1
|
Polfer R, Furukawa H. Biology, function and structure of the calcium homeostasis modulator family. J Physiol 2024. [PMID: 39470434 DOI: 10.1113/jp285197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 09/24/2024] [Indexed: 10/30/2024] Open
Abstract
Calcium homeostasis modulators (CALHMs) are the most recently discovered members of the large-pore channel family. They mediate the conductance of ions and larger molecules, such as ATP, and play critical roles in pathways related to Alzheimer's disease, neuroinflammation, neuromodulation, taste perception and innate immune responses. Since the inaugural report on CALHM1 in 2008, significant breakthroughs have revealed their biological roles, ion and ATP channel functions, and structures, positioning the field for further advancements. In this review, we discuss the overall progress and recent developments in understanding the biological roles, functions and molecular structures of CALHM proteins.
Collapse
Affiliation(s)
- Rachel Polfer
- Cold Spring Harbor Laboratory, School of Biological Science at Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Hiro Furukawa
- Cold Spring Harbor Laboratory, School of Biological Science at Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| |
Collapse
|
2
|
Biane JS, Ladow MA, Fan A, Choi HS, Zhou LZ, Hassan S, Apodaca-Montano DL, Kwon AO, Bratsch-Prince JX, Kheirbek MA. Representations of stimulus meaning in the hippocampus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.14.618280. [PMID: 39464010 PMCID: PMC11507678 DOI: 10.1101/2024.10.14.618280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
The ability to discriminate and categorize the meaning of environmental stimuli and respond accordingly is essential for survival. The ventral hippocampus (vHPC) controls emotional and motivated behaviors in response to environmental cues and is hypothesized to do so in part by deciphering the positive or negative quality of these cues. Yet, what features of the environment are represented in the activity patterns of vCA1 neurons, and whether the positive or negative meaning of a stimulus is present at this stage, remains unclear. Here, using 2-photon calcium imaging across six different experimental paradigms, we consistently found that vCA1 ensembles encode the identity, sensory features, and intensity of learned and innately salient stimuli, but not their overall valence. These results offer a reappraisal of vCA1 function, wherein information corresponding to individual stimulus features and their behavioral saliency predominates, while valence-related information is attached elsewhere.
Collapse
Affiliation(s)
- Jeremy S. Biane
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Max A. Ladow
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Austin Fan
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Hye Sun Choi
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Lexi Zichen Zhou
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Shazreh Hassan
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Daniel L. Apodaca-Montano
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Andrew O. Kwon
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Joshua X. Bratsch-Prince
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Mazen A. Kheirbek
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
- Kavli Institute for Fundamental Neuroscience, Weill Institute for Neurosciences and Center for Integrative Neuroscience, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
3
|
Flammer LJ, Ellis H, Rivers N, Caronia L, Ghidewon MY, Christensen CM, Jiang P, Breslin PAS, Tordoff MG. Topical application of a P2X2/P2X3 purine receptor inhibitor suppresses the bitter taste of medicines and other taste qualities. Br J Pharmacol 2024; 181:3282-3299. [PMID: 38745397 DOI: 10.1111/bph.16411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 04/01/2024] [Accepted: 04/05/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND AND PURPOSE Many medications taste intensely bitter. The innate aversion to bitterness affects medical compliance, especially in children. There is a clear need to develop bitter blockers to suppress the bitterness of vital medications. Bitter taste is mediated by TAS2R receptors. Because different pharmaceutical compounds activate distinct sets of TAS2Rs, targeting specific receptors may only suppress bitterness for certain, but not all, bitter-tasting compounds. Alternative strategies are needed to identify universal bitter blockers that will improve the acceptance of every medication. Taste cells in the mouth transmit signals to afferent gustatory nerve fibres through the release of ATP, which activates the gustatory nerve-expressed purine receptors P2X2/P2X3. We hypothesized that blocking gustatory nerve transmission with P2X2/P2X3 inhibitors (e.g. 5-(5-iodo-4-methoxy-2-propan-2-ylphenoxy)pyrimidine-2,4-diamine [AF-353]) would reduce bitterness for all medications and bitter compounds. EXPERIMENTAL APPROACH Human sensory taste testing and mouse behavioural analyses were performed to determine if oral application of AF-353 blocks perception of bitter taste and other taste qualities but not non-gustatory oral sensations (e.g. tingle). KEY RESULTS Rinsing the mouth with AF-353 in humans or oral swabbing it in mice suppressed the bitter taste and avoidance behaviours of all compounds tested. We further showed that AF-353 suppressed other taste qualities (i.e. salt, sweet, sour and savoury) but had no effects on other oral or nasal sensations (e.g, astringency and oral tingle). CONCLUSION AND IMPLICATIONS This is the first time a universal, reversible taste blocker in humans has been reported. Topical application of P2X2/P2X3 inhibitor to suppress bitterness may improve medical compliance.
