1
|
Choi JS, Ahn YJ, Lee S, Park DJ, Park J, Ha SM, Seo YJ. Role of Kir4.1 Channels in Aminoglycoside-Induced Ototoxicity of Hair Cells. BIOMED RESEARCH INTERNATIONAL 2023; 2023:4191999. [PMID: 38143588 PMCID: PMC10748730 DOI: 10.1155/2023/4191999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/27/2023] [Accepted: 11/14/2023] [Indexed: 12/26/2023]
Abstract
The Kir4.1 channel, an inwardly rectifying potassium ion (K+) channel, is located in the hair cells of the organ of Corti as well as the intermediate cells of the stria vascularis. The Kir4.1 channel has a crucial role in the generation of endolymphatic potential and maintenance of the resting membrane potential. However, the role and functions of the Kir4.1 channel in the progenitor remain undescribed. To observe the role of Kir4.1 in the progenitor treated with the one-shot ototoxic drugs (kanamycin and furosemide), we set the proper condition in culturing Immortomouse-derived HEI-OC1 cells to express the potassium-related channels well. And also, that was reproduced in mice experiments to show the important role of Kir4.1 in the survival of hair cells after treating the ototoxicity drugs. In our results, when kanamycin and furosemide drugs were cotreated with HEI-OC1 cells, the Kir4.1 channel did not change, but the expression levels of the NKCC1 cotransporter and KCNQ4 channel are decreased. This shows that inward and outward channels were blocked by the two drugs (kanamycin and furosemide). However, noteworthy here is that the expression level of Kir4.1 channel increased when kanamycin was treated alone. This shows that Kir4.1, an inwardly rectifying potassium channel, acts as an outward channel in place of the corresponding channel when the KCNQ4 channel, an outward channel, is blocked. These results suggest that the Kir4.1 channel has a role in maintaining K+ homeostasis in supporting cells, with K+ concentration compensator when the NKCC1 cotransporter and Kv7.4 (KCNQ4) channels are deficient.
Collapse
Affiliation(s)
- Jin Sil Choi
- Research Institute of Hearing Enhancement, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
- Department of Otorhinolaryngology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Ye Ji Ahn
- Research Institute of Hearing Enhancement, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
- Department of Otorhinolaryngology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - SuHoon Lee
- Research Institute of Hearing Enhancement, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
- Department of Otorhinolaryngology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Dong Jun Park
- Research Institute of Hearing Enhancement, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
- Department of Otorhinolaryngology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - JeongEun Park
- Research Institute of Hearing Enhancement, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
- Department of Otorhinolaryngology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Sun Mok Ha
- Research Institute of Hearing Enhancement, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
- Department of Otorhinolaryngology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Young Joon Seo
- Research Institute of Hearing Enhancement, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
- Department of Otorhinolaryngology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| |
Collapse
|
2
|
Jang MW, Lim J, Park MG, Lee JH, Lee CJ. Active role of glia-like supporting cells in the organ of Corti: Membrane proteins and their roles in hearing. Glia 2022; 70:1799-1825. [PMID: 35713516 DOI: 10.1002/glia.24229] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/23/2022] [Accepted: 05/30/2022] [Indexed: 12/13/2022]
Abstract
The organ of Corti, located in the cochlea in the inner ear, is one of the major sensory organs involved in hearing. The organ of Corti consists of hair cells, glia-like supporting cells, and the cochlear nerve, which work in harmony to receive sound from the outer ear and transmit auditory signals to the cochlear nucleus in the auditory ascending pathway. In this process, maintenance of the endocochlear potential, with a high potassium gradient and clearance of electrolytes and biochemicals in the inner ear, is critical for normal sound transduction. There is an emerging need for a thorough understanding of each cell type involved in this process to understand the sophisticated mechanisms of the organ of Corti. Hair cells have long been thought to be active, playing a primary role in the cochlea in actively detecting and transmitting signals. In contrast, supporting cells are thought to be silent and function to support hair cells. However, growing lines of evidence regarding the membrane proteins that mediate ionic movement in supporting cells have demonstrated that supporting cells are not silent, but actively play important roles in normal signal transduction. In this review, we summarize studies that characterize diverse membrane proteins according to the supporting cell subtypes involved in cochlear physiology and hearing. This review contributes to a better understanding of supporting cell functions and facilitates the development of potential therapeutic tools for hearing loss.
Collapse
Affiliation(s)
- Minwoo Wendy Jang
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.,Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Jiwoon Lim
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea.,IBS School, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Mingu Gordon Park
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.,Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Jae-Hun Lee
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - C Justin Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.,Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea.,IBS School, University of Science and Technology (UST), Daejeon, Republic of Korea
| |
Collapse
|
3
|
The Interplay of Cx26, Cx32, Cx37, Cx40, Cx43, Cx45, and Panx1 in Inner-Ear Development of Yotari (dab1−/−) Mice and Humans. Biomedicines 2022; 10:biomedicines10030589. [PMID: 35327391 PMCID: PMC8945117 DOI: 10.3390/biomedicines10030589] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 01/01/2023] Open
Abstract
We investigated DAB1-protein deficiency in the inner-ear development of yotari in comparison to humans and wild-type (wt) mice by immunofluorescence for the expression of connexins (Cxs) and the pannexin Panx1. The spatial and temporal dynamics of Cx26, Cx32, Cx37, Cx40, Cx43, Cx45, and Panx1 were determined in the sixth and eighth weeks of human development and at the corresponding mouse embryonic E13.5 and E15.5, in order to examine gap junction intercellular communication (GJIC) and hemichannel formation. The quantification of the area percentage covered by positive signal was performed for the epithelium and mesenchyme of the cochlear and semicircular ducts and is expressed as the mean ± SD. The data were analysed by one-way ANOVA. Almost all of the examined Cxs were significantly decreased in the cochlear and semicircular ducts of yotari compared to wt and humans, except for Cx32, which was significantly higher in yotari. Cx40 dominated in human inner-ear development, while yotari and wt had decreased expression. The Panx1 expression in yotari was significantly lower than that in the wt and human inner ear, except at E13.5 in the mesenchyme of the wt and epithelium and mesenchyme of humans. Our results emphasize the relevance of GJIC during the development of vestibular and cochlear functions, where they can serve as potential therapeutic targets in inner-ear impairments.
Collapse
|
4
|
Sanchez-Pupo RE, O'Donnell BL, Johnston D, Gyenis L, Litchfield DW, Penuela S. Pannexin 2 is expressed in murine skin and promotes UVB-induced apoptosis of keratinocytes. Mol Biol Cell 2022; 33:ar24. [PMID: 34985913 PMCID: PMC9250380 DOI: 10.1091/mbc.e21-08-0387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Pannexins (PANX) are a family of three channel-forming membrane glycoproteins expressed in the skin. Previous studies have focused on the role of PANX1 and PANX3 in the regulation of cellular functions in skin cells while PANX2, the largest member of this protein family, has not been investigated. In the current study, we explored the temporal PANX2 expression in murine skin and found that one Panx2 splice variant (Panx2-202) tends to be more abundant at the protein level and is continuously expressed in developed skin. PANX2 was detected in the suprabasal layers of the mouse epidermis and up-regulated in an in vitro model of rat epidermal keratinocyte differentiation. Furthermore, we show that in apoptotic rat keratinocytes, upon UV light B (UVB)-induced caspase-3/7 activation, ectopically overexpressed PANX2 is cleaved in its C-terminal domain at the D416 residue without increasing the apoptotic rate measured by caspase-3/7 activation. Notably, CRISPR-Cas9 mediated genetic deletion of rat Panx2 delays but does not impair caspase-3/7 activation and cytotoxicity in UVB-irradiated keratinocytes. We propose that endogenous PANX2 expression in keratinocytes promotes cell death after UVB insult and may contribute to skin homeostasis.
Collapse
Affiliation(s)
- Rafael E Sanchez-Pupo
- Department of Anatomy and Cell Biology, Division of Experimental Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Brooke L O'Donnell
- Department of Anatomy and Cell Biology, Division of Experimental Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Danielle Johnston
- Department of Anatomy and Cell Biology, Division of Experimental Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Laszlo Gyenis
- Department of Biochemistry, Division of Experimental Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - David W Litchfield
- Department of Biochemistry, Division of Experimental Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario N6A 5C1, Canada.,Department of Oncology, Division of Experimental Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, Division of Experimental Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario N6A 5C1, Canada.,Department of Oncology, Division of Experimental Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario N6A 5C1, Canada
| |
Collapse
|
5
|
Larrañaga-Vera A, Marco-Bonilla M, Largo R, Herrero-Beaumont G, Mediero A, Cronstein B. ATP transporters in the joints. Purinergic Signal 2021; 17:591-605. [PMID: 34392490 PMCID: PMC8677878 DOI: 10.1007/s11302-021-09810-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/09/2021] [Indexed: 02/08/2023] Open
Abstract
Extracellular adenosine triphosphate (ATP) plays a central role in a wide variety of joint diseases. ATP is generated intracellularly, and the concentration of the extracellular ATP pool is determined by the regulation of its transport out of the cell. A variety of ATP transporters have been described, with connexins and pannexins the most commonly cited. Both form intercellular channels, known as gap junctions, that facilitate the transport of various small molecules between cells and mediate cell-cell communication. Connexins and pannexins also form pores, or hemichannels, that are permeable to certain molecules, including ATP. All joint tissues express one or more connexins and pannexins, and their expression is altered in some pathological conditions, such as osteoarthritis (OA) and rheumatoid arthritis (RA), indicating that they may be involved in the onset and progression of these pathologies. The aging of the global population, along with increases in the prevalence of obesity and metabolic dysfunction, is associated with a rising frequency of joint diseases along with the increased costs and burden of related illness. The modulation of connexins and pannexins represents an attractive therapeutic target in joint disease, but their complex regulation, their combination of gap-junction-dependent and -independent functions, and their interplay between gap junction and hemichannel formation are not yet fully elucidated. In this review, we try to shed light on the regulation of these proteins and their roles in ATP transport to the extracellular space in the context of joint disease, and specifically OA and RA.
Collapse
Affiliation(s)
- Ane Larrañaga-Vera
- Department of Medicine, Division of Translational Medicine, NYU Langone Health, New York, NY, USA
| | - Miguel Marco-Bonilla
- Bone and Joint Research Unit, IIS-Fundación Jiménez Díaz UAM, 28040, Madrid, Spain
| | - Raquel Largo
- Bone and Joint Research Unit, IIS-Fundación Jiménez Díaz UAM, 28040, Madrid, Spain
| | | | - Aránzazu Mediero
- Bone and Joint Research Unit, IIS-Fundación Jiménez Díaz UAM, 28040, Madrid, Spain.
