneous Ca2+ activity in both sensory and non-sensory cells. Calcium signalling in neonatal OHCs can be modulated by oncomodulin (OCM, β-parvalbumin), an EF-hand calcium-binding protein. Here, we investigated whether OCM regulates OHC spontaneous Ca2+ activity and afferent connectivity during development. Using a genetically encoded Ca2+ sensor (GCaMP6s) expressed in OHCs in wild-type (Ocm+/+ ) and Ocm knockout (Ocm-/- ) littermates, we found increased spontaneous Ca2+ activity and upregulation of purinergic receptors in OHCs from Ocm-/- cochlea immediately following birth. The afferent synaptic maturation of OHCs was delayed in the absence of OCM, leading to an increased number of ribbon synapses and afferent fibres on Ocm-/- OHCs before hearing onset. We propose that OCM regulates the spontaneous Ca2+ signalling in the developing cochlea and the maturation of OHC afferent innervation. KEY POINTS: Cochlear outer hair cells (OHCs) exhibit spontaneous Ca2+ activity during a narrow period of neonatal development. OHC afferent maturation and connectivity requires spontaneous Ca2+ activity. Oncomodulin (OCM, β-parvalbumin), an EF-hand calcium-binding protein, modulates Ca2+ signals in immature OHCs. Using transgenic mice that endogenously expressed a Ca2+ sensor, GCaMP6s, we found increased spontaneous Ca2+ activity and upregulated purinergic receptors in Ocm-/- OHCs. The maturation of afferent synapses in Ocm-/- OHCs was also delayed, leading to an upregulation of ribbon synapses and afferent fibres in Ocm-/- OHCs before hearing onset. We propose that OCM plays an important role in modulating Ca2+ activity, expression of Ca2+ channels and afferent innervation in developing OHCs.
Collapse
Affiliation(s)
- Yang Yang
- Department of Biology, Baylor University, 101 Bagby Ave, Waco, TX
| | - Kaitlin Murtha
- Department of Biology, Baylor University, 101 Bagby Ave, Waco, TX
| | - Leslie K. Climer
- Department of Biology, Baylor University, 101 Bagby Ave, Waco, TX
| | - Federico Ceriani
- School of Biosciences, University of Sheffield, S10 2TN Sheffield, United Kingdom
| | - Pierce Thompson
- Department of Biology, Baylor University, 101 Bagby Ave, Waco, TX
| | - Aubrey J. Hornak
- Department of Biology, Baylor University, 101 Bagby Ave, Waco, TX
| | - Walter Marcotti
- School of Biosciences, University of Sheffield, S10 2TN Sheffield, United Kingdom
- Sheffield Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Dwayne D. Simmons
- Department of Biology, Baylor University, 101 Bagby Ave, Waco, TX
- School of Biosciences, University of Sheffield, S10 2TN Sheffield, United Kingdom
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
- Department of Psychology and Neuroscience, Baylor University, Waco, TX
| |
Collapse
|
7
|
Kameyama M, Minobe E, Shao D, Xu J, Gao Q, Hao L. Regulation of Cardiac Cav1.2 Channels by Calmodulin. Int J Mol Sci 2023; 24:ijms24076409. [PMID: 37047381 PMCID: PMC10094977 DOI: 10.3390/ijms24076409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/19/2023] [Accepted: 03/23/2023] [Indexed: 03/31/2023] Open
Abstract
Cav1.2 Ca2+ channels, a type of voltage-gated L-type Ca2+ channel, are ubiquitously expressed, and the predominant Ca2+ channel type, in working cardiac myocytes. Cav1.2 channels are regulated by the direct interactions with calmodulin (CaM), a Ca2+-binding protein that causes Ca2+-dependent facilitation (CDF) and inactivation (CDI). Ca2+-free CaM (apoCaM) also contributes to the regulation of Cav1.2 channels. Furthermore, CaM indirectly affects channel activity by activating CaM-dependent enzymes, such as CaM-dependent protein kinase II and calcineurin (a CaM-dependent protein phosphatase). In this article, we review the recent progress in identifying the role of apoCaM in the channel ‘rundown’ phenomena and related repriming of channels, and CDF, as well as the role of Ca2+/CaM in CDI. In addition, the role of CaM in channel clustering is reviewed.
Collapse
Affiliation(s)
- Masaki Kameyama
- Department of Physiology, Graduate School of Medical & Dental Sciences, Kagoshima University, Sakura-ga-oka, Kagoshima 890-8544, Japan
- Correspondence:
| | - Etsuko Minobe
- Department of Physiology, Graduate School of Medical & Dental Sciences, Kagoshima University, Sakura-ga-oka, Kagoshima 890-8544, Japan
| | - Dongxue Shao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang 110012, China (L.H.)
| | - Jianjun Xu
- Department of Physiology, Graduate School of Medical & Dental Sciences, Kagoshima University, Sakura-ga-oka, Kagoshima 890-8544, Japan
| | - Qinghua Gao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang 110012, China (L.H.)
| | - Liying Hao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang 110012, China (L.H.)
| |
Collapse
|
8
|
Yang Y, Murtha K, Climer LK, Ceriani F, Thompson P, Hornak AJ, Marcotti W, Simmons DD. Oncomodulin Regulates Spontaneous Calcium Signaling and Maturation of Afferent Innervation in Cochlear Outer Hair Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.01.529895. [PMID: 36909575 PMCID: PMC10002690 DOI: 10.1101/2023.03.01.529895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Cochlear outer hair cells (OHCs) are responsible for the exquisite frequency selectivity and sensitivity of mammalian hearing. During development, the maturation of OHC afferent connectivity is refined by coordinated spontaneous Ca 2+ activity in both sensory and non-sensory cells. Calcium signaling in neonatal OHCs can be modulated by Oncomodulin (OCM, β-parvalbumin), an EF-hand calcium-binding protein. Here, we investigated whether OCM regulates OHC spontaneous Ca 2+ activity and afferent connectivity during development. Using a genetically encoded Ca 2+ sensor (GCaMP6s) expressed in OHCs in wild-type (Ocm +/+ ) and Ocm knockout (Ocm -/- ) littermates, we found increased spontaneous Ca 2+ activity and upregulation of purinergic receptors in OHCs from GCaMP6s Ocm -/- cochlea immediately following birth. The afferent synaptic maturation of OHCs was delayed in the absence of OCM, leading to an increased number of ribbon synapses and afferent fibers on GCaMP6s Ocm -/- OHCs before hearing onset. We propose that OCM regulates the spontaneous Ca 2+ signaling in the developing cochlea and the maturation of OHC afferent innervation.
Collapse
|
9
|
Calcium signaling and genetic rare diseases: An auditory perspective. Cell Calcium 2023; 110:102702. [PMID: 36791536 DOI: 10.1016/j.ceca.2023.102702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/07/2023]
Abstract
Deafness is a highly heterogeneous disorder which stems, for 50%, from genetic origins. Sensory transduction relies mainly on sensory hair cells of the cochlea, in the inner ear. Calcium is key for the function of these cells and acts as a fundamental signal transduction. Its homeostasis depends on three factors: the calcium influx, through the mechanotransduction channel at the apical pole of the hair cell as well as the voltage-gated calcium channel at the base of the cells; the calcium buffering via Ca2+-binding proteins in the cytoplasm, but also in organelles such as mitochondria and the reticulum endoplasmic mitochondria-associated membranes with specialized proteins; and the calcium extrusion through the Ca-ATPase pump, located all over the plasma membrane. In addition, the synaptic transmission to the central nervous system is also controlled by calcium. Genetic studies of inherited deafness have tremendously helped understand the underlying molecular pathways of calcium signaling. In this review, we discuss these different factors in light of the associated genetic diseases (syndromic and non-syndromic deafness) and the causative genes.
Collapse
|
10
|
Lopez JA, Yamamoto A, Vecchi JT, Hagen J, Lee K, Sonka M, Hansen MR, Lee A. Caldendrin represses neurite regeneration and growth in dorsal root ganglion neurons. Sci Rep 2023; 13:2608. [PMID: 36788334 PMCID: PMC9929226 DOI: 10.1038/s41598-023-29622-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/07/2023] [Indexed: 02/16/2023] Open
Abstract
Caldendrin is a Ca2+ binding protein that interacts with multiple effectors, such as the Cav1 L-type Ca2+ channel, which play a prominent role in regulating the outgrowth of dendrites and axons (i.e., neurites) during development and in response to injury. Here, we investigated the role of caldendrin in Cav1-dependent pathways that impinge upon neurite growth in dorsal root ganglion neurons (DRGNs). By immunofluorescence, caldendrin was localized in medium- and large- diameter DRGNs. Compared to DRGNs cultured from WT mice, DRGNs of caldendrin knockout (KO) mice exhibited enhanced neurite regeneration and outgrowth. Strong depolarization, which normally represses neurite growth through activation of Cav1 channels, had no effect on neurite growth in DRGN cultures from female caldendrin KO mice. Remarkably, DRGNs from caldendrin KO males were no different from those of WT males in terms of depolarization-dependent neurite growth repression. We conclude that caldendrin opposes neurite regeneration and growth, and this involves coupling of Cav1 channels to growth-inhibitory pathways in DRGNs of females but not males.
Collapse
Affiliation(s)
- Josue A Lopez
- Department of Neuroscience, University of Texas-Austin, 100 E. 24th St., Austin, TX, 78712, USA
| | - Annamarie Yamamoto
- Department of Neuroscience, University of Texas-Austin, 100 E. 24th St., Austin, TX, 78712, USA
| | - Joseph T Vecchi
- Department of Molecular Physiology and Biophysics and Otolaryngology Head-Neck Surgery, Iowa Neuroscience Institute, Pappajohn Biomedical Institute, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Jussara Hagen
- Department of Molecular Physiology and Biophysics and Otolaryngology Head-Neck Surgery, Iowa Neuroscience Institute, Pappajohn Biomedical Institute, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Kyungmoo Lee
- Electrical and Computer Engineering, Iowa Institute for Biomedical Imaging, University of Iowa, 51 Newton Rd. Iowa City, Iowa, 52242, USA
| | - Milan Sonka
- Electrical and Computer Engineering, Iowa Institute for Biomedical Imaging, University of Iowa, 51 Newton Rd. Iowa City, Iowa, 52242, USA
| | - Marlan R Hansen
- Department of Molecular Physiology and Biophysics and Otolaryngology Head-Neck Surgery, Iowa Neuroscience Institute, Pappajohn Biomedical Institute, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Amy Lee
- Department of Neuroscience, University of Texas-Austin, 100 E. 24th St., Austin, TX, 78712, USA.
| |
Collapse
|
11
|
Shi T, Beaulieu MO, Saunders LM, Fabian P, Trapnell C, Segil N, Crump JG, Raible DW. Single-cell transcriptomic profiling of the zebrafish inner ear reveals molecularly distinct hair cell and supporting cell subtypes. eLife 2023; 12:82978. [PMID: 36598134 PMCID: PMC9851615 DOI: 10.7554/elife.82978] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 01/04/2023] [Indexed: 01/05/2023] Open
Abstract
A major cause of human deafness and vestibular dysfunction is permanent loss of the mechanosensory hair cells of the inner ear. In non-mammalian vertebrates such as zebrafish, regeneration of missing hair cells can occur throughout life. While a comparative approach has the potential to reveal the basis of such differential regenerative ability, the degree to which the inner ears of fish and mammals share common hair cells and supporting cell types remains unresolved. Here, we perform single-cell RNA sequencing of the zebrafish inner ear at embryonic through adult stages to catalog the diversity of hair cells and non-sensory supporting cells. We identify a putative progenitor population for hair cells and supporting cells, as well as distinct hair and supporting cell types in the maculae versus cristae. The hair cell and supporting cell types differ from those described for the lateral line system, a distributed mechanosensory organ in zebrafish in which most studies of hair cell regeneration have been conducted. In the maculae, we identify two subtypes of hair cells that share gene expression with mammalian striolar or extrastriolar hair cells. In situ hybridization reveals that these hair cell subtypes occupy distinct spatial domains within the three macular organs, the utricle, saccule, and lagena, consistent with the reported distinct electrophysiological properties of hair cells within these domains. These findings suggest that primitive specialization of spatially distinct striolar and extrastriolar hair cells likely arose in the last common ancestor of fish and mammals. The similarities of inner ear cell type composition between fish and mammals validate zebrafish as a relevant model for understanding inner ear-specific hair cell function and regeneration.