Collapse
Affiliation(s)
- Linda J Flammer
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
| | - Hillary Ellis
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
| | - Natasha Rivers
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
| | - Lauren Caronia
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
| | - Misgana Y Ghidewon
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
- University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Peihua Jiang
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
| | - Paul A S Breslin
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
- Department of Nutritional Sciences, Rutgers University, New Brunswick, New Jersey, USA
| | | |
Collapse
|
4
|
Sweet Taste Signaling: The Core Pathways and Regulatory Mechanisms. Int J Mol Sci 2022; 23:ijms23158225. [PMID: 35897802 PMCID: PMC9329783 DOI: 10.3390/ijms23158225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/23/2022] [Accepted: 07/25/2022] [Indexed: 12/10/2022] Open
Abstract
Sweet taste, a proxy for sugar-derived calories, is an important driver of food intake, and animals have evolved robust molecular and cellular machinery for sweet taste signaling. The overconsumption of sugar-derived calories is a major driver of obesity and other metabolic diseases. A fine-grained appreciation of the dynamic regulation of sweet taste signaling mechanisms will be required for designing novel noncaloric sweeteners with better hedonic and metabolic profiles and improved consumer acceptance. Sweet taste receptor cells express at least two signaling pathways, one mediated by a heterodimeric G-protein coupled receptor encoded by taste 1 receptor members 2 and 3 (TAS1R2 + TAS1R3) genes and another by glucose transporters and the ATP-gated potassium (KATP) channel. Despite these important discoveries, we do not fully understand the mechanisms regulating sweet taste signaling. We will introduce the core components of the above sweet taste signaling pathways and the rationale for having multiple pathways for detecting sweet tastants. We will then highlight the roles of key regulators of the sweet taste signaling pathways, including downstream signal transduction pathway components expressed in sweet taste receptor cells and hormones and other signaling molecules such as leptin and endocannabinoids.
Collapse
|
5
|
Finger T, Kinnamon S. Purinergic neurotransmission in the gustatory system. Auton Neurosci 2021; 236:102874. [PMID: 34536906 DOI: 10.1016/j.autneu.2021.102874] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/13/2021] [Accepted: 08/22/2021] [Indexed: 11/26/2022]
Abstract
Taste buds consist of specialized epithelial cells which detect particular tastants and synapse onto the afferent taste nerve innervating the endorgan. The nature of the neurotransmitter released by taste cells onto the nerve fiber was enigmatic early in this century although neurotransmitters for other sensory receptor cell types, e.g. hair cells, photoreceptors, was known for at least a decade. A 1999 paper by Burnstock and co-workers (Bo et al., 1999) showing the presence of P2X receptors on the afferent nerves served as a springboard for research that ultimately led to the discovery of ATP as the crucial neurotransmitter in the taste system (Finger et al., 2005). Subsequent work showed that a subpopulation of taste cells utilize a unique release channel, CALHM1/3, to release ATP in a voltage-dependent manner. Despite these advances, several aspects of purinergic transmission in this system remain to be elucidated.
Collapse
Affiliation(s)
- T Finger
- Dept. Cell & Developmental Biology, Dept. Otolaryngology, Univ. Colorado School of Medicine, Anschutz Medical Campus, MS 8108, Room L18-11118, RC-1, 12801 E. 17th Ave., Aurora, CO 80045, United States of America.
| | - Sue Kinnamon
- Dept. Cell & Developmental Biology, Dept. Otolaryngology, Univ. Colorado School of Medicine, Anschutz Medical Campus, MS 8108, Room L18-11118, RC-1, 12801 E. 17th Ave., Aurora, CO 80045, United States of America
| |
Collapse
|
6
|
Lin JY, Mukherjee N, Bernstein MJ, Katz DB. Perturbation of amygdala-cortical projections reduces ensemble coherence of palatability coding in gustatory cortex. eLife 2021; 10:e65766. [PMID: 34018924 PMCID: PMC8139825 DOI: 10.7554/elife.65766] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 04/30/2021] [Indexed: 01/01/2023] Open
Abstract
Taste palatability is centrally involved in consumption decisions-we ingest foods that taste good and reject those that don't. Gustatory cortex (GC) and basolateral amygdala (BLA) almost certainly work together to mediate palatability-driven behavior, but the precise nature of their interplay during taste decision-making is still unknown. To probe this issue, we discretely perturbed (with optogenetics) activity in rats' BLA→GC axons during taste deliveries. This perturbation strongly altered GC taste responses, but while the perturbation itself was tonic (2.5 s), the alterations were not-changes preferentially aligned with the onset times of previously-described taste response epochs, and reduced evidence of palatability-related activity in the 'late-epoch' of the responses without reducing the amount of taste identity information available in the 'middle epoch.' Finally, BLA→GC perturbations changed behavior-linked taste response dynamics themselves, distinctively diminishing the abruptness of ensemble transitions into the late epoch. These results suggest that BLA 'organizes' behavior-related GC taste dynamics.
