51
|
Monaghan TF, Dmochowski RR, Verbalis JG, Wein AJ, Lazar JM, Birder LA, Everaert K, Weiss JP, Bliwise DL. First voided volume: A novel approach to characterize nocturia. Neurourol Urodyn 2021; 40:848-854. [PMID: 33604915 DOI: 10.1002/nau.24633] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/15/2021] [Accepted: 02/04/2021] [Indexed: 02/06/2023]
Abstract
AIMS Nocturnal polyuria syndrome (NPS) denotes nocturnal polyuria (NP) in the absence of identifiable contributory factors. The trajectory of nocturnal urine production (NUP; typically expressed as ml/hour) may be useful in delineating between NP patients with versus without NPS, but changes in absolute urine volume, the directly measured substrate for behavioral and pharmacologic interventions targeting nocturnal urine production, have not been well characterized. This study compares the ratio of the first nocturnal voided volume (FNVV) to the nocturnal average voided volume (NAVV) in patients with versus without NPS. METHODS Secondary analysis of 24-h voiding diaries from male patients greater than or equal to 18 years of age with two or more nocturnal voids and NP using two different criteria for NP: NUP greater than or equal to 90 ml/h and nocturnal polyuria index (NPi) greater than or equal to 0.33. Patients with diabetes insipidus and CPAP-adherent obstructive sleep apnea (OSA) were excluded. Patients were divided into 2 groups: secondary NP (OSA, congestive heart failure, and chronic kidney disease) and NPS (absence of edema, diuretic use, and the aforementioned comorbidities). FNVV was defined as the volume of urine accompanying the first nocturic episode. NAVV was defined as nocturnal urine volume/(number of nocturnal voids + 1). The nocturnal urine trajectory ratio (NUTR) was defined as FNVV/NAVV. RESULTS At NUP greater than or equal to 90 ml/h, NUTR was significantly greater in patients with (n = 73) versus without (n = 28) NPS (1.10 [0.89-1.33] vs. 0.91 [0.55-1.15], p = .012). At NPi greater than or equal to 0.33, NUTR was likewise significantly greater in patients with (n = 92) versus without (n = 32) NPS (1.09 [0.90-1.33] vs. 0.91 [0.57-1.17], p = .010). CONCLUSIONS The volume of urine produced in the early hours of sleep is central to identification of NPS in patients with nocturia.
Collapse
Affiliation(s)
- Thomas F Monaghan
- Department of Urology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA.,Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Roger R Dmochowski
- Department of Urological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Joseph G Verbalis
- Division of Endocrinology and Metabolism, Department of Medicine, Georgetown University Medical Center, Washington DC, USA
| | - Alan J Wein
- Division of Urology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jason M Lazar
- Division of Cardiovascular Medicine, Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - Lori A Birder
- Departments of Medicine and Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Karel Everaert
- Department of Urology, Ghent University Hospital, Ghent, Belgium
| | - Jeffrey P Weiss
- Department of Urology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - Donald L Bliwise
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
52
|
Hu G, Wang Z, Zhang R, Sun W, Chen X. The Role of Apelin/Apelin Receptor in Energy Metabolism and Water Homeostasis: A Comprehensive Narrative Review. Front Physiol 2021; 12:632886. [PMID: 33679444 PMCID: PMC7928310 DOI: 10.3389/fphys.2021.632886] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 01/21/2021] [Indexed: 11/13/2022] Open
Abstract
The apelin receptor (APJ) is a member of the family A of G-protein-coupled receptors (GPCRs) and is involved in range of physiological and pathological functions, including fluid homeostasis, anxiety, and depression, as well as cardiovascular and metabolic disorders. APJ was classically described as a monomeric transmembrane receptor that forms a ternary complex together with its ligand and associated G proteins. More recently, increasing evidence indicates that APJ may interact with other GPCRs to form heterodimers, which may selectively modulate distinct intracellular signal transduction pathways. Besides, the apelin/APJ system plays important roles in the physiology and pathophysiology of several organs, including regulation of blood pressure, cardiac contractility, angiogenesis, metabolic balance, and cell proliferation, apoptosis, or inflammation. Additionally, the apelin/APJ system is widely expressed in the central nervous system, especially in neurons and oligodendrocytes. This article reviews the role of apelin/APJ in energy metabolism and water homeostasis. Compared with the traditional diuretics, apelin exerts a positive inotropic effect on the heart, while increases water excretion. Therefore, drugs targeting apelin/APJ system undoubtedly provide more therapeutic options for patients with congestive heart failure accompanied with hyponatremia. To provide more precise guidance for the development of clinical drugs, further in-depth studies are warranted on the metabolism and signaling pathways associated with apelin/APJ system.
Collapse
Affiliation(s)
- Gonghui Hu
- Department of Physiology, Shandong First Medical University (Shandong Academy of Medical Sciences), Taian, China
| | - Zhen Wang
- Neurobiology Institute, Jining Medical University, Jining, China
| | - Rumin Zhang
- Neurobiology Institute, Jining Medical University, Jining, China
| | - Wenping Sun
- Department of Pathology, Shandong First Medical University (Shandong Academy of Medical Sciences), Taian, China
| | - Xiaoyu Chen
- Department of Physiology, Shandong First Medical University (Shandong Academy of Medical Sciences), Taian, China
| |
Collapse
|
53
|
Inutsuka A, Ino D, Onaka T. Detection of neuropeptides in vivo and open questions for current and upcoming fluorescent sensors for neuropeptides. Peptides 2021; 136:170456. [PMID: 33245950 DOI: 10.1016/j.peptides.2020.170456] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/27/2020] [Accepted: 11/10/2020] [Indexed: 12/12/2022]
Abstract
During a stress response, various neuropeptides are secreted in a spatiotemporally coordinated way in the brain. For a precise understanding of peptide functions in a stress response, it is important to investigate when and where they are released, how they diffuse, and how they are broken down in the brain. In the past two decades, genetically encoded fluorescent calcium indicators have greatly advanced our knowledge of the functions of specific neuronal activity in regulation of behavioral changes and physiological responses during stress. In addition, various kinds of structural information on G-protein-coupled receptors (GPCRs) for neuropeptides have been revealed. Recently, genetically encoded fluorescent sensors have been developed for detection of neurotransmitters by making use of conformational changes induced by ligand binding. In this review, we summarize the recent and upcoming advances of techniques for detection of neuropeptides and then present several open questions that will be solved by application of recent or upcoming technical advances in detection of neuropeptides in vivo.
Collapse
Affiliation(s)
- Ayumu Inutsuka
- Department of Physiology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan.
| | - Daisuke Ino
- Department of Histology and Cell Biology, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa 920-8640, Japan
| | - Tatsushi Onaka
- Department of Physiology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan.
| |
Collapse
|
54
|
Sedaghat G, Montazerifar F, Keykhaie MA, Karajibani M, Shourestani S, Dashipour A. Effect of pre-meal water intake on the serum levels of Copeptin, glycemic control, lipid profile and anthropometric indices in patients with type 2 diabetes mellitus: a randomized, controlled trial. J Diabetes Metab Disord 2021; 20:171-177. [PMID: 34178828 DOI: 10.1007/s40200-020-00724-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/28/2020] [Indexed: 12/15/2022]
Abstract
Background Water pre-load affects insulin secretion by altering the level of copeptin (C-terminal component of the arginine vasopressin hormone (AVP)) and preventing obesity by reducing food intake. Aims The present randomized controlled trial (RCT) aimed to investigate the effects of pre-meal water intake on type 2 diabetes Mellitus (T2DM). Materials and methods In this study, 40 patients with T2DM were randomly assigned to two intervention groups for 8 weeks; a) drinking 1 liter of water per day before each main meal (PW group)., and b) no water consumption before any meal (NPW group). At the beginning and at the end of the study, blood samples were taken to assess glycemic indices, lipid profile, copeptin and anthropometric indices. Results Pre-meal water intake was associated with lower energy intake, BMI, waist circumference (WC) and greater weight loss, in compared with the controls (P < 0.0001) after 8 weeks. At the end of the trial, the concentrations of fasting blood sugar (FBS) (P < 0.0001), triglyceride (TG) (P < 0.05), low-density lipoprotein cholesterol (LDL-C) (P < 0.05) and copeptin (P < 0.05) were significantly reduced following water drinking before meals. Conclusion To sum up, the present study revealed that pre-meal water intake is associated with lower BMI, body weight, WC, FBS, TG, LDL-C and copeptin levels in patients with T2DM.
Collapse
Affiliation(s)
- Gohar Sedaghat
- Department of Nutrition, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Farzaneh Montazerifar
- Pregnancy Health Research Center, Department of Nutrition, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mahmood Ali Keykhaie
- Genetics of Non-Communicable Disease Research Center, Department of Internal, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mansour Karajibani
- Health Promotion Research Center, Department of Nutrition, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Shadi Shourestani
- Department of Nutrition, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Alireza Dashipour
- Cellular and Molecular Research Center, Department of Food and Technology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
55
|
Hoffiz YC, Castillo-Ruiz A, Hall MAL, Hite TA, Gray JM, Cisternas CD, Cortes LR, Jacobs AJ, Forger NG. Birth elicits a conserved neuroendocrine response with implications for perinatal osmoregulation and neuronal cell death. Sci Rep 2021; 11:2335. [PMID: 33504846 PMCID: PMC7840942 DOI: 10.1038/s41598-021-81511-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/05/2021] [Indexed: 12/17/2022] Open
Abstract
Long-standing clinical findings report a dramatic surge of vasopressin in umbilical cord blood of the human neonate, but the neural underpinnings and function(s) of this phenomenon remain obscure. We studied neural activation in perinatal mice and rats, and found that birth triggers activation of the suprachiasmatic, supraoptic, and paraventricular nuclei of the hypothalamus. This was seen whether mice were born vaginally or via Cesarean section (C-section), and when birth timing was experimentally manipulated. Neuronal phenotyping showed that the activated neurons were predominantly vasopressinergic, and vasopressin mRNA increased fivefold in the hypothalamus during the 2–3 days before birth. Copeptin, a surrogate marker of vasopressin, was elevated 30-to 50-fold in plasma of perinatal mice, with higher levels after a vaginal than a C-section birth. We also found an acute decrease in plasma osmolality after a vaginal, but not C-section birth, suggesting that the difference in vasopressin release between birth modes is functionally meaningful. When vasopressin was administered centrally to newborns, we found an ~ 50% reduction in neuronal cell death in specific brain areas. Collectively, our results identify a conserved neuroendocrine response to birth that is sensitive to birth mode, and influences peripheral physiology and neurodevelopment.
Collapse
Affiliation(s)
- Yarely C Hoffiz
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA
| | | | - Megan A L Hall
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA
| | - Taylor A Hite
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA
| | - Jennifer M Gray
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA
| | - Carla D Cisternas
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA.,Instituto de Investigación Médica M Y M Ferreyra, INIMEC-CONICET-UNC, Córdoba, Argentina
| | - Laura R Cortes
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA
| | - Andrew J Jacobs
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA
| | - Nancy G Forger
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA.
| |
Collapse
|
56
|
Wood C. Fluid management: An update for perioperative practitioners. J Perioper Pract 2021; 31:71-79. [PMID: 33472531 DOI: 10.1177/1750458920964174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
An interprofessional team approach is required to achieve optimum fluid balance for patients during the perioperative period. Incorrect management of fluid assessment and monitoring is associated with adverse outcomes. The scientific understanding of perioperative fluid balance has improved over recent years leading to changes in clinical practice with regard to volume and choice of intravenous fluid. It is important that perioperative practitioners have an understanding of intravenous fluid, fluid compartmentalisation, fluid mechanics and intravascular fluid control mechanisms. Optimum fluid status not only shortens hospital stay but also reduces the incidence of postoperative nausea and vomiting and complication profiles. This article aims to provide perioperative practitioners with a comprehensive overview of fluid management. It will cover important issues surrounding physiological control of fluid balance, choice of intravenous fluid therapy, methods to monitor intravascular volume and factors which influence delivery.
