1
|
Naganuma J, Koyama S, Watabe Y, Yoshihara S. A Boy with Reset Osmostat Who Developed Chronic Hyponatremia due to Hypothalamic Injury Caused By a Giant Arachnoid Cyst. J Clin Res Pediatr Endocrinol 2024; 16:489-494. [PMID: 36794751 PMCID: PMC11629720 DOI: 10.4274/jcrpe.galenos.2023.2022-7-16] [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: 09/12/2022] [Accepted: 01/10/2023] [Indexed: 02/17/2023] Open
Abstract
Reset osmostat (RO) is classified as type C among the four subtypes of the syndrome of inappropriate secretion of antidiuretic hormone based on antidiuretic hormone (ADH) secretion. It is characterized by a lower plasma osmolality threshold for ADH excretion when plasma sodium concentration is reduced. We report the case of a boy with RO and a giant arachnoid cyst (AC). The patient had been suspected of having AC since the fetal period, and a giant AC in the prepontine cistern was confirmed by brain magnetic resonance imaging seven days after birth. During the neonatal period, there were no abnormalities in the general condition or blood tests, and he was discharged from neonatal intensive care at 27 days after birth. He was born with a -2 standard deviation score birth length and mild mental retardation. When he was six years old, he was diagnosed with infectious impetigo and had hyponatremia of 121 mmol/L. Investigations revealed normal adrenal and thyroid functions, plasma hypo-osmolality, high urinary sodium, and high urinary osmolality. The 5% hypertonic saline and water load tests confirmed that ADH was secreted under low sodium and osmolality conditions, and the ability to concentrate urine and excrete a standard water load; therefore, RO was diagnosed. In addition, an anterior pituitary hormone secretion stimulation test was performed, which confirmed growth hormone secretion deficiency and gonadotropin hyperreactivity. Hyponatremia was untreated, but fluid restriction and salt loading were started at 12 years old because of the risk of growth obstacles. The diagnosis of RO is important from the viewpoint of clinical hyponatremia treatment options.
Collapse
Affiliation(s)
- Junko Naganuma
- Dokkyo Medical University Faculty of Medicine Department of Pediatrics, Tochigi, Japan
| | - Satomi Koyama
- Dokkyo Medical University Faculty of Medicine Department of Pediatrics, Tochigi, Japan
| | - Yoshiyuki Watabe
- Dokkyo Medical University Faculty of Medicine Department of Pediatrics, Tochigi, Japan
| | - Shigemi Yoshihara
- Dokkyo Medical University Faculty of Medicine Department of Pediatrics, Tochigi, Japan
| |
Collapse
|
2
|
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] [MESH Headings] [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
|
3
|
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
|
4
|
Kakigi A, Egami N, Fujita T, Uehara N, Yokoi J, Koda K, Nibu KI, Yamashita S, Yamasoba T. Live Imaging of the Dehydration Effect of Isosorbide on the Normal and Hydropic Guinea Pig Cochleae Using Optical Coherence Tomography. Otol Neurotol 2023; 44:833-837. [PMID: 37464452 DOI: 10.1097/mao.0000000000003962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
OBJECTIVE To investigate the isosorbide-induced dehydration effect on the endolymphatic space by intratympanic administration of isosorbide. BACKGROUND Isosorbide, an osmotic diuretic, is used orally as a typical conservative therapy for Menière's disease (MD) in Japan. The dehydration effect occurs 6 hours after isosorbide ingestion. Intratympanic administration of isosorbide resolves endolymphatic hydrops faster than oral ingestion. In addition, the dehydration effect has never been shown directly. Therefore, we investigated the dehydration effect of intratympanic administration of isosorbide on endolymphatic hydrops using optical coherence tomography. METHODS We used eight Hartley guinea pigs, divided into normal and hydrops groups. In the hydrops group, the animals underwent endolymphatic sac obliteration to create endolymphatic hydrops. We obtained midmodiolar section images of the cochleae using optical coherence tomography. Then, 50 to 70% isosorbide was sequentially administered intratympanically for 5 minutes, and the apical turn of the cochlea was observed. The relative midmodiolar cross-sectional area of the scala media was calculated for quantitative assessment of the endolymphatic space. RESULTS In the normal group, 50% isosorbide had a slight but significant dehydration effect on the scala media; at 55 to 70%, Reissner's membrane became flat. In the hydrops group, 50% isosorbide slightly reduced endolymphatic hydrops; 65% flattened Reissner's membrane, and 70% slightly concaved it toward the basilar membrane. CONCLUSION The results suggest that we could select the concentration of isosorbide according to the stage or severity of MD and relief from acute attack. Intratympanic administration of isosorbide may be a promising treatment for patients with MD.
Collapse
Affiliation(s)
- Akinobu Kakigi
- Department of Otolaryngology-Head and Neck Surgery, Kobe University, Graduate School of Medicine, Kobe, Hyogo
| | - Naoya Egami
- Department of Otolaryngology, Faculty of Medicine, The University of Tokyo, Hongo
| | - Takeshi Fujita
- Department of Otolaryngology-Head and Neck Surgery, Kobe University, Graduate School of Medicine, Kobe, Hyogo
| | - Natsumi Uehara
- Department of Otolaryngology-Head and Neck Surgery, Kobe University, Graduate School of Medicine, Kobe, Hyogo
| | - Jun Yokoi
- Department of Otolaryngology-Head and Neck Surgery, Kobe University, Graduate School of Medicine, Kobe, Hyogo
| | - Kento Koda
- Department of Otolaryngology, Faculty of Medicine, The University of Tokyo, Hongo
| | - Ken-Ichi Nibu
- Department of Otolaryngology-Head and Neck Surgery, Kobe University, Graduate School of Medicine, Kobe, Hyogo
| | - Shinji Yamashita
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro-ku, Tokyo, Japan
| | - Tatsuya Yamasoba
- Department of Otolaryngology, Faculty of Medicine, The University of Tokyo, Hongo
| |
Collapse
|
5
|
Limited Effect of Dehydrating via Active vs. Passive Heat Stress on Plasma Volume or Osmolality, Relative to the Effect of These Stressors per Se. Nutrients 2023; 15:nu15040904. [PMID: 36839262 PMCID: PMC9959915 DOI: 10.3390/nu15040904] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/05/2023] [Accepted: 02/06/2023] [Indexed: 02/15/2023] Open
Abstract
The physiological, perceptual, and functional effects of dehydration may depend on how it is incurred (e.g., intense exercise releases endogenous water via glycogenolysis) but this basic notion has rarely been examined. We investigated the effects of active (exercise) heat- vs. passive heat-induced dehydration, and the kinetics of ad libitum rehydration following each method. Twelve fit participants (five females and seven males) completed four trials in randomised order: DEHydration to -3% change in body mass (∆BM) under passive or active heat stress, and EUHydration to prevent ∆BM under passive or active heat stress. In all trials, participants then sat in a temperate-controlled environment, ate a standard snack and had free access to water and sports drink during their two-hour recovery. During mild dehydration (≤2% ∆BM), active and passive heating caused comparable increases in plasma osmolality (Posm: ~4 mOsmol/kg, interaction: p = 0.138) and reductions in plasma volume (PV: ~10%, interaction: p = 0.718), but heat stress per se was the main driver of hypovolaemia. Thirst in DEHydration was comparably stimulated by active than passive heat stress (p < 0.161) and shared the same relation to Posm (r ≥ 0.744) and ∆BM (r ≥ 0.882). Following heat exposures, at 3% gross ∆BM, PV reduction was approximately twice as large from passive versus active heating (p = 0.003), whereas Posm perturbations were approximately twice as large from EUHydration versus DEHydration (p < 0.001). Rehydrating ad libitum resulted in a similar net fluid balance between passive versus active heat stress and restored PV despite the incomplete replacement of ∆BM. In conclusion, dehydrating by 2% ∆BM via passive heat stress generally did not cause larger changes to PV or Posm than via active heat stress. The heat stressors themselves caused a greater reduction in PV than dehydration did, whereas ingesting water to maintain euhydration produced large reductions in Posm in recovery and therefore appears to be of more physiological significance.
Collapse
|
6
|
Lu HAJ, He J. Aquaporins in Diabetes Insipidus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1398:267-279. [PMID: 36717500 DOI: 10.1007/978-981-19-7415-1_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Disruption of water and electrolyte balance is frequently encountered in clinical medicine. Regulating water metabolism is critically important. Diabetes insipidus (DI) presented with excessive water loss from the kidney is a major disorder of water metabolism. To understanding the molecular and cellular mechanisms and pathophysiology of DI and rationales of clinical management of DI is important for both research and clinical practice. This chapter will first review various forms of DI focusing on central diabetes insipidus (CDI) and nephrogenic diabetes insipidus (NDI). This is followed by a discussion of regulatory mechanisms underlying CDI and NDI, with a focus on the regulatory axis of vasopressin, vasopressin receptor 2 (V2R) and the water channel molecule, aquaporin 2 (AQP2). The clinical manifestation, diagnosis, and management of various forms of DI will also be discussed with highlights of some of the latest therapeutic strategies that are developed from in vitro experiments and animal studies.
Collapse
Affiliation(s)
- H A Jenny Lu
- Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Jinzhao He
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
7
|
Dmitrieva NI, Gagarin A, Liu D, Wu CO, Boehm M. Middle-age high normal serum sodium as a risk factor for accelerated biological aging, chronic diseases, and premature mortality. EBioMedicine 2023; 87:104404. [PMID: 36599719 PMCID: PMC9873684 DOI: 10.1016/j.ebiom.2022.104404] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 11/22/2022] [Accepted: 11/22/2022] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND It is known that some people age faster than others, some people live into old age disease-free, while others develop age-related chronic diseases. With a rapidly aging population and an emerging chronic diseases epidemic, finding mechanisms and implementing preventive measures that could slow down the aging process has become a new challenge for biomedical research and public health. In mice, lifelong water restriction shortens the lifespan and promotes degenerative changes. Here, we test the hypothesis that optimal hydration may slow down the aging process in humans. METHODS We performed a cohort analysis of data from the Atherosclerosis Risk in Communities study with middle-age enrollment (45-66 years, n = 15,752) and 25 years follow-up. We used serum sodium, as a proxy for hydration habits. To estimate the relative speed of aging, we calculated the biological age (BA) from age-dependent biomarkers and assessed risks of chronic diseases and premature mortality. FINDINGS The analysis showed that middle age serum sodium >142 mmol/l is associated with a 39% increased risk to develop chronic diseases (hazard ratio [HR] = 1.39, 95% confidence interval [CI]:1.18-1.63) and >144 mmol/l with 21% elevated risk of premature mortality (HR = 1.21, 95% CI:1.02-1.45). People with serum sodium >142 mmol/l had up to 50% higher odds to be older than their chronological age (OR = 1.50, 95% CI:1.14-1.96). A higher BA was associated with an increased risk of chronic diseases (HR = 1.70, 95% CI:1.50-1.93) and premature mortality (HR = 1.59, 95% CI 1.39-1.83). INTERPRETATION People whose middle-age serum sodium exceeds 142 mmol/l have increased risk to be biologically older, develop chronic diseases and die at younger age. Intervention studies are needed to confirm the link between hydration and aging. FUNDING This work was funded by Intramural Research program of the National Heart, Lung, and Blood Institute (NHLBI). The ARIC study has been funded in whole or in part with federal funds from the NHLBI; the National Institutes of Health (NIH); and the Department of Health and Human Services.
Collapse
Affiliation(s)
- Natalia I Dmitrieva
- The Laboratory of Cardiovascular Regenerative Medicine, National Heart Lung and Blood Institute, Bethesda, MD, 20892, USA.
| | - Alessandro Gagarin
- The Laboratory of Cardiovascular Regenerative Medicine, National Heart Lung and Blood Institute, Bethesda, MD, 20892, USA
| | - Delong Liu
- The Laboratory of Vascular and Matrix Genetics, National Heart Lung and Blood Institute, Bethesda, MD, 20892, USA
| | - Colin O Wu
- Office of Biostatistics Research, National Heart Lung and Blood Institute, Bethesda, MD, 20892, USA
| | - Manfred Boehm
- The Laboratory of Cardiovascular Regenerative Medicine, National Heart Lung and Blood Institute, Bethesda, MD, 20892, USA
| |
Collapse
|
8
|
Brown EF, Fronius M, Brown CH. Vasopressin regulation of maternal body fluid balance in pregnancy and lactation: A role for TRPV channels? Mol Cell Endocrinol 2022; 558:111764. [PMID: 36038076 DOI: 10.1016/j.mce.2022.111764] [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: 01/26/2022] [Revised: 06/16/2022] [Accepted: 08/22/2022] [Indexed: 12/15/2022]
Abstract
Renal water reabsorption increases in pregnancy and lactation to expand maternal blood volume to cope with the cardiovascular demands of the developing fetus and new-born baby. Vasopressin (antidiuretic hormone) promotes renal water reabsorption and its secretion is principally stimulated by body fluid osmolality. Hence, lowered osmolality normally decreases vasopressin secretion. However, despite water retention profoundly reducing osmolality in pregnancy and lactation, vasopressin levels are maintained to drive blood volume expansion. Despite its importance for successful reproduction, the cellular mechanisms that maintain vasopressin secretion in the face of decreased osmolality during pregnancy and lactation are unknown. Vasopressin is secreted by neurons that are intrinsically osmosensitive through expression of N-terminal truncated-transient receptor potential vanilloid-1 channel, ΔN-TRPV1, which is mechanically activated by osmotically-induced cell shrinkage to increase vasopressin neuron activity. Vasopressin neurons also express TRPV4 but the role of TRPV4 in vasopressin neuron function is not well characterised. Here, we summarise our novel evidence showing that TRPV4 forms functional channels with ΔN-TRPV1 that have a greater single-channel conductance compared to channels with ΔN-TRPV1 alone. We propose that upregulation of TRPV4 heteromerisation with ΔN-TRPV1 might maintain vasopressin secretion in pregnancy and lactation to expand blood volume for successful reproduction.
Collapse
Affiliation(s)
- Emily F Brown
- Brain Health Research Centre, University of Otago, Dunedin, Aotearoa New Zealand; Centre for Neuroendocrinology, University of Otago, Dunedin, Aotearoa New Zealand; HeartOtago, University of Otago, Dunedin, Aotearoa New Zealand; Department of Physiology, University of Otago, Dunedin, Aotearoa New Zealand.
| | - Martin Fronius
- HeartOtago, University of Otago, Dunedin, Aotearoa New Zealand; Department of Physiology, University of Otago, Dunedin, Aotearoa New Zealand.
| | - Colin H Brown
- Brain Health Research Centre, University of Otago, Dunedin, Aotearoa New Zealand; Centre for Neuroendocrinology, University of Otago, Dunedin, Aotearoa New Zealand; HeartOtago, University of Otago, Dunedin, Aotearoa New Zealand; Department of Physiology, University of Otago, Dunedin, Aotearoa New Zealand.
| |
Collapse
|
9
|
Sudbury JR, Zaelzer C, Trudel E, Bumagin A, Bourque CW. Synaptic control of rat magnocellular neurosecretory cells by warm-sensing neurons in the organum vasculosum lamina terminalis. J Neuroendocrinol 2022; 34:e13214. [PMID: 36426844 DOI: 10.1111/jne.13214] [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: 04/22/2022] [Revised: 10/07/2022] [Accepted: 10/27/2022] [Indexed: 11/11/2022]
Abstract
Increases in core body temperature cause secretion of vasopressin (vasopressin, antidiuretic hormone) to promote water reabsorption and blunt water losses incurred through homeostatic evaporative cooling. Subtypes of transient receptor potential vanilloid (Trpv) channels have been shown to contribute to the intrinsic regulation of vasopressin-releasing magnocellular neurosecretory cells (MNCs) in the supraoptic nucleus (SON) and paraventricular nucleus (PVN). However, MNCs in vivo can also be excited by local heating of the adjacent preoptic area, indicating they receive thermosensory information from other areas. Here, we investigated whether neurons in the organum vasculosum lamina terminalis (OVLT) contribute to this process using in vitro electrophysiological approaches in male rats. We found that the majority of OVLT neurons are thermosensitive in the physiological range (36-39°C) and that this property is retained under conditions blocking synaptic transmission. A subset of these neurons could be antidromically activated by electrical stimulation in the SON. Whole cell recordings from SON MNCs revealed that heating significantly increases the rate of spontaneous excitatory postsynaptic currents (sEPCSs), and that this response is abolished by lesions targeting the OVLT, but not by bilateral lesions placed in the adjacent preoptic area. Finally, local heating of the OVLT caused a significant excitation of MNCs in the absence of temperature changes in the SON, and this effect was blocked by inhibitors of ionotropic glutamate receptors. These findings indicate that the OVLT serves as an important thermosensory nucleus and contributes to the activation of MNCs during physiological heating.