Collapse
Affiliation(s)
- Jian-You Lin
- Department of PsychologyWalthamUnited States
- The Volen National Center for Complex Systems, Brandeis UniversityWalthamUnited States
| | - Narendra Mukherjee
- The Volen National Center for Complex Systems, Brandeis UniversityWalthamUnited States
| | - Max J Bernstein
- Department of PsychologyWalthamUnited States
- The Volen National Center for Complex Systems, Brandeis UniversityWalthamUnited States
| | - Donald B Katz
- Department of PsychologyWalthamUnited States
- The Volen National Center for Complex Systems, Brandeis UniversityWalthamUnited States
| |
Collapse
|
7
|
Taruno A, Nomura K, Kusakizako T, Ma Z, Nureki O, Foskett JK. Taste transduction and channel synapses in taste buds. Pflugers Arch 2020; 473:3-13. [PMID: 32936320 DOI: 10.1007/s00424-020-02464-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 07/29/2020] [Accepted: 09/07/2020] [Indexed: 12/31/2022]
Abstract
The variety of taste sensations, including sweet, umami, bitter, sour, and salty, arises from diverse taste cells, each of which expresses specific taste sensor molecules and associated components for downstream signal transduction cascades. Recent years have witnessed major advances in our understanding of the molecular mechanisms underlying transduction of basic tastes in taste buds, including the identification of the bona fide sour sensor H+ channel OTOP1, and elucidation of transduction of the amiloride-sensitive component of salty taste (the taste of sodium) and the TAS1R-independent component of sweet taste (the taste of sugar). Studies have also discovered an unconventional chemical synapse termed "channel synapse" which employs an action potential-activated CALHM1/3 ion channel instead of exocytosis of synaptic vesicles as the conduit for neurotransmitter release that links taste cells to afferent neurons. New images of the channel synapse and determinations of the structures of CALHM channels have provided structural and functional insights into this unique synapse. In this review, we discuss the current view of taste transduction and neurotransmission with emphasis on recent advances in the field.
Collapse
Affiliation(s)
- Akiyuki Taruno
- Department of Molecular Cell Physiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan. .,Japan Science and Technology Agency, PRESTO, Kawaguchi, Saitama, Japan.
| | - Kengo Nomura
- Department of Molecular Cell Physiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tsukasa Kusakizako
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Zhongming Ma
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Osamu Nureki
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - J Kevin Foskett
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
8
|
Singh N, Driessen AK, McGovern AE, Moe AAK, Farrell MJ, Mazzone SB. Peripheral and central mechanisms of cough hypersensitivity. J Thorac Dis 2020; 12:5179-5193. [PMID: 33145095 PMCID: PMC7578480 DOI: 10.21037/jtd-2020-icc-007] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Chronic cough is a difficult to treat symptom of many respiratory and some non-respiratory diseases, indicating that varied pathologies can underpin the development of chronic cough. However, clinically and experimentally it has been useful to collate these different pathological processes into the single unifying concept of cough hypersensitivity. Cough hypersensitivity syndrome is reflected by troublesome cough often precipitated by levels of stimuli that ordinarily don't cause cough in healthy people, and this appears to be a hallmark feature in many patients with chronic cough. Accordingly, a strong argument has emerged that changes in the excitability and/or normal regulation of the peripheral and central neural circuits responsible for cough are instrumental in establishing cough hypersensitivity and for causing excessive cough in disease. In this review, we explore the current peripheral and central neural mechanisms that are believed to be involved in altered cough sensitivity and present possible links to the mechanism of action of novel therapies that are currently undergoing clinical trials for chronic cough.
Collapse
Affiliation(s)
- Nabita Singh
- Department of Medical Imaging and Radiation Sciences, Monash University, Clayton, Australia
| | - Alexandria K. Driessen
- Department of Anatomy and Neuroscience, School of Biomedical Science, The University of Melbourne, Parkville, Australia
| | - Alice E. McGovern
- Department of Anatomy and Neuroscience, School of Biomedical Science, The University of Melbourne, Parkville, Australia
| | - Aung Aung Kywe Moe
- Department of Anatomy and Neuroscience, School of Biomedical Science, The University of Melbourne, Parkville, Australia
| | - Michael J. Farrell
- Department of Medical Imaging and Radiation Sciences, Monash University, Clayton, Australia
- Monash Biomedical Imaging, Monash University, Clayton, Australia
| | - Stuart B. Mazzone
- Department of Anatomy and Neuroscience, School of Biomedical Science, The University of Melbourne, Parkville, Australia
| |
Collapse
|
9
|
Iwamoto M, Takashima M, Ohtubo Y. A subset of taste receptor cells express biocytin-permeable channels activated by reducing extracellular Ca 2+ concentration. Eur J Neurosci 2020; 51:1605-1623. [PMID: 31912931 DOI: 10.1111/ejn.14672] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 12/03/2019] [Accepted: 01/02/2020] [Indexed: 12/11/2022]
Abstract
Taste receptor cells (type II cells) transmit taste information to taste nerve fibres via ATP-permeable channels, including calcium homeostasis modulator (CALHM), connexin and/or pannexin1 channels, via the paracrine release of adenosine triphosphate (ATP) as a predominant transmitter. In the present study, we demonstrate that extracellular Ca2+ -dependent biocytin-permeable channels are present in a subset of type II cells in mouse fungiform taste buds using biocytin uptake, immunohistochemistry and in situ whole-cell recordings. Type II cells were labelled with biocytin in an extracellular Ca2+ concentration ([Ca2+ ]out )-sensitive manner. We found that the ratio of biocytin-labelled type II cells to type II cells per taste bud was approximately 20% in 2 mM Ca2+ saline, and this ratio increased to approximately 50% in nominally Ca2+ -free saline. The addition of 300 µM GdCl3 , which inhibits various channels including CALHM1 channels, significantly inhibited biocytin labelling in nominally Ca2+ -free saline, whereas the addition of 20 µM ruthenium red did not. Moreover, Cs+ -insensitive currents increased in nominally Ca2+ -free saline in approximately 40% of type II cells. These increased currents appeared at a potential of above -35 mV, reversed at approximately +10 mV and increased with depolarization. These results suggest that biocytin labels type II cells via ion channels activated by [Ca2+ ]out reduction, probably "CALHM-like" channels, on the basolateral membrane and that taste receptor cells can be categorized into two groups based on differences in the expression levels of [Ca2+ ]out -dependent biocytin-permeable channels. These data indicate electrophysiological and pharmacologically relevant properties of biocytin-permeable channels and suggest their contributions to taste signal transduction.