Collapse
Affiliation(s)
- Tuo Shi
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
- Caruso Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Marielle O Beaulieu
- Department of Otolaryngology-Head and Neck Surgery, University of WashingtonSeattleUnited States
| | - Lauren M Saunders
- Department of Genome Sciences, University of WashingtonSeattleUnited States
| | - Peter Fabian
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Cole Trapnell
- Department of Genome Sciences, University of WashingtonSeattleUnited States
| | - Neil Segil
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
- Caruso Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - J Gage Crump
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - David W Raible
- Department of Otolaryngology-Head and Neck Surgery, University of WashingtonSeattleUnited States
- Department of Genome Sciences, University of WashingtonSeattleUnited States
- Department of Biological Structure, University of WashingtonSeattleUnited States
| |
Collapse
|
12
|
Zong P, Yue L. Regulation of Presynaptic Calcium Channels. ADVANCES IN NEUROBIOLOGY 2023; 33:171-202. [PMID: 37615867 DOI: 10.1007/978-3-031-34229-5_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Voltage-gated calcium channels (VGCCs), especially Cav2.1 and Cav2.2, are the major mediators of Ca2+ influx at the presynaptic membrane in response to neuron excitation, thereby exerting a predominant control on synaptic transmission. To guarantee the timely and precise release of neurotransmitters at synapses, the activity of presynaptic VGCCs is tightly regulated by a variety of factors, including auxiliary subunits, membrane potential, G protein-coupled receptors (GPCRs), calmodulin (CaM), Ca2+-binding proteins (CaBP), protein kinases, various interacting proteins, alternative splicing events, and genetic variations.
Collapse
Affiliation(s)
- Pengyu Zong
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Lixia Yue
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine, Farmington, CT, USA.
| |
Collapse
|
13
|
Bharadwaj T, Schrauwen I, Acharya A, Nouel‐Saied LM, Väisänen M, Kraatari M, Rahikkala E, Jarvela I, Kotimäki J, Leal SM. Autosomal recessive nonsyndromic hearing impairment in two Finnish families due to the population enriched CABP2 c.637+1G>T variant. Mol Genet Genomic Med 2022; 10:e1866. [PMID: 35150090 PMCID: PMC8922966 DOI: 10.1002/mgg3.1866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 11/10/2021] [Accepted: 12/14/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND The genetic architecture of hearing impairment in Finland is largely unknown. Here, we investigated two Finnish families with autosomal recessive nonsyndromic symmetrical moderate-to-severe hearing impairment. METHODS Exome and custom capture next-generation sequencing were used to detect the underlying cause of hearing impairment. RESULTS In both Finnish families, we identified a homozygous pathogenic splice site variant c.637+1G>T in CAPB2 that is known to cause autosomal recessive nonsyndromic hearing impairment. Four CABP2 variants have been reported to underlie autosomal recessive nonsyndromic hearing impairment in eight families from Iran, Turkey, Pakistan, Italy, and Denmark. Of these variants, the pathogenic splice site variant c.637+1G>T is the most prevalent. The c.637+1G>T variant is enriched in the Finnish population, which has undergone multiple bottlenecks that can lead to the higher frequency of certain variants including those involved in disease. CONCLUSION We report two Finnish families with hearing impairment due to the CABP2 splice site variant c.637+1G>T.
Collapse
Affiliation(s)
- Thashi Bharadwaj
- Center for Statistical GeneticsGertrude H. Sergievsky Center, and the Department of NeurologyColumbia University Medical CenterNew YorkNYUSA
| | - Isabelle Schrauwen
- Center for Statistical GeneticsGertrude H. Sergievsky Center, and the Department of NeurologyColumbia University Medical CenterNew YorkNYUSA
| | - Anushree Acharya
- Center for Statistical GeneticsGertrude H. Sergievsky Center, and the Department of NeurologyColumbia University Medical CenterNew YorkNYUSA
| | - Liz M. Nouel‐Saied
- Center for Statistical GeneticsGertrude H. Sergievsky Center, and the Department of NeurologyColumbia University Medical CenterNew YorkNYUSA
| | - Marja‐Leena Väisänen
- Northern Finland Laboratory Centre NordLab and Medical Research CentreOulu University Hospital and University of OuluOuluFinland
| | - Minna Kraatari
- Department of Clinical GeneticsPEDEGO Research Unit and Medical Research Center OuluOulu University Hospital and University of OuluOuluFinland
| | - Elisa Rahikkala
- Department of Clinical GeneticsPEDEGO Research Unit and Medical Research Center OuluOulu University Hospital and University of OuluOuluFinland
- Institute of BiomedicineUniversity of TurkuTurkuFinland
| | - Irma Jarvela
- Department of Medical GeneticsUniversity of HelsinkiHelsinkiFinland
| | - Jouko Kotimäki
- Department of OtorhinolaryngologyKainuu Central HospitalKajaaniFinland
| | - Suzanne M. Leal
- Center for Statistical GeneticsGertrude H. Sergievsky Center, and the Department of NeurologyColumbia University Medical CenterNew YorkNYUSA
- Taub Institute for Alzheimer’s Disease and the Aging BrainColumbia University Medical CenterNew YorkNYUSA
| |
Collapse
|
14
|
Oestreicher D, Picher MM, Rankovic V, Moser T, Pangrsic T. Cabp2-Gene Therapy Restores Inner Hair Cell Calcium Currents and Improves Hearing in a DFNB93 Mouse Model. Front Mol Neurosci 2021; 14:689415. [PMID: 34489639 PMCID: PMC8417311 DOI: 10.3389/fnmol.2021.689415] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/28/2021] [Indexed: 12/02/2022] Open
Abstract
Clinical management of auditory synaptopathies like other genetic hearing disorders is currently limited to the use of hearing aids or cochlear implants. However, future gene therapy promises restoration of hearing in selected forms of monogenic hearing impairment, in which cochlear morphology is preserved over a time window that enables intervention. This includes non-syndromic autosomal recessive hearing impairment DFNB93, caused by defects in the CABP2 gene. Calcium-binding protein 2 (CaBP2) is a potent modulator of inner hair cell (IHC) voltage-gated calcium channels CaV1.3. Based on disease modeling in Cabp2–/– mice, DFNB93 hearing impairment has been ascribed to enhanced steady-state inactivation of IHC CaV1.3 channels, effectively limiting their availability to trigger synaptic transmission. This, however, does not seem to interfere with cochlear development and does not cause early degeneration of hair cells or their synapses. Here, we studied the potential of a gene therapeutic approach for the treatment of DFNB93. We used AAV2/1 and AAV-PHP.eB viral vectors to deliver the Cabp2 coding sequence into IHCs of early postnatal Cabp2–/– mice and assessed the level of restoration of hair cell function and hearing. Combining in vitro and in vivo approaches, we observed high transduction efficiency, and restoration of IHC CaV1.3 function resulting in improved hearing of Cabp2–/– mice. These preclinical results prove the feasibility of DFNB93 gene therapy.
Collapse
Affiliation(s)
- David Oestreicher
- Experimental Otology Group, InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany.,Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Maria Magdalena Picher
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
| | - Vladan Rankovic
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Restorative Cochlear Genomics Group, Auditory Neuroscience and Optogenetics Laboratory, German Primate Center, Göttingen, Germany
| | - Tobias Moser
- Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Collaborative Research Center 889, University of Göttingen, Göttingen, Germany.,Multiscale Bioimaging Cluster of Excellence (MBExC), University of Göttingen, Göttingen, Germany
| | - Tina Pangrsic
- Experimental Otology Group, InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany.,Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Collaborative Research Center 889, University of Göttingen, Göttingen, Germany.,Multiscale Bioimaging Cluster of Excellence (MBExC), University of Göttingen, Göttingen, Germany
| |
Collapse
|
15
|
Sheyanth IN, Højland AT, Okkels H, Lolas I, Thorup C, Petersen MB. First reported CABP2-related non-syndromic hearing loss in Northern Europe. Mol Genet Genomic Med 2021; 9:e1639. [PMID: 33666369 PMCID: PMC8123739 DOI: 10.1002/mgg3.1639] [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: 06/11/2020] [Revised: 08/06/2020] [Accepted: 02/10/2021] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND CABP2-related non-syndromic hearing loss have only been reported in a few families worldwide (Iran, Turkey, Pakistan and Italy). The hearing loss was in these cases described as prelingual, symmetrical, and moderate to severe. METHODS Following DNA isolation, exome sequencing was performed in 123 genes related to non-syndromic hearing loss. Variant verification and carrier testing were performed by direct sequencing. RESULTS We report the first Northern European individual with CABP2-related hearing loss: an 8-year-old Danish Caucasian boy with non-syndromic, prelingual, and sensorineural hearing loss, who is homozygous for the splice site variant CABP2: c. 637+1G>T previously found in three Iranian families and in one Pakistani family. Both parents are of Danish Caucasian origin with no known history of consanguinity. This is in contrast to the four reported Middle Eastern families, who all were consanguineous. However, loss of heterozygosity in a 3.2 Mb area on chromosome 11 including CABP2 was observed, suggesting a common parental ancestor. CONCLUSION We report the first case of CABP2-related autosomal recessive hearing loss in Northern Europe. The index is of Danish Caucasian origin and found to be homozygous for the splice site variant c.637+1G>T.
Collapse
Affiliation(s)
- Inger Norlyk Sheyanth
- Research and Knowledge Center in Sensory Genetics, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Genetics, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Allan Thomas Højland
- Research and Knowledge Center in Sensory Genetics, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Genetics, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Henrik Okkels
- Research and Knowledge Center in Sensory Genetics, Aalborg University Hospital, Aalborg, Denmark.,Department of Molecular Diagnostics, Aalborg University Hospital, Aalborg, Denmark
| | - Ihab Lolas
- Research and Knowledge Center in Sensory Genetics, Aalborg University Hospital, Aalborg, Denmark.,Department of Molecular Diagnostics, Aalborg University Hospital, Aalborg, Denmark
| | - Christian Thorup
- Research and Knowledge Center in Sensory Genetics, Aalborg University Hospital, Aalborg, Denmark.,Department of Audiology, Aarhus University Hospital, Aarhus, Denmark
| | - Michael Bjørn Petersen
- Research and Knowledge Center in Sensory Genetics, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Genetics, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
16
|
Dolphin AC. Functions of Presynaptic Voltage-gated Calcium Channels. FUNCTION (OXFORD, ENGLAND) 2020; 2:zqaa027. [PMID: 33313507 PMCID: PMC7709543 DOI: 10.1093/function/zqaa027] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/16/2020] [Accepted: 10/20/2020] [Indexed: 01/06/2023]
Abstract
Voltage-gated calcium channels are the principal conduits for depolarization-mediated Ca2+ entry into excitable cells. In this review, the biophysical properties of the relevant members of this family of channels, those that are present in presynaptic terminals, will be discussed in relation to their function in mediating neurotransmitter release. Voltage-gated calcium channels have properties that ensure they are specialized for particular roles, for example, differences in their activation voltage threshold, their various kinetic properties, and their voltage-dependence of inactivation. All these attributes play into the ability of the various voltage-gated calcium channels to participate in different patterns of presynaptic vesicular release. These include synaptic transmission resulting from single action potentials, and longer-term changes mediated by bursts or trains of action potentials, as well as release resulting from graded changes in membrane potential in specialized sensory synapses.
