1
|
Baumer-Harrison C, Patel S, Scott KA, Krause EG, de Kloet AD. Optical perturbation of Agtr1a-containing neurons and afferents within the caudal nucleus of the solitary tract modulates sodium intake. Physiol Behav 2024; 284:114624. [PMID: 38959991 DOI: 10.1016/j.physbeh.2024.114624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 06/30/2024] [Accepted: 06/30/2024] [Indexed: 07/05/2024]
Abstract
Angiotensin-II (Ang-II) production is driven by deviations in blood volume and osmolality, and serves the role of regulating blood pressure and fluid intake to maintain cardiovascular and hydromineral homeostasis. These actions are mediated by Ang-II acting on its type 1a receptor (AT1aR) within the central nervous system and periphery. Of relevance, AT1aR are expressed on sensory afferents responsible for conveying cardiovascular information to the nucleus of the solitary tract (NTS). We have previously determined that optical excitation of neurons and vagal afferents within the NTS that express AT1aR (referred to as NTSAT1aR) mimics the perception of increased vascular stretch and induces compensatory responses to restore blood pressure. Here, we test whether NTSAT1aR are also involved in the modulation of water and sodium intake. We directed the light-sensitive excitatory channelrhodopsin-2 (ChR2) or inhibitory halorhodopsin (Halo) to Agtr1a-containing neurons and measured water and sodium chloride (NaCl) intake in the presence and absence of optical stimulation within the NTS during various challenges to fluid homeostasis. Optical perturbation of NTSAT1aR modulates NaCl intake, such that excitation attenuates, whereas inhibition increases intake. This effect is only observed in the water-deprived condition, suggesting that NTSAT1aR are involved in the regulation of sodium intake during an imbalance in both the intracellular and extracellular fluid compartments. Furthermore, optical excitation of NTSAT1aR increases c-Fos expression within oxytocinergic neurons of the paraventricular nucleus of the hypothalamus (PVN), indicating that the regulation of sodium intake by NTSAT1aR may be mediated by oxytocin. Collectively, these results reveal that NTSAT1aR are sufficient and necessary to modulate sodium intake relative to perceived changes in vascular stretch.
Collapse
Affiliation(s)
- Caitlin Baumer-Harrison
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, 32611, United States; Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL, 32611, United States; Center for Smell and Taste, University of Florida, Gainesville, FL, 32611, United States; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States.
| | - Sagar Patel
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, 32611, United States
| | - Karen A Scott
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL, 32611, United States; Department of Pharmacodynamics, College of Pharmacy, University of Florida, 32611, United States; Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, United States
| | - Eric G Krause
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL, 32611, United States; Department of Pharmacodynamics, College of Pharmacy, University of Florida, 32611, United States; Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, United States
| | - Annette D de Kloet
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, 32611, United States; Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL, 32611, United States; Center for Smell and Taste, University of Florida, Gainesville, FL, 32611, United States; Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, United States
| |
Collapse
|
2
|
Jang H, Sharma AB, Dan U, Wong JH, Knight ZA, Garrison JL. Dysregulation of the fluid homeostasis system by aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.26.615271. [PMID: 39386575 PMCID: PMC11463352 DOI: 10.1101/2024.09.26.615271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Chronic dehydration is a leading cause of morbidity for the elderly, but how aging alters the fluid homeostasis system is not well understood. Here, we used a combination of physiologic, behavioral and circuit analyses to characterize how fluid balance is affected by aging in mice. We found that old mice have a primary defect in sensing and producing the anti-diuretic hormone vasopressin, which results in chronic dehydration. Recordings and manipulations of the thirst circuitry revealed that old mice retain the ability to sense systemic cues of dehydration but are impaired in detecting presystemic, likely oropharyngeal, cues generated during eating and drinking, resulting in disorganized drinking behavior on short timescales. Surprisingly, old mice had increased drinking and motivation after 24-hour water deprivation, indicating that aging does not result in a general impairment in the thirst circuit. These findings reveal how a homeostatic system undergoes coordinated changes during aging.
Collapse
Affiliation(s)
- Heeun Jang
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Alexis B. Sharma
- Department of Physiology; University of California San Francisco, San Francisco, CA 94158, USA
| | - Usan Dan
- Department of Physiology; University of California San Francisco, San Francisco, CA 94158, USA
| | - Jasmine H. Wong
- Department of Physiology; University of California San Francisco, San Francisco, CA 94158, USA
| | - Zachary A. Knight
- Department of Physiology; University of California San Francisco, San Francisco, CA 94158, USA
- Kavli Institute for Fundamental Neuroscience; University of California San Francisco, San Francisco, CA 94158, USA
- Howard Hughes Medical Institute; University of California San Francisco, San Francisco, CA 94158, USA
| | - Jennifer L. Garrison
- Buck Institute for Research on Aging, Novato, CA 94945, USA
- Center for Healthy Aging in Women, Novato, CA 94945, USA
- Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
- Leonard Davis School of Gerontology, University of Southern California; Los Angeles, CA 90089, USA
| |
Collapse
|
3
|
Elliott KB, Keefe MS, Rolloque JJS, Jiwan NC, Dunn RA, Luk HY, Sekiguchi Y. Relationships between Morning Thirst and Later Hydration Status and Total Water Intake. Nutrients 2024; 16:3212. [PMID: 39339812 PMCID: PMC11435390 DOI: 10.3390/nu16183212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/13/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
Purpose: To investigate the effects of thirst on later hydration status, total water intake (TWI-MA), and its potential sex differences. Methods: Twelve men (mean ± standard deviation; age: 21 ± 2 years; mass: 81.0 ± 15.9 kg) and twelve women (age: 22 ± 3 years; mass: 68.8 ± 15.2 kg) visited the laboratory in the morning (first thing in the morning) and afternoon (2:00-4:00 p.m.) for three consecutive days under a free-living condition. At each visit, urine osmolality (UOSM), urine specific gravity (USG), urine color (UCOL), body mass loss (BML), thirst, and plasma osmolality (POSM) were collected and analyzed. The participants recorded their food and fluid intake between the visits to determine TWI-MA. Linear regression was used to predict the effect of morning thirst on the afternoon hydration indices for all the participants, as well as for males and females separately. Results: Higher morning thirst predicted lower UOSM (r2 = 0.056, p = 0.045), USG (r2 = 0.096, p = 0.008), UCOL (r2 = 0.074, p = 0.021), and higher thirst (r2 = 0.074, p = 0.021) in the afternoon. However, morning thirst did not predict afternoon BML, POSM, or TWI-MA (p > 0.05). In males, higher morning thirst predicted lower afternoon UOSM (r2 = 0.130, p = 0.031) and USG (r2 = 0.153, p = 0.018). Additionally, higher morning thirst predicted higher TWI-MA (r2 = 0.154, p = 0.018) in females. Conclusions: Morning thirst had a negligible impact on later hydration status, specifically with afternoon urine indices. Furthermore, higher thirst sensation did not impact BML, POSM, or TWI-MA. However, thirst sensation minimally contributed to drinking behavior in females. Overall, individuals may not rely solely on thirst sensation to manipulate their drinking behavior to optimize their fluid balance during their daily lives due to the complexity of thirst mechanisms.
Collapse
Affiliation(s)
- Kelly B. Elliott
- Sports Performance Laboratory, Department of Kinesiology & Sport Management, Texas Tech University, Lubbock, TX 79409, USA; (K.B.E.); (M.S.K.); (J.-J.S.R.); (R.A.D.)
| | - Marcos S. Keefe
- Sports Performance Laboratory, Department of Kinesiology & Sport Management, Texas Tech University, Lubbock, TX 79409, USA; (K.B.E.); (M.S.K.); (J.-J.S.R.); (R.A.D.)
| | - Jan-Joseph S. Rolloque
- Sports Performance Laboratory, Department of Kinesiology & Sport Management, Texas Tech University, Lubbock, TX 79409, USA; (K.B.E.); (M.S.K.); (J.-J.S.R.); (R.A.D.)
| | - Nigel C. Jiwan
- Applied Physiology Laboratory, Department of Kinesiology & Sport Management, Texas Tech University, Lubbock, TX 79409, USA; (N.C.J.); (H.-Y.L.)
| | - Ryan A. Dunn
- Sports Performance Laboratory, Department of Kinesiology & Sport Management, Texas Tech University, Lubbock, TX 79409, USA; (K.B.E.); (M.S.K.); (J.-J.S.R.); (R.A.D.)
| | - Hui-Ying Luk
- Applied Physiology Laboratory, Department of Kinesiology & Sport Management, Texas Tech University, Lubbock, TX 79409, USA; (N.C.J.); (H.-Y.L.)
| | - Yasuki Sekiguchi
- Sports Performance Laboratory, Department of Kinesiology & Sport Management, Texas Tech University, Lubbock, TX 79409, USA; (K.B.E.); (M.S.K.); (J.-J.S.R.); (R.A.D.)
| |
Collapse
|
4
|
Santollo J, Daniels D. Fluid transitions. Neuropharmacology 2024; 256:110009. [PMID: 38823577 PMCID: PMC11184821 DOI: 10.1016/j.neuropharm.2024.110009] [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: 02/15/2024] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 06/03/2024]
Abstract
Water is critical for survival and thirst is a powerful way of ensuring that fluid levels remain in balance. Overconsumption, however, can have deleterious effects, therefore optimization requires a need to balance the drive for water with the satiation of that water drive. This review will highlight our current understanding of how thirst is both generated and quenched, with particular focus on the roles of angiotensin II, glucagon like-peptide 1, and estradiol in turning on and off the thirst drive. Our understanding of the roles these bioregulators play has benefited from modern behavioral analyses, which have improved the time resolution of intake measures, allowing for attention to the details of the patterns within a bout of intake. This has led to behavioral interpretation in ways that are helpful in understanding the many controls of water intake and has expanded our understanding beyond the dichotomy that something which increases water intake is simply a "stimulator" while something that decreases water intake is simply a "satiety" factor. Synthesizing the available information, we describe a framework in which thirst is driven directly by perturbations in fluid intake and indirectly modified by several bioregulators. This allows us to better highlight areas that are in need of additional attention to form a more comprehensive understanding of how the system transitions between states of thirst and satiety.
Collapse
Affiliation(s)
- Jessica Santollo
- Department of Biology, University of Kentucky, Lexington, KY 40506, USA.
| | - Derek Daniels
- Department of Biology, University at Buffalo, State University of New York, Buffalo, NY 14260, USA; Center for Ingestive Behavior Research, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| |
Collapse
|
5
|
Greene ES, Tabler TW, Orlowski SK, Dridi S. Effect of heat stress on the hypothalamic expression of water channel- and noncoding RNA biogenesis-related genes in modern broilers and their ancestor red jungle fowl. Brain Res 2024; 1830:148810. [PMID: 38365130 DOI: 10.1016/j.brainres.2024.148810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/12/2024] [Accepted: 02/13/2024] [Indexed: 02/18/2024]
Abstract
Genetic selection for high growth rate has resulted in spectacular progress in feed efficiency in chickens. As feed intake and water consumption (WC) are associated and both are affected by environmental conditions, we evaluated WC and its hypothalamic regulation in three broiler-based research lines and their ancestor jungle fowl (JF) under heat stress (HS) conditions. Slow growing ACRB, moderate growing 95RB, fast growing MRB, and JF were exposed to daily chronic cyclic HS (36 °C, 9 h/d) or thermoneutral temperature (24 °C). HS increased WC in the MRB only. Arginine vasopressin (AVP) mRNA levels were decreased by HS in the MRB. Within the renin-angiotensin-aldosterone system (RAAS) system, renin expression was increased by HS in the JF, ACRB, and 95RB, while angiotensin I-converting enzyme (ACE), angiotensin II receptors (type 1, AT1, and type 2, AT2) were affected by line. The expression of aquaporin (AQP2, 7, 9, 10, 11, and 12) genes was upregulated by HS, whereas AQP4 and AQP5 expressions were influenced by line. miRNA processing components (Dicer1, Ago2, Drosha) were significantly different among the lines, but were unaffected by HS. In summary, this is the first report showing the effect of HS on hypothalamic water channel- and noncoding RNA biogenesis-related genes in modern chicken populations and their ancestor JF. These results provide a novel framework for future research to identify new molecular mechanisms and signatures involved in water homeostasis and adaptation to HS.