Collapse
|
57
|
Sparapani S, Millet-Boureima C, Oliver J, Mu K, Hadavi P, Kalostian T, Ali N, Avelar CM, Bardies M, Barrow B, Benedikt M, Biancardi G, Bindra R, Bui L, Chihab Z, Cossitt A, Costa J, Daigneault T, Dault J, Davidson I, Dias J, Dufour E, El-Khoury S, Farhangdoost N, Forget A, Fox A, Gebrael M, Gentile MC, Geraci O, Gnanapragasam A, Gomah E, Haber E, Hamel C, Iyanker T, Kalantzis C, Kamali S, Kassardjian E, Kontos HK, Le TBU, LoScerbo D, Low YF, Mac Rae D, Maurer F, Mazhar S, Nguyen A, Nguyen-Duong K, Osborne-Laroche C, Park HW, Parolin E, Paul-Cole K, Peer LS, Philippon M, Plaisir CA, Porras Marroquin J, Prasad S, Ramsarun R, Razzaq S, Rhainds S, Robin D, Scartozzi R, Singh D, Fard SS, Soroko M, Soroori Motlagh N, Stern K, Toro L, Toure MW, Tran-Huynh S, Trépanier-Chicoine S, Waddingham C, Weekes AJ, Wisniewski A, Gamberi C. The Biology of Vasopressin. Biomedicines 2021; 9:89. [PMID: 33477721 PMCID: PMC7832310 DOI: 10.3390/biomedicines9010089] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/29/2020] [Accepted: 01/06/2021] [Indexed: 02/07/2023] Open
Abstract
Vasopressins are evolutionarily conserved peptide hormones. Mammalian vasopressin functions systemically as an antidiuretic and regulator of blood and cardiac flow essential for adapting to terrestrial environments. Moreover, vasopressin acts centrally as a neurohormone involved in social and parental behavior and stress response. Vasopressin synthesis in several cell types, storage in intracellular vesicles, and release in response to physiological stimuli are highly regulated and mediated by three distinct G protein coupled receptors. Other receptors may bind or cross-bind vasopressin. Vasopressin is regulated spatially and temporally through transcriptional and post-transcriptional mechanisms, sex, tissue, and cell-specific receptor expression. Anomalies of vasopressin signaling have been observed in polycystic kidney disease, chronic heart failure, and neuropsychiatric conditions. Growing knowledge of the central biological roles of vasopressin has enabled pharmacological advances to treat these conditions by targeting defective systemic or central pathways utilizing specific agonists and antagonists.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Chiara Gamberi
- Biology Department, Concordia University, Montreal, QC H4B 1R6, Canada; (S.S.); (C.M.-B.); (J.O.); (K.M.); (P.H.); (T.K.); (N.A.); (C.M.A.); (M.B.); (B.B.); (M.B.); (G.B.); (R.B.); (L.B.); (Z.C.); (A.C.); (J.C.); (T.D.); (J.D.); (I.D.); (J.D.); (E.D.); (S.E.-K.); (N.F.); (A.F.); (A.F.); (M.G.); (M.C.G.); (O.G.); (A.G.); (E.G.); (E.H.); (C.H.); (T.I.); (C.K.); (S.K.); (E.K.); (H.K.K.); (T.B.U.L.); (D.L.); (Y.F.L.); (D.M.R.); (F.M.); (S.M.); (A.N.); (K.N.-D.); (C.O.-L.); (H.W.P.); (E.P.); (K.P.-C.); (L.S.P.); (M.P.); (C.-A.P.); (J.P.M.); (S.P.); (R.R.); (S.R.); (S.R.); (D.R.); (R.S.); (D.S.); (S.S.F.); (M.S.); (N.S.M.); (K.S.); (L.T.); (M.W.T.); (S.T.-H.); (S.T.-C.); (C.W.); (A.J.W.); (A.W.)
| |
Collapse
|
58
|
Schiller M, Ben-Shaanan TL, Rolls A. Neuronal regulation of immunity: why, how and where? Nat Rev Immunol 2021; 21:20-36. [PMID: 32811994 DOI: 10.1038/s41577-020-0387-1] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2020] [Indexed: 02/07/2023]
Abstract
Neuroimmunology is one of the fastest-growing fields in the life sciences, and for good reason; it fills the gap between two principal systems of the organism, the nervous system and the immune system. Although both systems affect each other through bidirectional interactions, we focus here on one direction - the effects of the nervous system on immunity. First, we ask why is it beneficial to allow the nervous system any control over immunity? We evaluate the potential benefits to the immune system that arise by taking advantage of some of the brain's unique features, such as its capacity to integrate and synchronize physiological functions, its predictive capacity and its speed of response. Second, we explore how the brain communicates with the peripheral immune system, with a focus on the endocrine, sympathetic, parasympathetic, sensory and meningeal lymphatic systems. Finally, we examine where in the brain this immune information is processed and regulated. We chart a partial map of brain regions that may be relevant for brain-immune system communication, our goal being to introduce a conceptual framework for formulating new hypotheses to study these interactions.
Collapse
Affiliation(s)
- Maya Schiller
- Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Tamar L Ben-Shaanan
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Asya Rolls
- Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
59
|
Monaghan TF, Weiss JP, Everaert K, Wein AJ. Pharmacologic management of nocturnal polyuria: a contemporary assessment of efficacy, safety, and progress toward individualized treatment. Ther Adv Urol 2021; 13:1756287220988438. [PMID: 33796148 PMCID: PMC7970679 DOI: 10.1177/1756287220988438] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 12/26/2020] [Indexed: 12/18/2022] Open
Abstract
This narrative review synthesizes current evidence on the medical management of nocturnal polyuria, including antidiuretic replacement therapy as well as other emerging modalities, with particular emphasis on areas of active investigation and future research directions. Relative to earlier formulations, the pharmacological profiles of novel desmopressin acetate nasal spray and orally disintegrating tablet formulations appear favorable in optimizing the balance between efficacy and safety. Additionally, several highly selective small-molecule arginine vasopressin 2 receptor agonists are under active development, while appropriately timed short-acting diuretics, pharmacotherapy for hypertension, nonsteroidal anti-inflammatory drugs, and sex hormone replacement therapy are also a focal point of extensive ongoing nocturnal polyuria research. Emerging laboratory technologies now make feasible a sub-stratification of nocturnal polyuria patients into substrate-based phenotypes for individualized treatment. An increasingly refined understanding of the pathogenesis of nocturnal polyuria, and arginine vasopressin dysregulation in particular, has also introduced new opportunities for point-of-care testing in patients with nocturnal polyuria.
Collapse
Affiliation(s)
- Thomas F. Monaghan
- Department of Urology SUNY Downstate Health Sciences University, 450 Clarkson Avenue, Box 79, Brooklyn, New York 11203, USA
| | - Jeffrey P. Weiss
- Department of Urology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - Karel Everaert
- Department of Urology, Ghent University Hospital, Ghent, Belgium
| | - Alan J. Wein
- Division of Urology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
60
|
Himmel NJ, Rogers RT, Redd SK, Wang Y, Blount MA. Purinergic signaling is enhanced in the absence of UT-A1 and UT-A3. Physiol Rep 2021; 9:e14636. [PMID: 33369887 PMCID: PMC7769175 DOI: 10.14814/phy2.14636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 11/24/2022] Open
Abstract
ATP is an important paracrine regulator of renal tubular water and urea transport. The activity of P2Y2 , the predominant P2Y receptor of the medullary collecting duct, is mediated by ATP, and modulates urinary concentration. To investigate the role of purinergic signaling in the absence of urea transport in the collecting duct, we studied wild-type (WT) and UT-A1/A3 null (UT-A1/A3 KO) mice in metabolic cages to monitor urine output, and collected tissue samples for analysis. We confirmed that UT-A1/A3 KO mice are polyuric, and concurrently observed lower levels of urinary cAMP as compared to WT, despite elevated serum vasopressin (AVP) levels. Because P2Y2 inhibits AVP-stimulated transport by dampening cAMP synthesis, we suspected that, similar to other models of AVP-resistant polyuria, purinergic signaling is increased in UT-A1/A3 KO mice. In fact, we observed that both urinary ATP and purinergic-mediated prostanoid (PGE2 ) levels were elevated. Collectively, our data suggest that the reduction of medullary osmolality due to the lack of UT-A1 and UT-A3 induces an AVP-resistant polyuria that is possibly exacerbated by, or at least correlated with, enhanced purinergic signaling.
Collapse
Affiliation(s)
- Nathaniel J. Himmel
- Renal DivisionDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
| | - Richard T. Rogers
- Renal DivisionDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
| | - Sara K. Redd
- Renal DivisionDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
| | - Yirong Wang
- Renal DivisionDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
| | - Mitsi A. Blount
- Renal DivisionDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
- Department of PhysiologyEmory University School of MedicineAtlantaGAUSA
| |
Collapse
|
61
|
Saini AK, Saini R, Singh S. Autosomal dominant polycystic kidney disease and pioglitazone for its therapy: a comprehensive review with an emphasis on the molecular pathogenesis and pharmacological aspects. Mol Med 2020; 26:128. [PMID: 33308138 PMCID: PMC7731470 DOI: 10.1186/s10020-020-00246-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is an inherited chronic kidney disorder (CKD) that is characterized by the development of numerous fluid-filled cysts in kidneys. It is caused either due to the mutations in the PKD1 or PKD2 gene that encodes polycystin-1 and polycystin-2, respectively. This condition progresses into end-stage renal disorder if the renal or extra-renal clinical manifestations remain untreated. Several clinical trials with a variety of drugs have failed, and the only Food and Drugs Administration (FDA) approved drug to treat ADPKD to date is tolvaptan that works by antagonizing the vasopressin-2 receptor (V2R). The pathology of ADPKD is complex and involves the malfunction of different signaling pathways like cAMP, Hedgehog, and MAPK/ERK pathway owing to the mutated product that is polycystin-1 or 2. A measured yet substantial number of preclinical studies have found pioglitazone to decrease the cystic burden and improve the renal function in ADPKD. The peroxisome proliferator-activated receptor-gamma is found on the epithelial cells of renal collecting tubule and when it gets agonized by pioglitazone, confers efficacy in ADPKD treatment through multiple mechanisms. There is only one clinical trial (ongoing) wherein it is being assessed for its benefits and risk in patients with ADPKD, and is expected to get approval from the regulatory body owing to its promising therapeutic effects. This article would encompass the updated information on the epidemiology, pathophysiology of ADPKD, different mechanisms of action of pioglitazone in the treatment of ADPKD with preclinical and clinical shreds of evidence, and related safety updates.
Collapse
Affiliation(s)
- Aryendu Kumar Saini
- Department of Pharmacy, Chaudhary Sughar Singh College of Pharmacy, Etawah, Uttar Pradesh, India.
| | - Rakesh Saini
- Department of Pharmacy, Chaudhary Sughar Singh College of Pharmacy, Etawah, Uttar Pradesh, India
| | - Shubham Singh
- Department of Pharmacy, Shri Ram Lakhan Tiwari College of Pharmacy, Etawah, Uttar Pradesh, India
| |
Collapse
|
62
|
Rotem-Grunbaum B, Landau D. Genetic renal disease classification by hormonal axes. Pediatr Nephrol 2020; 35:2211-2219. [PMID: 31828468 DOI: 10.1007/s00467-019-04437-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 11/23/2019] [Accepted: 11/26/2019] [Indexed: 12/31/2022]
Abstract
The kidneys, which regulate many homeostatic pathways, are also a major endocrinological target organ. Many genetic renal diseases can be classified according to the affected protein along such endocrinological pathways. In this review, we examine the hypothesis that a more severe phenotype is expected as the affected protein is located more distally along such pathways. Thus, the location of a defect along its endocrinological pathway should be taken into consideration, in addition to the mutation type, when assessing genetic renal disease severity.
