1
|
Blumstein DM, Colella JP, Linder E, MacManes MD. High total water loss driven by low-fat diet in desert-adapted mice. J Mammal 2025; 106:293-303. [PMID: 40206330 PMCID: PMC11979454 DOI: 10.1093/jmammal/gyae093] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/30/2024] [Indexed: 04/11/2025] Open
Abstract
Availability of food resources is an important driver of survival. Populations must either relocate or adapt to persist in environments where food availability is changing. An optimal diet balances energy gain, water regulation, and nutrition. We used flow-through respirometry to characterize metabolic phenotypes of the desert-adapted Cactus Mouse (Peromyscus eremicus) under diurnally variable environmental conditions that mimic that of the Sonoran Desert. We treated mice with 2 different energetically equivalent diets, a standard diet and a low-fat diet, and measured energy expenditure, water loss rate, respiratory quotient, weight, and electrolyte levels. Mice fed the low-fat diet lost significantly more water than those on the standard diet. Despite being desert-adapted, our results suggest that cactus mice may have limited capacity to tolerate water deprivation if optimal foods become less abundant. Given that climate change is predicted to modify the distribution of food items, understanding these links may have important implications for long-term population viability for desert and non-desert-adapted animals alike.
Collapse
Affiliation(s)
- Danielle M Blumstein
- Molecular, Cellular, and Biomedical Sciences Department, University of New Hampshire, Durham, NH 03824, United States
| | - Jocelyn P Colella
- Biodiversity Institute and Department of Ecology and Evolutionary Biology, University of Kansas, Lawrence, KS 66045, United States
| | - Ernst Linder
- Department of Mathematics and Statistics, University of New Hampshire, Durham, NH 03824, United States
| | - Matthew D MacManes
- Molecular, Cellular, and Biomedical Sciences Department, University of New Hampshire, Durham, NH 03824, United States
| |
Collapse
|
2
|
Zhang D, Shen Z, He P, Wang J, Li D, Meng J, Zhang D, You J, Luo Y, Wang X, Zhuang X, Mu L, Zhang S, Han W, Cai H. The synergistic roles of MsRCI2B and MsRCI2E in the regulation of ion balance and ROS homeostasis in alfalfa under salt stress. Int J Biol Macromol 2025; 300:140093. [PMID: 39863229 DOI: 10.1016/j.ijbiomac.2025.140093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025]
Abstract
Under salt stress, plasma membrane proteins regulate ion homeostasis and the balance between reactive oxygen species (ROS). In this study, we investigated the functions of two small membrane proteins-MsRCI2B (tailless) and MsRCI2E (tailed)-encoded by the RCI2 (Rare Cold Inducible 2) gene family in Medicago sativa (alfalfa). We identified the distinct subcellular localization and expression patterns of these proteins under salt stress. Using yeast two-hybrid (Y2H), GST pull-down, and bimolecular fluorescence complementation (BiFC) assays, we confirmed the physical interactions between MsRCI2B and MsRCI2E. Transgenic alfalfa lines overexpressing MsRCI2(OE#RCI2) and co-expressing both MsRCI2B and MsRCI2E (OE#RCI2E-2B) were developed to explore their roles in salt tolerance. Interestingly, the C-terminal tail of MsRCI2E negatively affects salt tolerance; however, its interaction with MsRCI2B mitigates this adverse effect. To further understand the regulatory mechanisms, we screened for plasma membrane proteins (PMPs) that interact with MsRCI2B or MsRCI2E using a DUALmembrane yeast two-hybrid system. MsCaM1 interacts with MsRCI2B, whereas MsPIP1;4 and MsHVP1 specifically interact with MsRCI2E. Notably, the MsRCI2E-PIP1;4 interaction influenced the intracellular trafficking of PIP1;4, reducing its presence on the plasma membrane and thereby limiting the export of H2O2, which helps maintain ROS homeostasis. Additionally, the interaction between MsRCI2E and HVP1 stabilized ion homeostasis by decreasing Na+ concentration in the cytoplasm under salt stress. Overall, our study provides new insights into the molecular mechanisms through which MsRCI2B and MsRCI2E coordinate the ion and ROS balance under salt stress and offering promising strategies for enhancing crop tolerance to salinity.
Collapse
Affiliation(s)
- Depeng Zhang
- College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Zhongbao Shen
- Institute of Forage and Grassland Science, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Pin He
- College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Jianli Wang
- Institute of Forage and Grassland Science, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Donghuan Li
- College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Jing Meng
- College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Dongmei Zhang
- Institute of Forage and Grassland Science, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Jia You
- Institute of Forage and Grassland Science, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Yaqin Luo
- College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Xinsheng Wang
- College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Xu Zhuang
- Institute of Forage and Grassland Science, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Linlin Mu
- Institute of Forage and Grassland Science, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Shichao Zhang
- College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Weibo Han
- Institute of Forage and Grassland Science, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China.
| | - Hua Cai
- College of Life Science, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
3
|
Mendez-Hernandez R, Braga I, Bali A, Yang M, de Lartigue G. Vagal Sensory Gut-Brain Pathways That Control Eating-Satiety and Beyond. Compr Physiol 2025; 15:e70010. [PMID: 40229922 DOI: 10.1002/cph4.70010] [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: 02/03/2025] [Revised: 03/20/2025] [Accepted: 03/31/2025] [Indexed: 04/16/2025]
Abstract
The vagus nerve is the body's primary sensory conduit from gut to brain, traditionally viewed as a passive relay for satiety signals. However, emerging evidence reveals a far more complex system-one that actively encodes diverse aspects of meal-related information, from mechanical stretch to nutrient content, metabolic state, and even microbial metabolites. This review challenges the view of vagal afferent neurons (VANs) as simple meal-termination sensors and highlights their specialized subpopulations, diverse sensory modalities, and downstream brain circuits, which shape feeding behavior, metabolism, and cognition. We integrate recent advances from single-cell transcriptomics, neural circuit mapping, and functional imaging to examine how VANs contribute to gut-brain communication beyond satiety, including their roles in food reward and memory formation. By synthesizing the latest research and highlighting emerging directions for the field, this review provides a comprehensive update on vagal sensory pathways and their role as integrators of meal information.
Collapse
Affiliation(s)
- Rebeca Mendez-Hernandez
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Isadora Braga
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Avnika Bali
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mingxin Yang
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Guillaume de Lartigue
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
4
|
Murtaz A, Bourque CW. Actin fenestrae amplify the membrane response to hypertonic stress in osmosensory neurons. iScience 2025; 28:112042. [PMID: 40109380 PMCID: PMC11919590 DOI: 10.1016/j.isci.2025.112042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 11/20/2024] [Accepted: 02/13/2025] [Indexed: 03/22/2025] Open
Abstract
Osmosensory neurons detect hypertonic stress when ΔN-TRPV1 channels are activated through a push force delivered by microtubules during cell shrinking, and this process requires an essential yet unknown contribution from actin filaments. Here, we show that the actin cortex of these neurons feature fenestrations that allow for the formation of pits that magnify the osmotically induced displacement of the plasma membrane compared to that expected from uniform shrinking. Furthermore, we found that many N-terminal variant of the transient receptor potential vanilloid 1 (ΔN-TRPV1) channels attached to microtubules are aligned with fenestrae and such sites undergo greater hypertonicity-induced displacement than predicted by geometrically uniform cell shrinking. These results indicate that actin filaments contribute to the establishment of nanoscale architecture at sites which may optimize osmosensory transduction.
Collapse
Affiliation(s)
- Anzala Murtaz
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, 1650 Cedar Avenue, Montreal, QC H3G1A4, Canada
| | - Charles W Bourque
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, 1650 Cedar Avenue, Montreal, QC H3G1A4, Canada
| |
Collapse
|
5
|
Zou W, Deng S, Chen X, Ruan J, Wang H, Zhan W, Wang J, Liu Z, Yan Z. TMEM63B functions as a mammalian hyperosmolar sensor for thirst. Neuron 2025:S0896-6273(25)00130-8. [PMID: 40107268 DOI: 10.1016/j.neuron.2025.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/28/2025] [Accepted: 02/12/2025] [Indexed: 03/22/2025]
Abstract
Thirst drives animals to reinstate water homeostasis by fluid intake. An increase in blood osmolality is thought to induce thirst by activating a hyperosmolar sensor expressed in the subfornical organ (SFO), but the molecular identity of this sensor remains elusive. Here, we provide behavioral and functional evidence to show that TMEM63B functions as a mammalian hyperosmolar sensor for thirst in SFO neurons. First, we showed that TMEM63B is expressed in SFO excitatory neurons and required for the neuronal responses to hypertonic stimulation. More importantly, heterologously expressed TMEM63B is activated by hypertonic stimuli, and point mutations can alter the reversal potential of the channel. Additionally, purified TMEM63B in liposomes exhibits osmolarity-gated currents. Finally, Tmem63b knockout mice have profound deficits in thirst, and deleting TMEM63B within SFO neurons recapitulated this phenotype. Taken together, these results provide a molecular basis for thirst and suggest that TMEM63B is a mammalian hyperosmolar sensor for thirst.
Collapse
Affiliation(s)
- Wenjie Zou
- Department of Neurobiology, School of Basic Medicine, Capital Medical University, Beijing, China; Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Siqi Deng
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China; Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Xingyu Chen
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China; Department of Chemistry, Southern University of Science and Technology, Shenzhen, China
| | - Jiamin Ruan
- Department of Neurobiology, School of Basic Medicine, Capital Medical University, Beijing, China; Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Huize Wang
- Institute for Medical Physiology, Chinese Institutes for Medical Research, Beijing, China
| | - Wuqiang Zhan
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Jingxin Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Zhiyong Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Zhiqiang Yan
- Department of Neurobiology, School of Basic Medicine, Capital Medical University, Beijing, China; Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China; Institute for Medical Physiology, Chinese Institutes for Medical Research, Beijing, China.
| |
Collapse
|
6
|
Pohl F, Egan BM, Schneider DL, Mosley MC, Garcia MA, Hou S, Chiu CH, Kornfeld K. Environmental NaCl affects C. elegans development and aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.09.641258. [PMID: 40161617 PMCID: PMC11952357 DOI: 10.1101/2025.03.09.641258] [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: 04/02/2025]
Abstract
Sodium is an essential nutrient, but is toxic in excess. In humans, excessive dietary sodium can cause high blood pressure, which contributes to age-related diseases including stroke and heart disease. We used C. elegans to elucidate how sodium levels influence animal aging. Most experiments on this animal are conducted in standard culture conditions: Nematode Growth Medium (NGM) agar with a lawn of E. coli. Here, we report that the supplemental NaCl in standard NGM, 50 mM, accelerates aging and decreases lifespan. For comparison, we prepared NGM with reduced NaCl or excess NaCl. Considering reduced NaCl as a baseline, wild-type worms on standard NGM displayed normal development and fertility but reduced lifespan and health span, indicating toxicity in old animals. The long-lived mutants daf-2, age-1, and nuo-6, cultured on standard NGM, also displayed reduced lifespan. Thus, NaCl in standard NGM accelerates aging in multiple genetic backgrounds. Wild-type worms on excess NaCl displayed delayed development and reduced fertility, and reduced lifespan and health span, indicating toxicity in both young and old animals. These results suggest that young animals are relatively resistant to NaCl toxicity, but that aging causes progressive sensitivity, such that old animals display toxicity to both standard and excess NaCl. We investigated pathways that respond to NaCl. Young animals cultured with excess NaCl activated gpdh-1, a specific response to NaCl stress. Old animals cultured with excess NaCl activated gpdh-1 and hsp-6, a reporter for the mitochondrial unfolded protein response. Thus, excess NaCl activates multiple stress response pathways in older animals.