Collapse
Affiliation(s)
- Elizabeth S Greene
- Department of Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Travis W Tabler
- Department of Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Sara K Orlowski
- Department of Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Sami Dridi
- Department of Poultry Science, University of Arkansas, Fayetteville, AR, United States.
| |
Collapse
|
6
|
Yang D, Wang Y, Qi T, Zhang X, Shen L, Ma J, Pang Z, Lal NK, McClatchy DB, Seradj SH, Leung VH, Wang K, Xie Y, Polli FS, Maximov A, Gonzalez OC, de Lecea L, Cline HT, Augustine V, Yates JR, Ye L. Phosphorylation of pyruvate dehydrogenase inversely associates with neuronal activity. Neuron 2024; 112:959-971.e8. [PMID: 38266644 PMCID: PMC11021214 DOI: 10.1016/j.neuron.2023.12.015] [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: 10/12/2022] [Revised: 08/24/2023] [Accepted: 12/21/2023] [Indexed: 01/26/2024]
Abstract
For decades, the expression of immediate early genes (IEGs) such as FOS has been the most widely used molecular marker representing neuronal activation. However, to date, there is no equivalent surrogate available for the decrease of neuronal activity. Here, we developed an optogenetic-based biochemical screen in which population neural activities can be controlled by light with single action potential precision, followed by unbiased phosphoproteomic profiling. We identified that the phosphorylation of pyruvate dehydrogenase (pPDH) inversely correlated with the intensity of action potential firing in primary neurons. In in vivo mouse models, monoclonal antibody-based pPDH immunostaining detected activity decreases across the brain, which were induced by a wide range of factors including general anesthesia, chemogenetic inhibition, sensory experiences, and natural behaviors. Thus, as an inverse activity marker (IAM) in vivo, pPDH can be used together with IEGs or other cell-type markers to profile and identify bi-directional neural dynamics induced by experiences or behaviors.
Collapse
Affiliation(s)
- Dong Yang
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Yu Wang
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Tianbo Qi
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Xi Zhang
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Leyao Shen
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jingrui Ma
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Department of Neurobiology, University of California San Diego, La Jolla, CA 92093, USA
| | - Zhengyuan Pang
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Neeraj K Lal
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Daniel B McClatchy
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Saba Heydari Seradj
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Verina H Leung
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Kristina Wang
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Yi Xie
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Filip S Polli
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Anton Maximov
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Hollis T Cline
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Vineet Augustine
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Department of Neurobiology, University of California San Diego, La Jolla, CA 92093, USA
| | - John R Yates
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Li Ye
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
7
|
Cao Y, Li R, Bai L. Vagal sensory pathway for the gut-brain communication. Semin Cell Dev Biol 2024; 156:228-243. [PMID: 37558522 DOI: 10.1016/j.semcdb.2023.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 06/07/2023] [Accepted: 07/20/2023] [Indexed: 08/11/2023]
Abstract
The communication between the gut and brain is crucial for regulating various essential physiological functions, such as energy balance, fluid homeostasis, immune response, and emotion. The vagal sensory pathway plays an indispensable role in connecting the gut to the brain. Recently, our knowledge of the vagal gut-brain axis has significantly advanced through molecular genetic studies, revealing a diverse range of vagal sensory cell types with distinct peripheral innervations, response profiles, and physiological functions. Here, we review the current understanding of how vagal sensory neurons contribute to gut-brain communication. First, we highlight recent transcriptomic and genetic approaches that have characterized different vagal sensory cell types. Then, we focus on discussing how different subtypes encode numerous gut-derived signals and how their activities are translated into physiological and behavioral regulations. The emerging insights into the diverse cell types and functional properties of vagal sensory neurons have paved the way for exciting future directions, which may provide valuable insights into potential therapeutic targets for disorders involving gut-brain communication.
Collapse
Affiliation(s)
- Yiyun Cao
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Rui Li
- Chinese Institute for Brain Research, Beijing 102206, China; State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China
| | - Ling Bai
- Chinese Institute for Brain Research, Beijing 102206, China.
| |
Collapse
|
8
|
Stocker SD, Kinsman BJ, Farquhar WB, Gyarmati G, Peti-Peterdi J, Sved AF. Physiological Mechanisms of Dietary Salt Sensing in the Brain, Kidney, and Gastrointestinal Tract. Hypertension 2024; 81:447-455. [PMID: 37671571 PMCID: PMC10915107 DOI: 10.1161/hypertensionaha.123.19488] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2023]
Abstract
Excess dietary salt (NaCl) intake is strongly correlated with cardiovascular disease and is a major contributing factor to the pathogenesis of hypertension. NaCl-sensitive hypertension is a multisystem disorder that involves renal dysfunction, vascular abnormalities, and neurogenically-mediated increases in peripheral resistance. Despite a major research focus on organ systems and these effector mechanisms causing NaCl-induced increases in arterial blood pressure, relatively less research has been directed at elucidating how NaCl is sensed by various tissues to elicit these downstream effects. The purpose of this review is to discuss how the brain, kidney, and gastrointestinal tract sense NaCl including key cell types, the role of NaCl versus osmolality, and the underlying molecular and electrochemical mechanisms.
Collapse
Affiliation(s)
- Sean D. Stocker
- Department of Neurobiology, University of Pittsburgh School of Medicine
| | - Brian J Kinsman
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital
| | | | - Georgina Gyarmati
- Department of Physiology and Neuroscience and Medicine, Zilkha Neurogenetic Institute, University of Southern California
| | - Janos Peti-Peterdi
- Department of Physiology and Neuroscience and Medicine, Zilkha Neurogenetic Institute, University of Southern California
| | - Alan F. Sved
- Department of Neuroscience, University of Pittsburgh
| |
Collapse
|
9
|
Teckentrup V, Kroemer NB. Mechanisms for survival: vagal control of goal-directed behavior. Trends Cogn Sci 2024; 28:237-251. [PMID: 38036309 DOI: 10.1016/j.tics.2023.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 12/02/2023]
Abstract
Survival is a fundamental physiological drive, and neural circuits have evolved to prioritize actions that meet the energy demands of the body. This fine-tuning of goal-directed actions based on metabolic states ('allostasis') is deeply rooted in our brain, and hindbrain nuclei orchestrate the vital communication between the brain and body through the vagus nerve. Despite mounting evidence for vagal control of allostatic behavior in animals, its broader function in humans is still contested. Based on stimulation studies, we propose that the vagal afferent pathway supports transitions between survival modes by gating the integration of ascending bodily signals, thereby regulating reward-seeking. By reconceptualizing vagal signals as catalysts for goal-directed behavior, our perspective opens new avenues for theory-driven translational work in mental disorders.
Collapse
Affiliation(s)
- Vanessa Teckentrup
- Department of Psychiatry and Psychotherapy, Tübingen Center for Mental Health, University of Tübingen, 72076 Tübingen, Germany; School of Psychology and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Nils B Kroemer
- Department of Psychiatry and Psychotherapy, Tübingen Center for Mental Health, University of Tübingen, 72076 Tübingen, Germany; Section of Medical Psychology, Department of Psychiatry and Psychotherapy, Faculty of Medicine, University of Bonn, 53127 Bonn, Germany; German Center for Mental Health (DZPG), 72076 Tübingen, Germany.
| |
Collapse
|
10
|
Matsuda T, Kobayashi K, Kobayashi K, Noda M. Two parabrachial Cck neurons involved in the feedback control of thirst or salt appetite. Cell Rep 2024; 43:113619. [PMID: 38157299 DOI: 10.1016/j.celrep.2023.113619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 11/20/2023] [Accepted: 12/08/2023] [Indexed: 01/03/2024] Open
Abstract
Thirst and salt appetite are temporarily suppressed after water and salt ingestion, respectively, before absorption; however, the underlying neural mechanisms remain unclear. The parabrachial nucleus (PBN) is the relay center of ingestion signals from the digestive organs. We herein identify two distinct neuronal populations expressing cholecystokinin (Cck) mRNA in the lateral PBN that are activated in response to water and salt intake, respectively. The two Cck neurons in the dorsal-lateral compartment of the PBN project to the median preoptic nucleus and ventral part of the bed nucleus of the stria terminalis, respectively. The optogenetic stimulation of respective Cck neurons suppresses thirst or salt appetite under water- or salt-depleted conditions. The combination of optogenetics and in vivo Ca2+ imaging during ingestion reveals that both Cck neurons control GABAergic neurons in their target nuclei. These findings provide the feedback mechanisms for the suppression of thirst and salt appetite after ingestion.
Collapse
Affiliation(s)
- Takashi Matsuda
- Homeostatic Mechanism Research Unit, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Kanagawa 226-8503, Japan
| | - Kenta Kobayashi
- Section of Viral Vector Development, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Fukushima 960-1295, Japan
| | - Masaharu Noda
- Homeostatic Mechanism Research Unit, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Kanagawa 226-8503, Japan.
| |
Collapse
|
11
|
Smith J, Honig-Frand A, Antila H, Choi A, Kim H, Beier KT, Weber F, Chung S. Regulation of stress-induced sleep fragmentation by preoptic glutamatergic neurons. Curr Biol 2024; 34:12-23.e5. [PMID: 38096820 PMCID: PMC10872481 DOI: 10.1016/j.cub.2023.11.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 09/28/2023] [Accepted: 11/15/2023] [Indexed: 01/11/2024]
Abstract
Sleep disturbances are detrimental to our behavioral and emotional well-being. Stressful events disrupt sleep, in particular by inducing brief awakenings (microarousals, MAs), resulting in sleep fragmentation. The preoptic area of the hypothalamus (POA) is crucial for sleep control. However, how POA neurons contribute to the regulation of MAs and thereby impact sleep quality is unknown. Using fiber photometry in mice, we examine the activity of genetically defined POA subpopulations during sleep. We find that POA glutamatergic neurons are rhythmically activated in synchrony with an infraslow rhythm in the spindle band of the electroencephalogram during non-rapid eye movement sleep (NREMs) and are transiently activated during MAs. Optogenetic stimulation of these neurons promotes MAs and wakefulness. Exposure to acute social defeat stress fragments NREMs and significantly increases the number of transients in the calcium activity of POA glutamatergic neurons during NREMs. By reducing MAs, optogenetic inhibition during spontaneous sleep and after stress consolidates NREMs. Monosynaptically restricted rabies tracing reveals that POA glutamatergic neurons are innervated by brain regions regulating stress and sleep. In particular, presynaptic glutamatergic neurons in the lateral hypothalamus become activated after stress, and stimulating their projections to the POA promotes MAs and wakefulness. Our findings uncover a novel circuit mechanism by which POA excitatory neurons regulate sleep quality after stress.
Collapse
Affiliation(s)
- Jennifer Smith
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Adam Honig-Frand
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hanna Antila
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ashley Choi
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hannah Kim
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kevin T Beier
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA 92617, USA
| | - Franz Weber
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shinjae Chung
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
12
|
Salgado-Mozo S, Thirouin ZS, Wyrosdic JC, García-Hernández U, Bourque CW. Na X Channel Is a Physiological [Na +] Detector in Oxytocin- and Vasopressin-Releasing Magnocellular Neurosecretory Cells of the Rat Supraoptic Nucleus. J Neurosci 2023; 43:8306-8316. [PMID: 37783507 PMCID: PMC10711705 DOI: 10.1523/jneurosci.1203-23.2023] [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: 06/29/2023] [Revised: 09/08/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023] Open
Abstract
The Scn7A gene encodes NaX, an atypical noninactivating Na+ channel, whose expression in sensory circumventricular organs is essential to maintain homeostatic responses for body fluid balance. However, NaX has also been detected in homeostatic effector neurons, such as vasopressin (VP)-releasing magnocellular neurosecretory cells (MNCVP) that secrete VP (antidiuretic hormone) into the bloodstream in response to hypertonicity and hypernatremia. Yet, the physiological relevance of NaX expression in these effector cells remains unclear. Here, we show that rat MNCVP in males and females is depolarized and excited in proportion with isosmotic increases in [Na+]. These responses were caused by an inward current resulting from a cell-autonomous increase in Na+ conductance. The Na+-evoked current was unaffected by blockers of other Na+-permeable ion channels but was significantly reduced by shRNA-mediated knockdown of Scn7A expression. Furthermore, reducing the density of NaX channels selectively impaired the activation of MNCVP by systemic hypernatremia without affecting their responsiveness to hypertonicity in vivo These results identify NaX as a physiological Na+ sensor, whose expression in MNCVP contributes to the generation of homeostatic responses to hypernatremia.SIGNIFICANCE STATEMENT In this study, we provide the first direct evidence showing that the sodium-sensing channel encoded by the Scn7A gene (NaX) mediates cell-autonomous sodium detection by MNCs in the low millimolar range and that selectively reducing the expression of these channels in MNCs impairs their activation in response to a physiologically relevant sodium stimulus in vitro and in vivo These data reveal that NaX operates as a sodium sensor in these cells and that the endogenous sensory properties of osmoregulatory effector neurons contribute to their homeostatic activation in vivo.