| | - Bruce Cronstein
- Department of Medicine, Division of Translational Medicine, NYU Langone Health, New York, NY, USA
| |
Collapse
|
6
|
Grimmer B, Krauszman A, Hu X, Kabir G, Connelly KA, Li M, Grune J, Madry C, Isakson BE, Kuebler WM. Pannexin 1-a novel regulator of acute hypoxic pulmonary vasoconstriction. Cardiovasc Res 2021; 118:2535-2547. [PMID: 34668529 DOI: 10.1093/cvr/cvab326] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 09/08/2021] [Indexed: 12/16/2022] Open
Abstract
AIMS Hypoxic pulmonary vasoconstriction (HPV) is a physiological response to alveolar hypoxia that diverts blood flow from poorly ventilated to better aerated lung areas to optimize ventilation-perfusion matching. Yet, the exact sensory and signaling mechanisms by which hypoxia triggers pulmonary vasoconstriction remain incompletely understood. Recently, ATP release via pannexin 1 (Panx1) and subsequent signaling via purinergic P2Y receptors has been identified as regulator of vasoconstriction in systemic arterioles. Here, we probed for the role of Panx1-mediated ATP release in HPV and chronic hypoxic pulmonary hypertension (PH). METHODS AND RESULTS Pharmacological inhibition of Panx1 by probenecid, spironolactone, the Panx1 specific inhibitory peptide (10Panx1) and genetic deletion of Panx1 specifically in smooth muscle attenuated HPV in isolated perfused mouse lungs. In pulmonary artery smooth muscle cells (PASMC), both spironolactone and 10Panx1 attenuated the increase in intracellular Ca2+ concentration ([Ca2+]i) in response to hypoxia. Yet, genetic deletion of Panx1 in either endothelial or smooth muscle cells did not prevent the development of PH in mice. Unexpectedly, ATP release in response to hypoxia was not detectable in PASMC, and inhibition of purinergic receptors or ATP degradation by ATPase failed to attenuate HPV. Rather, transient receptor potential vanilloid 4 (TRPV4) antagonism and Panx1 inhibition inhibited the hypoxia-induced [Ca2+]i increase in PASMC in an additive manner, suggesting that Panx1 regulates [Ca2+]i independently of the ATP-P2Y-TRPV4 pathway. In line with this notion, Panx1 overexpression increased the [Ca2+]i response to hypoxia in HeLa cells. CONCLUSION In the present study we identify Panx1 as novel regulator of HPV. Yet, the role of Panx1 in HPV was not attributable to ATP release and downstream signaling via P2Y receptors or TRPV4 activation, but relates to a role of Panx1 as direct or indirect modulator of the PASMC Ca2+ response to hypoxia. Panx1 did not affect the development of chronic hypoxic PH. TRANSLATIONAL PERSPECTIVE Hypoxic pulmonary vasoconstriction (HPV) optimizes lung ventilation-perfusion matching, but also contributes to pulmonary pathologies including high altitude pulmonary edema (HAPE) or chronic hypoxic pulmonary hypertension. Here, we demonstrate that pharmaceutical inhibition as well as genetic deletion of the hemichannel pannexin-1 (Panx1) in pulmonary artery smooth muscle cells attenuates the physiological HPV response. Panx1 deficiency did, however, not prevent the development of chronic hypoxic pulmonary hypertension in mice. Panx1 inhibitors such as the mineralocorticoid receptor antagonist spironolactone may thus present a putative strategy for the prevention or treatment of HAPE, yet not for chronic hypoxic lung disease.
Collapse
Affiliation(s)
- Benjamin Grimmer
- Institute of Physiology, Charité-Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK)
| | - Adrienn Krauszman
- Institute of Physiology, Charité-Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada
| | - Xudong Hu
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada
| | - Golam Kabir
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada
| | - Kim A Connelly
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada
| | - Mei Li
- Institute of Physiology, Charité-Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Jana Grune
- Institute of Physiology, Charité-Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Christian Madry
- Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Brant E Isakson
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia, USA.,Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité-Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK).,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada.,Departments of Physiology and Surgery, University of Toronto, ON, Canada
| |
Collapse
|
7
|
Mei L, Liu LM, Chen K, Zhao HB. Early Functional and Cognitive Declines Measured by Auditory-Evoked Cortical Potentials in Mice With Alzheimer's Disease. Front Aging Neurosci 2021; 13:710317. [PMID: 34588972 PMCID: PMC8473830 DOI: 10.3389/fnagi.2021.710317] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 08/09/2021] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s disease (AD) is characterized by a progressive loss of memory and cognitive decline. However, the assessment of AD-associated functional and cognitive changes is still a big challenge. Auditory-evoked cortical potential (AECP) is an event-related potential reflecting not only neural activation in the auditory cortex (AC) but also cognitive activity in the brain. In this study, we used the subdermal needle electrodes with the same electrode setting as the auditory brainstem response (ABR) recording and recorded AECP in normal aging CBA/CaJ mice and APP/PS1 AD mice. AECP in mice usually appeared as three positive peaks, i.e., P1, P2, and P3, and three corresponding negative peaks, i.e., N1, N2, and N3. In normal aging CBA mice, the early sensory peaks P1, N1, and P2 were reduced as age increased, whereas the later cognitive peaks N2, P3, and N3 were increased or had no changes with aging. Moreover, the latency of the P1 peak was increased as age increased, although the latencies of later peaks had a significant reduction with aging. In AD mice, peak P1 was significantly reduced in comparison with wild-type (WT) littermates at young ages, proceeding AD phenotype presentation. In particular, the later cognitive peak P3 was diminished after 3 months old, different from the normal aging effect. However, the latencies of AECP peaks in AD mice generally had no significant delay or changes with aging. Finally, consistent with AECP changes, the accumulation of amyloid precursor protein (APP) at the AC was visible in AD mice as early as 2 months old. These data suggest that AECP could serve as an early, non-invasive, and objective biomarker for detecting AD and AD-related dementia (ADRD).
Collapse
Affiliation(s)
- Ling Mei
- Department of Otolaryngology, University of Kentucky Medical Center, Lexington, KY, United States
| | - Li-Man Liu
- Department of Otolaryngology, University of Kentucky Medical Center, Lexington, KY, United States
| | - Kaitian Chen
- Department of Otolaryngology, University of Kentucky Medical Center, Lexington, KY, United States
| | - Hong-Bo Zhao
- Department of Otolaryngology, University of Kentucky Medical Center, Lexington, KY, United States
| |
Collapse
|
8
|
Syrjanen J, Michalski K, Kawate T, Furukawa H. On the molecular nature of large-pore channels. J Mol Biol 2021; 433:166994. [PMID: 33865869 PMCID: PMC8409005 DOI: 10.1016/j.jmb.2021.166994] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 04/08/2021] [Accepted: 04/08/2021] [Indexed: 12/25/2022]
Abstract
Membrane transport is a fundamental means to control basic cellular processes such as apoptosis, inflammation, and neurodegeneration and is mediated by a number of transporters, pumps, and channels. Accumulating evidence over the last half century has shown that a type of so-called "large-pore channel" exists in various tissues and organs in gap-junctional and non-gap-junctional forms in order to flow not only ions but also metabolites such as ATP. They are formed by a number of protein families with little or no evolutionary linkages including connexin, innexin, pannexin, leucine-rich repeat-containing 8 (LRRC8), and calcium homeostasis modulator (CALHM). This review summarizes the history and concept of large-pore channels starting from connexin gap junction channels to the more recent developments in innexin, pannexin, LRRC8, and CALHM. We describe structural and functional features of large-pore channels that are crucial for their diverse functions on the basis of available structures.
Collapse
Affiliation(s)
- Johanna Syrjanen
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Kevin Michalski
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Toshimitsu Kawate
- Department of Molecular Medicine, Fields of Biochemistry, Molecular, and Cell Biology (BMCB), and Biophysics, Cornell University, Ithaca, NY 14853, USA
| | - Hiro Furukawa
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
9
|
O'Donnell BL, Penuela S. Pannexin 3 channels in health and disease. Purinergic Signal 2021; 17:577-589. [PMID: 34250568 DOI: 10.1007/s11302-021-09805-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/30/2021] [Indexed: 01/07/2023] Open
Abstract
Pannexin 3 (PANX3) is a member of the pannexin family of single membrane channel-forming glycoproteins. Originally thought to have a limited localization in cartilage, bone, and skin, PANX3 has now been detected in a variety of other tissues including skeletal muscle, mammary glands, the male reproductive tract, the cochlea, blood vessels, small intestines, teeth, and the vomeronasal organ. In many cell types of the musculoskeletal system, such as osteoblasts, chondrocytes, and odontoblasts, PANX3 has been shown to regulate the balance of proliferation and differentiation. PANX3 can be induced during progenitor cell differentiation, functioning at the cell surface as a conduit for ATP and/or in the endoplasmic reticulum as a calcium leak channel. Evidence in osteoblasts and monocytes also highlight a role for PANX3 in purinergic signalling through its function as an ATP release channel. PANX3 is critical in the development and ageing of bone and cartilage, with its levels temporally regulated in other tissues such as skeletal muscle, skin, and the cochlea. In diseases such as osteoarthritis and intervertebral disc degeneration, PANX3 can have either protective or detrimental roles depending on if the disease is age-related or injury-induced. This review will discuss PANX3 function in tissue growth and regeneration, its role in cellular differentiation, and how it becomes dysregulated in disease conditions such as obesity, Duchenne's muscular dystrophy, osteosarcoma, and non-melanoma skin cancer, where most of the findings on PANX3 function can be attributed to the characterization of Panx3 KO mouse models.
Collapse
Affiliation(s)
- Brooke L O'Donnell
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada.
- Department of Oncology, Division of Experimental Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada.
| |
Collapse
|
10
|
Mao H, Chen Y. Noise-Induced Hearing Loss: Updates on Molecular Targets and Potential Interventions. Neural Plast 2021; 2021:4784385. [PMID: 34306060 PMCID: PMC8279877 DOI: 10.1155/2021/4784385] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 06/12/2021] [Indexed: 12/18/2022] Open
Abstract
Noise overexposure leads to hair cell loss, synaptic ribbon reduction, and auditory nerve deterioration, resulting in transient or permanent hearing loss depending on the exposure severity. Oxidative stress, inflammation, calcium overload, glutamate excitotoxicity, and energy metabolism disturbance are the main contributors to noise-induced hearing loss (NIHL) up to now. Gene variations are also identified as NIHL related. Glucocorticoid is the only approved medication for NIHL treatment. New pharmaceuticals targeting oxidative stress, inflammation, or noise-induced neuropathy are emerging, highlighted by the nanoparticle-based drug delivery system. Given the complexity of the pathogenesis behind NIHL, deeper and more comprehensive studies still need to be fulfilled.
Collapse
Affiliation(s)
- Huanyu Mao
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai 200031, China
| | - Yan Chen
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai 200031, China
| |
Collapse
|
11
|
Visuomotor deficiency in panx1a knockout zebrafish is linked to dopaminergic signaling. Sci Rep 2020; 10:9538. [PMID: 32533080 PMCID: PMC7293225 DOI: 10.1038/s41598-020-66378-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 05/15/2020] [Indexed: 12/23/2022] Open
Abstract
Pannexin 1 (Panx1) forms ATP-permeable membrane channels that play roles in the nervous system. The analysis of roles in both standard and pathological conditions benefits from a model organism with rapid development and early onset of behaviors. Such a model was developed by ablating the zebrafish panx1a gene using TALEN technology. Here, RNA-seq analysis of 6 days post fertilization larvae were confirmed by Real-Time PCR and paired with testing visual-motor behavior and in vivo electrophysiology. Results demonstrated that loss of panx1a specifically affected the expression of gene classes representing the development of the visual system and visual processing. Abnormal swimming behavior in the dark and the expression regulation of pre-and postsynaptic biomarkers suggested changes in dopaminergic signaling. Indeed, altered visuomotor behavior in the absence of functional Panx1a was evoked through D1/D2-like receptor agonist treatment and rescued with the D2-like receptor antagonist Haloperidol. Local field potentials recorded from superficial areas of the optic tectum receiving input from the retina confirmed abnormal responses to visual stimuli, which resembled treatments with a dopamine receptor agonist or pharmacological blocking of Panx1a. We conclude that Panx1a functions are relevant at a time point when neuronal networks supporting visual-motor functions undergo modifications preparing for complex behaviors of freely swimming fish.
Collapse
|
12
|
Ishikawa M, Williams G, Forcinito P, Ishikawa M, Petrie RJ, Saito K, Fukumoto S, Yamada Y. Pannexin 3 ER Ca 2+ channel gating is regulated by phosphorylation at the Serine 68 residue in osteoblast differentiation. Sci Rep 2019; 9:18759. [PMID: 31822768 PMCID: PMC6904572 DOI: 10.1038/s41598-019-55371-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 11/19/2019] [Indexed: 12/31/2022] Open
Abstract
Pannexin 3 (Panx3) is a regulator of bone formation. Panx3 forms three distinct functional channels: hemichannels, gap junctions, and endoplasmic reticulum (ER) Ca2+ channels. However, the gating mechanisms of the Panx3 channels remain unclear. Here, we show that the Panx3 ER Ca2+ channel is modulated by phosphorylation of the serine 68 residue (Ser68) to promote osteoblast differentiation. Among the 17 candidate phosphorylation sites identified, the mutation of Ser68 to Ala (Ser68Ala) was sufficient to inhibit Panx3-mediated osteoblast differentiation via reduction of Osterix and ALP expression. Using a Ser68 phospho-specific antibody (P-Panx3) revealed Panx3 was phosphorylated in prehypertrophic, hypertrophic chondrocytes, and bone areas of the newborn growth plate. In osteogenic C2C12 cells, P-Panx3 was located on the ER membranes. Importantly, the Ser68Ala mutation only affected Panx3 ER Ca2+ channel function. Ser68 on Panx3 was phosphorylated by ATP stimulation and PI3K/Akt signaling. Finally, real-time FRET imaging and ratio analysis revealed that the Panx3 channel conformation was sensitive to ATP. Together, the phosphorylation of Panx3 at Ser68 is an essential step controlling the gating of the Panx3 ER Ca2+ channel to promote osteogenesis.