Collapse
Affiliation(s)
- Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, WC1E 6BT, UK,Address correspondence to A.C.D. (e-mail: )
| |
Collapse
|
17
|
Sameera, Shah FA, Rashid S. Conformational ensembles of non-peptide ω-conotoxin mimetics and Ca +2 ion binding to human voltage-gated N-type calcium channel Ca v2.2. Comput Struct Biotechnol J 2020; 18:2357-2372. [PMID: 32994894 PMCID: PMC7498737 DOI: 10.1016/j.csbj.2020.08.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/24/2020] [Accepted: 08/26/2020] [Indexed: 02/07/2023] Open
Abstract
Chronic neuropathic pain is the most complex and challenging clinical problem of a population that sets a major physical and economic burden at the global level. Ca2+-permeable channels functionally orchestrate the processing of pain signals. Among them, N-type voltage-gated calcium channels (VGCC) hold prominent contribution in the pain signal transduction and serve as prime targets for synaptic transmission block and attenuation of neuropathic pain. Here, we present detailed in silico analysis to comprehend the underlying conformational changes upon Ca2+ ion passage through Cav2.2 to differentially correlate subtle transitions induced via binding of a conopeptide-mimetic alkylphenyl ether-based analogue MVIIA. Interestingly, pronounced conformational changes were witnessed at the proximal carboxyl-terminus of Cav2.2 that attained an upright orientation upon Ca+2 ion permeability. Moreover, remarkable changes were observed in the architecture of channel tunnel. These findings illustrate that inhibitor binding to Cav2.2 may induce more narrowing in the pore size as compared to Ca2+ binding through modulating the hydrophilicity pattern at the selectivity region. A significant reduction in the tunnel volume at the selectivity filter and its enhancement at the activation gate of Ca+2-bound Cav2.2 suggests that ion binding modulates the outward splaying of pore-lining S6 helices to open the voltage gate. Overall, current study delineates dynamic conformational ensembles in terms of Ca+2 ion and MVIIA-associated structural implications in the Cav2.2 that may help in better therapeutic intervention to chronic and neuropathic pain management.
Collapse
Affiliation(s)
- Sameera
- National Center for Bioinformatics, Quaid-i-Azam University, Islamabad, Pakistan
| | - Fawad Ali Shah
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Sajid Rashid
- National Center for Bioinformatics, Quaid-i-Azam University, Islamabad, Pakistan
| |
Collapse
|
18
|
Wang S, Cortes CJ. Interactions with PDZ proteins diversify voltage-gated calcium channel signaling. J Neurosci Res 2020; 99:332-348. [PMID: 32476168 DOI: 10.1002/jnr.24650] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 04/29/2020] [Accepted: 05/07/2020] [Indexed: 11/12/2022]
Abstract
Voltage-gated Ca2+ (CaV ) channels are crucial for neuronal excitability and synaptic transmission upon depolarization. Their properties in vivo are modulated by their interaction with a variety of scaffolding proteins. Such interactions can influence the function and localization of CaV channels, as well as their coupling to intracellular second messengers and regulatory pathways, thus amplifying their signaling potential. Among these scaffolding proteins, a subset of PDZ (postsynaptic density-95, Drosophila discs-large, and zona occludens)-domain containing proteins play diverse roles in modulating CaV channel properties. At the presynaptic terminal, PDZ proteins enrich CaV channels in the active zone, enabling neurotransmitter release by maintaining a tight and vital link between channels and vesicles. In the postsynaptic density, these interactions are essential in regulating dendritic spine morphology and postsynaptic signaling cascades. In this review, we highlight the studies that demonstrate dynamic regulations of neuronal CaV channels by PDZ proteins. We discuss the role of PDZ proteins in controlling channel activity, regulating channel cell surface density, and influencing channel-mediated downstream signaling events. We highlight the importance of PDZ protein regulations of CaV channels and evaluate the link between this regulatory effect and human disease.
Collapse
Affiliation(s)
- Shiyi Wang
- Department of Cell Biology, Duke University, Durham, NC, USA.,Department of Neurology, Duke University, Durham, NC, USA
| | - Constanza J Cortes
- Department of Neurology, Duke University, Durham, NC, USA.,Department of Cell, Developmental and Integrative Biology, University of Alabama Birmingham, Birmingham, AL, USA
| |
Collapse
|
19
|
Dolphin AC, Lee A. Presynaptic calcium channels: specialized control of synaptic neurotransmitter release. Nat Rev Neurosci 2020; 21:213-229. [PMID: 32161339 PMCID: PMC7873717 DOI: 10.1038/s41583-020-0278-2] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2020] [Indexed: 11/09/2022]
Abstract
Chemical synapses are heterogeneous junctions formed between neurons that are specialized for the conversion of electrical impulses into the exocytotic release of neurotransmitters. Voltage-gated Ca2+ channels play a pivotal role in this process as they are the major conduits for the Ca2+ ions that trigger the fusion of neurotransmitter-containing vesicles with the presynaptic membrane. Alterations in the intrinsic function of these channels and their positioning within the active zone can profoundly alter the timing and strength of synaptic output. Advances in optical and electron microscopic imaging, structural biology and molecular techniques have facilitated recent breakthroughs in our understanding of the properties of voltage-gated Ca2+ channels that support their presynaptic functions. Here we examine the nature of these channels, how they are trafficked to and anchored within presynaptic boutons, and the mechanisms that allow them to function optimally in shaping the flow of information through neural circuits.
Collapse
Affiliation(s)
- Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK.
| | - Amy Lee
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
20
|
Ortner NJ, Pinggera A, Hofer NT, Siller A, Brandt N, Raffeiner A, Vilusic K, Lang I, Blum K, Obermair GJ, Stefan E, Engel J, Striessnig J. RBP2 stabilizes slow Cav1.3 Ca 2+ channel inactivation properties of cochlear inner hair cells. Pflugers Arch 2019; 472:3-25. [PMID: 31848688 PMCID: PMC6960213 DOI: 10.1007/s00424-019-02338-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 10/18/2019] [Accepted: 12/04/2019] [Indexed: 01/31/2023]
Abstract
Cav1.3 L-type Ca2+ channels (LTCCs) in cochlear inner hair cells (IHCs) are essential for hearing as they convert sound-induced graded receptor potentials into tonic postsynaptic glutamate release. To enable fast and indefatigable presynaptic Ca2+ signaling, IHC Cav1.3 channels exhibit a negative activation voltage range and uniquely slow inactivation kinetics. Interaction with CaM-like Ca2+-binding proteins inhibits Ca2+-dependent inactivation, while the mechanisms underlying slow voltage-dependent inactivation (VDI) are not completely understood. Here we studied if the complex formation of Cav1.3 LTCCs with the presynaptic active zone proteins RIM2α and RIM-binding protein 2 (RBP2) can stabilize slow VDI. We detected both RIM2α and RBP isoforms in adult mouse IHCs, where they co-localized with Cav1.3 and synaptic ribbons. Using whole-cell patch-clamp recordings (tsA-201 cells), we assessed their effect on the VDI of the C-terminal full-length Cav1.3 (Cav1.3L) and a short splice variant (Cav1.342A) that lacks the C-terminal RBP2 interaction site. When co-expressed with the auxiliary β3 subunit, RIM2α alone (Cav1.342A) or RIM2α/RBP2 (Cav1.3L) reduced Cav1.3 VDI to a similar extent as observed in IHCs. Membrane-anchored β2 variants (β2a, β2e) that inhibit inactivation on their own allowed no further modulation of inactivation kinetics by RIM2α/RBP2. Moreover, association with RIM2α and/or RBP2 consolidated the negative Cav1.3 voltage operating range by shifting the channel's activation threshold toward more hyperpolarized potentials. Taken together, the association with "slow" β subunits (β2a, β2e) or presynaptic scaffolding proteins such as RIM2α and RBP2 stabilizes physiological gating properties of IHC Cav1.3 LTCCs in a splice variant-dependent manner ensuring proper IHC function.
Collapse
Affiliation(s)
- Nadine J Ortner
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria.
| | - Alexandra Pinggera
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Nadja T Hofer
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Anita Siller
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Niels Brandt
- Department of Biophysics and CIPMM, Saarland University, Homburg, Germany
| | - Andrea Raffeiner
- Institute of Biochemistry, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Kristina Vilusic
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Isabelle Lang
- Department of Biophysics and CIPMM, Saarland University, Homburg, Germany
| | - Kerstin Blum
- Department of Biophysics and CIPMM, Saarland University, Homburg, Germany
| | - Gerald J Obermair
- Division of Physiology, Medical University Innsbruck, Innsbruck, Austria.,Division Physiology, Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Eduard Stefan
- Institute of Biochemistry, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Jutta Engel
- Department of Biophysics and CIPMM, Saarland University, Homburg, Germany
| | - Jörg Striessnig
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
21
|
Koohiyan M, Noori-Daloii MR, Hashemzadeh-Chaleshtori M, Salehi M, Abtahi H, Tabatabaiefar MA. A Novel Pathogenic Variant in the CABP2 Gene Causes Severe Nonsyndromic Hearing Loss in a Consanguineous Iranian Family. Audiol Neurootol 2019; 24:258-263. [PMID: 31661684 DOI: 10.1159/000502251] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 07/19/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Hereditary hearing loss (HL) can originate from mutations in one of many genes involved in the complex process of hearing. CABP2 mutations have been reported to cause moderate HL. Here, we report the whole exome sequencing (WES) of a proband presenting with prelingual, severe HL in an Iranian family. METHODS A comprehensive family history was obtained, and clinical evaluations and pedigree analysis were performed in the family with 2 affected members. After excluding mutations in the GJB2 gene and 7 other most common autosomal recessive nonsyndromic HL (ARNSHL) genes via Sanger sequencing and genetic linkage analysis in the family, WES was utilized to find the possible etiology of the disease. RESULTS WES results showed a novel rare variant (c.311G>A) in the CABP2gene.This missense variant in the exon 4 of the CABP2gene meets the criteria of being pathogenic according to the American College of Medical Genetics and Genomics (ACMG) interpretation guidelines. CONCLUSIONS Up to now, 3 mutations have been reported for the CABP2gene to cause moderate ARNSHL in different populations. Our results show that CABP2variantsalso cause severe ARNSHL, adding CABP2to the growing list of genes that exhibit phenotypic heterogeneity. Expanding our understanding of the mutational spectrum of HL genes is an important step in providing the correct clinical molecular interpretation and diagnosis for patients.
Collapse
Affiliation(s)
- Mahbobeh Koohiyan
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.,Cancer Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | | | - Morteza Hashemzadeh-Chaleshtori
- Cellular and Molecular Research Center, Basic Health Research Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mansoor Salehi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamidreza Abtahi
- Department of Otolaryngology, Al-Zahra Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Amin Tabatabaiefar
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran, .,Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Noncommunicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran,
| |
Collapse
|
22
|
Burgoyne RD, Helassa N, McCue HV, Haynes LP. Calcium Sensors in Neuronal Function and Dysfunction. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a035154. [PMID: 30833454 DOI: 10.1101/cshperspect.a035154] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Calcium signaling in neurons as in other cell types can lead to varied changes in cellular function. Neuronal Ca2+ signaling processes have also become adapted to modulate the function of specific pathways over a wide variety of time domains and these can have effects on, for example, axon outgrowth, neuronal survival, and changes in synaptic strength. Ca2+ also plays a key role in synapses as the trigger for fast neurotransmitter release. Given its physiological importance, abnormalities in neuronal Ca2+ signaling potentially underlie many different neurological and neurodegenerative diseases. The mechanisms by which changes in intracellular Ca2+ concentration in neurons can bring about diverse responses is underpinned by the roles of ubiquitous or specialized neuronal Ca2+ sensors. It has been established that synaptotagmins have key functions in neurotransmitter release, and, in addition to calmodulin, other families of EF-hand-containing neuronal Ca2+ sensors, including the neuronal calcium sensor (NCS) and the calcium-binding protein (CaBP) families, play important physiological roles in neuronal Ca2+ signaling. It has become increasingly apparent that these various Ca2+ sensors may also be crucial for aspects of neuronal dysfunction and disease either indirectly or directly as a direct consequence of genetic variation or mutations. An understanding of the molecular basis for the regulation of the targets of the Ca2+ sensors and the physiological roles of each protein in identified neurons may contribute to future approaches to the development of treatments for a variety of human neuronal disorders.