Collapse
Affiliation(s)
- Sandra Salgado-Mozo
- Brain Repair and Integrative Neuroscience Program, Research Institute of McGill University Health Center, Montréal, Québec H3G1A4, Canada
- Department of Physiology, Biophysics and Neurosciences, Centre for Research and Advanced Studies, Instituto Politecnico Nacional, 07360 Mexico City, Mexico
| | - Zahra S Thirouin
- Brain Repair and Integrative Neuroscience Program, Research Institute of McGill University Health Center, Montréal, Québec H3G1A4, Canada
| | - Joshua C Wyrosdic
- Brain Repair and Integrative Neuroscience Program, Research Institute of McGill University Health Center, Montréal, Québec H3G1A4, Canada
| | - Ubaldo García-Hernández
- Department of Physiology, Biophysics and Neurosciences, Centre for Research and Advanced Studies, Instituto Politecnico Nacional, 07360 Mexico City, Mexico
| | - Charles W Bourque
- Brain Repair and Integrative Neuroscience Program, Research Institute of McGill University Health Center, Montréal, Québec H3G1A4, Canada
| |
Collapse
|
13
|
Picó-Munyoz R, Tárrega A, Laguna L. Origins of thirstiness sensation and current food solutions. Compr Rev Food Sci Food Saf 2023; 22:4433-4450. [PMID: 37583300 DOI: 10.1111/1541-4337.13229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 06/15/2023] [Accepted: 08/01/2023] [Indexed: 08/17/2023]
Abstract
The sensation of thirstiness is the desire to drink water. In certain situations, the ingestion of liquid water can be restricted. As a result, thirstiness is not relieved, resulting in an uncomfortable and distressing situation. The present review describes thirstiness and hydration, the food products and beverages that cause thirstiness, and the beverages and food products currently available to quench thirstiness in individuals with restricted access to liquid ingestion. It also discusses how to measure the effectiveness of calming thirstiness. To diminish thirstiness distress, different alternatives to liquids are proposed. Individuals with swallowing disorders are given thickened water, individuals with restricted water ingestion are given ice cubes or ice popsicles of different flavors, and sportspeople are given energy gels. However, current beverage solutions seem not to relieve thirst fully, although some stimuli like iced water, flavors (especially lemon and mint), or acids seem to work better than plain stimuli and could be added to existing products. Therefore, there is still a need to incorporate these strategies into beverage and food formulations and to test their effectiveness.
Collapse
Affiliation(s)
- Ruth Picó-Munyoz
- Consumer Perception and Behavior and Adapted Nutrition Laboratory, Institute of Agrochemistry and Food Technology (IATA, CSIC), Paterna, Spain
| | - Amparo Tárrega
- Consumer Perception and Behavior and Adapted Nutrition Laboratory, Institute of Agrochemistry and Food Technology (IATA, CSIC), Paterna, Spain
| | - Laura Laguna
- Consumer Perception and Behavior and Adapted Nutrition Laboratory, Institute of Agrochemistry and Food Technology (IATA, CSIC), Paterna, Spain
| |
Collapse
|
14
|
Aggrey SE, Ghareeb AFA, Milfort MC, Ariyo OW, Aryal B, Hartono E, Kwakye J, Sovi S, Hipple SA, Stevenson C, Fuller AL, El Sabry MI, Stino F, Rekaya R. Quantitative and molecular aspects of water intake in meat-type chickens. Poult Sci 2023; 102:102973. [PMID: 37633082 PMCID: PMC10474491 DOI: 10.1016/j.psj.2023.102973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/28/2023] Open
Abstract
Even though water is the most essential nutrient for poultry production, adequate data on individual water intake in broiler chickens and its relationship with other traits of economic importance is scant. Water is provided to chickens in an unrestricted manner in spite of being a finite resource. Climate change continues to affect water sources and efficient bird use of water is long overdue. Understanding the biological basis of water intake is essential for sustainability of the poultry industry. Individual water and feed intake, and growth data was collected on 520 commercial broilers aged 14 to 42 days. We introduced the concepts of water conversion ratio (WCR) and residual water intake (RWI) as parameters that can be used to assess water intake efficiency. Water conversion ratio was defined as the amount of water consumed per unit of body weight gain, and RWI was defined as the difference between the actual water intake (WI) of a given bird and the expected WI by an average bird from the population with the same metabolic body weight, feed intake (FI) and body weight gain (BWG). The correlation between WI and FI was positive (r=0.77; P<0.0001), and the correlation between WI and BWG was positive (r=0.80; P<0.0001). Based on the distribution of RWI, the bottom 5 birds (LRWI) and the top 5 birds (HRWI) for RWI were selected for mRNA expression differences. The average broiler consumed about 7.8 L (± 1L) of water from 14 to 42 days of age. The mRNA expression of arginine vasopressin (AVP) antidiuretic hormone, calcium sensing receptor (CasR), sodium channel epithelial 1 subunit alpha (SCNN1A) and SCNN1D in the hypothalamus was upregulated in the LRWI group compared to the HRWI group. Similarly, kidney aquaporins (AQP) 2, 3, and 4 were upregulated in the LRWI group compared with the HRWI group. Given that water was provided ad libitum, the up-regulation of AVP and AQP gene mRNA expressions seem to indicate that the LRWI birds were more efficient in water reabsorption in the kidney compared to their HRWI counterparts. Increased water reabsorption will reduce the amount of water consumed to attain hydration. The water reabsorption potential was reflected in the excreta moisture levels as the LRWI birds had significantly lower excreta moisture than the HRWI birds. Excreta moisture level require further studies and could be considered as a potential proxy trait for water intake.
Collapse
Affiliation(s)
- Samuel E Aggrey
- Water Intake Genomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA, 30602.
| | - Ahmed F A Ghareeb
- Water Intake Genomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA, 30602
| | - Marie C Milfort
- Water Intake Genomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA, 30602
| | - Oluwatomide W Ariyo
- Water Intake Genomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA, 30602
| | - Bikash Aryal
- Water Intake Genomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA, 30602
| | - Evan Hartono
- Water Intake Genomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA, 30602
| | - Josephine Kwakye
- Water Intake Genomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA, 30602
| | - Selorm Sovi
- Water Intake Genomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA, 30602
| | - Sommer A Hipple
- Water Intake Genomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA, 30602
| | - Carrienton Stevenson
- Department of Agricultural Engineering Technology, Fort Valley State University, Fort Valley, GA, 31030
| | - Alberta L Fuller
- Water Intake Genomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA, 30602
| | | | - Farid Stino
- Department of Animal Production, Cairo University, Giza, Egypt
| | - Romdhane Rekaya
- Department of Animal and Dairy Science, University of Georgia, Athens, GA, 30602
| |
Collapse
|
15
|
Stuber GD. Neurocircuits for motivation. Science 2023; 382:394-398. [PMID: 37883553 DOI: 10.1126/science.adh8287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/22/2023] [Indexed: 10/28/2023]
Abstract
The nervous system coordinates various motivated behaviors such as feeding, drinking, and escape to promote survival and evolutionary fitness. Although the precise behavioral repertoires required for distinct motivated behaviors are diverse, common features such as approach or avoidance suggest that common brain substrates are required for a wide range of motivated behaviors. In this Review, I describe a framework by which neural circuits specified for some innate drives regulate the activity of ventral tegmental area (VTA) dopamine neurons to reinforce ongoing or planned actions to fulfill motivational demands. This framework may explain why signaling from VTA dopamine neurons is ubiquitously involved in many types of diverse volitional motivated actions, as well as how sensory and interoceptive cues can initiate specific goal-directed actions.
Collapse
Affiliation(s)
- Garret D Stuber
- Center for the Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
16
|
Pool AH, Poldsam H, Chen S, Thomson M, Oka Y. Recovery of missing single-cell RNA-sequencing data with optimized transcriptomic references. Nat Methods 2023; 20:1506-1515. [PMID: 37697162 DOI: 10.1038/s41592-023-02003-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 08/15/2023] [Indexed: 09/13/2023]
Abstract
Single-cell RNA-sequencing (scRNA-seq) is an indispensable tool for characterizing cellular diversity and generating hypotheses throughout biology. Droplet-based scRNA-seq datasets often lack expression data for genes that can be detected with other methods. Here we show that the observed sensitivity deficits stem from three sources: (1) poor annotation of 3' gene ends; (2) issues with intronic read incorporation; and (3) gene overlap-derived read loss. We show that missing gene expression data can be recovered by optimizing the reference transcriptome for scRNA-seq through recovering false intergenic reads, implementing a hybrid pre-mRNA mapping strategy and resolving gene overlaps. We demonstrate, with a diverse collection of mouse and human tissue data, that reference optimization can substantially improve cellular profiling resolution and reveal missing cell types and marker genes. Our findings argue that transcriptomic references need to be optimized for scRNA-seq analysis and warrant a reanalysis of previously published datasets and cell atlases.
Collapse
Affiliation(s)
- Allan-Hermann Pool
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Peter O'Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Anesthesiology and Pain Management, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Helen Poldsam
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Sisi Chen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Matt Thomson
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Yuki Oka
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
17
|
Warren AM, Grossmann M, Christ-Crain M, Russell N. Syndrome of Inappropriate Antidiuresis: From Pathophysiology to Management. Endocr Rev 2023; 44:819-861. [PMID: 36974717 PMCID: PMC10502587 DOI: 10.1210/endrev/bnad010] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 02/19/2023] [Accepted: 03/27/2023] [Indexed: 03/29/2023]
Abstract
Hyponatremia is the most common electrolyte disorder, affecting more than 15% of patients in the hospital. Syndrome of inappropriate antidiuresis (SIAD) is the most frequent cause of hypotonic hyponatremia, mediated by nonosmotic release of arginine vasopressin (AVP, previously known as antidiuretic hormone), which acts on the renal V2 receptors to promote water retention. There are a variety of underlying causes of SIAD, including malignancy, pulmonary pathology, and central nervous system pathology. In clinical practice, the etiology of hyponatremia is frequently multifactorial and the management approach may need to evolve during treatment of a single episode. It is therefore important to regularly reassess clinical status and biochemistry, while remaining alert to potential underlying etiological factors that may become more apparent during the course of treatment. In the absence of severe symptoms requiring urgent intervention, fluid restriction (FR) is widely endorsed as the first-line treatment for SIAD in current guidelines, but there is considerable controversy regarding second-line therapy in instances where FR is unsuccessful, which occurs in around half of cases. We review the epidemiology, pathophysiology, and differential diagnosis of SIAD, and summarize recent evidence for therapeutic options beyond FR, with a focus on tolvaptan, urea, and sodium-glucose cotransporter 2 inhibitors.