Collapse
Affiliation(s)
- Bar Rotem-Grunbaum
- Department of Pediatrics B, Schneider Children's Medical Center of Israel, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Daniel Landau
- Department of Pediatrics B, Schneider Children's Medical Center of Israel, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
63
|
Bahadoram M, Mahmoudian-Sani MR, Keikhaei B, Alikhani K, Bahadoram S. The antimigraine action of arginine-vasopressin: a theoretical basis. FUTURE NEUROLOGY 2020. [DOI: 10.2217/fnl-2020-0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Mohammad Bahadoram
- Thalassemia & Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad-Reza Mahmoudian-Sani
- Thalassemia & Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Bijan Keikhaei
- Thalassemia & Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Kosar Alikhani
- Thalassemia & Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sara Bahadoram
- Thalassemia & Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Pediatrics, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
64
|
Zuo J, Guo W, Wang S, Lang Y, Wang S, Shi X, Zhang R, Zhao X, Han Y, Shao L. Eight novel KCNJ1 variants and parathyroid hormone overaction or resistance in 5 probands with Bartter syndrome type 2. Clin Chim Acta 2020; 511:248-254. [PMID: 33058840 DOI: 10.1016/j.cca.2020.10.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/07/2020] [Accepted: 10/02/2020] [Indexed: 01/19/2023]
Abstract
PURPOSE Bartter syndrome type 2 (BS2) is an autosomal recessive renal tubular disorder, which is caused by the mutations in KCNJ1. This study was designed to analyze and describe the genotype and clinical features of five Chinese probands with BS2. METHODS Identify KCNJ1 gene variants by the next generation sequencing and evaluate their mutation effects according to 2015 American College of Medical Genetics and Genomics (ACMG) standards and guidelines. RESULTS Ten variants including eight novel ones of KCNJ1 gene were found, the most common type was missense variant. The common symptoms and signs from high to low incidence were: polydipsia and polyuria (5/5), one of them (1/5) presented with diabetes insipidus; maternal polyhydramnios and premature delivery (4/5); growth retardation (3/5). Two patients presented with hypochloremic metabolic alkalosis and hypokalemia; whereas the acid-base disturbance was absent in the others. One patient had evident parathyroid hormone (PTH) resistance (hypocalcemia, hyperphosphatemia and markedly elevated PTH levels), three presented with PTH overacting (hypercalcemia, hypophosphatemia and mild elevated PTH levels), and one showed normal blood calcium and phosphorus concentrations with high-normal PTH levels. All patients had nephrocalcinosis and/or hypercalciuria, and one of them complicated with nephrolithiasis. Indomethacin has significant therapeutic effect on the growth retardation, polydipsia and polyuria and treatment was associated with a decrease in urine calcium excretion, normalization of electrolyte disturbance and PTH parameters. CONCLUSIONS Ten variants of KCNJ1 gene were identified in five Chinese probands. These patients had atypical BS phenotype lacking evident metabolic alkalosis and/or manifesting with PTH overaction/resistance, which reminds clinicians to carefully differentiate BS2 with other parathyroid disorders. This is the first report of BS2 from Chinese populations.
Collapse
Affiliation(s)
- Jianxin Zuo
- Department of Obstetrics, the Affiliated Hospital of Qingdao University, Qingdao 266003, People's Republic of China; Department of Nephrology, the Affiliated Qingdao Municipal Hospital of Qingdao University, No.5 Donghai Middle Road, Qingdao 266071, People's Republic of China
| | - Wencong Guo
- Department of Nephrology, the Affiliated Qingdao Municipal Hospital of Qingdao University, No.5 Donghai Middle Road, Qingdao 266071, People's Republic of China; Department of Nephrology, the Affiliated Qingdao Municipal Hospital of Shandong University, No.5 Donghai Middle Road, Qingdao 266071, People's Republic of China; Central Laboratory, the Affiliated Hospital of Qingdao University, Qingdao 266003, People's Republic of China
| | - Shujuan Wang
- Department of Nursing, the Affiliated Hospital of Qingdao University, Qingdao 266003, People's Republic of China
| | - Yanhua Lang
- Department of Nursing, the Affiliated Hospital of Qingdao University, Qingdao 266003, People's Republic of China
| | - Sai Wang
- Department of Nephrology, the Affiliated Qingdao Municipal Hospital of Qingdao University, No.5 Donghai Middle Road, Qingdao 266071, People's Republic of China; Department of Nephrology, the Affiliated Qingdao Municipal Hospital of Shandong University, No.5 Donghai Middle Road, Qingdao 266071, People's Republic of China; Central Laboratory, the Affiliated Hospital of Qingdao University, Qingdao 266003, People's Republic of China
| | - Xiaomeng Shi
- Department of Nephrology, the Affiliated Qingdao Municipal Hospital of Qingdao University, No.5 Donghai Middle Road, Qingdao 266071, People's Republic of China; Department of Nephrology, the Affiliated Qingdao Municipal Hospital of Shandong University, No.5 Donghai Middle Road, Qingdao 266071, People's Republic of China; Central Laboratory, the Affiliated Hospital of Qingdao University, Qingdao 266003, People's Republic of China
| | - Ruixiao Zhang
- Department of Nephrology, the Affiliated Qingdao Municipal Hospital of Qingdao University, No.5 Donghai Middle Road, Qingdao 266071, People's Republic of China; Department of Nephrology, the Affiliated Qingdao Municipal Hospital of Shandong University, No.5 Donghai Middle Road, Qingdao 266071, People's Republic of China; Central Laboratory, the Affiliated Hospital of Qingdao University, Qingdao 266003, People's Republic of China
| | - Xiangzhong Zhao
- Central Laboratory, the Affiliated Hospital of Qingdao University, Qingdao 266003, People's Republic of China
| | - Yue Han
- Department of Nephrology, the Affiliated Qingdao Municipal Hospital of Qingdao University, No.5 Donghai Middle Road, Qingdao 266071, People's Republic of China; Department of Nephrology, the Affiliated Qingdao Municipal Hospital of Shandong University, No.5 Donghai Middle Road, Qingdao 266071, People's Republic of China; Central Laboratory, the Affiliated Hospital of Qingdao University, Qingdao 266003, People's Republic of China.
| | - Leping Shao
- Department of Nephrology, the Affiliated Qingdao Municipal Hospital of Qingdao University, No.5 Donghai Middle Road, Qingdao 266071, People's Republic of China; Department of Nephrology, the Affiliated Qingdao Municipal Hospital of Shandong University, No.5 Donghai Middle Road, Qingdao 266071, People's Republic of China; Central Laboratory, the Affiliated Hospital of Qingdao University, Qingdao 266003, People's Republic of China.
| |
Collapse
|
65
|
The mosaic genome of indigenous African cattle as a unique genetic resource for African pastoralism. Nat Genet 2020; 52:1099-1110. [PMID: 32989325 DOI: 10.1038/s41588-020-0694-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 08/18/2020] [Indexed: 02/07/2023]
Abstract
Cattle pastoralism plays a central role in human livelihood in Africa. However, the genetic history of its success remains unknown. Here, through whole-genome sequence analysis of 172 indigenous African cattle from 16 breeds representative of the main cattle groups, we identify a major taurine × indicine cattle admixture event dated to circa 750-1,050 yr ago, which has shaped the genome of today's cattle in the Horn of Africa. We identify 16 loci linked to African environmental adaptations across crossbred animals showing an excess of taurine or indicine ancestry. These include immune-, heat-tolerance- and reproduction-related genes. Moreover, we identify one highly divergent locus in African taurine cattle, which is putatively linked to trypanotolerance and present in crossbred cattle living in trypanosomosis-infested areas. Our findings indicate that a combination of past taurine and recent indicine admixture-derived genetic resources is at the root of the present success of African pastoralism.
Collapse
|
66
|
Kim K, Mody N, Chernoff A, Gupta S, Lekprasert P, Patarroyo-Aponte G. Transient diabetes insipidus after vasopressin use in a patient with Wernicke's encephalopathy. JOURNAL OF CLINICAL AND TRANSLATIONAL ENDOCRINOLOGY CASE REPORTS 2020. [DOI: 10.1016/j.jecr.2020.100067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
67
|
Dwivedi N, Tao S, Jamadar A, Sinha S, Howard C, Wallace DP, Fields TA, Leask A, Calvet JP, Rao R. Epithelial Vasopressin Type-2 Receptors Regulate Myofibroblasts by a YAP-CCN2-Dependent Mechanism in Polycystic Kidney Disease. J Am Soc Nephrol 2020; 31:1697-1710. [PMID: 32554753 DOI: 10.1681/asn.2020020190] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/13/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Fibrosis is a major cause of loss of renal function in autosomal dominant polycystic kidney disease (ADPKD). In this study, we examined whether vasopressin type-2 receptor (V2R) activity in cystic epithelial cells can stimulate interstitial myofibroblasts and fibrosis in ADPKD kidneys. METHODS We treated Pkd1 gene knockout (Pkd1KO) mice with dDAVP, a V2R agonist, for 3 days and evaluated the effect on myofibroblast deposition of extracellular matrix (ECM). We also analyzed the effects of conditioned media from primary cultures of human ADPKD cystic epithelial cells on myofibroblast activation. Because secretion of the profibrotic connective tissue growth factor (CCN2) increased significantly in dDAVP-treated Pkd1KO mouse kidneys, we examined its role in V2R-dependent fibrosis in ADPKD as well as that of yes-associated protein (YAP). RESULTS V2R stimulation using dDAVP increased the renal interstitial myofibroblast population and ECM deposition. Similarly, conditioned media from human ADPKD cystic epithelial cells increased myofibroblast activation in vitro, suggesting a paracrine mechanism. Renal collecting duct-specific gene deletion of CCN2 significantly reduced cyst growth and myofibroblasts in Pkd1KO mouse kidneys. We found that YAP regulates CCN2, and YAP inhibition or gene deletion reduces renal fibrosis in Pkd1KO mouse kidneys. Importantly, YAP inactivation blocks the dDAVP-induced increase in myofibroblasts in Pkd1KO kidneys. Further in vitro studies showed that V2R regulates YAP by an ERK1/2-dependent mechanism in human ADPKD cystic epithelial cells. CONCLUSIONS Our results demonstrate a novel mechanism by which cystic epithelial cells stimulate myofibroblasts in the pericystic microenvironment, leading to fibrosis in ADPKD. The V2R-YAP-CCN2 cell signaling pathway may present a potential therapeutic target for fibrosis in ADPKD.