Collapse
Affiliation(s)
- Franziska Pohl
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- current affiliation: Nutrition Innovation Center, Standard Process, Inc., Kannapolis, North Carolina, USA
| | - Brian M. Egan
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- current affiliation: School of Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Daniel L. Schneider
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Matthew C. Mosley
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Micklaus A. Garcia
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sydney Hou
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Chen-Hao Chiu
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kerry Kornfeld
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
7
|
Gao J, Wang Y, Ren X, Nie Y, Li Y, Zhang Y, Huang S, Zhu D. Study on the Differences in the Thirst-Quenching Effects of Different Beverages Supplemented Before Exercise: A Randomized Crossover Trial. Nutrients 2025; 17:760. [PMID: 40077631 PMCID: PMC11901982 DOI: 10.3390/nu17050760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/13/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Background/Objectives: Different beverages may vary in their effectiveness at quenching thirst. This study aims to explore the impact of pre-exercise consumption of different types of beverages on thirst relief, providing scientific evidence to guide the selection of the most suitable beverage type. Methods: A randomized crossover design was used, recruiting 13 healthy male college students as participants. Each participant completed five exercise trials, with a 7-day interval between trials. In random order, participants consumed 6 mL/kg body weight of water, carbonated beverage, juice, electrolyte drink, or tea before exercise in each trial. Blood, saliva, and urine samples were collected before and after exercise; body weight was measured, and thirst sensation was recorded. Results: Body weight significantly decreased in all groups post-exercise (p < 0.05), with no significant differences between the beverage groups (p > 0.05). Post-exercise, serum Na+ concentrations significantly decreased in all beverage groups, with the electrolyte drink group showing a significantly different change compared to the other groups (p < 0.05). Serum K+ concentrations significantly increased post-exercise only in the electrolyte drink group (p < 0.05). No consistent trend was observed in the changes in serum Ca2+ concentrations before and after exercise. Serum Cl- concentrations post-exercise were significantly lower than pre-exercise in all groups except the electrolyte drink group (Group E) (p < 0.05). All five hydration protocols resulted in a decrease in plasma volume. There was no consistent pattern in the changes in urine osmolality before and after exercise. Salivary osmolality significantly increased post-exercise in all groups (p < 0.05). In terms of subjective thirst, the water supplementation group had the highest score. Conclusions: This study indicates that electrolyte drinks are more effective in maintaining physiological balance, while water is most effective in alleviating subjective thirst. The impact of different beverages on subjective thirst did not fully align with changes in physiological markers, suggesting that future research should comprehensively evaluate the relationship between subjective sensations and physiological changes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Dazhou Zhu
- Institute of Food and Nutrition Development, Ministry of Agriculture and Rural Affairs, Beijing 100081, China; (J.G.); (Y.W.); (X.R.); (Y.N.); (Y.L.); (Y.Z.); (S.H.)
| |
Collapse
|
8
|
Doua S, Germain N, Geandrot A, Defour C, Gay A, Massoubre C, Lang F, Estour B, Galusca B. A scoping review of circulating peptides assessments in anorexia nervosa: Uncovering diversity and nuanced findings. J Proteomics 2025; 312:105370. [PMID: 39716569 DOI: 10.1016/j.jprot.2024.105370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 12/15/2024] [Accepted: 12/17/2024] [Indexed: 12/25/2024]
Abstract
Understanding biological mechanisms underlying anorexia nervosa (AN) is necessary to develop care strategies. Despite many articles dedicated to peptides assessment in AN, there is no systematic review. A scoping review of circulating peptides published in relation to AN, comparing their results with those of controls, was conducted. Embase and PubMed databases were search from 1966 to 2022 (PROSPERO CRD42022323716). All original English articles, assessing peptides in AN (except classical markers) were analyzed. 1151 studies for 207 peptides, in 486 published articles were selected, and evidences/trends in AN were compared to controls. Fifteen clusters of function gathering peptides covering physiopathological aspects of AN were identified. This scoping review revealed a large variety of circulating peptides explored in AN. Some peptides presented with convincing results and helped understanding pathophysiologic aspects. Other peptides presented with nuanced results, partly due to insufficient number of studies, multiple assay techniques, inadequate sampling time, and lack of phenotyping. Conversion from bench-to-bed remains difficult and may explain why peptides evaluations did not currently lead to specific international recommendations or tailored therapeutic/preventive strategies. Peptide evaluation in anorexia nervosa could explore secretion profiles, and test it in well-phenotyped patients with AN, to conclude for potential clinical use, and finally design therapeutic tests.
Collapse
Affiliation(s)
- Sandra Doua
- TAPE Research Group, Jean Monnet University, Lyon University, Saint-Etienne, France; Endocrinology Department, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Natacha Germain
- TAPE Research Group, Jean Monnet University, Lyon University, Saint-Etienne, France; Endocrinology Department, University Hospital of Saint-Etienne, Saint-Etienne, France; Eating Disorder Reference Center, University hospital of Saint-Etienne, Saint-Etienne, France.
| | - Amale Geandrot
- TAPE Research Group, Jean Monnet University, Lyon University, Saint-Etienne, France
| | - Cloé Defour
- TAPE Research Group, Jean Monnet University, Lyon University, Saint-Etienne, France
| | - Aurélia Gay
- TAPE Research Group, Jean Monnet University, Lyon University, Saint-Etienne, France; Psychiatry Department, University hospital of Saint-Etienne, Saint-Etienne, France
| | - Catherine Massoubre
- TAPE Research Group, Jean Monnet University, Lyon University, Saint-Etienne, France; Eating Disorder Reference Center, University hospital of Saint-Etienne, Saint-Etienne, France; Psychiatry Department, University hospital of Saint-Etienne, Saint-Etienne, France
| | - Francois Lang
- TAPE Research Group, Jean Monnet University, Lyon University, Saint-Etienne, France; Eating Disorder Reference Center, University hospital of Saint-Etienne, Saint-Etienne, France; Psychiatry Department, University hospital of Saint-Etienne, Saint-Etienne, France
| | - Bruno Estour
- TAPE Research Group, Jean Monnet University, Lyon University, Saint-Etienne, France; Endocrinology Department, University Hospital of Saint-Etienne, Saint-Etienne, France; Eating Disorder Reference Center, University hospital of Saint-Etienne, Saint-Etienne, France
| | - Bogdan Galusca
- TAPE Research Group, Jean Monnet University, Lyon University, Saint-Etienne, France; Endocrinology Department, University Hospital of Saint-Etienne, Saint-Etienne, France; Eating Disorder Reference Center, University hospital of Saint-Etienne, Saint-Etienne, France
| |
Collapse
|
9
|
Qifti A, Adeeko A, Rennie M, McGlaughlin E, McKinnon D, Rosati B, Scarlata S. Hypoosmotic stress shifts transcription of circadian genes. Biophys J 2025; 124:565-573. [PMID: 39754358 PMCID: PMC11866948 DOI: 10.1016/j.bpj.2024.12.027] [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: 06/18/2024] [Revised: 09/26/2024] [Accepted: 12/23/2024] [Indexed: 01/06/2025] Open
Abstract
Cells respond to hypoosmotic stress by initial swelling followed by intracellular increases in the number of osmolytes and initiation of gene transcription that allow cells to adapt to the stress. Here, we have studied the genes that change expression under mild hypoosmotic stress for 12 and 24 h in rat cultured smooth muscle cells (WKO-3M22). We find shifts in the transcription of many genes, several of which are associated with circadian rhythm, such as per1, nr1d1, per2, dbp, and Ciart. To determine whether there is a connection between osmotic stress and circadian rhythm, we first subjected cells to hypoosmotic stress for 12 h, and find that Bmal1, a transcription factor whose nuclear localization promotes transit through the cell cycle, localizes to the cytoplasm, which may connect osmotic stress to cell cycle. Bmal1 nuclear localization recovers after 24 h and cell cycle resumes even though the osmotic stress remains elevated. We hypothesized that osmotic force is transmitted into the cell by deforming caveolae membrane domains releasing one of its structural proteins, cavin-1, which can travel to the nucleus and affect gene transcription. In support of this idea, we find that Bmal1 localization becomes independent of osmotic stress with cavin-1 downregulation, and Bmal1 localization is independent of osmotic stress in a cell line with low caveolae expression. These studies indicate that osmotic stress transiently arrests circadian rhythm and cell-cycle progression through caveolae deformation.
Collapse
Affiliation(s)
- Androniqi Qifti
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, Massachusetts
| | - Ayobami Adeeko
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, Massachusetts
| | - Madison Rennie
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, Massachusetts
| | - Elizabeth McGlaughlin
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, Massachusetts
| | - David McKinnon
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, New York; Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, New York
| | - Barbara Rosati
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, New York; Veterans Affairs Medical Center, Northport, New York
| | - Suzanne Scarlata
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, Massachusetts.
| |
Collapse
|
10
|
Krieger JP, Daniels D, Lee S, Mastitskaya S, Langhans W. Glucagon-Like Peptide-1 Links Ingestion, Homeostasis, and the Heart. Compr Physiol 2025; 15:e7. [PMID: 39887844 PMCID: PMC11790259 DOI: 10.1002/cph4.7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 11/02/2024] [Accepted: 11/05/2024] [Indexed: 02/01/2025]
Abstract
Glucagon-like peptide-1 (GLP-1), a hormone released from enteroendocrine cells in the distal small and large intestines in response to nutrients and other stimuli, not only controls eating and insulin release, but is also involved in drinking control as well as renal and cardiovascular functions. Moreover, GLP-1 functions as a central nervous system peptide transmitter, produced by preproglucagon (PPG) neurons in the hindbrain. Intestinal GLP-1 inhibits eating by activating vagal sensory neurons directly, via GLP-1 receptors (GLP-1Rs), but presumably also indirectly, by triggering the release of serotonin from enterochromaffin cells. GLP-1 enhances glucose-dependent insulin release via a vago-vagal reflex and by direct action on beta cells. Finally, intestinal GLP-1 acts on the kidneys to modulate electrolyte and water movements, and on the heart, where it provides numerous benefits, including anti-inflammatory, antiatherogenic, and vasodilatory effects, as well as protection against ischemia/reperfusion injury and arrhythmias. Hindbrain PPG neurons receive multiple inputs and project to many GLP-1R-expressing brain areas involved in reward, autonomic functions, and stress. PPG neuron-derived GLP-1 is involved in the termination of large meals and is implicated in the inhibition of water intake. This review details GLP-1's roles in these interconnected systems, highlighting recent findings and unresolved issues, and integrating them to discuss the physiological and pathological relevance of endogenous GLP-1 in coordinating these functions. As eating poses significant threats to metabolic, fluid, and immune homeostasis, the body needs mechanisms to mitigate these challenges while sustaining essential nutrient intake. Endogenous GLP-1 plays a crucial role in this "ingestive homeostasis."
Collapse
Affiliation(s)
- Jean-Philippe Krieger
- Jean-Philippe Krieger, Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, Winterthurerstr. 260, 8057 Zurich
| | - Derek Daniels
- Department of Biological Sciences and the Center for Ingestive Behavior Research, University at Buffalo, the State University of New York, Buffalo NY 14260 USA
| | - Shin Lee
- Shin J. Lee, Neurimmune AG, Wagistrasse 18, 8952 Schlieren, Switzerland
| | - Svetlana Mastitskaya
- Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, Dept. of Health Sciences and Technology, ETH Zurich, 8603 Schwerzenbach, Switzerland
| |
Collapse
|
11
|
Doua S, Germain N, Merabet M, Redouté J, Boutet C, Schneider F, Hammour A, Gay A, Massoubre C, Estour B, Galusca B. Circadian copeptin and oxytocin profiles in anorexia nervosa: Exploring the interplay with neurohypophysis opioid tone. EUROPEAN EATING DISORDERS REVIEW 2025; 33:53-66. [PMID: 39032117 PMCID: PMC11617818 DOI: 10.1002/erv.3125] [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: 03/03/2024] [Revised: 05/21/2024] [Accepted: 06/29/2024] [Indexed: 07/22/2024]
Abstract
CONTEXT Neurohypophysis (NH) function in eating disorders (ED) remains poorly elucidated. Studies on vasopressin and oxytocin display inconclusive findings regarding their levels and associations with psychological complications in ED. The profile of opioid tone, a crucial NH activity regulator, is also unknown. OBJECTIVE To characterise the circadian profile of NH hormones and NH opioid tone using positron emission tomography/MRI (PET/MRI) imaging in patients with ED compared to healthy controls. METHODS Twelve-point plasma circadian profiles of copeptin and oxytocin, alongside nutritional and psychological scores, were assessed in age-matched female participants: 13 patients with anorexia nervosa restrictive-type (ANR), 12 patients recovered from AN (ANrec), 14 patients with bulimia nervosa and 12 controls. Neurohypophysis PET/MRI [11C] diprenorphin binding potential (BPND) was evaluated in AN, ANrec and controls. RESULTS Results revealed lower copeptin circadian levels in both ANR and ANrec compared to controls, with no oxytocin differences. Bulimia nervosa exhibited elevated copeptin and low oxytocin levels. [11C] diprenorphin pituitary binding was fully localised in NH. Anorexia nervosa restrictive-type displayed lower NH [11C] diprenorphin BPND (indicating higher opioid tone) and volume than controls. In ANR, copeptin inversely correlated with osmolarity. Neurohypophysis [11C] diprenorphin BPND did not correlated with copeptin or oxytocin. CONCLUSION Copeptin demonstrated significant group differences, highlighting its potential diagnostic and prognostic value. Oxytocin levels exhibited conflicting results, questioning the reliability of peripheral blood assessment. Increased NH opioid tone in anorexia nervosa may influence the vasopressin or oxytocin release, suggesting potential therapeutic applications.