Collapse
Affiliation(s)
- Annabelle M Warren
- Department of Medicine, University of Melbourne, Victoria 3010, Australia
- Department of Endocrinology, The Austin Hospital, Victoria 3084, Australia
| | - Mathis Grossmann
- Department of Medicine, University of Melbourne, Victoria 3010, Australia
- Department of Endocrinology, The Austin Hospital, Victoria 3084, Australia
| | - Mirjam Christ-Crain
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel 4031, Switzerland
- Department of Clinical Research, University of Basel and University Hospital Basel, Basel 4031, Switzerland
| | - Nicholas Russell
- Department of Medicine, University of Melbourne, Victoria 3010, Australia
- Department of Endocrinology, The Austin Hospital, Victoria 3084, Australia
| |
Collapse
|
18
|
Todini L, Fantuz F. Thirst: neuroendocrine regulation in mammals. Vet Res Commun 2023; 47:1085-1101. [PMID: 36932281 DOI: 10.1007/s11259-023-10104-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 03/13/2023] [Indexed: 03/19/2023]
Abstract
Animals can sense their changing internal needs and then generate specific physiological and behavioural responses in order to restore homeostasis. Water-saline homeostasis derives from balances of water and sodium intake and output (drinking and diuresis, salt appetite and natriuresis), maintaining an appropriate composition and volume of extracellular fluid. Thirst is the sensation which drives to seek and consume water, regulated in the central nervous system by both neural and chemical signals. Water and electrolyte homeostasis depends on finely tuned physiological mechanisms, mainly susceptible to plasma Na+ concentration and osmotic pressure, but also to blood volume and arterial pressure. Increases of osmotic pressure as slight as 1-2% are enough to induce thirst ("homeostatic" or cellular), by activation of specialized osmoreceptors in the circumventricular organs, outside the blood-brain barrier. Presystemic anticipatory signals (by oropharyngeal or gastrointestinal receptors) inhibit thirst when fluids are ingested, or stimulate thirst associated with food intake. Hypovolemia, arterial hypotension, Angiotensin II stimulate thirst ("hypovolemic thirst", "extracellular dehydration"). Hypervolemia, hypertension, Atrial Natriuretic Peptide inhibit thirst. Circadian rhythms of thirst are also detectable, driven by suprachiasmatic nucleus in the hypothalamus. Such homeostasis and other fundamental physiological functions (cardiocircolatory, thermoregulation, food intake) are highly interdependent.
Collapse
Affiliation(s)
- Luca Todini
- Scuola di Bioscienze e Medicina Veterinaria, Università di Camerino, Via della Circonvallazione 93/95, 62024, Matelica, MC, Italy.
| | - Francesco Fantuz
- Scuola di Bioscienze e Medicina Veterinaria, Università di Camerino, Via della Circonvallazione 93/95, 62024, Matelica, MC, Italy
| |
Collapse
|
19
|
Mayeli A, Al Zoubi O, White EJ, Chappelle S, Kuplicki R, Morton A, Bruce J, Smith R, Feinstein JS, Bodurka J, Paulus MP, Khalsa SS. Parieto-occipital ERP indicators of gut mechanosensation in humans. Nat Commun 2023; 14:3398. [PMID: 37311748 PMCID: PMC10264354 DOI: 10.1038/s41467-023-39058-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 05/24/2023] [Indexed: 06/15/2023] Open
Abstract
Understanding the neural processes governing the human gut-brain connection has been challenging due to the inaccessibility of the body's interior. Here, we investigated neural responses to gastrointestinal sensation using a minimally invasive mechanosensory probe by quantifying brain, stomach, and perceptual responses following the ingestion of a vibrating capsule. Participants successfully perceived capsule stimulation under two vibration conditions (normal and enhanced), as evidenced by above chance accuracy scores. Perceptual accuracy improved significantly during the enhanced relative to normal stimulation, which was associated with faster stimulation detection and reduced reaction time variability. Capsule stimulation induced late neural responses in parieto-occipital electrodes near the midline. Moreover, these 'gastric evoked potentials' showed intensity-dependent increases in amplitude and were significantly correlated with perceptual accuracy. Our results replicated in a separate experiment, and abdominal X-ray imaging localized most capsule stimulations to the gastroduodenal segments. Combined with our prior observation that a Bayesian model is capable of estimating computational parameters of gut-brain mechanosensation, these findings highlight a unique form of enterically-focused sensory monitoring within the human brain, with implications for understanding gut feelings and gut-brain interactions in healthy and clinical populations.
Collapse
Affiliation(s)
- Ahmad Mayeli
- Laureate Institute for Brain Research, Tulsa, OK, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Obada Al Zoubi
- Laureate Institute for Brain Research, Tulsa, OK, USA
- Harvard Medical School/McLean Hospital, Boston, MA, USA
| | - Evan J White
- Laureate Institute for Brain Research, Tulsa, OK, USA
| | | | | | - Alexa Morton
- Laureate Institute for Brain Research, Tulsa, OK, USA
| | - Jaimee Bruce
- Laureate Institute for Brain Research, Tulsa, OK, USA
| | - Ryan Smith
- Laureate Institute for Brain Research, Tulsa, OK, USA
| | | | - Jerzy Bodurka
- Laureate Institute for Brain Research, Tulsa, OK, USA
- Stephenson School of Biomedical Engineering, University of Oklahoma, Tulsa, OK, USA
| | - Martin P Paulus
- Laureate Institute for Brain Research, Tulsa, OK, USA
- Oxley College of Health Sciences, University of Tulsa, Tulsa, OK, USA
| | - Sahib S Khalsa
- Laureate Institute for Brain Research, Tulsa, OK, USA.
- Oxley College of Health Sciences, University of Tulsa, Tulsa, OK, USA.
| |
Collapse
|
20
|
Brakey DJ, Schatz KC, Paul MJ, Daniels D. The role of glucagon-like peptide-1 (GLP-1) in fluid and food intakes in vasopressin-deficient Brattleboro rats. Physiol Behav 2023; 262:114093. [PMID: 36706972 PMCID: PMC9974868 DOI: 10.1016/j.physbeh.2023.114093] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023]
Abstract
Eating and drinking co-occur and many of the same mechanisms that control one are involved in the control of the other, making it difficult to isolate specific mechanisms for the control of fluid intake. Glucagon-like peptide-1 (GLP-1) is a peptide that seems to be involved in the endogenous control of both ingestive behaviors, but we lack a thorough understanding of how and where GLP-1 is acting to control fluid intake. Vasopressin-deficient Brattleboro rats are a model of hereditary hypothalamic diabetes insipidus that have been used extensively for the study of vasopressin actions in behavior and physiology. Here, we propose that these rats, that eat normally but drink excessively, provide a useful model to dissociate central controls of food and fluid intakes. As an initial step toward establishing this model for these purposes, we focused on GLP-1. Similar to the effect observed after treatment with a GLP-1 receptor (GLP-1R) agonist, the intake difference between wildtype and Brattleboro rats was largely a function in the number of licking bursts, indicating differences in post-ingestive feedback (e.g., satiation). When given central injections of a GLP-1R agonist, the effect on feeding was comparable between wildtype and Brattleboro rats, but the effect of drug on fluid intake was markedly exaggerated in Brattleboro rats. Additionally, Brattleboro rats did not respond to GLP-1R antagonism, whereas wildtype rats did. Taken together, these results suggest that Brattleboro rats exhibit a selective disruption to GLP-1's control of water intake. Overall, these experiments provide foundational studies of the ingestive behavior of Brattleboro rats and demonstrate the potential to use these rats to disentangle the effects of GLP-1 on food and fluid intakes.
Collapse
Affiliation(s)
- Destiny J Brakey
- Department of Biological Sciences, State University of New York at Buffalo, Buffalo, NY, USA; Department of Psychology, State University of New York at Buffalo, Buffalo, NY, USA
| | - Kelcie C Schatz
- Department of Psychology, State University of New York at Buffalo, Buffalo, NY, USA
| | - Matthew J Paul
- Department of Psychology, State University of New York at Buffalo, Buffalo, NY, USA
| | - Derek Daniels
- Department of Biological Sciences, State University of New York at Buffalo, Buffalo, NY, USA; Department of Psychology, State University of New York at Buffalo, Buffalo, NY, USA; Center for Ingestive Behavior Research, State University of New York at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
21
|
Yang D, Wang Y, Qi T, Zhang X, Shen L, Ma J, Pang Z, Lal NK, McClatchy DB, Wang K, Xie Y, Polli F, Maximov A, Augustine V, Cline HT, Yates JR, Ye L. Phosphorylation of pyruvate dehydrogenase marks the inhibition of in vivo neuronal activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.13.532494. [PMID: 36993270 PMCID: PMC10054949 DOI: 10.1101/2023.03.13.532494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
For decades, the expression of immediate early genes (IEGs) such as c- fos has been the most widely used molecular marker representing neuronal activation. However, to date, there is no equivalent surrogate available for the decrease of neuronal activity (i.e., inhibition). Here, we developed an optogenetic-based biochemical screen in which population neural activities can be controlled by light with single action potential precision, followed by unbiased phosphoproteomic profiling. We identified that the phosphorylation of pyruvate dehydrogenase (pPDH) inversely correlated with the intensity of action potential firing in primary neurons. In in vivo mouse models, monoclonal antibody-based pPDH immunostaining detected neuronal inhibition across the brain induced by a wide range of factors including general anesthesia, sensory experiences, and natural behaviors. Thus, as an in vivo marker for neuronal inhibition, pPDH can be used together with IEGs or other cell-type markers to profile and identify bi-directional neural dynamics induced by experiences or behaviors.
Collapse
|
22
|
Liu Y, Wei JA, Luo Z, Cui J, Luo Y, Mak SOK, Wang S, Zhang F, Yang Y, So KF, Shi L, Zhang L, Chow BKC. A gut-brain axis mediates sodium appetite via gastrointestinal peptide regulation on a medulla-hypothalamic circuit. SCIENCE ADVANCES 2023; 9:eadd5330. [PMID: 36791202 PMCID: PMC9931223 DOI: 10.1126/sciadv.add5330] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 01/17/2023] [Indexed: 05/29/2023]
Abstract
Salt homeostasis is orchestrated by both neural circuits and peripheral endocrine factors. The colon is one of the primary sites for electrolyte absorption, while its potential role in modulating sodium intake remains unclear. Here, we revealed that a gastrointestinal hormone, secretin, is released from colon endocrine cells under body sodium deficiency and is indispensable for inducing salt appetite. As the neural substrate, circulating secretin activates specific receptors in the nucleus of the solitary tracts, which further activates the downstream paraventricular nucleus of the hypothalamus, resulting in enhanced sodium intake. These results demonstrated a previously unrecognized gut-brain pathway for the timely regulation of sodium homeostasis.
Collapse
Affiliation(s)
- Yuchu Liu
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
- School of Biological Sciences, University of Hong Kong, Hong Kong SAR, China
| | - Ji-an Wei
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
- School of Biological Sciences, University of Hong Kong, Hong Kong SAR, China
| | - Zhihua Luo
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Jing Cui
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Yifan Luo
- School of Biological Sciences, University of Hong Kong, Hong Kong SAR, China
| | - Sarah Oi Kwan Mak
- School of Biological Sciences, University of Hong Kong, Hong Kong SAR, China
| | - Siqi Wang
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Fengwei Zhang
- School of Biological Sciences, University of Hong Kong, Hong Kong SAR, China
| | - Yan Yang
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Kwok-Fai So
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
- Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou, China
- Neuroscience and Neurorehabilitation Institute, University of Health and Rehabilitation Sciences, Qingdao, China
- State Key Laboratory of Brain and Cognitive Science, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Lingling Shi
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Li Zhang
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
- Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou, China
- Neuroscience and Neurorehabilitation Institute, University of Health and Rehabilitation Sciences, Qingdao, China
| | | |
Collapse
|
23
|
Hao MM, Stamp LA. The many means of conversation between the brain and the gut. Nat Rev Gastroenterol Hepatol 2023; 20:73-74. [PMID: 36517617 DOI: 10.1038/s41575-022-00725-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Marlene M Hao
- Department of Anatomy and Physiology, the University of Melbourne, Parkville, Victoria, Australia.
| | - Lincon A Stamp
- Department of Anatomy and Physiology, the University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
24
|
Arginine-vasopressin-expressing neurons in the murine suprachiasmatic nucleus exhibit a circadian rhythm in network coherence in vivo. Proc Natl Acad Sci U S A 2023; 120:e2209329120. [PMID: 36656857 PMCID: PMC9942887 DOI: 10.1073/pnas.2209329120] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The suprachiasmatic nucleus (SCN) is composed of functionally distinct subpopulations of GABAergic neurons which form a neural network responsible for synchronizing most physiological and behavioral circadian rhythms in mammals. To date, little is known regarding which aspects of SCN rhythmicity are generated by individual SCN neurons, and which aspects result from neuronal interaction within a network. Here, we utilize in vivo miniaturized microscopy to measure fluorescent GCaMP-reported calcium dynamics in arginine vasopressin (AVP)-expressing neurons in the intact SCN of awake, behaving mice. We report that SCN AVP neurons exhibit periodic, slow calcium waves which we demonstrate, using in vivo electrical recordings, likely reflect burst firing. Further, we observe substantial heterogeneity of function in that AVP neurons exhibit unstable rhythms, and relatively weak rhythmicity at the population level. Network analysis reveals that correlated cellular behavior, or coherence, among neuron pairs also exhibited stochastic rhythms with about 33% of pairs rhythmic at any time. Unlike single-cell variables, coherence exhibited a strong rhythm at the population level with time of maximal coherence among AVP neuronal pairs at CT/ZT 6 and 9, coinciding with the timing of maximal neuronal activity for the SCN as a whole. These results demonstrate robust circadian variation in the coordination between stochastically rhythmic neurons and that interactions between AVP neurons in the SCN may be more influential than single-cell activity in the regulation of circadian rhythms. Furthermore, they demonstrate that cells in this circuit, like those in many other circuits, exhibit profound heterogenicity of function over time and space.