Collapse
Affiliation(s)
- Robert D Burgoyne
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Nordine Helassa
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Hannah V McCue
- Centre for Genomic Research, University of Liverpool, Liverpool, United Kingdom
| | - Lee P Haynes
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
23
|
Intrinsic planar polarity mechanisms influence the position-dependent regulation of synapse properties in inner hair cells. Proc Natl Acad Sci U S A 2019; 116:9084-9093. [PMID: 30975754 DOI: 10.1073/pnas.1818358116] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Encoding the wide range of audible sounds in the mammalian cochlea is collectively achieved by functionally diverse type I spiral ganglion neurons (SGNs) at each tonotopic position. The firing of each SGN is thought to be driven by an individual active zone (AZ) of a given inner hair cell (IHC). These AZs present distinct properties according to their position within the IHC, to some extent forming a gradient between the modiolar and the pillar IHC side. In this study, we investigated whether signaling involved in planar polarity at the apical surface can influence position-dependent AZ properties at the IHC base. Specifically, we tested the role of Gαi proteins and their binding partner LGN/Gpsm2 implicated in cytoskeleton polarization and hair cell (HC) orientation along the epithelial plane. Using high and superresolution immunofluorescence microscopy as well as patch-clamp combined with confocal Ca2+ imaging we analyzed IHCs in which Gαi signaling was blocked by Cre-induced expression of the pertussis toxin catalytic subunit (PTXa). PTXa-expressing IHCs exhibited larger CaV1.3 Ca2+-channel clusters and consequently greater Ca2+ influx at the whole-cell and single-synapse levels, which also showed a hyperpolarized shift of activation. Moreover, PTXa expression collapsed the modiolar-pillar gradients of ribbon size and maximal synaptic Ca2+ influx. Finally, genetic deletion of Gαi3 and LGN/Gpsm2 also disrupted the modiolar-pillar gradient of ribbon size. We propose a role for Gαi proteins and LGN in regulating the position-dependent AZ properties in IHCs and suggest that this signaling pathway contributes to setting up the diverse firing properties of SGNs.
Collapse
|
24
|
Single-Channel Resolution of the Interaction between C-Terminal Ca V1.3 Isoforms and Calmodulin. Biophys J 2019; 116:836-846. [PMID: 30773296 DOI: 10.1016/j.bpj.2019.01.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/05/2019] [Accepted: 01/16/2019] [Indexed: 12/21/2022] Open
Abstract
Voltage-dependent calcium (CaV) 1.3 channels are involved in the control of cellular excitability and pacemaking in neuronal, cardiac, and sensory cells. Various proteins interact with the alternatively spliced channel C-terminus regulating gating of CaV1.3 channels. Binding of a regulatory calcium-binding protein calmodulin (CaM) to the proximal C-terminus leads to the boosting of channel activity and promotes calcium-dependent inactivation (CDI). The C-terminal modulator domain (CTM) of CaV1.3 channels can interfere with the CaM binding, thereby inhibiting channel activity and CDI. Here, we compared single-channel gating behavior of two natural CaV1.3 splice isoforms: the long CaV1.342 with the full-length CTM and the short CaV1.342A with the C-terminus truncated before the CTM. We found that CaM regulation of CaV1.3 channels is dynamic on a minute timescale. We observed that at equilibrium, single CaV1.342 channels occasionally switched from low to high open probability, which perhaps reflects occasional binding of CaM despite the presence of CTM. Similarly, when the amount of the available CaM in the cell was reduced, the short CaV1.342A isoform showed patterns of the low channel activity. CDI also underwent periodic changes with corresponding kinetics in both isoforms. Our results suggest that the competition between CTM and CaM is influenced by calcium, allowing further fine-tuning of CaV1.3 channel activity for particular cellular needs.
Collapse
|
25
|
Pangrsic T, Singer JH, Koschak A. Voltage-Gated Calcium Channels: Key Players in Sensory Coding in the Retina and the Inner Ear. Physiol Rev 2019; 98:2063-2096. [PMID: 30067155 DOI: 10.1152/physrev.00030.2017] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Calcium influx through voltage-gated Ca (CaV) channels is the first step in synaptic transmission. This review concerns CaV channels at ribbon synapses in primary sense organs and their specialization for efficient coding of stimuli in the physical environment. Specifically, we describe molecular, biochemical, and biophysical properties of the CaV channels in sensory receptor cells of the retina, cochlea, and vestibular apparatus, and we consider how such properties might change over the course of development and contribute to synaptic plasticity. We pay particular attention to factors affecting the spatial arrangement of CaV channels at presynaptic, ribbon-type active zones, because the spatial relationship between CaV channels and release sites has been shown to affect synapse function critically in a number of systems. Finally, we review identified synaptopathies affecting sensory systems and arising from dysfunction of L-type, CaV1.3, and CaV1.4 channels or their protein modulatory elements.
Collapse
Affiliation(s)
- Tina Pangrsic
- Synaptic Physiology of Mammalian Vestibular Hair Cells Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen and Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine , Göttingen, Germany ; Department of Biology, University of Maryland , College Park, Maryland ; and Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck , Innsbruck , Austria
| | - Joshua H Singer
- Synaptic Physiology of Mammalian Vestibular Hair Cells Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen and Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine , Göttingen, Germany ; Department of Biology, University of Maryland , College Park, Maryland ; and Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck , Innsbruck , Austria
| | - Alexandra Koschak
- Synaptic Physiology of Mammalian Vestibular Hair Cells Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen and Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine , Göttingen, Germany ; Department of Biology, University of Maryland , College Park, Maryland ; and Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck , Innsbruck , Austria
| |
Collapse
|
26
|
Yang T, Britt JK, Cintrón-Pérez CJ, Vázquez-Rosa E, Tobin KV, Stalker G, Hardie J, Taugher RJ, Wemmie J, Pieper AA, Lee A. Ca 2+-Binding Protein 1 Regulates Hippocampal-dependent Memory and Synaptic Plasticity. Neuroscience 2018; 380:90-102. [PMID: 29660444 DOI: 10.1016/j.neuroscience.2018.04.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 03/16/2018] [Accepted: 04/05/2018] [Indexed: 11/25/2022]
Abstract
Ca2+-binding protein 1 (CaBP1) is a Ca2+-sensing protein similar to calmodulin that potently regulates voltage-gated Ca2+ channels. Unlike calmodulin, however, CaBP1 is mainly expressed in neuronal cell-types and enriched in the hippocampus, where its function is unknown. Here, we investigated the role of CaBP1 in hippocampal-dependent behaviors using mice lacking expression of CaBP1 (C-KO). By western blot, the largest CaBP1 splice variant, caldendrin, was detected in hippocampal lysates from wild-type (WT) but not C-KO mice. Compared to WT mice, C-KO mice exhibited mild deficits in spatial learning and memory in both the Barnes maze and in Morris water maze reversal learning. In contextual but not cued fear-conditioning assays, C-KO mice showed greater freezing responses than WT mice. In addition, the number of adult-born neurons in the hippocampus of C-KO mice was ∼40% of that in WT mice, as measured by bromodeoxyuridine labeling. Moreover, hippocampal long-term potentiation was significantly reduced in C-KO mice. We conclude that CaBP1 is required for cellular mechanisms underlying optimal encoding of hippocampal-dependent spatial and fear-related memories.
Collapse
Affiliation(s)
- Tian Yang
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Neurology, University of Iowa, Iowa City, IA 52242, USA
| | - Jeremiah K Britt
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Psychiatry, University of Iowa, Iowa City, IA 52242, USA
| | - Coral J Cintrón-Pérez
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Psychiatry, University of Iowa, Iowa City, IA 52242, USA
| | - Edwin Vázquez-Rosa
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Psychiatry, University of Iowa, Iowa City, IA 52242, USA
| | - Kevin V Tobin
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Neurology, University of Iowa, Iowa City, IA 52242, USA
| | - Grant Stalker
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Neurology, University of Iowa, Iowa City, IA 52242, USA
| | - Jason Hardie
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Neurology, University of Iowa, Iowa City, IA 52242, USA
| | - Rebecca J Taugher
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Psychiatry, University of Iowa, Iowa City, IA 52242, USA
| | - John Wemmie
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Psychiatry, University of Iowa, Iowa City, IA 52242, USA
| | - Andrew A Pieper
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Neurology, University of Iowa, Iowa City, IA 52242, USA; Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Psychiatry, University of Iowa, Iowa City, IA 52242, USA; Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Free Radical, University of Iowa, Iowa City, IA 52242, USA; Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Radiation Biology Program, University of Iowa, Iowa City, IA 52242, USA; Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Radiation Oncology Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA; Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Veterans Affairs, University of Iowa, Iowa City, IA 52242, USA; Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Pappajohn Biomedical Institute and Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242, USA
| | - Amy Lee
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Neurology, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
27
|
Yang T, Hu N, Pangršič T, Green S, Hansen M, Lee A. Functions of CaBP1 and CaBP2 in the peripheral auditory system. Hear Res 2018; 364:48-58. [PMID: 29661613 DOI: 10.1016/j.heares.2018.04.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 03/13/2018] [Accepted: 04/02/2018] [Indexed: 12/29/2022]
Abstract
CaBPs are a family of Ca2+ binding proteins related to calmodulin. Two CaBP family members, CaBP1 and CaBP2, are highly expressed in the cochlea. Here, we investigated the significance of CaBP1 and CaBP2 for hearing in mice lacking expression of these proteins (CaBP1 KO and CaBP2 KO) using auditory brain responses (ABRs) and distortion product otoacoustic emissions (DPOAEs). In CaBP1 KO mice, ABR wave I was larger in amplitude, and shorter in latency and faster in decay, suggestive of enhanced synchrony of auditory nerve fibers. This interpretation was supported by the greater excitability of CaBP1 KO than WT neurons in whole-cell patch clamp recordings of spiral ganglion neurons in culture, and normal presynaptic function of CaBP1 KO IHCs. DPOAEs and ABR thresholds were normal in 4-week old CaBP1 KO mice, but elevated ABR thresholds became evident at 32 kHz at 9 weeks, and at 8 and 16 kHz by 6 months of age. In contrast, CaBP2 KO mice exhibited significant ABR threshold elevations at 4 weeks of age that became more severe in the mid-frequency range by 9 weeks. Though normal at 4 weeks, DPOAEs in CaBP2 KO mice were significantly reduced in the mid-frequency range by 9 weeks. Our results reveal requirements for CaBP1 and CaBP2 in the peripheral auditory system and highlight the diverse modes by which CaBPs influence sensory processing.