Collapse
Affiliation(s)
- Nidhi Dwivedi
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas.,Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Shixin Tao
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas.,Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Abeda Jamadar
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas.,Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Sonali Sinha
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas.,Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Christianna Howard
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas.,Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Darren P Wallace
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas.,Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Timothy A Fields
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas.,Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Andrew Leask
- School of Dentistry, University of Saskatchewan, Saskatoon, SK, Canada
| | - James P Calvet
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas.,Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - Reena Rao
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas .,Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
68
|
Sorting Nexin 27 Regulates the Lysosomal Degradation of Aquaporin-2 Protein in the Kidney Collecting Duct. Cells 2020; 9:cells9051208. [PMID: 32413996 PMCID: PMC7290579 DOI: 10.3390/cells9051208] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/01/2020] [Accepted: 05/11/2020] [Indexed: 12/16/2022] Open
Abstract
Sorting nexin 27 (SNX27), a PDZ (Postsynaptic density-95/Discs large/Zonula occludens 1) domain-containing protein, cooperates with a retromer complex, which regulates intracellular trafficking and the abundance of membrane proteins. Since the carboxyl terminus of aquaporin-2 (AQP2c) has a class I PDZ-interacting motif (X-T/S-X-Φ), the role of SNX27 in the regulation of AQP2 was studied. Co-immunoprecipitation assay of the rat kidney demonstrated an interaction of SNX27 with AQP2. Glutathione S-transferase (GST) pull-down assays revealed an interaction of the PDZ domain of SNX27 with AQP2c. Immunocytochemistry of HeLa cells co-transfected with FLAG-SNX27 and hemagglutinin (HA)-AQP2 also revealed co-localization throughout the cytoplasm. When the PDZ domain was deleted, punctate HA-AQP2 labeling was localized in the perinuclear region. The labeling was intensively overlaid by Lysotracker staining but not by GM130 labeling, a cis-Golgi marker. In rat kidneys and primary cultured inner medullary collecting duct cells, the subcellular redistribution of SNX27 was similar to AQP2 under 1-deamino-8-D-arginine vasopressin (dDAVP) stimulation/withdrawal. Cell surface biotinylation assay showed that dDAVP-induced AQP2 translocation to the apical plasma membrane was unaffected after SNX27 knockdown in mpkCCD cells. In contrast, the dDAVP-induced AQP2 protein abundance was significantly attenuated without changes in AQP2 mRNA expression. Moreover, the AQP2 protein abundance was markedly declined during the dDAVP withdrawal period after stimulation under SNX27 knockdown, which was inhibited by lysosome inhibitors. Autophagy was induced after SNX27 knockdown in mpkCCD cells. Lithium-induced nephrogenic diabetes insipidus in rats revealed a significant downregulation of SNX27 in the kidney inner medulla. Taken together, the PDZ domain-containing SNX27 interacts with AQP2 and depletion of SNX27 contributes to the autophagy-lysosomal degradation of AQP2.
Collapse
|
69
|
Liang H, Li W, Yang H, Cao Y, Ge L, Shi R, Fan Z, Dong R, Zhang C. FAM96B inhibits the senescence of dental pulp stem cells. Cell Biol Int 2020; 44:1193-1203. [DOI: 10.1002/cbin.11319] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 02/02/2020] [Indexed: 01/12/2023]
Affiliation(s)
- Hanbing Liang
- Department of EndodonticsCapital Medical University School of Stomatology Beijing 100050 China
| | - Wenzhi Li
- Department of EndodonticsCapital Medical University School of Stomatology Beijing 100050 China
| | - Haoqing Yang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function ReconstructionCapital Medical University School of Stomatology Beijing 100050 China
| | - Yangyang Cao
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function ReconstructionCapital Medical University School of Stomatology Beijing 100050 China
| | - Lihua Ge
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function ReconstructionCapital Medical University School of Stomatology Beijing 100050 China
| | - Ruitang Shi
- Department of EndodonticsCapital Medical University School of Stomatology Beijing 100050 China
| | - Zhipeng Fan
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function ReconstructionCapital Medical University School of Stomatology Beijing 100050 China
| | - Rui Dong
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function ReconstructionCapital Medical University School of Stomatology Beijing 100050 China
| | - Chen Zhang
- Department of EndodonticsCapital Medical University School of Stomatology Beijing 100050 China
| |
Collapse
|
70
|
Patel S, Volpe AB, Awwad S, Schätzlein AG, Haider S, Liu B, Uchegbu IF. A Self-Assembling Lipidic Peptide and Selective Partial V2 Receptor Agonist Inhibits Urine Production. Sci Rep 2020; 10:7269. [PMID: 32350300 PMCID: PMC7190706 DOI: 10.1038/s41598-020-64070-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 02/21/2020] [Indexed: 11/09/2022] Open
Abstract
Lipidised analgesic peptide prodrugs self-assemble into peptide nanofibers; with the nanofiber morphology protecting the peptide from plasma degradation and improving therapeutic efficacy. Extending this learning, we hypothesised that a self-assembling lipidized peptide arginine vasopressin (AVP) receptor agonist, that had not been designed as a prodrug, could prove pharmacologically active and control urine production. The only approved AVP receptor agonist, desmopressin is indicated for the treatment of central diabetes insipidus (DI), bedwetting, haemophilia A and von Willebrand disease. Desmopressin is well tolerated by most patients, however adverse effects, such as hyponatraemia and water intoxication necessitate a strict fluid intake, thus motivating the search for alternative DI treatments. Selective V2 receptor agonism is required for anti-DI activity and we hypothesised that our new lipidized peptide (METx) would lead to selective AVP receptor agonism. METx was synthesised and characterised and then tested for activity against the V2, V1a and OT uterine receptors and not tested against the V1b receptor as METx was not expected to cross the blood brain barrier. METx was also tested in vivo in a healthy rat model. METx forms nanofibers and is a partial V2 receptor agonist (determined by measuring MDCK cell line cAMP accumulation), producing 57% of AVP's maximal activity (EC50 = 2.7 nM) and is not a V1a agonist up to a concentration of 1 μM (determined by measuring A7r5 cell line D-myo-inositol-1-phosphate accumulation). METx is a weak OT receptor antagonist, reducing the frequency of OT induced contractions (EC50 = 350 nM) and increasing the OT EC50 from 0.081 nM to 21 nM at a concentration of 600 nM. METx (41 nM) had no effect on spontaneous uterine contractions and METx (100 nM) had no effect on OT induced uterine contractions. Simulated binding studies show that binding avidity to the receptors follows the trend: V2 > OT > V1a. On intravenous injection, a nanoparticle formulation of METx reduced urine production in a healthy rat model in a dose responsive manner, with 40 mg kg-1 METx resulting in no urine production over 4 hours. The lipidized self-assembling peptide - METx - is a selective competitive V2 receptor agonist and an anti-diuretic.
Collapse
Affiliation(s)
- Sunish Patel
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | | | - Sahar Awwad
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Andreas G Schätzlein
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, UK.,Nanomerics Ltd. 30-34 New Bridge Street, London, EC4V 6BJ, UK
| | - Shozeb Haider
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Boqian Liu
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Ijeoma F Uchegbu
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, UK. .,Nanomerics Ltd. 30-34 New Bridge Street, London, EC4V 6BJ, UK.
| |
Collapse
|
71
|
Barranco R, Ventura F, Fracasso T. Immunohistochemical renal expression of aquaporin 2, arginine-vasopressin, vasopressin receptor 2, and renin in saltwater drowning and freshwater drowning. Int J Legal Med 2020; 134:1733-1740. [PMID: 32240384 DOI: 10.1007/s00414-020-02274-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 03/12/2020] [Indexed: 01/03/2023]
Abstract
The diagnosis of drowning is considered one of the most difficult in forensic medicine. Due to the paucity of signs, it is a classical diagnosis by exclusion. For this reason, specific immunohistochemical markers would be useful. Far too little has been done to analyze in-depth the differences between SWD and FWD. We focused on the renal immunohistochemical expression of aquaporin 2, AVP, V2R, and renin in cases of drowning. This study has two purposes: (1) to better understand the differences between saltwater drowning (SWD) and freshwater drowning (FWD), which may indicate different pathophysiology and (2) to eventually identify markers useful for the diagnosis of drowning. We retrospectively investigated 10 cases of SWD gathered from the Institute of Legal Medicine in Genoa (Italy), and 10 cases of FWD from the University Center of Legal Medicine in Geneva (Switzerland). As a control group, we investigated 10 cases of death by gunshot to the head. A strong expression of AQP2 and AVP was significantly (p < 0.05) more evident in cases of SWD than in FWD and control cases. Regarding the V2R, no statistically significant differences were found between the studied groups. The renin tubular expression was particularly intense (p < 0.05) both in SWD and in FWD compared controls. According to our results, AQP2 and AVP represent potential useful markers for the differential diagnosis between SWD and other causes of death, including FWD. Renin may be a useful marker in the diagnosis of drowning but it does not allow for differentiation between FWD and SWD.
Collapse
Affiliation(s)
- Rosario Barranco
- Department of Legal and Forensic Medicine, University of Genova, via De' Toni 12, 16132, Genoa, Italy.
| | - Francesco Ventura
- Department of Legal and Forensic Medicine, University of Genova, via De' Toni 12, 16132, Genoa, Italy
| | - Tony Fracasso
- Centre universitaire romand de Médecine Légale, Rue Michel-Servet 1, 1206, Geneva, Chemin de la Vulliette 4, 1000, Lausanne, Switzerland
- Centre Universitaire Romand de Médecine Légale, Chemin de la Vulliette 4, 1000, Lausanne, Switzerland
| |
Collapse
|
72
|
Sinha S, Dwivedi N, Tao S, Jamadar A, Kakade VR, Neil MO, Weiss RH, Enders J, Calvet JP, Thomas SM, Rao R. Targeting the vasopressin type-2 receptor for renal cell carcinoma therapy. Oncogene 2020; 39:1231-1245. [PMID: 31616061 PMCID: PMC7007354 DOI: 10.1038/s41388-019-1059-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 09/30/2019] [Accepted: 10/03/2019] [Indexed: 02/07/2023]
Abstract
Arginine vasopressin (AVP) and its type-2 receptor (V2R) play an essential role in the regulation of salt and water homeostasis by the kidneys. V2R activation also stimulates proliferation of renal cell carcinoma (RCC) cell lines in vitro. The current studies investigated V2R expression and activity in human RCC tumors, and its role in RCC tumor growth. Examination of the cancer genome atlas (TCGA) database, and analysis of human RCC tumor tissue microarrays, cDNA arrays and tumor biopsy samples demonstrated V2R expression and activity in clear cell RCC (ccRCC). In vitro, V2R antagonists OPC31260 and Tolvaptan, or V2R gene silencing reduced wound closure and cell viability of 786-O and Caki-1 human ccRCC cell lines. Similarly in mouse xenograft models, Tolvaptan and OPC31260 decreased RCC tumor growth by reducing cell proliferation and angiogenesis, while increasing apoptosis. In contrast, the V2R agonist dDAVP significantly increased tumor growth. High intracellular cAMP levels and ERK1/2 activation were observed in human ccRCC tumors. In mouse tumors and Caki-1 cells, V2R agonists reduced cAMP and ERK1/2 activation, while dDAVP treatment had the reverse effect. V2R gene silencing in Caki-1 cells also reduced cAMP and ERK1/2 activation. These results provide novel evidence for a pathogenic role of V2R signaling in ccRCC, and suggest that inhibitors of the AVP-V2R pathway, including the FDA-approved drug Tolvaptan, could be utilized as novel ccRCC therapeutics.
Collapse
Affiliation(s)
- Sonali Sinha
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Nidhi Dwivedi
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Shixin Tao
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Abeda Jamadar
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Vijayakumar R Kakade
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Maura O' Neil
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Robert H Weiss
- Division of Nephrology and Comprehensive Cancer Center, University of California, Davis, CA, USA
- Medical Service, VA Northern California Health Care System, Sacramento, CA, USA
| | - Jonathan Enders
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - James P Calvet
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Sufi M Thomas
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Reena Rao
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA.
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA.
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
73
|
Chauvigné F, Yilmaz O, Ferré A, Fjelldal PG, Finn RN, Cerdà J. The vertebrate Aqp14 water channel is a neuropeptide-regulated polytransporter. Commun Biol 2019; 2:462. [PMID: 31840107 PMCID: PMC6906440 DOI: 10.1038/s42003-019-0713-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 11/26/2019] [Indexed: 12/18/2022] Open
Abstract
Water channels (aquaporins) were originally discovered in mammals with fourteen subfamilies now identified (AQP0-13). Here we show that a functional Aqp14 subfamily phylogenetically related to AQP4-type channels exists in all vertebrate lineages except hagfishes and eutherian mammals. In contrast to the water-selective classical aquaporins, which have four aromatic-arginine constriction residues, Aqp14 proteins present five non-aromatic constriction residues and facilitate the permeation of water, urea, ammonia, H2O2 and glycerol. Immunocytochemical assays suggest that Aqp14 channels play important osmoregulatory roles in piscine seawater adaptation. Our data indicate that Aqp14 intracellular trafficking is tightly regulated by the vasotocinergic/isotocinergic neuropeptide and receptor systems, whereby protein kinase C and A transduction pathways phosphorylate highly conserved C-terminal residues to control channel plasma membrane insertion. The neuropeptide regulation of Aqp14 channels thus predates the vasotocin/vasopressin regulation of AQP2-5-6 orthologs observed in tetrapods. These findings demonstrate that vertebrate Aqp14 channels represent an ancient subfamily of neuropeptide-regulated polytransporters.