Collapse
|
25
|
Baumer-Harrison C, Breza JM, Sumners C, Krause EG, de Kloet AD. Sodium Intake and Disease: Another Relationship to Consider. Nutrients 2023; 15:535. [PMID: 36771242 PMCID: PMC9921152 DOI: 10.3390/nu15030535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/14/2023] [Accepted: 01/15/2023] [Indexed: 01/22/2023] Open
Abstract
Sodium (Na+) is crucial for numerous homeostatic processes in the body and, consequentially, its levels are tightly regulated by multiple organ systems. Sodium is acquired from the diet, commonly in the form of NaCl (table salt), and substances that contain sodium taste salty and are innately palatable at concentrations that are advantageous to physiological homeostasis. The importance of sodium homeostasis is reflected by sodium appetite, an "all-hands-on-deck" response involving the brain, multiple peripheral organ systems, and endocrine factors, to increase sodium intake and replenish sodium levels in times of depletion. Visceral sensory information and endocrine signals are integrated by the brain to regulate sodium intake. Dysregulation of the systems involved can lead to sodium overconsumption, which numerous studies have considered causal for the development of diseases, such as hypertension. The purpose here is to consider the inverse-how disease impacts sodium intake, with a focus on stress-related and cardiometabolic diseases. Our proposition is that such diseases contribute to an increase in sodium intake, potentially eliciting a vicious cycle toward disease exacerbation. First, we describe the mechanism(s) that regulate each of these processes independently. Then, we highlight the points of overlap and integration of these processes. We propose that the analogous neural circuitry involved in regulating sodium intake and blood pressure, at least in part, underlies the reciprocal relationship between neural control of these functions. Finally, we conclude with a discussion on how stress-related and cardiometabolic diseases influence these circuitries to alter the consumption of sodium.
Collapse
Affiliation(s)
- Caitlin Baumer-Harrison
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32603, USA
- Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, FL 32610, USA
- Center for Smell and Taste, University of Florida, Gainesville, FL 32610, USA
- Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Joseph M. Breza
- Department of Psychology, College of Arts and Sciences, Eastern Michigan University, Ypsilanti, MI 48197, USA
| | - Colin Sumners
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32603, USA
- Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, FL 32610, USA
- Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Eric G. Krause
- Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, FL 32610, USA
- Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Annette D. de Kloet
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32603, USA
- Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, FL 32610, USA
- Center for Smell and Taste, University of Florida, Gainesville, FL 32610, USA
- Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
26
|
Malaguarnera G, Graute M, Homs Corbera A. The translational roadmap of the gut models, focusing on gut-on-chip. OPEN RESEARCH EUROPE 2023; 1:62. [PMID: 37645178 PMCID: PMC10445823 DOI: 10.12688/openreseurope.13709.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/09/2022] [Indexed: 08/31/2023]
Abstract
It is difficult to model in vitro the intestine when seeking to include crosstalk with the gut microbiota, immune and neuroendocrine systems. Here we present a roadmap of the current models to facilitate the choice in preclinical and translational research with a focus on gut-on-chip. These micro physiological systems (MPS) are microfluidic devices that recapitulate in vitro the physiology of the intestine. We reviewed the gut-on-chips that had been developed in academia and industries as single chip and that have three main purpose: replicate the intestinal physiology, the intestinal pathological features, and for pharmacological tests.
Collapse
Affiliation(s)
| | - Miriam Graute
- R&D department, Cherry Biotech SAS, Rennes, Brittany, 35000, France
| | | |
Collapse
|
27
|
Malaguarnera G, Graute M, Homs Corbera A. The translational roadmap of the gut models, focusing on gut-on-chip. OPEN RESEARCH EUROPE 2023; 1:62. [PMID: 37645178 PMCID: PMC10445823 DOI: 10.12688/openreseurope.13709.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/09/2022] [Indexed: 08/31/2023]
Abstract
It is difficult to model in vitro the intestine when seeking to include crosstalk with the gut microbiota, immune and neuroendocrine systems. Here we present a roadmap of the current models to facilitate the choice in preclinical and translational research with a focus on gut-on-chip. These micro physiological systems (MPS) are microfluidic devices that recapitulate in vitro the physiology of the intestine. We reviewed the gut-on-chips that had been developed in academia and industries as single chip and that have three main purpose: replicate the intestinal physiology, the intestinal pathological features, and for pharmacological tests.
Collapse
Affiliation(s)
| | - Miriam Graute
- R&D department, Cherry Biotech SAS, Rennes, Brittany, 35000, France
| | | |
Collapse
|
28
|
Kamel MH, Upadhyay A, Borkan SC. Intractable hyponatremia complicated by a reset osmostat: a case report. J Med Case Rep 2023; 17:13. [PMID: 36639685 PMCID: PMC9840259 DOI: 10.1186/s13256-022-03732-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 12/21/2022] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Hyponatremia associated with a low serum osmolality is a common and confounding electrolyte disorder. Correcting hyponatremia is also complicated, especially in the setting of chronic hyponatremia. Here, we provide a rational approach to accurately detecting and safely treating acute on chronic euvolemic hyponatremia in the setting of acute polydipsia with a chronic reset osmostat. CASE PRESENTATION A 71-year-old hispanic gentleman with chronic hyponatremia presented with hiccups, polydipsia, and a serum sodium concentration of 120 mEq/L associated with diffuse weakness, inattentiveness, and suicidal ideation. Symptomatic euvolemic hyponatremia warranted hypertonic saline treatment in the acute phase and water restriction in the chronic phase. Both interventions resulted in improvement in symptoms and/or the serum sodium concentration, but to a serum sodium level that persistently remained below the normal range. Remarkably, the urine osmolality appropriately fell when the serum sodium concentration fell below 126 mEq/L. Also remarkable was the appropriate increase in urine osmolality when the serum sodium concentration exceeded 126 mEq/L. The preservation of both concentration and dilution, albeit at a lower-than-normal serum osmolality, shows that the osmostat regulating antidiuretic hormone release had been "reset." Both physiologic and pharmacologic resetting of the osmostat are discussed. CONCLUSIONS Preservation of urinary concentrating and diluting ability at a lower-than-normal serum sodium concentration, especially in the setting of chronic hyponatremia, is diagnostic of a reset osmostat. The presence of a reset osmostat often confounds the treatment of concomitant acute hyponatremia. Early recognition of a reset osmostat avoids the need to normalize serum sodium concentration, expedites hospital discharge, and limits potential harm from overcorrecting acute hyponatremia.
Collapse
Affiliation(s)
- Mohamed Hassan Kamel
- grid.189504.10000 0004 1936 7558Section of Nephrology, Department of Medicine, Boston Medical Center and Boston University School of Medicine, Boston, MA USA
| | - Ashish Upadhyay
- grid.189504.10000 0004 1936 7558Section of Nephrology, Department of Medicine, Boston Medical Center and Boston University School of Medicine, Boston, MA USA
| | - Steven C. Borkan
- grid.189504.10000 0004 1936 7558Section of Nephrology, Department of Medicine, Boston Medical Center and Boston University School of Medicine, Boston, MA USA ,Evans Biomedical Research Center, 650 Albany St, Room 546, Boston, MA 02118-2518 USA
| |
Collapse
|
29
|
Sodium Homeostasis, a Balance Necessary for Life. Nutrients 2023; 15:nu15020395. [PMID: 36678265 PMCID: PMC9862583 DOI: 10.3390/nu15020395] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Body sodium (Na) levels must be maintained within a narrow range for the correct functioning of the organism (Na homeostasis). Na disorders include not only elevated levels of this solute (hypernatremia), as in diabetes insipidus, but also reduced levels (hyponatremia), as in cerebral salt wasting syndrome. The balance in body Na levels therefore requires a delicate equilibrium to be maintained between the ingestion and excretion of Na. Salt (NaCl) intake is processed by receptors in the tongue and digestive system, which transmit the information to the nucleus of the solitary tract via a neural pathway (chorda tympani/vagus nerves) and to circumventricular organs, including the subfornical organ and area postrema, via a humoral pathway (blood/cerebrospinal fluid). Circuits are formed that stimulate or inhibit homeostatic Na intake involving participation of the parabrachial nucleus, pre-locus coeruleus, medial tuberomammillary nuclei, median eminence, paraventricular and supraoptic nuclei, and other structures with reward properties such as the bed nucleus of the stria terminalis, central amygdala, and ventral tegmental area. Finally, the kidney uses neural signals (e.g., renal sympathetic nerves) and vascular (e.g., renal perfusion pressure) and humoral (e.g., renin-angiotensin-aldosterone system, cardiac natriuretic peptides, antidiuretic hormone, and oxytocin) factors to promote Na excretion or retention and thereby maintain extracellular fluid volume. All these intake and excretion processes are modulated by chemical messengers, many of which (e.g., aldosterone, angiotensin II, and oxytocin) have effects that are coordinated at peripheral and central level to ensure Na homeostasis.
Collapse
|
30
|
Bárez-López S, Scanlon L, Murphy D, Greenwood MP. Imaging the Hypothalamo-Neurohypophysial System. Neuroendocrinology 2023; 113:168-178. [PMID: 34438401 DOI: 10.1159/000519233] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/23/2021] [Indexed: 11/19/2022]
Abstract
The hypothalamo-neurohypophysial system (HNS) is a brain peptidergic neurosecretory apparatus which is composed of arginine vasopressin (AVP) and oxytocin (OXT) magnocellular neurones and their neuronal processes in the posterior pituitary (PP). In response to specific stimuli, AVP and OXT are secreted into the systemic circulation at the neurovascular interface of the PP, where they act as hormones, but they can also behave as neurotransmitters when released at the somatodendritic compartment or by axon collaterals to other brain regions. Because these peptides are crucial for several physiological processes, including fluid homoeostasis and reproduction, it is of great importance to map the HNS connectome in its entirety in order to understand its functions. In recent years, advances in imaging technologies have provided considerable new information about the HNS. These approaches include the use of reporter proteins under the control of specific promoters, viral tracers, brain-clearing methods, genetically encoded indicators, sniffer cells, mass spectrometry imaging, and spatially resolved transcriptomics. In this review, we illustrate how these latest approaches have enhanced our understanding of the structure and function of the HNS and how they might contribute further in the coming years.