Collapse
Affiliation(s)
- Masafumi Iwamoto
- Graduate school of Life Science and Systems Engineering, Kyushu Institute of Technology, Kitakyushu-shi, Japan
| | - Madoka Takashima
- Graduate school of Life Science and Systems Engineering, Kyushu Institute of Technology, Kitakyushu-shi, Japan
| | - Yoshitaka Ohtubo
- Graduate school of Life Science and Systems Engineering, Kyushu Institute of Technology, Kitakyushu-shi, Japan
| |
Collapse
|
10
|
Levitan D, Lin JY, Wachutka J, Mukherjee N, Nelson SB, Katz DB. Single and population coding of taste in the gustatory cortex of awake mice. J Neurophysiol 2019; 122:1342-1356. [PMID: 31339800 DOI: 10.1152/jn.00357.2019] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Electrophysiological analysis has revealed much about the broad coding and neural ensemble dynamics that characterize gustatory cortical (GC) taste processing in awake rats and about how these dynamics relate to behavior. With regard to mice, however, data concerning cortical taste coding have largely been restricted to imaging, a technique that reveals average levels of neural responsiveness but that (currently) lacks the temporal sensitivity necessary for evaluation of fast response dynamics; furthermore, the few extant studies have thus far failed to provide consensus on basic features of coding. We have recorded the spiking activity of ensembles of GC neurons while presenting representatives of the basic taste modalities (sweet, salty, sour, and bitter) to awake mice. Our first central result is the identification of similarities between rat and mouse taste processing: most mouse GC neurons (~66%) responded distinctly to multiple (3-4) tastes; temporal coding analyses further reveal, for the first time, that single mouse GC neurons sequentially code taste identity and palatability, the latter responses emerging ~0.5 s after the former, with whole GC ensembles transitioning suddenly and coherently from coding taste identity to coding taste palatability. The second finding is that spatial location plays very little role in any aspect of taste responses: neither between- (anterior-posterior) nor within-mouse (dorsal-ventral) mapping revealed anatomic regions with narrow or temporally simple taste responses. These data confirm recent results showing that mouse cortical taste responses are not "gustotopic" but also go beyond these imaging results to show that mice process tastes through time.NEW & NOTEWORTHY Here, we analyzed taste-related spiking activity in awake mouse gustatory cortical (GC) neural ensembles, revealing deep similarities between mouse cortical taste processing and that repeatedly demonstrated in rat: mouse GC ensembles code multiple aspects of taste in a coarse-coded, time-varying manner that is essentially invariant across the spatial extent of GC. These data demonstrate that, contrary to some reports, cortical network processing is distributed, rather than being separated out into spatial subregion.