Collapse
Affiliation(s)
- Masaki Ishikawa
- Operative Dentistry, Tohoku University Graduate School of Dentistry, Sendai, 980-8575, Japan.
| | - Geneva Williams
- Molecular Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Philadelphia, USA
| | - Patricia Forcinito
- Molecular Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Philadelphia, USA
| | - Momoko Ishikawa
- Department of Pediatric Dentistry, Tohoku University Graduate School of Dentistry, Sendai, 980-8576, Japan
| | - Ryan J Petrie
- Department of Biology, Drexel University, Philadelphia, PA, 19104, USA
| | - Kan Saito
- Department of Pediatric Dentistry, Tohoku University Graduate School of Dentistry, Sendai, 980-8576, Japan
| | - Satoshi Fukumoto
- Department of Pediatric Dentistry, Tohoku University Graduate School of Dentistry, Sendai, 980-8576, Japan
| | - Yoshihiko Yamada
- Molecular Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Philadelphia, USA
| |
Collapse
|
13
|
Wu X, Zhang W, Li Y, Lin X. Structure and Function of Cochlear Gap Junctions and Implications for the Translation of Cochlear Gene Therapies. Front Cell Neurosci 2019; 13:529. [PMID: 31827424 PMCID: PMC6892400 DOI: 10.3389/fncel.2019.00529] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 11/13/2019] [Indexed: 12/23/2022] Open
Abstract
Connexins (Cxs) are ubiquitous membrane proteins that are found throughout vertebrate organs, acting as building blocks of the gap junctions (GJs) known to play vital roles in the normal function of many organs. Mutations in Cx genes (particularly GJB2, which encodes Cx26) cause approximately half of all cases of congenital hearing loss in newborns. Great progress has been made in understanding GJ function and the molecular mechanisms for the role of Cxs in the cochlea. Data reveal that multiple types of Cxs work together to ensure normal development and function of the cochlea. These findings include many aspects not proposed in the classic K+ recycling theory, such as the formation of normal cochlear morphology (e.g., the opening of the tunnel of Corti), the fine-tuning of the innervation of nerve fibers to the hair cells (HCs), the maturation of the ribbon synapses, and the initiation of the endocochlear potential (EP). New data, especially those collected from targeted modification of major Cx genes in the mouse cochlea, have demonstrated that Cx26 plays an essential role in the postnatal maturation of the cochlea. Studies also show that Cx26 and Cx30 assume very different roles in the EP generation, given that only Cx26 is required for normal hearing. This article will review our current understanding of the molecular structure, cellular distribution, and major functions of cochlear GJs. Potential implications of the knowledge of cochlear GJs on the design and implementation of translational studies of cochlear gene therapies for Cx mutations are also discussed.
Collapse
Affiliation(s)
- Xuewen Wu
- Department of Otolaryngology, Head-Neck and Surgery, Xiangya Hospital of Central South University, Changsha, China
- Department of Otolaryngology, Emory University School of Medicine, Atlanta, GA, United States
| | - Wenjuan Zhang
- Department of Otolaryngology, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yihui Li
- Department of Pharmacy, Changsha Hospital of Traditional Medicine, Changsha, China
| | - Xi Lin
- Department of Otolaryngology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
14
|
Köles L, Szepesy J, Berekméri E, Zelles T. Purinergic Signaling and Cochlear Injury-Targeting the Immune System? Int J Mol Sci 2019; 20:ijms20122979. [PMID: 31216722 PMCID: PMC6627352 DOI: 10.3390/ijms20122979] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/14/2019] [Accepted: 06/14/2019] [Indexed: 02/06/2023] Open
Abstract
Hearing impairment is the most common sensory deficit, affecting more than 400 million people worldwide. Sensorineural hearing losses currently lack any specific or efficient pharmacotherapy largely due to the insufficient knowledge of the pathomechanism. Purinergic signaling plays a substantial role in cochlear (patho)physiology. P2 (ionotropic P2X and the metabotropic P2Y) as well as adenosine receptors expressed on cochlear sensory and non-sensory cells are involved mostly in protective mechanisms of the cochlea. They are implicated in the sensitivity adjustment of the receptor cells by a K+ shunt and can attenuate the cochlear amplification by modifying cochlear micromechanics. Cochlear blood flow is also regulated by purines. Here, we propose to comprehend this field with the purine-immune interactions in the cochlea. The role of harmful immune mechanisms in sensorineural hearing losses has been emerging in the horizon of cochlear pathologies. In addition to decreasing hearing sensitivity and increasing cochlear blood supply, influencing the immune system can be the additional avenue for pharmacological targeting of purinergic signaling in the cochlea. Elucidating this complexity of purinergic effects on cochlear functions is necessary and it can result in development of new therapeutic approaches in hearing disabilities, especially in the noise-induced ones.
Collapse
Affiliation(s)
- László Köles
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary.
| | - Judit Szepesy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary.
| | - Eszter Berekméri
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary.
- Department of Ecology, University of Veterinary Medicine, H-1078 Budapest, Hungary.
| | - Tibor Zelles
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary.
- Department of Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, H-1083 Budapest, Hungary.
| |
Collapse
|
15
|
Double deletion of Panx1 and Panx3 affects skin and bone but not hearing. J Mol Med (Berl) 2019; 97:723-736. [PMID: 30918989 DOI: 10.1007/s00109-019-01779-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 03/11/2019] [Accepted: 03/13/2019] [Indexed: 12/15/2022]
Abstract
Pannexins (Panxs), large-pore channel forming glycoproteins, are expressed in a wide variety of tissues including the skin, bone, and cochlea. To date, the use of single knock-out mouse models of both Panx1 and Panx3 have demonstrated their roles in skin development, bone formation, and auditory phenotypes. Due to sequence homology between Panx1 and Panx3, when one Panx is ablated from germline, the other may be upregulated in a compensatory mechanism to maintain tissue homeostasis and function. To evaluate the roles of Panx1 and Panx3 in the skin, bone, and cochlea, we created the first Panx1/Panx3 double knock-out mouse model (dKO). These mice had smaller litters and reduced body weight compared to wildtype controls. The dKO dorsal skin had decreased epidermal and dermal area as well as decreased hypodermal area in neonatal but not in older mice. In addition, mouse skull shape and size were altered, and long bone length was decreased in neonatal dKO mice. Finally, auditory tests revealed that dKO mice did not exhibit hearing loss and were even slightly protected against noise-induced hearing damage at mid-frequency regions. Taken together, our findings suggest that Panx1 and Panx3 are important at early stages of development in the skin and bone but may be redundant in the auditory system. KEY MESSAGES: Panx double KO mice had smaller litters and reduced body weight. dKO skin had decreased epidermal and dermal area in neonatal mice. Skull shape and size changed plus long bone length decreased in neonatal dKO mice. dKO had no hearing loss and were slightly protected against noise-induced damage.
Collapse
|
16
|
Berekméri E, Szepesy J, Köles L, Zelles T. Purinergic signaling in the organ of Corti: Potential therapeutic targets of sensorineural hearing losses. Brain Res Bull 2019; 151:109-118. [PMID: 30721767 DOI: 10.1016/j.brainresbull.2019.01.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 01/10/2019] [Accepted: 01/25/2019] [Indexed: 01/04/2023]
Abstract
Purinergic signaling is deeply involved in the development, functions and protective mechanisms of the cochlea. Release of ATP and activation of purinergic receptors on sensory and supporting/epithelial cells play a substantial role in cochlear (patho)physiology. Both the ionotropic P2X and the metabotropic P2Y receptors are widely distributed on the inner and outer hair cells as well as on the different supporting cells in the organ of Corti and on other epithelial cells in the scala media. Among others, they are implicated in the sensitivity adjustment of the receptor cells by a K+ shunt and can attenuate the cochlear amplification by modifying cochlear micromechanics acting on outer hair cells and supporting cells. Cochlear blood flow is also regulated by purines. Sensorineural hearing losses currently lack any specific or efficient pharmacotherapy. Decreasing hearing sensitivity and increasing cochlear blood supply by pharmacological targeting of purinergic signaling in the cochlea are potential new therapeutic approaches in these hearing disabilities, especially in the noise-induced ones.
Collapse
Affiliation(s)
- Eszter Berekméri
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Judit Szepesy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - László Köles
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Tibor Zelles
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Department of Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary.
| |
Collapse
|
17
|
Boucher J, Simonneau C, Denet G, Clarhaut J, Balandre AC, Mesnil M, Cronier L, Monvoisin A. Pannexin-1 in Human Lymphatic Endothelial Cells Regulates Lymphangiogenesis. Int J Mol Sci 2018; 19:ijms19061558. [PMID: 29882918 PMCID: PMC6032340 DOI: 10.3390/ijms19061558] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 05/17/2018] [Accepted: 05/22/2018] [Indexed: 12/23/2022] Open
Abstract
The molecular mechanisms governing the formation of lymphatic vasculature are not yet well understood. Pannexins are transmembrane proteins that form channels which allow for diffusion of ions and small molecules (<1 kDa) between the extracellular space and the cytosol. The expression and function of pannexins in blood vessels have been studied in the last few decades. Meanwhile, no studies have been conducted to evaluate the role of pannexins during human lymphatic vessel formation. Here we show, using primary human dermal lymphatic endothelial cells (HDLECs), pharmacological tools (probenecid, Brilliant Blue FCF, mimetic peptides [10Panx]) and siRNA-mediated knockdown that Pannexin-1 is necessary for capillary tube formation on Matrigel and for VEGF-C-induced invasion. These results newly identify Pannexin-1 as a protein highly expressed in HDLECs and its requirement during in vitro lymphangiogenesis.
Collapse
Affiliation(s)
- Jonathan Boucher
- CNRS ERL 7003, Laboratoire "Signalisation & Transports Ioniques Membranaires", University of Poitiers, 86073 Poitiers, France.
| | - Claire Simonneau
- CNRS ERL 7003, Laboratoire "Signalisation & Transports Ioniques Membranaires", University of Poitiers, 86073 Poitiers, France.
| | - Golthlay Denet
- CNRS ERL 7003, Laboratoire "Signalisation & Transports Ioniques Membranaires", University of Poitiers, 86073 Poitiers, France.
| | - Jonathan Clarhaut
- CNRS UMR 7285, Institut de Chimie des Milieux et des Matériaux de Poitiers (IC2MP), University of Poitiers, 86073 Poitiers, France.
