1
|
Naraki K, Ghasemzadeh Rahbardar M, Ajiboye BO, Hosseinzadeh H. The effect of ellagic acid on the metabolic syndrome: A review article. Heliyon 2023; 9:e21844. [PMID: 38027887 PMCID: PMC10661066 DOI: 10.1016/j.heliyon.2023.e21844] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 10/29/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023] Open
Abstract
Objective (s): Metabolic syndrome is a collection of metabolic abnormalities that includes hyperglycemia, dyslipidemia, hypertension, and obesity. Ellagic acid is found in various fruits and vegetables. It has been reported to have several pharmacological properties, such as antibacterial, antifungal, antiviral, anti-inflammatory, hepatoprotective, cardioprotective, chemopreventive, neuroprotective, gastroprotective, and antidiabetic. Our current study aims to shed light on the probable efficiency of ellagic acid in managing metabolic syndrome and its complications. Materials and methods To prepare the present review, the databases or search engines utilized included Scopus, PubMed, Science Direct, and Google Scholar, and relevant articles have been gathered with no time limit until March 2023. Results Several investigations indicated that ellagic acid could be a potent compound for the treatment of many disorders such as diabetes, hypertension, and hyperlipidemia by various mechanisms, including increasing insulin secretion, insulin receptor substrate protein 1 expression, regulating glucose transporter 4, triglyceride, total cholesterol, low-density lipoprotein (LDL), high-density lipoprotein (HDL), attenuating tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), reactive oxygen species (ROS), malondialdehyde (MDA), and oxidative stress in related tissues. Furthermore, ellagic acid ameliorates mitochondrial function, upregulates uncoupling protein 1 (found in brown and white adipose tissues), and regulates blood levels of nitrate/nitrite and vascular relaxations in response to acetylcholine and sodium nitroprusside. Conclusion Ellagic acid can treat or manage metabolic syndrome and associated complications, according to earlier studies. To validate the beneficial effects of ellagic acid on metabolic syndrome, additional preclinical and clinical research is necessary.
Collapse
Affiliation(s)
- Karim Naraki
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Basiru Olaitan Ajiboye
- Phytomedicine and Molecular Toxicology Research Laboratory, Department of Biochemistry, Federal University Oye-Ekiti, Ekiti State, Nigeria
| | - Hossein Hosseinzadeh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
2
|
Lebedeva S, Margaryan A, Smolyarchuk E, Nedorubov A, Materenchuk M, Tonevitsky A, Mutig K. Metabolic effects of vasopressin in pathophysiology of diabetic kidney disease. Front Endocrinol (Lausanne) 2023; 14:1176199. [PMID: 37790608 PMCID: PMC10545091 DOI: 10.3389/fendo.2023.1176199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 08/23/2023] [Indexed: 10/05/2023] Open
Abstract
The diabetic kidney disease (DKD) is the major cause of the chronic kidney disease (CKD). Enhanced plasma vasopressin (VP) levels have been associated with the pathophysiology of DKD and CKD. Stimulation of VP release in DKD is caused by glucose-dependent reset of the osmostat leading to secondary pathophysiologic effects mediated by distinct VP receptor types. VP is a stress hormone exhibiting the antidiuretic action in the kidney along with broad adaptive effects in other organs. Excessive activation of the vasopressin type 2 (V2) receptor in the kidney leads to glomerular hyperfiltration and nephron loss, whereas stimulation of vasopressin V1a or V1b receptors in the liver, pancreas, and adrenal glands promotes catabolic metabolism for energy mobilization, enhancing glucose production and aggravating DKD. Increasing availability of selective VP receptor antagonists opens new therapeutic windows separating the renal and extra-renal VP effects for the concrete applications. Improved understanding of these paradigms is mandatory for further drug design and translational implementation. The present concise review focuses on metabolic effects of VP affecting DKD pathophysiology.
Collapse
Affiliation(s)
- Svetlana Lebedeva
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Arus Margaryan
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Elena Smolyarchuk
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Andrey Nedorubov
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Maria Materenchuk
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | | | - Kerim Mutig
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
- Department of Translational Physiology, Charité-Universitätsmedizin, Berlin, Germany
| |
Collapse
|
3
|
Watts JA, Arroyo JP. Rethinking Vasopressin: New Insights into Vasopressin Signaling and Its Implications. KIDNEY360 2023; 4:1174-1180. [PMID: 37357355 PMCID: PMC10476687 DOI: 10.34067/kid.0000000000000194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/06/2023] [Indexed: 06/27/2023]
Abstract
Vasopressin is a highly conserved peptide hormone that has been traditionally associated with water homeostasis. There is accumulating evidence in both humans and animal models that vasopressin is implicated in the regulation of metabolism. This review focuses on the effects that vasopressin exerts on the regulation of glucose and fatty acids with a particular emphasis on the potential repercussions of metabolic dysregulation in kidney disease.
Collapse
Affiliation(s)
- Jason A. Watts
- Epigenetics and Stem Cell Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Juan Pablo Arroyo
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
4
|
Arroyo JP, Terker AS, Zuchowski Y, Watts JA, Bock F, Meyer C, Luo W, Kapp ME, Gould ER, Miranda AX, Carty J, Jiang M, Vanacore RM, Hammock E, Wilson MH, Zent R, Zhang M, Bhave G, Harris RC. Kidney collecting duct cells make vasopressin in response to NaCl-induced hypertonicity. JCI Insight 2022; 7:e161765. [PMID: 36326835 PMCID: PMC9869977 DOI: 10.1172/jci.insight.161765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
Vasopressin has traditionally been thought to be produced by the neurohypophyseal system and then released into the circulation where it regulates water homeostasis. The questions of whether vasopressin could be produced outside of the brain and if the kidney could be a source of vasopressin are raised by the syndrome of inappropriate antidiuretic hormone secretion (vasopressin). We found that mouse and human kidneys expressed vasopressin mRNA. Using an antibody that detects preprovasopressin, we found that immunoreactive preprovasopressin protein was found in mouse and human kidneys. Moreover, we found that murine collecting duct cells made biologically active vasopressin, which increased in response to NaCl-mediated hypertonicity, and that water restriction increased the abundance of kidney-derived vasopressin mRNA and protein expression in mouse kidneys. Thus, we provide evidence of biologically active production of kidney-derived vasopressin in kidney tubular epithelial cells.
Collapse
Affiliation(s)
- Juan Pablo Arroyo
- Division of Nephrology and Hypertension, Department of Medicine, and
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Andrew S. Terker
- Division of Nephrology and Hypertension, Department of Medicine, and
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Yvonne Zuchowski
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Jason A. Watts
- Epigenetics and Stem Cell Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, North Carolina, USA
| | - Fabian Bock
- Division of Nephrology and Hypertension, Department of Medicine, and
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Cameron Meyer
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Wentian Luo
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Meghan E. Kapp
- Division of Renal Pathology, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology, Case Western Reserve University, University Hospitals, Cleveland, Ohio, USA
| | - Edward R. Gould
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Adam X. Miranda
- Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Joshua Carty
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Ming Jiang
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Roberto M. Vanacore
- Division of Nephrology and Hypertension, Department of Medicine, and
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Elizabeth Hammock
- Department of Psychology, Florida State University, Tallahassee, Florida, USA
| | - Matthew H. Wilson
- Division of Nephrology and Hypertension, Department of Medicine, and
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Roy Zent
- Division of Nephrology and Hypertension, Department of Medicine, and
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Psychology, Florida State University, Tallahassee, Florida, USA
| | - Mingzhi Zhang
- Division of Nephrology and Hypertension, Department of Medicine, and
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Gautam Bhave
- Division of Nephrology and Hypertension, Department of Medicine, and
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Raymond C. Harris
- Division of Nephrology and Hypertension, Department of Medicine, and
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
5
|
Mitrosz-Gołębiewska K, Rydzewska-Rosołowska A, Kakareko K, Zbroch E, Hryszko T. Water - A life-giving toxin - A nephrological oxymoron. Health consequences of water and sodium balance disorders. A review article. Adv Med Sci 2022; 67:55-65. [PMID: 34979423 DOI: 10.1016/j.advms.2021.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 08/24/2021] [Accepted: 12/15/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND This article aims to reveal misconceptions about methods of assessment of hydration status and impact of the water disorders on the progression of kidney disease or renal dysfunction. MATERIALS AND METHODS The PubMed database was searched for reviews, meta-analyses and original articles on hydration, volume depletion, fluid overload and diagnostic methods of hydration status, which were published in English. RESULTS Based on the results of available literature the relationship between the amount of fluid consumed, and the rate of progression of chronic kidney disease, autosomal dominant polycystic kidney disease, and kidney stones disease was discussed. Selected aspects of the assessment of the hydration level in clinical practice based on physical examination, laboratory tests, and imaging are presented. The subject of in-hospital fluid therapy is discussed. Based on available randomized studies, an attempt was made to assess, which fluids should be selected for intravenous treatment. CONCLUSIONS There is some evidence for the beneficial effect of increased water intake in preventing recurrent cystitis and kidney stones, but there are still no convincing data for chronic kidney disease and autosomal dominant polycystic kidney disease. Further studies are needed to clarify the aforementioned issues and establish a reliable way to assess the volemia and perform suitable fluid therapy.
Collapse
Affiliation(s)
- Katarzyna Mitrosz-Gołębiewska
- 2nd Department of Nephrology and Hypertension with Dialysis Unit, Medical University of Bialystok, Bialystok, Poland.
| | - Alicja Rydzewska-Rosołowska
- 2nd Department of Nephrology and Hypertension with Dialysis Unit, Medical University of Bialystok, Bialystok, Poland
| | - Katarzyna Kakareko
- 2nd Department of Nephrology and Hypertension with Dialysis Unit, Medical University of Bialystok, Bialystok, Poland
| | - Edyta Zbroch
- Department of Internal Medicine and Hypertension, Medical University od Bialystok, Bialystok, Poland
| | - Tomasz Hryszko
- 2nd Department of Nephrology and Hypertension with Dialysis Unit, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
6
|
Kourtidou C, Stangou M, Marinaki S, Tziomalos K. Novel Cardiovascular Risk Factors in Patients with Diabetic Kidney Disease. Int J Mol Sci 2021; 22:11196. [PMID: 34681856 PMCID: PMC8537513 DOI: 10.3390/ijms222011196] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/09/2021] [Accepted: 10/15/2021] [Indexed: 02/06/2023] Open
Abstract
Patients with diabetic kidney disease (DKD) are at very high risk for cardiovascular events. Only part of this increased risk can be attributed to the presence of diabetes mellitus (DM) and to other DM-related comorbidities, including hypertension and obesity. The identification of novel risk factors that underpin the association between DKD and cardiovascular disease (CVD) is essential for risk stratification, for individualization of treatment and for identification of novel treatment targets.In the present review, we summarize the current knowledge regarding the role of emerging cardiovascular risk markers in patients with DKD. Among these biomarkers, fibroblast growth factor-23 and copeptin were studied more extensively and consistently predicted cardiovascular events in this population. Therefore, it might be useful to incorporate them in risk stratification strategies in patients with DKD to identify those who would possibly benefit from more aggressive management of cardiovascular risk factors.
Collapse
Affiliation(s)
- Christodoula Kourtidou
- First Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA Hospital, 54636 Thessaloniki, Greece;
| | - Maria Stangou
- Department of Nephrology, Medical School, Aristotle University of Thessaloniki, Hippokration Hospital, 54642 Thessaloniki, Greece;
| | - Smaragdi Marinaki
- Department of Nephrology and Renal Transplantation, Medical School, National and Kapodistrian University of Athens, Laiko Hospital, 11527 Athens, Greece;
| | - Konstantinos Tziomalos
- First Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA Hospital, 54636 Thessaloniki, Greece;
| |
Collapse
|
7
|
Valenti G, Tamma G. The vasopressin-aquaporin-2 pathway syndromes. HANDBOOK OF CLINICAL NEUROLOGY 2021; 181:249-259. [PMID: 34238461 DOI: 10.1016/b978-0-12-820683-6.00018-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Vasopressin is the key hormone involved in water conservation and regulation of water balance, essential for life. In the renal collecting duct, vasopressin binds to the V2 receptor, increasing water permeability through activation of aquaporin-2 redistribution to the luminal membrane. This mechanism promotes rapid water reabsorption, important for immediate survival; however, only recently it has become clear that long-term adverse effects are associated with alterations of the vasopressin-aquaporin-2 pathway, leading to several syndromes associated with water balance disorders. The kidney resistance to the vasopressin action may cause severe dehydration for patients and, conversely, nonosmotic release of vasopressin is associated with water retention and increasing the circulatory blood volume. This chapter discusses the relevance of the altered vasopressin-aquaporin-2 pathway in some diseases associated with water balance disorders, including congenital nephrogenic diabetes insipidus, syndrome of inappropriate secretion of antidiuretic hormone, nephrogenic syndrome of inappropriate antidiuresis, and autosomal dominant polycystic kidney disease. The emerging picture suggests that targeting the vasopressin-AQP2 axis can provide therapeutic benefits in those patients.