Collapse
Affiliation(s)
- Tian Yang
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA; Department of Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, IA 52242, USA
| | - Ning Hu
- Department of Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, IA 52242, USA; Department of Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Tina Pangršič
- Synaptic Physiology of Mammalian Vestibular Hair Cells Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
| | - Steven Green
- Department of Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, IA 52242, USA; Department of Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Marlan Hansen
- Department of Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, IA 52242, USA; Department of Neurosurgery, University of Iowa, Iowa City, IA 52242, USA
| | - Amy Lee
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA; Department of Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, IA 52242, USA; Department of Neurology, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
28
|
Yang T, Choi JE, Soh D, Tobin K, Joiner ML, Hansen M, Lee A. CaBP1 regulates Ca v1 L-type Ca 2+ channels and their coupling to neurite growth and gene transcription in mouse spiral ganglion neurons. Mol Cell Neurosci 2018; 88:342-352. [PMID: 29548764 DOI: 10.1016/j.mcn.2018.03.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 03/07/2018] [Accepted: 03/12/2018] [Indexed: 12/19/2022] Open
Abstract
CaBP1 is a Ca2+ binding protein that is widely expressed in neurons in the brain, retina, and cochlea. In heterologous expression systems, CaBP1 interacts with and regulates voltage-gated Cav Ca2+ channels but whether this is the case in neurons is unknown. Here, we investigated the cellular functions of CaBP1 in cochlear spiral ganglion neurons (SGNs), which express high levels of CaBP1. Consistent with the role of CaBP1 as a suppressor of Ca2+-dependent inactivation (CDI) of Cav1 (L-type) channels, Cav1 currents underwent greater CDI in SGNs from mice lacking CaBP1 (C-KO) than in wild-type (WT) SGNs. The coupling of Cav1 channels to downstream signaling pathways was also disrupted in C-KO SGNs. Activity-dependent repression of neurite growth was significantly blunted and unresponsive to Cav1 antagonists in C-KO SGNs in contrast to WT SGNs. Moreover, Cav1-mediated Ca2+ signals and phosphorylation of cAMP-response element binding protein were reduced in C-KO SGNs compared to WT SGNs. Our findings establish a role for CaBP1 as an essential regulator of Cav1 channels in SGNs and their coupling to downstream pathways controlling activity-dependent transcription and neurite growth.
Collapse
Affiliation(s)
- Tian Yang
- Departments of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA
| | - Ji-Eun Choi
- Departments of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA
| | - Daniel Soh
- Departments of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA
| | - Kevin Tobin
- Departments of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA
| | - Mei-Ling Joiner
- Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, IA 52242, USA
| | - Marlan Hansen
- Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, IA 52242, USA; Neurosurgery, University of Iowa, Iowa City, IA 52242, USA
| | - Amy Lee
- Departments of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA; Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, IA 52242, USA; Neurology, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
29
|
Compartmentalization of antagonistic Ca 2+ signals in developing cochlear hair cells. Proc Natl Acad Sci U S A 2018; 115:E2095-E2104. [PMID: 29439202 DOI: 10.1073/pnas.1719077115] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
During a critical developmental period, cochlear inner hair cells (IHCs) exhibit sensory-independent activity, featuring action potentials in which Ca2+ ions play a fundamental role in driving both spiking and glutamate release onto synapses with afferent auditory neurons. This spontaneous activity is controlled by a cholinergic input to the IHC, activating a specialized nicotinic receptor with high Ca2+ permeability, and coupled to the activation of hyperpolarizing SK channels. The mechanisms underlying distinct excitatory and inhibitory Ca2+ roles within a small, compact IHC are unknown. Making use of Ca2+ imaging, afferent auditory bouton recordings, and electron microscopy, the present work shows that unusually high intracellular Ca2+ buffering and "subsynaptic" cisterns provide efficient compartmentalization and tight control of cholinergic Ca2+ signals. Thus, synaptic efferent Ca2+ spillover and cross-talk are prevented, and the cholinergic input preserves its inhibitory signature to ensure normal development of the auditory system.
Collapse
|
30
|
Jean P, Lopez de la Morena D, Michanski S, Jaime Tobón LM, Chakrabarti R, Picher MM, Neef J, Jung S, Gültas M, Maxeiner S, Neef A, Wichmann C, Strenzke N, Grabner C, Moser T. The synaptic ribbon is critical for sound encoding at high rates and with temporal precision. eLife 2018; 7:29275. [PMID: 29328020 PMCID: PMC5794258 DOI: 10.7554/elife.29275] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 12/19/2017] [Indexed: 11/30/2022] Open
Abstract
We studied the role of the synaptic ribbon for sound encoding at the synapses between inner hair cells (IHCs) and spiral ganglion neurons (SGNs) in mice lacking RIBEYE (RBEKO/KO). Electron and immunofluorescence microscopy revealed a lack of synaptic ribbons and an assembly of several small active zones (AZs) at each synaptic contact. Spontaneous and sound-evoked firing rates of SGNs and their compound action potential were reduced, indicating impaired transmission at ribbonless IHC-SGN synapses. The temporal precision of sound encoding was impaired and the recovery of SGN-firing from adaptation indicated slowed synaptic vesicle (SV) replenishment. Activation of Ca2+-channels was shifted to more depolarized potentials and exocytosis was reduced for weak depolarizations. Presynaptic Ca2+-signals showed a broader spread, compatible with the altered Ca2+-channel clustering observed by super-resolution immunofluorescence microscopy. We postulate that RIBEYE disruption is partially compensated by multi-AZ organization. The remaining synaptic deficit indicates ribbon function in SV-replenishment and Ca2+-channel regulation. Our sense of hearing relies on our ears quickly and tirelessly processing information in a precise manner. Sounds cause vibrations in a part of the inner ear called the cochlea. Inside the cochlea, the vibrations move hair-like structures on sensory cells that translate these movements into electrical signals. These hair cells are connected to specialized nerve cells that relay the signals to the brain, which then interprets them as sounds. Hair cells communicate with the specialized nerve cells via connections known as chemical synapses. This means that the electrical signals in the hair cell activate channel proteins that allow calcium ions to flow in. This in turn triggers membrane-bound packages called vesicles inside the hair cell to fuse with its surface membrane and release their contents to the outside. The contents, namely chemicals called neurotransmitters, then travels across the space between the cells, relaying the signal to the nerve cell. The junctions between the hair cells and the nerve cells are more specifically known as ribbon synapses. This is because they have a ribbon-like structure that appears to tether a halo of vesicles close to the active zone where neurotransmitters are released. However, the exact role of this synaptic ribbon has remained mysterious despite decades of study. The ribbon is mainly composed of a protein called Ribeye, and now Jean, Lopez de la Morena, Michanski, Jaime Tobón et al. show that mutant mice that lack this protein do not have any ribbons at their “ribbon synapses”. Hair cells without synaptic ribbons are less able to timely and reliably send signals to the nerve cells, most likely because they cannot replenish the vesicles at the synapse quickly enough. Further analysis showed that the synaptic ribbon also helps to regulate the calcium channels at the synapse, which is important for linking the electrical signals in the hair cell to the release of the neurotransmitters. Jean et al. also saw that hair cells without ribbons reorganize their synapses to form multiple active zones that could transfer neurotransmitter to the nerve cells. This could partially compensate for the loss of the ribbons, meaning the impact of their loss may have been underestimated. Future studies could explore this by eliminating the Ribeye protein only after the ribbon synapses are fully formed. These findings may help scientists to better understand deafness and other hearing disorders in humans. They will also be of interest to neuroscientists who research synapses, hearing and other sensory processes.
Collapse
Affiliation(s)
- Philippe Jean
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Collaborative Research Center, University of Göttingen, Göttingen, Germany.,Göttingen Graduate School for Neurosciences and Molecular Biosciences, University of Göttingen, Göttingen, Germany.,InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
| | - David Lopez de la Morena
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Göttingen Graduate School for Neurosciences and Molecular Biosciences, University of Göttingen, Göttingen, Germany.,InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
| | - Susann Michanski
- Collaborative Research Center, University of Göttingen, Göttingen, Germany.,InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany.,Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen, Germany.,Institute for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Lina María Jaime Tobón
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Collaborative Research Center, University of Göttingen, Göttingen, Germany.,Göttingen Graduate School for Neurosciences and Molecular Biosciences, University of Göttingen, Göttingen, Germany.,InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany.,Synaptic Nanophysiology Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.,Auditory Neuroscience Group, Max Planck Institute for Experimental Medicine, Göttingen, Germany
| | - Rituparna Chakrabarti
- Collaborative Research Center, University of Göttingen, Göttingen, Germany.,Göttingen Graduate School for Neurosciences and Molecular Biosciences, University of Göttingen, Göttingen, Germany.,InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany.,Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen, Germany.,Institute for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Maria Magdalena Picher
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Collaborative Research Center, University of Göttingen, Göttingen, Germany.,InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
| | - Jakob Neef
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Collaborative Research Center, University of Göttingen, Göttingen, Germany.,InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany.,Synaptic Nanophysiology Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.,Auditory Neuroscience Group, Max Planck Institute for Experimental Medicine, Göttingen, Germany
| | - SangYong Jung
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany.,Neuro Modulation and Neuro Circuitry Group, Singapore Bioimaging Consortium (SBIC), Biomedical Sciences Institutes, Singapore, Singapore
| | - Mehmet Gültas
- Department of Breeding Informatics, Georg-August-University Göttingen, Göttingen, Germany
| | - Stephan Maxeiner
- Institute for Anatomy and Cell Biology, University of the Saarland, Homburg, Germany
| | - Andreas Neef
- Bernstein Group Biophysics of Neural Computation, Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
| | - Carolin Wichmann
- Collaborative Research Center, University of Göttingen, Göttingen, Germany.,InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany.,Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen, Germany.,Institute for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Nicola Strenzke
- Collaborative Research Center, University of Göttingen, Göttingen, Germany.,InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany.,Auditory Systems Physiology Group, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
| | - Chad Grabner
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany.,Synaptic Nanophysiology Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Collaborative Research Center, University of Göttingen, Göttingen, Germany.,InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany.,Synaptic Nanophysiology Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.,Auditory Neuroscience Group, Max Planck Institute for Experimental Medicine, Göttingen, Germany.,Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
31
|
Different Ca V1.3 Channel Isoforms Control Distinct Components of the Synaptic Vesicle Cycle in Auditory Inner Hair Cells. J Neurosci 2017; 37:2960-2975. [PMID: 28193694 DOI: 10.1523/jneurosci.2374-16.2017] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 01/27/2017] [Accepted: 02/01/2017] [Indexed: 12/16/2022] Open
Abstract
The mechanisms orchestrating transient and sustained exocytosis in auditory inner hair cells (IHCs) remain largely unknown. These exocytotic responses are believed to mobilize sequentially a readily releasable pool of vesicles (RRP) underneath the synaptic ribbons and a slowly releasable pool of vesicles (SRP) at farther distance from them. They are both governed by Cav1.3 channels and require otoferlin as Ca2+ sensor, but whether they use the same Cav1.3 isoforms is still unknown. Using whole-cell patch-clamp recordings in posthearing mice, we show that only a proportion (∼25%) of the total Ca2+ current in IHCs displaying fast inactivation and resistance to 20 μm nifedipine, a l-type Ca2+ channel blocker, is sufficient to trigger RRP but not SRP exocytosis. This Ca2+ current is likely conducted by short C-terminal isoforms of Cav1.3 channels, notably Cav1.342A and Cav1.343S, because their mRNA is highly expressed in wild-type IHCs but poorly expressed in Otof-/- IHCs, the latter having Ca2+ currents with considerably reduced inactivation. Nifedipine-resistant RRP exocytosis was poorly affected by 5 mm intracellular EGTA, suggesting that the Cav1.3 short isoforms are closely associated with the release site at the synaptic ribbons. Conversely, our results suggest that Cav1.3 long isoforms, which carry ∼75% of the total IHC Ca2+ current with slow inactivation and confer high sensitivity to nifedipine and to internal EGTA, are essentially involved in recruiting SRP vesicles. Intracellular Ca2+ imaging showed that Cav1.3 long isoforms support a deep intracellular diffusion of Ca2+SIGNIFICANCE STATEMENT Auditory inner hair cells (IHCs) encode sounds into nerve impulses through fast and indefatigable Ca2+-dependent exocytosis at their ribbon synapses. We show that this synaptic process involves long and short C-terminal isoforms of the Cav1.3 Ca2+ channel that differ in the kinetics of their Ca2+-dependent inactivation and their relative sensitivity to the l-type Ca2+ channel blocker nifedipine. The short C-terminal isoforms, having fast inactivation and low sensitivity to nifedipine, mainly control the fast fusion of the readily releasable pool (RRP); that is, they encode the phasic exocytotic component. The long isoforms, with slow inactivation and great sensitivity to nifedipine, mainly regulate the vesicular replenishment of the RRP; that is, the sustained or tonic exocytosis.