- CHU de Poitiers, 86021 Poitiers, France.
| | - Annie-Claire Balandre
- CNRS ERL 7003, Laboratoire "Signalisation & Transports Ioniques Membranaires", University of Poitiers, 86073 Poitiers, France.
| | - Marc Mesnil
- CNRS ERL 7003, Laboratoire "Signalisation & Transports Ioniques Membranaires", University of Poitiers, 86073 Poitiers, France.
| | - Laurent Cronier
- CNRS ERL 7003, Laboratoire "Signalisation & Transports Ioniques Membranaires", University of Poitiers, 86073 Poitiers, France.
| | - Arnaud Monvoisin
- CNRS ERL 7003, Laboratoire "Signalisation & Transports Ioniques Membranaires", University of Poitiers, 86073 Poitiers, France.
| |
Collapse
|
18
|
Knockout of Pannexin-1 Induces Hearing Loss. Int J Mol Sci 2018; 19:ijms19051332. [PMID: 29710868 PMCID: PMC5983795 DOI: 10.3390/ijms19051332] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/20/2018] [Accepted: 04/23/2018] [Indexed: 12/31/2022] Open
Abstract
Mutations of gap junction connexin genes induce a high incidence of nonsyndromic hearing loss. Pannexin genes also encode gap junctional proteins in vertebrates. Recent studies demonstrated that Pannexin-1 (Panx1) deficiency in mice and mutation in humans are also associated with hearing loss. So far, several Panx1 knockout (KO) mouse lines were established. In general, these Panx1 KO mouse lines demonstrate consistent phenotypes in most aspects, including hearing loss. However, a recent study reported that a Panx1 KO mouse line, which was created by Genentech Inc., had no hearing loss as measured by the auditory brainstem response (ABR) threshold at low-frequency range (<24 kHz). Here, we used multiple auditory function tests and re-examined hearing function in the Genentech Panx1 (Gen-Panx1) KO mouse. We found that ABR thresholds in the Gen-Panx1 KO mouse were significantly increased, in particular, in the high-frequency region. Moreover, consistent with the increase in ABR threshold, distortion product otoacoustic emission (DPOAE) and cochlear microphonics (CM), which reflect active cochlear amplification and auditory receptor current, respectively, were significantly reduced. These data demonstrated that the Gen-Panx1 KO mouse has hearing loss and further confirmed that Panx1 deficiency can cause deafness.
Collapse
|
19
|
Whyte-Fagundes P, Kurtenbach S, Zoidl C, Shestopalov VI, Carlen PL, Zoidl G. A Potential Compensatory Role of Panx3 in the VNO of a Panx1 Knock Out Mouse Model. Front Mol Neurosci 2018; 11:135. [PMID: 29780304 PMCID: PMC5946002 DOI: 10.3389/fnmol.2018.00135] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 04/03/2018] [Indexed: 12/30/2022] Open
Abstract
Pannexins (Panx) are integral membrane proteins, with Panx1 being the best-characterized member of the protein family. Panx1 is implicated in sensory processing, and knockout (KO) animal models have become the primary tool to investigate the role(s) of Panx1 in sensory systems. Extending previous work from our group on primary olfaction, the expression patterns of Panxs in the vomeronasal organ (VNO), an auxiliary olfactory sense organ with a role in reproduction and social behavior, were compared. Using qRT-PCR and Immunohistochemistry (IHC), we confirmed the loss of Panx1, found similar Panx2 expression levels in both models, and a significant upregulation of Panx3 in mice with a global ablation of Panx1. Specifically, Panx3 showed upregulated expression in nerve fibers of the non-sensory epithelial layer in juvenile and adult KO mice and in the sensory layer of adults, which overlaps with Panx1 expression areas in WT populations. Since both social behavior and evoked ATP release in the VNO was not compromised in KO animals, we hypothesized that Panx3 could compensate for the loss of Panx1. This led us to compare Panx1 and Panx3 channels in vitro, demonstrating similar dye uptake and ATP release properties. Outcomes of this study strongly suggest that Panx3 may functionally compensate for the loss of Panx1 in the VNO of the olfactory system, ensuring sustained chemosensory processing. This finding extends previous reports on the upregulation of Panx3 in arterial walls and the skin of Panx1 KO mice, suggesting that roles of Panx1 warrant uncharacterized safeguarding mechanisms involving Panx3.
Collapse
Affiliation(s)
- Paige Whyte-Fagundes
- Department of Biology, York University, Toronto, ON, Canada.,Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Stefan Kurtenbach
- Department of Biology, York University, Toronto, ON, Canada.,Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | | | - Valery I Shestopalov
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Peter L Carlen
- Krembil Research Institute, University Health Network, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Georg Zoidl
- Department of Biology, York University, Toronto, ON, Canada.,Krembil Research Institute, University Health Network, Toronto, ON, Canada.,Department of Psychology, York University, Toronto, ON, Canada
| |
Collapse
|
20
|
Abstract
Adenosine triphosphate (ATP) has been well established as an important extracellular ligand of autocrine signaling, intercellular communication, and neurotransmission with numerous physiological and pathophysiological roles. In addition to the classical exocytosis, non-vesicular mechanisms of cellular ATP release have been demonstrated in many cell types. Although large and negatively charged ATP molecules cannot diffuse across the lipid bilayer of the plasma membrane, conductive ATP release from the cytosol into the extracellular space is possible through ATP-permeable channels. Such channels must possess two minimum qualifications for ATP permeation: anion permeability and a large ion-conducting pore. Currently, five groups of channels are acknowledged as ATP-release channels: connexin hemichannels, pannexin 1, calcium homeostasis modulator 1 (CALHM1), volume-regulated anion channels (VRACs, also known as volume-sensitive outwardly rectifying (VSOR) anion channels), and maxi-anion channels (MACs). Recently, major breakthroughs have been made in the field by molecular identification of CALHM1 as the action potential-dependent ATP-release channel in taste bud cells, LRRC8s as components of VRACs, and SLCO2A1 as a core subunit of MACs. Here, the function and physiological roles of these five groups of ATP-release channels are summarized, along with a discussion on the future implications of understanding these channels.
Collapse
|
21
|
Zorzi V, Paciello F, Ziraldo G, Peres C, Mazzarda F, Nardin C, Pasquini M, Chiani F, Raspa M, Scavizzi F, Carrer A, Crispino G, Ciubotaru CD, Monyer H, Fetoni AR, M Salvatore A, Mammano F. Mouse Panx1 Is Dispensable for Hearing Acquisition and Auditory Function. Front Mol Neurosci 2017; 10:379. [PMID: 29234270 PMCID: PMC5712377 DOI: 10.3389/fnmol.2017.00379] [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: 10/02/2017] [Accepted: 10/30/2017] [Indexed: 11/13/2022] Open
Abstract
Panx1 forms plasma membrane channels in brain and several other organs, including the inner ear. Biophysical properties, activation mechanisms and modulators of Panx1 channels have been characterized in detail, however the impact of Panx1 on auditory function is unclear due to conflicts in published results. To address this issue, hearing performance and cochlear function of the Panx1−/− mouse strain, the first with a reported global ablation of Panx1, were scrutinized. Male and female homozygous (Panx1−/−), hemizygous (Panx1+/−) and their wild type (WT) siblings (Panx1+/+) were used for this study. Successful ablation of Panx1 was confirmed by RT-PCR and Western immunoblotting in the cochlea and brain of Panx1−/− mice. Furthermore, a previously validated Panx1-selective antibody revealed strong immunoreactivity in WT but not in Panx1−/− cochleae. Hearing sensitivity, outer hair cell-based “cochlear amplifier” and cochlear nerve function, analyzed by auditory brainstem response (ABR) and distortion product otoacoustic emission (DPOAE) recordings, were normal in Panx1+/− and Panx1−/− mice. In addition, we determined that global deletion of Panx1 impacts neither on connexin expression, nor on gap-junction coupling in the developing organ of Corti. Finally, spontaneous intercellular Ca2+ signal (ICS) activity in organotypic cochlear cultures, which is key to postnatal development of the organ of Corti and essential for hearing acquisition, was not affected by Panx1 ablation. Therefore, our results provide strong evidence that, in mice, Panx1 is dispensable for hearing acquisition and auditory function.
Collapse
Affiliation(s)
- Veronica Zorzi
- CNR Institute of Cell Biology and Neurobiology, Monterotondo, Italy.,School of Medicine, Institute of Otolaryngology, Catholic University, Rome, Italy
| | - Fabiola Paciello
- CNR Institute of Cell Biology and Neurobiology, Monterotondo, Italy
| | - Gaia Ziraldo
- CNR Institute of Cell Biology and Neurobiology, Monterotondo, Italy.,School of Medicine, Institute of Otolaryngology, Catholic University, Rome, Italy
| | - Chiara Peres
- CNR Institute of Cell Biology and Neurobiology, Monterotondo, Italy
| | - Flavia Mazzarda
- CNR Institute of Cell Biology and Neurobiology, Monterotondo, Italy.,Department of Science, Roma Tre University, Rome, Italy
| | - Chiara Nardin
- CNR Institute of Cell Biology and Neurobiology, Monterotondo, Italy.,Department of Science, Roma Tre University, Rome, Italy
| | - Miriam Pasquini
- CNR Institute of Cell Biology and Neurobiology, Monterotondo, Italy.,Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Rome, Italy
| | - Francesco Chiani
- CNR Institute of Cell Biology and Neurobiology, Monterotondo, Italy
| | - Marcello Raspa
- CNR Institute of Cell Biology and Neurobiology, Monterotondo, Italy
| | | | - Andrea Carrer
- Department of Physics and Astronomy G. Galilei, University of Padua, Padua, Italy
| | - Giulia Crispino
- Department of Physics and Astronomy G. Galilei, University of Padua, Padua, Italy
| | | | - Hannah Monyer
- Department of Clinical Neurobiology, Deutches Krebforschungzentrum, University of Heidelberg, Heidelberg, Germany
| | - Anna R Fetoni
- School of Medicine, Institute of Otolaryngology, Catholic University, Rome, Italy
| | - Anna M Salvatore
- CNR Institute of Cell Biology and Neurobiology, Monterotondo, Italy
| | - Fabio Mammano
- CNR Institute of Cell Biology and Neurobiology, Monterotondo, Italy.,Department of Physics and Astronomy G. Galilei, University of Padua, Padua, Italy.,Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| |
Collapse
|
22
|
Verselis VK. Connexin hemichannels and cochlear function. Neurosci Lett 2017; 695:40-45. [PMID: 28917982 DOI: 10.1016/j.neulet.2017.09.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 07/24/2017] [Accepted: 09/10/2017] [Indexed: 01/01/2023]
Abstract
Connexins play vital roles in hearing, including promoting cochlear development and sustaining auditory function in the mature cochlea. Mutations in connexins expressed in the cochlear epithelium, Cx26 and Cx30, cause sensorineural deafness and in the case of Cx26, is one of the most common causes of non-syndromic, hereditary deafness. Connexins function as gap junction channels and as hemichannels, which mediate intercellular and transmembrane signaling, respectively. Both channel configurations can play important, but very different roles in the cochlea. The potential roles connexin hemichannels can play are discussed both in normal cochlear function and in promoting pathogenesis that can lead to hearing loss.
Collapse
Affiliation(s)
- Vytas K Verselis
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, United States.
| |
Collapse
|
23
|
Whyte-Fagundes P, Zoidl G. Mechanisms of pannexin1 channel gating and regulation. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:65-71. [PMID: 28735901 DOI: 10.1016/j.bbamem.2017.07.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 07/17/2017] [Accepted: 07/18/2017] [Indexed: 01/07/2023]
Abstract
Pannexins are a family of integral membrane proteins with distinct post-translational modifications, sub-cellular localization and tissue distribution. Panx1 is the most studied and best-characterized isoform of this gene family. The ubiquitous expression, as well as its function as a major ATP release and nucleotide permeation channel, makes Panx1 a primary candidate for participating in the pathophysiology of CNS disorders. While many investigations revolve around Panx1 functions in health and disease, more recently, details started emerging about mechanisms that control Panx1 channel activity. These advancements in Panx1 biology have revealed that beyond its classical role as an unopposed plasma membrane channel, it participates in alternative pathways involving multiple intracellular compartments, protein complexes and a myriad of extracellular participants. Here, we review recent progress in our understanding of Panx1 at the center of these pathways, highlighting its modulation in a context specific manner. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
| | - Georg Zoidl
- Biology, York University, Toronto, Canada; Psychology, York University, Toronto, Canada.
| |
Collapse
|
24
|
Whyte-Fagundes P, Siu R, Brown C, Zoidl G. Pannexins in vision, hearing, olfaction and taste. Neurosci Lett 2017; 695:32-39. [PMID: 28495272 DOI: 10.1016/j.neulet.2017.05.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 04/06/2017] [Accepted: 05/05/2017] [Indexed: 12/25/2022]
Abstract
In mammals, the pannexin gene family consists of three members (Panx1, 2, 3), which represent a class of integral membrane channel proteins sharing some structural features with chordate gap junction proteins, the connexins. Since their discovery in the early 21st century, pannexin expression has been detected throughout the vertebrate body including eye, ear, nose and tongue, making the investigation of the roles of this new class of channel protein in health and disease very appealing. The localization in sensory organs, coupled with unique channel properties and associations with major signaling pathways make Panx1, and its relative's, significant contributors for fundamental functions in sensory perception. Until recently, cell-based studies were at the forefront of pannexin research. Lately, the availability of mice with genetic ablation of pannexins opened new avenues for testing pannexin functions and behavioural phenotyping. Although we are only at the beginning of understanding the roles of pannexins in health and disease, this review summarizes recent advances in elucidating the various emerging roles pannexins play in sensory systems, with an emphasis on unresolved conflicts.