Collapse
Affiliation(s)
- Sandra Doua
- TAPE Research GroupJean Monnet UniversityLyon UniversitySaint‐EtienneFrance
- Endocrinology DepartmentUniversity Hospital of Saint‐EtienneSaint‐EtienneFrance
| | - Natacha Germain
- TAPE Research GroupJean Monnet UniversityLyon UniversitySaint‐EtienneFrance
- Endocrinology DepartmentUniversity Hospital of Saint‐EtienneSaint‐EtienneFrance
- Eating Disorder Reference CenterUniversity Hospital of Saint‐EtienneSaint‐EtienneFrance
| | - Manel Merabet
- TAPE Research GroupJean Monnet UniversityLyon UniversitySaint‐EtienneFrance
- Endocrinology DepartmentUniversity Hospital of Saint‐EtienneSaint‐EtienneFrance
| | | | - Claire Boutet
- TAPE Research GroupJean Monnet UniversityLyon UniversitySaint‐EtienneFrance
- Imaging DepartmentUniversity Hospital of Saint‐EtienneSaint‐EtienneFrance
| | - Fabien Schneider
- TAPE Research GroupJean Monnet UniversityLyon UniversitySaint‐EtienneFrance
- Imaging DepartmentUniversity Hospital of Saint‐EtienneSaint‐EtienneFrance
| | - Amira Hammour
- TAPE Research GroupJean Monnet UniversityLyon UniversitySaint‐EtienneFrance
- Endocrinology DepartmentUniversity Hospital of Saint‐EtienneSaint‐EtienneFrance
- Eating Disorder Reference CenterUniversity Hospital of Saint‐EtienneSaint‐EtienneFrance
| | - Aurélia Gay
- TAPE Research GroupJean Monnet UniversityLyon UniversitySaint‐EtienneFrance
- Eating Disorder Reference CenterUniversity Hospital of Saint‐EtienneSaint‐EtienneFrance
- Psychiatry DepartmentUniversity Hospital of Saint‐EtienneSaint‐EtienneFrance
| | - Catherine Massoubre
- TAPE Research GroupJean Monnet UniversityLyon UniversitySaint‐EtienneFrance
- Eating Disorder Reference CenterUniversity Hospital of Saint‐EtienneSaint‐EtienneFrance
- Psychiatry DepartmentUniversity Hospital of Saint‐EtienneSaint‐EtienneFrance
| | - Bruno Estour
- TAPE Research GroupJean Monnet UniversityLyon UniversitySaint‐EtienneFrance
- Endocrinology DepartmentUniversity Hospital of Saint‐EtienneSaint‐EtienneFrance
- Eating Disorder Reference CenterUniversity Hospital of Saint‐EtienneSaint‐EtienneFrance
| | - Bogdan Galusca
- TAPE Research GroupJean Monnet UniversityLyon UniversitySaint‐EtienneFrance
- Endocrinology DepartmentUniversity Hospital of Saint‐EtienneSaint‐EtienneFrance
- Eating Disorder Reference CenterUniversity Hospital of Saint‐EtienneSaint‐EtienneFrance
| |
Collapse
|
12
|
Howard PG, Zou P, Zhang Y, Huang F, Tesic V, Wu CYC, Lee RHC. Serum/glucocorticoid regulated kinase 1 (SGK1) in neurological disorders: pain or gain. Exp Neurol 2024; 382:114973. [PMID: 39326820 PMCID: PMC11536509 DOI: 10.1016/j.expneurol.2024.114973] [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: 08/26/2024] [Revised: 09/18/2024] [Accepted: 09/21/2024] [Indexed: 09/28/2024]
Abstract
Serum/Glucocorticoid Regulated Kinase 1 (SGK1), a serine/threonine kinase, is ubiquitous across a wide range of tissues, orchestrating numerous signaling pathways and associated with various human diseases. SGK1 has been extensively explored in diverse types of immune and inflammatory diseases, cardiovascular disorders, as well as cancer metastasis. These studies link SGK1 to cellular proliferation, survival, metabolism, membrane transport, and drug resistance. Recently, increasing research has focused on SGK1's role in neurological disorders, including a variety of neurodegenerative diseases (e.g., Alzheimer's disease, Huntington's disease and Parkinson's disease), brain injuries (e.g., cerebral ischemia and traumatic brain injury), psychiatric conditions (e.g., depression and drug addiction). SGK1 is emerging as an increasingly compelling therapeutic target across the spectrum of neurological disorders, supported by the availability of several effective agents. However, the conclusions of many studies observing the prevalence and function of SGK1 in neurological disorders are contradictory, necessitating a review of the SGK1 research within neurological disorders. Herein, we review recent literature on SGK1's primary functions within the nervous system and its impacts within different neurological disorders. We summarize significant findings, identify research gaps, and outline possible future research directions based on the current understanding of SGK1 to help further progress the understanding and treatment of neurological disorders.
Collapse
Affiliation(s)
- Peyton Grace Howard
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA; Department of Neurology, Shreveport, Louisiana State University Health, LA, USA
| | - Peibin Zou
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA; Department of Neurology, Shreveport, Louisiana State University Health, LA, USA
| | - Yulan Zhang
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA; Department of Neurology, Shreveport, Louisiana State University Health, LA, USA
| | - Fang Huang
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA; Department of Neurology, Shreveport, Louisiana State University Health, LA, USA
| | - Vesna Tesic
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA; Department of Neurology, Shreveport, Louisiana State University Health, LA, USA
| | - Celeste Yin-Chieh Wu
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA; Department of Neurology, Shreveport, Louisiana State University Health, LA, USA.
| | - Reggie Hui-Chao Lee
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA; Department of Neurology, Shreveport, Louisiana State University Health, LA, USA; Department of Department of Cell Biology & Anatomy, Louisiana State University Health, Shreveport, LA, USA.
| |
Collapse
|
13
|
Takeda T, Takeda S, Kakigi A. The clinical manifestation and treatment of Meniere's Disease from the viewpoint of the water homeostasis of the inner ear. Auris Nasus Larynx 2024; 51:905-910. [PMID: 39244939 DOI: 10.1016/j.anl.2024.08.002] [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: 06/25/2024] [Revised: 07/29/2024] [Accepted: 08/08/2024] [Indexed: 09/10/2024]
Abstract
Endolymphatic hydrops, a pathological feature of Ménière's disease, has been experimentally and clinically confirmed to be influenced by the blood circulation of vasopressin (VP). VP is a well-known hormonal regulator of water homeostasis. In addition, VP is influenced by various environmental changes, dehydration, fluctuation of atmospheric pressure, pregnancy, and other factors. Furthermore, VP is a key regulator of the hypothalamic-pituitary-adrenal (HPA) axis. The HPA axis is a major neuroendocrine system that controls reactions to emotional and physical stresses, as well as the sleep/wake cycle (circadian rhythm). Therefore, VP is susceptible to change via the HPA axis. This review considers possible mechanisms of the formation of endolymphatic hydrops from the perspective of the vasopressin-aquaporin 2 system.
Collapse
Affiliation(s)
- Taizo Takeda
- Department of Otolaryngology, Kochi Medical School, Nankoku, Kochi, Japan
| | | | - Akinobu Kakigi
- Department of Otolaryngology-Head & Neck Surgery, Kobe University, Graduate School of Medicine, Hyogo, Japan.
| |
Collapse
|
14
|
Junkins MS, Feng NY, Merriman DK, Bagriantsev SN, Gracheva EO. Suppression of neurons in circumventricular organs enables months-long survival without water in thirteen-lined ground squirrels. Science 2024; 386:1048-1055. [PMID: 39607925 PMCID: PMC11853976 DOI: 10.1126/science.adp8358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 10/28/2024] [Indexed: 11/30/2024]
Abstract
Water deprivation is a life-threatening condition that engages a protective physiological response to couple osmolyte retention with potentiation of thirst. This response, typical for most mammals, safeguards against short-term water deprivation but fails in the long term. Thirteen-lined ground squirrels (Ictidomys tridecemlineatus) use the short-term response during summer, whereas during winter, they lack thirst and survive without water for months. In this work, we show that long-term thirst suppression occurs despite hormonal and behavioral signs of a substantial fluid deficit and originates from hypoactivity of neurons in the circumventricular organs, which exhibit marked functional suppression during winter that blunts their sensitivity to thirst cues. Our work reveals a notable capacity of the evolutionarily conserved brain regions that control fluid homeostasis in mammals to enable long-term survival without water.
Collapse
Affiliation(s)
- Madeleine S. Junkins
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Neuroscience and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, USA
| | - Ni Y. Feng
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Neuroscience and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, USA
| | - Dana K. Merriman
- Department of Biology, University of Wisconsin–Oshkosh, Oshkosh, WI, USA
| | - Sviatoslav N. Bagriantsev
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Elena O. Gracheva
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Neuroscience and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, USA
- Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
15
|
Augusto I, Girard-Dias W, Schoijet A, Alonso GD, Portugal RV, de Souza W, Jimenez V, Miranda K. Quantitative assessment of the nanoanatomy of the contractile vacuole complex in Trypanosoma cruzi. Life Sci Alliance 2024; 7:e202402826. [PMID: 39074903 PMCID: PMC11287019 DOI: 10.26508/lsa.202402826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 07/31/2024] Open
Abstract
Trypanosoma cruzi uses various mechanisms to cope with osmotic fluctuations during infection, including the remodeling of organelles such as the contractile vacuole complex (CVC). Little is known about the morphological changes of the CVC during pulsation cycles occurring upon osmotic stress. Here, we investigated the structure-function relationship between the CVC and the flagellar pocket domain where fluid discharge takes place-the adhesion plaque-during the CVC pulsation cycle. Using TcrPDEC2 and TcVps34 overexpressing mutants, known to have low and high efficiency for osmotic responses, we described a structural phenotype for the CVC that matches their corresponding physiological responses. Quantitative tomography provided data on the volume of the CVC and spongiome connections. Changes in the adhesion plaque during the pulsation cycle were also quantified and a dense filamentous network was observed. Together, the results suggest that the adhesion plaque mediates fluid discharge from the central vacuole, revealing new aspects of the osmoregulatory system in T. cruzi.
Collapse
Affiliation(s)
- Ingrid Augusto
- Laboratório de Ultraestrutura Celular Hertha Meyer, Centro de Pesquisa em Medicina de Precisão, Instituto de Biofísica Carlos Chagas Filho and Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Wendell Girard-Dias
- Laboratório de Ultraestrutura Celular Hertha Meyer, Centro de Pesquisa em Medicina de Precisão, Instituto de Biofísica Carlos Chagas Filho and Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Plataforma de Microscopia Eletrônica Rudolf Barth, Instituto Oswaldo Cruz - Fiocruz, Rio de Janeiro, Brazil
| | - Alejandra Schoijet
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres", Buenos Aires, Argentina
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Guillermo Daniel Alonso
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres", Buenos Aires, Argentina
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Rodrigo V Portugal
- Laboratório Nacional de Nanotecnologia, Centro Nacional de Pesquisa em Energia e Materiais, Campinas, Brazil
- Programa de Biotecnologia, Universidade Federal do ABC, Santo André, Brazil
| | - Wanderley de Souza
- Laboratório de Ultraestrutura Celular Hertha Meyer, Centro de Pesquisa em Medicina de Precisão, Instituto de Biofísica Carlos Chagas Filho and Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagem - Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Centro Multiusuário para Análise de Fenômenos Biomédicos, Universidade do Estado do Amazonas, Manaus, Brazil
| | - Veronica Jimenez
- Department of Biological Sciences, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA, USA
| | - Kildare Miranda
- Laboratório de Ultraestrutura Celular Hertha Meyer, Centro de Pesquisa em Medicina de Precisão, Instituto de Biofísica Carlos Chagas Filho and Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagem - Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Centro Multiusuário para Análise de Fenômenos Biomédicos, Universidade do Estado do Amazonas, Manaus, Brazil
| |
Collapse
|
16
|
De Luca LA, Laurin M, Menani JV. Control of fluid intake in dehydrated rats and evolution of sodium appetite. Physiol Behav 2024; 284:114642. [PMID: 39032667 DOI: 10.1016/j.physbeh.2024.114642] [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: 12/17/2023] [Revised: 07/04/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
The objective of the present work is to examine from a new perspective the existence of causal factors not predicted by the classical theory that thirst and sodium appetite are two distinct motivations. For example, we ask why water deprivation induces sodium appetite, thirst is not "water appetite", and intracellular dehydration potentially causes sodium appetite. Contrary to the classical theory, we suggest that thirst first, and sodium appetite second, designate a temporal sequence underlying the same motivation. The single motivation becomes an "intervenient variable" a concept borrowed from the literature, fully explained in the text, between causes of dehydration (extracellular, intracellular, or both together), and respective behavioral responses subserved by hindbrain-dependent inhibition (e.g., lateral parabrachial nucleus) and forebrain facilitation (e.g., angiotensin II). A corollary is homology between rat sodium appetite and marine teleost thirst-like motivation that we name "protodipsia". The homology argument rests on similarities between behavior (salty water intake) and respective neuroanatomical as well as functional mechanisms. Tetrapod origin in a marine environment provides additional support for the homology. The single motivation hypothesis is also consistent with ingestive behaviors in nature given similarities (e.g., thirst producing brackish water intake) between the behavior of the laboratory rat and wild animals, rodents included. The hypotheses of single motivation and homology might explain why hyperosmotic rats, or eventually any other hyperosmotic tetrapod, shows paradoxical signs of sodium appetite. They might also explain how ingestive behaviors determined by dehydration and subserved by hindbrain inhibitory mechanisms contributed to tetrapod transition from sea to land.
Collapse
Affiliation(s)
- Laurival A De Luca
- Department of Physiology & Pathology, School of Dentistry, São Paulo State University (UNESP), 14801-903 Araraquara, São Paulo, Brazil.
| | - Michel Laurin
- CR2P, UMR 7207, CNRS/MNHN/SU, Muséum National d'Histoire Naturelle, Bâtiment de Géologie, CP 48, F-75231 Paris cedex 05, France
| | - José Vanderlei Menani
- Department of Physiology & Pathology, School of Dentistry, São Paulo State University (UNESP), 14801-903 Araraquara, São Paulo, Brazil
| |
Collapse
|
17
|
Çelik SŞ, Mert S, Arslan HN. The Effect of Cold Oral Applications in the Management of Postoperative Thirst: A Systematic Review. J Perianesth Nurs 2024; 39:907-914. [PMID: 38556965 DOI: 10.1016/j.jopan.2023.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 11/16/2023] [Accepted: 11/28/2023] [Indexed: 04/02/2024]
Abstract
PURPOSE Thirst is one of the most bothersome symptoms experienced by surgical patients. Effective thirst intervention and management in the Post Anesthesia Care Unit (PACU) and hospital wards is critical because patients are less sedated and more aware than in the past. There is a need to review the literature on the identification and management of thirst in the inpatient and PACU settings. The aim of this systematic review was to examine the available evidence on the effectiveness of oral cold applications on thirst in postoperative patients. DESIGN This was a systematic review study. Articles in PUBMED, Web of Science, ScienceDirect, TÜBİTAK-ULAKBİM, and TRDizin databases between January 2008 and January 2023 that included oral cold applications to relieve the thirst of patients in the postoperative period were included. METHODS The PICOT-SD (Patients Interventions Comparison Outcome Time-Study Design) method was used as an eligibility criterion for inclusion in the study. The eligibility criteria included that the articles were written in English-Turkish and within the target dates, the studies included nursing interventions, the primary outcome of the studies was thirst, and the study sample included postoperative patients. The risk of bias was assessed using the RoB2 tool developed by Cochrane. FINDINGS A total of 254 articles were retrieved from the databases using the specified keywords. 244 articles did not meet the study criteria: 30 were excluded because they were not interventional studies, 61 were not conducted in a postoperative population, 56 were duplicates, and 79 were not on a related topic. A total of 10 studies consisting of randomized controlled trials and quasi-experimental articles met the criteria for our review. Oral cold applications effectively reduced the thirst rate of postoperative patients and improved their health-related quality of life. The intervention has also been shown to reduce other anesthesia-related complications. CONCLUSIONS This systematic review concluded that cold oral applications have promising effects on thirst, dry mouth, and health-related quality of life. Cold oral applications are cost-effective and suitable for large-scale health care applications.