Collapse
Affiliation(s)
- François Chauvigné
- IRTA-Institute of Biotechnology and Biomedicine (IBB), Universitat Autònoma de Barcelona, 08193 Bellaterra, (Cerdanyola del Vallès) Spain
| | - Ozlem Yilmaz
- Department of Biological Sciences, Bergen High Technology Centre, University of Bergen, 5020 Bergen, Norway
| | - Alba Ferré
- IRTA-Institute of Biotechnology and Biomedicine (IBB), Universitat Autònoma de Barcelona, 08193 Bellaterra, (Cerdanyola del Vallès) Spain
| | - Per Gunnar Fjelldal
- Institute of Marine Research, Matre Aquaculture Research Station, 5984 Matredal, Norway
| | - Roderick Nigel Finn
- IRTA-Institute of Biotechnology and Biomedicine (IBB), Universitat Autònoma de Barcelona, 08193 Bellaterra, (Cerdanyola del Vallès) Spain
- Department of Biological Sciences, Bergen High Technology Centre, University of Bergen, 5020 Bergen, Norway
| | - Joan Cerdà
- IRTA-Institute of Biotechnology and Biomedicine (IBB), Universitat Autònoma de Barcelona, 08193 Bellaterra, (Cerdanyola del Vallès) Spain
| |
Collapse
|
74
|
Mazaheri M, Assadi F, Sadeghi-Bojd S. Adjunctive acetazolamide therapy for the treatment of Bartter syndrome. Int Urol Nephrol 2019; 52:121-128. [PMID: 31820361 DOI: 10.1007/s11255-019-02351-7] [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: 09/12/2019] [Accepted: 11/24/2019] [Indexed: 12/17/2022]
Abstract
PURPOSE Bartter syndrome is a rare hereditary salt-losing tubulopathy caused by mutations of several genes in the thick ascending limb of Henle's loop, characterized by polyuria, hypokalemic metabolic alkalosis, growth retardation and normal blood pressure. Cyclooxygenase inhibitors, potassium-sparing diuretics and angiotensin-converting enzyme inhibitors are currently used to treat electrolyte derangements, but with poor response. Whether treatment with acetazolamide, a carbonic-anhydrase inhibitor, would result in better clinical outcomes is unknown. METHODS We randomly assigned children with Bartter syndrome in a 1:1 ratio to either receive indomethacin, enalapril, and spironolactone or indomethacin, enalapril, and spironolactone plus acetazolamide once daily in the morning for 4 weeks. After 2 days of washout, participants crossed over to receive the alternative intervention for 4 weeks. The present study examines the serum bicarbonate lowering effect of acetazolamide as an adjunctive therapy in children with Batter syndrome. RESULTS Of the 43 patients screened for eligibility, 22 (51%), between the ages 6 and 42 months, were randomized to intervention. Baseline characteristics were similar between the two groups. Addition of acetazolamide for a period of 4 weeks significantly reduced serum bicarbonate and increased serum potassium levels, parallel with a reduction in serum aldosterone and plasma renin concentration. The 24-h urine volume, sodium, potassium, and chloride decreased significantly. CONCLUSION Our data define a new physiologic and therapeutic role of acetazolamide for the management of children with Bartter syndrome.
Collapse
Affiliation(s)
- Mojgan Mazaheri
- Department of Pediatrics, Section of Nephrology, Semnan University of Medical Science, Semnan, Iran
| | - Farahnak Assadi
- Department of Pediatrics, Division of Nephrology, Rush University Medical Center, 445 East North Water Street, Chicago, IL, USA.
| | - Simin Sadeghi-Bojd
- Department of Pediatrics, Division of Nephrology, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
75
|
Ranieri M, Tamma G, Pellegrino T, Vezzi V, Ambrosio C, Grò C, Di Mise A, Costa T, Valenti G, Cotecchia S. Gain-of-function mutations of the V2 vasopressin receptor in nephrogenic syndrome of inappropriate antidiuresis (NSIAD): a cell-based assay to assess constitutive water reabsorption. Pflugers Arch 2019; 471:1291-1304. [PMID: 31486901 DOI: 10.1007/s00424-019-02307-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 08/28/2019] [Accepted: 08/29/2019] [Indexed: 12/31/2022]
Abstract
Nephrogenic syndrome of inappropriate antidiuresis (NSIAD) is a recently identified chromosome X-linked disease associated with gain-of-function mutations of the V2 vasopressin receptor (V2R), a G-protein-coupled receptor. It is characterized by inability to excrete a free water load, hyponatremia, and undetectable vasopressin-circulating levels. Hyponatremia can be quite severe in affected male children. To gain a deeper insight into the functional properties of the V2R active mutants and how they might translate into the pathological outcome of NSIAD, in this study, we have expressed the wild-type V2R and three constitutively active V2R mutants associated with NSIAD (R137L, R137C, and the F229V) in MCD4 cells, a cell line derived from renal mouse collecting duct, stably expressing the vasopressin-sensitive water channel aquaporin-2 (AQP2). Our findings indicate that in cells expressing each active mutant, AQP2 was constitutively localized to the apical plasma membrane in the absence of vasopressin stimulation. In line with these observations, under basal conditions, osmotic water permeability in cells expressing the constitutively active mutants was significantly higher compared to that of cells expressing the wild-type V2R. Our findings demonstrate a direct link between activating mutations of the V2R and the perturbation of water balance in NSIAD. In addition, this study provides a useful cell-based assay system to assess the functional consequences of newly discovered activating mutations of the V2R on water permeability in kidney cells and to screen the effect of drugs on the mutated receptors.
Collapse
Affiliation(s)
- Marianna Ranieri
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70125, Bari, Italy
| | - Grazia Tamma
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70125, Bari, Italy.,Istituto Nazionale di Biostrutture e Biosistemi, 00136, Rome, Italy
| | - Tommaso Pellegrino
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70125, Bari, Italy
| | - Vanessa Vezzi
- Department of Pharmacology, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Caterina Ambrosio
- Department of Pharmacology, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Cristina Grò
- Department of Pharmacology, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Annarita Di Mise
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70125, Bari, Italy
| | - Tommaso Costa
- Department of Pharmacology, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Giovanna Valenti
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70125, Bari, Italy. .,Istituto Nazionale di Biostrutture e Biosistemi, 00136, Rome, Italy. .,Center of Excellence in Comparative Genomics (CEGBA), University of Bari, 70125, Bari, Italy.
| | - Susanna Cotecchia
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70125, Bari, Italy.
| |
Collapse
|
76
|
Vallon V, Unwin R, Inscho EW, Leipziger J, Kishore BK. Extracellular Nucleotides and P2 Receptors in Renal Function. Physiol Rev 2019; 100:211-269. [PMID: 31437091 DOI: 10.1152/physrev.00038.2018] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The understanding of the nucleotide/P2 receptor system in the regulation of renal hemodynamics and transport function has grown exponentially over the last 20 yr. This review attempts to integrate the available data while also identifying areas of missing information. First, the determinants of nucleotide concentrations in the interstitial and tubular fluids of the kidney are described, including mechanisms of cellular release of nucleotides and their extracellular breakdown. Then the renal cell membrane expression of P2X and P2Y receptors is discussed in the context of their effects on renal vascular and tubular functions. Attention is paid to effects on the cortical vasculature and intraglomerular structures, autoregulation of renal blood flow, tubuloglomerular feedback, and the control of medullary blood flow. The role of the nucleotide/P2 receptor system in the autocrine/paracrine regulation of sodium and fluid transport in the tubular and collecting duct system is outlined together with its role in integrative sodium and fluid homeostasis and blood pressure control. The final section summarizes the rapidly growing evidence indicating a prominent role of the extracellular nucleotide/P2 receptor system in the pathophysiology of the kidney and aims to identify potential therapeutic opportunities, including hypertension, lithium-induced nephropathy, polycystic kidney disease, and kidney inflammation. We are only beginning to unravel the distinct physiological and pathophysiological influences of the extracellular nucleotide/P2 receptor system and the associated therapeutic perspectives.
Collapse
Affiliation(s)
- Volker Vallon
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Robert Unwin
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Edward W Inscho
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Jens Leipziger
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Bellamkonda K Kishore
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| |
Collapse
|
77
|
Wouda RD, Dekker SEI, Reijm J, Olde Engberink RHG, Vogt L. Effects of Water Loading on Observed and Predicted Plasma Sodium, and Fluid and Urine Cation Excretion in Healthy Individuals. Am J Kidney Dis 2019; 74:320-327. [PMID: 31005371 DOI: 10.1053/j.ajkd.2019.02.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 02/23/2019] [Indexed: 11/11/2022]
Abstract
RATIONALE & OBJECTIVE The discovery of sodium storage without concurrent water retention suggests the presence of an additional compartment for sodium distribution in the body. The osmoregulatory role of this compartment under hypotonic conditions is not known. STUDY DESIGN Experimental interventional study. SETTING & PARTICIPANTS Single-center study of 12 apparently healthy men. INTERVENTION To investigate whether sodium can be released from its nonosmotic stores after a hypotonic fluid load, a water-loading test (20mL water/kg in 20 minutes) was performed. OUTCOMES During a 240-minute follow-up, we compared the observed plasma sodium concentration ([Na+]) and fluid and urine cation excretion with values predicted by the Barsoum-Levine and Nguyen-Kurtz formulas. These formulas are used for guidance of fluid therapy during dysnatremia, but do not account for nonosmotic sodium stores. RESULTS 30 minutes after water loading, mean plasma [Na+] decreased 3.2±1.6 (SD) mmol/L, after which plasma [Na+] increased gradually. 120 minutes after water loading, plasma [Na+] was significantly underestimated by the Barsoum-Levine (-1.3±1.4mmol/L; P=0.05) and Nguyen-Kurtz (-1.5±1.5mmol/L; P=0.03) formulas. In addition, the Barsoum-Levine and Nguyen-Kurtz formulas overestimated urine volume, while cation excretion was significantly underestimated, with a cation gap of 57±62 (P=0.009) and 63±63mmol (P=0.005), respectively. After 240 minutes, this gap was 28±59 (P=0.2) and 34±60mmol (P=0.08), respectively. LIMITATIONS The compartment from which the mobilized sodium originated was not identified, and heterogeneity in responses to water loading was observed across participants. CONCLUSIONS These data suggest that healthy individuals are able to mobilize osmotically inactivated sodium after an acute hypotonic fluid load. Further research is needed to expand knowledge about the compartment of osmotically inactivated sodium and its role in osmoregulation and therapy for dysnatremias. FUNDING This investigator-initiated study was partly supported by a grant from Unilever Research and Development Vlaardingen, The Netherlands B.V. (MA-2014-01914).