Collapse
Affiliation(s)
- Paige Whyte-Fagundes
- Graduate Program In Biology, Faculty of Science, York University, Toronto, ON, Canada
| | - Ryan Siu
- Graduate Program In Biology, Faculty of Science, York University, Toronto, ON, Canada
| | - Cherie Brown
- Graduate Program In Biology, Faculty of Science, York University, Toronto, ON, Canada
| | - Georg Zoidl
- Department of Biology, Faculty of Science, York University, Toronto, ON, Canada; Center for Vision Research, York University, Toronto, ON, Canada.
| |
Collapse
|
25
|
Mittal R, Debs LH, Nguyen D, Patel AP, Grati M, Mittal J, Yan D, Eshraghi AA, Liu XZ. Signaling in the Auditory System: Implications in Hair Cell Regeneration and Hearing Function. J Cell Physiol 2017; 232:2710-2721. [DOI: 10.1002/jcp.25695] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 11/18/2016] [Indexed: 02/06/2023]
Affiliation(s)
- Rahul Mittal
- Department of Otolaryngology; University of Miami Miller School of Medicine; Miami Florida
| | - Luca H. Debs
- Department of Otolaryngology; University of Miami Miller School of Medicine; Miami Florida
| | - Desiree Nguyen
- Department of Otolaryngology; University of Miami Miller School of Medicine; Miami Florida
| | - Amit P. Patel
- Department of Otolaryngology; University of Miami Miller School of Medicine; Miami Florida
| | - M'hamed Grati
- Department of Otolaryngology; University of Miami Miller School of Medicine; Miami Florida
| | - Jeenu Mittal
- Department of Otolaryngology; University of Miami Miller School of Medicine; Miami Florida
| | - Denise Yan
- Department of Otolaryngology; University of Miami Miller School of Medicine; Miami Florida
| | - Adrien A. Eshraghi
- Department of Otolaryngology; University of Miami Miller School of Medicine; Miami Florida
| | - Xue Zhong Liu
- Department of Otolaryngology; University of Miami Miller School of Medicine; Miami Florida
| |
Collapse
|
26
|
Boyce AKJ, Epp AL, Nagarajan A, Swayne LA. Transcriptional and post-translational regulation of pannexins. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:72-82. [PMID: 28279657 DOI: 10.1016/j.bbamem.2017.03.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 03/02/2017] [Accepted: 03/03/2017] [Indexed: 12/21/2022]
Abstract
Pannexins are a 3-membered family of proteins that form large pore ion and metabolite channels in vertebrates. The impact of pannexins on vertebrate biology is intricately tied to where and when they are expressed, and how they are modified, once produced. The purpose of this review is therefore to outline our current understanding of transcriptional and post-translational regulation of pannexins. First, we briefly summarize their discovery and characteristics. Next, we describe several aspects of transcriptional regulation, including cell and tissue-specific expression, dynamic expression over development and disease, as well as new insights into the underlying molecular machinery involved. Following this, we delve into the role of post-translational modifications in the regulation of trafficking and channel properties, highlighting important work on glycosylation, phosphorylation, S-nitrosylation and proteolytic cleavage. Embedded throughout, we also highlight important knowledge gaps and avenues of future research. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- Andrew K J Boyce
- Division of Medical Sciences and Island Medical Program, University of Victoria, Victoria V8P 5C2, Canada
| | - Anna L Epp
- Division of Medical Sciences and Island Medical Program, University of Victoria, Victoria V8P 5C2, Canada
| | - Archana Nagarajan
- Division of Medical Sciences and Island Medical Program, University of Victoria, Victoria V8P 5C2, Canada
| | - Leigh Anne Swayne
- Division of Medical Sciences and Island Medical Program, University of Victoria, Victoria V8P 5C2, Canada; Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver V6T 1Z3, Canada.
| |
Collapse
|
27
|
Meehan DT, Delimont D, Dufek B, Zallocchi M, Phillips G, Gratton MA, Cosgrove D. Endothelin-1 mediated induction of extracellular matrix genes in strial marginal cells underlies strial pathology in Alport mice. Hear Res 2016; 341:100-108. [PMID: 27553900 PMCID: PMC5086449 DOI: 10.1016/j.heares.2016.08.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 07/21/2016] [Accepted: 08/15/2016] [Indexed: 12/20/2022]
Abstract
Alport syndrome, a type IV collagen disorder, manifests as glomerular disease associated with hearing loss with thickening of the glomerular and strial capillary basement membranes (SCBMs). We have identified a role for endothelin-1 (ET-1) activation of endothelin A receptors (ETARs) in glomerular pathogenesis. Here we explore whether ET-1 plays a role in strial pathology. Wild type (WT) and Alport mice were treated with the ETAR antagonist, sitaxentan. The stria vascularis was analyzed for SCBM thickness and for extracellular matrix (ECM) proteins. Additional WT and Alport mice were exposed to noise or hypoxia and the stria analyzed for hypoxia-related and ECM genes. A strial marginal cell line cultured under hypoxic conditions, or stimulated with ET-1 was analyzed for expression of hypoxia-related and ECM transcripts. Noise exposure resulted in significantly elevated ABR thresholds in Alport mice relative to wild type littermates. Alport stria showed elevated expression of collagen α1(IV), laminin α2, and laminin α5 proteins relative to WT. SCBM thickening and elevated ECM protein expression was ameliorated by ETAR blockade. Stria from normoxic Alport mice and hypoxic WT mice showed upregulation of hypoxia-related, ECM, and ET-1 transcripts. Both ET-1 stimulation and hypoxia up-regulated ECM transcripts in cultured marginal cells. We conclude that ET-1 mediated activation of ETARs on strial marginal cells results in elevated expression of ECM genes and thickening of the SCBMs in Alport mice. SCBM thickening results in hypoxic stress further elevating ECM and ET-1 gene expression, exacerbating strial pathology.
Collapse
Affiliation(s)
| | | | - Brianna Dufek
- Boys Town National Research Hospital, Omaha, NE, USA
| | | | | | | | - Dominic Cosgrove
- Boys Town National Research Hospital, Omaha, NE, USA; University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
28
|
Differential effects of pannexins on noise-induced hearing loss. Biochem J 2016; 473:4665-4680. [PMID: 27784763 DOI: 10.1042/bcj20160668] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 10/14/2016] [Accepted: 10/24/2016] [Indexed: 12/20/2022]
Abstract
Hearing loss, including noise-induced hearing loss, is highly prevalent and severely hinders an individual's quality of life, yet many of the mechanisms that cause hearing loss are unknown. The pannexin (Panx) channel proteins, Panx1 and Panx3, are regionally expressed in many cell types along the auditory pathway, and mice lacking Panx1 in specific cells of the inner ear exhibit hearing loss, suggesting a vital role for Panxs in hearing. We proposed that Panx1 and/or Panx3 null mice would exhibit severe hearing loss and increased susceptibility to noise-induced hearing loss. Using the auditory brainstem response, we surprisingly found that Panx1-/- and Panx3-/- mice did not harbor hearing or cochlear nerve deficits. Furthermore, while Panx1-/- mice displayed no protection against loud noise-induced hearing loss, Panx3-/- mice exhibited enhanced 16- and 24-kHz hearing recovery 7 days after a loud noise exposure (NE; 12 kHz tone, 115 dB sound pressure level, 1 h). Interestingly, Cx26, Cx30, Cx43, and Panx2 were up-regulated in Panx3-/- mice compared with wild-type and/or Panx1-/- mice, and assessment of the auditory tract revealed morphological changes in the middle ear bones of Panx3-/- mice. It is unclear if these changes alone are sufficient to provide protection against loud noise-induced hearing loss. Contrary to what we expected, these data suggest that Panx1 and Panx3 are not essential for baseline hearing in mice tested, but the therapeutic targeting of Panx3 may prove protective against mid-high-frequency hearing loss caused by loud NE.
Collapse
|
29
|
Abstract
Pannexin (Panx) is a gene family encoding gap junction proteins in vertebrates. So far, three isoforms (Panx1, 2 and 3) have been identified. All of three Panx isoforms express in the cochlea with distinct expression patterns. Panx1 expresses in the cochlea extensively, including the spiral limbus, the organ of Corti, and the cochlear lateral wall, whereas Panx2 and Panx3 restrict to the basal cells of the stria vascularis in the lateral wall and the cochlear bony structure, respectively. However, there is no pannexin expression in auditory sensory hair cells. Recent studies demonstrated that like connexin gap junction gene, Panx1 deficiency causes hearing loss. Panx1 channels dominate ATP release in the cochlea. Deletion of Panx1 abolishes ATP release in the cochlea and reduces endocochlear potential (EP), auditory receptor current/potential, and active cochlear amplification. Panx1 deficiency in the cochlea also activates caspase-3 cell apoptotic pathway leading to cell degeneration. These new findings suggest that pannexins have a critical role in the cochlea in regard to hearing. However, detailed information about pannexin function in the cochlea and Panx mutation induced hearing loss still remain largely undetermined. Further studies are required.
Collapse
Affiliation(s)
- Hong-Bo Zhao
- Department of Otolaryngology, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY, 40536, USA.
| |
Collapse
|
30
|
Shao Q, Lindstrom K, Shi R, Kelly J, Schroeder A, Juusola J, Levine KL, Esseltine JL, Penuela S, Jackson MF, Laird DW. A Germline Variant in the PANX1 Gene Has Reduced Channel Function and Is Associated with Multisystem Dysfunction. J Biol Chem 2016; 291:12432-12443. [PMID: 27129271 DOI: 10.1074/jbc.m116.717934] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Indexed: 12/20/2022] Open
Abstract
Pannexin1 (PANX1) is probably best understood as an ATP release channel involved in paracrine signaling. Given its ubiquitous expression, PANX1 pathogenic variants would be expected to lead to disorders involving multiple organ systems. Using whole exome sequencing, we discovered the first patient with a homozygous PANX1 variant (c.650G→A) resulting in an arginine to histidine substitution at position 217 (p.Arg217His). The 17-year-old female has intellectual disability, sensorineural hearing loss requiring bilateral cochlear implants, skeletal defects, including kyphoscoliosis, and primary ovarian failure. Her consanguineous parents are each heterozygous for this variant but are not affected by the multiorgan syndromes noted in the proband. Expression of the p.Arg217His mutant in HeLa, N2A, HEK293T, and Ad293 cells revealed normal PANX1 glycosylation and cell surface trafficking. Dye uptake, ATP release, and electrophysiological measurements revealed p.Arg217His to be a loss-of-function variant. Co-expression of the mutant with wild-type PANX1 suggested the mutant was not dominant-negative to PANX1 channel function. Collectively, we demonstrate a PANX1 missense change associated with human disease in the first report of a "PANX1-related disorder."