Collapse
Affiliation(s)
- David Levitan
- Department of Biology, Brandeis University, Waltham, Massachusetts
| | - Jian-You Lin
- Department of Psychology, Brandeis University, Waltham, Massachusetts.,Volen Center for Complex Systems, Brandeis University, Waltham, Massachusetts
| | - Joseph Wachutka
- Department of Psychology, Brandeis University, Waltham, Massachusetts
| | | | - Sacha B Nelson
- Department of Biology, Brandeis University, Waltham, Massachusetts.,Volen Center for Complex Systems, Brandeis University, Waltham, Massachusetts
| | - Donald B Katz
- Department of Psychology, Brandeis University, Waltham, Massachusetts.,Volen Center for Complex Systems, Brandeis University, Waltham, Massachusetts
| |
Collapse
|
11
|
Pronin A, Pham D, An W, Dvoriantchikova G, Reshetnikova G, Qiao J, Kozhekbaeva Z, Reiser AE, Slepak VZ, Shestopalov VI. Inflammasome Activation Induces Pyroptosis in the Retina Exposed to Ocular Hypertension Injury. Front Mol Neurosci 2019; 12:36. [PMID: 30930743 PMCID: PMC6425693 DOI: 10.3389/fnmol.2019.00036] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Accepted: 01/29/2019] [Indexed: 12/22/2022] Open
Abstract
Mechanical stress and hypoxia during episodes of ocular hypertension (OHT) trigger glial activation and neuroinflammation in the retina. Glial activation and release of pro-inflammatory cytokines TNFα and IL-1β, complement, and other danger factors was shown to facilitate injury and loss of retinal ganglion cells (RGCs) that send visual information to the brain. However, cellular events linking neuroinflammation and neurotoxicity remain poorly characterized. Several pro-inflammatory and danger signaling pathways, including P2X7 receptors and Pannexin1 (Panx1) channels, are known to activate inflammasome caspases that proteolytically activate gasdermin D channel-formation to export IL-1 cytokines and/or induce pyroptosis. In this work, we used molecular and genetic approaches to map and characterize inflammasome complexes and detect pyroptosis in the OHT-injured retina. Acute activation of distinct inflammasome complexes containing NLRP1, NLRP3 and Aim2 sensor proteins was detected in RGCs, retinal astrocytes and Muller glia of the OHT-challenged retina. Inflammasome-mediated activation of caspases-1 and release of mature IL-1β were detected within 6 h and peaked at 12–24 h after OHT injury. These coincided with the induction of pyroptotic pore protein gasdermin D in neurons and glia in the ganglion cell layer (GCL) and inner nuclear layer (INL). The OHT-induced release of cytokines and RGC death were significantly decreased in the retinas of Casp1−/−Casp4(11)del, Panx1−/− and in Wild-type (WT) mice treated with the Panx1 inhibitor probenecid. Our results showed a complex spatio-temporal pattern of innate immune responses in the retina. Furthermore, they indicate an active contribution of neuronal NLRP1/NLRP3 inflammasomes and the pro-pyroptotic gasdermin D pathway to pathophysiology of the OHT injury. These results support the feasibility of inflammasome modulation for neuroprotection in OHT-injured retinas.
Collapse
Affiliation(s)
- Alexey Pronin
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Dien Pham
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Weijun An
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Galina Dvoriantchikova
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Galina Reshetnikova
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Jianzhong Qiao
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Zhanna Kozhekbaeva
- Department of Medicine, The Division of Hematology and Oncology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Ashlyn E Reiser
- Geisinger Commonwealth School of Medicine, Scranton, PA, United States
| | - Vladlen Z Slepak
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Valery I Shestopalov
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, United States.,Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, Russia.,Department of Cell Biology, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
12
|
Gupta A, Li X, DiCicco-Bloom E, Bello NT. Altered salt taste response and increased tongue epithelium Scnna1 expression in adult Engrailed-2 null mice. Physiol Behav 2018; 194:410-419. [PMID: 29953887 DOI: 10.1016/j.physbeh.2018.06.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 06/19/2018] [Accepted: 06/20/2018] [Indexed: 02/06/2023]
Abstract
Sensory impairments are critical for diagnosing and characterizing neurodevelopmental disorders. Taste is a sensory modality often not well characterized. Engrailed-2 (En2) is a transcription factor critical for neural development, and mice lacking En2 (En2-/-) display signs of impaired social interaction, cognitive processes (e.g., learning and memory, conditioned fear), and neurodevelopmental alterations. As such, En2-/- mice display the behavioral deficits and neural impairments characteristic of the core symptoms associated with autism spectrum disorder (ASD). The objective of this study was to characterize the taste function in En2-/- compared with En2+/+ in adult male mice. Measuring taste responsiveness by an automated gustometer, En2 null mice had decreased lick responses for 1.6 M fructose, whereas they demonstrated an increased taste responsivity (i.e., relative to water) at 0.3 M sodium chloride and 1 M monosodium glutamate. In a separate cohort of mice, En2-/- mice had an increased preference for sodium chloride over a range of concentrations (0.032-0.3 M) compared with En2+/+ mice. Regional gene expression of the tongue epithelium demonstrated an increase in Scnn1a, T2R140, T1R3, and Trpm5 and a decrease in Pkd1l3 in En2 null mice. Taken together, such data indicate that deficits in En2 can produce sensory impairments that can have a measurable impact on taste, particularly salt taste.
Collapse
Affiliation(s)
- Ankita Gupta
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Xinyi Li
- Nutritional Sciences Graduate Program, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Emanuel DiCicco-Bloom
- Department of Neuroscience and Cell Biology/Pediatrics, Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Nicholas T Bello
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA; Nutritional Sciences Graduate Program, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.
| |
Collapse
|
13
|
CALHM3 Is Essential for Rapid Ion Channel-Mediated Purinergic Neurotransmission of GPCR-Mediated Tastes. Neuron 2018; 98:547-561.e10. [PMID: 29681531 DOI: 10.1016/j.neuron.2018.03.043] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 02/26/2018] [Accepted: 03/26/2018] [Indexed: 11/21/2022]
Abstract
Binding of sweet, umami, and bitter tastants to G protein-coupled receptors (GPCRs) in apical membranes of type II taste bud cells (TBCs) triggers action potentials that activate a voltage-gated nonselective ion channel to release ATP to gustatory nerves mediating taste perception. Although calcium homeostasis modulator 1 (CALHM1) is necessary for ATP release, the molecular identification of the channel complex that provides the conductive ATP-release mechanism suitable for action potential-dependent neurotransmission remains to be determined. Here we show that CALHM3 interacts with CALHM1 as a pore-forming subunit in a CALHM1/CALHM3 hexameric channel, endowing it with fast voltage-activated gating identical to that of the ATP-release channel in vivo. Calhm3 is co-expressed with Calhm1 exclusively in type II TBCs, and its genetic deletion abolishes taste-evoked ATP release from taste buds and GPCR-mediated taste perception. Thus, CALHM3, together with CALHM1, is essential to form the fast voltage-gated ATP-release channel in type II TBCs required for GPCR-mediated tastes.