Collapse
Affiliation(s)
| | - Sabiha Mert
- Istanbul Galata University, Vocational School, Anesthesia Program, Istanbul, Turkey
| | | |
Collapse
|
18
|
Hou C, Chen G, Li D, Wang X, Liu X, Cui D, Jiang Y, Liu Y, Wang P, Wang YF, Meng D, Jia S. Intranasal Application of Diluted Saline Alleviates Ischemic Brain Injury in Association with Suppression of Vasopressin Neurons. Neuroendocrinology 2024; 114:1090-1111. [PMID: 39348823 DOI: 10.1159/000541648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 09/25/2024] [Indexed: 10/02/2024]
Abstract
INTRODUCTION Cerebral swelling and brain injury in ischemic stroke are closely related to increased vasopressin (VP) secretion. How to alleviate ischemic brain injury by suppressing VP hypersecretion through simply available approaches remains to be established. METHODS Using a rat model of middle cerebral artery occlusion (MCAO), testing effects of the intranasal application of low concentration saline-0.09% NaCl (IAL) on brain damage, VP neuronal activity, synaptic inputs, astrocytic plasticity, and olfactory bulb (OB) activity in immunohistochemistry, patch-clamp recording, Western blotting, and co-immunoprecipitation. RESULTS IAL reduced MCAO-evoked neurological disorders, brain swelling, injury and loss of neurons, increase in c-Fos expression, and excitation of supraoptic VP neurons. The effects of IAL on VP neurons were associated with its suppression of MCAO-evoked increase in the frequency of excitatory synaptic inputs and decrease in the expression of glial fibrillary acidic protein (GFAP) filaments around VP neurons. MCAO and IAL also caused similar but weaker reactions in putative oxytocin neurons. In the OB, MCAO increased the firing rate of mitral cells on the MCAO side, which was reduced by IAL. A direct hypotonic challenge of OB slices increased the expression of glutamine synthetase and GFAP filaments in the glomerular bodies while reducing the firing rate of mitral cells. Blocking aquaporin 4 activity in the supraoptic and paraventricular nuclei on the MCAO side reduced MCAO-evoked VP increase and brain damage. CONCLUSION IAL reduces ischemic stroke-evoked brain injury in association with suppression of VP neuronal activity through reducing excitatory synaptic inputs and astrocytic process retraction, which likely result from reducing mitral cell activation in ischemic side.
Collapse
Affiliation(s)
- Chunmei Hou
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Guichuan Chen
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
- Department of Medicine, Ziyang College of Dental Technology, Ziyang, China
| | - Di Li
- Department of Otolaryngology, The First Affiliated Hospital, Jiamusi University, Jiamusi, China
| | - Xiaoran Wang
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Xiaoyu Liu
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Dan Cui
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
- School of Nursing, Dali University, Dali, China
| | - Yunhao Jiang
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yang Liu
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
- Department of Clinical Laboratory, Quzhou People's Hospital, Quzhou, China
| | - Ping Wang
- Department of Genetics, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Yu-Feng Wang
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Dexin Meng
- Department of Physiology, School of Basic Medical Sciences, Jiamusi University, Jiamusi, China
| | - Shuwei Jia
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| |
Collapse
|
19
|
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
|
20
|
Rabinowitz J, Darawshi M, Burak N, Boehm M, Dmitrieva NI. Risk for hypertension and heart failure linked to high normal serum sodium and tonicity in electronic medical records. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.05.24309996. [PMID: 39072033 PMCID: PMC11275695 DOI: 10.1101/2024.07.05.24309996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Background and Aims Population aging is fueling an epidemic of age-related chronic diseases. Managing risk factors and lifestyle interventions have proven effective in disease prevention. Epidemiological studies have linked markers of poor hydration with higher risk of chronic diseases and premature mortality. Many individuals do not adhere to recommended hydration levels and could benefit from improved hydration habits. Our study evaluates the use of electronic medical records to confirm the relationship between inadequate hydration and the risk of chronic diseases, which may inform hydration-focused interventions in general healthcare. Methods We analyzed 20-year electronic medical records for 411,029 adults from Israel's Leumit Healthcare Services. Hydration status was assessed using serum sodium and tonicity. We included adults without significant chronic diseases or water balance issues, defined as having normal serum sodium (135-146 mmol/l) and no diagnosis of diabetes. We used Cox proportional hazards models, adjusted for age, to assess the risk of developing hypertension and heart failure. Results Our findings showed an increased risk of hypertension with elevated serum sodium levels: a 12% rise for the 140-142 mmol/l group and 30% for levels above 143 mmol/l (HR1.30, 95%CI:1.26-1.34). Tonicity over 287 mosmol/kg was associated with a 19% increased risk of hypertension (HR1.19, 95%CI:1.17-1.22). The risk of heart failure also increased, reaching 20% for sodium levels above 143 mmol/l (HR1.20,95%CI:1.12-1.29) and 16% for tonicity above 289 mosmol/kg (HR1.16, 95%CI: 1.10-1.22). The association between sodium and hypertension was observed across genders, while the risk of heart failure was more pronounced in females. Within the healthy Leumit cohort, 19% had serum sodium levels within the 143-146 mmol/l range, and 39% were in the 140-142 mmol/l range. Conclusions Data analysis from electronic medical records identified a link between serum sodium of 140 mmol/l and above and increased risk of hypertension and heart failure in the general Israeli population. Identifying individuals with high-normal sodium values in healthcare records could guide improvements in hydration habits, potentially leading to better health outcomes.
Collapse
|
21
|
Nair-Collins M. The Uniform Determination of Death Act is Not Changing. Will Physicians Continue to Misdiagnose Brain Death? THE AMERICAN JOURNAL OF BIOETHICS : AJOB 2024:1-12. [PMID: 38967488 DOI: 10.1080/15265161.2024.2371129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/06/2024]
Abstract
Efforts to revise the Uniform Determination of Death Act in order to align law with medical practice have failed. Medical practice must now align with the law. People who are not dead under the law that defines death should not be declared dead. There is no compelling reason to continue the practice of declaring legally living persons to be dead.
Collapse
|
22
|
Rein JL, Mackie K, Kleyman TR, Satlin LM. Cannabinoid receptor type 1 activation causes a water diuresis by inducing an acute central diabetes insipidus in mice. Am J Physiol Renal Physiol 2024; 326:F917-F930. [PMID: 38634131 PMCID: PMC12040313 DOI: 10.1152/ajprenal.00320.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 04/01/2024] [Accepted: 04/05/2024] [Indexed: 04/19/2024] Open
Abstract
Cannabis and synthetic cannabinoid consumption are increasing worldwide. Cannabis contains numerous phytocannabinoids that act on the G protein-coupled cannabinoid receptor type 1 (CB1R) and cannabinoid receptor type 2 expressed throughout the body, including the kidney. Essentially every organ, including the kidney, produces endocannabinoids, which are endogenous ligands to these receptors. Cannabinoids acutely increase urine output in rodents and humans, thus potentially influencing total body water and electrolyte homeostasis. As the kidney collecting duct (CD) regulates total body water, acid/base, and electrolyte balance through specific functions of principal cells (PCs) and intercalated cells (ICs), we examined the cell-specific immunolocalization of CB1R in the mouse CD. Antibodies against either the C-terminus or N-terminus of CB1R consistently labeled aquaporin 2 (AQP2)-negative cells in the cortical and medullary CD and thus presumably ICs. Given the well-established role of ICs in urinary acidification, we used a clearance approach in mice that were acid loaded with 280 mM NH4Cl for 7 days and nonacid-loaded mice treated with the cannabinoid receptor agonist WIN55,212-2 (WIN) or a vehicle control. Although WIN had no effect on urinary acidification, these WIN-treated mice had less apical + subapical AQP2 expression in PCs compared with controls and developed acute diabetes insipidus associated with the excretion of large volumes of dilute urine. Mice maximally concentrated their urine when WIN and 1-desamino-8-d-arginine vasopressin [desmopressin (DDAVP)] were coadministered, consistent with central rather than nephrogenic diabetes insipidus. Although ICs express CB1R, the physiological role of CB1R in this cell type remains to be determined.NEW & NOTEWORTHY The CB1R agonist WIN55,212-2 induces central diabetes insipidus in mice. This research integrates existing knowledge regarding the diuretic effects of cannabinoids and the influence of CB1R on vasopressin secretion while adding new mechanistic insights about total body water homeostasis. Our findings provide a deeper understanding about the potential clinical impact of cannabinoids on human physiology and may help identify targets for novel therapeutics to treat water and electrolyte disorders such as hyponatremia and volume overload.
Collapse
Affiliation(s)
- Joshua L Rein
- Renal Section, Department of Medicine, James J. Peters Veterans Affairs Medical Center, Bronx, New York, United States
- Barbara T. Murphy Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Ken Mackie
- Gill Center for Biomolecular Medicine, Indiana University, Bloomington, Indiana, United States
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana, United States
| | - Thomas R Kleyman
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Lisa M Satlin
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| |
Collapse
|
23
|
Zhao Z, Covelo A, Couderc Y, Mitra A, Varilh M, Wu Y, Jacky D, Fayad R, Cannich A, Bellocchio L, Marsicano G, Beyeler A. Cannabinoids regulate an insula circuit controlling water intake. Curr Biol 2024; 34:1918-1929.e5. [PMID: 38636514 DOI: 10.1016/j.cub.2024.03.053] [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: 03/11/2022] [Revised: 02/29/2024] [Accepted: 03/25/2024] [Indexed: 04/20/2024]
Abstract
The insular cortex, or insula, is a large brain region involved in the detection of thirst and the regulation of water intake. However, our understanding of the topographical, circuit, and molecular mechanisms for controlling water intake within the insula remains parcellated. We found that type-1 cannabinoid (CB1) receptors in the insular cortex cells participate in the regulation of water intake and deconstructed the circuit mechanisms of this control. Topographically, we revealed that the activity of excitatory neurons in both the anterior insula (aIC) and posterior insula (pIC) increases in response to water intake, yet only the specific removal of CB1 receptors in the pIC decreases water intake. Interestingly, we found that CB1 receptors are highly expressed in insula projections to the basolateral amygdala (BLA), while undetectable in the neighboring central part of the amygdala. Thus, we recorded the neurons of the aIC or pIC targeting the BLA (aIC-BLA and pIC-BLA) and found that they decreased their activity upon water drinking. Additionally, chemogenetic activation of pIC-BLA projection neurons decreased water intake. Finally, we uncovered CB1-dependent short-term synaptic plasticity (depolarization-induced suppression of excitation [DSE]) selectively in pIC-BLA, compared with aIC-BLA synapses. Altogether, our results support a model where CB1 receptor signaling promotes water intake by inhibiting the pIC-BLA pathway, thereby contributing to the fine top-down control of thirst responses.
Collapse
Affiliation(s)
- Zhe Zhao
- INSERM 1215, Neurocentre Magendie, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France; Max Planck Florida Institute for Neuroscience, 1 Max Planck Way, Jupiter, FL 33458, USA
| | - Ana Covelo
- INSERM 1215, Neurocentre Magendie, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France
| | - Yoni Couderc
- INSERM 1215, Neurocentre Magendie, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France
| | - Arojit Mitra
- INSERM 1215, Neurocentre Magendie, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France
| | - Marjorie Varilh
- INSERM 1215, Neurocentre Magendie, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France
| | - Yifan Wu
- INSERM 1215, Neurocentre Magendie, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France
| | - Débora Jacky
- INSERM 1215, Neurocentre Magendie, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France
| | - Rim Fayad
- INSERM 1215, Neurocentre Magendie, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France
| | - Astrid Cannich
- INSERM 1215, Neurocentre Magendie, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France
| | - Luigi Bellocchio
- INSERM 1215, Neurocentre Magendie, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France
| | - Giovanni Marsicano
- INSERM 1215, Neurocentre Magendie, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France.
| | - Anna Beyeler
- INSERM 1215, Neurocentre Magendie, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France.
| |
Collapse
|
24
|
Dmitrieva NI, Boehm M, Yancey PH, Enhörning S. Long-term health outcomes associated with hydration status. Nat Rev Nephrol 2024; 20:275-294. [PMID: 38409366 DOI: 10.1038/s41581-024-00817-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2024] [Indexed: 02/28/2024]
Abstract
Body water balance is determined by fundamental homeostatic mechanisms that maintain stable volume, osmolality and the composition of extracellular and intracellular fluids. Water balance is maintained by multiple mechanisms that continuously match water losses through urine, the skin, the gastrointestinal tract and respiration with water gains achieved through drinking, eating and metabolic water production. Hydration status is determined by the state of the water balance. Underhydration occurs when a decrease in body water availability, due to high losses or low gains, stimulates adaptive responses within the water balance network that are aimed at decreasing losses and increasing gains. This stimulation is also accompanied by cardiovascular adjustments. Epidemiological and experimental studies have linked markers of low fluid intake and underhydration - such as increased plasma concentration of vasopressin and sodium, as well as elevated urine osmolality - with an increased risk of new-onset chronic diseases, accelerated aging and premature mortality, suggesting that persistent activation of adaptive responses may be detrimental to long-term health outcomes. The causative nature of these associations is currently being tested in interventional trials. Understanding of the physiological responses to underhydration may help to identify possible mechanisms that underlie potential adverse, long-term effects of underhydration and inform future research to develop preventative and treatment approaches to the optimization of hydration status.