Collapse
Affiliation(s)
- Giovanna Valenti
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, Bari, Italy.
| | - Grazia Tamma
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, Bari, Italy
| |
Collapse
|
8
|
Wang HW, Jiang MY. Higher volume of water intake is associated with lower risk of albuminuria and chronic kidney disease. Medicine (Baltimore) 2021; 100:e26009. [PMID: 34011099 PMCID: PMC8137104 DOI: 10.1097/md.0000000000026009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/30/2021] [Indexed: 01/05/2023] Open
Abstract
Increased water intake correlated to lower vasopressin level and may benefit kidney function. However, results of previous studies were conflicted and inconclusive. We aimed to investigate the association between water intake and risk of chronic kidney disease (CKD) and albuminuria.In this cross-sectional study, the study population were adult participants of 2011-2012 National Health and Nutrition Examination Survey (NHANES) whose estimated glomerular filtration rate (eGFR) were ≥30 ml/min/1.73 m2. Data of water intake were obtained from the NHANES 24-h dietary recall questionnaire. Participants were divided into three groups based on volume of water intake: <500 (low, n = 1589), ≥500 to <1200 (moderate, n = 1359), and ≥1200 ml/day (high, n = 1685). CKD was defined as eGFR <60 ml/min/1.73 m2, and albuminuria as albumin-to-creatinine ratio (ACR) ≥30 mg/g.Our results showed that 377 out of 4633 participants had CKD; the prevalence inversely correlated to volume of water intake: 10.7% in low, 8.2% in moderate, and 5.6% in high intake groups (P < .001). Prevalence of albuminuria was also lower in high (9.5%) compared with moderate (12.8%) and low intake groups (14.1%), P < .001. Additionally, water intake positively correlated to eGFR and negatively correlated to urinary ACR, as well as plasma and urine osmolality. Multivariable logistic regression showed that low water intake group had higher risk of CKD (OR 1.35, 95% CI 1.01-1.82) and albuminuria when compared to high water intake group (OR 1.42, 95% CI 1.13-1.79).In conclusion, increased water intake was associated lower risk of CKD and albuminuria. Meticulous studies are needed to elucidate the underlying mechanisms.
Collapse
|
9
|
Horie S, Muto S, Kawano H, Okada T, Shibasaki Y, Nakajima K, Ibuki T. Preservation of kidney function irrelevant of total kidney volume growth rate with tolvaptan treatment in patients with autosomal dominant polycystic kidney disease. Clin Exp Nephrol 2021; 25:467-478. [PMID: 33471240 PMCID: PMC8038960 DOI: 10.1007/s10157-020-02009-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 12/02/2020] [Indexed: 11/29/2022]
Abstract
Background Tolvaptan slowed the rates of total kidney volume (TKV) growth and renal function decline over a 3-year period in patients with autosomal dominant polycystic kidney disease (ADPKD) enrolled in the Tolvaptan Efficacy and Safety in Management of Autosomal Dominant Polycystic Kidney Disease and Its Outcomes (TEMPO) 3:4 trial (NCT00428948). In this post hoc analysis of Japanese patients from TEMPO 3:4, we evaluated whether the effects of tolvaptan on TKV and on renal function are interrelated. Methods One hundred and forty-seven Japanese patients from TEMPO 3:4 were included in this analysis (placebo, n = 55; tolvaptan, n = 92). Tolvaptan-treated patients were stratified into the responder group (n = 37), defined as tolvaptan-treated patients with a net decrease in TKV from baseline to year 3, and the non-responder group (n = 55), defined as tolvaptan-treated patients with a net increase in TKV. Results Mean changes during follow-up in the placebo, responder, and non-responder groups were 16.99%, − 8.33%, and 13.95%, respectively, for TKV and − 12.61, − 8.47, and − 8.58 mL/min/1.73 m2, respectively, for estimated glomerular filtration rate (eGFR). Compared with the placebo group, eGFR decline was significantly slowed in both the responder and non-responder groups (P < 0.05). Conclusion Tolvaptan was effective in slowing eGFR decline, regardless of TKV response, over 3 years in patients with ADPKD in Japan. Treatment with tolvaptan may have beneficial effects on slowing of renal function decline even in patients who have not experienced a reduction in the rate of TKV growth by treatment with tolvaptan. Supplementary Information The online version contains supplementary material available at 10.1007/s10157-020-02009-0.
Collapse
Affiliation(s)
- Shigeo Horie
- Department of Urology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan. .,Department of Advanced Informatics for Genetic Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| | - Satoru Muto
- Department of Urology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.,Department of Advanced Informatics for Genetic Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Haruna Kawano
- Department of Urology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.,Department of Advanced Informatics for Genetic Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tadashi Okada
- Department of Clinical Development, Otsuka Pharmaceutical Co., Ltd, Osaka, Japan
| | | | - Koji Nakajima
- Medical Affairs, Otsuka Pharmaceutical Co., Ltd, Tokyo, Japan
| | - Tatsuki Ibuki
- Medical Affairs, Otsuka Pharmaceutical Co., Ltd, Tokyo, Japan
| |
Collapse
|
10
|
El Dayem SMA, Battah AA, El Bohy AEM, Yousef RN, Talaat A. Copeptin as a Biomarker of Atherosclerosis in Type 1 Diabetic Patients. Open Access Maced J Med Sci 2020; 7:3975-3978. [PMID: 33318769 PMCID: PMC7061391 DOI: 10.3889/oamjms.2019.643] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 11/05/2022] Open
Abstract
AIM: To evaluate copeptin as an early marker of atherosclerosis in adolescent type 1 diabetics. METHODS: Sixty-two type 1 diabetic patients and 50 healthy volunteers were enrolled in the study. Serum copeptin, glycosylated haemoglobin (HbA1c), lipid profile, oxidised low-density lipoprotein (OxLDL), urinary albumin/creatinine ratio, carotid intimal medial thickness (cIMT), aortic intimal medial thickness (aIMT) and resistivity index were assessed for all participants in the study. RESULTS: HbA1c, albumin/creatinine ratio, lipid profile, OxlDL, copeptin, cIMT and aIMT were significantly higher in diabetic patients. Copeptin was higher in patients with positive cIMT and aIMT. Copeptin correlated with cIMT and aIMT. Stepwise multiple regression analysis found that copeptin correlated with aIMT. ROC curve showed that copeptin had 100 % specificity with aIMT and cIMT and 95.2 and 60,7 sensitivity with aIMT and cIMT respectively. CONCLUSION: Copeptin can be used as a marker for early detection of atherosclerosis of type 1 diabetic patients.
Collapse
Affiliation(s)
| | - Ahmed A Battah
- Critical Care Department, Cairo University, Cairo, Egypt
| | | | | | - Ahmed Talaat
- Pediatrics Department, National Research Centre, Cairo, Egypt
| |
Collapse
|
11
|
Pavlov TS, Palygin O, Isaeva E, Levchenko V, Khedr S, Blass G, Ilatovskaya DV, Cowley AW, Staruschenko A. NOX4-dependent regulation of ENaC in hypertension and diabetic kidney disease. FASEB J 2020; 34:13396-13408. [PMID: 32799394 DOI: 10.1096/fj.202000966rr] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/17/2020] [Accepted: 07/20/2020] [Indexed: 12/11/2022]
Abstract
NADPH oxidase 4 (NOX4) is the most abundant NOX isoform in the kidney; however, its importance for renal function has only recently emerged. The NOX4-dependent pathway regulates many factors essential for proper sodium handling in the distal nephron. However, the functional significance of this pathway in the control of sodium reabsorption during the initiation of chronic kidney disease is not established. The goal of this study was to test Nox4-dependent ENaC regulation in two models: SS hypertension and STZ-induced type 1 diabetes. First, we showed that genetic ablation of Nox4 in Dahl salt-sensitive (SS) rat attenuated a high-salt (HS)-induced increase in epithelial Na+ channel (ENaC) activity in the cortical collecting duct. We also found that H2 O2 upregulated ENaC activity, and H2 O2 production was reduced in both the renal cortex and medulla in SSNox4-/- rats fed an HS diet. Second, in the streptozotocin model of hyperglycemia-induced renal injury ENaC activity in hyperglycemic animals was elevated in SS but not SSNox4-/- rats. NaCl cotransporter (NCC) expression was increased compared to healthy controls, while expression values between SS and SSNox4-/- groups were similar. These data emphasize a critical contribution of the NOX4-mediated pathway in maladaptive upregulation of ENaC-mediated sodium reabsorption in the distal nephron in the conditions of HS- and hyperglycemia-induced kidney injury.
Collapse
Affiliation(s)
- Tengis S Pavlov
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA.,Division of Hypertension and Vascular Research, Henry Ford Health System, Detroit, MI, USA
| | - Oleg Palygin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Elena Isaeva
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Sherif Khedr
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA.,Department of Physiology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Gregory Blass
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Allen W Cowley
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Alexander Staruschenko
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA.,Clement J. Zablocki VA Medical Center, Milwaukee, WI, USA
| |
Collapse
|
12
|
Perrier ET, Armstrong LE, Bottin JH, Clark WF, Dolci A, Guelinckx I, Iroz A, Kavouras SA, Lang F, Lieberman HR, Melander O, Morin C, Seksek I, Stookey JD, Tack I, Vanhaecke T, Vecchio M, Péronnet F. Hydration for health hypothesis: a narrative review of supporting evidence. Eur J Nutr 2020; 60:1167-1180. [PMID: 32632658 PMCID: PMC7987589 DOI: 10.1007/s00394-020-02296-z] [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: 02/24/2020] [Accepted: 05/28/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE An increasing body of evidence suggests that excreting a generous volume of diluted urine is associated with short- and long-term beneficial health effects, especially for kidney and metabolic function. However, water intake and hydration remain under-investigated and optimal hydration is poorly and inconsistently defined. This review tests the hypothesis that optimal chronic water intake positively impacts various aspects of health and proposes an evidence-based definition of optimal hydration. METHODS Search strategy included PubMed and Google Scholar using relevant keywords for each health outcome, complemented by manual search of article reference lists and the expertise of relevant practitioners for each area studied. RESULTS The available literature suggest the effects of increased water intake on health may be direct, due to increased urine flow or urine dilution, or indirect, mediated by a reduction in osmotically -stimulated vasopressin (AVP). Urine flow affects the formation of kidney stones and recurrence of urinary tract infection, while increased circulating AVP is implicated in metabolic disease, chronic kidney disease, and autosomal dominant polycystic kidney disease. CONCLUSION In order to ensure optimal hydration, it is proposed that optimal total water intake should approach 2.5 to 3.5 L day-1 to allow for the daily excretion of 2 to 3 L of dilute (< 500 mOsm kg-1) urine. Simple urinary markers of hydration such as urine color or void frequency may be used to monitor and adjust intake.
Collapse
Affiliation(s)
- Erica T Perrier
- Health, Hydration & Nutrition Science, Danone Research, Route Départementale 128, 91767, Palaiseau cedex, France.
| | - Lawrence E Armstrong
- Department of Kinesiology, University of Connecticut, Storrs, CT, USA.,Hydration & Nutrition, LLC, Newport News, VA, USA
| | - Jeanne H Bottin
- Health, Hydration & Nutrition Science, Danone Research, Route Départementale 128, 91767, Palaiseau cedex, France
| | - William F Clark
- London Health Sciences Centre and Western University, London, ON, Canada
| | - Alberto Dolci
- Health, Hydration & Nutrition Science, Danone Research, Route Départementale 128, 91767, Palaiseau cedex, France
| | - Isabelle Guelinckx
- Health, Hydration & Nutrition Science, Danone Research, Route Départementale 128, 91767, Palaiseau cedex, France
| | - Alison Iroz
- Health, Hydration & Nutrition Science, Danone Research, Route Départementale 128, 91767, Palaiseau cedex, France
| | - Stavros A Kavouras
- College of Health Solutions and Hydration Science Lab, Arizona State University, Phoenix, AZ, USA
| | - Florian Lang
- Department of Physiology, Eberhard Karls University, Tübingen, Germany
| | | | - Olle Melander
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Clementine Morin
- Health, Hydration & Nutrition Science, Danone Research, Route Départementale 128, 91767, Palaiseau cedex, France
| | - Isabelle Seksek
- Health, Hydration & Nutrition Science, Danone Research, Route Départementale 128, 91767, Palaiseau cedex, France
| | - Jodi D Stookey
- Children's Hospital Oakland Research Institute, Oakland, CA, USA
| | - Ivan Tack
- Explorations Fonctionnelles Physiologiques, Hôpital Rangueil, Toulouse, France
| | - Tiphaine Vanhaecke
- Health, Hydration & Nutrition Science, Danone Research, Route Départementale 128, 91767, Palaiseau cedex, France
| | - Mariacristina Vecchio
- Health, Hydration & Nutrition Science, Danone Research, Route Départementale 128, 91767, Palaiseau cedex, France
| | - François Péronnet
- École de Kinésiologie et des Sciences de l'activité Physique, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
13
|
Tanaka S, Nakano T, Tokumoto M, Masutani K, Tsuchimoto A, Ooboshi H, Kitazono T. Estimated plasma osmolarity and risk of end-stage kidney disease in patients with IgA nephropathy. Clin Exp Nephrol 2020; 24:910-918. [PMID: 32594371 DOI: 10.1007/s10157-020-01919-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 06/07/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND Several experimental studies have indicated that increased plasma osmolarity caused by recurrent dehydration is involved in kidney injury via a mechanism, mediated by vasopressin secretion and activation of the aldose reductase pathway. Epidemiologic evidence linking increased plasma osmolarity and the onset of end-stage kidney disease (ESKD), in patients with primary glomerulonephritis, is lacking. METHODS We retrospectively examined 663 patients with IgA nephropathy (IgAN) diagnosed by kidney biopsy and evaluated the association between estimated plasma osmolarity and ESKD prevalence, using a Cox proportional hazards model. RESULTS During follow-up (median 80.4 months; interquartile range 22.2-120.1), 73 patients developed ESKD. In a baseline survey, plasma osmolarity was correlated negatively with the mean value of the estimated glomerular filtration rate, but correlated positively with the mean value of urinary protein excretion, systolic blood pressure, and pathologic severity of extracapillary proliferation, in addition to tissue fibrosis and sclerosis. The incidence rate of ESKD increased linearly with increase in plasma osmolarity (P < 0.05 for trend). In multivariate analyses, plasma osmolarity was an independent risk factor for ESKD (hazard ratio for each increment of 5 mOsm/kg in plasma osmolarity 1.56; 95% confidence interval 1.18-2.07) even after adjustment for potential confounders. CONCLUSIONS Increased plasma osmolarity was associated significantly with an increased risk of ESKD in patients with IgAN. Maintenance of plasma osmolarity by appropriate control of the balance between salt and water may contribute to kidney protection.