Collapse
Affiliation(s)
- Qing Shao
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Kristin Lindstrom
- Division of Genetics and Metabolism, Phoenix Children's Hospital, Phoenix, Arizona 85016
| | - Ruoyang Shi
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Manitoba R3E 0Z3, Canada,; Kleysen Institute for Advanced Medicine, Health Sciences Centre, Winnipeg, Manitoba R3E 0Z3, Canada
| | - John Kelly
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Audrey Schroeder
- Division of Genetics, University of Rochester Medical Center, Rochester, New York 14642
| | | | | | - Jessica L Esseltine
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Michael F Jackson
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Manitoba R3E 0Z3, Canada,; Kleysen Institute for Advanced Medicine, Health Sciences Centre, Winnipeg, Manitoba R3E 0Z3, Canada
| | - Dale W Laird
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario N6A 5C1, Canada.
| |
Collapse
|
31
|
Caskenette D, Penuela S, Lee V, Barr K, Beier F, Laird DW, Willmore KE. Global deletion of Panx3 produces multiple phenotypic effects in mouse humeri and femora. J Anat 2016; 228:746-56. [PMID: 26749194 DOI: 10.1111/joa.12437] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2015] [Indexed: 02/04/2023] Open
Abstract
Pannexins form single-membrane channels that allow passage of small molecules between the intracellular and extracellular compartments. Of the three pannexin family members, Pannexin3 (Panx3) is the least studied but is highly expressed in skeletal tissues and is thought to play a role in the regulation of chondrocyte and osteoblast proliferation and differentiation. The purpose of our study is to closely examine the in vivo effects of Panx3 ablation on long bone morphology using micro-computed tomography. Using Panx3 knockout (KO) and wildtype (WT) adult mice, we measured and compared aspects of phenotypic shape, bone mineral density (BMD), cross-sectional geometric properties of right femora and humeri, and lean mass. We found that KO mice have absolutely and relatively shorter diaphyseal shafts compared with WT mice, and relatively larger areas of muscle attachment sites. No differences in BMD or lean mass were found between WT and KO mice. Interestingly, KO mice had more robust femora and humeri compared with WT mice when assessed in cross-section at the midshaft. Our results clearly show that Panx3 ablation produces phenotypic effects in mouse femora and humeri, and support the premise that Panx3 has a role in regulating long bone growth and development.
Collapse
Affiliation(s)
- Deidre Caskenette
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Vanessa Lee
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Kevin Barr
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Frank Beier
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada.,Children's Health Research Institute, London, ON, Canada
| | - Dale W Laird
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada.,Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada
| | - Katherine E Willmore
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| |
Collapse
|
32
|
Raslan A, Hainz N, Beckmann A, Tschernig T, Meier C. Pannexin-1 expression in developing mouse nervous system: new evidence for expression in sensory ganglia. Cell Tissue Res 2015; 364:29-41. [PMID: 26453396 DOI: 10.1007/s00441-015-2294-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 09/07/2015] [Indexed: 12/14/2022]
Abstract
Pannexin1 (Panx1) is one of three members of the pannexin protein family. The expression of Panx1 mRNA has been extensively investigated from late embryonic to adult stages. In contrast, expression during early embryonic development is largely unknown. Our aim is to examine the temporal and spatial expression of Panx1 in mouse embryonic development by focusing on embryonic days (E) 9.5 to 12.5. Whole embryos are investigated in order to provide a comprehensive survey. Analyses were performed at the mRNA level by using reverse transcription plus the polymerase chain reaction and whole-mount in situ hybridization. Panx1 mRNA was detected in the heads and bodies of embryos at all developmental stages investigated (E9.5, E10.5, E11.5, E12.5). In particular, the nervous system expressed Panx1 at an early time point. Interestingly, Panx1 expression was found in afferent ganglia of the cranial nerves and spinal cord. This finding is of particular interest in the context of neuropathic pain and other Panx1-related neurological disorders. Our study shows, for the first time, that Panx1 is expressed in the central and peripheral nervous system during early developmental stages. The consequences of Panx1 deficiency or inhibition in a number of experimental paradigms might therefore be predicated on changes during early development.
Collapse
Affiliation(s)
- Abdulrahman Raslan
- Department of Anatomy and Cell Biology, Saarland University, Building 61, 66424, Homburg, Saar, Germany
| | - Nadine Hainz
- Department of Anatomy and Cell Biology, Saarland University, Building 61, 66424, Homburg, Saar, Germany
| | - Anja Beckmann
- Department of Anatomy and Cell Biology, Saarland University, Building 61, 66424, Homburg, Saar, Germany
| | - Thomas Tschernig
- Department of Anatomy and Cell Biology, Saarland University, Building 61, 66424, Homburg, Saar, Germany
| | - Carola Meier
- Department of Anatomy and Cell Biology, Saarland University, Building 61, 66424, Homburg, Saar, Germany.
| |
Collapse
|
33
|
Putative role of border cells in generating spontaneous morphological activity within Kölliker's organ. Hear Res 2015; 330:90-7. [PMID: 26119178 DOI: 10.1016/j.heares.2015.06.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 05/21/2015] [Accepted: 06/23/2015] [Indexed: 11/21/2022]
Abstract
Kölliker's organ is a transient epithelial structure, comprising a major part of the organ of Corti during pre-hearing stages of development. The auditory system is spontaneously active during development, which serves to retain and refine neural connections. Kölliker's organ is considered a key candidate for generating such spontaneous activity, most likely through purinergic (P2 receptor) signalling and inner hair cell (IHC) activation. Associated with the spontaneous neural activity, ATP released locally by epithelial cells induces rhythmic morphological changes within Kölliker's organ, the purpose of which is not understood. These changes are accompanied by a shift in cellular refractive index, allowing optical detection of this activity in real-time. Using this principle, we investigated the origin of spontaneous morphological activity within Kölliker's organ. Apical turns of Wistar rat cochleae (P9-11) were dissected, and the purinergic involvement was studied following acute tissue exposure to a P2 receptor agonist (ATPγS) and antagonist (suramin). ATPγS induced a sustained darkening throughout Kölliker's organ, reversed by suramin. This effect was most pronounced in the region closest to the inner hair cells, which also displayed the highest frequency of intrinsic morphological events. Additionally, suramin alone induced swelling of this region, suggesting a tight regulation of cell volume by ATP-mediated mechanisms. Histological analysis of cochlear tissues demonstrates the most profound volume changes in the border cell region immediately adjacent to the IHCs. Together, these results underline the role of purinergic signalling in initiating morphological events within Kölliker's organ, and suggest a key involvement of border cells surrounding IHCs in regulating this spontaneous activity.
Collapse
|
34
|
Pannexin1 channels dominate ATP release in the cochlea ensuring endocochlear potential and auditory receptor potential generation and hearing. Sci Rep 2015; 5:10762. [PMID: 26035172 PMCID: PMC4451810 DOI: 10.1038/srep10762] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 04/28/2015] [Indexed: 01/04/2023] Open
Abstract
Pannexin1 (Panx1) is a gap junction gene in vertebrates whose proteins mainly function as non-junctional channels on the cell surface. Panx1 channels can release ATP under physiological conditions and play critical roles in many physiological and pathological processes. Here, we report that Panx1 deficiency can reduce ATP release and endocochlear potential (EP) generation in the cochlea inducing hearing loss. Panx1 extensively expresses in the cochlea, including the cochlear lateral wall. We found that deletion of Panx1 in the cochlear lateral wall almost abolished ATP release under physiological conditions. Positive EP is a driving force for current through hair cells to produce auditory receptor potential. EP generation requires ATP. In the Panx1 deficient mice, EP and auditory receptor potential as measured by cochlear microphonics (CM) were significantly reduced. However, no apparent hair cell loss was detected. Moreover, defect of connexin hemichannels by deletion of connexin26 (Cx26) and Cx30, which are predominant connexin isoforms in the cochlea, did not reduce ATP release under physiological conditions. These data demonstrate that Panx1 channels dominate ATP release in the cochlea ensuring EP and auditory receptor potential generation and hearing. Panx1 deficiency can reduce ATP release and EP generation causing hearing loss.
Collapse
|
35
|
Li L, He L, Wu D, Chen L, Jiang Z. Pannexin-1 channels and their emerging functions in cardiovascular diseases. Acta Biochim Biophys Sin (Shanghai) 2015; 47:391-6. [PMID: 25921414 DOI: 10.1093/abbs/gmv028] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Accepted: 02/04/2015] [Indexed: 11/15/2022] Open
Abstract
Pannexin-1, Pannexin-2, and Pannexin-3 are three members of the Pannexin family of channel-forming glycoprotein. Their primary function is defined by their ability to form single-membrane channels. Pannexin-1 ubiquitously exists in many cells and organs throughout the body and is specially distributed in the circulatory system, while the expressions of Pannexin-2 and Pannexin-3 are mostly restricted to organs and tissues. Pannexin-1 oligomers have been shown to be functional single membrane channels that connect intracellular and extracellular compartments and are not intercellular channels in appositional membranes. The physiological functions of Pannexin-1 are to link to the adenosine triphosphate efflux that acts as a paracrine signal, and regulate cellular inflammasomes in a variety of cell types under physiological and pathophysiological conditions. However, there are still many functions to be explored. This review summarizes recent reports and discusses the role of Pannexin-1 in cardiovascular diseases, including ischemia, arrhythmia, cardiac fibrosis, and hypertension. Pannexin-1 has been suggested as an exciting, clinically relevant target in cardiovascular diseases.
Collapse
Affiliation(s)
- Lanfang Li
- Post-doctoral Mobile Stations for Basic Medicine, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang 421001, China Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, University of South China, Hengyang 421001, China
| | - Lu He
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, University of South China, Hengyang 421001, China
| | - Di Wu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, University of South China, Hengyang 421001, China
| | - Linxi Chen
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, University of South China, Hengyang 421001, China
| | - Zhisheng Jiang
- Post-doctoral Mobile Stations for Basic Medicine, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang 421001, China
| |
Collapse
|
36
|
Zhao HB, Zhu Y, Liang C, Chen J. Pannexin 1 deficiency can induce hearing loss. Biochem Biophys Res Commun 2015; 463:143-7. [PMID: 26002464 DOI: 10.1016/j.bbrc.2015.05.049] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 05/10/2015] [Indexed: 11/19/2022]
Abstract
Gap junctions play a critical role in hearing. Connexin gap junction gene mutations can induce a high incidence of hearing loss. Pannexin (Panx) gene also encodes gap junction proteins in vertebrates. Panx1 is a predominant pannexin isoform and has extensive expression in the cochlea. Here, we report that deletion of Panx1 in the cochlea could produce a progressive hearing loss. The auditory brainstem response (ABR) recording showed that hearing loss was moderate to severe and severe at high-frequencies. Distortion product otoacoustic emission (DPOAE), which reflects the activity of active cochlear mechanics that can amply acoustic stimulation to enhance hearing sensitivity and frequency selectivity, was also reduced. We further found that Panx1 deficiency could activate Caspase-3 cell apoptotic pathway in the cochlea to cause hair cells and other types of cells degeneration. These data indicate that like connexins Panx1 deficiency can also induce hearing loss. These data also suggest that pannexins play important rather than redundant roles in the cochlea and hearing.
Collapse
Affiliation(s)
- Hong-Bo Zhao
- Department of Otolaryngology, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, United States.
| | - Yan Zhu
- Department of Otolaryngology, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, United States
| | - Chun Liang
- Department of Otolaryngology, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, United States
| | - Jin Chen
- Department of Otolaryngology, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, United States
| |
Collapse
|
37
|
Boassa D, Nguyen P, Hu J, Ellisman MH, Sosinsky GE. Pannexin2 oligomers localize in the membranes of endosomal vesicles in mammalian cells while Pannexin1 channels traffic to the plasma membrane. Front Cell Neurosci 2015; 8:468. [PMID: 25698922 PMCID: PMC4313697 DOI: 10.3389/fncel.2014.00468] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 12/27/2014] [Indexed: 12/13/2022] Open
Abstract
Pannexin2 (Panx2) is the largest of three members of the pannexin proteins. Pannexins are topologically related to connexins and innexins, but serve different functional roles than forming gap junctions. We previously showed that pannexins form oligomeric channels but unlike connexins and innexins, they form only single membrane channels. High levels of Panx2 mRNA and protein in the Central Nervous System (CNS) have been documented. Whereas Pannexin1 (Panx1) is fairly ubiquitous and Pannexin3 (Panx3) is found in skin and connective tissue, both are fully glycosylated, traffic to the plasma membrane and have functions correlated with extracellular ATP release. Here, we describe trafficking and subcellular localizations of exogenous Panx2 and Panx1 protein expression in MDCK, HeLa, and HEK 293T cells as well as endogenous Panx1 and Panx2 patterns in the CNS. Panx2 was found in intracellular localizations, was partially N-glycosylated, and localizations were non-overlapping with Panx1. Confocal images of hippocampal sections immunolabeled for the astrocytic protein GFAP, Panx1 and Panx2 demonstrated that the two isoforms, Panx1 and Panx2, localized at different subcellular compartments in both astrocytes and neurons. Using recombinant fusions of Panx2 with appended genetic tags developed for correlated light and electron microscopy and then expressed in different cell lines, we determined that Panx2 is localized in the membrane of intracellular vesicles and not in the endoplasmic reticulum as initially indicated by calnexin colocalization experiments. Dual immunofluorescence imaging with protein markers for specific vesicle compartments showed that Panx2 vesicles are early endosomal in origin. In electron tomographic volumes, cross-sections of these vesicles displayed fine structural details and close proximity to actin filaments. Thus, pannexins expressed at different subcellular compartments likely exert distinct functional roles, particularly in the nervous system.