Collapse
|
32
|
Ca 2+-binding protein 2 inhibits Ca 2+-channel inactivation in mouse inner hair cells. Proc Natl Acad Sci U S A 2017; 114:E1717-E1726. [PMID: 28183797 DOI: 10.1073/pnas.1617533114] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Ca2+-binding protein 2 (CaBP2) inhibits the inactivation of heterologously expressed voltage-gated Ca2+ channels of type 1.3 (CaV1.3) and is defective in human autosomal-recessive deafness 93 (DFNB93). Here, we report a newly identified mutation in CABP2 that causes a moderate hearing impairment likely via nonsense-mediated decay of CABP2-mRNA. To study the mechanism of hearing impairment resulting from CABP2 loss of function, we disrupted Cabp2 in mice (Cabp2LacZ/LacZ ). CaBP2 was expressed by cochlear hair cells, preferentially in inner hair cells (IHCs), and was lacking from the postsynaptic spiral ganglion neurons (SGNs). Cabp2LacZ/LacZ mice displayed intact cochlear amplification but impaired auditory brainstem responses. Patch-clamp recordings from Cabp2LacZ/LacZ IHCs revealed enhanced Ca2+-channel inactivation. The voltage dependence of activation and the number of Ca2+ channels appeared normal in Cabp2LacZ/LacZ mice, as were ribbon synapse counts. Recordings from single SGNs showed reduced spontaneous and sound-evoked firing rates. We propose that CaBP2 inhibits CaV1.3 Ca2+-channel inactivation, and thus sustains the availability of CaV1.3 Ca2+ channels for synaptic sound encoding. Therefore, we conclude that human deafness DFNB93 is an auditory synaptopathy.
Collapse
|
33
|
Lack of CaBP1/Caldendrin or CaBP2 Leads to Altered Ganglion Cell Responses. eNeuro 2016; 3:eN-NWR-0099-16. [PMID: 27822497 PMCID: PMC5083949 DOI: 10.1523/eneuro.0099-16.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 09/24/2016] [Accepted: 10/08/2016] [Indexed: 12/01/2022] Open
Abstract
Calcium-binding proteins (CaBPs) form a subfamily of calmodulin-like proteins that were cloned from the retina. CaBP4 and CaBP5 have been shown to be important for normal visual function. Although CaBP1/caldendrin and CaBP2 have been shown to modulate various targets in vitro, it is not known whether they contribute to the transmission of light responses through the retina. Therefore, we generated mice that lack CaBP2 or CaBP1/caldendrin (Cabp2–/– and Cabp1–/–) to test whether these CaBPs are essential for normal retinal function. By immunohistochemistry, the overall morphology of Cabp1–/– and Cabp2–/– retinas and the number of synaptic ribbons appear normal; transmission electron microscopy shows normal tethered ribbon synapses and synaptic vesicles as in wild-type retinas. However, whole-cell patch clamp recordings showed that light responses of retinal ganglion cells of Cabp2–/– and Cabp1–/– mice differ in amplitude and kinetics from those of wild-type mice. We conclude that CaBP1/caldendrin and CaBP2 are not required for normal gross retinal and synapse morphology but are necessary for the proper transmission of light responses through the retina; like other CaBPs, CaBP1/caldendrin and CaBP2 likely act by modulating presynaptic Ca2+-dependent signaling mechanisms.
Collapse
|
34
|
Hair cells use active zones with different voltage dependence of Ca2+ influx to decompose sounds into complementary neural codes. Proc Natl Acad Sci U S A 2016; 113:E4716-25. [PMID: 27462107 DOI: 10.1073/pnas.1605737113] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
For sounds of a given frequency, spiral ganglion neurons (SGNs) with different thresholds and dynamic ranges collectively encode the wide range of audible sound pressures. Heterogeneity of synapses between inner hair cells (IHCs) and SGNs is an attractive candidate mechanism for generating complementary neural codes covering the entire dynamic range. Here, we quantified active zone (AZ) properties as a function of AZ position within mouse IHCs by combining patch clamp and imaging of presynaptic Ca(2+) influx and by immunohistochemistry. We report substantial AZ heterogeneity whereby the voltage of half-maximal activation of Ca(2+) influx ranged over ∼20 mV. Ca(2+) influx at AZs facing away from the ganglion activated at weaker depolarizations. Estimates of AZ size and Ca(2+) channel number were correlated and larger when AZs faced the ganglion. Disruption of the deafness gene GIPC3 in mice shifted the activation of presynaptic Ca(2+) influx to more hyperpolarized potentials and increased the spontaneous SGN discharge. Moreover, Gipc3 disruption enhanced Ca(2+) influx and exocytosis in IHCs, reversed the spatial gradient of maximal Ca(2+) influx in IHCs, and increased the maximal firing rate of SGNs at sound onset. We propose that IHCs diversify Ca(2+) channel properties among AZs and thereby contribute to decomposing auditory information into complementary representations in SGNs.
Collapse
|
35
|
Haeseleer F, Williams B, Lee A. Characterization of C-terminal Splice Variants of Cav1.4 Ca2+ Channels in Human Retina. J Biol Chem 2016; 291:15663-73. [PMID: 27226626 DOI: 10.1074/jbc.m116.731737] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Indexed: 11/06/2022] Open
Abstract
Voltage-gated Ca(2+) channels (Cav) undergo extensive alternative splicing that greatly enhances their functional diversity in excitable cells. Here, we characterized novel splice variants of the cytoplasmic C-terminal domain of Cav1.4 Ca(2+) channels that regulate neurotransmitter release in photoreceptors in the retina. These variants lack a portion of exon 45 and/or the entire exon 47 (Cav1.4Δex p45, Cav1.4Δex 47, Cav1.4Δex p45,47) and are expressed in the retina of primates but not mice. Although the electrophysiological properties of Cav1.4Δex p45 are similar to those of full-length channels (Cav1.4FL), skipping of exon 47 dramatically alters Cav1.4 function. Deletion of exon 47 removes part of a C-terminal automodulatory domain (CTM) previously shown to suppress Ca(2+)-dependent inactivation (CDI) and to cause a positive shift in the voltage dependence of channel activation. Exon 47 is crucial for these effects of the CTM because variants lacking this exon show intense CDI and activate at more hyperpolarized voltages than Cav1.4FL The robust CDI of Cav1.4Δex 47 is suppressed by CaBP4, a regulator of Cav1.4 channels in photoreceptors. Although CaBP4 enhances activation of Cav1.4FL, Cav1.4Δex 47 shows similar voltage-dependent activation in the presence and absence of CaBP4. We conclude that exon 47 encodes structural determinants that regulate CDI and voltage-dependent activation of Cav1.4, and is necessary for modulation of channel activation by CaBP4.
Collapse
Affiliation(s)
- Françoise Haeseleer
- From the Department of Physiology and Biophysics, University of Washington, Seattle, Washington 98195 and
| | - Brittany Williams
- the Departments of Molecular Physiology and Biophysics, Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, Iowa 52242
| | - Amy Lee
- the Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Neurology, and
| |
Collapse
|
36
|
Yang T, Scholl ES, Pan N, Fritzsch B, Haeseleer F, Lee A. Expression and Localization of CaBP Ca2+ Binding Proteins in the Mouse Cochlea. PLoS One 2016; 11:e0147495. [PMID: 26809054 PMCID: PMC4725724 DOI: 10.1371/journal.pone.0147495] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 01/05/2016] [Indexed: 11/19/2022] Open
Abstract
CaBPs are a family of EF-hand Ca2+ binding proteins that are structurally similar to calmodulin. CaBPs can interact with, and yet differentially modulate, effectors that are regulated by calmodulin, such as Cav1 voltage-gated Ca2+ channels. Immunolabeling studies suggest that multiple CaBP family members (CaBP1, 2, 4, and 5) are expressed in the cochlea. To gain insights into the respective auditory functions of these CaBPs, we characterized the expression and cellular localization of CaBPs in the mouse cochlea. By quantitative reverse transcription PCR, we show that CaBP1 and CaBP2 are the major CaBPs expressed in mouse cochlea both before and after hearing onset. Of the three alternatively spliced variants of CaBP1 (caldendrin, CaBP1-L, and CaBP1-S) and CaBP2 (CaBP2-alt, CaBP2-L, CaBP2-S), caldendrin and CaBP2-alt are the most abundant. By in situ hybridization, probes recognizing caldendrin strongly label the spiral ganglion, while probes designed to recognize all three isoforms of CaBP1 weakly label both the inner and outer hair cells as well as the spiral ganglion. Within the spiral ganglion, caldendrin/CaBP1 labeling is associated with cells resembling satellite glial cells. CaBP2-alt is strongly expressed in inner hair cells both before and after hearing onset. Probes designed to recognize all three variants of CaBP2 strongly label inner hair cells before hearing onset and outer hair cells after the onset of hearing. Thus, CaBP1 and CaBP2 may have overlapping roles in regulating Ca2+ signaling in the hair cells, and CaBP1 may have an additional function in the spiral ganglion. Our findings provide a framework for understanding the role of CaBP family members in the auditory periphery.
Collapse
Affiliation(s)
- Tian Yang
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa, United States of America
- Department of Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, Iowa, United States of America
- Department of Neurology, University of Iowa, Iowa City, Iowa, United States of America
| | - Elizabeth S. Scholl
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa, United States of America
- Department of Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, Iowa, United States of America
- Department of Neurology, University of Iowa, Iowa City, Iowa, United States of America
| | - Ning Pan
- Department of Biology, University of Iowa, Iowa City, Iowa, United States of America
| | - Bernd Fritzsch
- Department of Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, Iowa, United States of America
- Department of Biology, University of Iowa, Iowa City, Iowa, United States of America
| | - Françoise Haeseleer
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington, United States of America
| | - Amy Lee
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa, United States of America
- Department of Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, Iowa, United States of America
- Department of Neurology, University of Iowa, Iowa City, Iowa, United States of America
- * E-mail:
| |
Collapse
|
37
|
Zamponi GW, Striessnig J, Koschak A, Dolphin AC. The Physiology, Pathology, and Pharmacology of Voltage-Gated Calcium Channels and Their Future Therapeutic Potential. Pharmacol Rev 2015; 67:821-70. [PMID: 26362469 PMCID: PMC4630564 DOI: 10.1124/pr.114.009654] [Citation(s) in RCA: 748] [Impact Index Per Article: 83.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Voltage-gated calcium channels are required for many key functions in the body. In this review, the different subtypes of voltage-gated calcium channels are described and their physiologic roles and pharmacology are outlined. We describe the current uses of drugs interacting with the different calcium channel subtypes and subunits, as well as specific areas in which there is strong potential for future drug development. Current therapeutic agents include drugs targeting L-type Ca(V)1.2 calcium channels, particularly 1,4-dihydropyridines, which are widely used in the treatment of hypertension. T-type (Ca(V)3) channels are a target of ethosuximide, widely used in absence epilepsy. The auxiliary subunit α2δ-1 is the therapeutic target of the gabapentinoid drugs, which are of value in certain epilepsies and chronic neuropathic pain. The limited use of intrathecal ziconotide, a peptide blocker of N-type (Ca(V)2.2) calcium channels, as a treatment of intractable pain, gives an indication that these channels represent excellent drug targets for various pain conditions. We describe how selectivity for different subtypes of calcium channels (e.g., Ca(V)1.2 and Ca(V)1.3 L-type channels) may be achieved in the future by exploiting differences between channel isoforms in terms of sequence and biophysical properties, variation in splicing in different target tissues, and differences in the properties of the target tissues themselves in terms of membrane potential or firing frequency. Thus, use-dependent blockers of the different isoforms could selectively block calcium channels in particular pathologies, such as nociceptive neurons in pain states or in epileptic brain circuits. Of important future potential are selective Ca(V)1.3 blockers for neuropsychiatric diseases, neuroprotection in Parkinson's disease, and resistant hypertension. In addition, selective or nonselective T-type channel blockers are considered potential therapeutic targets in epilepsy, pain, obesity, sleep, and anxiety. Use-dependent N-type calcium channel blockers are likely to be of therapeutic use in chronic pain conditions. Thus, more selective calcium channel blockers hold promise for therapeutic intervention.