Collapse
Affiliation(s)
- Shigeru Tanaka
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Toshiaki Nakano
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Masanori Tokumoto
- Department of Internal Medicine, Fukuoka Dental College, Fukuoka, Japan
| | - Kosuke Masutani
- Department of Nephrology and Rheumatology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Akihiro Tsuchimoto
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hiroaki Ooboshi
- Department of Internal Medicine, Fukuoka Dental College, Fukuoka, Japan
| | - Takanari Kitazono
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
14
|
Sakai K, Yamazaki O, Ishizawa K, Tamura Y, Wang Q, Ueno M, Hayama Y, Fujigaki Y, Shibata S. Upregulation of renal Na-K-2Cl cotransporter 2 in obese diabetes mellitus via a vasopressin receptor 2-dependent pathway. Biochem Biophys Res Commun 2020; 524:710-715. [PMID: 32035616 DOI: 10.1016/j.bbrc.2020.01.142] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 01/26/2020] [Indexed: 11/30/2022]
Abstract
Na-K-2Cl cotransporter 2 (NKCC2) in thick ascending limb (TAL) in the kidney plays a central role in tubuloglomerular feedback (TGF) system by sensing NaCl delivery to the distal tubules. Although accumulating data indicate that dysregulated TGF contributes to the progression of diabetic complications, the regulation of NKCC2 in diabetes mellitus (DM) remains unclear. We here show that NKCC2 is overactivated via a vasopressin receptor 2 (V2R)-dependent mechanism in db/db mice, a mouse model of obese DM. Compared with db/+ mice, we found that both aquaporin 2 and NKCC2 levels were significantly increased in the kidney in db/db mice. Immunohistochemical analysis of V2R and NKCC2 in the kidney demonstrated that V2R is present in the TAL, as well as in the collecting duct. Moreover, the administration of tolvaptan, a selective V2R antagonist, sharply decreased aquaporin 2 and NKCC2 in db/db mice, confirming the causal role of V2R signaling in NKCC2 induction in this model. Although tolvaptan reduced aquaporin 2 abundance also in db/+ mice, its effect on NKCC2 was modest compared with db/db mice. In total kidney lysates, uromodulin expression was not altered between db/+ and db/db mice, suggesting that V2R signaling alters NKCC2 without altering uromodulin levels. These data implicate the dysregulation of NKCC2 in the pathophysiology of type 2 DM, and underscore the complex nature of fluid volume disorders in diabetic kidney disease.
Collapse
Affiliation(s)
- Kazuhiro Sakai
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, 173-8605, Japan
| | - Osamu Yamazaki
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, 173-8605, Japan
| | - Kenichi Ishizawa
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, 173-8605, Japan
| | - Yoshifuru Tamura
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, 173-8605, Japan
| | - Qin Wang
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, 173-8605, Japan; Department of Nephrology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Masaki Ueno
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, 173-8605, Japan
| | - Yuto Hayama
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, 173-8605, Japan
| | - Yoshihide Fujigaki
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, 173-8605, Japan
| | - Shigeru Shibata
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, 173-8605, Japan.
| |
Collapse
|
15
|
Lytvyn Y, Bjornstad P, van Raalte DH, Heerspink HL, Cherney DZI. The New Biology of Diabetic Kidney Disease-Mechanisms and Therapeutic Implications. Endocr Rev 2020; 41:5601424. [PMID: 31633153 PMCID: PMC7156849 DOI: 10.1210/endrev/bnz010] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 10/17/2019] [Indexed: 02/07/2023]
Abstract
Diabetic kidney disease remains the most common cause of end-stage kidney disease in the world. Despite reductions in incidence rates of myocardial infarction and stroke in people with diabetes over the past 3 decades, the risk of diabetic kidney disease has remained unchanged, and may even be increasing in younger individuals afflicted with this disease. Accordingly, changes in public health policy have to be implemented to address the root causes of diabetic kidney disease, including the rise of obesity and diabetes, in addition to the use of safe and effective pharmacological agents to prevent cardiorenal complications in people with diabetes. The aim of this article is to review the mechanisms of pathogenesis and therapies that are either in clinical practice or that are emerging in clinical development programs for potential use to treat diabetic kidney disease.
Collapse
Affiliation(s)
- Yuliya Lytvyn
- Department of Medicine, Division of Nephrology, Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Petter Bjornstad
- Department of Medicine, Division of Nephrology, Department of Pediatrics, Section of Endocrinology, University of Colorado School of Medicine, Aurora, Colorado
| | - Daniel H van Raalte
- Diabetes Center, Department of Internal Medicine, VU University Medical Center, Netherlands
| | - Hiddo L Heerspink
- The George Institute for Global Health, Sydney, Australia.,Department of Clinical Pharmacology, University of Groningen, Groningen, Netherlands
| | - David Z I Cherney
- Department of Medicine, Division of Nephrology, Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
16
|
Butler-Dawson J, Dally M, Johnson RJ, Johnson EC, Krisher L, Sánchez-Lozada LG, Griffin BR, Brindley S, Newman LS. Association of Copeptin, a Surrogate Marker of Arginine Vasopressin, with Decreased Kidney Function in Sugarcane Workers in Guatemala. ANNALS OF NUTRITION AND METABOLISM 2020; 76:30-36. [PMID: 32172243 DOI: 10.1159/000506619] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/16/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Vasopressin is elevated in response to heat and dehydration and has been postulated to have a role in the chronic kidney disease of unknown origin being observed in Central America. The aims of this study were to examine whether the vasopressin pathway, as measured by copeptin, is associated with the presence of kidney dysfunction, and to examine whether higher fluid intake is associated with lower circulating copeptin and thereby preserves kidney health among sugarcane workers exposed to hot conditions. METHODS Utilizing a longitudinal study of 105 workers in Guatemala, we examined relationships between hydration indices, plasma copeptin concentrations, and kidney function markers at 3 times during the 6-month harvest. We also examined whether baseline copeptin concentrations increased the odds of developing an estimated glomerular filtration rate (eGFR) <60 mL/min/1.73 m2. RESULTS Copeptin concentrations were positively associated with serum creatinine (β 1.41, 95% CI 0.88-2.03) and negatively associated with eGFR (β -1.07, 95% CI -1.43 to -0.70). In addition, as workers improved their hydration (measured by increases in fluid balance), copeptin concentrations were reduced, and this reduction was associated with an improvement in kidney function. CONCLUSIONS Results suggest that copeptin should be studied as a potential prognostic biomarker.
Collapse
Affiliation(s)
- Jaime Butler-Dawson
- Center for Health, Work, and Environment, Colorado School of Public Health, University of Colorado, Aurora, Colorado, USA, .,Department of Environmental and Occupational Health, Colorado School of Public Health, University of Colorado, Aurora, Colorado, USA, .,Colorado Consortium on Climate Change and Human Health, University of Colorado, Aurora, Colorado, USA,
| | - Miranda Dally
- Center for Health, Work, and Environment, Colorado School of Public Health, University of Colorado, Aurora, Colorado, USA.,Department of Environmental and Occupational Health, Colorado School of Public Health, University of Colorado, Aurora, Colorado, USA.,Colorado Consortium on Climate Change and Human Health, University of Colorado, Aurora, Colorado, USA
| | - Richard J Johnson
- Colorado Consortium on Climate Change and Human Health, University of Colorado, Aurora, Colorado, USA.,Division of Renal Diseases and Hypertension, Department of Medicine, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Evan C Johnson
- Division of Kinesiology and Health, University of Wyoming, Laramie, Wyoming, USA
| | - Lyndsay Krisher
- Center for Health, Work, and Environment, Colorado School of Public Health, University of Colorado, Aurora, Colorado, USA.,Colorado Consortium on Climate Change and Human Health, University of Colorado, Aurora, Colorado, USA
| | | | - Benjamin R Griffin
- Division of Renal Diseases and Hypertension, Department of Medicine, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Stephen Brindley
- Department of Environmental and Occupational Health, Colorado School of Public Health, University of Colorado, Aurora, Colorado, USA
| | - Lee S Newman
- Center for Health, Work, and Environment, Colorado School of Public Health, University of Colorado, Aurora, Colorado, USA.,Department of Environmental and Occupational Health, Colorado School of Public Health, University of Colorado, Aurora, Colorado, USA.,Colorado Consortium on Climate Change and Human Health, University of Colorado, Aurora, Colorado, USA.,Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, School of Medicine, Aurora, Colorado, USA.,Department of Epidemiology, Colorado School of Public Health, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
17
|
Li X, Gao Z, Xu L, Li B, Gao H. Over-expression of arginine vasopressin in magnocellular neurosecretory cells of hypothalamus and its potential relationship with development of diabetic nephropathy. Arch Med Sci 2020; 16:1130-1139. [PMID: 32864002 PMCID: PMC7444698 DOI: 10.5114/aoms.2020.92402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 01/27/2018] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION We aimed to assess our hypothesis that the expression changes of arginine vasopressin (AVP) in the magnocellular neurosecretory cells (MNCs) of hypothalamus and V2 receptor for AVP (RVP) in kidney may contribute to the pathogenesis of diabetic nephropathy (DN). MATERIAL AND METHODS Twenty-five male Wistar rats were randomly assigned to the control group and the diabetes mellitus (DM) group. Periodic acid-Schiff (PAS) staining and electron microscopy were used for morphological studies. Immunohistochemical staining for glial fibrillary acidic protein (GFAP) is standard for visualization of reactive astrocytes in the hypothalamus. Hypothalamus was used for immunofluorescence of AVP. Kidney was used for immunohistochemical staining of RVP. Quantitative real-time reverse transcriptase polymerase chain reaction (qRT-PCR) was used for quantitative determinations of AVP mRNA in hypothalamus and RVP mRNA in kidney. Western blot was used to measure the protein expression of AVP in hypothalamus and RVP in kidney. RESULTS Morphological studies showed abnormalities in kidney and hypothalamus in the DM group. The number of neurons and the gray value of astrocytes in hypothalamus in the DM group were markedly decreased. The expression level of AVP in hypothalamus and the expression level of RVP in kidney of DM rats were significantly increased. The positive correlations between the proteinuria and expression (mRNA and protein) of AVP, proteinuria and expression (mRNA and protein) of RVP, and the expression of AVP and RVP levels were found. CONCLUSIONS AVP was upregulated in the MNCs of hypothalamus and RVP was upregulated in kidney in streptozotocin-induced DM rats, indicating their potential roles in the development of DN.
Collapse
Affiliation(s)
- Xianhua Li
- Department of Nephrology, Qi Lu Hospital of Shandong University, Jinan, China
| | - Zhaoli Gao
- Department of Geriatrics, Qi Lu Hospital of Shandong University, Jinan, China
| | - Ling Xu
- Department of Geriatrics, Qi Lu Hospital of Shandong University, Jinan, China
| | - Baoying Li
- Department of Geriatrics, Qi Lu Hospital of Shandong University, Jinan, China
| | - Haiqing Gao
- Department of Geriatrics, Qi Lu Hospital of Shandong University, Jinan, China
| |
Collapse
|
18
|
Enhörning S, Christensson A, Melander O. Plasma copeptin as a predictor of kidney disease. Nephrol Dial Transplant 2019; 34:74-82. [PMID: 29471407 PMCID: PMC6322441 DOI: 10.1093/ndt/gfy017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 01/06/2018] [Indexed: 12/16/2022] Open
Abstract
Background Plasma copeptin, a marker of vasopressin, is associated with renal function decline in the general population. Our aim was to study the links between elevated copeptin and future risk of kidney disease. Methods Copeptin was measured in a sample of the Malmö Preventive Project (MPP) Reinvestigation (n = 5158) and in the Malmö Diet and Cancer Cardiovascular Cohort (MDC-CC) (n = 5162). According to national registers, 89 subjects in MPP and 180 in MDC-CC developed chronic kidney disease (CKD) during follow-up (8.7 and 19.6 years, respectively). Results After multivariate adjustment (gender, age, body mass index, smoking status, estimated glomerular filtration rate, prevalent diabetes, systolic blood pressure and prevalent antihypertensive treatment), copeptin (beta-coefficient per 1 standard deviation increment of ln copeptin) was independently associated with increased risk of CKD during follow-up in both cohorts (MPP: (HR) 1.46, 95% confidence interval (CI) 1.18–1.80, P < 0.001; MDC-CC: HR 1.25, 95% CI 1.02–1.54, P = 0.03) among subjects free from prevalent kidney disease at baseline. Furthermore, in MPP, elevated copeptin predicted a specified diagnosis of kidney disease other than CKD (HR 1.31, 95% CI 1.08–1.59, P = 0.006) after multivariate adjustment. In a corresponding analysis in MDC-CC, copeptin was associated with a 10% increased risk, which, however, was non-significant (P = 0.25). A meta-analysis of the MPP and MDC-CC data showed significant association between elevated copeptin and a specified diagnosis of kidney disease other than CKD (HR 1.18, 95% CI 1.05–1.34, P = 0.008). Conclusion An increased level of copeptin independently predicts development of both CKD and other specified kidney diseases, suggesting that copeptin can be used to identify individuals at risk for kidney disease development.