Collapse
Affiliation(s)
- Natalia I Dmitrieva
- Laboratory of Cardiovascular Regenerative Medicine, National Heart Lung and Blood Institute, NIH, Bethesda, Maryland, USA.
| | - Manfred Boehm
- Laboratory of Cardiovascular Regenerative Medicine, National Heart Lung and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Paul H Yancey
- Biology Department, Whitman College, Walla Walla, Washington, USA
| | - Sofia Enhörning
- Perinatal and Cardiovascular Epidemiology, Lund University Diabetes Centre, Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
- Department of Internal Medicine, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
25
|
Pei S, Tao Q, Li W, Qi G, Wang B, Wang Y, Dai S, Shen Q, Wang X, Wu X, Xu S, Theprungsirikul L, Zhang J, Liang L, Liu Y, Chen K, Shen Y, Crawford BM, Cheng M, Zhang Q, Wang Y, Liu H, Yang B, Krichilsky B, Pei J, Song K, Johnson DM, Jiang Z, Wu F, Swift GB, Yang H, Liu Z, Zou X, Vo-Dinh T, Liu F, Pei ZM, Yuan F. Osmosensor-mediated control of Ca 2+ spiking in pollen germination. Nature 2024; 629:1118-1125. [PMID: 38778102 PMCID: PMC11136663 DOI: 10.1038/s41586-024-07445-6] [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: 08/08/2022] [Accepted: 04/19/2024] [Indexed: 05/25/2024]
Abstract
Higher plants survive terrestrial water deficiency and fluctuation by arresting cellular activities (dehydration) and resuscitating processes (rehydration). However, how plants monitor water availability during rehydration is unknown. Although increases in hypo-osmolarity-induced cytosolic Ca2+ concentration (HOSCA) have long been postulated to be the mechanism for sensing hypo-osmolarity in rehydration1,2, the molecular basis remains unknown. Because osmolarity triggers membrane tension and the osmosensing specificity of osmosensing channels can only be determined in vivo3-5, these channels have been classified as a subtype of mechanosensors. Here we identify bona fide cell surface hypo-osmosensors in Arabidopsis and find that pollen Ca2+ spiking is controlled directly by water through these hypo-osmosensors-that is, Ca2+ spiking is the second messenger for water status. We developed a functional expression screen in Escherichia coli for hypo-osmosensitive channels and identified OSCA2.1, a member of the hyperosmolarity-gated calcium-permeable channel (OSCA) family of proteins6. We screened single and high-order OSCA mutants, and observed that the osca2.1/osca2.2 double-knockout mutant was impaired in pollen germination and HOSCA. OSCA2.1 and OSCA2.2 function as hypo-osmosensitive Ca2+-permeable channels in planta and in HEK293 cells. Decreasing osmolarity of the medium enhanced pollen Ca2+ oscillations, which were mediated by OSCA2.1 and OSCA2.2 and required for germination. OSCA2.1 and OSCA2.2 convert extracellular water status into Ca2+ spiking in pollen and may serve as essential hypo-osmosensors for tracking rehydration in plants.
Collapse
Affiliation(s)
- Songyu Pei
- College of Horticulture and Landscape, Hunan Agricultural University, Changsha, China
- Department of Biology, Duke University, Durham, NC, USA
- Fitzpatrick Institute for Photonics, Duke University, Durham, NC, USA
- College of Life Sciences, Zhejiang University, Hangzhou, China
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Qi Tao
- College of Life Sciences, Zhejiang University, Hangzhou, China
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Wenke Li
- College of Life Sciences, Zhejiang University, Hangzhou, China
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Guoning Qi
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Borong Wang
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Yan Wang
- College of Horticulture and Landscape, Hunan Agricultural University, Changsha, China
| | - Shiwen Dai
- College of Horticulture and Landscape, Hunan Agricultural University, Changsha, China
| | - Qiujing Shen
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Xi Wang
- College of Life Sciences, Zhejiang University, Hangzhou, China
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Xiaomei Wu
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Shijian Xu
- Department of Biology, Duke University, Durham, NC, USA
| | | | | | - Liang Liang
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Yuantao Liu
- College of Life Sciences, Zhejiang University, Hangzhou, China
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Kena Chen
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Yang Shen
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | | | - Mengjia Cheng
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Qi Zhang
- College of Life Sciences, Zhejiang University, Hangzhou, China
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Yiqi Wang
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Hongli Liu
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Benguang Yang
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | | | - Jessica Pei
- Department of Biology, Duke University, Durham, NC, USA
- Fuqua School of Business, Duke University, Durham, NC, USA
| | - Karen Song
- Department of Biology, Duke University, Durham, NC, USA
| | | | | | - Feihua Wu
- Department of Biology, Duke University, Durham, NC, USA
| | - Gary B Swift
- Department of Physics, Duke University, Durham, NC, USA
| | - Huanghe Yang
- Department of Biochemistry, Duke University, Durham, NC, USA
| | - Zhonghua Liu
- College of Horticulture and Landscape, Hunan Agricultural University, Changsha, China
| | - Xuexiao Zou
- College of Horticulture and Landscape, Hunan Agricultural University, Changsha, China
| | - Tuan Vo-Dinh
- Fitzpatrick Institute for Photonics, Duke University, Durham, NC, USA
| | - Feng Liu
- College of Horticulture and Landscape, Hunan Agricultural University, Changsha, China.
- Department of Biology, Duke University, Durham, NC, USA.
| | - Zhen-Ming Pei
- Department of Biology, Duke University, Durham, NC, USA.
- Fitzpatrick Institute for Photonics, Duke University, Durham, NC, USA.
| | - Fang Yuan
- College of Horticulture and Landscape, Hunan Agricultural University, Changsha, China.
- Department of Biology, Duke University, Durham, NC, USA.
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China.
| |
Collapse
|
26
|
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
|
27
|
Robayo-Amortegui H, Quintero-Altare A, Florez-Navas C, Serna-Palacios I, Súarez-Saavedra A, Buitrago-Bernal R, Casallas-Barrera JO. Fluid dynamics of life: exploring the physiology and importance of water in the critical illness. Front Med (Lausanne) 2024; 11:1368502. [PMID: 38745736 PMCID: PMC11092983 DOI: 10.3389/fmed.2024.1368502] [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: 01/12/2024] [Accepted: 04/12/2024] [Indexed: 05/16/2024] Open
Abstract
Water acknowledged as a vital component for life and the universal solvent, is crucial for diverse physiological processes in the human body. While essential for survival, the human body lacks the capacity to produce water, emphasizing the need for regular ingestion to maintain a homeostatic environment. The human body, predominantly composed of water, exhibits remarkable biochemical properties, playing a pivotal role in processes such as protein transport, thermoregulation, the cell cycle, and acid–base balance. This review delves into comprehending the molecular characteristics of water and its interactions within the human body. The article offers valuable insights into the intricate relationship between water and critical illness. Through a comprehensive exploration, it seeks to enhance our understanding of water’s pivotal role in sustaining overall human health.
Collapse
Affiliation(s)
- Henry Robayo-Amortegui
- Department of Critical Care Medicine, Fundación Clínica Shaio, Bogotá, DC, Colombia
- Department of Medicine, Critical Care Resident, Universidad de La Sabana, Chía Cundinamarca, Colombia
| | - Alejandro Quintero-Altare
- Department of Critical Care Medicine, Fundación Clínica Shaio, Bogotá, DC, Colombia
- Department of Medicine, Critical Care Resident, Universidad de La Sabana, Chía Cundinamarca, Colombia
| | - Catalina Florez-Navas
- Department of Critical Care Medicine, Fundación Clínica Shaio, Bogotá, DC, Colombia
- Department of Medicine, Critical Care Resident, Universidad de La Sabana, Chía Cundinamarca, Colombia
| | - Isacio Serna-Palacios
- Department of Medicine, Critical Care Resident, Universidad de La Sabana, Chía Cundinamarca, Colombia
| | | | - Ricardo Buitrago-Bernal
- Department of Critical Care Medicine, Fundación Clínica Shaio, Bogotá, DC, Colombia
- Exploratorium group, Fundación Clínica Shaio, Bogotá, DC, Colombia
| | | |
Collapse
|
28
|
Pudelko L, Cabianca DS. The influencers' era: how the environment shapes chromatin in 3D. Curr Opin Genet Dev 2024; 85:102173. [PMID: 38417271 DOI: 10.1016/j.gde.2024.102173] [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: 09/08/2023] [Revised: 01/26/2024] [Accepted: 02/06/2024] [Indexed: 03/01/2024]
Abstract
Environment-epigenome interactions are emerging as contributors to disease risk and health outcomes. In fact, organisms outside of the laboratory are constantly exposed to environmental changes that can influence chromatin regulation at multiple levels, potentially impacting on genome function. In this review, we will summarize recent findings on how major external cues impact on 3D chromatin organization in different experimental systems. We will describe environment-induced 3D genome alterations ranging from chromatin accessibility to the spatial distribution of the genome and discuss their role in regulating gene expression.
Collapse
Affiliation(s)
- Lorenz Pudelko
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, Germany; Faculty of Medicine, Ludwig-Maximilians Universität München, Munich, Germany. https://twitter.com/@lorenz_pudelko
| | - Daphne S Cabianca
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, Germany.
| |
Collapse
|
29
|
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
|
30
|
Schill F, Engström G, Melander O, Timpka S, Enhörning S. The possible role of the vasopressin system in hematopoiesis. Sci Rep 2024; 14:5085. [PMID: 38429469 PMCID: PMC10907562 DOI: 10.1038/s41598-024-55772-5] [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: 11/23/2023] [Accepted: 02/27/2024] [Indexed: 03/03/2024] Open
Abstract
Vasopressin is a pleiotropic hormone that controls body fluid homeostasis. Vasopressin has also been proposed to be involved in erythropoiesis, thrombocyte activity and inflammation. However, whether increasing vasopressin is associated with changes in hematopoietic markers is not known. To evaluate this gap of knowledge we measured the vasopressin marker copeptin and markers of erythropoiesis (erythrocyte count, hemoglobin (Hb), red blood cell distribution width (RDW), mean corpuscular volume (MCV), erythrocyte volume fraction (EVF)), leukocyte count (total count, lymphocytes, neutrophils) and thrombocyte count in 5312 participants from the Swedish CArdioPulmonary bioImage Study (SCAPIS). The associations between increasing copeptin tertile and the hematopoietic markers were analyzed in multivariate linear regression analyses. We found that increasing copeptin tertile was significantly (p < 0.001) associated with increasing erythrocytes, RDW, EVF, Hb, leukocytes and neutrophils after adjustment for age, sex, current smoking, prevalent diabetes, hypertension, creatinine, body mass index and physical activity. Increasing copeptin tertile was, however, not associated with change in MCV, lymphocyte or thrombocyte count. In conclusion, we found that increasing copeptin levels are positively associated with markers of erythropoiesis and leukocyte count in the general population. These results warrant further research on possible mechanistic effects of vasopressin on hematopoiesis.
Collapse
Affiliation(s)
- Fredrika Schill
- Department of Cardiology, Skåne University Hospital, Carl-Bertil Laurells Gata 9, 214 28, Malmö, Sweden.
- Department of Clinical Sciences, Lund University, Jan Waldenströms Gata 35, 214 28, Malmö, Sweden.
| | - Gunnar Engström
- Department of Clinical Sciences, Lund University, Jan Waldenströms Gata 35, 214 28, Malmö, Sweden
| | - Olle Melander
- Department of Clinical Sciences, Lund University, Jan Waldenströms Gata 35, 214 28, Malmö, Sweden
- Department of Internal Medicine, Skåne University Hospital, Jan Waldenströms Gata 11 A, 214 28, Malmö, Sweden
| | - Simon Timpka
- Perinatal and Cardiovascular Epidemiology, Department of Clinical Sciences Malmö, Lund University, Jan Waldenströms gata 35, 214 28, Malmö, Sweden
- Department of Obstetrics and Gynecology, Skåne University Hospital, Jan Waldenströms gata 47, 214 28, Malmö, Sweden
| | - Sofia Enhörning
- Department of Clinical Sciences, Lund University, Jan Waldenströms Gata 35, 214 28, Malmö, Sweden
- Department of Internal Medicine, Skåne University Hospital, Jan Waldenströms Gata 11 A, 214 28, Malmö, Sweden
| |
Collapse
|
31
|
Junkins MS, Feng NY, Murphy LA, Curtis G, Merriman DK, Bagriantsev SN, Gracheva EO. Neural control of fluid homeostasis is engaged below 10°C in hibernation. Curr Biol 2024; 34:923-930.e5. [PMID: 38325375 PMCID: PMC11232715 DOI: 10.1016/j.cub.2024.01.035] [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: 05/14/2023] [Revised: 11/29/2023] [Accepted: 01/12/2024] [Indexed: 02/09/2024]
Abstract
Thirteen-lined ground squirrels (Ictidomys tridecemlineatus) hibernate for several months each winter without access to water,1 but the mechanisms that maintain fluid homeostasis during hibernation are poorly understood. In torpor, when body temperature (TB) reaches 4°C, squirrels decrease metabolism, slow heart rate, and reduce plasma levels of the antidiuretic hormones arginine vasopressin (AVP) and oxytocin (OXT).1 Squirrels spontaneously undergo interbout arousal (IBA) every 2 weeks, temporarily recovering an active-like metabolism and a TB of 37°C for up to 48 h.1,2 Despite the low levels of AVP and OXT during torpor, profound increases in blood pressure and heart rate during the torpor-IBA transition are not associated with massive fluid loss, suggesting the existence of a mechanism that protects against diuresis at a low TB. Here, we demonstrate that the antidiuretic hormone release pathway is activated by hypothalamic supraoptic nucleus (SON) neurons early in the torpor-arousal transition. SON neuron activity, dense-core vesicle release from the posterior pituitary, and plasma hormone levels all begin to increase before TB reaches 10°C. In vivo fiber photometry of SON neurons from hibernating squirrels, together with RNA sequencing and c-FOS immunohistochemistry, confirms that SON is electrically, transcriptionally, and translationally active to monitor blood osmolality throughout the dynamic torpor-arousal transition. Our work emphasizes the importance of the antidiuretic pathway during the torpor-arousal transition and reveals that the neurophysiological mechanism that coordinates the hormonal response to retain fluid is active at an extremely low TB, which is prohibitive for these processes in non-hibernators.