Collapse
Affiliation(s)
- Daniela Boassa
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California San Diego, La Jolla, CA, USA
| | - Phuong Nguyen
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California San Diego, La Jolla, CA, USA
| | - Junru Hu
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California San Diego, La Jolla, CA, USA
| | - Mark H Ellisman
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California San Diego, La Jolla, CA, USA ; Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Gina E Sosinsky
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California San Diego, La Jolla, CA, USA ; Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
38
|
Kurtenbach S, Kurtenbach S, Zoidl G. Emerging functions of pannexin 1 in the eye. Front Cell Neurosci 2014; 8:263. [PMID: 25309318 PMCID: PMC4163987 DOI: 10.3389/fncel.2014.00263] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 08/14/2014] [Indexed: 01/23/2023] Open
Abstract
Pannexin 1 (Panx1) is a high-conductance, voltage-gated channel protein found in vertebrates. Panx1 is widely expressed in many organs and tissues, including sensory systems. In the eye, Panx1 is expressed in major divisions including the retina, lens and cornea. Panx1 is found in different neuronal and non-neuronal cell types. The channel is mechanosensitive and responds to changes in extracellular ATP, intracellular calcium, pH, or ROS/nitric oxide. Since Panx1 channels operate at the crossroad of major signaling pathways, physiological functions in important autocrine and paracrine feedback signaling mechanisms were hypothesized. This review starts with describing in depth the initial Panx1 expression and localization studies fostering functional studies that uncovered distinct roles in processing visual information in subsets of neurons in the rodent and fish retina. Panx1 is expressed along the entire anatomical axis from optical nerve to retina and cornea in glia, epithelial and endothelial cells as well as in neurons. The expression and diverse localizations throughout the eye points towards versatile functions of Panx1 in neuronal and non-neuronal cells, implicating Panx1 in the crosstalk between immune and neural cells, pressure related pathological conditions like glaucoma, wound repair or neuronal cell death caused by ischemia. Summarizing the literature on Panx1 in the eye highlights the diversity of emerging Panx1 channel functions in health and disease.
Collapse
Affiliation(s)
- Sarah Kurtenbach
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada
| | - Stefan Kurtenbach
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada
| | - Georg Zoidl
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada ; Department of Biology, Faculty of Science, York University Toronto, ON, Canada
| |
Collapse
|
39
|
Kurtenbach S, Whyte-Fagundes P, Gelis L, Kurtenbach S, Brazil E, Zoidl C, Hatt H, Shestopalov VI, Zoidl G. Investigation of olfactory function in a Panx1 knock out mouse model. Front Cell Neurosci 2014; 8:266. [PMID: 25309319 PMCID: PMC4162419 DOI: 10.3389/fncel.2014.00266] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 08/18/2014] [Indexed: 01/01/2023] Open
Abstract
Pannexin 1 (Panx1), the most extensively investigated member of a channel-forming protein family, is able to form pores conducting molecules up to 1.5 kDa, like ATP, upon activation. In the olfactory epithelium (OE), ATP modulates olfactory responsiveness and plays a role in proliferation and differentiation of olfactory sensory neurons (OSNs). This process continuously takes place in the OE, as neurons are replaced throughout the whole lifespan. The recent discovery of Panx1 expression in the OE raises the question whether Panx1 mediates ATP release responsible for modulating chemosensory function. In this study, we analyzed pannexin expression in the OE and a possible role of Panx1 in olfactory function using a Panx1−/− mouse line with a global ablation of Panx1. This mouse model has been previously used to investigate Panx1 functions in the retina and adult hippocampus. Here, qPCR, in-situ hybridization, and immunohistochemistry (IHC) demonstrated that Panx1 is expressed in axon bundles deriving from sensory neurons of the OE. The localization, distribution, and expression of major olfactory signal transduction proteins were not significantly altered in Panx1−/− mice. Further, functional analysis of Panx1−/− animals does not reveal any major impairment in odor perception, indicated by electroolfactogram (EOG) measurements and behavioral testing. However, ATP release evoked by potassium gluconate application was reduced in Panx1−/− mice. This result is consistent with previous reports on ATP release in isolated erythrocytes and spinal or lumbar cord preparations from Panx1−/− mice, suggesting that Panx1 is one of several alternative pathways to release ATP in the olfactory system.
Collapse
Affiliation(s)
- Stefan Kurtenbach
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada
| | - Paige Whyte-Fagundes
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada
| | - Lian Gelis
- Department of Cell Physiology, Ruhr University Bochum Bochum, Germany
| | - Sarah Kurtenbach
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada
| | - Emerson Brazil
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada
| | - Christiane Zoidl
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada
| | - Hanns Hatt
- Department of Cell Physiology, Ruhr University Bochum Bochum, Germany
| | - Valery I Shestopalov
- Department of Ophthalmology, Bascom Palmer Eye Institute, Miller School of Medicine, University of Miami Miami, FL, USA ; Vavilov Institute of General Genetics, Russian Academy of Sciences Moscow, Russia
| | - Georg Zoidl
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada
| |
Collapse
|
40
|
Kölliker's organ and the development of spontaneous activity in the auditory system: implications for hearing dysfunction. BIOMED RESEARCH INTERNATIONAL 2014; 2014:367939. [PMID: 25210710 PMCID: PMC4156998 DOI: 10.1155/2014/367939] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 08/07/2014] [Indexed: 11/25/2022]
Abstract
Prior to the “onset of hearing,” developing cochlear inner hair cells (IHCs) and primary auditory neurons undergo experience-independent activity, which is thought to be important in retaining and refining neural connections in the absence of sound. One of the major hypotheses regarding the origin of such activity involves a group of columnar epithelial supporting cells forming Kölliker's organ, which is only present during this critical period of auditory development. There is strong evidence for a purinergic signalling mechanism underlying such activity. ATP released through connexin hemichannels may activate P2 purinergic receptors in both Kölliker's organ and the adjacent IHCs, leading to generation of electrical activity throughout the auditory system. However, recent work has suggested an alternative origin, by demonstrating the ability of IHCs to generate this spontaneous activity without activation by ATP. Regardless, developmental abnormalities of Kölliker's organ may lead to congenital hearing loss, considering that mutations in ion channels (hemichannels, gap junctions, and calcium channels) involved in Kölliker's organ activity share strong links with such types of deafness.
Collapse
|
41
|
Kwon TJ, Kim DB, Bae JW, Sagong B, Choi SY, Cho HJ, Kim UK, Lee KY. Molecular cloning, characterization, and expression of pannexin genes in chicken. Poult Sci 2014; 93:2253-61. [PMID: 25002553 DOI: 10.3382/ps.2013-03867] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pannexins (Panx) are a family of proteins that share sequences with the invertebrate gap junction proteins, innexins, and have a similar structure to that of the vertebrate gap junction proteins, connexins. To date, the Panx family consists of 3 members, but their genetic sequences have only been completely determined in a few vertebrate species. Moreover, expression of the Panx family has been reported in several rodent tissues: Panx1 is ubiquitously expressed in mammals, whereas Panx2 and Panx3 expressions are more restricted. Although members of the Panx family have been detected in mammals, their genetic sequences in avian species have not yet been fully elucidated. Here, we obtained the full-length mRNA sequences of chicken PANX genes and evaluated the homology of the amino acids from these sequences with those of other species. Furthermore, PANX gene expression in several chicken tissues was investigated based on mRNA levels. PANX1 was detected in the brain, cochlea, chondrocytes, eye, lung, skin, and intestine, and PANX2 was expressed in the brain, eye, and intestine. PANX3 was observed in the cochlea, chondrocytes, and bone. In addition, expression of PANX3 was higher than PANX1 in the cochlea. Immunofluorescent staining revealed PANX1 in hair cells, as well as the supporting cells, ganglion neurons, and the tegmentum vasculosum in chickens, whereas PANX3 was only detected in the bone surrounding the cochlea. Overall, the results of this study provide the first identification and characterization of the sequence and expression of the PANX family in an avian species, and fundamental data for confirmation of Panx function.
Collapse
Affiliation(s)
- Tae-Jun Kwon
- School of Life Sciences, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, Daegu, 702-701, South Korea
| | - Dong-Bin Kim
- School of Life Sciences, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, Daegu, 702-701, South Korea
| | - Jae Woong Bae
- School of Life Sciences, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, Daegu, 702-701, South Korea
| | - Borum Sagong
- School of Life Sciences, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, Daegu, 702-701, South Korea
| | - Soo-Young Choi
- Department of Medicine, University of Pennsylvania, Philadelphia 19104-4539
| | - Hyun-Ju Cho
- School of Life Sciences, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, Daegu, 702-701, South Korea
| | - Un-Kyung Kim
- School of Life Sciences, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, Daegu, 702-701, South Korea
| | - Kyu-Yup Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, School of Medicine, Kyungpook National University, Daegu, 700-721, South Korea
| |
Collapse
|
42
|
Pannexins form gap junctions with electrophysiological and pharmacological properties distinct from connexins. Sci Rep 2014; 4:4955. [PMID: 24828343 PMCID: PMC4021813 DOI: 10.1038/srep04955] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 04/22/2014] [Indexed: 11/17/2022] Open
Abstract
Stable expression of pannexin 1 (Panx1) and pannexin 3 (Panx3) resulted in functional gap junctions (GJs) in HeLa cells, but not in Neuro-2a (N2a) or PC-12 cells. The glycosylation pattern of expressed Panx1 varied greatly among different cell lines. In contrast to connexin (Cx) containing GJs (Cx-GJs), junctional conductance (Gj) of pannexin GJs (Panx-GJs) is very less sensitive to junctional voltage. Both Panx1 and Panx3 junctions favoured anionic dyes over cations to permeate. Though, carbenoxolone (CBX) and probenecid blocked Panx1 hemichannel activity, they had no effect on Panx1-GJs or Panx3-GJs. Extracellular loop 1 (E1) of Panx1 possibly bears the binding pocket. The Cx-GJ blocker heptanol blocked neither Panx1 hemichannel nor Panx-GJs. Unlike the GJs formed by most Cxs, CO2 did not uncouple Panx-GJs completely. Oxygen and glucose deprivation (OGD) caused lesser uncoupling of Panx-GJs compared to Cx43-GJs. These findings demonstrate properties of Panx-GJs that are distinctly different from Cx-GJs.
Collapse
|
43
|
Shoji KF, Sáez PJ, Harcha PA, Aguila HL, Sáez JC. Pannexin1 channels act downstream of P2X 7 receptors in ATP-induced murine T-cell death. Channels (Austin) 2014; 8:142-56. [PMID: 24590064 DOI: 10.4161/chan.28122] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Death of murine T cells induced by extracellular ATP is mainly triggered by activation of purinergic P2X 7 receptors (P2X 7Rs). However, a link between P2X 7Rs and pannexin1 (Panx1) channels, which are non-selective, has been recently demonstrated in other cell types. In this work, we characterized the expression and cellular distribution of pannexin family members (Panxs 1, 2 and 3) in isolated T cells. Panx1 was the main pannexin family member clearly detected in both helper (CD4+) and cytotoxic (CD8+) T cells, whereas low levels of Panx2 were found in both T-cell subsets. Using pharmacological and genetic approaches, Panx1 channels were found to mediate most ATP-induced ethidium uptake since this was drastically reduced by Panx1 channel blockers (10Panx1, Probenecid and low carbenoxolone concentration) and absent in T cells derived from Panx1-/- mice. Moreover, electrophysiological measurements in wild-type CD4+ cells treated with ATP unitary current events and pharmacological sensitivity compatible with Panx1 channels were found. In addition, ATP release from T cells treated with 4Br-A23187, a calcium ionophore, was completely blocked with inhibitors of both connexin hemichannels and Panx1 channels. Panx1 channel blockers drastically reduced the ATP-induced T-cell mortality, indicating that Panx1 channels mediate the ATP-induced T-cell death. However, mortality was not reduced in T cells of Panx1-/- mice, in which levels of P2X 7Rs and ATP-induced intracellular free Ca2+ responses were enhanced suggesting that P2X 7Rs take over Panx1 channels lose-function in mediating the onset of cell death induced by extracellular ATP.