Collapse
Affiliation(s)
- Sofia Enhörning
- Department of Endocrinology, Skåne University Hospital, Lund, Sweden.,Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Anders Christensson
- Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Nephrology, Skåne University Hospital, Lund, Sweden
| | - Olle Melander
- Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Internal Medicine, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
19
|
Gonzalez AA, Salinas-Parra N, Cifuentes-Araneda F, Reyes-Martinez C. Vasopressin actions in the kidney renin angiotensin system and its role in hypertension and renal disease. VITAMINS AND HORMONES 2019; 113:217-238. [PMID: 32138949 DOI: 10.1016/bs.vh.2019.09.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Vasopressin, also named antidiuretic hormone (ADH), arginine vasopressin (AVP) is the main hormone responsible for water maintenance in the body through the antidiuretic actions in the kidney. The posterior pituitary into the blood releases vasopressin formed in the hypothalamus. Hypothalamic osmotic neurons are responsible to initiate the cascade for AVP actions. The effects of AVP peptide includes activation of V2 receptors which stimulate the formation of cyclic AMP (cAMP) and phosphorylation of water channels aquaporin 2 (AQP2) in the collecting duct. AVP also has vasoconstrictor effects through V1a receptors in the vasculature, while V1b is found in the nervous system. V1a and b receptors increases intracellular Ca2+ while activation of V2 receptors of signaling pathways are related to cAMP-dependent phosphorylation in kidney collecting ducts acting in coordination to stimulate water and electrolyte homeostasis. AVP potentiate formation of intratubular angiotensin II (Ang II) through V2 receptors-dependent distal tubular renin formation, contributing to Na+ reabsorption. On the same way, Ang II receptors are able to potentiate the effects of V2-dependent stimulation of AQP2 abundance in the plasma membrane. The role of AVP in hypertension and renal disease has been demonstrated in pathological states with the involvement of V2 receptors in the progression of kidney damage in diabetes and also on the stimulation of intracellular pathways linked to the development of polycystic kidney.
Collapse
Affiliation(s)
- Alexis A Gonzalez
- Instituto de Química Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile.
| | - Nicolas Salinas-Parra
- Instituto de Química Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | | | | |
Collapse
|
20
|
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
|
21
|
Montero D, Diaz-Canestro C, Oberholzer L, Lundby C. The role of blood volume in cardiac dysfunction and reduced exercise tolerance in patients with diabetes. Lancet Diabetes Endocrinol 2019; 7:807-816. [PMID: 31255583 DOI: 10.1016/s2213-8587(19)30119-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/20/2019] [Accepted: 03/27/2019] [Indexed: 11/18/2022]
Abstract
Blood volume is an integral component of the cardiovascular system, and fundamental to discerning the pathophysiology of multiple cardiovascular conditions leading to exercise intolerance. Based on a systematic search of controlled studies assessing blood volume, in this Personal View we describe how hypovolaemia is a prevalent characteristic of patients with diabetes, irrespective of sex, age, and physical activity levels. Multiple endocrine and haematological mechanisms contribute to hypovolaemia in diabetes. The regulation of intravascular volumes is altered by sustained hyperglycaemia and hypertension. Chronic activation of endocrine systems controlling fluid homeostasis, such as the renin-angiotensin-aldosterone system and vasopressin axis, has a role in progressive kidney desensitisation and diabetic nephropathy. Furthermore, albumin loss from the intravascular compartment reduces the osmotic potential of plasma to retain water. Hypovolaemia also affects the loading conditions and filling of the heart in diabetes. The elucidation of modifiable volumetric traits will plausibly have major health benefits in the diabetes population.
Collapse
Affiliation(s)
- David Montero
- Faculty of Kinesiology, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada.
| | - Candela Diaz-Canestro
- Faculty of Kinesiology, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada
| | - Laura Oberholzer
- Department of Clinical Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Carsten Lundby
- Inland Norway University of Applied Sciences, Lillehammer, Norway
| |
Collapse
|
22
|
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
|
23
|
Abstract
Cardiorenal syndrome commonly refers to the collective dysfunction of heart and kidney resulting in a cascade of feedback mechanism causing damage to both the organs and is associated with adverse clinical outcomes. The pathophysiology of cardiorenal syndrome is complex, multifactorial, and dynamic. Improving the understanding of disease mechanisms will aid in developing targeted pharmacologic and nonpharmacologic therapies for the management of this syndrome. This article discusses the various mechanisms involved in the pathophysiology of the cardiorenal syndrome.
Collapse
Affiliation(s)
- Ujjala Kumar
- Division of Nephrology-Hypertension, University of California San Diego, 9500 Gilman Drive# 9111H, La Jolla, CA 92093-9111, USA
| | - Nicholas Wettersten
- Division of Cardiology, University of California San Diego, 9434 Medical Center Drive, La Jolla, CA 92037, USA
| | - Pranav S Garimella
- Division of Nephrology-Hypertension, University of California San Diego, 9500 Gilman Drive# 9111H, La Jolla, CA 92093-9111, USA.
| |
Collapse
|
24
|
Giesecke T, Himmerkus N, Leipziger J, Bleich M, Koshimizu TA, Fähling M, Smorodchenko A, Shpak J, Knappe C, Isermann J, Ayasse N, Kawahara K, Schmoranzer J, Gimber N, Paliege A, Bachmann S, Mutig K. Vasopressin Increases Urinary Acidification via V1a Receptors in Collecting Duct Intercalated Cells. J Am Soc Nephrol 2019; 30:946-961. [PMID: 31097611 DOI: 10.1681/asn.2018080816] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 03/11/2019] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Antagonists of the V1a vasopressin receptor (V1aR) are emerging as a strategy for slowing progression of CKD. Physiologically, V1aR signaling has been linked with acid-base homeostasis, but more detailed information is needed about renal V1aR distribution and function. METHODS We used a new anti-V1aR antibody and high-resolution microscopy to investigate Va1R distribution in rodent and human kidneys. To investigate whether V1aR activation promotes urinary H+ secretion, we used a V1aR agonist or antagonist to evaluate V1aR function in vasopressin-deficient Brattleboro rats, bladder-catheterized mice, isolated collecting ducts, and cultured inner medullary collecting duct (IMCD) cells. RESULTS Localization of V1aR in rodent and human kidneys produced a basolateral signal in type A intercalated cells (A-ICs) and a perinuclear to subapical signal in type B intercalated cells of connecting tubules and collecting ducts. Treating vasopressin-deficient Brattleboro rats with a V1aR agonist decreased urinary pH and tripled net acid excretion; we observed a similar response in C57BL/6J mice. In contrast, V1aR antagonist did not affect urinary pH in normal or acid-loaded mice. In ex vivo settings, basolateral treatment of isolated perfused medullary collecting ducts with the V1aR agonist or vasopressin increased intracellular calcium levels in ICs and decreased luminal pH, suggesting V1aR-dependent calcium release and stimulation of proton-secreting proteins. Basolateral treatment of IMCD cells with the V1aR agonist increased apical abundance of vacuolar H+-ATPase in A-ICs. CONCLUSIONS Our results show that activation of V1aR contributes to urinary acidification via H+ secretion by A-ICs, which may have clinical implications for pharmacologic targeting of V1aR.
Collapse
Affiliation(s)
- Torsten Giesecke
- Institute of Vegetative Anatomy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; .,Berlin Institute of Health (BIH), Berlin, Germany
| | - Nina Himmerkus
- Institute of Physiology, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Jens Leipziger
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Markus Bleich
- Institute of Physiology, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Taka-Aki Koshimizu
- Division of Molecular Pharmacology, Department of Pharmacology, Jichi Medical University, Shimotsuke-shi, Tochigi-ken, Japan
| | - Michael Fähling
- Institute of Vegetative Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Alina Smorodchenko
- Institute of Vegetative Anatomy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Julia Shpak
- Institute of Vegetative Anatomy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Carolin Knappe
- Institute of Vegetative Anatomy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Julian Isermann
- Institute of Physiology, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Niklas Ayasse
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Katsumasa Kawahara
- Department of Physiology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Jan Schmoranzer
- Advanced Medical BioImaging Core Facility, Charite-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Niclas Gimber
- Advanced Medical BioImaging Core Facility, Charite-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Alexander Paliege
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; and
| | - Sebastian Bachmann
- Institute of Vegetative Anatomy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Kerim Mutig
- Institute of Vegetative Anatomy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; .,Department of Pharmacology, I.M. Sechenov First Moscow State Medical University of the Ministry of Healthcare of the Russian Federation (Sechenovskiy University), Moscow, Russian Federation
| |
Collapse
|
25
|
Renal response to an oral protein load in patients with central diabetes insipidus before and after treatment with vasopressin. J Nephrol 2019; 32:411-415. [PMID: 30778919 DOI: 10.1007/s40620-018-00575-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 12/18/2018] [Indexed: 10/27/2022]
Abstract
AIM Different factors have been hypothesized to play a role in the cascade of events associated with the protein-induced glomerular response. However, scant data are available on the possible functional effect of vasopressin (VP) on the glomerular filtration rate (GFR) in humans with central diabetes insipidus (CDI), which was the aim of the present study. METHOD Renal function was studied under fasting conditions (baseline) and after a meat meal in 16 patients with CDI before and after treatment with desmopressin (DDAVP) and in 16 control subjects. GFR was measured by the inulin method. RESULTS At baseline, the GFR was lower in patients with CDI. Treatment with DDAVP resulted in an insignificant increase in GFR, which was not statistically different from untreated patients. After an acute oral protein load, the GFR increased, peaking at 45 min post meal in controls, and at 135 min post meal in treated and untreated CDI patients. CONCLUSION After a meat meal, the peak GFR response is delayed in CDI patients suggesting that VP might indirectly affect tubule-glomerular feedback.
Collapse
|
26
|
Muscogiuri G, Barrea L, Annunziata G, Vecchiarini M, Orio F, Di Somma C, Colao A, Savastano S. Water intake keeps type 2 diabetes away? Focus on copeptin. Endocrine 2018; 62:292-298. [PMID: 30027433 DOI: 10.1007/s12020-018-1680-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 07/09/2018] [Indexed: 12/11/2022]
Abstract
INTRODUCTION In both diabetic subjects and animal models high levels of vasopressin (AVP) have beendetected. The relationship between AVP and glucose metabolism is mediated through several direct andindirect effects and most of them are still unknown. METHODS We have reviewed 100 manuscripts retrieved from Cochrane Library, Embase and Pubmeddatabases in order to highlight a possible relationship between copeptin and type 2 diabetes and to provideinsights on the molecular mechanism that could explain this association. RESULTS AND CONCLUSIONS AVP potentiates CRH action at pituitary level resulting in an increased ACTH secretion and in turn in an increased cortisol secretion that escapes the negative feedback loop. Further, AVP regulates insulin and glucagon secretion through V1b receptor and promotes hepatic glycogenolysis and gluconeogenesis through V1a receptor. In addition to worsen glucose metabolism, AVP has been reported to have a role in the pathogenesis of diabetic complications such as cardiovascular diseases, kidney and ocular complications. Due to the very low concentration of AVP in the blood, the small size and poor stability, the assay of AVP is very difficult to perform. Thus, copeptin, the stable C-terminal portion of the prepro-vasopressin peptide has been identified as an easier assay to be measured and that mirrors AVP activity. Although there are promising evidence that copeptin could be involved in the pathogenesis of type 2 diabetes, further studies need to demonstrate the importance of copeptin as clinical marker to predict glucose metabolism derangements.
Collapse
Affiliation(s)
- Giovanna Muscogiuri
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy.
| | - Luigi Barrea
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy
| | - Giuseppe Annunziata
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy
| | | | - Francesco Orio
- Dipartimento di Scienze Motorie e del Benessere, Università Partenope di Napoli, Naples, Italy
| | | | - Annamaria Colao
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy
| | - Silvia Savastano
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy
| |
Collapse
|
27
|
El Boustany R, Tasevska I, Meijer E, Kieneker LM, Enhörning S, Lefèvre G, Mohammedi K, Marre M, Fumeron F, Balkau B, Bouby N, Bankir L, Bakker SJ, Roussel R, Melander O, Gansevoort RT, Velho G. Plasma copeptin and chronic kidney disease risk in 3 European cohorts from the general population. JCI Insight 2018; 3:121479. [PMID: 29997293 PMCID: PMC6124520 DOI: 10.1172/jci.insight.121479] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 05/23/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The prevalence of chronic kidney disease (CKD) is increasing worldwide. The identification of factors contributing to its progression is important for designing preventive measures. Previous studies have suggested that chronically high vasopressin is deleterious to renal function. Here, we evaluated the association of plasma copeptin, a surrogate of vasopressin, with the incidence of CKD in the general population. METHODS We studied 3 European cohorts: DESIR (n = 5,047; France), MDCS-CC (n = 3,643; Sweden), and PREVEND (n = 7,684; the Netherlands). Median follow-up was 8.5, 16.5, and 11.3 years, respectively. Pooled data were analyzed at an individual level for 4 endpoints during follow-up: incidence of stage 3 CKD (estimated glomerular filtration rate [eGFR] < 60 ml/min/1.73 m2); the KDIGO criterion "certain drop in eGFR"; rapid kidney function decline (eGFR slope steeper than -3 ml/min/1.73 m2/yr); and incidence of microalbuminuria. RESULTS The upper tertile of plasma copeptin was significantly and independently associated with a 49% higher risk for stage 3 CKD (P < 0.0001); a 64% higher risk for kidney function decline, as defined by the KDIGO criterion (P < 0.0001); a 79% higher risk for rapid kidney function decline (P < 0.0001); and a 24% higher risk for microalbuminuria (P = 0.008). CONCLUSIONS High copeptin levels are associated with the development and the progression of CKD in the general population. Intervention studies are needed to assess the potential beneficial effect on kidney health in the general population of reducing vasopressin secretion or action. FUNDING INSERM and Danone Research Centre for Specialized Nutrition.