Collapse
Affiliation(s)
- Madeleine S Junkins
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA; Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA; Department of Neuroscience and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| | - Ni Y Feng
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA; Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA; Department of Neuroscience and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA; Neuroscience & Behavior Program, Wesleyan University, 52 Lawn Ave, Middletown, CT 06459, USA.
| | - Lyle A Murphy
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| | - Genevieve Curtis
- Department of Biology, Wesleyan University, 52 Lawn Ave, Middletown, CT 06459, USA
| | - Dana K Merriman
- Department of Biology, University of Wisconsin-Oshkosh, 800 Algoma Blvd, Oshkosh, WI 54901, USA
| | - Sviatoslav N Bagriantsev
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA.
| | - Elena O Gracheva
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA; Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA; Department of Neuroscience and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA; Kavli Institute for Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA.
| |
Collapse
|
32
|
Zuloaga R, Ahumada-Langer L, Aedo JE, Molina A, Valdés JA. 11-Deoxycorticosterone (DOC)'s Action on the Gill Osmoregulation of Juvenile Rainbow Trout ( Oncorhynchus mykiss). BIOLOGY 2024; 13:107. [PMID: 38392325 PMCID: PMC10886319 DOI: 10.3390/biology13020107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/24/2024]
Abstract
In aquaculture, stress can negatively affect fish growth. For years, the cortisol hormone has been thought to play both glucocorticoid and mineralocorticoid functions. Nevertheless, recent research has suggested that 11-deoxycorticosterone (DOC) released during stress could contribute to cortisol actions, though this process is still misunderstood. Here, we evaluated the DOC effects on physiological and early transcriptional responses by RNA-seq. Juvenile rainbow trout were treated with DOC and/or glucocorticoids (mifepristone) or mineralocorticoid (eplerenone) receptor antagonists. Subsequently, plasma was collected, and cDNA libraries were generated from the gills of vehicle (control), DOC, mifepristone, mifepristone with DOC, eplerenone, and eplerenone with DOC groups. Calcium and phosphate levels in plasma were changed. Results revealed 914 differentially expressed transcripts (DETs) induced by DOC compared with control, mainly associated with sodium ion transmembrane transport, gluconeogenesis, negative regulation of transmembrane transport, and activation of innate immune response. DOC versus eplerenone with DOC comparison displayed 444 DETs related to cell-cell junction organization, canonical glycolysis, positive regulation of immune response, and potassium ion transport. Conversely, no DETs were detected in DOC versus mifepristone with DOC comparison. These data suggest that DOC has a relevant role in gill stress response and ion transport, which is differentially regulated by mineralocorticoid receptors.
Collapse
Affiliation(s)
- Rodrigo Zuloaga
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago 8370146, Chile
- Interdisciplinary Center for Aquaculture Research (INCAR), Concepción 4030000, Chile
| | - Luciano Ahumada-Langer
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago 8370146, Chile
| | - Jorge Eduardo Aedo
- Departamento de Biología y Química, Facultad de Ciencias Básicas, Universidad Católica del Maule, Talca 3466706, Chile
| | - Alfredo Molina
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago 8370146, Chile
- Interdisciplinary Center for Aquaculture Research (INCAR), Concepción 4030000, Chile
| | - Juan Antonio Valdés
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago 8370146, Chile
- Interdisciplinary Center for Aquaculture Research (INCAR), Concepción 4030000, Chile
| |
Collapse
|
33
|
Duan C, Anderson JL, Schepers LE, Damen FW, Cox A, Goergen CJ, Sivasankar PM. In Vivo Visualization and Quantification of Rat Laryngeal Blood Supply After Hydration Challenge. Laryngoscope 2024; 134:779-785. [PMID: 37584333 PMCID: PMC10842383 DOI: 10.1002/lary.30965] [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: 02/15/2023] [Revised: 05/31/2023] [Accepted: 08/01/2023] [Indexed: 08/17/2023]
Abstract
OBJECTIVES Systemic dehydration decreases total body blood volume; however, hemodynamic alterations at the level of local organs, such as the larynx, remain unclear. Here we sought to quantify superior thyroid artery (STA) blood flow after dehydration and rehydration using in vivo magnetic resonance angiography (MRA) and ultrasound imaging in a rat model. METHODS Male Sprague-Dawley rats (N = 17) were included in this prospective, repeated measures design. Rats first underwent MRA to determine baseline STA cross-sectional area, followed by high-frequency in vivo ultrasound imaging to measure STA blood velocity at baseline. Next, rats were systemically dehydrated (water withholding), followed by rehydration (water ad-lib). Ultrasound imaging was repeated immediately after dehydration and following rehydration. The STA blood velocity and STA cross-sectional area were used to compute STA blood flow. Three rats served as temporal controls for ultrasound imaging. To determine if the challenges to hydration status affected the STA cross-sectional area, four rats underwent only MRA at baseline, dehydration, and rehydration. RESULTS Systemic dehydration resulted in 10.5% average body weight loss. Rehydration resulted in average body weight gain of 10.9%. Statistically significant reductions were observed in STA mean blood flow rate after dehydration. Rehydration reversed these changes to pre-dehydration levels. No significant differences were observed in STA cross-sectional area with dehydration or rehydration. CONCLUSION Systemic dehydration decreased blood flow in the superior thyroid artery. Rehydration restored blood flow in the STA. Change in hydration status did not alter the STA cross-sectional area. These preliminary findings demonstrate the feasibility of using ultrasound and MRA to quantify hemodynamic changes and visualize laryngeal blood vessels. LEVEL OF EVIDENCE NA Laryngoscope, 134:779-785, 2024.
Collapse
Affiliation(s)
- Chenwei Duan
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN
- Department of Speech, Language, and Hearing Sciences, Purdue University, West Lafayette, IN
| | | | - Luke E. Schepers
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN
| | - Frederick W. Damen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN
- Indiana University School of Medicine, Indianapolis, IN
| | - Abigail Cox
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN
| | - Craig J. Goergen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN
- Indiana University School of Medicine, Indianapolis, IN
| | - Preeti M. Sivasankar
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN
- Department of Speech, Language, and Hearing Sciences, Purdue University, West Lafayette, IN
| |
Collapse
|
34
|
Yoshikawa K, Saito S, Kadonosono T, Tanaka M, Okochi M. Osmotic stress induces the formation of migrasome-like vesicles. FEBS Lett 2024; 598:437-445. [PMID: 38339800 DOI: 10.1002/1873-3468.14816] [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: 12/05/2023] [Revised: 01/10/2024] [Accepted: 01/10/2024] [Indexed: 02/12/2024]
Abstract
Migrasomes are extracellular vesicles that form on the retraction fibers of migrating cells. In this study, we report the formation of migrasome-like vesicles enriched in tetraspanin 4 and containing cytoplasmic components in response to hypoosmotic stress. When migrating cells were subjected to hypoosmotic stress, vesicles with a size distribution of 0.5 to 2 μm formed on the retraction fibers, and vanished in a few minutes. The vesicles are rich in cholesterol, and their number was reduced when cells were pretreated with lipoprotein-deficient serum. The formation of migrasome-like vesicles upon hypoosmotic stress may provide biophysical cues regarding the cellular response to this external stimulus in cells and tissues.
Collapse
Grants
- 23K17843 Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science, and Technology of Japan
- 22K19913 Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science, and Technology of Japan
- 21H01726 Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science, and Technology of Japan
- 21H01725 Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science, and Technology of Japan
- JP21zf0127004 Moonshot Research and Development Program from the Japan Agency for Medical Research and Development (AMED)
Collapse
Affiliation(s)
- Koki Yoshikawa
- Department of Chemical Science and Engineering, Tokyo Institute of Technology, Japan
| | - Shogo Saito
- Department of Chemical Science and Engineering, Tokyo Institute of Technology, Japan
| | - Tetsuya Kadonosono
- Department of Life Science and Technology, Tokyo Institute of Technology, Yokohama-shi, Japan
| | - Masayoshi Tanaka
- Department of Chemical Science and Engineering, Tokyo Institute of Technology, Yokohama-shi, Japan
| | - Mina Okochi
- Department of Chemical Science and Engineering, Tokyo Institute of Technology, Japan
| |
Collapse
|
35
|
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
|
36
|
Gong J, Li Q. Comparative Transcriptome and WGCNA Analysis Reveal Molecular Responses to Salinity Change in Larvae of the Iwagaki Oyster Crassostrea Nippona. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2023; 25:1031-1042. [PMID: 37872465 DOI: 10.1007/s10126-023-10257-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 10/09/2023] [Indexed: 10/25/2023]
Abstract
The Iwagaki oyster Crassostrea nippona is an important aquaculture species with significant potential for large-scale oyster farming. It is susceptible to the fluctuated salinity in the coastal area. In this study, we compared the transcriptome of Crassostrea nippona larvae under variant conditions with low-salinity stress (28, 20, 15, 10, and 5 practical salinity units (psu)) for 24 h. KEGG enrichment analysis of differentially expressed genes (DEGs) from pairwise comparisons identified several free amino acid metabolism pathway (taurine and hypotaurine, arginine and proline, glycine, and beta-alanine) contributing to the salinity change adaptation and activated "lysosome" and "apoptosis" pathway in response to the low-salinity stress (10 and 5 psu). Trend analysis revealed sustained upregulation of transmembrane transport-related genes (such as SLC family) and downregulation of ribosomal protein synthesis genes faced with decreasing salinities. In addition, 9 biomarkers in response to low-salinity stress were identified through weighted gene co-expression network analysis (WGCNA) and validated by qRT-PCR. Our transcriptome analysis provides a comprehensive view of the molecular mechanisms and regulatory networks underlying the adaptive responses of oyster larvae to hypo-salinity conditions. These findings contribute to our understanding of the complex biological processes involved in oyster resilience and adaptation to changing environmental conditions.
Collapse
Affiliation(s)
- Jianwen Gong
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Qi Li
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China.
| |
Collapse
|
37
|
Notley SR, Mitchell D, Taylor NAS. A century of exercise physiology: concepts that ignited the study of human thermoregulation. Part 1: Foundational principles and theories of regulation. Eur J Appl Physiol 2023; 123:2379-2459. [PMID: 37702789 DOI: 10.1007/s00421-023-05272-7] [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: 12/30/2022] [Accepted: 06/30/2023] [Indexed: 09/14/2023]
Abstract
This contribution is the first of a four-part, historical series encompassing foundational principles, mechanistic hypotheses and supported facts concerning human thermoregulation during athletic and occupational pursuits, as understood 100 years ago and now. Herein, the emphasis is upon the physical and physiological principles underlying thermoregulation, the goal of which is thermal homeostasis (homeothermy). As one of many homeostatic processes affected by exercise, thermoregulation shares, and competes for, physiological resources. The impact of that sharing is revealed through the physiological measurements that we take (Part 2), in the physiological responses to the thermal stresses to which we are exposed (Part 3) and in the adaptations that increase our tolerance to those stresses (Part 4). Exercising muscles impose our most-powerful heat stress, and the physiological avenues for redistributing heat, and for balancing heat exchange with the environment, must adhere to the laws of physics. The first principles of internal and external heat exchange were established before 1900, yet their full significance is not always recognised. Those physiological processes are governed by a thermoregulatory centre, which employs feedback and feedforward control, and which functions as far more than a thermostat with a set-point, as once was thought. The hypothalamus, today established firmly as the neural seat of thermoregulation, does not regulate deep-body temperature alone, but an integrated temperature to which thermoreceptors from all over the body contribute, including the skin and probably the muscles. No work factor needs to be invoked to explain how body temperature is stabilised during exercise.