Collapse
Affiliation(s)
- Jessica R Sudbury
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Cristian Zaelzer
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Eric Trudel
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Anna Bumagin
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Charles W Bourque
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
10
|
Dmitrieva NI, Liu D, Wu CO, Boehm M. Middle age serum sodium levels in the upper part of normal range and risk of heart failure. Eur Heart J 2022; 43:3335-3348. [PMID: 35348651 PMCID: PMC10263272 DOI: 10.1093/eurheartj/ehac138] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 02/01/2022] [Accepted: 03/03/2022] [Indexed: 07/29/2023] Open
Abstract
AIMS With increasing prevalence of heart failure (HF) owing to the ageing population, identification of modifiable risk factors is important. In a mouse model, chronic hypohydration induced by lifelong water restriction promotes cardiac fibrosis. Hypohydration elevates serum sodium. Here, we evaluate the association of serum sodium at middle age as a measure of hydration habits with risk to develop HF. METHODS AND RESULTS We analysed data from Atherosclerosis Risk in Communities study with middle age enrolment (45-66 years) and 25 years of follow-up. Participants without water balance dysregulation were selected: serum sodium within normal range (135-146 mmol/L), not diabetic, not obese and free of HF at baseline (N = 11 814). In time-to-event analysis, HF risk was increased by 39% if middle age serum sodium exceeded 143 mmol/L corresponding to 1% body weight water deficit [hazard ratio 1.39, 95% confidence interval (CI) 1.14-1.70]. In a retrospective case-control analysis performed on 70- to 90-year-old attendees of Visit 5 (N = 4961), serum sodium of 142.5-143 mmol/L was associated with 62% increase in odds of left ventricular hypertrophy (LVH) diagnosis [odds ratio (OR) 1.62, 95% CI 1.03-2.55]. Serum sodium above 143 mmol/L was associated with 107% increase in odds of LVH (OR 2.07, 95% CI 1.30-3.28) and 54% increase in odds of HF (OR 1.54, 95% CI 1.06-2.23). As a result, prevalence of HF and LVH was increased among 70- to 90-year-old participants with higher middle age serum sodium. CONCLUSION Middle age serum sodium above 142 mmol is a risk factor for LVH and HF. Maintaining good hydration throughout life may slow down decline in cardiac function and decrease prevalence of HF.
Collapse
Affiliation(s)
- Natalia I Dmitrieva
- The Laboratory of Cardiovascular Regenerative Medicine, National Heart Lung and Blood Institute, Bethesda, MD 20892, USA
| | - Delong Liu
- The Laboratory of Vascular and Matrix Genetics, National Heart Lung and Blood Institute, Bethesda, MD 20892, USA
| | - Colin O Wu
- Office of Biostatistics Research, National Heart Lung and Blood Institute, Bethesda, MD 20892, USA
| | - Manfred Boehm
- The Laboratory of Cardiovascular Regenerative Medicine, National Heart Lung and Blood Institute, Bethesda, MD 20892, USA
| |
Collapse
|
11
|
Baranauskas M, Kupčiūnaitė I, Stukas R. The Association between Rapid Weight Loss and Body Composition in Elite Combat Sports Athletes. Healthcare (Basel) 2022; 10:665. [PMID: 35455842 PMCID: PMC9031560 DOI: 10.3390/healthcare10040665] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/18/2022] [Accepted: 03/31/2022] [Indexed: 02/07/2023] Open
Abstract
Rapid Weight Loss (RWL) is a rapid reduction in weight over a short period of time seeking to attain the norm required for a competition in a particular weight category. RWL has a negative health impact on athletes including the significant muscle damage induced by RWL. This study aimed to identify the association between RWL and body composition among competitive combat athletes (n = 43) in Lithuania. Our focus was laid on the disclosure of their RWL practice by using a previously standardized RWL Questionnaire. The body composition of the athletes was measured by means of the standing-posture 8-12-electrode multi-frequency bioelectrical impedance analysis (BIA) and the electrical signals of 5, 50, 250, 550 and 1000 kHz. This non-experimental cross-sectional study resulted in preliminary findings on the prevalence and profile of RWL among combat athletes in Lithuania. 88% of the athletes surveyed in our study had lost weight in order to compete, with the average weight loss of 4.6 ± 2% of the habitual body mass. The athletes started to resort to weight cycling as early as 9 years old, with a mean age of 12.8 ± 2.1 years. The combination of practiced weight loss techniques such as skipping meals (adjusted Odd Ratio (AOR) 6.3; 95% CI: 1.3−31.8), restricting fluids (AOR 5.5; 95% CI: 1.0−31.8), increased exercise (AOR 3.6; 95% CI: 1.0−12.5), training with rubber/plastic suits (AOR 3.2; 95% CI: 0.9−11.3) predicted the risk of RWL aggressiveness. RWL magnitude potentially played an important role in maintaining the loss of muscle mass in athletes during the preparatory training phase (β −0.01 kg, p < 0.001). Therefore, an adequate regulatory programme should be integrated into the training plans of high-performance combat sports athletes to keep not only the athletes but also their coaches responsible for a proper weight control.
Collapse
Affiliation(s)
- Marius Baranauskas
- Faculty of Biomedical Sciences, Panevėžys University of Applied Sciences, 35200 Panevėžys, Lithuania;
| | - Ingrida Kupčiūnaitė
- Faculty of Biomedical Sciences, Panevėžys University of Applied Sciences, 35200 Panevėžys, Lithuania;
| | - Rimantas Stukas
- Department of Public Health, Institute of Health Sciences, Faculty of Medicine, Vilnius University, 01513 Vilnius, Lithuania;
| |
Collapse
|
12
|
Comparing thirst and spot urine concentrations in humans of differing body sizes: An observational study. Physiol Behav 2021; 245:113673. [PMID: 34921838 DOI: 10.1016/j.physbeh.2021.113673] [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: 10/21/2021] [Revised: 12/02/2021] [Accepted: 12/12/2021] [Indexed: 11/21/2022]
Abstract
Elevated body mass index (BMI) has been associated with elevated urine osmolality (UOsm), despite having higher total water intake, but it is unclear if overweight/obese individuals have reduced thirst. In this observational study, we found that overweight/obese individuals had higher UOsm compared to normal-weight individuals (749 ± 37 vs. 624 ± 35 mmol•kg-1; P < 0.01) while possessing similar thirst ratings (56.4 ± 3 vs. 51.6 ± 3 mm; P = 0.3). In this observational study, overweight/obese individuals possessed more concentrated urine in the absence of higher thirst perception.
Collapse
|
13
|
Che K, Muttenthaler M, Kurzbach D. Conformational selection of vasopressin upon V 1a receptor binding. Comput Struct Biotechnol J 2021; 19:5826-5833. [PMID: 34765097 PMCID: PMC8567363 DOI: 10.1016/j.csbj.2021.10.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 10/14/2021] [Accepted: 10/14/2021] [Indexed: 11/30/2022] Open
Abstract
The neuropeptide vasopressin (VP) and its three G protein-coupled receptors (V1aR, V1bR and V2R) are of high interest in a wide array of drug discovery programs. V1aR is of particular importance due to its cardiovascular functions and diverse roles in the central nervous system. The structure–activity relationships underpinning ligand-receptor interactions remain however largely unclear, hindering rational drug design. This is not least due to the high structural flexibility of VP in its free as well as receptor-bound states. In this work, we developed a novel approach to reveal features of conformational selectivity upon VP-V1aR complex formation. We employed virtual screening strategies to probe VP’s conformational space for transiently adopted structures that favor binding to V1aR. To this end, we dissected the VP conformational space into three sub-ensembles, each containing distinct structural sets for VP’s three-residue C-terminal tail. We validated the computational results with experimental nuclear magnetic resonance (NMR) data and docked each sub-ensemble to V1aR. We observed that the conformation of VP’s three-residue tail significantly modulated the complex dissociation constants. Solvent-exposed and proline trans-configured VP tail conformations bound to the receptor with three-fold enhanced affinities compared to compacted or cis-configured conformations. The solvent-exposed and more flexible structures facilitated unique interaction patterns between VP and V1aR transmembrane helices 3, 4, and 6 which led to high binding energies. The presented “virtual conformational space screening” approach, integrated with NMR spectroscopy, thus enabled identification and characterization of a conformational selection-type complex formation mechanism that confers novel perspectives on targeting the VP-V1aR interactions at the level of the encounter complex – an aspect that opens novel research avenues for understanding the functionality of the evolutionary selected conformational properties of VP, as well as guidance for ligand design strategies to provide more potent and selective VP analogues.
Collapse
Affiliation(s)
- Kateryna Che
- University Vienna, Faculty of Chemistry, Institute of Biological Chemistry, Währinger Str. 38, A-1090 Vienna, Austria
| | - Markus Muttenthaler
- University Vienna, Faculty of Chemistry, Institute of Biological Chemistry, Währinger Str. 38, A-1090 Vienna, Austria
- The University of Queensland, Institute for Molecular Bioscience, 306 Carmody Rd, 4072 St Lucia, Brisbane, Queensland, Australia
| | - Dennis Kurzbach
- University Vienna, Faculty of Chemistry, Institute of Biological Chemistry, Währinger Str. 38, A-1090 Vienna, Austria
- Corresponding author.
| |
Collapse
|
14
|
Opichka MA, Rappelt MW, Gutterman DD, Grobe JL, McIntosh JJ. Vascular Dysfunction in Preeclampsia. Cells 2021; 10:3055. [PMID: 34831277 PMCID: PMC8616535 DOI: 10.3390/cells10113055] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/01/2021] [Accepted: 11/04/2021] [Indexed: 01/22/2023] Open
Abstract
Preeclampsia is a life-threatening pregnancy-associated cardiovascular disorder characterized by hypertension and proteinuria at 20 weeks of gestation. Though its exact underlying cause is not precisely defined and likely heterogenous, a plethora of research indicates that in some women with preeclampsia, both maternal and placental vascular dysfunction plays a role in the pathogenesis and can persist into the postpartum period. Potential abnormalities include impaired placentation, incomplete spiral artery remodeling, and endothelial damage, which are further propagated by immune factors, mitochondrial stress, and an imbalance of pro- and antiangiogenic substances. While the field has progressed, current gaps in knowledge include detailed initial molecular mechanisms and effective treatment options. Newfound evidence indicates that vasopressin is an early mediator and biomarker of the disorder, and promising future therapeutic avenues include mitigating mitochondrial dysfunction, excess oxidative stress, and the resulting inflammatory state. In this review, we provide a detailed overview of vascular defects present during preeclampsia and connect well-established notions to newer discoveries at the molecular, cellular, and whole-organism levels.
Collapse
Affiliation(s)
- Megan A. Opichka
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (M.A.O.); (D.D.G.); (J.L.G.)
| | - Matthew W. Rappelt
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| | - David D. Gutterman
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (M.A.O.); (D.D.G.); (J.L.G.)
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Justin L. Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (M.A.O.); (D.D.G.); (J.L.G.)
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jennifer J. McIntosh
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (M.A.O.); (D.D.G.); (J.L.G.)
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
15
|
Gumbel JH, Yang CB, Hubscher CH. Timeline of Changes in Biomarkers Associated with Spinal Cord Injury-Induced Polyuria. Neurotrauma Rep 2021; 2:462-475. [PMID: 34901942 PMCID: PMC8655813 DOI: 10.1089/neur.2021.0046] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Deficits in upper and lower urinary tract function, which include detrusor overactivity, urinary incontinence, detrusor-sphincter dyssynergia, and polyuria, are among the leading issues that arise after spinal cord injury (SCI) affecting quality of life. Given that overproduction of urine (polyuria) has been shown to be associated with an imbalance in key regulators of body fluid homeostasis, the current study examined the timing of changes in levels of various relevant hormones, peptides, receptors, and channels post-contusion injury in adult male Wistar rats. The results show significant up- or downregulation at various time points, beginning at 7 days post-injury, in levels of urinary atrial natriuretic peptide, serum arginine vasopressin (AVP), kidney natriuretic peptide receptor-A, kidney vasopressin-2 receptor, kidney aquaporin-2 channels, and kidney epithelial sodium channels (β- and γ-, but not α-, subunits). The number of AVP-labeled neurons in the hypothalamus (supraoptic and -chiasmatic, but not paraventricular, nuclei) was also significantly altered at one or more time points. These data show significant fluctuations in key biomarkers involved in body fluid homeostasis during the post-SCI secondary injury phase, suggesting that therapeutic interventions (e.g., desmopressin, a synthetic analogue of AVP) should be considered early post-SCI.
Collapse
Affiliation(s)
- Jason H. Gumbel
- Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, Kentucky, USA
| | - Cui Bo Yang
- Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, Kentucky, USA
| | - Charles H. Hubscher
- Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, Kentucky, USA
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
16
|
Abstract
Diabetes insipidus (DI) is a disorder characterized by a high hypotonic urinary output of more than 50ml per kg body weight per 24 hours, with associated polydipsia of more than 3 liters a day [1,2]. Central DI results from inadequate secretion and usually deficient synthesis of Arginine vasopressin (AVP) in the hypothalamus or pituitary gland. Besides central DI further underlying etiologies of DI can be due to other primary forms (renal origin) or secondary forms of polyuria (resulting from primary polydipsia). All these forms belong to the Polyuria Polydipsia Syndrom (PPS). In most cases central and nephrogenic DI are acquired, but there are also congenital forms caused by genetic mutations of the AVP gene (central DI) [3] or by mutations in the gene for the AVP V2R or the AQP2 water channel (nephrogenic DI) [4]. Primary polydipsia (PP) as secondary form of polyuria includes an excessive intake of large amounts of fluid leading to polyuria in the presence of intact AVP secretion and appropriate antidiuretic renal response. Differentiation between the three mentioned entities is difficult [5], especially in patients with Primary polydipsia or partial, mild forms of DI [1,6], but different tests for differential diagnosis, most recently based on measurement of copeptin, and a thorough medical history mostly lead to the correct diagnosis. This is important since treatment strategies vary and application of the wrong treatment can be dangerous [7]. Treatment of central DI consists of fluid management and drug therapy with the synthetic AVP analogue Desmopressin (DDAVP), that is used as nasal or oral preparation in most cases. Main side effect can be dilutional hyponatremia [8]. In this review we will focus on central diabetes insipidus and describe the prevalence, the clinical manifestations, the etiology as well as the differential diagnosis and management of central diabetes insipidus in the out- and inpatient setting.
Collapse
|
17
|
Sekiguchi Y, Benjamin CL, Butler CR, Morrissey MC, Filep EM, Stearns RL, Lee EC, Casa DJ. Relationships Between WUT (Body Weight, Urine Color, and Thirst Level) Criteria and Urine Indices of Hydration Status. Sports Health 2021; 14:566-574. [PMID: 34465235 DOI: 10.1177/19417381211038494] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND A Venn diagram consisting of percentage body mass loss, urine color, and thirst perception (weight, urine, thirst [WUT]) has been suggested as a practical method to assess hydration status. However, no study to date has examined relationships between WUT and urine hydration indices. Thus, the purpose of this study was to investigate relationships between urine specific gravity, urine osmolality, and the WUT criteria. HYPOTHESIS Urine specific gravity and urine osmolality indicate hypohydration when the WUT criteria demonstrate hypohydration (≥2 markers). STUDY DESIGN Laboratory cohort study. LEVEL OF EVIDENCE Level 3. METHODS A total of 22 women (mean ± SD; age, 20 ± 1 years; mass, 65.4 ± 12.6 kg) and 21 men (age, 21 ± 1 years; body mass, 78.7 ± 14.6 kg) participated in this study. First morning body mass, urine color, urine specific gravity, urine osmolality, and thirst level were collected for 10 consecutive days in a free-living situation. Body mass loss >1%, urine color >5, and thirst level ≥5 were used as the dehydration thresholds. The number of markers that indicated dehydration levels were counted and categorized into either 3, 2, 1, or 0 WUT markers that indicated dehydration. One-way analysis of variance with Tukey pairwise comparisons was used to assess the differences in urine specific gravity and urine osmolality between the different number of WUT markers. RESULTS Urine specific gravity in 3 WUT markers (mean ± SD [effect size], 1.021 ± 0.007 [0.57]; P = 0.025) and 2 WUT markers (1.019 ± 0.010 [0.31]; P = 0.026) was significantly higher than 1 WUT marker (1.016 ± 0.009). Urine mosmolality in 2 WUT markers (705 ± 253 mOsmol [0.43]; P = 0.018) was significantly higher than 1 WUT (597 ± 253 mOsmol). Meeting at least 2 WUT markers resulted in sensitivities of 0.652 (2 WUT criteria met) and 0.933 (3 WUT criteria met) to detect urine osmolality >700 mOsmol. CONCLUSION These results suggest that when 3 WUT markers are met, urine specific gravity and urine osmolality were greater than euhydration cutoff points. The WUT criterion is a useful tool to use in field settings to assess hydration status when first morning urine sample was used. CLINICAL RELEVANCE Athletes, coaches, sports scientists, and medical professionals can use WUT criteria to monitor dehydration with reduced cost and time.