Collapse
Affiliation(s)
- Ray El Boustany
- Inserm Research Unit 1138, Centre de Recherche des Cordeliers, Paris, France
- Danone Nutricia Research, Palaiseau, France
| | - Irina Tasevska
- Departments of Internal Medicine and Clinical Sciences, Lund University, Malmö, Sweden
| | - Esther Meijer
- Department of Internal Medicine, University Medical Center, Division of Nephrology, University of Groningen, Groningen, Netherlands
| | - Lyanne M. Kieneker
- Department of Internal Medicine, University Medical Center, Division of Nephrology, University of Groningen, Groningen, Netherlands
| | - Sofia Enhörning
- Departments of Internal Medicine and Clinical Sciences, Lund University, Malmö, Sweden
| | - Guillaume Lefèvre
- Service de Biochimie et Hormonologie, Assistance Publique — Hôpitaux de Paris, Hôpitaux Universitaires Est Parisien–Tenon, Paris, France
| | - Kamel Mohammedi
- Inserm Research Unit 1138, Centre de Recherche des Cordeliers, Paris, France
- Department of Diabetology, Endocrinology and Nutrition, DHU Fire, Assistance Publique — Hôpitaux de Paris, Bichat Hospital, Paris, France
| | - Michel Marre
- Inserm Research Unit 1138, Centre de Recherche des Cordeliers, Paris, France
- Department of Diabetology, Endocrinology and Nutrition, DHU Fire, Assistance Publique — Hôpitaux de Paris, Bichat Hospital, Paris, France
- UFR de Médecine, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Frédéric Fumeron
- Inserm Research Unit 1138, Centre de Recherche des Cordeliers, Paris, France
- UFR de Médecine, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Beverley Balkau
- Inserm Research Unit 1018, Center for Research in Epidemiology and Population Health, Villejuif, France
- Université Paris Sud, Villejuif, France
| | - Nadine Bouby
- Inserm Research Unit 1138, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Lise Bankir
- Inserm Research Unit 1138, Centre de Recherche des Cordeliers, Paris, France
- UPMC University Paris 6, Sorbonne Universités, Paris, France
| | - Stephan J.L. Bakker
- Department of Internal Medicine, University Medical Center, Division of Nephrology, University of Groningen, Groningen, Netherlands
| | - Ronan Roussel
- Inserm Research Unit 1138, Centre de Recherche des Cordeliers, Paris, France
- Department of Diabetology, Endocrinology and Nutrition, DHU Fire, Assistance Publique — Hôpitaux de Paris, Bichat Hospital, Paris, France
- UFR de Médecine, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Olle Melander
- Departments of Internal Medicine and Clinical Sciences, Lund University, Malmö, Sweden
| | - Ron T. Gansevoort
- Department of Internal Medicine, University Medical Center, Division of Nephrology, University of Groningen, Groningen, Netherlands
| | - Gilberto Velho
- Inserm Research Unit 1138, Centre de Recherche des Cordeliers, Paris, France
| |
Collapse
|
28
|
Kim YB, Kim WB, Jung WW, Jin X, Kim YS, Kim B, Han HC, Block GD, Colwell CS, Kim YI. Excitatory GABAergic Action and Increased Vasopressin Synthesis in Hypothalamic Magnocellular Neurosecretory Cells Underlie the High Plasma Level of Vasopressin in Diabetic Rats. Diabetes 2018; 67:486-495. [PMID: 29212780 DOI: 10.2337/db17-1042] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 11/20/2017] [Indexed: 11/13/2022]
Abstract
Diabetes mellitus (DM) is associated with increased plasma levels of arginine-vasopressin (AVP), which may aggravate hyperglycemia and nephropathy. However, the mechanisms by which DM may cause the increased AVP levels are not known. Electrophysiological recordings in supraoptic nucleus (SON) slices from streptozotocin (STZ)-induced DM rats and vehicle-treated control rats revealed that γ-aminobutyric acid (GABA) functions generally as an excitatory neurotransmitter in the AVP neurons of STZ rats, whereas it usually evokes inhibitory responses in the cells of control animals. Furthermore, Western blotting analyses of Cl- transporters in the SON tissues indicated that Na+-K+-2Cl- cotransporter isotype 1 (a Cl- importer) was upregulated and K+-Cl- cotransporter isotype 2 (KCC2; a Cl- extruder) was downregulated in STZ rats. Treatment with CLP290 (a KCC2 activator) significantly lowered blood AVP and glucose levels in STZ rats. Last, investigation that used rats expressing an AVP-enhanced green fluorescent protein fusion gene showed that AVP synthesis in AVP neurons was much more intense in STZ rats than in control rats. We conclude that altered Cl- homeostasis that makes GABA excitatory and enhanced AVP synthesis are important changes in AVP neurons that would increase AVP secretion in DM. Our data suggest that Cl- transporters in AVP neurons are potential targets of antidiabetes treatments.
Collapse
Affiliation(s)
- Young-Beom Kim
- Department of Physiology, Korea University College of Medicine, Seoul, Republic of Korea
- Neuroscience Research Institute, Korea University College of Medicine, Seoul, Republic of Korea
| | - Woong Bin Kim
- Department of Physiology, Korea University College of Medicine, Seoul, Republic of Korea
- Neuroscience Research Institute, Korea University College of Medicine, Seoul, Republic of Korea
| | - Won Woo Jung
- Department of Physiology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Xiangyan Jin
- Department of Physiology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Yoon Sik Kim
- Department of Physiology, Korea University College of Medicine, Seoul, Republic of Korea
- Department of Psychiatry & Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA
| | - Byoungjae Kim
- Department of Physiology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hee Chul Han
- Department of Physiology, Korea University College of Medicine, Seoul, Republic of Korea
- Neuroscience Research Institute, Korea University College of Medicine, Seoul, Republic of Korea
| | - Gene D Block
- Department of Psychiatry & Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA
| | - Christopher S Colwell
- Department of Psychiatry & Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA
| | - Yang In Kim
- Department of Physiology, Korea University College of Medicine, Seoul, Republic of Korea
- Neuroscience Research Institute, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
29
|
García-Arroyo FE, Tapia E, Blas-Marron MG, Gonzaga G, Silverio O, Cristóbal M, Osorio H, Arellano-Buendía AS, Zazueta C, Aparicio-Trejo OE, Reyes-García JG, Pedraza-Chaverri J, Soto V, Roncal-Jiménez C, Johnson RJ, Sánchez-Lozada LG. Vasopressin Mediates the Renal Damage Induced by Limited Fructose Rehydration in Recurrently Dehydrated Rats. Int J Biol Sci 2017; 13:961-975. [PMID: 28924378 PMCID: PMC5599902 DOI: 10.7150/ijbs.20074] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 05/26/2017] [Indexed: 12/26/2022] Open
Abstract
Recurrent dehydration and heat stress cause chronic kidney damage in experimental animals. The injury is exacerbated by rehydration with fructose-containing beverages. Fructose may amplify dehydration-induced injury by directly stimulating vasopressin release and also by acting as a substrate for the aldose reductase-fructokinase pathway, as both of these systems are active during dehydration. The role of vasopressin in heat stress associated injury has not to date been explored. Here we show that the amplification of renal damage mediated by fructose in thermal dehydration is mediated by vasopressin. Fructose rehydration markedly enhanced vasopressin (copeptin) levels and activation of the aldose reductase-fructokinase pathway in the kidney. Moreover, the amplification of the renal functional changes (decreased creatinine clearance and tubular injury with systemic inflammation, renal oxidative stress, and mitochondrial dysfunction) were prevented by the blockade of V1a and V2 vasopressin receptors with conivaptan. On the other hand, there are also other operative mechanisms when water is used as rehydration fluid that produce milder renal damage that is not fully corrected by vasopressin blockade. Therefore, we clearly showed evidence of the cross-talk between fructose, even at small doses, and vasopressin that interact to amplify the renal damage induced by dehydration. These data may be relevant for heat stress nephropathy as well as for other renal pathologies due to the current generalized consumption of fructose and deficient hydration habits.
Collapse
Affiliation(s)
| | - Edilia Tapia
- Laboratory of Renal Physiopathology. INC Ignacio Chávez. Mexico City. Mexico.,Dept. of Nephrology. INC Ignacio Chávez. Mexico City. Mexico
| | | | - Guillermo Gonzaga
- Laboratory of Renal Physiopathology. INC Ignacio Chávez. Mexico City. Mexico
| | - Octaviano Silverio
- Laboratory of Renal Physiopathology. INC Ignacio Chávez. Mexico City. Mexico
| | - Magdalena Cristóbal
- Laboratory of Renal Physiopathology. INC Ignacio Chávez. Mexico City. Mexico.,Dept. of Nephrology. INC Ignacio Chávez. Mexico City. Mexico
| | - Horacio Osorio
- Laboratory of Renal Physiopathology. INC Ignacio Chávez. Mexico City. Mexico.,Dept. of Nephrology. INC Ignacio Chávez. Mexico City. Mexico
| | - Abraham S Arellano-Buendía
- Laboratory of Renal Physiopathology. INC Ignacio Chávez. Mexico City. Mexico.,Dept. of Nephrology. INC Ignacio Chávez. Mexico City. Mexico
| | - Cecilia Zazueta
- Dept. of Cardiovascular Biomedicine. INC Ignacio Chávez. Mexico City. Mexico
| | | | - Juan G Reyes-García
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, IPN Mexico City. Mexico
| | | | - Virgilia Soto
- Dept. of Pathology. INC Ignacio Chávez. Mexico City. Mexico
| | | | | | - Laura G Sánchez-Lozada
- Laboratory of Renal Physiopathology. INC Ignacio Chávez. Mexico City. Mexico.,Dept. of Nephrology. INC Ignacio Chávez. Mexico City. Mexico
| |
Collapse
|
30
|
|
31
|
Bachmann S, Mutig K. Regulation of renal Na-(K)-Cl cotransporters by vasopressin. Pflugers Arch 2017; 469:889-897. [DOI: 10.1007/s00424-017-2002-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 05/16/2017] [Accepted: 05/17/2017] [Indexed: 10/19/2022]
|
32
|
Bjornstad P, Johnson RJ, Snell-Bergeon JK, Pyle L, Davis A, Foster N, Cherney DZ, Maahs DM. Albuminuria is associated with greater copeptin concentrations in men with type 1 diabetes: A brief report from the T1D exchange Biobank. J Diabetes Complications 2017; 31:387-389. [PMID: 27979439 PMCID: PMC5303164 DOI: 10.1016/j.jdiacomp.2016.11.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 11/25/2016] [Accepted: 11/28/2016] [Indexed: 11/20/2022]
Abstract
Vasopressin exerts important cardio-renal effects, but remains problematic to measure. Copeptin is a more stable peptide derived from the same precursor molecule. In this case-control study from the Type 1 Diabetes Exchange (T1DX) Biobank registry, men with T1D and albuminuria had greater copeptin concentrations than men with normoalbuminuria.
Collapse
Affiliation(s)
- Petter Bjornstad
- Department of Pediatric Endocrinology, University of Colorado School of Medicine, Aurora, CO; Barbara Davis Center for Diabetes, University of Colorado Denver, Aurora, CO.
| | | | | | - Laura Pyle
- Department of Pediatric Endocrinology, University of Colorado School of Medicine, Aurora, CO
| | - Asa Davis
- Benaroya Research Institute, Seattle, WA
| | | | - David Z Cherney
- Department of Nephrology, University of Toronto School of Medicine, Toronto, ON
| | - David M Maahs
- Department of Pediatrics, Division of Endocrinology, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
33
|
Roncal-Jimenez CA, Milagres T, Andres-Hernando A, Kuwabara M, Jensen T, Song Z, Bjornstad P, Garcia GE, Sato Y, Sanchez-Lozada LG, Lanaspa MA, Johnson RJ. Effects of exogenous desmopressin on a model of heat stress nephropathy in mice. Am J Physiol Renal Physiol 2016; 312:F418-F426. [PMID: 28003190 PMCID: PMC5374310 DOI: 10.1152/ajprenal.00495.2016] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 12/06/2016] [Accepted: 12/09/2016] [Indexed: 11/22/2022] Open
Abstract
Recurrent heat stress and dehydration have recently been shown experimentally to cause chronic kidney disease (CKD). One potential mediator may be vasopressin, acting via the type 2 vasopressin receptor (V2 receptor). We tested the hypothesis that desmopressin accelerates CKD in mice subjected to heat stress and recurrent dehydration. Recurrent exposure to heat with limited water availability was performed in male mice over a 5-wk period, with one group receiving desmopressin two times daily and the other group receiving vehicle. Two additional control groups were not exposed to heat or dehydration and received vehicle or desmopressin. The effects of the treatment on renal injury were assessed. Heat stress and recurrent dehydration induced functional changes (albuminuria, elevated urinary neutrophil gelatinase-associated protein), glomerular changes (mesangiolysis, matrix expansion), and tubulointerstitial changes (fibrosis, inflammation). Desmopressin also induced albuminuria, glomerular changes, and tubulointerstitial fibrosis in normal animals and also exacerbated injury in mice with heat stress nephropathy. Both heat stress and/or desmopressin were also associated with activation of the polyol pathway in the renal cortex, likely due to increased interstitial osmolarity. Our studies document both glomerular and tubulointerstitial injury and inflammation in heat stress nephropathy and may be clinically relevant to the pathogenesis of Mesoamerican nephropathy. Our data also suggest that vasopressin may play a role in the pathogenesis of the renal injury of heat stress nephropathy, likely via a V2 receptor-dependent pathway.