Collapse
|
14
|
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
|
15
|
Abstract
The past decade has witnessed a consolidation and refinement of the extraordinary progress made in taste research. This Review describes recent advances in our understanding of taste receptors, taste buds, and the connections between taste buds and sensory afferent fibres. The article discusses new findings regarding the cellular mechanisms for detecting tastes, new data on the transmitters involved in taste processing and new studies that address longstanding arguments about taste coding.
Collapse
|
16
|
WNT10A mutation causes ectodermal dysplasia by impairing progenitor cell proliferation and KLF4-mediated differentiation. Nat Commun 2017; 8:15397. [PMID: 28589954 PMCID: PMC5467248 DOI: 10.1038/ncomms15397] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Accepted: 03/27/2017] [Indexed: 02/06/2023] Open
Abstract
Human WNT10A mutations are associated with developmental tooth abnormalities and adolescent onset of a broad range of ectodermal defects. Here we show that β-catenin pathway activity and adult epithelial progenitor proliferation are reduced in the absence of WNT10A, and identify Wnt-active self-renewing stem cells in affected tissues including hair follicles, sebaceous glands, taste buds, nails and sweat ducts. Human and mouse WNT10A mutant palmoplantar and tongue epithelia also display specific differentiation defects that are mimicked by loss of the transcription factor KLF4. We find that β-catenin interacts directly with region-specific LEF/TCF factors, and with KLF4 in differentiating, but not proliferating, cells to promote expression of specialized keratins required for normal tissue structure and integrity. Our data identify WNT10A as a critical ligand controlling adult epithelial proliferation and region-specific differentiation, and suggest downstream β-catenin pathway activation as a potential approach to ameliorate regenerative defects in WNT10A patients. Human WNT10A mutations are associated with dental defects and adult onset ectodermal dysplasia. Xu et al. show that WNT10A-activated ß-catenin plays dual roles in adult epithelial progenitor proliferation and differentiation by complexing with KLF4 in differentiating, but not proliferating, cells.
Collapse
|
17
|
Abstract
Many people avidly consume foods and drinks containing caffeine, despite its bitter taste. Here, we review what is known about caffeine as a bitter taste stimulus. Topics include caffeine's action on the canonical bitter taste receptor pathway and caffeine's action on noncanonical receptor-dependent and -independent pathways in taste cells. Two conclusions are that (1) caffeine is a poor prototypical bitter taste stimulus because it acts on bitter taste receptor-independent pathways, and (2) caffeinated products most likely stimulate "taste" receptors in nongustatory cells. This review is relevant for taste researchers, manufacturers of caffeinated products, and caffeine consumers.
Collapse
Affiliation(s)
- Rachel L Poole
- Monell Chemical Senses Center, Philadelphia, Pennsylvania
| | | |
Collapse
|
18
|
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: 0.9] [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
|
19
|
Bigiani A. Calcium Homeostasis Modulator 1-Like Currents in Rat Fungiform Taste Cells Expressing Amiloride-Sensitive Sodium Currents. Chem Senses 2017; 42:343-359. [DOI: 10.1093/chemse/bjx013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
20
|
Ma Z, Saung WT, Foskett JK. Action potentials and ion conductances in wild-type and CALHM1-knockout type II taste cells. J Neurophysiol 2017; 117:1865-1876. [PMID: 28202574 DOI: 10.1152/jn.00835.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 02/09/2017] [Accepted: 02/09/2017] [Indexed: 11/22/2022] Open
Abstract
Taste bud type II cells fire action potentials in response to tastants, triggering nonvesicular ATP release to gustatory neurons via voltage-gated CALHM1-associated ion channels. Whereas CALHM1 regulates mouse cortical neuron excitability, its roles in regulating type II cell excitability are unknown. In this study, we compared membrane conductances and action potentials in single identified TRPM5-GFP-expressing circumvallate papillae type II cells acutely isolated from wild-type (WT) and Calhm1 knockout (KO) mice. The activation kinetics of large voltage-gated outward currents were accelerated in cells from Calhm1 KO mice, and their associated nonselective tail currents, previously shown to be highly correlated with ATP release, were completely absent in Calhm1 KO cells, suggesting that CALHM1 contributes to all of these currents. Calhm1 deletion did not significantly alter resting membrane potential or input resistance, the amplitudes and kinetics of Na+ currents either estimated from action potentials or recorded from steady-state voltage pulses, or action potential threshold, overshoot peak, afterhyperpolarization, and firing frequency. However, Calhm1 deletion reduced the half-widths of action potentials and accelerated the deactivation kinetics of transient outward currents, suggesting that the CALHM1-associated conductance becomes activated during the repolarization phase of action potentials.NEW & NOTEWORTHY CALHM1 is an essential ion channel component of the ATP neurotransmitter release mechanism in type II taste bud cells. Its contribution to type II cell resting membrane properties and excitability is unknown. Nonselective voltage-gated currents, previously associated with ATP release, were absent in cells lacking CALHM1. Calhm1 deletion was without effects on resting membrane properties or voltage-gated Na+ and K+ channels but contributed modestly to the kinetics of action potentials.