Collapse
|
18
|
Jia D, Peroni M, Khalapyan T, Esmaily M. An Efficient Assisted Bidirectional Glenn Design With Lowered Superior Vena Cava Pressure for Stage-One Single Ventricle Patients. J Biomech Eng 2021; 143:071008. [PMID: 33590839 DOI: 10.1115/1.4050170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Indexed: 11/08/2022]
Abstract
Recently, the assisted bidirectional Glenn (ABG) procedure has been proposed as an alternative to the modified Blalock-Taussig shunt (mBTS) operation for neonates with single-ventricle physiology. Despite success in reducing heart workload and maintaining sufficient pulmonary flow, the ABG also raised the superior vena cava (SVC) pressure to a level that may not be tolerated by infants. To lower the SVC pressure, we propose a modified version of the ABG (mABG), in which a shunt with a slit-shaped nozzle exit is inserted at the junction of the right and left brachiocephalic veins. The proposed operation is compared against the ABG, the mBTS, and the bidirectional Glenn (BDG) operations using closed-loop multiscale simulations. Both normal (2.3 Wood units-m2) and high (7 Wood units-m2) pulmonary vascular resistance (PVR) values are simulated. The mABG provides the highest oxygen saturation, oxygen delivery, and pulmonary flow rate in comparison to the BDG and the ABG. At normal PVR, the SVC pressure is significantly reduced below that of the ABG and the BDG (mABG: 4; ABG: 8; BDG: 6; mBTS: 3 mmHg). However, the SVC pressure remains high at high PVR (mABG: 15; ABG: 16; BDG: 12; mBTS: 3 mmHg), motivating an optimization study to improve the ABG hemodynamics efficiency for a broader range of conditions in the future. Overall, the mABG preserves all advantages of the original ABG procedure while reducing the SVC pressure at normal PVR.
Collapse
Affiliation(s)
- Dongjie Jia
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY 14850
| | - Matthew Peroni
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY 14850
| | | | - Mahdi Esmaily
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY 14850
| |
Collapse
|
19
|
Adams JD, Darcy C, DeGrasse AG, Jordan R, Boscia CS. Crosstalk of Pain and Thirst Perception: A Brief Review. CHEMOSENS PERCEPT 2021. [DOI: 10.1007/s12078-021-09286-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
20
|
Nakagawa A, Yasumura Y, Yoshida C, Okumura T, Tateishi J, Yoshida J, Tamaki S, Yano M, Hayashi T, Nakagawa Y, Yamada T, Nakatani D, Hikoso S, Sakata Y. Prognostic relevance of elevated plasma osmolality on admission in acute decompensated heart failure with preserved ejection fraction: insights from PURSUIT-HFpEF registry. BMC Cardiovasc Disord 2021; 21:281. [PMID: 34098878 PMCID: PMC8182926 DOI: 10.1186/s12872-021-02098-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 06/03/2021] [Indexed: 12/11/2022] Open
Abstract
Background Complicated pathophysiology makes it difficult to identify the prognosis of heart failure with preserved ejection fraction (HFpEF). While plasma osmolality has been reported to have prognostic importance, mainly in heart failure with reduced ejection fraction (HFrEF), its prognostic meaning for HFpEF has not been elucidated. Methods We prospectively studied 960 patients in PURSUIT-HFpEF, a multicenter observational study of acute decompensated HFpEF inpatients. We divided patients into three groups according to the quantile values of plasma osmolality on admission. During a follow-up averaging 366 days, we examined the primary composite endpoint of cardiac mortality or heart failure re-admission using Kaplan–Meier curve analysis and Cox proportional hazard testing. Results 216 (22.5%) patients reached the primary endpoint. Kaplan–Meier curve analysis revealed that the highest quantile of plasma osmolality on admission (higher than 300.3 mOsm/kg) was significantly associated with adverse outcomes (Log-rank P = 0.0095). Univariable analysis in the Cox proportional hazard model also revealed significantly higher rates of adverse outcomes in the higher plasma osmolality on admission (hazard ratio [HR] 7.29; 95% confidence interval [CI] 2.25–23.92, P = 0.0009). Multivariable analysis in the Cox proportional hazard model also showed that higher plasma osmolality on admission was significantly associated with adverse outcomes (HR 5.47; 95% CI 1.46–21.56, P = 0.0113) independently from other confounding factors such as age, gender, comorbid of atrial fibrillation, hypertension history, diabetes, anemia, malnutrition, E/e′, and N-terminal pro-B-type natriuretic peptide elevation. Conclusions Higher plasma osmolality on admission was prognostically important for acute decompensated HFpEF inpatients. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-021-02098-z.
Collapse
Affiliation(s)
- Akito Nakagawa
- Division of Cardiology, Amagasaki Chuo Hospital, 1-12-1 Shioe, Amagasaki, Hyogo, 661-0976, Japan. .,Department of Medical Informatics, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, 565-0871, Japan.
| | - Yoshio Yasumura
- Division of Cardiology, Amagasaki Chuo Hospital, 1-12-1 Shioe, Amagasaki, Hyogo, 661-0976, Japan
| | - Chikako Yoshida
- Division of Cardiology, Amagasaki Chuo Hospital, 1-12-1 Shioe, Amagasaki, Hyogo, 661-0976, Japan
| | - Takahiro Okumura
- Division of Cardiology, Amagasaki Chuo Hospital, 1-12-1 Shioe, Amagasaki, Hyogo, 661-0976, Japan
| | - Jun Tateishi
- Division of Cardiology, Amagasaki Chuo Hospital, 1-12-1 Shioe, Amagasaki, Hyogo, 661-0976, Japan
| | - Junichi Yoshida
- Division of Cardiology, Amagasaki Chuo Hospital, 1-12-1 Shioe, Amagasaki, Hyogo, 661-0976, Japan
| | - Shunsuke Tamaki
- Division of Cardiology, Osaka General Medical Center, 3-1-56 Mandaihigashi, Sumiyoshi-ku, Osaka, 558-8558, Japan
| | - Masamichi Yano
- Division of Cardiology, Osaka Rosai Hospital, 3-1179 Nagasonecho, Kita-ku, Sakai, 591-8025, Japan
| | - Takaharu Hayashi
- Cardiovascular Division, Osaka Police Hospital, 10-31 Kitayamacho, Tennojiku, Osaka, 543-0035, Japan
| | - Yusuke Nakagawa
- Division of Cardiology, Kawanishi City Hospital, 5-21-1, Kawanishi, Hyogo, 666-0195, Japan
| | - Takahisa Yamada
- Division of Cardiology, Osaka General Medical Center, 3-1-56 Mandaihigashi, Sumiyoshi-ku, Osaka, 558-8558, Japan
| | - Daisaku Nakatani
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, 565-0871, Japan
| | - Shungo Hikoso
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, 565-0871, Japan
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, 565-0871, Japan
| | | |
Collapse
|
21
|
Zandawala M, Nguyen T, Balanyà Segura M, Johard HAD, Amcoff M, Wegener C, Paluzzi JP, Nässel DR. A neuroendocrine pathway modulating osmotic stress in Drosophila. PLoS Genet 2021; 17:e1009425. [PMID: 33684132 PMCID: PMC7971876 DOI: 10.1371/journal.pgen.1009425] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 03/18/2021] [Accepted: 02/15/2021] [Indexed: 12/19/2022] Open
Abstract
Environmental factors challenge the physiological homeostasis in animals, thereby evoking stress responses. Various mechanisms have evolved to counter stress at the organism level, including regulation by neuropeptides. In recent years, much progress has been made on the mechanisms and neuropeptides that regulate responses to metabolic/nutritional stress, as well as those involved in countering osmotic and ionic stresses. Here, we identified a peptidergic pathway that links these types of regulatory functions. We uncover the neuropeptide Corazonin (Crz), previously implicated in responses to metabolic stress, as a neuroendocrine factor that inhibits the release of a diuretic hormone, CAPA, and thereby modulates the tolerance to osmotic and ionic stress. Both knockdown of Crz and acute injections of Crz peptide impact desiccation tolerance and recovery from chill-coma. Mapping of the Crz receptor (CrzR) expression identified three pairs of Capa-expressing neurons (Va neurons) in the ventral nerve cord that mediate these effects of Crz. We show that Crz acts to restore water/ion homeostasis by inhibiting release of CAPA neuropeptides via inhibition of cAMP production in Va neurons. Knockdown of CrzR in Va neurons affects CAPA signaling, and consequently increases tolerance for desiccation, ionic stress and starvation, but delays chill-coma recovery. Optogenetic activation of Va neurons stimulates excretion and simultaneous activation of Crz and CAPA-expressing neurons reduces this response, supporting the inhibitory action of Crz. Thus, Crz inhibits Va neurons to maintain osmotic and ionic homeostasis, which in turn affects stress tolerance. Earlier work demonstrated that systemic Crz signaling restores nutrient levels by promoting food search and feeding. Here we additionally propose that Crz signaling also ensures osmotic homeostasis by inhibiting release of CAPA neuropeptides and suppressing diuresis. Thus, Crz ameliorates stress-associated physiology through systemic modulation of both peptidergic neurosecretory cells and the fat body in Drosophila.
Collapse
Affiliation(s)
- Meet Zandawala
- Department of Zoology, Stockholm University, Stockholm, Sweden
| | - Thomas Nguyen
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Marta Balanyà Segura
- Neurobiology and Genetics, Würzburg Insect Research (WIR), Theodor-Boveri-Institute, Biocenter, University of Würzburg, Germany
| | | | - Mirjam Amcoff
- Department of Zoology, Stockholm University, Stockholm, Sweden
| | - Christian Wegener
- Neurobiology and Genetics, Würzburg Insect Research (WIR), Theodor-Boveri-Institute, Biocenter, University of Würzburg, Germany
| | | | - Dick R Nässel
- Department of Zoology, Stockholm University, Stockholm, Sweden
| |
Collapse
|
22
|
Dharmayanti C, Gillam TA, Klingler-Hoffmann M, Albrecht H, Blencowe A. Strategies for the Development of pH-Responsive Synthetic Polypeptides and Polymer-Peptide Hybrids: Recent Advancements. Polymers (Basel) 2021; 13:624. [PMID: 33669548 PMCID: PMC7921987 DOI: 10.3390/polym13040624] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 02/11/2021] [Accepted: 02/15/2021] [Indexed: 12/11/2022] Open
Abstract
Synthetic polypeptides and polymer-peptide hybrid materials have been successfully implemented in an array of biomedical applications owing to their biocompatibility, biodegradability and ability to mimic natural proteins. In addition, these materials have the capacity to form complex supramolecular structures, facilitate specific biological interactions, and incorporate a diverse selection of functional groups that can be used as the basis for further synthetic modification. Like conventional synthetic polymers, polypeptide-based materials can be designed to respond to external stimuli (e.g., light and temperature) or changes in the environmental conditions (e.g., redox reactions and pH). In particular, pH-responsive polypeptide-based systems represent an interesting avenue for the preparation of novel drug delivery systems that can exploit physiological or pathological pH variations within the body, such as those that arise in the extracellular tumour microenvironment, intracellularly within endosomes/lysosomes, or during tissue inflammation. Here, we review the significant progress made in advancing pH-responsive polypeptides and polymer-peptide hybrid materials during the last five years, with a particular emphasis on the manipulation of ionisable functional groups, pH-labile linkages, pH-sensitive changes to secondary structure, and supramolecular interactions.
Collapse
Affiliation(s)
- Cintya Dharmayanti
- Applied Chemistry and Translational Biomaterials Group, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (C.D.); (T.A.G.)
| | - Todd A. Gillam
- Applied Chemistry and Translational Biomaterials Group, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (C.D.); (T.A.G.)
- Surface Interactions and Soft Matter Group, Future Industries Institute, University of South Australia, Mawson Lakes, SA 5095, Australia
| | | | - Hugo Albrecht
- Drug Discovery and Development Group, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia;
| | - Anton Blencowe
- Applied Chemistry and Translational Biomaterials Group, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (C.D.); (T.A.G.)
| |
Collapse
|
23
|
Hoffiz YC, Castillo-Ruiz A, Hall MAL, Hite TA, Gray JM, Cisternas CD, Cortes LR, Jacobs AJ, Forger NG. Birth elicits a conserved neuroendocrine response with implications for perinatal osmoregulation and neuronal cell death. Sci Rep 2021; 11:2335. [PMID: 33504846 PMCID: PMC7840942 DOI: 10.1038/s41598-021-81511-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/05/2021] [Indexed: 12/17/2022] Open
Abstract
Long-standing clinical findings report a dramatic surge of vasopressin in umbilical cord blood of the human neonate, but the neural underpinnings and function(s) of this phenomenon remain obscure. We studied neural activation in perinatal mice and rats, and found that birth triggers activation of the suprachiasmatic, supraoptic, and paraventricular nuclei of the hypothalamus. This was seen whether mice were born vaginally or via Cesarean section (C-section), and when birth timing was experimentally manipulated. Neuronal phenotyping showed that the activated neurons were predominantly vasopressinergic, and vasopressin mRNA increased fivefold in the hypothalamus during the 2–3 days before birth. Copeptin, a surrogate marker of vasopressin, was elevated 30-to 50-fold in plasma of perinatal mice, with higher levels after a vaginal than a C-section birth. We also found an acute decrease in plasma osmolality after a vaginal, but not C-section birth, suggesting that the difference in vasopressin release between birth modes is functionally meaningful. When vasopressin was administered centrally to newborns, we found an ~ 50% reduction in neuronal cell death in specific brain areas. Collectively, our results identify a conserved neuroendocrine response to birth that is sensitive to birth mode, and influences peripheral physiology and neurodevelopment.
Collapse
Affiliation(s)
- Yarely C Hoffiz
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA
| | | | - Megan A L Hall
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA
| | - Taylor A Hite
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA
| | - Jennifer M Gray
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA
| | - Carla D Cisternas
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA.,Instituto de Investigación Médica M Y M Ferreyra, INIMEC-CONICET-UNC, Córdoba, Argentina
| | - Laura R Cortes
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA
| | - Andrew J Jacobs
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA
| | - Nancy G Forger
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA.
| |
Collapse
|
24
|
Castle-Kirszbaum M, Kyi M, Wright C, Goldschlager T, Danks RA, Parkin WG. Hyponatraemia and hypernatraemia: Disorders of Water Balance in Neurosurgery. Neurosurg Rev 2021; 44:2433-2458. [PMID: 33389341 DOI: 10.1007/s10143-020-01450-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/26/2020] [Accepted: 11/25/2020] [Indexed: 12/23/2022]
Abstract
Disorders of tonicity, hyponatraemia and hypernatraemia, are common in neurosurgical patients. Tonicity is sensed by the circumventricular organs while the volume state is sensed by the kidney and peripheral baroreceptors; these two signals are integrated in the hypothalamus. Volume is maintained through the renin-angiotensin-aldosterone axis, while tonicity is defended by arginine vasopressin (antidiuretic hormone) and the thirst response. Edelman found that plasma sodium is dependent on the exchangeable sodium, potassium and free-water in the body. Thus, changes in tonicity must be due to disproportionate flux of these species in and out of the body. Sodium concentration may be measured by flame photometry and indirect, or direct, ion-sensitive electrodes. Only the latter method is not affected by changes in plasma composition. Classification of hyponatraemia by the volume state is imprecise. We compare the tonicity of the urine, given by the sodium potassium sum, to that of the plasma to determine the renal response to the dysnatraemia. We may then assess the activity of the renin-angiotensin-aldosterone axis using urinary sodium and fractional excretion of sodium, urate or urea. Together, with clinical context, these help us determine the aetiology of the dysnatraemia. Symptomatic individuals and those with intracranial catastrophes require prompt treatment and vigilant monitoring. Otherwise, in the absence of hypovolaemia, free-water restriction and correction of any reversible causes should be the mainstay of treatment for hyponatraemia. Hypernatraemia should be corrected with free-water, and concurrent disorders of volume should be addressed. Monitoring for overcorrection of hyponatraemia is necessary to avoid osmotic demyelination.