Collapse
Affiliation(s)
| | - Tamara Milagres
- Division of Renal Diseases and Hypertension, University of Colorado, Aurora, Colorado
| | - Ana Andres-Hernando
- Division of Renal Diseases and Hypertension, University of Colorado, Aurora, Colorado
| | - Masanari Kuwabara
- Division of Renal Diseases and Hypertension, University of Colorado, Aurora, Colorado
| | - Thomas Jensen
- Division of Renal Diseases and Hypertension, University of Colorado, Aurora, Colorado.,Division of Endocrinology, Metabolism, and Diabetes, University of Colorado, Aurora, Colorado
| | - Zhilin Song
- Division of Renal Diseases and Hypertension, University of Colorado, Aurora, Colorado.,Division of Endocrinology, Metabolism, and Diabetes, University of Colorado, Aurora, Colorado
| | - Petter Bjornstad
- Division of Pediatric Endocrinology, University of Colorado, Aurora, Colorado; and
| | - Gabriela E Garcia
- Division of Renal Diseases and Hypertension, University of Colorado, Aurora, Colorado
| | - Yuka Sato
- Division of Renal Diseases and Hypertension, University of Colorado, Aurora, Colorado
| | | | - Miguel A Lanaspa
- Division of Renal Diseases and Hypertension, University of Colorado, Aurora, Colorado
| | - Richard J Johnson
- Division of Renal Diseases and Hypertension, University of Colorado, Aurora, Colorado;
| |
Collapse
|
34
|
Zhu FX, Wu HL, Tu KS, Chen JX, Zhang M, Shi C. Serum levels of copeptin are associated with type 2 diabetes and diabetic complications in Chinese population. J Diabetes Complications 2016; 30:1566-1570. [PMID: 27497684 DOI: 10.1016/j.jdiacomp.2016.07.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 07/18/2016] [Accepted: 07/19/2016] [Indexed: 01/19/2023]
Abstract
PURPOSE The aim of this study was to investigate copeptin levels in serum, and assess their associations with type 2 diabetes (T2DM) and diabetic complications. METHODS In this post hoc analysis, serum levels of copeptin were tested in 306 patients with T2DM. Clinical information including diabetic retinopathy (DR) and diabetic nephropathy (DN) were collected. The relation of serum copeptin with DR and DN were investigated with the use of logistic regression models according to equal quartiles of the distributions of serum copeptin. RESULTS We found that serum copeptin levels were significantly higher in diabetes as compared to normal controls [9.4(IQR, 7.4-12.5) pmol/L vs. 4.1(IQR, 2.5-6.2) pmol/L; P<0.0001]. In multivariate analysis, there was an increased risk of T2DM associated with copeptin levels (OR 1.312, 95% CI: 1.204-1.403; P<0.0001) after adjusting for possible confounders. After adjustment for possible confounders, serum copeptin levels were positively associated with the DR (odds ratio [OR], 1.117; 95% confidence interval [CI], 1.072-1.241; P<0.001) and DN (OR, 1.259; 95% CI, 1.198-1.323; P<0.001). Compared with the first quartile of serum copeptin levels, the ORs for DR and DN were as follows: second quartile, 1.19 (95% CI, 0.94-1.51, P=0.12) and 1.37 (95% CI, 0.78-2.37, P=0.28); third quartile, 1.61 (95% CI, 1.18-2.43, P=0.005) and 2.12 (95% CI, 1.32-3.27, P=0.003); fourth quartile, 2.83 (95% CI, 2.04-4.93; P<0.001) and 3.48 (95% CI, 1.77-7.03; P<0.001), respectively. CONCLUSIONS Using a post-hoc analysis our data show that elevated serum levels of copeptin are associated with type 2 diabetes and diabetic complications in Chinese population, suggesting a potential role of the AVP system (copeptin) in the pathophysiology of diabetes.
Collapse
Affiliation(s)
- Fu-Xiang Zhu
- Department of Nephrology, the First Hospital of Jiaxing, Jiaxing 314001, Zhejiang Province, PR China.
| | - Heng-Lan Wu
- Department of Nephrology, the First Hospital of Jiaxing, Jiaxing 314001, Zhejiang Province, PR China
| | - Kai-Sheng Tu
- Department of Endocrinology, the Affiliated Beijing Rehabilitation Hospital of Capital Medical University, Beijing 100144, PR China
| | - Jian-Xiang Chen
- Department of Nephrology, the First Hospital of Jiaxing, Jiaxing 314001, Zhejiang Province, PR China
| | - Min Zhang
- Department of Nephrology, the First Hospital of Jiaxing, Jiaxing 314001, Zhejiang Province, PR China
| | - Chao Shi
- Department of Nephrology, the First Hospital of Jiaxing, Jiaxing 314001, Zhejiang Province, PR China
| |
Collapse
|
35
|
Johnson RJ, Stenvinkel P, Jensen T, Lanaspa MA, Roncal C, Song Z, Bankir L, Sánchez-Lozada LG. Metabolic and Kidney Diseases in the Setting of Climate Change, Water Shortage, and Survival Factors. J Am Soc Nephrol 2016; 27:2247-56. [PMID: 27283495 PMCID: PMC4978060 DOI: 10.1681/asn.2015121314] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Climate change (global warming) is leading to an increase in heat extremes and coupled with increasing water shortage, provides a perfect storm for a new era of environmental crises and potentially, new diseases. We use a comparative physiologic approach to show that one of the primary mechanisms by which animals protect themselves against water shortage is to increase fat mass as a means for providing metabolic water. Strong evidence suggests that certain hormones (vasopressin), foods (fructose), and metabolic products (uric acid) function as survival signals to help reduce water loss and store fat (which also provides a source of metabolic water). These mechanisms are intricately linked with each other and stimulated by dehydration and hyperosmolarity. Although these mechanisms were protective in the setting of low sugar and low salt intake in our past, today, the combination of diets high in fructose and salty foods, increasing temperatures, and decreasing available water places these survival signals in overdrive and may be accelerating the obesity and diabetes epidemics. The recent discovery of multiple epidemics of CKD occurring in agricultural workers in hot and humid environments may represent harbingers of the detrimental consequences of the combination of climate change and overactivation of survival pathways.
Collapse
Affiliation(s)
- Richard J Johnson
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, Colorado;
| | - Peter Stenvinkel
- Division of Renal Medicine, Department of Clinical Science Intervention and Technology, Karolinska University Hospital, Stockholm, Sweden
| | - Thomas Jensen
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, Colorado
| | - Miguel A Lanaspa
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, Colorado
| | - Carlos Roncal
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, Colorado
| | - Zhilin Song
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, Colorado
| | - Lise Bankir
- Institut National de las Santé et de la Recherche Medicalé UMRS 1138, Centre de Recherche des Cordeliers, Paris, France; and
| | - Laura G Sánchez-Lozada
- Laboratory of Renal Physiopathology, Instituto Nacional de Cardiologia Ignacio Chávez, Mexico City, Mexico
| |
Collapse
|
36
|
Bjornstad P, Maahs DM, Jensen T, Lanaspa MA, Johnson RJ, Rewers M, Snell-Bergeon JK. Elevated copeptin is associated with atherosclerosis and diabetic kidney disease in adults with type 1 diabetes. J Diabetes Complications 2016; 30:1093-6. [PMID: 27141815 PMCID: PMC4949105 DOI: 10.1016/j.jdiacomp.2016.04.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 04/14/2016] [Accepted: 04/15/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND Vasopressin exerts important cardio-renal effects, but remains problematic to measure. Copeptin is a more stable peptide derived from the same precursor molecule. We examined the associations between copeptin, coronary artery calcium (CAC), albuminuria and impaired glomerular filtration rate (GFR) in adults with type 1 diabetes (T1D). METHODS Participants with (n=209) and without T1D (n=244) in the Coronary Artery Calcification in Type 1 Diabetes (CACTI) study were assessed for serum copeptin, CAC measured using 128-slice spiral CT, urinary albumin-to-creatinine ratio (UACR) and eGFR calculated by CKD-EPI creatinine. Impaired GFR was defined as eGFR <60mL/min/1.73m(2), albuminuria as UACR ≥30mg/g, high and very high CAC score as ≥100 and ≥300AU, and elevated copeptin as >13pmol/L (>97.5th percentile for healthy adults). Unadjusted and adjusted (age, sex, HbA1c, SBP and LDL-C) logistic models were applied to examine the relationships. RESULTS Participants with T1D had greater ultrasensitive copeptin concentrations than non-diabetics (3.5 [95% CI 2.3-3.8] vs. 2.8 [2.7-3.1], p=0.003). In participants with T1D, elevated copeptin was associated with greater odds of impaired eGFR (OR: 18.52, 95% CI 4.03-85.02), albuminuria (10.55, 2.24-49.62), high CAC (6.61, 1.39-31.31) and very high CAC (6.24, 1.51-25.90) in multivariable models. Similar linear relationships were obtained with ultrasensitive copeptin, eGFR, UACR, CAC volume and CAC score in adjusted models. CONCLUSION In this cross-sectional analysis, copeptin was strongly associated with diabetic kidney disease and coronary atherosclerosis in adults with T1D. Further research is needed to determine whether these relationships hold true longitudinally in people with T1D.
Collapse
Affiliation(s)
- Petter Bjornstad
- Department of Pediatric Endocrinology, University of Colorado School of Medicine, Aurora, CO; Barbara Davis Center for Diabetes, University of Colorado Denver, Aurora, CO.
| | - David M Maahs
- Department of Pediatric Endocrinology, University of Colorado School of Medicine, Aurora, CO; Barbara Davis Center for Diabetes, University of Colorado Denver, Aurora, CO; Department of Nephrology, University of Colorado Denver, Aurora, CO
| | - Thomas Jensen
- Department of Adult Endocrinology, University of Colorado School of Medicine, Aurora, CO
| | - Miguel A Lanaspa
- Department of Nephrology, University of Colorado Denver, Aurora, CO
| | | | - Marian Rewers
- Department of Pediatric Endocrinology, University of Colorado School of Medicine, Aurora, CO; Barbara Davis Center for Diabetes, University of Colorado Denver, Aurora, CO
| | - Janet K Snell-Bergeon
- Department of Pediatric Endocrinology, University of Colorado School of Medicine, Aurora, CO; Barbara Davis Center for Diabetes, University of Colorado Denver, Aurora, CO
| |
Collapse
|
37
|
Windt WAKM, Tahara A, Kluppel ACA, de Zeeuw D, Henning RH, van Dokkum RPE. Early, but not late therapy with a vasopressin V1a-antagonist ameliorates the development of renal damage after 5/6 nephrectomy. J Renin Angiotensin Aldosterone Syst 2016; 7:217-24. [PMID: 17347933 DOI: 10.3317/jraas.2006.041] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Introduction. Vasopressin, mainly through the V1a-receptor, is thought to be a major player in the maintenance of hyperfiltration. Its inhibition could therefore lead to a decrease in progression of chronic renal failure.To this end, the effect of the vasopressin V1a-receptor-selective antagonist, YM218, was studied on proteinuria and focal glomerulosclerosis in early and late intervention after 5/6 nephrectomy in rats, and compared with an angiotensin-converting enzyme inhibitor (ACE-I).Materials and methods. After 5/6 nephrectomy, early intervention was performed between week 2 and 10 thereafter with the V1a-receptor-selective antagonist (VRA, 10 mg/kg/day, n=10), enalapril (ACE-I, 10 mg/kg/day, n=9), or vehicle (n=8). Late intervention was performed in another group between week 6 and 12 with VRA (10 mg/kg/day, n=7), lisinopril (ACE-I, 5 mg/kg/day, n=7), or vehicle (n=7).Results. In early intervention, proteinuria and focal glomerulosclerosis were significantly decreased by VRA compared to vehicle (44 7% and 59+8% respectively). ACE-I significantly decreased proteinuria (67 7%) and a trend towards a decrease in focal glomerulosclerosis was observed (30 18%). In late intervention, VRA did not decrease proteinuria and focal glomerulosclerosis compared to vehicle (21 20% and 0%, respectively),ACE-I significantly lowered proteinuria (92 2%) and a focal glomerulosclerosis (69+1%) lowering trend was observed.Conclusion. These results indicate that VRA may protect against early progression of renal injury after 5/6 nephrectomy, whereas its effectiveness seems limited in established renal damage.