Collapse
Affiliation(s)
- Rosa D Wouda
- Section of Nephrology, Department of Internal Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Shosha E I Dekker
- Section of Nephrology, Department of Internal Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Joelle Reijm
- Section of Nephrology, Department of Internal Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Rik H G Olde Engberink
- Section of Nephrology, Department of Internal Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Liffert Vogt
- Section of Nephrology, Department of Internal Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
78
|
Effect of nonsteroidal anti-inflammatory drugs in children with Bartter syndrome. Pediatr Nephrol 2019; 34:679-684. [PMID: 30426218 DOI: 10.1007/s00467-018-4135-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 09/14/2018] [Accepted: 10/29/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Bartter syndrome (BS) is a salt-wasting tubulopathy with induced expression of cyclooxygenase-2 in the macula densa, leading to increased prostaglandin production and hyperreninemia. Nonsteroidal anti-inflammatory drugs (NSAIDs) are currently used in BS; however, there is limited information on the impact of NSAIDs at treatment initiation or the potential utility of plasma renin level to guide therapy in patients with BS. METHODS We included 19 patients with BS treated with NSAIDs between 1994 and 2016. We assessed serum levels of renin, aldosterone, electrolytes, calcium, phosphorus, vitamin D, and intact parathyroid hormone (iPTH) before and after treatment initiation. We also recorded modifications in sodium and potassium supplements and changes in urine calcium. RESULTS Median age at diagnosis was 0.9 months [IQR 0-6.9]. Seven patients had BS types 1 or 2, 12 had BS type 3 and two had no mutation identified. There was a trend towards a decrease in sodium chloride supplementation after initiation of NSAIDs. When defining response to treatment based on the normalization of plasma renin level, responders had a greater reduction in their electrolytes supplementation. NSAIDs treatment was associated with a reduction in urine calcium. Before treatment, half of the patients had elevated iPTH, but iPTH normalized following initiation of NSAIDs in all but one patient. CONCLUSIONS This study confirms that NSAIDs reduce urine wasting of sodium and calcium in patients with BS. Monitoring serum renin levels may be useful to identify the lowest effective dose of NSAIDs that optimizes reduction of urine electrolyte losses.
Collapse
|
79
|
Cilz NI, Cymerblit-Sabba A, Young WS. Oxytocin and vasopressin in the rodent hippocampus. GENES BRAIN AND BEHAVIOR 2018; 18:e12535. [PMID: 30378258 DOI: 10.1111/gbb.12535] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 11/02/2018] [Accepted: 11/06/2018] [Indexed: 12/25/2022]
Abstract
The role of the hippocampus in social memory and behavior is under intense investigation. Oxytocin (Oxt) and vasopressin (Avp) are two neuropeptides with many central actions related to social cognition. Oxt- and Avp-expressing fibers are abundant in the hippocampus and receptors for both peptides are seen throughout the different subfields, suggesting that Oxt and Avp modulate hippocampal-dependent processes. In this review, we first focus on the anatomical sources of Oxt and Avp input to the hippocampus and consider the distribution of their corresponding receptors in different hippocampal subfields and neuronal populations. We next discuss the behavioral outcomes related to social memory seen with perturbation of hippocampal Oxt and Avp signaling. Finally, we review Oxt and Avp modulatory mechanisms in the hippocampus that may underlie the behavioral roles for both peptides.
Collapse
Affiliation(s)
- Nicholas I Cilz
- Section on Neural Gene Expression, National Institute of Mental Health, Bethesda, Maryland
| | - Adi Cymerblit-Sabba
- Section on Neural Gene Expression, National Institute of Mental Health, Bethesda, Maryland
| | - W Scott Young
- Section on Neural Gene Expression, National Institute of Mental Health, Bethesda, Maryland
| |
Collapse
|
80
|
Er Shen Wan extract alleviates polyuria and regulates AQP 2 and AVPR 2 in a rat model of spleen-kidney Yang deficiency-induced diarrhea. Biomed Pharmacother 2018; 110:302-311. [PMID: 30522016 DOI: 10.1016/j.biopha.2018.11.147] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 11/27/2018] [Accepted: 11/28/2018] [Indexed: 02/08/2023] Open
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Er Shen Wan (ESW), has been empirically used for treating spleen-kidney Yang deficiency (SKYD) syndrome in Traditional Chinese medicine (TCM) for centuries and shows a variety of activities. The medicinal formula is a mixture of two component herbs, Psoraleae Fructus (PF, Bu-Gu-Zhi in Chinese) and Myristicae Semen (MS, Rou-Dou-Kou in Chinese). The current study was designed to evaluate ESWP antidiuretic treatment of polyuria and to explore potential mechanisms of renal water metabolism in the rat model of SKYD-induced diarrhea. MATERIALS AND METHODS An animal model of 'SKYD-induced diarrhea syndrome' has been established to evaluate the therapeutic effect and action mechanism according to the clinical syndrome and symptoms. The optimal dose (3.5 g/kg) of ESWP was given to rats by gavage for two weeks. Urinary volumes after 24 h were recorded. After the end of the trial, macroscopic morphological and histological examination of the kidney were conducted. Serum levels of Arginine vasopressin (AVP) and aldosterone (ALD) were also measured. Additionally, quantitative real-time RT-PCR (RT-qPCR) and immunohistochemistry (IHC) analyses were performed to clarify the regulation of aquaporin 2 (AQP 2) and arginine vasopressin type 2 receptor (AVPR 2) in the kidney at the gene and tissue expression levels respectively. RESULTS After the administration of ESWP, urinary output volume after 24 h was found to be significantly decreased in rats. Elevated plasma levels of AVP and ALD were detected. Histological kidney damage appeared to be impeded, and histological disease scores were reduced. In addition, the expression levels of AQP 2 and AVPR 2 were significantly increased. CONCLUSION This study suggests that ESWP may elicit significant effects on the treatment of polyuria. Potential mechanisms at least partially involve hormone regulation, and alleviating renal pathological damage. Simultaneously, ESWP may alter renal water absorption by increasing AQP 2 and AVPR 2 expression levels. Thus, the in vivo experimental evidence indicates that ESWP has a therapeutic effect on the SKYD syndrome, which is consistent with its traditional usage.
Collapse
|
81
|
Assan F, Vilaine E, Wagner S, Longvert C, Saiag P, Seidowsky A, Bourgault‐Villada I, Massy ZA. Hyponatremia and MAP‐kinase inhibitors in malignant melanoma: Frequency, pathophysiological aspects and clinical consequences. Pigment Cell Melanoma Res 2018; 32:326-331. [PMID: 30387922 DOI: 10.1111/pcmr.12749] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 09/27/2018] [Accepted: 10/24/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Florence Assan
- Division of Nephrology, Ambroise Paré Hospital APHP Boulogne Billancourt/Paris France
| | - Eve Vilaine
- Division of Nephrology, Ambroise Paré Hospital, APHP Paris‐Ile‐de‐France‐West University (UVSQ) Boulogne Billancourt/Paris France
- INSERM U1018 Team5 Villejuif France
| | | | - Christine Longvert
- Division of Dermatology, Ambroise Paré Hospital, APHP Paris‐Ile‐de‐France‐West University (UVSQ) Boulogne Billancourt/Paris France
| | - Philippe Saiag
- Division of Dermatology, Ambroise Paré Hospital, APHP Paris‐Ile‐de‐France‐West University (UVSQ) Boulogne Billancourt/Paris France
| | - Alexandre Seidowsky
- Division of Nephrology, Ambroise Paré Hospital, APHP Paris‐Ile‐de‐France‐West University (UVSQ) Boulogne Billancourt/Paris France
- INSERM U1018 Team5 Villejuif France
| | - Isabelle Bourgault‐Villada
- Division of Clinical Dermatology‐immunology, Ambroise Paré Hospital, APHP Paris‐Ile‐de‐France‐West University (UVSQ) Boulogne Billancourt/Paris France
| | - Ziad A. Massy
- Division of Nephrology, Ambroise Paré Hospital, APHP Paris‐Ile‐de‐France‐West University (UVSQ) Boulogne Billancourt/Paris France
- INSERM U1018 Team5 Villejuif France
| |
Collapse
|
82
|
Li Y, Zou Q, Zhang J. Vincamine exerts protective effect on spiral ganglion neurons in endolymphatic hydrops guinea pig models. Am J Transl Res 2018; 10:3650-3663. [PMID: 30662616 PMCID: PMC6291722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 10/16/2018] [Indexed: 06/09/2023]
Abstract
The aim of this study was to investigate the protective effects of vincamine in endolymphatic hydrops (ELH). After ELH guinea pigs treated by vincamine, the concentration of VAP in plasma, and the levels of cAMP, MDA, SOD, GSH-Px in right cochlea were measured using spectrophotometric method. The V2R, NMDAR1, p-NMDAR1, AQP2, p-AQP2, caspase3/9 and c-caspase3/9 expressions in right cochlea were detected using western blot analysis. The cochlear hydrops degree and SGNs density were evaluated by hemotoxylin and eosin staining (HE) test. Normal hearing and vestibular function were warranted by the tests of auditory brainstem response (ABR) and electronystagmography (ENG). After glutamate-injured SGNs treated with vincamine, the MDA, SOD GSH-Px, NGF, BDNF, NT3, NT4 and Trks levels were measured. Meanwhile, the Bcl2, Bax, NMDAR1, p-NMDAR1, PI3K, p-PI3K, Akt, p-Akt, caspase3/9 and cleaved-caspase3/9 expression levels were detected. Furthermore, the viability, apoptosis and necrosis of SGNs were tested by MTT and Hoechst/PI staining methods. The results indicated that vincamine could significantly inhibit the expression levels of cAMP, MDA, V2R, p-NMDAR1, p-AQP2 and c-caspase-3/-9 in cochlea, alleviate the cochlear hydrops degree, regulate the audiological and vestibular dysfunctions. The SGNs density, SOD and GSH-Px levels were also increased by vincamine. In vincamine-treating groups, the MDA, Bax, p-NMDAR1, and c-caspase3/9 levels were observably decreased, while SGNs survival, SOD, GSH, NGF, BDNF, NT3, NT4, Trks, Bcl2, p-PI3K, p-Akt expressions were improved. The present study indicated a novel use of vincamine in suppressing ELH formation by down-regulating the VAP/AQP2 signaling pathway. It also manifested that vincamine exerted protective effects on hearing via improving neurotrophin-dependent PI3K/Akt signaling pathway in SGNs.
Collapse
Affiliation(s)
- Yi Li
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren HospitalBeijing 100730, China
| | - Qijuan Zou
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren HospitalBeijing 100730, China
| | - Jie Zhang
- Shanghai University of TCMShanghai 201203, China
| |
Collapse
|
83
|
Nawata CM, Pannabecker TL. Mammalian urine concentration: a review of renal medullary architecture and membrane transporters. J Comp Physiol B 2018; 188:899-918. [PMID: 29797052 PMCID: PMC6186196 DOI: 10.1007/s00360-018-1164-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 04/23/2018] [Accepted: 05/14/2018] [Indexed: 01/10/2023]
Abstract
Mammalian kidneys play an essential role in balancing internal water and salt concentrations. When water needs to be conserved, the renal medulla produces concentrated urine. Central to this process of urine concentration is an osmotic gradient that increases from the corticomedullary boundary to the inner medullary tip. How this gradient is generated and maintained has been the subject of study since the 1940s. While it is generally accepted that the outer medulla contributes to the gradient by means of an active process involving countercurrent multiplication, the source of the gradient in the inner medulla is unclear. The last two decades have witnessed advances in our understanding of the urine-concentrating mechanism. Details of medullary architecture and permeability properties of the tubules and vessels suggest that the functional and anatomic relationships of these structures may contribute to the osmotic gradient necessary to concentrate urine. Additionally, we are learning more about the membrane transporters involved and their regulatory mechanisms. The role of medullary architecture and membrane transporters in the mammalian urine-concentrating mechanism are the focus of this review.