Collapse
Affiliation(s)
| | - Mervyn Kyi
- Department of Endocrinology, Melbourne Health, Melbourne, Australia
| | - Christopher Wright
- Department of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
| | - Tony Goldschlager
- Department of Neurosurgery, Monash Health, Melbourne, Australia.,Department of Surgery, Monash University, Melbourne, Australia
| | - R Andrew Danks
- Department of Neurosurgery, Monash Health, Melbourne, Australia.,Department of Surgery, Monash University, Melbourne, Australia
| | - W Geoffrey Parkin
- Department of Surgery, Monash University, Melbourne, Australia.,Department of Intensive Care, Monash Health, Melbourne, Australia
| |
Collapse
|
25
|
Giersch GEW, Colburn AT, Morrissey MC, Butler CR, Pruchnicki ML, Kavouras SA, Charkoudian N, Casa DJ. Effects of sex and menstrual cycle on volume-regulatory responses to 24-h fluid restriction. Am J Physiol Regul Integr Comp Physiol 2020; 319:R560-R565. [PMID: 32936680 DOI: 10.1152/ajpregu.00173.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Reproductive hormones have significant nonreproductive physiological effects, including altering fluid regulation. Our purpose was to explore the impact of sex and menstrual cycle (MC) phase on volume-regulatory responses to 24-h fluid restriction (24-h FR). Participants (men: n = 12, 20 ± 2 yr; women: n = 10, 20 ± 1 yr) were assigned two randomized and counterbalanced fluid prescriptions [Euhy: euhydrated, urine specific gravity (USG) < 1.020; Dehy: 24-h FR, USG > 1.020]. Men completed both (MEuhy, MDehy), while women completed both in the late-follicular (days 10-13; FDehy, FEuhy) and midluteal (days 18-22; LDehy, LEuhy) phases. We measured body mass, plasma and urine osmolality (Posm, Uosm), urine specific gravity (USG), urine color (Ucol), and serum copeptin; 24-h FR yielded mild dehydration without influence of sex or MC (P > 0.05). Copeptin increased in men following Dehy (pre: 8.2 ± 5.2, post: 15.8 ± 12.6, P = 0.04) but not in women (FDehy pre: 4.3 ± 1.6, post: 10.5 ± 6.9, P = 0.06; LDehy pre: 5.6 ± 3.5, post: 10.4 ± 6.2, P = 0.16). In FDehy, Posm increased following FR (pre: 288 ± 2, post: 292 ± 1, P = 0.03) but not in men (pre: 292 ± 3, post: 293 ± 2, P = 0.46). No MC differences were observed between body mass loss, Posm, Uosm, USG, and copeptin (P > 0.05). These results suggest that volume-regulatory responses to 24-h FR were present in men but not in women, without apparent effects of the menstrual cycle.
Collapse
Affiliation(s)
- Gabrielle E W Giersch
- United States Army Research Institute for Environmental Medicine, Natick, Massachusetts.,Korey Stringer Institute, Department of Kinesiology, University of Connecticut, Storrs, Connecticut
| | - Abigail T Colburn
- Arizona State University, Hydration Science Laboratory, Phoenix, Arizona
| | - Margaret C Morrissey
- Korey Stringer Institute, Department of Kinesiology, University of Connecticut, Storrs, Connecticut
| | - Cody R Butler
- Korey Stringer Institute, Department of Kinesiology, University of Connecticut, Storrs, Connecticut
| | - Michaela L Pruchnicki
- Korey Stringer Institute, Department of Kinesiology, University of Connecticut, Storrs, Connecticut
| | - Stavros A Kavouras
- Arizona State University, Hydration Science Laboratory, Phoenix, Arizona
| | - Nisha Charkoudian
- United States Army Research Institute for Environmental Medicine, Natick, Massachusetts
| | - Douglas J Casa
- Korey Stringer Institute, Department of Kinesiology, University of Connecticut, Storrs, Connecticut
| |
Collapse
|
26
|
Abstract
The two main differential diagnoses of central diabetes insipidus are nephrogenic diabetes insipidus and primary polydipsia. Reliable distinction between those entities is essential as treatment differs substantially with the wrong treatment potentially leading to serious complications. Past diagnostic measures using the indirect water deprivation test had several pitfalls, resulting in a low diagnostic accuracy. With the introduction of copeptin, a stable and reliable surrogate marker for arginine vasopressin, diagnosis of diabetes insipidus was new evaluated. While unstimulated basal copeptin measurement reliably diagnoses nephrogenic diabetes insipidus, a stimulation test is needed to differentiate patients with central diabetes insipidus from patients with primary polydipsia. Stimulation can either be achieved through hypertonic saline infusion or arginine infusion. While the former showed high diagnostic accuracy and superiority over the indirect water deprivation test in a recent validation study, the diagnostic accuracy for arginine-stimulated copeptin was slightly lower, but superior in test tolerance. In summary of the recent findings, a new copeptin based diagnostic algorithm is proposed for the reliable diagnosis of diabetes insipidus.
Collapse
Affiliation(s)
- Julie Refardt
- Departments of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Switzerland; University of Basel, Basel, Switzerland.
| |
Collapse
|
27
|
Abstract
The differential diagnosis of diabetes insipidus involves the distinction between central or nephrogenic diabetes insipidus and primary polydipsia. Differentiation is important because treatment strategies vary; the wrong treatment can be dangerous. Reliable differentiation is difficult especially in patients with primary polydipsia or partial forms of diabetes insipidus. New diagnostic algorithms are based on the measurement of copeptin after osmotic stimulation by hypertonic saline infusion or after nonosmotic stimulation by arginine and have a higher diagnostic accuracy than the water deprivation test. Treatment involves correcting preexisting water deficits, but is different for central diabetes insipidus, nephrogenic diabetes insipidus, and primary polydipsia.
Collapse
Affiliation(s)
- Julie Refardt
- Division of Endocrinology, Diabetes and Metabolism, Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland; Department of Endocrinology, University Hospital Basel, University of Basel, Petersgraben 4, Basel 4031, Switzerland
| | - Bettina Winzeler
- Division of Endocrinology, Diabetes and Metabolism, Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland; Department of Endocrinology, University Hospital Basel, University of Basel, Petersgraben 4, Basel 4031, Switzerland
| | - Mirjam Christ-Crain
- Division of Endocrinology, Diabetes and Metabolism, Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland; Department of Endocrinology, University Hospital Basel, University of Basel, Petersgraben 4, Basel 4031, Switzerland.
| |
Collapse
|
28
|
Takagi H, Hagiwara D, Handa T, Sugiyama M, Onoue T, Tsunekawa T, Ito Y, Iwama S, Goto M, Suga H, Banno R, Takahashi K, Matsui S, Arima H. Diagnosis of central diabetes insipidus using a vasopressin radioimmunoassay during hypertonic saline infusion. Endocr J 2020; 67:267-274. [PMID: 31748430 DOI: 10.1507/endocrj.ej19-0224] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Central diabetes insipidus (CDI) is characterized by polyuria and polydipsia caused by impairment of arginine vasopressin (AVP) secretion. In this study, we evaluated plasma AVP concentrations during a hypertonic saline infusion test using a new AVP radioimmunoassay (RIA) which is now available in Japan. Thirteen control subjects, mostly with hypothalamo-pituitary disease but without CDI, and 13 patients with CDI were enrolled in the study. Whether or not subjects had CDI was determined based on the totality of clinical data, which included urine volumes and osmolality. Regression analysis of plasma AVP and serum Na concentrations revealed that the gradient was significantly lower in the CDI group than in the control group. The area under the receiver-operating-characteristic (ROC) curve was 0.99, and the <0.1 gradient cut-off values for the simple regression line to distinguish CDI from control had a 100% sensitivity and a 77% specificity. The ROC analysis with estimated plasma AVP concentrations at a serum Na concentration of 149 mEq/L showed that the area under the ROC curve was 1.0 and the <1.0 pg/mL cut-off values of plasma AVP had a 99% sensitivity and a 95% specificity. We conclude that measurement of AVP by RIA during a hypertonic saline infusion test can differentiate patients with CDI from those without CDI with a high degree of accuracy. Further investigation is required to confirm whether the cut-off values shown in this study are also applicable to a diagnosis of partial CDI or a differential diagnosis between CDI and primary polydipsia.
Collapse
Affiliation(s)
- Hiroshi Takagi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Daisuke Hagiwara
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Tomoko Handa
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Mariko Sugiyama
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Takeshi Onoue
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Taku Tsunekawa
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Yoshihiro Ito
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Shintaro Iwama
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Motomitsu Goto
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Hidetaka Suga
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Ryoichi Banno
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Kunihiko Takahashi
- Department of Biological Statistics, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Shigeyuki Matsui
- Department of Biological Statistics, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Hiroshi Arima
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
29
|
Underhydration Is Associated with Obesity, Chronic Diseases, and Death Within 3 to 6 Years in the U.S. Population Aged 51-70 Years. Nutrients 2020; 12:nu12040905. [PMID: 32224908 PMCID: PMC7230456 DOI: 10.3390/nu12040905] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/23/2020] [Accepted: 03/23/2020] [Indexed: 12/14/2022] Open
Abstract
Nationally representative data from the National Health and Nutrition Examination Survey (NHANES) indicate that over 65% of adults aged 51–70 years in the U.S. do not meet hydration criteria. They have hyponatremia (serum sodium < 135 mmol/L) and/or underhydration (serum sodium >145 mmol/L, spot urine volume <50 mL, and/or spot urine osmolality ≥500 mmol/kg). To explore potential public health implications of not meeting hydration criteria, data from the NHANES 2009–2012 and National Center for Health Statistics Linked Mortality Files for fasting adults aged 51–70 years (sample n = 1200) were used to determine if hyponatremia and/or underhydration were cross-sectionally associated with chronic health conditions and/or longitudinally associated with chronic disease mortality. Underhydration accounted for 97% of the population group not meeting hydration criteria. In weighted multivariable adjusted Poisson models, underhydration was significantly associated with increased prevalence of obesity, high waist circumference, insulin resistance, diabetes, low HDL, hypertension, and metabolic syndrome. Over 3–6 years of follow-up, 33 chronic disease deaths occurred in the sample, representing an estimated 1,084,144 deaths in the U.S. Alongside chronic health conditions, underhydration was a risk factor for an estimated 863,305 deaths. Independent of the chronic health conditions evaluated, underhydration was a risk factor for 128,107 deaths. In weighted multivariable Cox models, underhydration was associated with 4.21 times greater chronic disease mortality (95% CI: 1.29–13.78, p = 0.019). Zero chronic disease deaths were observed for people who met the hydration criteria and did not already have a chronic condition in 2009–2012. Further work should consider effects of underhydration on population health.
Collapse
|
30
|
Armstrong LE, Muñoz CX, Armstrong EM. Distinguishing Low and High Water Consumers-A Paradigm of Disease Risk. Nutrients 2020; 12:E858. [PMID: 32210168 PMCID: PMC7146321 DOI: 10.3390/nu12030858] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/18/2020] [Accepted: 03/20/2020] [Indexed: 12/31/2022] Open
Abstract
A long-standing body of clinical observations associates low 24-h total water intake (TWI = water + beverages + food moisture) with acute renal disorders such as kidney stones and urinary tract infections. These findings prompted observational studies and experimental interventions comparing habitual low volume (LOW) and high volume (HIGH) drinkers. Investigators have learned that the TWI of LOW and HIGH differ by 1-2 L·d-1, their hematological values (e.g., plasma osmolality, plasma sodium) are similar and lie within the laboratory reference ranges of healthy adults and both groups appear to successfully maintain water-electrolyte homeostasis. However, LOW differs from HIGH in urinary biomarkers (e.g., reduced urine volume and increased osmolality or specific gravity), as well as higher plasma concentrations of arginine vasopressin (AVP) and cortisol. Further, evidence suggests that both a low daily TWI and/or elevated plasma AVP influence the development and progression of metabolic syndrome, diabetes, obesity, chronic kidney disease, hypertension and cardiovascular disease. Based on these studies, we propose a theory of increased disease risk in LOW that involves chronic release of fluid-electrolyte (i.e., AVP) and stress (i.e., cortisol) hormones. This narrative review describes small but important differences between LOW and HIGH, advises future investigations and provides practical dietary recommendations for LOW that are intended to decrease their risk of chronic diseases.
Collapse
Affiliation(s)
- Lawrence E. Armstrong
- Professor Emeritus, Human Performance Laboratory and Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Colleen X. Muñoz
- Assistant Professor, Department of Health Sciences, University of Hartford, West Hartford, CT 06117, USA;
| | | |
Collapse
|
31
|
Abstract
Activation of arginine–vasopressin is one of the hormonal responses to face vasodilation-related hypotension. Released from the post-pituitary gland, vasopressin induces vasoconstriction through the activation of V1a receptors located on vascular smooth muscle cells. Due to its non-selective receptor affinity arginine–vasopressin also activates V2 (located on renal tubular cells of collecting ducts) and V1b (located in the anterior pituitary and in the pancreas) receptors, thereby potentially promoting undesired side effects such as anti-diuresis, procoagulant properties due to release of the von Willebrand’s factor and platelet activation. Finally, it also cross-activates oxytocin receptors. During septic shock, vasopressin plasma levels were reported to be lower than expected, and a hypersensitivity to its vasopressor effect is reported in such situation. Terlipressin and selepressin are synthetic vasopressin analogues with a higher affinity for the V1 receptor, and, hence, potentially less side effects. In this narrative review, we present the current knowledge of the rationale, benefits and risks of vasopressin use in the setting of septic shock and vasoplegic shock following cardiac surgery. Clearly, vasopressin administration allows reducing norepinephrine requirements, but so far, no improvement of survival was reported and side effects are frequent, particularly ischaemic events. Finally, we will discuss the current indications for vasopressin and its agonists in the setting of septic shock, and the remaining unresolved questions.
Collapse
|
32
|
Christ-Crain M, Fenske WK. Copeptin in the differential diagnosis of hypotonic polyuria. J Endocrinol Invest 2020; 43:21-30. [PMID: 31368050 DOI: 10.1007/s40618-019-01087-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/17/2019] [Indexed: 11/26/2022]
Abstract
COPEPTIN Copeptin is secreted in equimolar amount to Arginine Vasopressin (AVP) but can easily be measured with a sandwich immunoassay. Both peptides, copeptin and AVP, show a high correlation. Accordingly, copeptin mirrors the amount of AVP in the circulation and its measurement provides an attractive marker in the differential diagnosis of diabetes insipidus. THE POLYURIA POLYDIPSIA SYNDROME Diabetes insipidus-either central or nephrogenic-has to be differentiated from primary polydipsia. Differentiation is crucial since wrong treatment can have deleterious consequences. Since many decades, the "gold standard" for differential diagnosis has been the classical water deprivation test, which has several limitations leading to an overall limited diagnostic accuracy. In addition, the test has a long duration of 17 hours and is cumbersome for patients. Clinical signs and symptoms as well as MRI characteristics overlap between patients with diabetes insipidus and primary polydipsia. Direct measurement of AVP upon osmotic stimulation was first shown to overcome these limitations, but failed to enter clinical practice mainly due to technical limitations of the AVP assay. COPEPTIN AS DIAGNOSTIC TOOL IN THE POLYURIA POLYDIPSIA SYNDROME We have recently shown that copeptin, without prior water deprivation, identifies patients with nephrogenic diabetes insipidus. On the other hand, for the more difficult differentiation between central diabetes insipidus and primary polydipsia, a copeptin level of 4.9 pmol/L stimulated with hypertonic saline infusion differentiates between these two entities with a high diagnostic accuracy, and is superior to the water deprivation test. It is important to note that close sodium monitoring during the hypertonic saline test is a prerequisite. CONCLUSION Therefore, we propose that copeptin upon hypertonic saline infusion should become the new standard test in the differential diagnosis of diabetes insipidus.
Collapse
Affiliation(s)
- M Christ-Crain
- Department of Endocrinology, Diabetology and Metabolism, University Hospital of Basel, University of Basel, Petersgraben 4, 4031, Basel, Switzerland.
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, University Hospital Leipzig, Leipzig, Germany.
| | - W K Fenske
- Department of Endocrinology, Diabetology and Metabolism, University Hospital of Basel, University of Basel, Petersgraben 4, 4031, Basel, Switzerland.