Collapse
Affiliation(s)
- Willemijn A K M Windt
- Department of Clinical Pharmacology, Groningen Institute for Drug Evaluation (GUIDE), University Medical Center Groningen, Antonius Deusinglaan 1. NL-9713 AV Groningen NL-9713, The Netherlands
| | | | | | | | | | | |
Collapse
|
38
|
Hassouneh R, Nasrallah R, Zimpelmann J, Gutsol A, Eckert D, Ghossein J, Burns KD, Hébert RL. PGE2 receptor EP3 inhibits water reabsorption and contributes to polyuria and kidney injury in a streptozotocin-induced mouse model of diabetes. Diabetologia 2016; 59:1318-28. [PMID: 26995650 DOI: 10.1007/s00125-016-3916-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 02/19/2016] [Indexed: 10/22/2022]
Abstract
AIMS/HYPOTHESIS The first clinical manifestation of diabetes is polyuria. The prostaglandin E2 (PGE2) receptor EP3 antagonises arginine vasopressin (AVP)-mediated water reabsorption and its expression is increased in the diabetic kidney. The purpose of this work was to study the contribution of EP3 to diabetic polyuria and renal injury. METHODS Male Ep 3 (-/-) (also known as Ptger3 (-/-)) mice were treated with streptozotocin (STZ) to generate a mouse model of diabetes and renal function was evaluated after 12 weeks. Isolated collecting ducts (CDs) were microperfused to study the contribution of EP3 to AVP-mediated fluid reabsorption. RESULTS Ep 3 (-/-)-STZ mice exhibited attenuated polyuria and increased urine osmolality compared with wild-type STZ (WT-STZ) mice, suggesting enhanced water reabsorption. Compared with WT-STZ mice, Ep 3 (-/-)-STZ mice also had increased protein expression of aquaporin-1, aquaporin-2, and urea transporter A1, and reduced urinary AVP excretion, but increased medullary V2 receptors. In vitro microperfusion studies indicated that Ep 3 (-/-) and WT-STZ CDs responded to AVP stimulation similarly to those of wild-type mice, with a 60% increase in fluid reabsorption. In WT non-injected and WT-STZ mice, EP3 activation with sulprostone (PGE2 analogue) abrogated AVP-mediated water reabsorption; this effect was absent in mice lacking EP3. A major finding of this work is that Ep 3 (-/-)-STZ mice showed blunted renal cyclooxygenase-2 protein expression, reduced renal hypertrophy, reduced hyperfiltration and reduced albuminuria, as well as diminished tubular dilation and nuclear cysts. CONCLUSIONS/INTERPRETATION Taken together, the data suggest that EP3 contributes to diabetic polyuria by inhibiting expression of aquaporins and that it promotes renal injury during diabetes. EP3 may prove to be a promising target for more selective management of diabetic kidney disease.
Collapse
Affiliation(s)
- Ramzi Hassouneh
- Department of Cellular and Molecular Medicine, Kidney Research Centre, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Room 2514, Ottawa, ON, Canada, K1H 8M5
| | - Rania Nasrallah
- Department of Cellular and Molecular Medicine, Kidney Research Centre, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Room 2514, Ottawa, ON, Canada, K1H 8M5
| | - Joe Zimpelmann
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Alex Gutsol
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - David Eckert
- Department of Cellular and Molecular Medicine, Kidney Research Centre, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Room 2514, Ottawa, ON, Canada, K1H 8M5
| | - Jamie Ghossein
- Department of Cellular and Molecular Medicine, Kidney Research Centre, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Room 2514, Ottawa, ON, Canada, K1H 8M5
| | - Kevin D Burns
- Department of Cellular and Molecular Medicine, Kidney Research Centre, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Room 2514, Ottawa, ON, Canada, K1H 8M5
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Richard L Hébert
- Department of Cellular and Molecular Medicine, Kidney Research Centre, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Room 2514, Ottawa, ON, Canada, K1H 8M5.
| |
Collapse
|
39
|
Bjornstad P, Cherney DZ, Maahs DM, Nadeau KJ. Diabetic Kidney Disease in Adolescents With Type 2 Diabetes: New Insights and Potential Therapies. Curr Diab Rep 2016; 16:11. [PMID: 26803647 PMCID: PMC5841446 DOI: 10.1007/s11892-015-0708-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Diabetic kidney disease (DKD) is the leading cause of end-stage renal disease (ESRD) and dialysis in the Western world. Early DKD, including microalbuminuria and renal hyperfiltration, is common in adolescents with type 2 diabetes (T2D). Furthermore, youth-onset T2D carries a higher risk of progressive DKD than adult-onset T2D of similar diabetes duration. DKD is characterized by a long clinically silent period without signs of disease. Therefore, a major challenge in preventing DKD is the difficulty in identifying high-risk T2D patients at an early stage. The Type 2 Diabetes in Adolescents and Youth (TODAY) study demonstrated a high initial prevalence that increased over time, irrespective of treatment arm. This key observation underscores the importance of discovering new therapeutic targets to supplement conventional management, in order to reduce DKD risk. In this review, we focus on early DKD in T2D and summarize potential novel biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Petter Bjornstad
- Department of Pediatric Endocrinology, University of Colorado School of Medicine, Aurora, CO, USA.
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, 13123 East 16th Ave, Box B265, Aurora, CO, 80045, USA.
| | - David Z Cherney
- Department of Medicine, Division of Nephrology, Toronto General Hospital, University of Toronto, Toronto, ON, Canada
| | - David M Maahs
- Department of Pediatric Endocrinology, University of Colorado School of Medicine, Aurora, CO, USA
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, 13123 East 16th Ave, Box B265, Aurora, CO, 80045, USA
- Department of Medicine, Division of Nephrology, University of Colorado, Aurora, CO, USA
| | - Kristen J Nadeau
- Department of Pediatric Endocrinology, University of Colorado School of Medicine, Aurora, CO, USA
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, 13123 East 16th Ave, Box B265, Aurora, CO, 80045, USA
| |
Collapse
|
40
|
Roncal-Jimenez C, García-Trabanino R, Barregard L, Lanaspa MA, Wesseling C, Harra T, Aragón A, Grases F, Jarquin ER, González MA, Weiss I, Glaser J, Sánchez-Lozada LG, Johnson RJ. Heat Stress Nephropathy From Exercise-Induced Uric Acid Crystalluria: A Perspective on Mesoamerican Nephropathy. Am J Kidney Dis 2015; 67:20-30. [PMID: 26455995 DOI: 10.1053/j.ajkd.2015.08.021] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Accepted: 08/07/2015] [Indexed: 11/11/2022]
Abstract
Mesoamerican nephropathy (MeN), an epidemic in Central America, is a chronic kidney disease of unknown cause. In this article, we argue that MeN may be a uric acid disorder. Individuals at risk for developing the disease are primarily male workers exposed to heat stress and physical exertion that predisposes to recurrent water and volume depletion, often accompanied by urinary concentration and acidification. Uric acid is generated during heat stress, in part consequent to nucleotide release from muscles. We hypothesize that working in the sugarcane fields may result in cyclic uricosuria in which uric acid concentrations exceed solubility, leading to the formation of dihydrate urate crystals and local injury. Consistent with this hypothesis, we present pilot data documenting the common presence of urate crystals in the urine of sugarcane workers from El Salvador. High end-of-workday urinary uric acid concentrations were common in a pilot study, particularly if urine pH was corrected to 7. Hyperuricemia may induce glomerular hypertension, whereas the increased urinary uric acid may directly injure renal tubules. Thus, MeN may result from exercise and heat stress associated with dehydration-induced hyperuricemia and uricosuria. Increased hydration with water and salt, urinary alkalinization, reduction in sugary beverage intake, and inhibitors of uric acid synthesis should be tested for disease prevention.
Collapse
Affiliation(s)
| | - Ramón García-Trabanino
- Scientific Board, Department of Investigation, Hospital Nacional Rosales, San Salvador, El Salvador
| | - Lars Barregard
- Occupational and Environmental Medicine, Sahlgrenska University Hospital and University of Gothenburg, Gothenburg, Sweden
| | - Miguel A Lanaspa
- Division of Kidney Diseases and Hypertension, University of Colorado, Denver, CO
| | - Catharina Wesseling
- Unit of Occupational Medicine, Institute of Environmental Medicine (IMM), Karolinska Institutet, Stockholm, Sweden
| | - Tamara Harra
- Division of Kidney Diseases and Hypertension, University of Colorado, Denver, CO
| | - Aurora Aragón
- Research Center on Health, Work and Environment (CISTA), National Autonomous University of Nicaragua at León (UNAN-León), León, Nicaragua
| | - Felix Grases
- University of Balearic Islands, Palma de Mallorca, Spain
| | - Emmanuel R Jarquin
- Agencia para el Desarrollo y la Salud Agropecuaria, San Salvador, El Salvador
| | - Marvin A González
- Research Center on Health, Work and Environment (CISTA), National Autonomous University of Nicaragua at León (UNAN-León), León, Nicaragua; Department of Non-communicable Disease Epidemiology of London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Ilana Weiss
- La Isla Foundation, San Salvador, El Salvador
| | | | - Laura G Sánchez-Lozada
- Laboratory of Renal Physiopathology and Nephrology Department, INC Ignacio Chavez, Mexico City, Mexico
| | - Richard J Johnson
- Division of Kidney Diseases and Hypertension, University of Colorado, Denver, CO; Division of Nephrology, Eastern Colorado Health Care System, Department of Veteran Affairs, Denver, CO.
| |
Collapse
|
41
|
Roussel R, Matallah N, Bouby N, El Boustany R, Potier L, Fumeron F, Mohammedi K, Balkau B, Marre M, Bankir L, Velho G. Plasma Copeptin and Decline in Renal Function in a Cohort from the Community: The Prospective D.E.S.I.R. Study. Am J Nephrol 2015; 42:107-14. [PMID: 26346607 DOI: 10.1159/000439061] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 07/27/2015] [Indexed: 01/12/2023]
Abstract
BACKGROUND/AIMS In recent days, chronic kidney disease (CKD) is becoming an increasing public health problem. Identification of factors contributing to its progression is crucial for designing preventive interventions. Previous studies suggested that chronically high vasopressin is deleterious to the renal function. We evaluated plasma copeptin, a surrogate of vasopressin, as a predictor for renal function decline in a community cohort. METHODS Plasma copeptin was measured at baseline in 1,234 participants from the D.E.S.I.R. study, a prospective cohort from the French general population. All participants were followed for 9 years. Progression towards CKD during follow-up was defined as an estimated glomerular filtration rate (eGFR) below 60 ml/min/1.73 m2 on at least one follow-up visit. We have also considered the criterion 'Certain Drop in eGFR' proposed by the Kidney Disease Improving Global Outcomes (KDIGO) group. RESULTS Progression towards CKD was observed in 86 (7.0%) participants. Factors like age, female gender, plasma copeptin and use of angiotensin converting enzyme inhibitor or angiotensin 2 receptor blocker at baseline were positively associated, and eGFR inversely associated with CKD progression during follow-up. The hazard ratio per unit of log10-transformed plasma copeptin was 1.65 (95% CI 1.06-2.54) and p=0.02. Copeptin was similarly associated with CKD and this was observed when we considered the KDIGO criterion: OR 3.03 (95% CI 1.21-7.57), p=0.02. CONCLUSION The plasma copeptin level was independently and positively associated with progression towards CKD in a community-based cohort. Our results add to the available evidence for a deleterious effect of high vasopressin on renal health not only in selected groups of patients with CKD but also in the general population.
Collapse
Affiliation(s)
- Ronan Roussel
- INSERM, Research Unit 1138, Centre de Recherche des Cordeliers, Paris, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Kortenoeven MLA, Pedersen NB, Rosenbaek LL, Fenton RA. Vasopressin regulation of sodium transport in the distal nephron and collecting duct. Am J Physiol Renal Physiol 2015; 309:F280-99. [DOI: 10.1152/ajprenal.00093.2015] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 05/27/2015] [Indexed: 12/22/2022] Open
Abstract
Arginine vasopressin (AVP) is released from the posterior pituitary gland during states of hyperosmolality or hypovolemia. AVP is a peptide hormone, with antidiuretic and antinatriuretic properties. It allows the kidneys to increase body water retention predominantly by increasing the cell surface expression of aquaporin water channels in the collecting duct alongside increasing the osmotic driving forces for water reabsorption. The antinatriuretic effects of AVP are mediated by the regulation of sodium transport throughout the distal nephron, from the thick ascending limb through to the collecting duct, which in turn partially facilitates osmotic movement of water. In this review, we will discuss the regulatory role of AVP in sodium transport and summarize the effects of AVP on various molecular targets, including the sodium-potassium-chloride cotransporter NKCC2, the thiazide-sensitive sodium-chloride cotransporter NCC, and the epithelial sodium channel ENaC.
Collapse
Affiliation(s)
- M. L. A. Kortenoeven
- Department of Biomedicine and Center for Interactions of Proteins in Epithelial Transport (InterPrET), Aarhus University, Aarhus, Denmark
| | - N. B. Pedersen
- Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark; and
| | - L. L. Rosenbaek
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - R. A. Fenton
- Department of Biomedicine and Center for Interactions of Proteins in Epithelial Transport (InterPrET), Aarhus University, Aarhus, Denmark
| |
Collapse
|
43
|
Bankir L, Roussel R, Bouby N. Protein- and diabetes-induced glomerular hyperfiltration: role of glucagon, vasopressin, and urea. Am J Physiol Renal Physiol 2015; 309:F2-23. [DOI: 10.1152/ajprenal.00614.2014] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 04/13/2015] [Indexed: 12/21/2022] Open
Abstract
A single protein-rich meal (or an infusion of amino acids) is known to increase the glomerular filtration rate (GFR) for a few hours, a phenomenon known as “hyperfiltration.” It is important to understand the factors that initiate this upregulation because it becomes maladaptive in the long term. Several mediators and paracrine factors have been shown to participate in this upregulation, but they are not directly triggered by protein intake. Here, we explain how a rise in glucagon and in vasopressin secretion, directly induced by protein ingestion, might be the initial factors triggering the hepatic and renal events leading to an increase in the GFR. Their effects include metabolic actions in the liver and stimulation of sodium chloride reabsorption in the thick ascending limb. Glucagon is not only a glucoregulatory hormone. It is also important for the excretion of nitrogen end products by stimulating both urea synthesis in the liver (along with gluconeogenesis from amino acids) and urea excretion by the kidney. Vasopressin allows the concentration of nitrogenous end products (urea, ammonia, etc.) and other protein-associated wastes in a hyperosmotic urine, thus allowing a very significant water economy characteristic of all terrestrial mammals. No hyperfiltration occurs in the absence of one or the other hormone. Experimental results suggest that the combined actions of these two hormones, along with the complex intrarenal handling of urea, lead to alter the composition of the tubular fluid at the macula densa and to reduce the intensity of the signal activating the tubuloglomerular feedback control of GFR, thus allowing GFR to raise. Altogether, glucagon, vasopressin, and urea contribute to set up the best compromise between efficient urea excretion and water economy.