Collapse
Affiliation(s)
- Sean R Notley
- Defence Science and Technology Group, Department of Defence, Melbourne, Australia
- School of Human Kinetics, University of Ottawa, Ottawa, Canada
| | - Duncan Mitchell
- Brain Function Research Group, School of Physiology, University of the Witwatersrand, Johannesburg, South Africa
- School of Human Sciences, University of Western Australia, Crawley, Australia
| | - Nigel A S Taylor
- Research Institute of Human Ecology, College of Human Ecology, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
38
|
Williams JT, Baker JJ, Zheng H, Dechow SJ, Fallon J, Murto M, Albrecht VJ, Gilliland HN, Olive AJ, Abramovitch RB. A genetic selection for Mycobacterium smegmatis mutants tolerant to killing by sodium citrate defines a combined role for cation homeostasis and osmotic stress in cell death. mSphere 2023; 8:e0035823. [PMID: 37681985 PMCID: PMC10597346 DOI: 10.1128/msphere.00358-23] [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: 07/19/2023] [Accepted: 07/23/2023] [Indexed: 09/09/2023] Open
Abstract
Mycobacteria can colonize environments where the availability of metal ions is limited. Biological or inorganic chelators play an important role in limiting metal availability, and we developed a model to examine Mycobacterium smegmatis survival in the presence of the chelator sodium citrate. We observed that instead of restricting M. smegmatis growth, concentrated sodium citrate killed M. smegmatis. RNAseq analysis during sodium citrate treatment revealed transcriptional signatures of metal starvation and hyperosmotic stress. Notably, metal starvation and hyperosmotic stress, individually, do not kill M. smegmatis under these conditions. A forward genetic transposon selection was conducted to examine why sodium citrate was lethal, and several sodium-citrate-tolerant mutants were isolated. Based on the identity of three tolerant mutants, mgtE, treZ, and fadD6, we propose a dual stress model of killing by sodium citrate, where sodium citrate chelate metals from the cell envelope and then osmotic stress in combination with a weakened cell envelope causes cell lysis. This sodium citrate tolerance screen identified mutants in several other genes with no known function, with most conserved in the pathogen M. tuberculosis. Therefore, this model will serve as a basis to define their functions, potentially in maintaining cell wall integrity, cation homeostasis, or osmotolerance. IMPORTANCE Bacteria require mechanisms to adapt to environments with differing metal availability. When Mycobacterium smegmatis is treated with high concentrations of the metal chelator sodium citrate, the bacteria are killed. To define the mechanisms underlying killing by sodium citrate, we conducted a genetic selection and observed tolerance to killing in mutants of the mgtE magnesium transporter. Further characterization studies support a model where killing by sodium citrate is driven by a weakened cell wall and osmotic stress, that in combination cause cell lysis.
Collapse
Affiliation(s)
- John T. Williams
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - Jacob J. Baker
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - Huiqing Zheng
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - Shelby J. Dechow
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - Jared Fallon
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - Megan Murto
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - Veronica J. Albrecht
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - Haleigh N. Gilliland
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - Andrew J. Olive
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - Robert B. Abramovitch
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
39
|
Nguyen DH, Duque V, Phillips N, Mecawi AS, Cunningham JT. Spatial transcriptomics reveal basal sex differences in supraoptic nucleus gene expression of adult rats related to cell signaling and ribosomal pathways. Biol Sex Differ 2023; 14:71. [PMID: 37858270 PMCID: PMC10585758 DOI: 10.1186/s13293-023-00554-3] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 09/25/2023] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND The supraoptic nucleus (SON) of the hypothalamus contains magnocellular neurosecretory cells that secrete the hormones vasopressin and oxytocin. Sex differences in SON gene expression have been relatively unexplored. Our study used spatially resolved transcriptomics to visualize gene expression profiles in the SON of adult male (n = 4) and female (n = 4) Sprague-Dawley rats using Visium Spatial Gene Expression (10x Genomics). METHODS Briefly, 10-μm coronal sections (~ 4 × 4 mm) containing the SON were collected from each rat and processed using Visium slides and recommended protocols. Data were analyzed using 10x Genomics' Space Ranger and Loupe Browser applications and other bioinformatic tools. Two unique differential expression (DE) analysis methods, Loupe Browser and DESeq2, were used. RESULTS Loupe Browser DE analysis of the SON identified 116 significant differentially expressed genes (DEGs) common to both sexes (e.g., Avp and Oxt), 31 significant DEGs unique to the males, and 73 significant DEGs unique to the females. DESeq2 analysis revealed 183 significant DEGs between the two groups. Gene Ontology (GO) enrichment and pathway analyses using significant genes identified via Loupe Browser revealed GO terms and pathways related to (1) neurohypophyseal hormone activity, regulation of peptide hormone secretion, and regulation of ion transport for the significant genes common to both males and females, (2) Gi signaling/G-protein mediated events for the significant genes unique to males, and (3) potassium ion transport/voltage-gated potassium channels for the significant genes unique to females, as some examples. GO/pathway analyses using significant genes identified via DESeq2 comparing female vs. male groups revealed GO terms/pathways related to ribosomal structure/function. Ingenuity Pathway Analysis (IPA) identified additional sex differences in canonical pathways (e.g., 'Mitochondrial Dysfunction', 'Oxidative Phosphorylation') and upstream regulators (e.g., CSF3, NFKB complex, TNF, GRIN3A). CONCLUSION There was little overlap in the IPA results for the two different DE methods. These results suggest sex differences in SON gene expression that are associated with cell signaling and ribosomal pathways.
Collapse
Affiliation(s)
- Dianna H Nguyen
- Department of Physiology and Anatomy, School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX, USA
- Texas College of Osteopathic Medicine, UNT Health Science Center, Fort Worth, TX, USA
| | - Victor Duque
- Department of Biophysics, Laboratory of Molecular Neuroendocrinology, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Nicole Phillips
- Department of Microbiology, Immunology, and Genetics, School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX, USA
| | - André Souza Mecawi
- Department of Biophysics, Laboratory of Molecular Neuroendocrinology, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - J Thomas Cunningham
- Department of Physiology and Anatomy, School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX, USA.
| |
Collapse
|
40
|
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
|
41
|
Nair-Collins M, Joffe AR. Frequent Preservation of Neurologic Function in Brain Death and Brainstem Death Entails False-Positive Misdiagnosis and Cerebral Perfusion. AJOB Neurosci 2023; 14:255-268. [PMID: 34586014 DOI: 10.1080/21507740.2021.1973148] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Some patients who have been diagnosed as "dead by neurologic criteria" continue to exhibit certain brain functions, most commonly, neuroendocrine functions. This preservation of neurologic function after the diagnosis of "brain death" or "brainstem death" is an ongoing source of controversy and concern in the medical, bioethics, and legal literatures. Most obviously, if some brain function persists, then it is not the case that all functions of the entire brain have ceased and hence, declaring such a patient to be "dead" would be a false positive, in any nation with so-called "whole brain death" laws. Furthermore, and perhaps more concerning, the preservation of any brain function necessarily entails the preservation of some amount of brain perfusion, thereby raising the concern as to whether additional areas of neural tissue may remain viable, including areas in the brainstem. These and other considerations cast significant doubt on the reliability of diagnosing either "brain death" or "brainstem death."
Collapse
Affiliation(s)
| | - Ari R Joffe
- University of Alberta and Stollery Children's Hospital
| |
Collapse
|
42
|
Woodson J, Bergan JF. Uncovering the brain-wide pattern of synaptic input to vasopressin-expressing neurons in the paraventricular nucleus of the hypothalamus. J Comp Neurol 2023; 531:1017-1031. [PMID: 37121600 PMCID: PMC10566340 DOI: 10.1002/cne.25476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 05/02/2023]
Abstract
Arginine vasopressin (AVP) is a neuropeptide critical for the mammalian stress response and social behavior. AVP produced in the hypothalamus regulates water osmolality and vasoconstriction in the body, and in the brain, it regulates social behavior, aggression, and anxiety. However, the circuit mechanisms that link AVP to social behavior, homeostatic function, and disease are not well understood. This study investigates the circuit configurations of AVP-expressing neurons in the rodent hypothalamus and characterizes synaptic input from the entire brain. We targeted the paraventricular nucleus (PVN) using retrograde viral tracing techniques to identify direct afferent synaptic connections made onto AVP-expressing neurons. AVP neurons in the PVN display region-specific anatomical configurations that reflect their unique contributions to homeostatic function, motor behaviors, feeding, and affiliative behavior. The afferent connections identified were similar in both sexes and subsequent molecular investigation of these inputs shows that those local hypothalamic inputs are overwhelmingly nonpeptidergic cells indicating a potential interneuron nexus between hormone cell activation and broader cortical connection. This proposed work reveals new insights into the organization of social behavior circuits in the brain, and how neuropeptides act centrally to modulate social behaviors.
Collapse
Affiliation(s)
- Jonathan Woodson
- Neuroscience and Behavior Program, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | - Joseph F Bergan
- Neuroscience and Behavior Program, University of Massachusetts Amherst, Amherst, Massachusetts, USA
- Department of Psychological and Brain Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| |
Collapse
|
43
|
Aikins AO, Farmer GE, Little JT, Cunningham JT. Effects of bile duct ligation on the inhibitory control of supraoptic vasopressin neurons. J Neuroendocrinol 2023; 35:e13312. [PMID: 37337093 PMCID: PMC10942741 DOI: 10.1111/jne.13312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/02/2023] [Accepted: 05/11/2023] [Indexed: 06/21/2023]
Abstract
Dilutional hyponatremia due to increased plasma arginine vasopressin (AVP) is associated with liver cirrhosis. However, plasma AVP remains elevated despite progressive hypoosmolality. This study investigated changes to inhibitory control of supraoptic nucleus (SON) AVP neurons during liver cirrhosis. Experiments were conducted with adult male Sprague-Dawley rats. Bile duct ligation was used as a model of chronic liver cirrhosis. An adeno-associated virus containing a construct with an AVP promoter and either green fluorescent protein (GFP) or a ratiometric chloride indicator, ClopHensorN, was bilaterally injected into the SON of rats. After 2 weeks, rats received either BDL or sham surgery, and liver cirrhosis was allowed to develop for 4 weeks. In vitro, loose patch recordings of action potentials were obtained from GFP-labeled and unlabeled SON neurons in response to a brief focal application of the GABAA agonist muscimol (100 μM). Changes to intracellular chloride ([Cl]i) following muscimol application were determined by changes to the fluorescence ratio of ClopHensorN. The contribution of cation chloride cotransporters NKCC1 and KCC2 to changes in intracellular chloride was investigated using their respective antagonists, bumetanide (BU, 10 μM) and VU0240551 (10 μM). Plasma osmolality and hematocrit were measured as a marker of dilutional hyponatremia. The results showed reduced or absent GABAA -mediated inhibition in a greater proportion of AVP neurons from BDL rats as compared to sham rats (100% inhibition in sham vs. 47% in BDL, p = .001). Muscimol application was associated with increased [Cl]i in most cells from BDL as compared to cells from sham rats (χ2 = 30.24, p < .001). NKCC1 contributed to the impaired inhibition observed in BDL rats (p < .001 BDL - BU vs. BDL + BU). The results show that impaired inhibition of SON AVP neurons and increased intracellular chloride contribute to the sustained dilutional hyponatremia in liver cirrhosis.
Collapse
Affiliation(s)
- Ato O Aikins
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - George E Farmer
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Joel T Little
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - J Thomas Cunningham
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, USA
| |
Collapse
|
44
|
Elsamad G, Mecawi AS, Pauža AG, Gillard B, Paterson A, Duque VJ, Šarenac O, Žigon NJ, Greenwood M, Greenwood MP, Murphy D. Ageing restructures the transcriptome of the hypothalamic supraoptic nucleus and alters the response to dehydration. NPJ AGING 2023; 9:12. [PMID: 37264028 PMCID: PMC10234251 DOI: 10.1038/s41514-023-00108-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 05/04/2023] [Indexed: 06/03/2023]
Abstract
Ageing is associated with altered neuroendocrine function. In the context of the hypothalamic supraoptic nucleus, which makes the antidiuretic hormone vasopressin, ageing alters acute responses to hyperosmotic cues, rendering the elderly more susceptible to dehydration. Chronically, vasopressin has been associated with numerous diseases of old age, including type 2 diabetes and metabolic syndrome. Bulk RNAseq transcriptome analysis has been used to catalogue the polyadenylated supraoptic nucleus transcriptomes of adult (3 months) and aged (18 months) rats in basal euhydrated and stimulated dehydrated conditions. Gene ontology and Weighted Correlation Network Analysis revealed that ageing is associated with alterations in the expression of extracellular matrix genes. Interestingly, whilst the transcriptomic response to dehydration is overall blunted in aged animals compared to adults, there is a specific enrichment of differentially expressed genes related to neurodegenerative processes in the aged cohort, suggesting that dehydration itself may provoke degenerative consequences in aged rats.
Collapse
Affiliation(s)
- Ghadir Elsamad
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, Dorothy Hodgkin Building, University of Bristol, Bristol, England
| | - André Souza Mecawi
- Laboratory of Molecular Neuroendocrinology, Department of Biophysics, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Audrys G Pauža
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, Dorothy Hodgkin Building, University of Bristol, Bristol, England
- Translational Cardio-Respiratory Research Group, Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Benjamin Gillard
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, Dorothy Hodgkin Building, University of Bristol, Bristol, England
| | - Alex Paterson
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, Dorothy Hodgkin Building, University of Bristol, Bristol, England
- Insilico Consulting Ltd., Wapping Wharf, Bristol, England
| | - Victor J Duque
- Laboratory of Molecular Neuroendocrinology, Department of Biophysics, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Olivera Šarenac
- Institute of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Department of Safety Pharmacology, Abbvie, North Chicago, Illinois, USA
| | - Nina Japundžić Žigon
- Institute of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Mingkwan Greenwood
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, Dorothy Hodgkin Building, University of Bristol, Bristol, England
| | - Michael P Greenwood
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, Dorothy Hodgkin Building, University of Bristol, Bristol, England
| | - David Murphy
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, Dorothy Hodgkin Building, University of Bristol, Bristol, England.
| |
Collapse
|
45
|
Zhang W, Bertinetti L, Yavuzsoy EC, Gao C, Schneck E, Fratzl P. Submicron-Sized In Situ Osmotic Pressure Sensors for In Vitro Applications in Biology. Adv Healthc Mater 2023; 12:e2202373. [PMID: 36541931 PMCID: PMC11468766 DOI: 10.1002/adhm.202202373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 12/06/2022] [Indexed: 12/24/2022]
Abstract
Physical forces are important cues in determining the development and the normal function of biological tissues. While forces generated by molecular motors have been widely studied, forces resulting from osmotic gradients have been less considered in this context. A possible reason is the lack of direct in situ measurement methods that can be applied to cell and organ culture systems. Herein, novel kinds of resonance energy transfer (FRET)-based liposomal sensors are developed, so that their sensing range and sensitivity can be adjusted to satisfy physiological osmotic conditions. Several types of sensors are prepared, either based on polyethylene glycol- (PEG)ylated liposomes with steric stabilization and stealth property or on crosslinked liposomes capable of enduring relatively harsh environments for liposomes (e.g., in the presence of biosurfactants). The sensors are demonstrated to be effective in the measurement of osmotic pressures in pre-osteoblastic in vitro cell culture systems by means of FRET microscopy. This development paves the way toward the in situ sensing of osmotic pressures in biological culture systems.