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, University Hospital Leipzig, Leipzig, Germany.
| |
Collapse
|
33
|
Christ-Crain M. Diabetes Insipidus: New Concepts for Diagnosis. Neuroendocrinology 2020; 110:859-867. [PMID: 31986514 DOI: 10.1159/000505548] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 12/18/2019] [Indexed: 11/19/2022]
Abstract
Diabetes insipidus (DI), be it from central or from nephrogenic origin, has to be differentiated from primary polydipsia. This differentiation is crucial since wrong treatment can have dangerous consequences. For decades, the "gold standard" for differential diagnosis has been the standard water deprivation test. However, this test has several limitations leading to an overall limited diagnostic accuracy. In addition, the test has a long duration of 17 h and is cumbersome for patients. Also clinical signs and symptoms and MRI characteristics overlap between patients with DI and primary polydipsia. Direct measurement of arginine vasopressin (AVP) upon osmotic stimulation was first shown to overcome these limitations, but failed to enter clinical practice mainly due to technical limitations of the AVP assay. Copeptin is secreted in equimolar ratio to AVP, mirroring AVP concentrations in the circulation. We have shown that copeptin, without prior fluid deprivation, identifies patients with nephrogenic DI. For the more difficult differentiation between central DI and primary polydipsia, a copeptin level of 4.9 pmol/L stimulated with hypertonic saline infusion differentiates between these 2 entities with a high diagnostic accuracy and is superior to the water deprivation test. However, it is important to note that close and regular sodium monitoring every 30 min during the hypertonic saline test is a prerequisite, which is not possible in all hospitals. Furthermore, side effects are common. Therefore, a nonosmotic stimulation test would be advantageous. Arginine significantly stimulates copeptin and therefore is a novel, so far unknown stimulus of this peptide. Consequently, infusion of arginine with subsequent copeptin measurement was shown to be an even simpler and better tolerated test, but head to head comparison is still lacking.
Collapse
Affiliation(s)
- Mirjam Christ-Crain
- Department of Endocrinology, Diabetology and Metabolism University Hospital Basel, Basel, Switzerland,
- Department of Clinical Research, University of Basel, Basel, Switzerland,
| |
Collapse
|
34
|
Kanbay M, Yilmaz S, Dincer N, Ortiz A, Sag AA, Covic A, Sánchez-Lozada LG, Lanaspa MA, Cherney DZI, Johnson RJ, Afsar B. Antidiuretic Hormone and Serum Osmolarity Physiology and Related Outcomes: What Is Old, What Is New, and What Is Unknown? J Clin Endocrinol Metab 2019; 104:5406-5420. [PMID: 31365096 DOI: 10.1210/jc.2019-01049] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/25/2019] [Indexed: 12/16/2022]
Abstract
CONTEXT Although the physiology of sodium, water, and arginine vasopressin (AVP), also known as antidiuretic hormone, has long been known, accumulating data suggest that this system operates as a more complex network than previously thought. EVIDENCE ACQUISITION English-language basic science and clinical studies of AVP and osmolarity on the development of kidney and cardiovascular disease and overall outcomes. EVIDENCE SYNTHESIS Apart from osmoreceptors and hypovolemia, AVP secretion is modified by novel factors such as tongue acid-sensing taste receptor cells and brain median preoptic nucleus neurons. Moreover, pharyngeal, esophageal, and/or gastric sensors and gut microbiota modulate AVP secretion. Evidence is accumulating that increased osmolarity, AVP, copeptin, and dehydration are all associated with worse outcomes in chronic disease states such as chronic kidney disease (CKD), diabetes, and heart failure. On the basis of these pathophysiological relationships, an AVP receptor 2 blocker is now licensed for CKD related to polycystic kidney disease. CONCLUSION From a therapeutic perspective, fluid intake may be associated with increased AVP secretion if it is driven by loss of urine concentration capacity or with suppressed AVP if it is driven by voluntary fluid intake. In the current review, we summarize the literature on the relationship between elevated osmolarity, AVP, copeptin, and dehydration with renal and cardiovascular outcomes and underlying classical and novel pathophysiologic pathways. We also review recent unexpected and contrasting findings regarding AVP physiology in an attempt to explain and understand some of these relationships.
Collapse
Affiliation(s)
- Mehmet Kanbay
- Division of Nephrology, Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Sezen Yilmaz
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Neris Dincer
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Alberto Ortiz
- Dialysis Unit, School of Medicine, IIS-Fundacion Jimenez Diaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Alan A Sag
- Division of Vascular and Interventional Radiology, Department of Radiology, Duke University Medical Center, Durham, North Carolina
| | - Adrian Covic
- Nephrology Department, Dialysis and Renal Transplant Center, "Dr. C. I. Parhon" University Hospital, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania
| | - Laura G Sánchez-Lozada
- Laboratory of Renal Physiopathology, Department of Nephrology, INC Ignacio Chávez, Mexico City, Mexico
| | - Miguel A Lanaspa
- Division of Renal Diseases and Hypertension, School of Medicine, University of Colorado Denver, Aurora, Colorado
| | - David Z I Cherney
- Department of Medicine, Division of Nephrology, Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Richard J Johnson
- Division of Renal Diseases and Hypertension, School of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Baris Afsar
- Division of Nephrology, Department of Medicine, Suleyman Demirel University School of Medicine, Isparta, Turkey
| |
Collapse
|
35
|
Allen MD, Springer DA, Burg MB, Boehm M, Dmitrieva NI. Suboptimal hydration remodels metabolism, promotes degenerative diseases, and shortens life. JCI Insight 2019; 4:130949. [PMID: 31484829 PMCID: PMC6777918 DOI: 10.1172/jci.insight.130949] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/06/2019] [Indexed: 01/30/2023] Open
Abstract
With increased life expectancy worldwide, there is an urgent need for improving preventive measures that delay the development of age-related degenerative diseases. Here, we report evidence from mouse and human studies that this goal can be achieved by maintaining optimal hydration throughout life. We demonstrate that restricting the amount of drinking water shortens mouse lifespan with no major warning signs up to 14 months of life, followed by sharp deterioration. Mechanistically, water restriction yields stable metabolism remodeling toward metabolic water production with greater food intake and energy expenditure, an elevation of markers of inflammation and coagulation, accelerated decline of neuromuscular coordination, renal glomerular injury, and the development of cardiac fibrosis. In humans, analysis of data from the Atherosclerosis Risk in Communities (ARIC) study revealed that hydration level, assessed at middle age by serum sodium concentration, is associated with markers of coagulation and inflammation and predicts the development of many age-related degenerative diseases 24 years later. The analysis estimates that improving hydration throughout life may greatly decrease the prevalence of degenerative diseases, with the most profound effect on dementia, heart failure (HF), and chronic lung disease (CLD), translating to the development of these diseases in 3 million fewer people in the United States alone. In mice, restricting the amount of drinking water shortens lifespan and may accelerate degenerative changes leading to age-related chronic diseases.
Collapse
Affiliation(s)
| | | | | | - Manfred Boehm
- Laboratory of Cardiovascular Regenerative Medicine, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Natalia I Dmitrieva
- Renal Cellular and Molecular Biology Section, and.,Laboratory of Cardiovascular Regenerative Medicine, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| |
Collapse
|
36
|
Abstract
Diabetes insipidus (DI) is a disorder characterized by excretion of large amounts of hypotonic urine. Central DI results from a deficiency of the hormone arginine vasopressin (AVP) in the pituitary gland or the hypothalamus, whereas nephrogenic DI results from resistance to AVP in the kidneys. Central and nephrogenic DI are usually acquired, but genetic causes must be evaluated, especially if symptoms occur in early childhood. Central or nephrogenic DI must be differentiated from primary polydipsia, which involves excessive intake of large amounts of water despite normal AVP secretion and action. Primary polydipsia is most common in psychiatric patients and health enthusiasts but the polydipsia in a small subgroup of patients seems to be due to an abnormally low thirst threshold, a condition termed dipsogenic DI. Distinguishing between the different types of DI can be challenging and is done either by a water deprivation test or by hypertonic saline stimulation together with copeptin (or AVP) measurement. Furthermore, a detailed medical history, physical examination and imaging studies are needed to ensure an accurate DI diagnosis. Treatment of DI or primary polydipsia depends on the underlying aetiology and differs in central DI, nephrogenic DI and primary polydipsia.
Collapse
|
37
|
Kim YB, Colwell CS, Kim YI. Long-term ionic plasticity of GABAergic signalling in the hypothalamus. J Neuroendocrinol 2019; 31:e12753. [PMID: 31166034 DOI: 10.1111/jne.12753] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/24/2019] [Accepted: 06/03/2019] [Indexed: 12/23/2022]
Abstract
The hypothalamus contains a number of nuclei that subserve a variety of functions, including generation of circadian rhythms, regulation of hormone secretion and maintenance of homeostatic levels for a variety of physiological parameters. Within the hypothalamus, γ-amino-butyric acid (GABA) is one of the major neurotransmitters responsible for cellular communication. Although GABA most commonly serves as an inhibitory neurotransmitter, a growing body of evidence indicates that it can evoke post-synaptic excitation as a result of the active regulation of intracellular chloride concentration. In this review, we consider the evidence for this ionic plasticity of GABAergic synaptic transmission in five distinct cases in hypothalamic cell populations. We argue that this plasticity serves as part of the functional response to or is at least associated with dehydration, lactation, hypertension and stress. As such, GABA excitation should be considered as part of the core homeostatic mechanisms of the hypothalamus.
Collapse
Affiliation(s)
- Young-Beom Kim
- Department of Physiology, Korea University College of Medicine, Seoul, Korea
- Neuroscience Research Institute, Korea University, Seoul, Korea
| | - Christopher S Colwell
- Department of Psychiatry and Biobehavioral Sciences, UCLA School of Medicine, Los Angeles, CA, USA
| | - Yang In Kim
- Department of Physiology, Korea University College of Medicine, Seoul, Korea
- Neuroscience Research Institute, Korea University, Seoul, Korea
| |
Collapse
|
38
|
Refardt J, Winzeler B, Christ-Crain M. Copeptin and its role in the diagnosis of diabetes insipidus and the syndrome of inappropriate antidiuresis. Clin Endocrinol (Oxf) 2019; 91:22-32. [PMID: 31004513 PMCID: PMC6850413 DOI: 10.1111/cen.13991] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 04/08/2019] [Accepted: 04/15/2019] [Indexed: 01/14/2023]
Abstract
Copeptin is secreted in an equimolar amount to arginine vasopressin (AVP) but can easily be measured in plasma or serum with a sandwich immunoassay. The main stimuli for copeptin are similar to AVP, that is an increase in osmolality and a decrease in arterial blood volume and pressure. A high correlation between copeptin and AVP has been shown. Accordingly, copeptin mirrors the amount of AVP in the circulation. Copeptin has, therefore, been evaluated as diagnostic biomarker in vasopressin-dependent disorders of body fluid homeostasis. Disorders of body fluid homeostasis are common and can be divided into hyper- and hypoosmolar circumstances: the classical hyperosmolar disorder is diabetes insipidus, while the most common hypoosmolar disorder is the syndrome of inappropriate antidiuresis (SIAD). Copeptin measurement has led to a "revival" of the direct test in the differential diagnosis of diabetes insipidus. Baseline copeptin levels, without prior thirsting, unequivocally identify patients with nephrogenic diabetes insipidus. In contrast, for the difficult differentiation between central diabetes insipidus and primary polydipsia, a stimulated copeptin level of 4.9 pmol/L upon hypertonic saline infusion differentiates these two entities with a high diagnostic accuracy and is clearly superior to the classical water deprivation test. On the contrary, in the SIAD, copeptin measurement is of only little diagnostic value. Copeptin levels widely overlap in patients with hyponatraemia and emphasize the heterogeneity of the disease. Additionally, a variety of factors lead to unspecific copeptin elevations in the acute setting further complicating its interpretation. The broad use of copeptin as diagnostic marker in hyponatraemia and specifically to detect cancer-related disease in SIADH patients can, therefore, not be recommended.
Collapse
Affiliation(s)
- Julie Refardt
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Bettina Winzeler
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Mirjam Christ-Crain
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel, Switzerland
- University of Basel, Basel, Switzerland
| |
Collapse
|
39
|
Christ-Crain M. EJE AWARD 2019: New diagnostic approaches for patients with polyuria polydipsia syndrome. Eur J Endocrinol 2019; 181:R11-R21. [PMID: 31067508 PMCID: PMC6598864 DOI: 10.1530/eje-19-0163] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/08/2019] [Indexed: 02/02/2023]
Abstract
Diabetes insipidus (DI), be it from central or nephrogenic origin, must be differentiated from secondary forms of hypotonic polyuria such as primary polydipsia. Differentiation is crucial since wrong treatment can have deleterious consequences. Since decades, the gold standard for differentiation has been the water deprivation test, which has limitations leading to an overall unsatisfying diagnostic accuracy. Furthermore, it is cumbersome for patients with a long test duration. Clinical signs and symptoms and MRI characteristics overlap between patients with DI and primary polydipsia. The direct test including vasopressin (AVP) measurement upon osmotic stimulation was meant to overcome these limitations, but failed to enter clinical practice mainly due to technical constraints of the AVP assay. Copeptin is secreted in equimolar amount to AVP but can easily be measured with a sandwich immunoassay. A high correlation between copeptin and AVP has been shown. Accordingly, copeptin mirrors the amount of AVP in the circulation and has led to a 'revival' of the direct test in the differential diagnosis of DI. We have shown that a baseline copeptin, without prior thirsting, unequivocally identifies patients with nephrogenic DI. In contrast, for the differentiation between central DI and primary polydipsia, a stimulated copeptin level of 4.9 pmol/L upon hypertonic saline infusion differentiates these two entities with a high diagnostic accuracy and is superior to the water deprivation test. Close sodium monitoring during the test is a prerequisite. Further new test methods are currently evaluated and might provide an even simpler way of differential diagnosis in the future.
Collapse
Affiliation(s)
- Mirjam Christ-Crain
- Department of Endocrinology, Diabetology and Metabolism and Department of Clinical Research, University Hospital Basel, Basel, Switzerland
- University of Basel, Basel, Switzerland
| |
Collapse
|
40
|
Berends YR, Tulen JH, Wierdsma AI, van Pelt J, Kushner SA, van Marle HJ. Oxytocin, vasopressin and trust: Associations with aggressive behavior in healthy young males. Physiol Behav 2019; 204:180-185. [DOI: 10.1016/j.physbeh.2019.02.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 02/20/2019] [Accepted: 02/21/2019] [Indexed: 11/30/2022]
|
41
|
Andersson KE, Longstreth J, Brucker BM, Campeau L, Cheng L, Francis L, Fein S. Pharmacokinetic and Pharmacodynamic Properties of a Micro-Dose Nasal Spray Formulation of Desmopressin (AV002) in Healthy Water-Loaded Subjects. Pharm Res 2019; 36:92. [PMID: 31037429 PMCID: PMC6488552 DOI: 10.1007/s11095-019-2628-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 04/10/2019] [Indexed: 11/24/2022]
Abstract
Purpose Antidiuretic therapy with desmopressin for nocturia has been hampered by formulations with high doses, low bioavailability and variable pharmacokinetics. AV002 (SER120), a novel, emulsified, microdose desmopressin nasal spray, with a permeation enhancer (cylcopentadecanolide), was developed to have pharmacokinetic characteristics suitable for nocturia treatment. Methods Twelve healthy subjects participated in an open-label, dose-escalating study. Water-loaded subjects were sequentially dosed every 48 h with AV002 0.5, 1.0, 2.0 μg and 0.12 μg desmopressin subcutaneous (SC) bolus injection. Results AV002 intranasal administration produced a time-to-maximum concentration (Tmax) between 15 and 30 min and a maximum concentration (Cmax) <10 pg/mL. Cmax and area under the curve showed dose proportionality. Coefficient of variation for AV002 was similar to that observed for the SC dose. Bioavailability of AV002 was approximately 8% compared to SC injection. AV002 demonstrated pharmacodynamic effects within 20 min of dosing and showed increasing magnitude and duration with escalating doses. AV002 2.0 μg had maximum median urine osmolality of 629 mOsm/kg and median urine output ≤2 mL/min for 5–6 h. Conclusions AV002 demonstrated rapid absorption, high bioavailability, limited duration of action, and low coefficient of variation, suggesting it may be a suitable formulation for nocturia treatment. Trial registration not required (single-center, phase 1).