Collapse
Affiliation(s)
- Zhongming Ma
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Wint Thu Saung
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - J Kevin Foskett
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and.,Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
21
|
Gaillard D, Stratford JM. Measurement of Behavioral Taste Responses in Mice: Two-Bottle Preference, Lickometer, and Conditioned Taste-Aversion Tests. ACTA ACUST UNITED AC 2016; 6:380-407. [PMID: 27906463 DOI: 10.1002/cpmo.18] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The natural like and dislike of foods based on taste is one of the most easily observed behaviors in animals. Animals eat palatable foods and reject aversive foods, which makes measurement of taste perception possible using various behavioral techniques. Three different methods to accurately measure taste behavior are described here. First, two-bottle preference tests evaluate whether a taste compound (tastant) is preferred over water. Second, lickometer tests quantify the like and dislike for multiple concentrations of the same tastant or multiple tastants at the same time. Finally, conditioned taste aversion tests accurately determine the perceived taste threshold for palatable tastants. Together, these diverse methods enable researchers to observe and measure behavioral taste responses in mice to any tastant. © 2016 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Dany Gaillard
- Department of Cell and Developmental Biology and the Rocky Mountain Taste and Smell Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jennifer M Stratford
- Department of Cell and Developmental Biology and the Rocky Mountain Taste and Smell Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
22
|
Kovalzon VM, Moiseenko LS, Ambaryan AV, Kurtenbach S, Shestopalov VI, Panchin YV. Sleep-wakefulness cycle and behavior in pannexin1 knockout mice. Behav Brain Res 2016; 318:24-27. [PMID: 27769744 DOI: 10.1016/j.bbr.2016.10.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 10/04/2016] [Accepted: 10/07/2016] [Indexed: 01/13/2023]
Abstract
Pannexins are membrane channel proteins that play a role in a number of critical biological processes (Panchin et al., 2000; Shestopalov, Panchin, 2008). Among other cellular functions, pannexin hemichannels serve as purine nucleoside conduits providing ATP efflux into the extracellular space (Dahl, 2015), where it is rapidly degraded to adenosine. Pannexin1 (Panx1) is abundantly expressed in the brain and has been shown to contribute to adenosine signaling in nervous system tissues (Prochnow et al., 2012). We hypothesized that pannexin1 may contribute to sleep-wake cycle regulation through extracellular adenosine, a well-established paracrine factor in slow wave sleep. To investigate this link, EEG and movement activity throughout the light/dark cycle were compared in Panx1-/- and Panx1+/+ mice. We found a significant increase in waking and a correspondent decrease in slow wave sleep percentages in the Panx1-/- animals. These changes were especially pronounced during the dark period. Furthermore, we found a significant increase in movement activity of Panx1-/- mice. These findings are consistent with the hypothesis that extracellular adenosine is relatively depleted in Panx1-/- animals due to the absence of the ATP-permeable hemichannels. At the same time, sleep rebound after a 6-h sleep deprivation remained unchanged in Panx1-/- mice as compared to the control animals. Behavioral tests revealed that Panx1-/- mice were significantly faster during their descent along the vertical pole but more sluggish during their run through the horizontal pole as compared to the control mice.
Collapse
Affiliation(s)
- V M Kovalzon
- Severtsov Institute Ecology and Evolution, Russian Academy of Sciences, Moscow, Russia
| | - L S Moiseenko
- Severtsov Institute Ecology and Evolution, Russian Academy of Sciences, Moscow, Russia
| | - A V Ambaryan
- Severtsov Institute Ecology and Evolution, Russian Academy of Sciences, Moscow, Russia
| | - S Kurtenbach
- Bascom Palmer Eye Institute, University of Miami School Medicine, Miami, Florida, USA
| | - V I Shestopalov
- Bascom Palmer Eye Institute, University of Miami School Medicine, Miami, Florida, USA; Vavilov Institute for General Genetics, Russian Academy of Sciences, Moscow, Russia
| | - Y V Panchin
- Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, Russia; Belozersky Institute for Physicochemical Biology, Lomonosov Moscow State University, Moscow, Russia.
| |
Collapse
|
23
|
Molecular mechanism of sweetness sensation. Physiol Behav 2016; 164:453-463. [DOI: 10.1016/j.physbeh.2016.03.015] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 03/12/2016] [Accepted: 03/14/2016] [Indexed: 11/17/2022]
|
24
|
Recent Advances in Molecular Mechanisms of Taste Signaling and Modifying. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 323:71-106. [PMID: 26944619 DOI: 10.1016/bs.ircmb.2015.12.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The sense of taste conveys crucial information about the quality and nutritional value of foods before it is ingested. Taste signaling begins with taste cells via taste receptors in oral cavity. Activation of these receptors drives the transduction systems in taste receptor cells. Then particular transmitters are released from the taste cells and activate corresponding afferent gustatory nerve fibers. Recent studies have revealed that taste sensitivities are defined by distinct taste receptors and modulated by endogenous humoral factors in a specific group of taste cells. Such peripheral taste generations and modifications would directly influence intake of nutritive substances. This review will highlight current understanding of molecular mechanisms for taste reception, signal transduction in taste bud cells, transmission between taste cells and nerves, regeneration from taste stem cells, and modification by humoral factors at peripheral taste organs.