Collapse
Affiliation(s)
- Soledad Bárez-López
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Liam Scanlon
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - David Murphy
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Michael Paul Greenwood
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| |
Collapse
|
31
|
Lv X, Gao F, Cao X. Skeletal interoception in bone homeostasis and pain. Cell Metab 2022; 34:1914-1931. [PMID: 36257317 PMCID: PMC9742337 DOI: 10.1016/j.cmet.2022.09.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/07/2022] [Accepted: 09/26/2022] [Indexed: 01/24/2023]
Abstract
Accumulating evidence indicates that interoception maintains proper physiological status and orchestrates metabolic homeostasis by regulating feeding behaviors, glucose balance, and lipid metabolism. Continuous skeletal remodeling consumes a tremendous amount of energy to provide skeletal scaffolding, support muscle movement, store vital minerals, and maintain a niche for hematopoiesis, which are processes that also contribute to overall metabolic balance. Although skeletal innervation has been described for centuries, recent work has shown that skeletal metabolism is tightly regulated by the nervous system and that skeletal interoception regulates bone homeostasis. Here, we provide a general discussion of interoception and its effects on the skeleton and whole-body metabolism. We also discuss skeletal interoception-mediated regulation in the context of pathological conditions and skeletal pain as well as future challenges to our understanding of these process and how they can be leveraged for more effective therapy.
Collapse
Affiliation(s)
- Xiao Lv
- Center for Musculoskeletal Research, Department of Orthopaedic Surgery and Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21205, USA
| | - Feng Gao
- Center for Musculoskeletal Research, Department of Orthopaedic Surgery and Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21205, USA
| | - Xu Cao
- Center for Musculoskeletal Research, Department of Orthopaedic Surgery and Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
32
|
Zhang F, Mak SOK, Liu Y, Ke Y, Rao F, Yung WH, Zhang L, Chow BKC. Secretin receptor deletion in the subfornical organ attenuates the activation of excitatory neurons under dehydration. Curr Biol 2022; 32:4832-4841.e5. [PMID: 36220076 DOI: 10.1016/j.cub.2022.09.037] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 07/22/2022] [Accepted: 09/19/2022] [Indexed: 11/06/2022]
Abstract
In mammals, thirst is strongly influenced by the subfornical organ (SFO), a forebrain structure that integrates circulating signals including osmotic pressure and sodium contents. Secretin (SCT), a classical gastrointestinal hormone, has been implicated as a humoral factor regulating body-fluid homeostasis. However, the neural mechanism of secretin in the central nervous system in managing thirst remains unclear. In this study, we report that the local ablation of SCT receptor (SCTR) in the SFO reduces water but not salt intake in dehydrated mice and this effect could not be rescued by exogenous SCT administration. Electrophysiology with single-cell RT-PCR indicates that SCT elicits inward currents in the SFO neuronal nitric oxide synthase (SFOnNOS) neurons via SCTR in the presence of glutamate receptor antagonists. We further show that the SCTR in the SFO permits the activation of SFOnNOS neurons under distinct thirst types. Projection-specific gene deletion of SCTR in SFO to the median preoptic nucleus (MnPO) pathway also reduces water intake in dehydrated animals. SCT signaling thus plays an indispensable role in driving thirst. These data not only expand the functional boundaries of SCTR but also provide insights into the central mechanisms of homeostatic regulation.
Collapse
Affiliation(s)
- Fengwei Zhang
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | - Sarah O K Mak
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | - Yuchu Liu
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | - Ya Ke
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Gerald Choa Neuroscience Centre, The Chinese University of Hong Kong, Hong Kong, China
| | - Feng Rao
- School of Life Sciences, Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Wing Ho Yung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Gerald Choa Neuroscience Centre, The Chinese University of Hong Kong, Hong Kong, China.
| | - Li Zhang
- Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, China; Neuroscience and Neurorehabilitation Institute, University of Health and Rehabilitation Sciences, Qingdao, China.
| | | |
Collapse
|
33
|
Durst und Trinken – Physiologie und Bedeutung für die Störungen des Wasserhaushalts. JOURNAL FÜR KLINISCHE ENDOKRINOLOGIE UND STOFFWECHSEL 2022. [DOI: 10.1007/s41969-022-00179-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
34
|
Park A, Croset V, Otto N, Agarwal D, Treiber CD, Meschi E, Sims D, Waddell S. Gliotransmission of D-serine promotes thirst-directed behaviors in Drosophila. Curr Biol 2022; 32:3952-3970.e8. [PMID: 35963239 PMCID: PMC9616736 DOI: 10.1016/j.cub.2022.07.038] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/04/2022] [Accepted: 07/15/2022] [Indexed: 12/13/2022]
Abstract
Thirst emerges from a range of cellular changes that ultimately motivate an animal to consume water. Although thirst-responsive neuronal signals have been reported, the full complement of brain responses is unclear. Here, we identify molecular and cellular adaptations in the brain using single-cell sequencing of water-deprived Drosophila. Water deficiency primarily altered the glial transcriptome. Screening the regulated genes revealed astrocytic expression of the astray-encoded phosphoserine phosphatase to bi-directionally regulate water consumption. Astray synthesizes the gliotransmitter D-serine, and vesicular release from astrocytes is required for drinking. Moreover, dietary D-serine rescues aay-dependent drinking deficits while facilitating water consumption and expression of water-seeking memory. D-serine action requires binding to neuronal NMDA-type glutamate receptors. Fly astrocytes contribute processes to tripartite synapses, and the proportion of astrocytes that are themselves activated by glutamate increases with water deprivation. We propose that thirst elevates astrocytic D-serine release, which awakens quiescent glutamatergic circuits to enhance water procurement.
Collapse
Affiliation(s)
- Annie Park
- Centre for Neural Circuits & Behaviour, University of Oxford, Oxford OX1 3TA, UK
| | - Vincent Croset
- Centre for Neural Circuits & Behaviour, University of Oxford, Oxford OX1 3TA, UK; Department of Biosciences, Durham University, Durham DH1 3LE, UK.
| | - Nils Otto
- Centre for Neural Circuits & Behaviour, University of Oxford, Oxford OX1 3TA, UK
| | - Devika Agarwal
- Centre for Neural Circuits & Behaviour, University of Oxford, Oxford OX1 3TA, UK; MRC Computational Genomics Analysis and Training Programme (CGAT), MRC Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK
| | - Christoph D Treiber
- Centre for Neural Circuits & Behaviour, University of Oxford, Oxford OX1 3TA, UK
| | - Eleonora Meschi
- Centre for Neural Circuits & Behaviour, University of Oxford, Oxford OX1 3TA, UK
| | - David Sims
- MRC Computational Genomics Analysis and Training Programme (CGAT), MRC Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK
| | - Scott Waddell
- Centre for Neural Circuits & Behaviour, University of Oxford, Oxford OX1 3TA, UK.
| |
Collapse
|
35
|
Grove JCR, Gray LA, La Santa Medina N, Sivakumar N, Ahn JS, Corpuz TV, Berke JD, Kreitzer AC, Knight ZA. Dopamine subsystems that track internal states. Nature 2022; 608:374-380. [PMID: 35831501 PMCID: PMC9365689 DOI: 10.1038/s41586-022-04954-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/08/2022] [Indexed: 12/11/2022]
Abstract
Food and water are rewarding in part because they satisfy our internal needs1,2. Dopaminergic neurons in the ventral tegmental area (VTA) are activated by gustatory rewards3-5, but how animals learn to associate these oral cues with the delayed physiological effects of ingestion is unknown. Here we show that individual dopaminergic neurons in the VTA respond to detection of nutrients or water at specific stages of ingestion. A major subset of dopaminergic neurons tracks changes in systemic hydration that occur tens of minutes after thirsty mice drink water, whereas different dopaminergic neurons respond to nutrients in the gastrointestinal tract. We show that information about fluid balance is transmitted to the VTA by a hypothalamic pathway and then re-routed to downstream circuits that track the oral, gastrointestinal and post-absorptive stages of ingestion. To investigate the function of these signals, we used a paradigm in which a fluid's oral and post-absorptive effects can be independently manipulated and temporally separated. We show that mice rapidly learn to prefer one fluid over another based solely on its rehydrating ability and that this post-ingestive learning is prevented if dopaminergic neurons in the VTA are selectively silenced after consumption. These findings reveal that the midbrain dopamine system contains subsystems that track different modalities and stages of ingestion, on timescales from seconds to tens of minutes, and that this information is used to drive learning about the consequences of ingestion.
Collapse
Affiliation(s)
- James C R Grove
- Department of Physiology, University of California, San Francisco, San Francisco, CA, USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | | | | | | | - Jamie S Ahn
- Howard Hughes Medical Institute, San Francisco, CA, USA
| | | | - Joshua D Berke
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Anatol C Kreitzer
- Department of Physiology, University of California, San Francisco, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Gladstone Institutes, San Francisco, CA, USA
| | - Zachary A Knight
- Department of Physiology, University of California, San Francisco, San Francisco, CA, USA.
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA.
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA.
- Howard Hughes Medical Institute, San Francisco, CA, USA.
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
36
|
Mercado-Perez A, Beyder A. Gut feelings: mechanosensing in the gastrointestinal tract. Nat Rev Gastroenterol Hepatol 2022; 19:283-296. [PMID: 35022607 PMCID: PMC9059832 DOI: 10.1038/s41575-021-00561-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/22/2021] [Indexed: 12/11/2022]
Abstract
The primary function of the gut is to procure nutrients. Synchronized mechanical activities underlie nearly all its endeavours. Coordination of mechanical activities depends on sensing of the mechanical forces, in a process called mechanosensation. The gut has a range of mechanosensory cells. They function either as specialized mechanoreceptors, which convert mechanical stimuli into coordinated physiological responses at the organ level, or as non-specialized mechanosensory cells that adjust their function based on the mechanical state of their environment. All major cell types in the gastrointestinal tract contain subpopulations that act as specialized mechanoreceptors: epithelia, smooth muscle, neurons, immune cells, and others. These cells are tuned to the physical properties of the surrounding tissue, so they can discriminate mechanical stimuli from the baseline mechanical state. The importance of gastrointestinal mechanosensation has long been recognized, but the latest discoveries of molecular identities of mechanosensors and technical advances that resolve the relevant circuitry have poised the field to make important intellectual leaps. This Review describes the mechanical factors relevant for normal function, as well as the molecules, cells and circuits involved in gastrointestinal mechanosensing. It concludes by outlining important unanswered questions in gastrointestinal mechanosensing.
Collapse
Affiliation(s)
- Arnaldo Mercado-Perez
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN, USA
- Medical Scientist Training Program (MSTP), Mayo Clinic, Rochester, MN, USA
| | - Arthur Beyder
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN, USA.
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
37
|
Gaffield MA, Sauerbrei BA, Christie JM. Cerebellum encodes and influences the initiation, performance, and termination of discontinuous movements in mice. eLife 2022; 11:e71464. [PMID: 35451957 PMCID: PMC9075950 DOI: 10.7554/elife.71464] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 04/21/2022] [Indexed: 11/23/2022] Open
Abstract
The cerebellum is hypothesized to represent timing information important for organizing salient motor events during periodically performed discontinuous movements. To provide functional evidence validating this idea, we measured and manipulated Purkinje cell (PC) activity in the lateral cerebellum of mice trained to volitionally perform periodic bouts of licking for regularly allocated water rewards. Overall, PC simple spiking modulated during task performance, mapping phasic tongue protrusions and retractions, as well as ramping prior to both lick-bout initiation and termination, two important motor events delimiting movement cycles. The ramping onset occurred earlier for the initiation of uncued exploratory licking that anticipated water availability relative to licking that was reactive to water allocation, suggesting that the cerebellum is engaged differently depending on the movement context. In a subpopulation of PCs, climbing-fiber-evoked responses also increased during lick-bout initiation, but not termination, highlighting differences in how cerebellar input pathways represent task-related information. Optogenetic perturbation of PC activity disrupted the behavior by degrading lick-bout rhythmicity in addition to initiating and terminating licking bouts confirming a causative role in movement organization. Together, these results substantiate that the cerebellum contributes to the initiation and timing of repeated motor actions.