Collapse
Affiliation(s)
- Karl-Erik Andersson
- Institute for Regenerative Medicine, Wake Forest University School of Medicine, Richard H. Dean Biomedical Building, 391 Technology Way, Winston-Salem, North Carolina, USA.
| | | | - Benjamin M Brucker
- Department of Urology, Langone Health, New York University, New York, New York, USA
| | - Lysanne Campeau
- Division of Urology, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Linda Cheng
- Serenity Pharmaceuticals LLC, Milford, Pennsylvania, USA
| | - Leo Francis
- Avadel Specialty Pharmaceuticals plc, Chesterfield, Missouri, USA
| | - Seymour Fein
- Serenity Pharmaceuticals LLC, Milford, Pennsylvania, USA
| |
Collapse
|
42
|
Lema SC, Washburn EH, Crowley ME, Carvalho PG, Egelston JN, McCormick SD. Evidence for a role of arginine vasotocin receptors in the gill during salinity acclimation by a euryhaline teleost fish. Am J Physiol Regul Integr Comp Physiol 2019; 316:R735-R750. [PMID: 30916577 DOI: 10.1152/ajpregu.00328.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The nonapeptide arginine vasotocin (AVT) regulates osmotic balance in teleost fishes, but its mechanisms of action are not fully understood. Recently, it was discovered that nonapeptide receptors in teleost fishes are differentiated into two V1a-type, several V2-type, and two isotocin (IT) receptors, but it remains unclear which receptors mediate AVT's effects on gill osmoregulation. Here, we examined the role of nonapeptide receptors in the gill of the euryhaline Amargosa pupfish (Cyprinodon nevadensis amargosae) during osmotic acclimation. Transcripts for the teleost V1a-type receptor v1a2 were upregulated over fourfold in gill 24 h after transferring pupfish from 7.5 ppt to seawater (35 ppt) or hypersaline (55 ppt) conditions and downregulated after transfer to freshwater (0.3 ppt). Gill transcripts for the nonapeptide degradation enzyme leucyl-cystinyl aminopeptidase (LNPEP) also increased in fish acclimating to 35 ppt. To test whether the effects of AVT on the gill might be mediated by a V1a-type receptor, we administered AVT or a V1-type receptor antagonist (Manning compound) intraperitoneally to pupfish before transfer to 0.4 ppt or 35 ppt. Pupfish transferred to 35 ppt exhibited elevated gill mRNA abundance for cystic fibrosis transmembrane conductance regulator (cftr), but that upregulation diminished under V1-receptor inhibition. AVT inhibited the increase in gill Na+/Cl- cotransporter 2 (ncc2) transcript abundance that occurs following transfer to hypoosmotic environments, whereas V1-type receptor antagonism increased ncc2 mRNAs even without a change in salinity. These findings indicate that AVT acts via a V1-type receptor to regulate gill Cl- transport by inhibiting Cl- uptake and facilitating Cl- secretion during seawater acclimation.
Collapse
Affiliation(s)
- Sean C Lema
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University , San Luis Obispo, California
| | - Elsie H Washburn
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University , San Luis Obispo, California
| | - Mary E Crowley
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University , San Luis Obispo, California
| | - Paul G Carvalho
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University , San Luis Obispo, California
| | - Jennifer N Egelston
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University , San Luis Obispo, California
| | - Stephen D McCormick
- United States Geological Survey, Leetown Science Center, Conte Anadromous Fish Research Laboratory, Turners Falls, Massachusetts
| |
Collapse
|
43
|
Hernández-Pérez OR, Hernández VS, Nava-Kopp AT, Barrio RA, Seifi M, Swinny JD, Eiden LE, Zhang L. A Synaptically Connected Hypothalamic Magnocellular Vasopressin-Locus Coeruleus Neuronal Circuit and Its Plasticity in Response to Emotional and Physiological Stress. Front Neurosci 2019; 13:196. [PMID: 30949017 PMCID: PMC6435582 DOI: 10.3389/fnins.2019.00196] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 02/19/2019] [Indexed: 01/01/2023] Open
Abstract
The locus coeruleus (LC)-norepinephrine (NE) system modulates a range of salient brain functions, including memory and response to stress. The LC-NE system is regulated by neurochemically diverse inputs, including a range of neuropeptides such as arginine-vasopressin (AVP). Whilst the origins of many of these LC inputs, their synaptic connectivity with LC neurons, and their contribution to LC-mediated brain functions, have been well characterized, this is not the case for the AVP-LC system. Therefore, our aims were to define the types of synapses formed by AVP+ fibers with LC neurons using immunohistochemistry together with confocal and transmission electron microscopy (TEM), the origins of such inputs, using retrograde tracers, and the plasticity of the LC AVP system in response to stress and spatial learning, using the maternal separation (MS) and Morris water maze (MWM) paradigms, respectively, in rat. Confocal microscopy revealed that AVP+ fibers contacting tyrosine hydroxylase (TH)+ LC neurons were also immunopositive for vesicular glutamate transporter 2, a marker of presynaptic glutamatergic axons. TEM confirmed that AVP+ axons formed Gray type I (asymmetric) synapses with TH+ dendrites thus confirming excitatory synaptic connections between these systems. Retrograde tracing revealed that these LC AVP+ fibers originate from hypothalamic vasopressinergic magnocellular neurosecretory neurons (AVPMNNs). MS induced a significant increase in the density of LC AVP+ fibers. Finally, AVPMNN circuit upregulation by water-deprivation improved MWM performance while increased Fos expression was found in LC and efferent regions such as hippocampus and prefrontal cortex, suggesting that AVPMMN projections to LC could integrate homeostatic responses modifying neuroplasticity.
Collapse
Affiliation(s)
- Oscar R Hernández-Pérez
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Vito S Hernández
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Alicia T Nava-Kopp
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Rafael A Barrio
- Instituto de Física, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Mohsen Seifi
- School of Pharmacy and Biomedical Sciences, Institute for Biomedical and Biomolecular Science, University of Portsmouth, Portsmouth, United Kingdom
| | - Jerome D Swinny
- School of Pharmacy and Biomedical Sciences, Institute for Biomedical and Biomolecular Science, University of Portsmouth, Portsmouth, United Kingdom
| | - Lee E Eiden
- Section on Molecular Neuroscience, National Institute of Mental Health-IRP, Bethesda, MD, United States
| | - Limei Zhang
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
44
|
Kass-Iliyya A, Hashim H. Nocturnal polyuria: Literature review of definition, pathophysiology, investigations and treatment. JOURNAL OF CLINICAL UROLOGY 2019. [DOI: 10.1177/2051415818756792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Nocturnal polyuria (NP) is characterised by increased urine production overnight in comparison to daytime. It has significant adverse events in adults including reduced quality of life, increased risk of falls and increased mortality.Although NP can be a manifestation of other significant conditions like heart failure and sleep apnoea, there are lots of unanswered questions about NP. What is the underlying pathophysiology? Is NP a physiological manifestation of ageing? Is the circadian change of vasopressin release the primary pathology? Or is it a secondary phenomenon to a low diurnal production of urine? Is the primary pathology at the kidney level or is there another humoral, cardiac, or endovascular element? In this article, we summarise the available English-language literature on the subject of NP, including its epidemiology, pathogenesis, diagnosis and treatment.Level of evidence: Not applicable for this multicentre audit.
Collapse
Affiliation(s)
- Antoine Kass-Iliyya
- Urology Department, Bristol Urological Institute, Southmead Hospital, Bristol, BS10 5NB, UK
| | - Hashim Hashim
- Urology Department, Bristol Urological Institute, Southmead Hospital, Bristol, BS10 5NB, UK
| |
Collapse
|
45
|
Armstrong LE, Johnson EC. Water Intake, Water Balance, and the Elusive Daily Water Requirement. Nutrients 2018; 10:E1928. [PMID: 30563134 PMCID: PMC6315424 DOI: 10.3390/nu10121928] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 11/11/2018] [Accepted: 11/27/2018] [Indexed: 12/12/2022] Open
Abstract
Water is essential for metabolism, substrate transport across membranes, cellular homeostasis, temperature regulation, and circulatory function. Although nutritional and physiological research teams and professional organizations have described the daily total water intakes (TWI, L/24h) and Adequate Intakes (AI) of children, women, and men, there is no widespread consensus regarding the human water requirements of different demographic groups. These requirements remain undefined because of the dynamic complexity inherent in the human water regulatory network, which involves the central nervous system and several organ systems, as well as large inter-individual differences. The present review analyzes published evidence that is relevant to these issues and presents a novel approach to assessing the daily water requirements of individuals in all sex and life-stage groups, as an alternative to AI values based on survey data. This empirical method focuses on the intensity of a specific neuroendocrine response (e.g., plasma arginine vasopressin (AVP) concentration) employed by the brain to regulate total body water volume and concentration. We consider this autonomically-controlled neuroendocrine response to be an inherent hydration biomarker and one means by which the brain maintains good health and optimal function. We also propose that this individualized method defines the elusive state of euhydration (i.e., water balance) and distinguishes it from hypohydration. Using plasma AVP concentration to analyze multiple published data sets that included both men and women, we determined that a mild neuroendocrine defense of body water commences when TWI is ˂1.8 L/24h, that 19⁻71% of adults in various countries consume less than this TWI each day, and consuming less than the 24-h water AI may influence the risk of dysfunctional metabolism and chronic diseases.
Collapse
Affiliation(s)
- Lawrence E Armstrong
- University of Connecticut, Human Performance Laboratory and Department of Nutritional Sciences, Storrs CT 06269-1110, USA.
| | - Evan C Johnson
- University of Wyoming, Human Integrated Physiology Laboratory, Division of Kinesiology and Health, Laramie, WY 82071, USA.
| |
Collapse
|
46
|
The Effect of Water Loading on Acute Weight Loss Following Fluid Restriction in Combat Sports Athletes. Int J Sport Nutr Exerc Metab 2018; 28:565-573. [DOI: 10.1123/ijsnem.2017-0183] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Novel methods of acute weight loss practiced by combat sport athletes include “water loading,” the consumption of large fluid volumes for several days prior to restriction. We examined claims that this technique increases total body water losses, while also assessing the risk of hyponatremia. Male athletes were separated into control (n = 10) and water loading (n = 11) groups and fed a standardized energy-matched diet for 6 days. Days 1–3 fluid intake was 40 and 100 ml/kg for control and water loading groups, respectively, with both groups consuming 15 ml/kg on Day 4 and following the same rehydration protocol on Days 5 and 6. We tracked body mass (BM), urine sodium, urine specific gravity and volume, training-related sweat losses and blood concentrations of renal hormones, and urea and electrolytes throughout. Physical performance was assessed preintervention and postintervention. Following fluid restriction, there were substantial differences between groups in the ratio of fluid input/output (39%, p < .01, effect size = 1.2) and BM loss (0.6% BM, p = .02, effect size = 0.82). Changes in urine specific gravity, urea and electrolytes, and renal hormones occurred over time (p < .05), with an interaction of time and intervention on blood sodium, potassium, chloride, urea, creatinine, urine specific gravity, and vasopressin (p < .05). Measurements of urea and electrolyte remained within reference ranges, and no differences in physical performance were detected over time or between groups. Water loading appears to be a safe and effective method of acute BM loss under the conditions of this study. Vasopressin-regulated changes in aquaporin channels may potentially partially explain the mechanism of increased body water loss with water loading.
Collapse
|
47
|
Fenske W, Refardt J, Chifu I, Schnyder I, Winzeler B, Drummond J, Ribeiro-Oliveira A, Drescher T, Bilz S, Vogt DR, Malzahn U, Kroiss M, Christ E, Henzen C, Fischli S, Tönjes A, Mueller B, Schopohl J, Flitsch J, Brabant G, Fassnacht M, Christ-Crain M. A Copeptin-Based Approach in the Diagnosis of Diabetes Insipidus. N Engl J Med 2018; 379:428-439. [PMID: 30067922 DOI: 10.1056/nejmoa1803760] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND The indirect water-deprivation test is the current reference standard for the diagnosis of diabetes insipidus. However, it is technically cumbersome to administer, and the results are often inaccurate. The current study compared the indirect water-deprivation test with direct detection of plasma copeptin, a precursor-derived surrogate of arginine vasopressin. METHODS From 2013 to 2017, we recruited 156 patients with hypotonic polyuria at 11 medical centers to undergo both water-deprivation and hypertonic saline infusion tests. In the latter test, plasma copeptin was measured when the plasma sodium level had increased to at least 150 mmol per liter after infusion of hypertonic saline. The primary outcome was the overall diagnostic accuracy of each test as compared with the final reference diagnosis, which was determined on the basis of medical history, test results, and treatment response, with copeptin levels masked. RESULTS A total of 144 patients underwent both tests. The final diagnosis was primary polydipsia in 82 patients (57%), central diabetes insipidus in 59 (41%), and nephrogenic diabetes insipidus in 3 (2%). Overall, among the 141 patients included in the analysis, the indirect water-deprivation test determined the correct diagnosis in 108 patients (diagnostic accuracy, 76.6%; 95% confidence interval [CI], 68.9 to 83.2), and the hypertonic saline infusion test (with a copeptin cutoff level of >4.9 pmol per liter) determined the correct diagnosis in 136 patients (96.5%; 95% CI, 92.1 to 98.6; P<0.001). The indirect water-deprivation test correctly distinguished primary polydipsia from partial central diabetes insipidus in 77 of 105 patients (73.3%; 95% CI, 63.9 to 81.2), and the hypertonic saline infusion test distinguished between the two conditions in 99 of 104 patients (95.2%; 95% CI, 89.4 to 98.1; adjusted P<0.001). One serious adverse event (desmopressin-induced hyponatremia that resulted in hospitalization) occurred during the water-deprivation test. CONCLUSIONS The direct measurement of hypertonic saline-stimulated plasma copeptin had greater diagnostic accuracy than the water-deprivation test in patients with hypotonic polyuria. (Funded by the Swiss National Foundation and others; ClinicalTrials.gov number, NCT01940614 .).