Collapse
Affiliation(s)
- C Michele Nawata
- Department of Physiology, Banner University Medical Center, University of Arizona, 1501 N. Campbell Avenue, Tucson, AZ, 85724-5051, USA.
| | - Thomas L Pannabecker
- Department of Physiology, Banner University Medical Center, University of Arizona, 1501 N. Campbell Avenue, Tucson, AZ, 85724-5051, USA
| |
Collapse
|
84
|
Elkhenany H, AlOkda A, El-Badawy A, El-Badri N. Tissue regeneration: Impact of sleep on stem cell regenerative capacity. Life Sci 2018; 214:51-61. [PMID: 30393021 DOI: 10.1016/j.lfs.2018.10.057] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 10/23/2018] [Accepted: 10/25/2018] [Indexed: 12/31/2022]
Abstract
The circadian rhythm orchestrates many cellular functions, such as cell division, cell migration, metabolism and numerous intracellular biological processes. The physiological changes during sleep are believed to promote a suitable microenvironment for stem cells to proliferate, migrate and differentiate. These effects are mediated either directly by circadian clock genes or indirectly via hormones and cytokines. Hormones, such as melatonin and cortisol, are secreted in response to neural optic signals and act in harmony to regulate many biological functions during sleep. Herein, we correlate the effects of the main circadian genes on the expression of certain stem cell genes responsible for the regeneration of different tissues, including bone, cartilage, skin, and intestine. We also review the effects of different hormones and cytokines on stem cell activation or suppression and their relationship to the day/night cycle. The correlation of circadian rhythm with tissue regeneration could have implications in understanding the biology of sleep and tissue regeneration and in enhancing the efficacy and timing of surgical procedures.
Collapse
Affiliation(s)
- Hoda Elkhenany
- Centre of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, 12588, Egypt; Department of Surgery, Faculty of Veterinary Medicine, Alexandria University, 22785, Egypt
| | - Abdelrahman AlOkda
- Centre of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, 12588, Egypt
| | - Ahmed El-Badawy
- Centre of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, 12588, Egypt
| | - Nagwa El-Badri
- Centre of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, 12588, Egypt.
| |
Collapse
|
85
|
Goel A, Farhat F, Zik C, Jeffery M. Triphasic response of pituitary stalk injury following TBI: a relevant yet uncommonly recognised endocrine phenomenon. BMJ Case Rep 2018; 2018:bcr-2018-226725. [PMID: 30361454 DOI: 10.1136/bcr-2018-226725] [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] [Indexed: 11/04/2022] Open
Abstract
The triphasic response of pituitary stalk injury has previously been described in a minority of patients following intracranial surgery, however, this phenomenon can also occur after traumatic brain injury. We present the case of a 20-year-old male who experienced the triphasic response of pituitary stalk injury (central diabetes insipidus, syndrome of inappropriate antidiuretic hormone and central diabetes insipidus again) after striking his head on a concrete curb. His history and presentation highlight the importance of recognising the distinctive symptoms of each individual stage of pituitary stalk injury, and using the appropriate diagnostic tools and therapies to guide further management.
Collapse
Affiliation(s)
- Ansha Goel
- Department of Medicine, Inova Health System, Falls Church, Virginia, USA
| | - Freba Farhat
- Department of Medicine, Inova Health System, Falls Church, Virginia, USA
| | - Chad Zik
- Department of Medicine, Inova Health System, Falls Church, Virginia, USA
| | - Michelle Jeffery
- Department of Medicine, Inova Health System, Falls Church, Virginia, USA
| |
Collapse
|
86
|
Bijkerk R, Trimpert C, van Solingen C, de Bruin RG, Florijn BW, Kooijman S, van den Berg R, van der Veer EP, Bredewold EOW, Rensen PCN, Rabelink TJ, Humphreys BD, Deen PMT, van Zonneveld AJ. MicroRNA-132 controls water homeostasis through regulating MECP2-mediated vasopressin synthesis. Am J Physiol Renal Physiol 2018; 315:F1129-F1138. [DOI: 10.1152/ajprenal.00087.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Fine-tuning of the body’s water balance is regulated by vasopressin (AVP), which induces the expression and apical membrane insertion of aquaporin-2 water channels and subsequent water reabsorption in the kidney. Here we demonstrate that silencing of microRNA-132 (miR-132) in mice causes severe weight loss due to acute diuresis coinciding with increased plasma osmolality, reduced renal total and plasma membrane expression of aquaporin-2, and abrogated increase in AVP levels. Infusion with synthetic AVP fully reversed the antagomir-132-induced diuresis, and low-dose intracerebroventricular administration of antagomir-132 similarly caused acute diuresis. Central and intracerebroventricular antagomir-132 injection both decreased hypothalamic AVP mRNA levels. At the molecular level, antagomir-132 increased the in vivo and in vitro mRNA expression of methyl-CpG-binding protein-2 (MECP2), which is a miR-132 target and which blocks AVP gene expression by binding its enhancer region. In line with this, treatment of hypothalamic N6 cells with a high-salt solution increased its miR-132 levels, whereas it attenuated endogenous Mecp2 mRNA levels. In conclusion, we identified miR-132 as a first miRNA regulating the osmotic balance by regulating the hypothalamic AVP gene mRNA expression.
Collapse
Affiliation(s)
- Roel Bijkerk
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Renal Division, Department of Medicine, Brigham & Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Christiane Trimpert
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Coen van Solingen
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine, New York University Medical Center, New York, New York
| | - Ruben G. de Bruin
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Barend W. Florijn
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Sander Kooijman
- Department of Internal Medicine (Endocrinology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Rosa van den Berg
- Department of Internal Medicine (Endocrinology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Eric P. van der Veer
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Edwin O. W. Bredewold
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Patrick C. N. Rensen
- Department of Internal Medicine (Endocrinology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Ton J. Rabelink
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Benjamin D. Humphreys
- Renal Division, Department of Medicine, Brigham & Women’s Hospital and Harvard Medical School, Boston, Massachusetts
- Renal Division, Washington University School of Medicine, St. Louis, Missouri
| | - Peter M. T. Deen
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anton Jan van Zonneveld
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
87
|
Mitochondria, Oxytocin, and Vasopressin: Unfolding the Inflammatory Protein Response. Neurotox Res 2018; 36:239-256. [PMID: 30259418 DOI: 10.1007/s12640-018-9962-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 09/11/2018] [Accepted: 09/17/2018] [Indexed: 01/07/2023]
Abstract
Neuroendocrine and immune signaling pathways are activated following insults such as stress, injury, and infection, in a systemic response aimed at restoring homeostasis. Mitochondrial metabolism and function have been implicated in the control of immune responses. Commonly studied along with mitochondrial function, reactive oxygen species (ROS) are closely linked to cellular inflammatory responses. It is also accepted that cells experiencing mitochondrial or endoplasmic reticulum (ER) stress induce response pathways in order to cope with protein-folding dysregulation, in homeostatic responses referred to as the unfolded protein responses (UPRs). Recent reports indicate that the UPRs may play an important role in immune responses. Notably, the homeostasis-regulating hormones oxytocin (OXT) and vasopressin (AVP) are also associated with the regulation of inflammatory responses and immune function. Intriguingly, OXT and AVP have been linked with ER unfolded protein responses (UPRER), and can impact ROS production and mitochondrial function. Here, we will review the evidence for interactions between these various factors and how these neuropeptides might influence mitochondrial processes.
Collapse
|
88
|
Perschbacher KJ, Deng G, Fisher RA, Gibson-Corley KN, Santillan MK, Grobe JL. Regulators of G protein signaling in cardiovascular function during pregnancy. Physiol Genomics 2018; 50:590-604. [PMID: 29702036 PMCID: PMC6139632 DOI: 10.1152/physiolgenomics.00037.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
G protein-coupled receptor signaling mechanisms are implicated in many aspects of cardiovascular control, and dysfunction of such signaling mechanisms is commonly associated with disease states. Investigators have identified a large number of regulator of G protein signaling (RGS) proteins that variously contribute to the modulation of intracellular second-messenger signaling kinetics. These many RGS proteins each interact with a specific set of second-messenger cascades and receptor types and exhibit tissue-specific expression patterns. Increasing evidence supports the contribution of RGS proteins, or their loss, in the pathogenesis of cardiovascular dysfunctions. This review summarizes the current understanding of the functional contributions of RGS proteins, particularly within the B/R4 family, in cardiovascular disorders of pregnancy including gestational hypertension, uterine artery dysfunction, and preeclampsia.
Collapse
Affiliation(s)
| | - Guorui Deng
- Department of Pharmacology, University of Iowa , Iowa City, Iowa
| | - Rory A Fisher
- Department of Pharmacology, University of Iowa , Iowa City, Iowa
| | - Katherine N Gibson-Corley
- Department of Pathology, University of Iowa , Iowa City, Iowa
- UIHC Center for Hypertension Research, University of Iowa , Iowa City, Iowa
| | - Mark K Santillan
- Department of Obstetrics & Gynecology, University of Iowa , Iowa City, Iowa
- UIHC Center for Hypertension Research, University of Iowa , Iowa City, Iowa
- Abboud Cardiovascular Research Center, University of Iowa , Iowa City, Iowa
| | - Justin L Grobe
- Department of Pharmacology, University of Iowa , Iowa City, Iowa
- UIHC Center for Hypertension Research, University of Iowa , Iowa City, Iowa
- Abboud Cardiovascular Research Center, University of Iowa , Iowa City, Iowa
- Fraternal Order of Eagles' Diabetes Research Center, University of Iowa , Iowa City, Iowa
- Obesity Education & Research Initiative, University of Iowa , Iowa City, Iowa
- Iowa Neuroscience Institute, University of Iowa , Iowa City, Iowa
| |
Collapse
|
89
|
Giorello FM, Feijoo M, D'Elía G, Naya DE, Valdez L, Opazo JC, Lessa EP. An association between differential expression and genetic divergence in the Patagonian olive mouse (Abrothrix olivacea). Mol Ecol 2018; 27:3274-3286. [PMID: 29940092 DOI: 10.1111/mec.14778] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 05/30/2018] [Accepted: 05/31/2018] [Indexed: 12/19/2022]
Abstract
Recent molecular studies have found striking differences between desert-adapted species and model mammals regarding water conservation. In particular, aquaporin 4, a classical gene involved in water regulation of model species, is absent or not expressed in the kidneys of desert-adapted species. To further understand the molecular response to water availability, we studied the Patagonian olive mouse Abrothrix olivacea, a species with an unusually broad ecological tolerance that exhibits a great urine concentration capability. The species is able to occupy both the arid Patagonian steppe and the Valdivian and Magellanic forests. We sampled 95 olive mouse specimens from four localities (two in the steppe and two in the forests) and analysed both phenotypic variables and transcriptomic data to investigate the response of this species to the contrasting environmental conditions. The relative size of the kidney and the ratio of urine to plasma concentrations were, as expected, negatively correlated with annual rainfall. Expression analyses uncovered nearly 3,000 genes that were differentially expressed between steppe and forest samples and indicated that this species resorts to the "classical" gene pathways for water regulation. Differential expression across biomes also involves genes that involved in immune and detoxification functions. Overall, genes that were differentially expressed showed a slight tendency to be more divergent and to display an excess of intermediate allele frequencies, relative to the remaining loci. Our results indicate that both differential expression in pathways involved in water conservation and geographical allelic variation are important in the occupation of contrasting habitats by the Patagonian olive mouse.