Collapse
Affiliation(s)
- Gerald W Zamponi
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Joerg Striessnig
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Alexandra Koschak
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Annette C Dolphin
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| |
Collapse
|
38
|
Scharinger A, Eckrich S, Vandael DH, Schönig K, Koschak A, Hecker D, Kaur G, Lee A, Sah A, Bartsch D, Benedetti B, Lieb A, Schick B, Singewald N, Sinnegger-Brauns MJ, Carbone E, Engel J, Striessnig J. Cell-type-specific tuning of Cav1.3 Ca(2+)-channels by a C-terminal automodulatory domain. Front Cell Neurosci 2015; 9:309. [PMID: 26379493 PMCID: PMC4547004 DOI: 10.3389/fncel.2015.00309] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 07/27/2015] [Indexed: 11/13/2022] Open
Abstract
Cav1.3 L-type Ca(2+)-channel function is regulated by a C-terminal automodulatory domain (CTM). It affects channel binding of calmodulin and thereby tunes channel activity by interfering with Ca(2+)- and voltage-dependent gating. Alternative splicing generates short C-terminal channel variants lacking the CTM resulting in enhanced Ca(2+)-dependent inactivation and stronger voltage-sensitivity upon heterologous expression. However, the role of this modulatory domain for channel function in its native environment is unkown. To determine its functional significance in vivo, we interrupted the CTM with a hemagglutinin tag in mutant mice (Cav1.3DCRD(HA/HA)). Using these mice we provide biochemical evidence for the existence of long (CTM-containing) and short (CTM-deficient) Cav1.3 α1-subunits in brain. The long (HA-labeled) Cav1.3 isoform was present in all ribbon synapses of cochlear inner hair cells. CTM-elimination impaired Ca(2+)-dependent inactivation of Ca(2+)-currents in hair cells but increased it in chromaffin cells, resulting in hyperpolarized resting potentials and reduced pacemaking. CTM disruption did not affect hearing thresholds. We show that the modulatory function of the CTM is affected by its native environment in different cells and thus occurs in a cell-type specific manner in vivo. It stabilizes gating properties of Cav1.3 channels required for normal electrical excitability.
Collapse
Affiliation(s)
- Anja Scharinger
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck Innsbruck, Austria
| | - Stephanie Eckrich
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine, Saarland University Homburg, Germany
| | - David H Vandael
- Laboratory of Cellular and Molecular Neuroscience, Department of Drug Science, Nanostructured Interfaces and Surfaces Center, University of Torino Torino, Italy
| | - Kai Schönig
- Department of Molecular Biology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University Mannheim, Germany
| | - Alexandra Koschak
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck Innsbruck, Austria
| | - Dietmar Hecker
- Department of Otorhinolaryngology, Saarland University Homburg, Germany
| | - Gurjot Kaur
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck Innsbruck, Austria
| | - Amy Lee
- Department of Molecular Physiology and Biophysics, University of Iowa Iowa City, IA, USA
| | - Anupam Sah
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck Innsbruck, Austria
| | - Dusan Bartsch
- Department of Molecular Biology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University Mannheim, Germany
| | - Bruno Benedetti
- Department of Physiology and Medical Physics, Innsbruck Medical University Innsbruck, Austria
| | - Andreas Lieb
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck Innsbruck, Austria
| | - Bernhard Schick
- Department of Otorhinolaryngology, Saarland University Homburg, Germany
| | - Nicolas Singewald
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck Innsbruck, Austria
| | - Martina J Sinnegger-Brauns
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck Innsbruck, Austria
| | - Emilio Carbone
- Laboratory of Cellular and Molecular Neuroscience, Department of Drug Science, Nanostructured Interfaces and Surfaces Center, University of Torino Torino, Italy
| | - Jutta Engel
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine, Saarland University Homburg, Germany
| | - Jörg Striessnig
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck Innsbruck, Austria
| |
Collapse
|
39
|
Wichmann C. Molecularly and structurally distinct synapses mediate reliable encoding and processing of auditory information. Hear Res 2015; 330:178-90. [PMID: 26188105 DOI: 10.1016/j.heares.2015.07.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 06/21/2015] [Accepted: 07/10/2015] [Indexed: 01/20/2023]
Abstract
Hearing impairment is the most common human sensory deficit. Considering the sophisticated anatomy and physiology of the auditory system, disease-related failures frequently occur. To meet the demands of the neuronal circuits responsible for processing auditory information, the synapses of the lower auditory pathway are anatomically and functionally specialized to process acoustic information indefatigably with utmost temporal precision. Despite sharing some functional properties, the afferent synapses of the cochlea and of auditory brainstem differ greatly in their morphology and employ distinct molecular mechanisms for regulating synaptic vesicle release. Calyceal synapses of the endbulb of Held and the calyx of Held profit from a large number of release sites that project onto one principal cell. Cochlear inner hair cell ribbon synapses exhibit a unique one-to-one relation of the presynaptic active zone to the postsynaptic cell and use hair-cell-specific proteins such as otoferlin for vesicle release. The understanding of the molecular physiology of the hair cell ribbon synapse has been advanced by human genetics studies of sensorineural hearing impairment, revealing human auditory synaptopathy as a new nosological entity.
Collapse
Affiliation(s)
- Carolin Wichmann
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience & InnerEarLab, University Medical Center, Göttingen, Germany.
| |
Collapse
|
40
|
Abstract
Ca2+-dependent inactivation (CDI) is a negative feedback regulation of voltage-gated Cav1 and Cav2 channels that is mediated by the Ca2+ sensing protein, calmodulin (CaM), binding to the pore-forming Cav α1 subunit. David Yue and his colleagues made seminal contributions to our understanding of this process, as well as factors that regulate CDI. Important in this regard are members of a family of Ca2+ binding proteins (CaBPs) that are related to calmodulin. CaBPs are expressed mainly in neural tissues and can antagonize CaM-dependent CDI for Cav1 L-type channels. This review will focus on the roles of CaBPs as Cav1-interacting proteins, and the significance of these interactions for vision, hearing, and neuronal Ca2+ signaling events.
Collapse
Affiliation(s)
- Jason Hardie
- a Departments of Molecular Physiology and Biophysics ; Otolaryngology-Head and Neck Surgery and Neurology; University of Iowa ; Iowa City , IA USA
| | - Amy Lee
- a Departments of Molecular Physiology and Biophysics ; Otolaryngology-Head and Neck Surgery and Neurology; University of Iowa ; Iowa City , IA USA
| |
Collapse
|
41
|
Soong TW, Mori MX. Post-transcriptional modifications and "Calmodulation" of voltage-gated calcium channel function: Reflections by two collaborators of David T Yue. Channels (Austin) 2015; 10:14-9. [PMID: 26054929 DOI: 10.1080/19336950.2015.1051271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
This review article is written to specially pay tribute to David T. Yue who was an outstanding human being and an excellent scientist who exuded passion and creativity. He exemplified an inter-disciplinary scientist who was able to cross scientific boundaries effortlessly in order to provide amazing understanding on how calcium channels work. This article provides a glimpse of some of the research the authors have the privilege to collaborate with David and it attempts to provide the thinking behind some of the research done. In a wider context, we highlight that calcium channel function could be exquisitely modulated by interaction with a tethered calmodulin. Post-transcriptional modifications such as alternative splicing and RNA editing further influence the Ca(2+)-CaM mediated processes such as calcium dependent inhibition and/or facilitation. Besides modifications of electrophysiological and pharmacological properties, protein interactions with the channels could also be influenced in a splice-variant dependent manner.
Collapse
Affiliation(s)
- Tuck Wah Soong
- a Department of Physiology ; Yong Loo Lin School of Medicine; National University of Singapore ; Singapore.,b NUS Graduate School for Integrative Science and Engineering, and Neurobiology/Aging Program ; Singapore.,c National Neuroscience Institute ; Singapore
| | - Masayuki X Mori
- d Kyoto University Department of Synthetic Chemistry and Biological Chemistry ; Graduate School of Engineering, Kyoto University ; Kyoto , Japan
| |
Collapse
|
42
|
Nishio SY, Hattori M, Moteki H, Tsukada K, Miyagawa M, Naito T, Yoshimura H, Iwasa YI, Mori K, Shima Y, Sakuma N, Usami SI. Gene expression profiles of the cochlea and vestibular endorgans: localization and function of genes causing deafness. Ann Otol Rhinol Laryngol 2015; 124 Suppl 1:6S-48S. [PMID: 25814645 DOI: 10.1177/0003489415575549] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
OBJECTIVES We sought to elucidate the gene expression profiles of the causative genes as well as the localization of the encoded proteins involved in hereditary hearing loss. METHODS Relevant articles (as of September 2014) were searched in PubMed databases, and the gene symbols of the genes reported to be associated with deafness were located on the Hereditary Hearing Loss Homepage using localization, expression, and distribution as keywords. RESULTS Our review of the literature allowed us to systematize the gene expression profiles for genetic deafness in the inner ear, clarifying the unique functions and specific expression patterns of these genes in the cochlea and vestibular endorgans. CONCLUSIONS The coordinated actions of various encoded molecules are essential for the normal development and maintenance of auditory and vestibular function.
Collapse
Affiliation(s)
- Shin-Ya Nishio
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan Department of Hearing Implant Sciences, Shinshu University School of Medicine, Matsumoto, Japan
| | - Mitsuru Hattori
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Hideaki Moteki
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Keita Tsukada
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Maiko Miyagawa
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan Department of Hearing Implant Sciences, Shinshu University School of Medicine, Matsumoto, Japan
| | - Takehiko Naito
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Hidekane Yoshimura
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yoh-Ichiro Iwasa
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Kentaro Mori
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yutaka Shima
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Naoko Sakuma
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan Department of Otorhinolaryngology and Head and Neck Surgery, Yokohama City University School of Medicine, Yokohama, Japan
| | - Shin-Ichi Usami
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan Department of Hearing Implant Sciences, Shinshu University School of Medicine, Matsumoto, Japan
| |
Collapse
|
43
|
Abstract
Voltage- and ligand-gated ion channels form the molecular basis of cellular excitability. With >400 members and accounting for ∼1.5% of the human genome, ion channels are some of the most well studied of all proteins in heterologous expression systems. Yet, ion channels often exhibit unexpected properties in vivo because of their interaction with a variety of signaling/scaffolding proteins. Such interactions can influence the function and localization of ion channels, as well as their coupling to intracellular second messengers and pathways, thus increasing the signaling potential of these ion channels in neurons. Moreover, functions have been ascribed to ion channels that are largely independent of their ion-conducting roles. Molecular and functional dissection of the ion channel proteome/interactome has yielded new insights into the composition of ion channel complexes and how their dysregulation leads to human disease.