Collapse
Affiliation(s)
- Wenbo Zhang
- Department of BiomaterialsMax Planck Institute of Colloids and Interfaces14476PotsdamGermany
| | - Luca Bertinetti
- Department of BiomaterialsMax Planck Institute of Colloids and Interfaces14476PotsdamGermany
- B CUBE Center for Molecular BioengineeringTechnical University of Darmstadt01307DresdenGermany
| | - Efe Cuma Yavuzsoy
- Department of BiomaterialsMax Planck Institute of Colloids and Interfaces14476PotsdamGermany
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang UniversityHangzhou310027China
| | - Emanuel Schneck
- Department of BiomaterialsMax Planck Institute of Colloids and Interfaces14476PotsdamGermany
- Department of PhysicsTechnical University of Darmstadt64289DarmstadtGermany
| | - Peter Fratzl
- Department of BiomaterialsMax Planck Institute of Colloids and Interfaces14476PotsdamGermany
| |
Collapse
|
46
|
Stocker SD. Altered Neuronal Discharge in the Organum Vasculosum of the Lamina Terminalis Contributes to Dahl Salt-Sensitive Hypertension. Hypertension 2023; 80:872-881. [PMID: 36752103 PMCID: PMC10023399 DOI: 10.1161/hypertensionaha.122.20798] [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: 12/13/2022] [Accepted: 01/30/2023] [Indexed: 02/09/2023]
Abstract
BACKGROUND Salt-sensitive hypertension in humans and experimental models is associated with higher plasma and cerebrospinal fluid sodium chloride (NaCl) concentrations. Changes in extracellular NaCl concentrations are sensed by specialized neurons in the organum vasculosum of the lamina terminalis (OVLT). Stimulation of OVLT neurons increases sympathetic nerve activity (SNA) and arterial blood pressure (ABP), whereas chronic activation produces hypertension. Therefore, the present study tested whether OVLT neuronal activity was elevated and contributed to SNA and ABP in salt-sensitive hypertension. METHODS Male Dahl salt-sensitive (Dahl S) and Dahl salt-resistant (Dahl R) rats were fed 0.1% or 4.0% NaCl diets for 3 to 4 weeks and used for single-unit recordings of OVLT neurons or simultaneous recording of multiple sympathetic nerves during pharmacological inhibition of the OVLT. RESULTS Plasma and cerebrospinal fluid Na+ and Cl- concentrations were higher in Dahl S rats fed 4% versus 0.1% or Dahl R rats fed either diet. In vivo single-unit recordings revealed a significantly higher discharge of NaCl-responsive OVLT neurons in Dahl S rats fed 4% versus 0.1% or Dahl R rats. Interestingly, intracarotid infusion of hypertonic NaCl evoked greater increases in OVLT neuronal discharge of Dahl S versus Dahl R rats regardless of NaCl diet. The activity of non-NaCl-responsive OVLT neurons was not different across strain or diets. Finally, inhibition of OVLT neurons by local injection of the gamma-aminobutyric acid agonist muscimol produced a greater decrease in renal SNA, splanchnic SNA, and ABP of Dahl S rats fed 4% versus 0.1% or Dahl R rats. CONCLUSIONS A high salt diet activates NaCl-responsive OVLT neurons to increase SNA and ABP in salt-sensitive hypertension.
Collapse
Affiliation(s)
- Sean D Stocker
- Department of Neurobiology, University of Pittsburgh School of Medicine, PA
| |
Collapse
|
47
|
Travers S, Bertoye C, Daudon M, Courbebaisse M, Baron S. How to Monitor Hydration Status and Urine Dilution in Patients with Nephrolithiasis. Nutrients 2023; 15:nu15071642. [PMID: 37049482 PMCID: PMC10097240 DOI: 10.3390/nu15071642] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/21/2023] [Accepted: 03/27/2023] [Indexed: 03/30/2023] Open
Abstract
Maintenance of hydration status requires a tight balance between fluid input and output. An increase in water loss or a decrease in fluid intake is responsible for dehydration status, leading to kidney water reabsorption. Thus, urine volume decreases and concentration of the different solutes increases. Urine dilution is the main recommendation to prevent kidney stone recurrence. Monitoring hydration status and urine dilution is key to preventing stone recurrence. This monitoring could either be performed via spot urine or 24 h urine collection with corresponding interpretation criteria. In laboratory conditions, urine osmolality measurement is the best tool to evaluate urine dilution, with less interference than urine-specific gravity measurement. However, this evaluation is only available during time lab examination. To improve urine dilution in nephrolithiasis patients in daily life, such monitoring should also be available at home. Urine color is of poor interest, but reagent strips with urine-specific gravity estimation are currently the only available tool, even with well-known interferences. Finally, at home, fluid intake monitoring could be an alternative to urine dilution monitoring. Eventually, the use of a connected device seems to be the most promising solution.
Collapse
|
48
|
Zagmutt S, Mera P, González-García I, Ibeas K, Romero MDM, Obri A, Martin B, Esteve-Codina A, Soler-Vázquez MC, Bastias-Pérez M, Cañes L, Augé E, Pelegri C, Vilaplana J, Ariza X, García J, Martinez-González J, Casals N, López M, Palmiter R, Sanz E, Quintana A, Herrero L, Serra D. CPT1A in AgRP neurons is required for sex-dependent regulation of feeding and thirst. Biol Sex Differ 2023; 14:14. [PMID: 36966335 PMCID: PMC10040140 DOI: 10.1186/s13293-023-00498-8] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 03/10/2023] [Indexed: 03/27/2023] Open
Abstract
BACKGROUND Fatty acid metabolism in the hypothalamus has an important role in food intake, but its specific role in AgRP neurons is poorly understood. Here, we examined whether carnitinea palmitoyltransferase 1A (CPT1A), a key enzyme in mitochondrial fatty acid oxidation, affects energy balance. METHODS To obtain Cpt1aKO mice and their control littermates, Cpt1a(flox/flox) mice were crossed with tamoxifen-inducible AgRPCreERT2 mice. Food intake and body weight were analyzed weekly in both males and females. At 12 weeks of age, metabolic flexibility was determined by ghrelin-induced food intake and fasting-refeeding satiety tests. Energy expenditure was analyzed by calorimetric system and thermogenic activity of brown adipose tissue. To study fluid balance the analysis of urine and water intake volumes; osmolality of urine and plasma; as well as serum levels of angiotensin and components of RAAS (renin-angiotensin-aldosterone system) were measured. At the central level, changes in AgRP neurons were determined by: (1) analyzing specific AgRP gene expression in RiboTag-Cpt1aKO mice obtained by crossing Cpt1aKO mice with RiboTag mice; (2) measuring presynaptic terminal formation in the AgRP neurons with the injection of the AAV1-EF1a-DIO-synaptophysin-GFP in the arcuate nucleus of the hypothalamus; (3) analyzing AgRP neuronal viability and spine formations by the injection AAV9-EF1a-DIO-mCherry in the arcuate nucleus of the hypothalamus; (4) analyzing in situ the specific AgRP mitochondria in the ZsGreen-Cpt1aKO obtained by breeding ZsGreen mice with Cpt1aKO mice. Two-way ANOVA analyses were performed to determine the contributions of the effect of lack of CPT1A in AgRP neurons in the sex. RESULTS Changes in food intake were just seen in male Cpt1aKO mice while only female Cpt1aKO mice increased energy expenditure. The lack of Cpt1a in the AgRP neurons enhanced brown adipose tissue activity, mainly in females, and induced a substantial reduction in fat deposits and body weight. Strikingly, both male and female Cpt1aKO mice showed polydipsia and polyuria, with more reduced serum vasopressin levels in females and without osmolality alterations, indicating a direct involvement of Cpt1a in AgRP neurons in fluid balance. AgRP neurons from Cpt1aKO mice showed a sex-dependent gene expression pattern, reduced mitochondria and decreased presynaptic innervation to the paraventricular nucleus, without neuronal viability alterations. CONCLUSIONS Our results highlight that fatty acid metabolism and CPT1A in AgRP neurons show marked sex differences and play a relevant role in the neuronal processes necessary for the maintenance of whole-body fluid and energy balance.
Collapse
Affiliation(s)
- Sebastián Zagmutt
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028, Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Paula Mera
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028, Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Ismael González-García
- NeurObesity Group, Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Kevin Ibeas
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028, Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - María Del Mar Romero
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028, Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Arnaud Obri
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Beatriz Martin
- CNAG-CRG, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, 08028, Barcelona, Spain
| | - Anna Esteve-Codina
- CNAG-CRG, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, 08028, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - M Carmen Soler-Vázquez
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028, Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Marianela Bastias-Pérez
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028, Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Laia Cañes
- Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Barcelona, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Biomédica Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Elisabeth Augé
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Carme Pelegri
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Institute of Neurosciences of the Universitat de Barcelona, Barcelona, Spain
| | - Jordi Vilaplana
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Institute of Neurosciences of the Universitat de Barcelona, Barcelona, Spain
| | - Xavier Ariza
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Inorganic & Organic Chemistry, Faculty of Chemistry, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Jordi García
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Inorganic & Organic Chemistry, Faculty of Chemistry, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - José Martinez-González
- Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Barcelona, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Biomédica Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Núria Casals
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Basic Sciences, Faculty of Medicine & Health Sciences, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Spain
| | - Miguel López
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- NeurObesity Group, Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Richard Palmiter
- Department of Biochemistry, Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - Elisenda Sanz
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Albert Quintana
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Laura Herrero
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028, Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Dolors Serra
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028, Barcelona, Spain.
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
49
|
Haan KD, Park SJ, Nakamura Y, Fukami K, Fisher TE. Osmotically evoked PLCδ1-dependent translocation of ΔN-TRPV1 channels in rat supraoptic neurons. iScience 2023; 26:106258. [PMID: 36926650 PMCID: PMC10011827 DOI: 10.1016/j.isci.2023.106258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/10/2022] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
Osmoregulation is an essential homeostatic process that requires constant release of vasopressin during sustained increases in plasma osmolality. The magnocellular neurosecretory cells (MNCs) respond to increases in external osmolality through increases in the activity of ΔN-TRPV1 channels, which leads to increased action potential firing and vasopressin release. We show that sustained exposure of acutely isolated rat and mouse MNCs to hypertonic solutions (90 min) causes a reversible translocation of ΔN-TRPV1 channels from internal stores to the plasma membrane that depends on the activation of phospholipase C and on SNARE-dependent exocytosis. ΔN-TRPV1 channel translocation is absent in MNCs isolated from transgenic mice lacking the PLCδ1 isoform, suggesting that PLCδ1 is essential for triggering this process. The translocation of ΔN-TRPV1 channels to the cell surface could contribute to the maintenance of MNC excitability during sustained increases in osmolality. Our data therefore have important implications for the mechanisms underlying mammalian osmoregulation.
Collapse
Affiliation(s)
- Kirk D Haan
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Sung Jin Park
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Yoshikazu Nakamura
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba, Japan
| | - Kiyoko Fukami
- Laboratory of Genome and Biosignals, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Thomas E Fisher
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
50
|
Liu H, Wu JJ, Li R, Wang PZ, Huang JH, Xu Y, Zhao JL, Wu PP, Li SJ, Wu ZX. Disexcitation in the ASH/RIM/ADL negative feedback circuit fine-tunes hyperosmotic sensation and avoidance in Caenorhabditis elegans. Front Mol Neurosci 2023; 16:1101628. [PMID: 37008778 PMCID: PMC10050701 DOI: 10.3389/fnmol.2023.1101628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 02/21/2023] [Indexed: 03/17/2023] Open
Abstract
Sensations, especially nociception, are tightly controlled and regulated by the central and peripheral nervous systems. Osmotic sensation and related physiological and behavioral reactions are essential for animal well-being and survival. In this study, we find that interaction between secondary nociceptive ADL and primary nociceptive ASH neurons upregulates Caenorhabditis elegans avoidance of the mild and medium hyperosmolality of 0.41 and 0.88 Osm but does not affect avoidance of high osmolality of 1.37 and 2.29 Osm. The interaction between ASH and ADL is actualized through a negative feedback circuit consisting of ASH, ADL, and RIM interneurons. In this circuit, hyperosmolality-sensitive ADL augments the ASH hyperosmotic response and animal hyperosmotic avoidance; RIM inhibits ADL and is excited by ASH; thus, ASH exciting RIM reduces ADL augmenting ASH. The neuronal signal integration modality in the circuit is disexcitation. In addition, ASH promotes hyperosmotic avoidance through ASH/RIC/AIY feedforward circuit. Finally, we find that in addition to ASH and ADL, multiple sensory neurons are involved in hyperosmotic sensation and avoidance behavior.
Collapse
|