Collapse
Affiliation(s)
- Kenji F Shoji
- Departamento de Fisiología; Pontificia Universidad Católica de Chile; Santiago, Chile
| | - Pablo J Sáez
- Departamento de Fisiología; Pontificia Universidad Católica de Chile; Santiago, Chile
| | - Paloma A Harcha
- Departamento de Fisiología; Pontificia Universidad Católica de Chile; Santiago, Chile
| | - Hector L Aguila
- Department of Immunology; University of Connecticut Health Center; Farmington, CT USA
| | - Juan C Sáez
- Departamento de Fisiología; Pontificia Universidad Católica de Chile; Santiago, Chile; Instituto Milenio; Centro Interdisciplinario de Neurociencias de Valparaíso; Valparaíso, Chile
| |
Collapse
|
44
|
Kurtenbach S, Kurtenbach S, Zoidl G. Gap junction modulation and its implications for heart function. Front Physiol 2014; 5:82. [PMID: 24578694 PMCID: PMC3936571 DOI: 10.3389/fphys.2014.00082] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 02/10/2014] [Indexed: 01/04/2023] Open
Abstract
Gap junction communication (GJC) mediated by connexins is critical for heart function. To gain insight into the causal relationship of molecular mechanisms of disease pathology, it is important to understand which mechanisms contribute to impairment of gap junctional communication. Here, we present an update on the known modulators of connexins, including various interaction partners, kinases, and signaling cascades. This gap junction network (GJN) can serve as a blueprint for data mining approaches exploring the growing number of publicly available data sets from experimental and clinical studies.
Collapse
Affiliation(s)
- Stefan Kurtenbach
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada
| | - Sarah Kurtenbach
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada
| | - Georg Zoidl
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada ; Department of Biology, Faculty of Science, York University Toronto, ON, Canada ; Center for Vision Research, York University Toronto, ON, Canada
| |
Collapse
|
45
|
Signal Transduction in Astrocytes during Chronic or Acute Treatment with Drugs (SSRIs, Antibipolar Drugs, GABA-ergic Drugs, and Benzodiazepines) Ameliorating Mood Disorders. JOURNAL OF SIGNAL TRANSDUCTION 2014; 2014:593934. [PMID: 24707399 PMCID: PMC3953578 DOI: 10.1155/2014/593934] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 12/16/2013] [Indexed: 01/29/2023]
Abstract
Chronic treatment with fluoxetine or other so-called serotonin-specific reuptake inhibitor antidepressants (SSRIs) or with a lithium salt “lithium”, carbamazepine, or valproic acid, the three classical antibipolar drugs, exerts a multitude of effects on astrocytes, which in turn modulate astrocyte-neuronal interactions and brain function. In the case of the SSRIs, they are to a large extent due to 5-HT2B-mediated upregulation and editing of genes. These alterations induce alteration in effects of cPLA2, GluK2, and the 5-HT2B receptor, probably including increases in both glucose metabolism and glycogen turnover, which in combination have therapeutic effect on major depression. The ability of increased levels of extracellular K+ to increase [Ca2+]i is increased as a sign of increased K+-induced excitability in astrocytes. Acute anxiolytic drug treatment with benzodiazepines or GABAA receptor stimulation has similar glycogenolysis-enhancing effects. The antibipolar drugs induce intracellular alkalinization in astrocytes with lithium acting on one acid extruder and carbamazepine and valproic acid on a different acid extruder. They inhibit K+-induced and transmitter-induced increase of astrocytic [Ca2+]i and thereby probably excitability. In several cases, they exert different changes in gene expression than SSRIs, determined both in cultured astrocytes and in freshly isolated astrocytes from drug-treated animals.
Collapse
|
46
|
Abstract
The pannexins (Panxs) are a family of chordate proteins homologous to the invertebrate gap junction forming proteins named innexins. Three distinct Panx paralogs (Panx1, Panx2, and Panx3) are shared among the major vertebrate phyla, but they appear to have suppressed (or even lost) their ability to directly couple adjacent cells. Connecting the intracellular and extracellular compartments is now widely accepted as Panx's primary function, facilitating the passive movement of ions and small molecules along electrochemical gradients. The tissue distribution of the Panxs ranges from pervasive to very restricted, depending on the paralog, and are often cell type-specific and/or developmentally regulated within any given tissue. In recent years, Panxs have been implicated in an assortment of physiological and pathophysiological processes, particularly with respect to ATP signaling and inflammation, and they are now considered to be a major player in extracellular purinergic communication. The following is a comprehensive review of the Panx literature, exploring the historical events leading up to their discovery, outlining our current understanding of their biochemistry, and describing the importance of these proteins in health and disease.
Collapse
Affiliation(s)
- Stephen R Bond
- Genome Technology Branch, Division of Intramural Research, National Human Genome Research Institute, National Institutes of Health Bethesda, MD, USA ; Department of Cellular and Physiological Science, Life Sciences Institute, University of British Columbia Vancouver, BC, Canada
| | - Christian C Naus
- Department of Cellular and Physiological Science, Life Sciences Institute, University of British Columbia Vancouver, BC, Canada
| |
Collapse
|
47
|
Chen J, Zhao HB. The role of an inwardly rectifying K(+) channel (Kir4.1) in the inner ear and hearing loss. Neuroscience 2014; 265:137-46. [PMID: 24480364 DOI: 10.1016/j.neuroscience.2014.01.036] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 01/16/2014] [Accepted: 01/18/2014] [Indexed: 11/18/2022]
Abstract
The KCNJ10 gene which encodes an inwardly rectifying K(+) channel Kir4.1 subunit plays an essential role in the inner ear and hearing. Mutations or deficiency of KCNJ10 can cause hearing loss with EAST or SeSAME syndromes. This review mainly focuses on the expression and function of Kir4.1 potassium channels in the inner ear and hearing. We first introduce general information about inwardly rectifying potassium (Kir) channels. Then, we review the expression and function of Kir4.1 channels in the inner ear, especially in endocochlear potential (EP) generation. Finally, we review KCNJ10 mutation-induced hearing loss and functional impairments. Kir4.1 is strongly expressed on the apical membrane of intermediate cells in the stria vascularis and in the satellite cells of cochlear ganglia. Functionally, Kir4.1 has critical roles in cochlear development and hearing through two distinct aspects of extracellular K(+) homeostasis: First, it participates in the generation and maintenance of EP and high K(+) concentration in the endolymph inside the scala media. Second, Kir4.1 is the major K(+) channel in satellite glial cells surrounding spiral ganglion neurons to sink K(+) ions expelled by the ganglion neurons during excitation. Kir4.1 deficiency leads to hearing loss with the absence of EP and spiral ganglion neuron degeneration. Deafness mutants show loss-of-function and reduced channel membrane-targeting and currents, which can be rescued upon by co-expression with wild-type Kir4.1. This review provides insights for further understanding Kir potassium channel function in the inner ear and the pathogenesis of deafness due to KCNJ10 deficiency, and also provides insights for developing therapeutic strategies targeting this deafness.
Collapse
Affiliation(s)
- J Chen
- Department of Morphology, Medical College of China Three Gorges University, Yichang, Hubei 443002, PR China; Department of Otolaryngology, University of Kentucky Medical Center, Lexington, KY 40536-0293, USA
| | - H-B Zhao
- Department of Otolaryngology, University of Kentucky Medical Center, Lexington, KY 40536-0293, USA.
| |
Collapse
|
48
|
Cloes M, Renson T, Johnen N, Thelen N, Thiry M. Differentiation of Boettcher’s cells during postnatal development of rat cochlea. Cell Tissue Res 2013; 354:707-16. [DOI: 10.1007/s00441-013-1705-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 07/12/2013] [Indexed: 11/30/2022]
|
49
|
Cone AC, Ambrosi C, Scemes E, Martone ME, Sosinsky GE. A comparative antibody analysis of pannexin1 expression in four rat brain regions reveals varying subcellular localizations. Front Pharmacol 2013; 4:6. [PMID: 23390418 PMCID: PMC3565217 DOI: 10.3389/fphar.2013.00006] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 01/09/2013] [Indexed: 11/13/2022] Open
Abstract
Pannexin1 (Panx1) channels release cytosolic ATP in response to signaling pathways. Panx1 is highly expressed in the central nervous system. We used four antibodies with different Panx1 anti-peptide epitopes to analyze four regions of rat brain. These antibodies labeled the same bands in Western blots and had highly similar patterns of immunofluorescence in tissue culture cells expressing Panx1, but Western blots of brain lysates from Panx1 knockout and control mice showed different banding patterns. Localizations of Panx1 in brain slices were generated using automated wide field mosaic confocal microscopy for imaging large regions of interest while retaining maximum resolution for examining cell populations and compartments. We compared Panx1 expression over the cerebellum, hippocampus with adjacent cortex, thalamus, and olfactory bulb. While Panx1 localizes to the same neuronal cell types, subcellular localizations differ. Two antibodies with epitopes against the intracellular loop and one against the carboxy terminus preferentially labeled cell bodies, while an antibody raised against an N-terminal peptide highlighted neuronal processes more than cell bodies. These labeling patterns may be a reflection of different cellular and subcellular localizations of full-length and/or modified Panx1 channels where each antibody is highlighting unique or differentially accessible Panx1 populations. However, we cannot rule out that one or more of these antibodies have specificity issues. All data associated with experiments from these four antibodies are presented in a manner that allows them to be compared and our claims thoroughly evaluated, rather than eliminating results that were questionable. Each antibody is given a unique identifier through the NIF Antibody Registry that can be used to track usage of individual antibodies across papers and all image and metadata are made available in the public repository, the Cell Centered Database, for on-line viewing, and download.
Collapse
Affiliation(s)
- Angela C Cone
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California San Diego, La Jolla, CA, USA
| | | | | | | | | |
Collapse
|
50
|
Abstract
In the developing nervous system, spontaneous neuronal activity arises independently of experience or any environmental input. This activity may play a major role in axonal pathfinding, refinement of topographic maps, dendritic morphogenesis, and the segregation of axonal terminal arbors. In the auditory system, endogenously released ATP in the cochlea activates inner hair cells to trigger bursts of action potentials (APs), which are transferred to the central auditory system. Here we show the modulatory role of purinergic signaling beyond the cochlea, i.e., the developmentally regulated and cell-type-specific depolarizing effects on auditory brainstem neurons of Mongolian gerbil. We assessed the effects of P2X receptors (P2XRs) on neuronal excitability from prehearing to early stages of auditory signal processing. Our results demonstrate that in neurons expressing P2XRs, extracellular ATP can evoke APs in sync with Ca(2+) signals. In cochlear nucleus (CN) bushy cells, ATP increases spontaneous and also acoustically evoked activity in vivo, but these effects diminish with maturity. Moreover, ATP not only augmented glutamate-driven firing, but it also evoked APs in the absence of glutamatergic transmission. In vivo recordings also revealed that endogenously released ATP in the CN contributes to neuronal firing activity by facilitating AP generation and prolonging AP duration. Given the enhancing effect of ATP on AP firing and confinement of P2XRs to certain auditory brainstem nuclei, and to distinct neurons within these nuclei, it is conceivable that purinergic signaling plays a specific role in the development of neuronal brainstem circuits.
Collapse
|