Collapse
Affiliation(s)
- Wiebke Fenske
- From the University of Leipzig, Department of Endocrinology and Nephrology (W.F., A.T.), and Leipzig University Medical Center, Integrated Research and Treatment Center Adiposity Diseases (W.F.), Leipzig, the Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg (I.C., M.K., M.F.), and the Clinical Trial Center (U.M.) and Central Laboratory (M.F.), University Hospital Würzburg, Würzburg, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität, Munich (J.S.), the Department of Neurosurgery, University Hospital Hamburg-Eppendorf, Hamburg (J.F.), and Experimental and Clinical Endocrinology, University Hospital Lübeck, Lübeck (G.B.) - all in Germany; the Clinic of Endocrinology, Diabetology and Metabolism (J.R., I.S., B.W., E.C., M.C.-C.) and the Clinical Trial Unit (D.R.V.), Department of Clinical Research, University of Basel and University Hospital Basel, Basel, the Department of Endocrinology, Kantonsspital St. Gallen, St. Gallen (T.D., S.B.), the Department of Endocrinology, Inselspital Bern, Bern (E.C.), the Department of Endocrinology, Luzerner Kantonsspital, Lucerne (C.H., S.F.), and the Division of Endocrinology, Diabetology and Metabolism, Medical University Clinic, Kantonsspital Aarau, Aarau (B.M.) - all in Switzerland; and Faculdade de Medicina da UFMG, Universidade Federal de Minas Gerais (J.D., A.R.-O.), and Hermes Pardini Institute (J.D.), Belo Horizonte, Brazil
| | - Julie Refardt
- From the University of Leipzig, Department of Endocrinology and Nephrology (W.F., A.T.), and Leipzig University Medical Center, Integrated Research and Treatment Center Adiposity Diseases (W.F.), Leipzig, the Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg (I.C., M.K., M.F.), and the Clinical Trial Center (U.M.) and Central Laboratory (M.F.), University Hospital Würzburg, Würzburg, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität, Munich (J.S.), the Department of Neurosurgery, University Hospital Hamburg-Eppendorf, Hamburg (J.F.), and Experimental and Clinical Endocrinology, University Hospital Lübeck, Lübeck (G.B.) - all in Germany; the Clinic of Endocrinology, Diabetology and Metabolism (J.R., I.S., B.W., E.C., M.C.-C.) and the Clinical Trial Unit (D.R.V.), Department of Clinical Research, University of Basel and University Hospital Basel, Basel, the Department of Endocrinology, Kantonsspital St. Gallen, St. Gallen (T.D., S.B.), the Department of Endocrinology, Inselspital Bern, Bern (E.C.), the Department of Endocrinology, Luzerner Kantonsspital, Lucerne (C.H., S.F.), and the Division of Endocrinology, Diabetology and Metabolism, Medical University Clinic, Kantonsspital Aarau, Aarau (B.M.) - all in Switzerland; and Faculdade de Medicina da UFMG, Universidade Federal de Minas Gerais (J.D., A.R.-O.), and Hermes Pardini Institute (J.D.), Belo Horizonte, Brazil
| | - Irina Chifu
- From the University of Leipzig, Department of Endocrinology and Nephrology (W.F., A.T.), and Leipzig University Medical Center, Integrated Research and Treatment Center Adiposity Diseases (W.F.), Leipzig, the Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg (I.C., M.K., M.F.), and the Clinical Trial Center (U.M.) and Central Laboratory (M.F.), University Hospital Würzburg, Würzburg, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität, Munich (J.S.), the Department of Neurosurgery, University Hospital Hamburg-Eppendorf, Hamburg (J.F.), and Experimental and Clinical Endocrinology, University Hospital Lübeck, Lübeck (G.B.) - all in Germany; the Clinic of Endocrinology, Diabetology and Metabolism (J.R., I.S., B.W., E.C., M.C.-C.) and the Clinical Trial Unit (D.R.V.), Department of Clinical Research, University of Basel and University Hospital Basel, Basel, the Department of Endocrinology, Kantonsspital St. Gallen, St. Gallen (T.D., S.B.), the Department of Endocrinology, Inselspital Bern, Bern (E.C.), the Department of Endocrinology, Luzerner Kantonsspital, Lucerne (C.H., S.F.), and the Division of Endocrinology, Diabetology and Metabolism, Medical University Clinic, Kantonsspital Aarau, Aarau (B.M.) - all in Switzerland; and Faculdade de Medicina da UFMG, Universidade Federal de Minas Gerais (J.D., A.R.-O.), and Hermes Pardini Institute (J.D.), Belo Horizonte, Brazil
| | - Ingeborg Schnyder
- From the University of Leipzig, Department of Endocrinology and Nephrology (W.F., A.T.), and Leipzig University Medical Center, Integrated Research and Treatment Center Adiposity Diseases (W.F.), Leipzig, the Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg (I.C., M.K., M.F.), and the Clinical Trial Center (U.M.) and Central Laboratory (M.F.), University Hospital Würzburg, Würzburg, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität, Munich (J.S.), the Department of Neurosurgery, University Hospital Hamburg-Eppendorf, Hamburg (J.F.), and Experimental and Clinical Endocrinology, University Hospital Lübeck, Lübeck (G.B.) - all in Germany; the Clinic of Endocrinology, Diabetology and Metabolism (J.R., I.S., B.W., E.C., M.C.-C.) and the Clinical Trial Unit (D.R.V.), Department of Clinical Research, University of Basel and University Hospital Basel, Basel, the Department of Endocrinology, Kantonsspital St. Gallen, St. Gallen (T.D., S.B.), the Department of Endocrinology, Inselspital Bern, Bern (E.C.), the Department of Endocrinology, Luzerner Kantonsspital, Lucerne (C.H., S.F.), and the Division of Endocrinology, Diabetology and Metabolism, Medical University Clinic, Kantonsspital Aarau, Aarau (B.M.) - all in Switzerland; and Faculdade de Medicina da UFMG, Universidade Federal de Minas Gerais (J.D., A.R.-O.), and Hermes Pardini Institute (J.D.), Belo Horizonte, Brazil
| | - Bettina Winzeler
- From the University of Leipzig, Department of Endocrinology and Nephrology (W.F., A.T.), and Leipzig University Medical Center, Integrated Research and Treatment Center Adiposity Diseases (W.F.), Leipzig, the Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg (I.C., M.K., M.F.), and the Clinical Trial Center (U.M.) and Central Laboratory (M.F.), University Hospital Würzburg, Würzburg, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität, Munich (J.S.), the Department of Neurosurgery, University Hospital Hamburg-Eppendorf, Hamburg (J.F.), and Experimental and Clinical Endocrinology, University Hospital Lübeck, Lübeck (G.B.) - all in Germany; the Clinic of Endocrinology, Diabetology and Metabolism (J.R., I.S., B.W., E.C., M.C.-C.) and the Clinical Trial Unit (D.R.V.), Department of Clinical Research, University of Basel and University Hospital Basel, Basel, the Department of Endocrinology, Kantonsspital St. Gallen, St. Gallen (T.D., S.B.), the Department of Endocrinology, Inselspital Bern, Bern (E.C.), the Department of Endocrinology, Luzerner Kantonsspital, Lucerne (C.H., S.F.), and the Division of Endocrinology, Diabetology and Metabolism, Medical University Clinic, Kantonsspital Aarau, Aarau (B.M.) - all in Switzerland; and Faculdade de Medicina da UFMG, Universidade Federal de Minas Gerais (J.D., A.R.-O.), and Hermes Pardini Institute (J.D.), Belo Horizonte, Brazil
| | - Juliana Drummond
- From the University of Leipzig, Department of Endocrinology and Nephrology (W.F., A.T.), and Leipzig University Medical Center, Integrated Research and Treatment Center Adiposity Diseases (W.F.), Leipzig, the Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg (I.C., M.K., M.F.), and the Clinical Trial Center (U.M.) and Central Laboratory (M.F.), University Hospital Würzburg, Würzburg, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität, Munich (J.S.), the Department of Neurosurgery, University Hospital Hamburg-Eppendorf, Hamburg (J.F.), and Experimental and Clinical Endocrinology, University Hospital Lübeck, Lübeck (G.B.) - all in Germany; the Clinic of Endocrinology, Diabetology and Metabolism (J.R., I.S., B.W., E.C., M.C.-C.) and the Clinical Trial Unit (D.R.V.), Department of Clinical Research, University of Basel and University Hospital Basel, Basel, the Department of Endocrinology, Kantonsspital St. Gallen, St. Gallen (T.D., S.B.), the Department of Endocrinology, Inselspital Bern, Bern (E.C.), the Department of Endocrinology, Luzerner Kantonsspital, Lucerne (C.H., S.F.), and the Division of Endocrinology, Diabetology and Metabolism, Medical University Clinic, Kantonsspital Aarau, Aarau (B.M.) - all in Switzerland; and Faculdade de Medicina da UFMG, Universidade Federal de Minas Gerais (J.D., A.R.-O.), and Hermes Pardini Institute (J.D.), Belo Horizonte, Brazil
| | - Antônio Ribeiro-Oliveira
- From the University of Leipzig, Department of Endocrinology and Nephrology (W.F., A.T.), and Leipzig University Medical Center, Integrated Research and Treatment Center Adiposity Diseases (W.F.), Leipzig, the Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg (I.C., M.K., M.F.), and the Clinical Trial Center (U.M.) and Central Laboratory (M.F.), University Hospital Würzburg, Würzburg, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität, Munich (J.S.), the Department of Neurosurgery, University Hospital Hamburg-Eppendorf, Hamburg (J.F.), and Experimental and Clinical Endocrinology, University Hospital Lübeck, Lübeck (G.B.) - all in Germany; the Clinic of Endocrinology, Diabetology and Metabolism (J.R., I.S., B.W., E.C., M.C.-C.) and the Clinical Trial Unit (D.R.V.), Department of Clinical Research, University of Basel and University Hospital Basel, Basel, the Department of Endocrinology, Kantonsspital St. Gallen, St. Gallen (T.D., S.B.), the Department of Endocrinology, Inselspital Bern, Bern (E.C.), the Department of Endocrinology, Luzerner Kantonsspital, Lucerne (C.H., S.F.), and the Division of Endocrinology, Diabetology and Metabolism, Medical University Clinic, Kantonsspital Aarau, Aarau (B.M.) - all in Switzerland; and Faculdade de Medicina da UFMG, Universidade Federal de Minas Gerais (J.D., A.R.-O.), and Hermes Pardini Institute (J.D.), Belo Horizonte, Brazil
| | - Tilman Drescher
- From the University of Leipzig, Department of Endocrinology and Nephrology (W.F., A.T.), and Leipzig University Medical Center, Integrated Research and Treatment Center Adiposity Diseases (W.F.), Leipzig, the Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg (I.C., M.K., M.F.), and the Clinical Trial Center (U.M.) and Central Laboratory (M.F.), University Hospital Würzburg, Würzburg, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität, Munich (J.S.), the Department of Neurosurgery, University Hospital Hamburg-Eppendorf, Hamburg (J.F.), and Experimental and Clinical Endocrinology, University Hospital Lübeck, Lübeck (G.B.) - all in Germany; the Clinic of Endocrinology, Diabetology and Metabolism (J.R., I.S., B.W., E.C., M.C.-C.) and the Clinical Trial Unit (D.R.V.), Department of Clinical Research, University of Basel and University Hospital Basel, Basel, the Department of Endocrinology, Kantonsspital St. Gallen, St. Gallen (T.D., S.B.), the Department of Endocrinology, Inselspital Bern, Bern (E.C.), the Department of Endocrinology, Luzerner Kantonsspital, Lucerne (C.H., S.F.), and the Division of Endocrinology, Diabetology and Metabolism, Medical University Clinic, Kantonsspital Aarau, Aarau (B.M.) - all in Switzerland; and Faculdade de Medicina da UFMG, Universidade Federal de Minas Gerais (J.D., A.R.-O.), and Hermes Pardini Institute (J.D.), Belo Horizonte, Brazil
| | - Stefan Bilz
- From the University of Leipzig, Department of Endocrinology and Nephrology (W.F., A.T.), and Leipzig University Medical Center, Integrated Research and Treatment Center Adiposity Diseases (W.F.), Leipzig, the Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg (I.C., M.K., M.F.), and the Clinical Trial Center (U.M.) and Central Laboratory (M.F.), University Hospital Würzburg, Würzburg, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität, Munich (J.S.), the Department of Neurosurgery, University Hospital Hamburg-Eppendorf, Hamburg (J.F.), and Experimental and Clinical Endocrinology, University Hospital Lübeck, Lübeck (G.B.) - all in Germany; the Clinic of Endocrinology, Diabetology and Metabolism (J.R., I.S., B.W., E.C., M.C.-C.) and the Clinical Trial Unit (D.R.V.), Department of Clinical Research, University of Basel and University Hospital Basel, Basel, the Department of Endocrinology, Kantonsspital St. Gallen, St. Gallen (T.D., S.B.), the Department of Endocrinology, Inselspital Bern, Bern (E.C.), the Department of Endocrinology, Luzerner Kantonsspital, Lucerne (C.H., S.F.), and the Division of Endocrinology, Diabetology and Metabolism, Medical University Clinic, Kantonsspital Aarau, Aarau (B.M.) - all in Switzerland; and Faculdade de Medicina da UFMG, Universidade Federal de Minas Gerais (J.D., A.R.-O.), and Hermes Pardini Institute (J.D.), Belo Horizonte, Brazil
| | - Deborah R Vogt
- From the University of Leipzig, Department of Endocrinology and Nephrology (W.F., A.T.), and Leipzig University Medical Center, Integrated Research and Treatment Center Adiposity Diseases (W.F.), Leipzig, the Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg (I.C., M.K., M.F.), and the Clinical Trial Center (U.M.) and Central Laboratory (M.F.), University Hospital Würzburg, Würzburg, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität, Munich (J.S.), the Department of Neurosurgery, University Hospital Hamburg-Eppendorf, Hamburg (J.F.), and Experimental and Clinical Endocrinology, University Hospital Lübeck, Lübeck (G.B.) - all in Germany; the Clinic of Endocrinology, Diabetology and Metabolism (J.R., I.S., B.W., E.C., M.C.-C.) and the Clinical Trial Unit (D.R.V.), Department of Clinical Research, University of Basel and University Hospital Basel, Basel, the Department of Endocrinology, Kantonsspital St. Gallen, St. Gallen (T.D., S.B.), the Department of Endocrinology, Inselspital Bern, Bern (E.C.), the Department of Endocrinology, Luzerner Kantonsspital, Lucerne (C.H., S.F.), and the Division of Endocrinology, Diabetology and Metabolism, Medical University Clinic, Kantonsspital Aarau, Aarau (B.M.) - all in Switzerland; and Faculdade de Medicina da UFMG, Universidade Federal de Minas Gerais (J.D., A.R.-O.), and Hermes Pardini Institute (J.D.), Belo Horizonte, Brazil
| | - Uwe Malzahn
- From the University of Leipzig, Department of Endocrinology and Nephrology (W.F., A.T.), and Leipzig University Medical Center, Integrated Research and Treatment Center Adiposity Diseases (W.F.), Leipzig, the Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg (I.C., M.K., M.F.), and the Clinical Trial Center (U.M.) and Central Laboratory (M.F.), University Hospital Würzburg, Würzburg, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität, Munich (J.S.), the Department of Neurosurgery, University Hospital Hamburg-Eppendorf, Hamburg (J.F.), and Experimental and Clinical Endocrinology, University Hospital Lübeck, Lübeck (G.B.) - all in Germany; the Clinic of Endocrinology, Diabetology and Metabolism (J.R., I.S., B.W., E.C., M.C.-C.) and the Clinical Trial Unit (D.R.V.), Department of Clinical Research, University of Basel and University Hospital Basel, Basel, the Department of Endocrinology, Kantonsspital St. Gallen, St. Gallen (T.D., S.B.), the Department of Endocrinology, Inselspital Bern, Bern (E.C.), the Department of Endocrinology, Luzerner Kantonsspital, Lucerne (C.H., S.F.), and the Division of Endocrinology, Diabetology and Metabolism, Medical University Clinic, Kantonsspital Aarau, Aarau (B.M.) - all in Switzerland; and Faculdade de Medicina da UFMG, Universidade Federal de Minas Gerais (J.D., A.R.-O.), and Hermes Pardini Institute (J.D.), Belo Horizonte, Brazil
| | - Matthias Kroiss
- From the University of Leipzig, Department of Endocrinology and Nephrology (W.F., A.T.), and Leipzig University Medical Center, Integrated Research and Treatment Center Adiposity Diseases (W.F.), Leipzig, the Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg (I.C., M.K., M.F.), and the Clinical Trial Center (U.M.) and Central Laboratory (M.F.), University Hospital Würzburg, Würzburg, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität, Munich (J.S.), the Department of Neurosurgery, University Hospital Hamburg-Eppendorf, Hamburg (J.F.), and Experimental and Clinical Endocrinology, University Hospital Lübeck, Lübeck (G.B.) - all in Germany; the Clinic of Endocrinology, Diabetology and Metabolism (J.R., I.S., B.W., E.C., M.C.-C.) and the Clinical Trial Unit (D.R.V.), Department of Clinical Research, University of Basel and University Hospital Basel, Basel, the Department of Endocrinology, Kantonsspital St. Gallen, St. Gallen (T.D., S.B.), the Department of Endocrinology, Inselspital Bern, Bern (E.C.), the Department of Endocrinology, Luzerner Kantonsspital, Lucerne (C.H., S.F.), and the Division of Endocrinology, Diabetology and Metabolism, Medical University Clinic, Kantonsspital Aarau, Aarau (B.M.) - all in Switzerland; and Faculdade de Medicina da UFMG, Universidade Federal de Minas Gerais (J.D., A.R.-O.), and Hermes Pardini Institute (J.D.), Belo Horizonte, Brazil
| | - Emanuel Christ
- From the University of Leipzig, Department of Endocrinology and Nephrology (W.F., A.T.), and Leipzig University Medical Center, Integrated Research and Treatment Center Adiposity Diseases (W.F.), Leipzig, the Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg (I.C., M.K., M.F.), and the Clinical Trial Center (U.M.) and Central Laboratory (M.F.), University Hospital Würzburg, Würzburg, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität, Munich (J.S.), the Department of Neurosurgery, University Hospital Hamburg-Eppendorf, Hamburg (J.F.), and Experimental and Clinical Endocrinology, University Hospital Lübeck, Lübeck (G.B.) - all in Germany; the Clinic of Endocrinology, Diabetology and Metabolism (J.R., I.S., B.W., E.C., M.C.-C.) and the Clinical Trial Unit (D.R.V.), Department of Clinical Research, University of Basel and University Hospital Basel, Basel, the Department of Endocrinology, Kantonsspital St. Gallen, St. Gallen (T.D., S.B.), the Department of Endocrinology, Inselspital Bern, Bern (E.C.), the Department of Endocrinology, Luzerner Kantonsspital, Lucerne (C.H., S.F.), and the Division of Endocrinology, Diabetology and Metabolism, Medical University Clinic, Kantonsspital Aarau, Aarau (B.M.) - all in Switzerland; and Faculdade de Medicina da UFMG, Universidade Federal de Minas Gerais (J.D., A.R.-O.), and Hermes Pardini Institute (J.D.), Belo Horizonte, Brazil