Collapse
Affiliation(s)
| | | | - Jason M Christie
- Max Planck Florida Institute for NeuroscienceJupiterUnited States
| |
Collapse
|
38
|
Kurt G, Kodur N, Quiles CR, Reynolds C, Eagle A, Mayer T, Brown J, Makela A, Bugescu R, Seo HD, Carroll QE, Daniels D, Robison AJ, Mazei-Robison M, Leinninger G. Time to drink: Activating lateral hypothalamic area neurotensin neurons promotes intake of fluid over food in a time-dependent manner. Physiol Behav 2022; 247:113707. [PMID: 35063424 PMCID: PMC8844224 DOI: 10.1016/j.physbeh.2022.113707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 11/24/2021] [Accepted: 01/16/2022] [Indexed: 10/19/2022]
Abstract
The lateral hypothalamic area (LHA) is essential for ingestive behavior but has primarily been studied in modulating feeding, with comparatively scant attention on drinking. This is partly because most LHA neurons simultaneously promote feeding and drinking, suggesting that ingestive behaviors track together. A notable exception are LHA neurons expressing neurotensin (LHANts neurons): activating these neurons promotes water intake but modestly restrains feeding. Here we investigated the connectivity of LHANts neurons, their necessity and sufficiency for drinking and feeding, and how timing and resource availability influence their modulation of these behaviors. LHANts neurons project broadly throughout the brain, including to the lateral preoptic area (LPO), a brain region implicated in modulating drinking behavior. LHANts neurons also receive inputs from brain regions implicated in sensing hydration and energy status. While activation of LHANts neurons is not required to maintain homeostatic water or food intake, it selectively promotes drinking during the light cycle, when ingestive drive is low. Activating LHANts neurons during this period also increases willingness to work for water or palatable fluids, regardless of their caloric content. By contrast, LHANts neuronal activation during the dark cycle does not promote drinking, but suppresses feeding during this time. Finally, we demonstrate that the activation of the LHANts → LPO projection is sufficient to mediate drinking behavior, but does not suppress feeding as observed after generally activating all LHANts neurons. Overall, our work suggests how and when LHANts neurons oppositely modulate ingestive behaviors.
Collapse
Key Words
- ARC, Arcuate nucleus
- CEA, Central amygdala
- CNO, Clozapine N-Oxide
- CPP, Conditioned place preference
- DR, Dorsal raphe
- DREADD
- DREADD, Designer receptor exclusively activated by designer drugs
- FR-1, Fixed ratio-1
- LHA
- LHA(Nts), Lateral hypothalamic area neuotensin-expressing
- LHA, Lateral hypothalamic area
- LPO, Lateral preoptic area
- LT, Lateral terminalis
- LepRb, Long form of the leptin receptor
- MnPO, Median preoptic area
- ModRabies, Genetically modified rabies virus, EnvA-∆G-Rabies-mCherry
- NTS, Nucleus of solitary tract
- Nts, Neurotensin
- NtsR1, Neurotensin receptor-1
- NtsR2, Neurotensin receptor-2
- OVLT, Organum vasculosum lamina terminalis
- PAG, Periaqueductal gray
- PB, Parabrachial area
- PR, Progressive ratio
- PVH, Paraventricular nucleus of hypothalamus
- SFO, Subfornical organ
- SNc, Substantia nigra compacta
- SO, Supraoptic nucleus
- TVA, avian viral receptor protein
- VEH, Vehicle
- VTA, Ventral tegmental area
- WT, Wild type
- Water
- aCSF, Artificial cerebrospinal fluid
- body weight
- feeding
- homeostasis
- lHb, Lateral habenula
- lateral preoptic area (LPO)
- neurotensin receptor
- reward
Collapse
Affiliation(s)
- Gizem Kurt
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Nandan Kodur
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | | | - Chelsea Reynolds
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Andrew Eagle
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Tom Mayer
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Juliette Brown
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | - Anna Makela
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Raluca Bugescu
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Harim Delgado Seo
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Quinn E Carroll
- Department of Psychology and the Center for Ingestive Behavior Research, University at Buffalo, the State University of New York, Buffalo, NY 14226, USA
| | - Derek Daniels
- Department of Psychology and the Center for Ingestive Behavior Research, University at Buffalo, the State University of New York, Buffalo, NY 14226, USA
| | - A J Robison
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | | | - Gina Leinninger
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
39
|
Prescott SL, Liberles SD. Internal senses of the vagus nerve. Neuron 2022; 110:579-599. [PMID: 35051375 PMCID: PMC8857038 DOI: 10.1016/j.neuron.2021.12.020] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/30/2021] [Accepted: 12/11/2021] [Indexed: 12/16/2022]
Abstract
The vagus nerve is an indispensable body-brain connection that controls vital aspects of autonomic physiology like breathing, heart rate, blood pressure, and gut motility, reflexes like coughing and swallowing, and survival behaviors like feeding, drinking, and sickness responses. Classical physiological studies and recent molecular/genetic approaches have revealed a tremendous diversity of vagal sensory neuron types that innervate different internal organs, with many cell types remaining poorly understood. Here, we review the state of knowledge related to vagal sensory neurons that innervate the respiratory, cardiovascular, and digestive systems. We focus on cell types and their response properties, physiological/behavioral roles, engaged neural circuits and, when possible, sensory receptors. We are only beginning to understand the signal transduction mechanisms used by vagal sensory neurons and upstream sentinel cells, and future studies are needed to advance the field of interoception to the level of mechanistic understanding previously achieved for our external senses.
Collapse
|
40
|
Ichiki T, Wang T, Kennedy A, Pool AH, Ebisu H, Anderson DJ, Oka Y. Sensory representation and detection mechanisms of gut osmolality change. Nature 2022; 602:468-474. [PMID: 35082448 DOI: 10.1038/s41586-021-04359-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 12/15/2021] [Indexed: 11/08/2022]
Abstract
Ingested food and water stimulate sensory systems in the oropharyngeal and gastrointestinal areas before absorption1,2. These sensory signals modulate brain appetite circuits in a feed-forward manner3-5. Emerging evidence suggests that osmolality sensing in the gut rapidly inhibits thirst neurons upon water intake. Nevertheless, it remains unclear how peripheral sensory neurons detect visceral osmolality changes, and how they modulate thirst. Here we use optical and electrical recording combined with genetic approaches to visualize osmolality responses from sensory ganglion neurons. Gut hypotonic stimuli activate a dedicated vagal population distinct from mechanical-, hypertonic- or nutrient-sensitive neurons. We demonstrate that hypotonic responses are mediated by vagal afferents innervating the hepatic portal area (HPA), through which most water and nutrients are absorbed. Eliminating sensory inputs from this area selectively abolished hypotonic but not mechanical responses in vagal neurons. Recording from forebrain thirst neurons and behavioural analyses show that HPA-derived osmolality signals are required for feed-forward thirst satiation and drinking termination. Notably, HPA-innervating vagal afferents do not sense osmolality itself. Instead, these responses are mediated partly by vasoactive intestinal peptide secreted after water ingestion. Together, our results reveal visceral hypoosmolality as an important vagal sensory modality, and that intestinal osmolality change is translated into hormonal signals to regulate thirst circuit activity through the HPA pathway.
Collapse
Affiliation(s)
- Takako Ichiki
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Tongtong Wang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Ann Kennedy
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Department of Physiology, Northwestern University, Chicago, IL, USA
| | - Allan-Hermann Pool
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Haruka Ebisu
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - David J Anderson
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Howard Hughes Medical Institute, California Institute of Technology, Pasadena, CA, USA
| | - Yuki Oka
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
41
|
Chen L, Jung HJ, Datta A, Park E, Poll BG, Kikuchi H, Leo KT, Mehta Y, Lewis S, Khundmiri SJ, Khan S, Chou CL, Raghuram V, Yang CR, Knepper MA. Systems Biology of the Vasopressin V2 Receptor: New Tools for Discovery of Molecular Actions of a GPCR. Annu Rev Pharmacol Toxicol 2022; 62:595-616. [PMID: 34579536 PMCID: PMC10676752 DOI: 10.1146/annurev-pharmtox-052120-011012] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Systems biology can be defined as the study of a biological process in which all of the relevant components are investigated together in parallel to discover the mechanism. Although the approach is not new, it has come to the forefront as a result of genome sequencing projects completed in the first few years of the current century. It has elements of large-scale data acquisition (chiefly next-generation sequencing-based methods and protein mass spectrometry) and large-scale data analysis (big data integration and Bayesian modeling). Here we discuss these methodologies and show how they can be applied to understand the downstream effects of GPCR signaling, specifically looking at how the neurohypophyseal peptide hormone vasopressin, working through the V2 receptor and PKA activation, regulates the water channel aquaporin-2. The emerging picture provides a detailedframework for understanding the molecular mechanisms involved in water balance disorders, pointing the way to improved treatment of both polyuric disorders and water-retention disorders causing dilutional hyponatremia.
Collapse
Affiliation(s)
- Lihe Chen
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20814, USA;
| | - Hyun Jun Jung
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20814, USA;
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
| | - Arnab Datta
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20814, USA;
- Yenepoya Research Center, Yenepoya, Mangalore 575018, Karnataka, India
| | - Euijung Park
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20814, USA;
| | - Brian G Poll
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20814, USA;
| | - Hiroaki Kikuchi
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20814, USA;
| | - Kirby T Leo
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20814, USA;
| | - Yash Mehta
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20814, USA;
| | - Spencer Lewis
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20814, USA;
| | - Syed J Khundmiri
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20814, USA;
| | - Shaza Khan
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20814, USA;
| | - Chung-Lin Chou
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20814, USA;
| | - Viswanathan Raghuram
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20814, USA;
| | - Chin-Rang Yang
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20814, USA;
| | - Mark A Knepper
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20814, USA;
| |
Collapse
|
42
|
NODA M, MATSUDA T. Central regulation of body fluid homeostasis. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2022; 98:283-324. [PMID: 35908954 PMCID: PMC9363595 DOI: 10.2183/pjab.98.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Extracellular fluids, including blood, lymphatic fluid, and cerebrospinal fluid, are collectively called body fluids. The Na+ concentration ([Na+]) in body fluids is maintained at 135-145 mM and is broadly conserved among terrestrial animals. Homeostatic osmoregulation by Na+ is vital for life because severe hyper- or hypotonicity elicits irreversible organ damage and lethal neurological trauma. To achieve "body fluid homeostasis" or "Na homeostasis", the brain continuously monitors [Na+] in body fluids and controls water/salt intake and water/salt excretion by the kidneys. These physiological functions are primarily regulated based on information on [Na+] and relevant circulating hormones, such as angiotensin II, aldosterone, and vasopressin. In this review, we discuss sensing mechanisms for [Na+] and hormones in the brain that control water/salt intake behaviors, together with the responsible sensors (receptors) and relevant neural pathways. We also describe mechanisms in the brain by which [Na+] increases in body fluids activate the sympathetic neural activity leading to hypertension.
Collapse
Affiliation(s)
- Masaharu NODA
- Homeostatic Mechanism Research Unit, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
- Correspondence should be addressed to: Homeostatic Mechanism Research Unit, Institute of Innovative Research, Tokyo Institute of Technology, Nagatsuta-cho 4259, Midori-ku, Yokohama, Kanagawa 226-8503, Japan (e-mail: )
| | - Takashi MATSUDA
- Homeostatic Mechanism Research Unit, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| |
Collapse
|
43
|
Neuroimaging and modulation in obesity and diabetes research: 10th anniversary meeting. Int J Obes (Lond) 2022; 46:718-725. [PMID: 34934178 PMCID: PMC8960390 DOI: 10.1038/s41366-021-01025-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 10/29/2021] [Accepted: 11/09/2021] [Indexed: 11/29/2022]
|
44
|
Abstract
We confirm that rats can act as rational economic agents, making choices about how much work to do to obtain a reward in a way that optimally trades off the value of the reward against the cost of the effort. Contrary to the notion that bigger rewards are more motivating, rats worked harder in economies where rewards were small, ensuring a sufficient minimum income of water. But they chose to earn and consume more water per day when water was “cheap” (available for little work). We present a mathematical model explaining why rats work when they do (surprisingly, not just when they are thirsty) and suggesting where in the brain animals might compute the current value of working for water. In the laboratory, animals’ motivation to work tends to be positively correlated with reward magnitude. But in nature, rewards earned by work are essential to survival (e.g., working to find water), and the payoff of that work can vary on long timescales (e.g., seasonally). Under these constraints, the strategy of working less when rewards are small could be fatal. We found that instead, rats in a closed economy did more work for water rewards when the rewards were stably smaller, a phenomenon also observed in human labor supply curves. Like human consumers, rats showed elasticity of demand, consuming far more water per day when its price in effort was lower. The neural mechanisms underlying such “rational” market behaviors remain largely unexplored. We propose a dynamic utility maximization model that can account for the dependence of rat labor supply (trials/day) on the wage rate (milliliter/trial) and also predict the temporal dynamics of when rats work. Based on data from mice, we hypothesize that glutamatergic neurons in the subfornical organ in lamina terminalis continuously compute the instantaneous marginal utility of voluntary work for water reward and causally determine the amount and timing of work.