Collapse
Affiliation(s)
- Lise Bankir
- INSERM UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Diderot, Sorbonne-Paris-Cité, Paris, France; and
| | - Ronan Roussel
- INSERM UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Diderot, Sorbonne-Paris-Cité, Paris, France; and
- Diabétologie Endocrinologie Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France
| | - Nadine Bouby
- INSERM UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Diderot, Sorbonne-Paris-Cité, Paris, France; and
| |
Collapse
|
44
|
Taveau C, Chollet C, Waeckel L, Desposito D, Bichet DG, Arthus MF, Magnan C, Philippe E, Paradis V, Foufelle F, Hainault I, Enhorning S, Velho G, Roussel R, Bankir L, Melander O, Bouby N. Vasopressin and hydration play a major role in the development of glucose intolerance and hepatic steatosis in obese rats. Diabetologia 2015; 58:1081-90. [PMID: 25622862 DOI: 10.1007/s00125-015-3496-9] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 12/22/2014] [Indexed: 12/11/2022]
Abstract
AIMS/HYPOTHESIS High plasma copeptin, a marker of vasopressin (VP) secretion, has been shown to be associated with the metabolic syndrome and development of type 2 diabetes in humans. The present study was designed to determine the long-term influence of plasma VP concentration in a rodent model prone to metabolic dysfunction. METHODS Obese Zucker rats and their lean counterparts were submitted for 4 weeks to one of three protocols inducing different levels of VP. Circulating VP was either reduced by increasing the daily water intake (low-VP), or increased by a chronic i.p. infusion of VP (high-VP). The control rats had normal VP levels that depended on their own regulation of water intake and VP secretion. RESULTS Compared with controls with normal VP, lean rats with high-VP had a higher fasting glycaemia after 4 weeks. In obese rats, high-VP promoted hyperinsulinaemia, glucose intolerance, assessed by glucose and insulin tolerance tests, and an impaired response to a pyruvate challenge. Conversely, treatment with a selective arginine vasopressin receptor 1A (V1aR) antagonist reduced glucose intolerance. Low-VP obese rats had unchanged glucose tolerance but exhibited a drastic decrease in liver steatosis compared with control obese rats, associated with low hepatic triacylglycerol and cholesterol content, and reduced expression of hepatic lipogenic genes. These effects were independent of changes in body adiposity, and plasma sodium and osmolality did not differ among groups. CONCLUSION/INTERPRETATION These findings show a causal relationship between the VP-hydration axis and the metabolic risk. Therapeutic perspectives include diet recommendations regarding hydration, but also potential pharmacological interventions targeting the VP V1aR.
Collapse
Affiliation(s)
- Christopher Taveau
- Inserm U1138, Centre de Recherche des Cordeliers, 15 rue de l'Ecole de Medecine, 75006, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Bjornstad P, Maahs DM. Diabetes Complications in Childhood Diabetes-New Biomarkers and Technologies. CURRENT PEDIATRICS REPORTS 2015; 3:177-186. [PMID: 26425403 DOI: 10.1007/s40124-015-0081-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A major challenge in preventing vascular complications in diabetes is the inability to identify high-risk patients at an early stage, emphasizing the importance of discovering new risk factors, technologies and therapeutic targets to reduce the development and progression of complications. Promising biomarkers which may improve risk stratification and serve as therapeutic targets, include: uric acid, insulin sensitivity, copeptin, SGLT-2 and Klotho/FGF-23. Non-invasive measures of macrovasuclar disease in youth, include: 1) pulse wave velocity to examine arterial stiffness; 2) carotid intima-media thickness to evaluate arterial thickness; 3) cardiac MRI to investigate cardiac function and structure. Novel microvascular measures include: GFR by iohexol clearance using filter paper to directly measure GFR, retinal vascular geometry to predict early retinal changes and corneal confocal microscopy to improve detection of early nerve loss to better predict diabetic neuropathy. Herein we will review technologies, novel biomarkers, and therapeutic targets in relation to vascular complications of diabetes.
Collapse
Affiliation(s)
- Petter Bjornstad
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, United States ; Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - David M Maahs
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, United States ; Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, Colorado, United States
| |
Collapse
|
46
|
Mavani GP, DeVita MV, Michelis MF. A review of the nonpressor and nonantidiuretic actions of the hormone vasopressin. Front Med (Lausanne) 2015; 2:19. [PMID: 25853137 PMCID: PMC4371647 DOI: 10.3389/fmed.2015.00019] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 03/09/2015] [Indexed: 01/02/2023] Open
Abstract
The pressor and antidiuretic actions of arginine vasopressin (AVP) have been well documented. This review focuses on the less widely appreciated actions of AVP which also have important physiologic functions and when better understood may provide important insights into common disease states. These actions include effects on pain perception and bone structure as well as important relationships to the varied components of metabolic syndrome. These include effects on blood glucose, lipid levels, and blood pressure. AVP may also play a role in the progression of chronic kidney disease and effect physiologic changes relating to aging, abnormal social behavior, and cognitive function. Important cellular responses including cell proliferation, inflammation, and control of infection and their relationship to AVP are described. Finally, the effects of AVP on hemostasis and the hypothalamic-pituitary-adrenal axis are noted. The goal of this summary of the various actions of AVP is to direct attention to the potential benefits of research in these underemphasized areas of importance.
Collapse
Affiliation(s)
- Gaurang P Mavani
- Division of Nephrology, Department of Medicine, Lenox Hill Hospital , New York, NY , USA
| | - Maria V DeVita
- Division of Nephrology, Department of Medicine, Lenox Hill Hospital , New York, NY , USA
| | - Michael F Michelis
- Division of Nephrology, Department of Medicine, Lenox Hill Hospital , New York, NY , USA
| |
Collapse
|
47
|
Weber M, Berglund D, Reule S, Jackson S, Matas AJ, Ibrahim HN. Daily fluid intake and outcomes in kidney recipients: post hoc analysis from the randomized ABCAN trial. Clin Transplant 2015; 29:261-7. [PMID: 25619874 DOI: 10.1111/ctr.12514] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2015] [Indexed: 11/28/2022]
Abstract
Generous and even excessive fluid intake is routinely recommended to kidney transplant recipients despite minimal evidence to support this practice. We hypothesized that increased fluid intake, ascertained by 24-h urine volume output, may adversely affect graft outcomes as it would impose an extra workload on a limited number of nephrons. Kidney transplant recipients who were randomized to losartan vs. placebo in the Angiotensin II Blockade for Chronic Allograft Nephropathy (ABCAN) trial (n = 153) underwent baseline, five-yr biopsies, and annual iothalamate glomerular filtration rate assessment. Recipients with higher urine volume at randomization had higher urinary sodium and also higher urinary protein. The proportion using diuretics or CNI based regimens were similar across urinary volume tertiles. The highest urinary volume tertile (>2.56 L/d) did not predict the development of interstitial volume doubling or end-stage renal disease (ESRD) from interstitial fibrosis/tubular atrophy (OR = 3.52, 95% CI 0.4, 31.24, p = 0.26), interstitial volume doubling or all-cause ESRD (OR = 7.04, 95% CI 0.66, 74.87, p = 0.11), and was not associated with the conventional endpoint of doubling serum creatinine, all-cause ESRD, or death (OR = 0.89, 95% CI 0.21, 3.71, p = 0.87). These results suggest that the current practice of liberal fluid intake may not be beneficial in low risk and mostly Caucasian transplant recipients.
Collapse
Affiliation(s)
- M Weber
- Division of Renal Diseases and Hypertension, University of Minnesota, Minneapolis, MN, USA
| | | | | | | | | | | |
Collapse
|
48
|
Abstract
The synthesis of nonpeptide orally bioavailable vasopressin antagonists devoid of agonistic activity (vaptans) has made possible the selective blockade of vasopressin receptor subtypes for therapeutic purposes. Vaptans acting on the vasopressin V2 receptors (aquaretics) have attracted attention as a possible therapy for heart failure and polycystic kidney disease. Despite a solid rationale and encouraging preclinical testing, aquaretics have not improved clinical outcomes in randomized clinical trials for heart failure. Additional clinical trials with select population targets, more flexible dosing schedules, and possibly a different drug type or combination (balanced V1a/V2 receptor antagonism) may be warranted. Aquaretics are promising for the treatment of autosomal dominant polycystic kidney disease and have been approved in Japan for this indication. More studies are needed to better define their long-term safety and efficacy and optimize their utilization.
Collapse
Affiliation(s)
- Vicente E Torres
- Division of Nephrology and Hypertension, Mayo Clinic College of Medicine, Rochester, Minnesota 55905;
| |
Collapse
|
49
|
Abstract
PURPOSE OF REVIEW Despite improvements in glycemic and blood pressure control in patients with type 1 diabetes, diabetic nephropathy remains the most common cause of chronic kidney disease worldwide. A major challenge in preventing diabetic nephropathy is the inability to identify high-risk patients at an early stage, emphasizing the importance of discovering new therapeutic targets and implementation of clinical trials to reduce diabetic nephropathy risk. RECENT FINDINGS Limitations of managing patients with diabetic nephropathy with renin-angiotensin-aldosterone system blockade have been identified in recent clinical trials, including the failure of primary prevention studies in T1D and the demonstration of harm with dual renin-angiotensin-aldosterone system blockade. Fortunately, several new targets, including serum uric acid, insulin sensitivity, vasopressin, and sodium-glucose cotransporter-2 inhibition, are promising in the prevention and treatment of diabetic nephropathy. SUMMARY Diabetic nephropathy is characterized by a long clinically silent period without signs or symptoms of disease. There is an urgent need for improved methods of detecting early mediators of renal injury, to ultimately prevent the initiation and progression of diabetic nephropathy. In this review, we will focus on early diabetic nephropathy and summarize potential new therapeutic targets.
Collapse
Affiliation(s)
- Petter Bjornstad
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - David Cherney
- Department of Medicine, Division of Nephrology, Toronto General Hospital, University of Toronto, Ontario, Canada
| | - David M. Maahs
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, United States
- Barbara Davis Center for Diabetes, University of Colorado Denver, Aurora, Colorado, United States
| |
Collapse
|
50
|
Johnson RJ, Rodriguez-Iturbe B, Roncal-Jimenez C, Lanaspa MA, Ishimoto T, Nakagawa T, Correa-Rotter R, Wesseling C, Bankir L, Sanchez-Lozada LG. Hyperosmolarity drives hypertension and CKD--water and salt revisited. Nat Rev Nephrol 2014; 10:415-20. [PMID: 24802066 DOI: 10.1038/nrneph.2014.76] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
An epidemic of chronic kidney disease (CKD) in Mesoamerica is providing new insights into the mechanisms by which salt and water might drive hypertension and CKD. Increasingly, evidence suggests that recurrent dehydration and salt loss might be a mechanism that causes CKD, and experimental studies suggest a key role for increased plasma osmolarity in activating both intrarenal (polyol-fructokinase) and extrarenal (vasopressin) pathways that drive renal injury. Thus, we propose that water and salt might influence blood pressure and kidney disease through the timing and combination of their intake, which affect plasma osmolarity as well as intrarenal and extrarenal mechanisms of renal injury. The type of fluid intake might also be important, as fluids containing fructose can trigger activation of these pathways. Future studies should investigate the effects of salt, sugar and fluid intake on plasma osmolarity as a potential pathogenetic mechanism in renal injury and high blood pressure.
Collapse
Affiliation(s)
- Richard J Johnson
- Division of Nephrology, Eastern Colorado Health Care System, Department of Veteran Affairs, 12700 East 19th Avenue, Room 7015, Aurora, CO 80045, USA
| | - Bernardo Rodriguez-Iturbe
- Universidad del Zulia, Instituto Venezolano de Investigaciones Científicas (IVIC)-Zulia, Maracaibo, Venezuela
| | - Carlos Roncal-Jimenez
- Division of Renal Diseases and Hypertension, University of Colorado, Denver, CO, USA
| | - Miguel A Lanaspa
- Division of Renal Diseases and Hypertension, University of Colorado, Denver, CO, USA
| | - Takuji Ishimoto
- Division of Renal Diseases and Hypertension, University of Colorado, Denver, CO, USA
| | - Takahiko Nakagawa
- Mitsubishi Tanabe-Kyoto (TMK) project, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ricardo Correa-Rotter
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Catharina Wesseling
- Program on Work, Environment and Health in Central America (SALTRA), Central American Institute for Studies on Toxic Substances (IRET), Universidad Nacional, Heredia, Costa Rica
| | - Lise Bankir
- INSERM Unité Mixte de Recherche (UMR)-S 1138/Equipe 2, Centre de Recherche des Cordeliers, Paris, France
| | - Laura G Sanchez-Lozada
- Laboratory of Renal Physiopathology, Intituto Nacional de Cardiología Ignacio Chavez, Mexico City, Mexico
| |
Collapse
|