Collapse
|
25
|
Ma Z, Tanis JE, Taruno A, Foskett JK. Calcium homeostasis modulator (CALHM) ion channels. Pflugers Arch 2015; 468:395-403. [PMID: 26603282 DOI: 10.1007/s00424-015-1757-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 10/31/2015] [Indexed: 10/22/2022]
Abstract
Calcium homeostasis modulator 1 (CALHM1), formerly known as FAM26C, was recently identified as a physiologically important plasma membrane ion channel. CALHM1 and its Caenorhabditis elegans homolog, CLHM-1, are regulated by membrane voltage and extracellular Ca(2+) concentration ([Ca(2+)]o). In the presence of physiological [Ca(2+)]o (∼1.5 mM), CALHM1 and CLHM-1 are closed at resting membrane potentials but can be opened by strong depolarizations. Reducing [Ca(2+)]o increases channel open probability, enabling channel activation at negative membrane potentials. Together, voltage and Ca(2+) o allosterically regulate CALHM channel gating. Through convergent evolution, CALHM has structural features that are reminiscent of connexins and pannexins/innexins/LRRC8 (volume-regulated anion channel (VRAC)) gene families, including four transmembrane helices with cytoplasmic amino and carboxyl termini. A CALHM1 channel is a hexamer of CALHM1 monomers with a functional pore diameter of ∼14 Å. CALHM channels discriminate poorly among cations and anions, with signaling molecules including Ca(2+) and ATP able to permeate through its pore. CALHM1 is expressed in the brain where it plays an important role in cortical neuron excitability induced by low [Ca(2+)]o and in type II taste bud cells in the tongue that sense sweet, bitter, and umami tastes where it functions as an essential ATP release channel to mediate nonsynaptic neurotransmitter release. CLHM-1 is expressed in C. elegans sensory neurons and body wall muscles, and its genetic deletion causes locomotion defects. Thus, CALHM is a voltage- and Ca(2+) o-gated ion channel, permeable to large cations and anions, that plays important roles in physiology.
Collapse
Affiliation(s)
- Zhongming Ma
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, 720 Clinical Research Bldg., 415 Curie Blvd., Philadelphia, PA, 19104, USA.
| | - Jessica E Tanis
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, 720 Clinical Research Bldg., 415 Curie Blvd., Philadelphia, PA, 19104, USA
| | - Akiyuki Taruno
- Department of Molecular Cell Physiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - J Kevin Foskett
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, 720 Clinical Research Bldg., 415 Curie Blvd., Philadelphia, PA, 19104, USA. .,Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
26
|
Vandenbeuch A, Anderson CB, Kinnamon SC. Mice Lacking Pannexin 1 Release ATP and Respond Normally to All Taste Qualities. Chem Senses 2015; 40:461-7. [PMID: 26136251 DOI: 10.1093/chemse/bjv034] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Adenosine triphosphate (ATP) is required for the transmission of all taste qualities from taste cells to afferent nerve fibers. ATP is released from Type II taste cells by a nonvesicular mechanism and activates purinergic receptors containing P2X2 and P2X3 on nerve fibers. Several ATP release channels are expressed in taste cells including CALHM1, Pannexin 1, Connexin 30, and Connexin 43, but whether all are involved in ATP release is not clear. We have used a global Pannexin 1 knock out (Panx1 KO) mouse in a series of in vitro and in vivo experiments. Our results confirm that Panx1 channels are absent in taste buds of the knockout mice and that other known ATP release channels are not upregulated. Using a luciferin/luciferase assay, we show that circumvallate taste buds from Panx1 KO mice normally release ATP upon taste stimulation compared with wild type (WT) mice. Gustatory nerve recordings in response to various tastants applied to the tongue and brief-access behavioral testing with SC45647 also show no difference between Panx1 KO and WT. These results confirm that Panx1 is not required for the taste evoked release of ATP or for neural and behavioral responses to taste stimuli.
Collapse
Affiliation(s)
- Aurelie Vandenbeuch
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, CO, USA and Rocky Mountain Taste and Smell Center, University of Colorado School of Medicine, Aurora, CO, USA
| | - Catherine B Anderson
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, CO, USA and Rocky Mountain Taste and Smell Center, University of Colorado School of Medicine, Aurora, CO, USA
| | - Sue C Kinnamon
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, CO, USA and Rocky Mountain Taste and Smell Center, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|