Collapse
|
45
|
Affiliation(s)
- David H Ellison
- From the Oregon Clinical and Translational Research Institute, Oregon Health and Science University (D.H.E) and the VA Portland Health Care System (D.H.E.) - both in Portland; and LeDucq Transatlantic Network of Excellence (D.H.E., P.W.) and the Departments of Medicine and Physiology, Johns Hopkins University (P.W.) - both in Baltimore
| | - Paul Welling
- From the Oregon Clinical and Translational Research Institute, Oregon Health and Science University (D.H.E) and the VA Portland Health Care System (D.H.E.) - both in Portland; and LeDucq Transatlantic Network of Excellence (D.H.E., P.W.) and the Departments of Medicine and Physiology, Johns Hopkins University (P.W.) - both in Baltimore
| |
Collapse
|
46
|
Stamatakis AM, Resendez SL, Chen KS, Favero M, Liang-Guallpa J, Nassi JJ, Neufeld SQ, Visscher K, Ghosh KK. Miniature microscopes for manipulating and recording in vivo brain activity. Microscopy (Oxf) 2021; 70:399-414. [PMID: 34283242 PMCID: PMC8491619 DOI: 10.1093/jmicro/dfab028] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/02/2021] [Accepted: 07/19/2021] [Indexed: 12/23/2022] Open
Abstract
Here we describe the development and application of miniature integrated microscopes (miniscopes) paired with microendoscopes that allow for the visualization and manipulation of neural circuits in superficial and subcortical brain regions in freely behaving animals. Over the past decade the miniscope platform has expanded to include simultaneous optogenetic capabilities, electrically-tunable lenses that enable multi-plane imaging, color-corrected optics, and an integrated data acquisition platform that streamlines multimodal experiments. Miniscopes have given researchers an unprecedented ability to monitor hundreds to thousands of genetically-defined neurons from weeks to months in both healthy and diseased animal brains. Sophisticated algorithms that take advantage of constrained matrix factorization allow for background estimation and reliable cell identification, greatly improving the reliability and scalability of source extraction for large imaging datasets. Data generated from miniscopes have empowered researchers to investigate the neural circuit underpinnings of a wide array of behaviors that cannot be studied under head-fixed conditions, such as sleep, reward seeking, learning and memory, social behaviors, and feeding. Importantly, the miniscope has broadened our understanding of how neural circuits can go awry in animal models of progressive neurological disorders, such as Parkinson's disease. Continued miniscope development, including the ability to record from multiple populations of cells simultaneously, along with continued multimodal integration of techniques such as electrophysiology, will allow for deeper understanding into the neural circuits that underlie complex and naturalistic behavior.
Collapse
Affiliation(s)
| | | | - Kai-Siang Chen
- Inscopix Inc., 2462 Embarcadero Way, Palo Alto, CA 94303, USA
| | - Morgana Favero
- Inscopix Inc., 2462 Embarcadero Way, Palo Alto, CA 94303, USA
| | | | | | - Shay Q Neufeld
- Inscopix Inc., 2462 Embarcadero Way, Palo Alto, CA 94303, USA
| | - Koen Visscher
- Inscopix Inc., 2462 Embarcadero Way, Palo Alto, CA 94303, USA
| | - Kunal K Ghosh
- Inscopix Inc., 2462 Embarcadero Way, Palo Alto, CA 94303, USA
| |
Collapse
|
47
|
Kim A, Madara JC, Wu C, Andermann ML, Lowell BB. Neural basis for regulation of vasopressin secretion by anticipated disturbances in osmolality. eLife 2021; 10:66609. [PMID: 34585668 PMCID: PMC8601670 DOI: 10.7554/elife.66609] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 09/28/2021] [Indexed: 12/01/2022] Open
Abstract
Water balance, tracked by extracellular osmolality, is regulated by feedback and feedforward mechanisms. Feedback regulation is reactive, occurring as deviations in osmolality are detected. Feedforward or presystemic regulation is proactive, occurring when disturbances in osmolality are anticipated. Vasopressin (AVP) is a key hormone regulating water balance and is released during hyperosmolality to limit renal water excretion. AVP neurons are under feedback and feedforward regulation. Not only do they respond to disturbances in blood osmolality, but they are also rapidly suppressed and stimulated, respectively, by drinking and eating, which will ultimately decrease and increase osmolality. Here, we demonstrate that AVP neuron activity is regulated by multiple anatomically and functionally distinct neural circuits. Notably, presystemic regulation during drinking and eating are mediated by non-overlapping circuits that involve the lamina terminalis and hypothalamic arcuate nucleus, respectively. These findings reveal neural mechanisms that support differential regulation of AVP release by diverse behavioral and physiological stimuli. Fine-tuning the amount of water present in the body at any given time is a tight balancing act. The hormone vasopressin helps to ensure that organisms do not get too dehydrated by allowing water in the urine to be reabsorbed into the bloodstream. A group of vasopressin neurons in the brain trigger the release of the hormone if water levels get too low (as reflected by an increase in osmolality, the level of substances dissolved in a unit of blood). However, these cells also receive additional information that allows them to predict and respond to upcoming changes in water levels. For example, drinking water while dehydrated ‘switches off’ the neurons, even before osmolality is restored in the blood to normal levels. Eating, on the other hand, rapidly activates vasopressin neurons before the food is digested and blood osmolality increases as a result. How vasopressin neurons receive this ‘anticipatory’ information remains unclear. Kim et al. explored this question in mice by inhibiting different sets of brain cells one by one, and then examining whether the neurons could still exhibit anticipatory responses. This revealed a remarkable division of labor in the neural circuits that regulate vasopressin neurons: two completely different sets of neurons from distinct areas of the brain are dedicated to relaying anticipatory information about either water or food intake. These findings help to understand how healthy levels of water can be maintained in the body. Overall, they give a glimpse into the neural mechanisms that underlie anticipatory forms of regulation, which can also take place when hunger or thirst neurons ‘foresee’ that food or water will be consumed.
Collapse
Affiliation(s)
- Angela Kim
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, United States.,Program in Neuroscience, Harvard Medical School, Boston, United States
| | - Joseph C Madara
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, United States
| | - Chen Wu
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, United States
| | - Mark L Andermann
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, United States.,Program in Neuroscience, Harvard Medical School, Boston, United States
| | - Bradford B Lowell
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, United States.,Program in Neuroscience, Harvard Medical School, Boston, United States
| |
Collapse
|
48
|
Hicks AI, Kobrinsky S, Zhou S, Yang J, Prager-Khoutorsky M. Anatomical Organization of the Rat Subfornical Organ. Front Cell Neurosci 2021; 15:691711. [PMID: 34552469 PMCID: PMC8450496 DOI: 10.3389/fncel.2021.691711] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 08/10/2021] [Indexed: 11/14/2022] Open
Abstract
The subfornical organ (SFO) is a sensory circumventricular organ located along the anterodorsal wall of the third ventricle. SFO lacks a complete blood-brain barrier (BBB), and thus peripherally-circulating factors can penetrate the SFO parenchyma. These signals are detected by local neurons providing the brain with information from the periphery to mediate central responses to humoral signals and physiological stressors. Circumventricular organs are characterized by the presence of unique populations of non-neuronal cells, such as tanycytes and fenestrated endothelium. However, how these populations are organized within the SFO is not well understood. In this study, we used histological techniques to analyze the anatomical organization of the rat SFO and examined the distribution of neurons, fenestrated and non-fenestrated vasculature, tanycytes, ependymocytes, glia cells, and pericytes within its confines. Our data show that the shell of SFO contains non-fenestrated vasculature, while fenestrated capillaries are restricted to the medial-posterior core region of the SFO and associated with a higher BBB permeability. In contrast to non-fenestrated vessels, fenestrated capillaries are encased in a scaffold created by pericytes and embedded in a network of tanycytic processes. Analysis of c-Fos expression following systemic injections of angiotensin II or hypertonic NaCl reveals distinct neuronal populations responding to these stimuli. Hypertonic NaCl activates ∼13% of SFO neurons located in the shell. Angiotensin II-sensitive neurons represent ∼35% of SFO neurons and their location varies between sexes. Our study provides a comprehensive description of the organization of diverse cellular elements within the SFO, facilitating future investigations in this important brain area.
Collapse
Affiliation(s)
| | - Simona Kobrinsky
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Suijian Zhou
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Jieyi Yang
- Department of Physiology, McGill University, Montreal, QC, Canada
| | | |
Collapse
|
49
|
Berntson GG, Khalsa SS. Neural Circuits of Interoception. Trends Neurosci 2021; 44:17-28. [PMID: 33378653 DOI: 10.1016/j.tins.2020.09.011] [Citation(s) in RCA: 156] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/30/2020] [Accepted: 09/25/2020] [Indexed: 12/11/2022]
Abstract
The present paper considers recent progress in our understanding of the afferent/ascending neural pathways and neural circuits of interoception. Of particular note is the extensive role of rostral neural systems, including cortical systems, in the recognition of internal body states, and the reciprocal role of efferent/descending systems in the regulation of those states. Together these reciprocal interacting networks entail interoceptive circuits that play an important role in a broad range of functions beyond the homeostatic maintenance of physiological steady-states. These include the regulation of behavioral, cognitive, and affective processes across conscious and nonconscious levels of processing. We highlight recent advances and knowledge gaps that are important for accelerating progress in the study of interoception.
Collapse
Affiliation(s)
- Gary G Berntson
- Department of Psychology, Ohio State University, Columbus, OH, USA.
| | - Sahib S Khalsa
- Laureate Institute for Brain Research, Tulsa, OK, USA; Oxley College of Health Sciences, University of Tulsa, Tulsa, OK, USA
| |
Collapse
|
50
|
Ho V, Goh G, Tang XR, See KC. Underrecognition and undertreatment of thirst among hospitalized patients with restricted oral feeding and drinking. Sci Rep 2021; 11:13636. [PMID: 34211006 PMCID: PMC8249500 DOI: 10.1038/s41598-021-93048-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 06/21/2021] [Indexed: 11/08/2022] Open
Abstract
Thirst is distressing but overlooked by healthcare professionals. Patients experience thirst due to comorbidities, physical or cognitive limitations, and iatrogenesis. Nasogastric tube (NGT) use and nil-by-mouth(NBM) orders are common practices that can lead to thirst. However, thirst in these populations has never been formally studied. We aim to examine prevalence of recognition and treatment of thirst among NGT + NBM and NBM patients. Our descriptive study was conducted intermittently over 25 weeks, across 1.5 years, in 12 adult general medicine wards of a tertiary hospital. Cognitively intact NGT + NBM or NBM inpatients, defined as Abbreviated Mental Test score ≥ 8, were studied. One-time questionnaire was administered. Variables included: demography, co-morbidities, clinical condition, feeding route, thirst defined by thirst distress and/or intensity ≥ 3, pain, hunger and volume status. 88 NGT + NBM and NBM patients were studied. 69.3% suffered from thirst. Thirsty patients experienced significant thirst-related distress (mean score ± SD: 5.7 ± 2.5). Subjects with previous stroke and who were euvolemic tended towards thirst. 13.6% were asked about thirst by doctors or nurses. Thirst was a major source of patient distress in our study. We suggest that thirst needs to be actively identified and targeted to achieve person-centred care.
Collapse
Affiliation(s)
- Vanda Ho
- Department of Geriatric Medicine, National University Hospital, 5 Lower Kent Ridge Rd, Singapore, 119074, Singapore.
| | - Gordon Goh
- Yong Loo Lin School of Medicine, National University, Singapore, Singapore
| | - Xuan Rong Tang
- Yong Loo Lin School of Medicine, National University, Singapore, Singapore
| | - Kay Choong See
- Division of Respiratory and Critical Care Medicine, Department of Medicine, National University Hospital, Singapore, Singapore
| |
Collapse
|