| | - Christoph Henzen
- From the University of Leipzig, Department of Endocrinology and Nephrology (W.F., A.T.), and Leipzig University Medical Center, Integrated Research and Treatment Center Adiposity Diseases (W.F.), Leipzig, the Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg (I.C., M.K., M.F.), and the Clinical Trial Center (U.M.) and Central Laboratory (M.F.), University Hospital Würzburg, Würzburg, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität, Munich (J.S.), the Department of Neurosurgery, University Hospital Hamburg-Eppendorf, Hamburg (J.F.), and Experimental and Clinical Endocrinology, University Hospital Lübeck, Lübeck (G.B.) - all in Germany; the Clinic of Endocrinology, Diabetology and Metabolism (J.R., I.S., B.W., E.C., M.C.-C.) and the Clinical Trial Unit (D.R.V.), Department of Clinical Research, University of Basel and University Hospital Basel, Basel, the Department of Endocrinology, Kantonsspital St. Gallen, St. Gallen (T.D., S.B.), the Department of Endocrinology, Inselspital Bern, Bern (E.C.), the Department of Endocrinology, Luzerner Kantonsspital, Lucerne (C.H., S.F.), and the Division of Endocrinology, Diabetology and Metabolism, Medical University Clinic, Kantonsspital Aarau, Aarau (B.M.) - all in Switzerland; and Faculdade de Medicina da UFMG, Universidade Federal de Minas Gerais (J.D., A.R.-O.), and Hermes Pardini Institute (J.D.), Belo Horizonte, Brazil
| | - Stefan Fischli
- From the University of Leipzig, Department of Endocrinology and Nephrology (W.F., A.T.), and Leipzig University Medical Center, Integrated Research and Treatment Center Adiposity Diseases (W.F.), Leipzig, the Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg (I.C., M.K., M.F.), and the Clinical Trial Center (U.M.) and Central Laboratory (M.F.), University Hospital Würzburg, Würzburg, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität, Munich (J.S.), the Department of Neurosurgery, University Hospital Hamburg-Eppendorf, Hamburg (J.F.), and Experimental and Clinical Endocrinology, University Hospital Lübeck, Lübeck (G.B.) - all in Germany; the Clinic of Endocrinology, Diabetology and Metabolism (J.R., I.S., B.W., E.C., M.C.-C.) and the Clinical Trial Unit (D.R.V.), Department of Clinical Research, University of Basel and University Hospital Basel, Basel, the Department of Endocrinology, Kantonsspital St. Gallen, St. Gallen (T.D., S.B.), the Department of Endocrinology, Inselspital Bern, Bern (E.C.), the Department of Endocrinology, Luzerner Kantonsspital, Lucerne (C.H., S.F.), and the Division of Endocrinology, Diabetology and Metabolism, Medical University Clinic, Kantonsspital Aarau, Aarau (B.M.) - all in Switzerland; and Faculdade de Medicina da UFMG, Universidade Federal de Minas Gerais (J.D., A.R.-O.), and Hermes Pardini Institute (J.D.), Belo Horizonte, Brazil
| | - Anke Tönjes
- From the University of Leipzig, Department of Endocrinology and Nephrology (W.F., A.T.), and Leipzig University Medical Center, Integrated Research and Treatment Center Adiposity Diseases (W.F.), Leipzig, the Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg (I.C., M.K., M.F.), and the Clinical Trial Center (U.M.) and Central Laboratory (M.F.), University Hospital Würzburg, Würzburg, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität, Munich (J.S.), the Department of Neurosurgery, University Hospital Hamburg-Eppendorf, Hamburg (J.F.), and Experimental and Clinical Endocrinology, University Hospital Lübeck, Lübeck (G.B.) - all in Germany; the Clinic of Endocrinology, Diabetology and Metabolism (J.R., I.S., B.W., E.C., M.C.-C.) and the Clinical Trial Unit (D.R.V.), Department of Clinical Research, University of Basel and University Hospital Basel, Basel, the Department of Endocrinology, Kantonsspital St. Gallen, St. Gallen (T.D., S.B.), the Department of Endocrinology, Inselspital Bern, Bern (E.C.), the Department of Endocrinology, Luzerner Kantonsspital, Lucerne (C.H., S.F.), and the Division of Endocrinology, Diabetology and Metabolism, Medical University Clinic, Kantonsspital Aarau, Aarau (B.M.) - all in Switzerland; and Faculdade de Medicina da UFMG, Universidade Federal de Minas Gerais (J.D., A.R.-O.), and Hermes Pardini Institute (J.D.), Belo Horizonte, Brazil
| | - Beat Mueller
- From the University of Leipzig, Department of Endocrinology and Nephrology (W.F., A.T.), and Leipzig University Medical Center, Integrated Research and Treatment Center Adiposity Diseases (W.F.), Leipzig, the Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg (I.C., M.K., M.F.), and the Clinical Trial Center (U.M.) and Central Laboratory (M.F.), University Hospital Würzburg, Würzburg, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität, Munich (J.S.), the Department of Neurosurgery, University Hospital Hamburg-Eppendorf, Hamburg (J.F.), and Experimental and Clinical Endocrinology, University Hospital Lübeck, Lübeck (G.B.) - all in Germany; the Clinic of Endocrinology, Diabetology and Metabolism (J.R., I.S., B.W., E.C., M.C.-C.) and the Clinical Trial Unit (D.R.V.), Department of Clinical Research, University of Basel and University Hospital Basel, Basel, the Department of Endocrinology, Kantonsspital St. Gallen, St. Gallen (T.D., S.B.), the Department of Endocrinology, Inselspital Bern, Bern (E.C.), the Department of Endocrinology, Luzerner Kantonsspital, Lucerne (C.H., S.F.), and the Division of Endocrinology, Diabetology and Metabolism, Medical University Clinic, Kantonsspital Aarau, Aarau (B.M.) - all in Switzerland; and Faculdade de Medicina da UFMG, Universidade Federal de Minas Gerais (J.D., A.R.-O.), and Hermes Pardini Institute (J.D.), Belo Horizonte, Brazil
| | - Jochen Schopohl
- From the University of Leipzig, Department of Endocrinology and Nephrology (W.F., A.T.), and Leipzig University Medical Center, Integrated Research and Treatment Center Adiposity Diseases (W.F.), Leipzig, the Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg (I.C., M.K., M.F.), and the Clinical Trial Center (U.M.) and Central Laboratory (M.F.), University Hospital Würzburg, Würzburg, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität, Munich (J.S.), the Department of Neurosurgery, University Hospital Hamburg-Eppendorf, Hamburg (J.F.), and Experimental and Clinical Endocrinology, University Hospital Lübeck, Lübeck (G.B.) - all in Germany; the Clinic of Endocrinology, Diabetology and Metabolism (J.R., I.S., B.W., E.C., M.C.-C.) and the Clinical Trial Unit (D.R.V.), Department of Clinical Research, University of Basel and University Hospital Basel, Basel, the Department of Endocrinology, Kantonsspital St. Gallen, St. Gallen (T.D., S.B.), the Department of Endocrinology, Inselspital Bern, Bern (E.C.), the Department of Endocrinology, Luzerner Kantonsspital, Lucerne (C.H., S.F.), and the Division of Endocrinology, Diabetology and Metabolism, Medical University Clinic, Kantonsspital Aarau, Aarau (B.M.) - all in Switzerland; and Faculdade de Medicina da UFMG, Universidade Federal de Minas Gerais (J.D., A.R.-O.), and Hermes Pardini Institute (J.D.), Belo Horizonte, Brazil
| | - Jörg Flitsch
- From the University of Leipzig, Department of Endocrinology and Nephrology (W.F., A.T.), and Leipzig University Medical Center, Integrated Research and Treatment Center Adiposity Diseases (W.F.), Leipzig, the Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg (I.C., M.K., M.F.), and the Clinical Trial Center (U.M.) and Central Laboratory (M.F.), University Hospital Würzburg, Würzburg, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität, Munich (J.S.), the Department of Neurosurgery, University Hospital Hamburg-Eppendorf, Hamburg (J.F.), and Experimental and Clinical Endocrinology, University Hospital Lübeck, Lübeck (G.B.) - all in Germany; the Clinic of Endocrinology, Diabetology and Metabolism (J.R., I.S., B.W., E.C., M.C.-C.) and the Clinical Trial Unit (D.R.V.), Department of Clinical Research, University of Basel and University Hospital Basel, Basel, the Department of Endocrinology, Kantonsspital St. Gallen, St. Gallen (T.D., S.B.), the Department of Endocrinology, Inselspital Bern, Bern (E.C.), the Department of Endocrinology, Luzerner Kantonsspital, Lucerne (C.H., S.F.), and the Division of Endocrinology, Diabetology and Metabolism, Medical University Clinic, Kantonsspital Aarau, Aarau (B.M.) - all in Switzerland; and Faculdade de Medicina da UFMG, Universidade Federal de Minas Gerais (J.D., A.R.-O.), and Hermes Pardini Institute (J.D.), Belo Horizonte, Brazil
| | - Georg Brabant
- From the University of Leipzig, Department of Endocrinology and Nephrology (W.F., A.T.), and Leipzig University Medical Center, Integrated Research and Treatment Center Adiposity Diseases (W.F.), Leipzig, the Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg (I.C., M.K., M.F.), and the Clinical Trial Center (U.M.) and Central Laboratory (M.F.), University Hospital Würzburg, Würzburg, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität, Munich (J.S.), the Department of Neurosurgery, University Hospital Hamburg-Eppendorf, Hamburg (J.F.), and Experimental and Clinical Endocrinology, University Hospital Lübeck, Lübeck (G.B.) - all in Germany; the Clinic of Endocrinology, Diabetology and Metabolism (J.R., I.S., B.W., E.C., M.C.-C.) and the Clinical Trial Unit (D.R.V.), Department of Clinical Research, University of Basel and University Hospital Basel, Basel, the Department of Endocrinology, Kantonsspital St. Gallen, St. Gallen (T.D., S.B.), the Department of Endocrinology, Inselspital Bern, Bern (E.C.), the Department of Endocrinology, Luzerner Kantonsspital, Lucerne (C.H., S.F.), and the Division of Endocrinology, Diabetology and Metabolism, Medical University Clinic, Kantonsspital Aarau, Aarau (B.M.) - all in Switzerland; and Faculdade de Medicina da UFMG, Universidade Federal de Minas Gerais (J.D., A.R.-O.), and Hermes Pardini Institute (J.D.), Belo Horizonte, Brazil
| | - Martin Fassnacht
- From the University of Leipzig, Department of Endocrinology and Nephrology (W.F., A.T.), and Leipzig University Medical Center, Integrated Research and Treatment Center Adiposity Diseases (W.F.), Leipzig, the Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg (I.C., M.K., M.F.), and the Clinical Trial Center (U.M.) and Central Laboratory (M.F.), University Hospital Würzburg, Würzburg, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität, Munich (J.S.), the Department of Neurosurgery, University Hospital Hamburg-Eppendorf, Hamburg (J.F.), and Experimental and Clinical Endocrinology, University Hospital Lübeck, Lübeck (G.B.) - all in Germany; the Clinic of Endocrinology, Diabetology and Metabolism (J.R., I.S., B.W., E.C., M.C.-C.) and the Clinical Trial Unit (D.R.V.), Department of Clinical Research, University of Basel and University Hospital Basel, Basel, the Department of Endocrinology, Kantonsspital St. Gallen, St. Gallen (T.D., S.B.), the Department of Endocrinology, Inselspital Bern, Bern (E.C.), the Department of Endocrinology, Luzerner Kantonsspital, Lucerne (C.H., S.F.), and the Division of Endocrinology, Diabetology and Metabolism, Medical University Clinic, Kantonsspital Aarau, Aarau (B.M.) - all in Switzerland; and Faculdade de Medicina da UFMG, Universidade Federal de Minas Gerais (J.D., A.R.-O.), and Hermes Pardini Institute (J.D.), Belo Horizonte, Brazil
| | - Mirjam Christ-Crain
- From the University of Leipzig, Department of Endocrinology and Nephrology (W.F., A.T.), and Leipzig University Medical Center, Integrated Research and Treatment Center Adiposity Diseases (W.F.), Leipzig, the Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg (I.C., M.K., M.F.), and the Clinical Trial Center (U.M.) and Central Laboratory (M.F.), University Hospital Würzburg, Würzburg, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität, Munich (J.S.), the Department of Neurosurgery, University Hospital Hamburg-Eppendorf, Hamburg (J.F.), and Experimental and Clinical Endocrinology, University Hospital Lübeck, Lübeck (G.B.) - all in Germany; the Clinic of Endocrinology, Diabetology and Metabolism (J.R., I.S., B.W., E.C., M.C.-C.) and the Clinical Trial Unit (D.R.V.), Department of Clinical Research, University of Basel and University Hospital Basel, Basel, the Department of Endocrinology, Kantonsspital St. Gallen, St. Gallen (T.D., S.B.), the Department of Endocrinology, Inselspital Bern, Bern (E.C.), the Department of Endocrinology, Luzerner Kantonsspital, Lucerne (C.H., S.F.), and the Division of Endocrinology, Diabetology and Metabolism, Medical University Clinic, Kantonsspital Aarau, Aarau (B.M.) - all in Switzerland; and Faculdade de Medicina da UFMG, Universidade Federal de Minas Gerais (J.D., A.R.-O.), and Hermes Pardini Institute (J.D.), Belo Horizonte, Brazil
| |
Collapse
|
48
|
Stookey JD, Hamer J, Killilea DW. Change in hydration indices associated with an increase in total water intake of more than 0.5 L/day, sustained over 4 weeks, in healthy young men with initial total water intake below 2 L/day. Physiol Rep 2018; 5:5/22/e13356. [PMID: 29150589 PMCID: PMC5704074 DOI: 10.14814/phy2.13356] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 06/09/2017] [Accepted: 06/23/2017] [Indexed: 01/11/2023] Open
Abstract
This secondary data analysis addressed gaps in knowledge about effects of chronic water intake. Longitudinal data from the Adapt Study were used to describe effects of prescribing a sustained increase in water intake relative to baseline, for 4 weeks, on multiple indices of total body water (TBW) flux, regulation, distribution, and volume in five healthy, free-living, young men, with mean total water intake initially below 2 L/day. Indices were measured weekly. Within-person fixed effect models tested for significant changes in indices over time and associations between changes in indices. Agreement between indices was described. Mixed models tested if baseline between-person differences in hydration indices modified changes in indices over time. Body water flux: The half-life of water in the body decreased significantly. Body water regulation: Serum osmolality decreased significantly. Urine anti-diuretic hormone, sodium, potassium, and osmolality decreased significantly. Plasma aldosterone and serum sodium increased significantly. Body water distribution: No significant changes were observed. Body water volume: Saliva osmolality decreased significantly. Body weight increased significantly by a mean ± SEM of 1.8% ± 0.5% from baseline over 4 weeks. Changes in indices were significantly inter-correlated. Agreement between indices changed over 4 weeks. Baseline saliva osmolality significantly modified responses to chronic water intake. The results motivate hypotheses for future studies: Chronic TBW deficit occurs in healthy individuals under daily life conditions and increases chronic disease risk; Sustained higher water intake restores TBW through gradual isotonic retention of potassium and/or sodium; Saliva osmolality is a sensitive and specific index of chronic hydration status.
Collapse
Affiliation(s)
- Jodi D Stookey
- Children's Hospital Oakland Research Institute, Oakland, California
| | - Janice Hamer
- Children's Hospital Oakland Research Institute, Oakland, California
| | - David W Killilea
- Children's Hospital Oakland Research Institute, Oakland, California
| |
Collapse
|
49
|
Baylis PH, Robertson GL. Plasma Vasopressin Response to Hypertonic Saline Infusion to Assess Posterior Pituitary Function. J R Soc Med 2018; 73:255-60. [PMID: 7241442 PMCID: PMC1437414 DOI: 10.1177/014107688007300408] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Hypertonic saline was infused into 11 volunteers to osmotically stimulate vasopressin secretion. A strong positive correlation between plasma arginine vasopressin (PAVP) and plasma osmolality (Pos) was obtained, defined by the function PAVP = 0.63 (Pos – 284), r = +0.80, P < 0.001. The sensitivity of vasopressin secretion to osmotic stimulation was represented by the slope of the expression and the theoretical threshold of vasopressin release by the abscissal intercept. Plasma osmolality at the onset of thirst was 298.5 ± 1.1 mmol/kg. Application of hypertonic saline infusion to 10 polyuric patients clearly separated those with normal osmoregulation of vasopressin secretion from those with cranial diabetes insipidus.
Collapse
|
50
|
Gizowski C, Trudel E, Bourque CW. Central and peripheral roles of vasopressin in the circadian defense of body hydration. Best Pract Res Clin Endocrinol Metab 2017; 31:535-546. [PMID: 29224666 DOI: 10.1016/j.beem.2017.11.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Vasopressin is a neuropeptide synthesized by specific subsets of neurons within the eye and brain. Studies in rats and mice have shown that vasopressin produced by magnocellular neurosecretory cells (MNCs) that project to the neurohypophysis is released into the blood circulation where it serves as an antidiuretic hormone to promote water reabsorption from the kidney. Moreover vasopressin is a neurotransmitter and neuromodulator that contributes to time-keeping within the master circadian clock (i.e. the suprachiasmatic nucleus, SCN) and is also used as an output signal by SCN neurons to direct centrally mediated circadian rhythms. In this chapter, we review recent cellular and network level studies in rodents that have provided insight into how circadian rhythms in vasopressin mediate changes in water intake behavior and renal water conservation that protect the body against dehydration during sleep.
Collapse
Affiliation(s)
- Claire Gizowski
- Center for Research in Neuroscience, Research Institute of the McGill University Health Center, 1650 Cedar Avenue, Montreal, QC, H3G1A4, Canada.
| | - Eric Trudel
- Center for Research in Neuroscience, Research Institute of the McGill University Health Center, 1650 Cedar Avenue, Montreal, QC, H3G1A4, Canada.
| | - Charles W Bourque
- Center for Research in Neuroscience, Research Institute of the McGill University Health Center, 1650 Cedar Avenue, Montreal, QC, H3G1A4, Canada.
| |
Collapse
|