Collapse
|
44
|
Lee A, Wang S, Williams B, Hagen J, Scheetz TE, Haeseleer F. Characterization of Cav1.4 complexes (α11.4, β2, and α2δ4) in HEK293T cells and in the retina. J Biol Chem 2014; 290:1505-21. [PMID: 25468907 DOI: 10.1074/jbc.m114.607465] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
In photoreceptor synaptic terminals, voltage-gated Cav1.4 channels mediate Ca(2+) signals required for transmission of visual stimuli. Like other high voltage-activated Cav channels, Cav1.4 channels are composed of a main pore-forming Cav1.4 α1 subunit and auxiliary β and α2δ subunits. Of the four distinct classes of β and α2δ, β2 and α2δ4 are thought to co-assemble with Cav1.4 α1 subunits in photoreceptors. However, an understanding of the functional properties of this combination of Cav subunits is lacking. Here, we provide evidence that Cav1.4 α1, β2, and α2δ4 contribute to Cav1.4 channel complexes in the retina and describe their properties in electrophysiological recordings. In addition, we identified a variant of β2, named here β2X13, which, along with β2a, is present in photoreceptor terminals. Cav1.4 α1, β2, and α2δ4 were coimmunoprecipitated from lysates of transfected HEK293 cells and mouse retina and were found to interact in the outer plexiform layer of the retina containing the photoreceptor synaptic terminals, by proximity ligation assays. In whole-cell patch clamp recordings of transfected HEK293T cells, channels (Cav1.4 α1 + β2X13) containing α2δ4 exhibited weaker voltage-dependent activation than those with α2δ1. Moreover, compared with channels (Cav1.4 α1 + α2δ4) with β2a, β2X13-containing channels exhibited greater voltage-dependent inactivation. The latter effect was specific to Cav1.4 because it was not seen for Cav1.2 channels. Our results provide the first detailed functional analysis of the Cav1.4 subunits that form native photoreceptor Cav1.4 channels and indicate potential heterogeneity in these channels conferred by β2a and β2X13 variants.
Collapse
Affiliation(s)
- Amy Lee
- From the Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, and Neurology, University of Iowa, Iowa City, Iowa 52242
| | - Shiyi Wang
- From the Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, and Neurology, University of Iowa, Iowa City, Iowa 52242
| | - Brittany Williams
- From the Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, and Neurology, University of Iowa, Iowa City, Iowa 52242
| | - Jussara Hagen
- From the Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, and Neurology, University of Iowa, Iowa City, Iowa 52242
| | - Todd E Scheetz
- the Departments of Ophthalmology and Visual Sciences and Biomedical Engineering, University of Iowa, Iowa City, Iowa 52242, and
| | - Françoise Haeseleer
- the Department of Physiology and Biophysics, University of Washington, Seattle, Washington 98195
| |
Collapse
|
45
|
Burgoyne RD, Haynes LP. Sense and specificity in neuronal calcium signalling. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1853:1921-32. [PMID: 25447549 PMCID: PMC4728190 DOI: 10.1016/j.bbamcr.2014.10.029] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 10/25/2014] [Accepted: 10/29/2014] [Indexed: 11/02/2022]
Abstract
Changes in the intracellular free calcium concentration ([Ca²⁺]i) in neurons regulate many and varied aspects of neuronal function over time scales from microseconds to days. The mystery is how a single signalling ion can lead to such diverse and specific changes in cell function. This is partly due to aspects of the Ca²⁺ signal itself, including its magnitude, duration, localisation and persistent or oscillatory nature. The transduction of the Ca²⁺ signal requires Ca²⁺binding to various Ca²⁺ sensor proteins. The different properties of these sensors are important for differential signal processing and determine the physiological specificity of Ca(2+) signalling pathways. A major factor underlying the specific roles of particular Ca²⁺ sensor proteins is the nature of their interaction with target proteins and how this mediates unique patterns of regulation. We review here recent progress from structural analyses and from functional analyses in model organisms that have begun to reveal the rules that underlie Ca²⁺ sensor protein specificity for target interaction. We discuss three case studies exemplifying different aspects of Ca²⁺ sensor/target interaction. This article is part of a special issue titled the 13th European Symposium on Calcium.
Collapse
Affiliation(s)
- Robert D Burgoyne
- Department of Cellular and Molecular Physiology, The Physiological Laboratory, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool, L69 3BX, United Kingdom.
| | - Lee P Haynes
- Department of Cellular and Molecular Physiology, The Physiological Laboratory, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool, L69 3BX, United Kingdom
| |
Collapse
|
46
|
ATP hydrolysis is critically required for function of CaV1.3 channels in cochlear inner hair cells via fueling Ca2+ clearance. J Neurosci 2014; 34:6843-8. [PMID: 24828638 DOI: 10.1523/jneurosci.4990-13.2014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Sound encoding is mediated by Ca(2+) influx-evoked release of glutamate at the ribbon synapse of inner hair cells. Here we studied the role of ATP in this process focusing on Ca(2+) current through CaV1.3 channels and Ca(2+) homeostasis in mouse inner hair cells. Patch-clamp recordings and Ca(2+) imaging demonstrate that hydrolyzable ATP is essential to maintain synaptic Ca(2+) influx in inner hair cells via fueling Ca(2+)-ATPases to avoid an increase in cytosolic [Ca(2+)] and subsequent Ca(2+)/calmodulin-dependent inactivation of CaV1.3 channels.
Collapse
|
47
|
Abstract
Synaptic vesicle recycling sustains high rates of neurotransmission at the ribbon-type active zones (AZs) of mouse auditory inner hair cells (IHCs), but its modes and molecular regulation are poorly understood. Electron microscopy indicated the presence of clathrin-mediated endocytosis (CME) and bulk endocytosis. The endocytic proteins dynamin, clathrin, and amphiphysin are expressed and broadly distributed in IHCs. We used confocal vglut1-pHluorin imaging and membrane capacitance (Cm) measurements to study the spatial organization and dynamics of IHC exocytosis and endocytosis. Viral gene transfer expressed vglut1-pHluorin in IHCs and targeted it to synaptic vesicles. The intravesicular pH was ∼6.5, supporting only a modest increase of vglut1-pHluorin fluorescence during exocytosis and pH neutralization. Ca(2+) influx triggered an exocytic increase of vglut1-pHluorin fluorescence at the AZs, around which it remained for several seconds. The endocytic Cm decline proceeded with constant rate (linear component) after exocytosis of the readily releasable pool (RRP). When exocytosis exceeded three to four RRP equivalents, IHCs additionally recruited a faster Cm decline (exponential component) that increased with the amount of preceding exocytosis and likely reflects bulk endocytosis. The dynamin inhibitor Dyngo-4a and the clathrin blocker pitstop 2 selectively impaired the linear component of endocytic Cm decline. A missense mutation of dynamin 1 (fitful) inhibited endocytosis to a similar extent as Dyngo-4a. We propose that IHCs use dynamin-dependent endocytosis via CME to support vesicle cycling during mild stimulation but recruit bulk endocytosis to balance massive exocytosis.
Collapse
|
48
|
Kim KY, Scholl ES, Liu X, Shepherd A, Haeseleer F, Lee A. Localization and expression of CaBP1/caldendrin in the mouse brain. Neuroscience 2014; 268:33-47. [PMID: 24631676 DOI: 10.1016/j.neuroscience.2014.02.052] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 02/26/2014] [Accepted: 02/27/2014] [Indexed: 12/31/2022]
Abstract
Ca(2+) binding protein 1 (CaBP1) and caldendrin are alternatively spliced variants of a subfamily of CaBPs with high homology to calmodulin. Although CaBP1 and caldendrin regulate effectors including plasma membrane and intracellular Ca(2+) channels in heterologous expression systems, little is known about their functions in vivo. Therefore, we generated mice deficient in CaBP1/caldendrin expression (C-KO) and analyzed the expression and cellular localization of CaBP1 and caldendrin in the mouse brain. Immunoperoxidase labeling with antibodies recognizing both CaBP1 and caldendrin was absent in the brain of C-KO mice, but was intense in multiple brain regions of wild-type mice. By Western blot, the antibodies detected two proteins that were absent in the C-KO mouse and consistent in size with caldendrin variants originating from alternative translation initiation sites. By quantitative PCR, caldendrin transcript levels were far greater than those for CaBP1, particularly in the cerebral cortex and hippocampus. In the frontal cortex but not in the hippocampus, caldendrin expression increased steadily from birth. By double-label immunofluorescence, CaBP1/caldendrin was localized in principal neurons and parvalbumin-positive interneurons. In the cerebellum, CaBP1/caldendrin antibodies labeled interneurons in the molecular layer and in basket cell terminals surrounding the soma and axon initial segment of Purkinje neurons, but immunolabeling was absent in Purkinje neurons. We conclude that CaBP1/caldendrin is localized both pre- and postsynaptically where it may regulate Ca(2+) signaling and excitability in select groups of excitatory and inhibitory neurons.
Collapse
Affiliation(s)
- K Y Kim
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA; Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, IA 52242, USA; Department of Neurology, University of Iowa, Iowa City, IA 52242, USA
| | - E S Scholl
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA; Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, IA 52242, USA; Department of Neurology, University of Iowa, Iowa City, IA 52242, USA
| | - X Liu
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA; Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, IA 52242, USA; Department of Neurology, University of Iowa, Iowa City, IA 52242, USA
| | - A Shepherd
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - F Haeseleer
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA
| | - A Lee
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA; Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, IA 52242, USA; Department of Neurology, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
49
|
Abstract
OBJECTIVE To review new insights into the pathophysiology of sensorineural hearing impairment. Specifically, we address defects of the ribbon synapses between inner hair cells and spiral ganglion neurons that cause auditory synaptopathy. DATA SOURCES AND STUDY SELECTION Here, we review original publications on the genetics, animal models, and molecular mechanisms of hair cell ribbon synapses and their dysfunction. CONCLUSION Hair cell ribbon synapses are highly specialized to enable indefatigable sound encoding with utmost temporal precision. Their dysfunctions, which we term auditory synaptopathies, impair audibility of sounds to varying degrees but commonly affect neural encoding of acoustic temporal cues essential for speech comprehension. Clinical features of auditory synaptopathies are similar to those accompanying auditory neuropathy, a group of genetic and acquired disorders of spiral ganglion neurons. Genetic auditory synaptopathies include alterations of glutamate loading of synaptic vesicles, synaptic Ca influx or synaptic vesicle turnover. Acquired synaptopathies include noise-induced hearing loss because of excitotoxic synaptic damage and subsequent gradual neural degeneration. Alterations of ribbon synapses likely also contribute to age-related hearing loss.
Collapse
|
50
|
Allostery in Ca²⁺ channel modulation by calcium-binding proteins. Nat Chem Biol 2014; 10:231-8. [PMID: 24441587 DOI: 10.1038/nchembio.1436] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 11/25/2013] [Indexed: 12/17/2022]
Abstract
Distinguishing between allostery and competition among modulating ligands is challenging for large target molecules. Out of practical necessity, inferences are often drawn from in vitro assays on target fragments, but such inferences may belie actual mechanisms. One key example of such ambiguity concerns calcium-binding proteins (CaBPs) that tune signaling molecules regulated by calmodulin (CaM). As CaBPs resemble CaM, CaBPs are believed to competitively replace CaM on targets. Yet, brain CaM expression far surpasses that of CaBPs, raising questions as to whether CaBPs can exert appreciable biological actions. Here, we devise a live-cell, holomolecule approach that reveals an allosteric mechanism for calcium channels whose CaM-mediated inactivation is eliminated by CaBP4. Our strategy is to covalently link CaM and/or CaBP to holochannels, enabling live-cell fluorescence resonance energy transfer assays to resolve a cyclical allosteric binding scheme for CaM and CaBP4 to channels, thus explaining how trace CaBPs prevail. This approach may apply generally for discerning allostery in live cells.
Collapse
|