Collapse
Affiliation(s)
- Facundo M Giorello
- Departamento de Ecología y Evolución, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
- Espacio de Biología Vegetal del Noreste, Centro Universitario de Tacuarembó, Universidad de la República, Tacuarembó, Uruguay
| | - Matias Feijoo
- Departamento de Ecología y Evolución, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Guillermo D'Elía
- Instituto de Ciencias Ambientales y Evolutivas, Universidad Austral de Chile, Valdivia, Chile
| | - Daniel E Naya
- Departamento de Ecología y Evolución, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Lourdes Valdez
- Instituto de Ciencias Ambientales y Evolutivas, Universidad Austral de Chile, Valdivia, Chile
| | - Juan C Opazo
- Instituto de Ciencias Ambientales y Evolutivas, Universidad Austral de Chile, Valdivia, Chile
| | - Enrique P Lessa
- Departamento de Ecología y Evolución, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
90
|
Wang R, Chow BKC, Zhang L. Distribution and Functional Implication of Secretin in Multiple Brain Regions. J Mol Neurosci 2018; 68:485-493. [PMID: 29882022 DOI: 10.1007/s12031-018-1089-z] [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: 03/31/2018] [Accepted: 05/11/2018] [Indexed: 12/15/2022]
Abstract
Secretin is a polypeptide hormone initially identified for its gastrointestinal functions. However, emerging evidences show wide distribution of secretin and secretin receptor across various brain regions from cerebral cortex, hippocampus, hypothalamus to cerebellum. In this mini review, we will firstly describe the region-specific expression pattern of secretin and secretin receptor in the brain, followed by a summary of central physiological and neurological functions mediated by secretin. Using genetic manipulation and pharmaceutical approaches, one can elucidate the role of secretin in mediating various neurological functions from simple behaviors, such as water and food intake, to more complex functions including emotion, motor, and learning or memory. At last, current weakness and future perspectives of secretin in the central nervous system will be discussed, aiming to provide the potency of using secretin or its analog for treating various neurological disorders.
Collapse
Affiliation(s)
- Ruanna Wang
- Joint International Research Laboratory of CNS Regeneration, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Billy K C Chow
- School of Biological Sciences, University of Hong Kong, Hong Kong SAR, China.
| | - Li Zhang
- Joint International Research Laboratory of CNS Regeneration, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, China.
| |
Collapse
|
91
|
Palmer BF, Clegg DJ. Altered Prostaglandin Signaling as a Cause of Thiazide-Induced Hyponatremia. Am J Kidney Dis 2018; 71:769-771. [PMID: 29501264 DOI: 10.1053/j.ajkd.2017.11.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 11/19/2017] [Indexed: 11/11/2022]
Affiliation(s)
- Biff F Palmer
- University of Texas Southwestern Medical Center, Dallas, TX.
| | | |
Collapse
|
92
|
Bongers CCWG, Alsady M, Nijenhuis T, Tulp ADM, Eijsvogels TMH, Deen PMT, Hopman MTE. Impact of acute versus prolonged exercise and dehydration on kidney function and injury. Physiol Rep 2018; 6:e13734. [PMID: 29890037 PMCID: PMC5995308 DOI: 10.14814/phy2.13734] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 05/18/2018] [Accepted: 05/18/2018] [Indexed: 01/17/2023] Open
Abstract
Exercise and dehydration may be associated with a compromised kidney function and potential signs of kidney injury. However, the kidney responses to exercise of different durations and hypohydration levels are not yet known. Therefore, we aimed to compare the effects of acute versus prolonged exercise and dehydration on estimated glomerular filtration rate (eGFR) and kidney injury biomarkers in healthy male adults. A total of 35 subjects (23 ± 3 years) were included and invited for two study visits. Visit 1 consisted of a maximal cycling test. On Visit 2, subjects performed a submaximal exercise test at 80% of maximal heart rate until 3% hypohydration. Blood and urine samples were taken at baseline, after 30 min of exercise (acute effects; low level of hypohydration) and after 150 min of exercise or when 3% hypohydration was achieved (prolonged effects, high level of hypohydration). Urinary outcome parameters were corrected for urinary cystatin C, creatinine, and osmolality. Subjects dehydrated on average 0.6 ± 0.3% and 2.9 ± 0.7% after acute and prolonged exercise, respectively (P < 0.001). The eGFRcystatin C did not differ between baseline and acute exercise (118 ± 11 vs. 116 ± 12 mL/min/1.73 m2 , P = 0.12), whereas eGFRcystatin C was significantly lower after prolonged exercise (103 ± 16 mL/min/1.73 m2 , P < 0.001). We found no difference in osmolality corrected uKIM1 concentrations after acute and prolonged exercise (P > 0.05), and elevated osmolality corrected uNGAL concentrations after acute and prolonged exercise (all P-values < 0.05). In conclusion, acute exercise did barely impact on eGFRcystatin C and kidney injury biomarkers, whereas prolonged exercise is associated with a decline in eGFRcystatin C and increased biomarkers for kidney injury.
Collapse
Affiliation(s)
- Coen C. W. G. Bongers
- Department of PhysiologyRadboud Institute for Health SciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Mohammad Alsady
- Department of PhysiologyRadboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Tom Nijenhuis
- Department of NephrologyRadboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Anouk D. M. Tulp
- Department of PhysiologyRadboud Institute for Health SciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Thijs M. H. Eijsvogels
- Department of PhysiologyRadboud Institute for Health SciencesRadboud University Medical CenterNijmegenThe Netherlands
- Research Institute for Sports and Exercise SciencesLiverpool John Moores UniversityLiverpoolUnited Kingdom
| | - Peter M. T. Deen
- Department of PhysiologyRadboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Maria T. E. Hopman
- Department of PhysiologyRadboud Institute for Health SciencesRadboud University Medical CenterNijmegenThe Netherlands
| |
Collapse
|
93
|
Huang J, Montani JP, Verrey F, Feraille E, Ming XF, Yang Z. Arginase-II negatively regulates renal aquaporin-2 and water reabsorption. FASEB J 2018; 32:5520-5531. [PMID: 29718707 PMCID: PMC6405175 DOI: 10.1096/fj.201701209r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Type-II l-arginine:ureahydrolase, arginase-II (Arg-II), is abundantly
expressed in the kidney. The physiologic role played by Arg-II in the kidney remains
unknown. Herein, we report that in mice that are deficient in Arg-II
(Arg-II−/−), total and membrane-associated aquaporin-2
(AQP2) protein levels were significantly higher compared with wild-type (WT)
controls. Water deprivation enhanced Arg-II expression, AQP2 levels, and membrane
association in collecting ducts. Effects of water deprivation on AQP2 were stronger
in Arg-II−/− mice than in WT mice. Accordingly, a decrease
in urine volume and an increase in urine osmolality under water deprivation were more
pronounced in Arg-II−/− mice than in WT mice, which
correlated with a weaker increase in plasma osmolality in
Arg-II−/− mice. There was no difference in vasopressin
release under water deprivation conditions between either genotype of mice. Although
total AQP2 and phosphorylated AQP2-S256 levels (mediated by PKA) in kidneys under
water deprivation conditions were significantly higher in
Arg-II−/− mice compared with WT animals, there is no
difference in the ratio of AQP2-S256:AQP2. In cultured mouse collecting duct
principal mCCDcl1 cells, expression of both Arg-II and AQP2 were enhanced
by the vasopressin type 2 receptor agonist, desamino-d-arginine
vasopressin (dDAVP). Silencing Arg-II enhanced the expression and membrane
association of AQP2 by dDAVP without influencing cAMP levels. In conclusion,
in vivo and in vitro experiments demonstrate
that Arg-II negatively regulates AQP2 and the urine-concentrating capability in
kidneys via a mechanism that is not associated with the modulation
of the cAMP pathway.—Huang, J., Montani, J.-P., Verrey, F., Feraille, E.,
Ming, X.-F., Yang, Z. Arginase-II negatively regulates renal aquaporin-2 and water
reabsorption.
Collapse
Affiliation(s)
- Ji Huang
- Division of Physiology, Department of Medicine, Cardiovascular and Aging Research, University of Fribourg, Fribourg, Switzerland.,Kidney Control of Homeostasis, National Center of Competence in Research, Zurich, Switzerland
| | - Jean-Pierre Montani
- Division of Physiology, Department of Medicine, Cardiovascular and Aging Research, University of Fribourg, Fribourg, Switzerland.,Kidney Control of Homeostasis, National Center of Competence in Research, Zurich, Switzerland
| | - François Verrey
- Kidney Control of Homeostasis, National Center of Competence in Research, Zurich, Switzerland.,Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Eric Feraille
- Kidney Control of Homeostasis, National Center of Competence in Research, Zurich, Switzerland.,Department of Cell Biology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Xiu-Fen Ming
- Division of Physiology, Department of Medicine, Cardiovascular and Aging Research, University of Fribourg, Fribourg, Switzerland.,Kidney Control of Homeostasis, National Center of Competence in Research, Zurich, Switzerland
| | - Zhihong Yang
- Division of Physiology, Department of Medicine, Cardiovascular and Aging Research, University of Fribourg, Fribourg, Switzerland.,Kidney Control of Homeostasis, National Center of Competence in Research, Zurich, Switzerland
| |
Collapse
|
94
|
Sung YJ, Winkler TW, de las Fuentes L, Bentley AR, Brown MR, Kraja AT, Schwander K, Ntalla I, Guo X, Franceschini N, Lu Y, Cheng CY, Sim X, Vojinovic D, Marten J, Musani SK, Li C, Feitosa MF, Kilpeläinen TO, Richard MA, Noordam R, Aslibekyan S, Aschard H, Bartz TM, Dorajoo R, Liu Y, Manning AK, Rankinen T, Smith AV, Tajuddin SM, Tayo BO, Warren HR, Zhao W, Zhou Y, Matoba N, Sofer T, Alver M, Amini M, Boissel M, Chai JF, Chen X, Divers J, Gandin I, Gao C, Giulianini F, Goel A, Harris SE, Hartwig FP, Horimoto ARVR, Hsu FC, Jackson AU, Kähönen M, Kasturiratne A, Kühnel B, Leander K, Lee WJ, Lin KH, 'an Luan J, McKenzie CA, Meian H, Nelson CP, Rauramaa R, Schupf N, Scott RA, Sheu WHH, Stančáková A, Takeuchi F, van der Most PJ, Varga TV, Wang H, Wang Y, Ware EB, Weiss S, Wen W, Yanek LR, Zhang W, Zhao JH, Afaq S, Alfred T, Amin N, Arking D, Aung T, Barr RG, Bielak LF, Boerwinkle E, Bottinger EP, Braund PS, Brody JA, Broeckel U, Cabrera CP, Cade B, Caizheng Y, Campbell A, Canouil M, Chakravarti A, Chauhan G, Christensen K, Cocca M, Collins FS, Connell JM, de Mutsert R, de Silva HJ, Debette S, Dörr M, Duan Q, Eaton CB, Ehret G, Evangelou E, Faul JD, Fisher VA, Forouhi NG, Franco OH, Friedlander Y, Gao H, Gigante B, Graff M, Gu CC, Gu D, Gupta P, Hagenaars SP, Harris TB, He J, Heikkinen S, Heng CK, Hirata M, Hofman A, Howard BV, Hunt S, Irvin MR, Jia Y, Joehanes R, Justice AE, Katsuya T, Kaufman J, Kerrison ND, Khor CC, Koh WP, Koistinen HA, Komulainen P, Kooperberg C, Krieger JE, Kubo M, Kuusisto J, Langefeld CD, Langenberg C, Launer LJ, Lehne B, Lewis CE, Li Y, Lim SH, Lin S, Liu CT, Liu J, Liu J, Liu K, Liu Y, Loh M, Lohman KK, Long J, Louie T, Mägi R, Mahajan A, Meitinger T, Metspalu A, Milani L, Momozawa Y, Morris AP, Mosley TH, Munson P, Murray AD, Nalls MA, Nasri U, Norris JM, North K, Ogunniyi A, Padmanabhan S, Palmas WR, Palmer ND, Pankow JS, Pedersen NL, Peters A, Peyser PA, Polasek O, Raitakari OT, Renström F, Rice TK, Ridker PM, Robino A, Robinson JG, Rose LM, Rudan I, Sabanayagam C, Salako BL, Sandow K, Schmidt CO, Schreiner PJ, Scott WR, Seshadri S, Sever P, Sitlani CM, Smith JA, Snieder H, Starr JM, Strauch K, Tang H, Taylor KD, Teo YY, Tham YC, Uitterlinden AG, Waldenberger M, Wang L, Wang YX, Wei WB, Williams C, Wilson G, Wojczynski MK, Yao J, Yuan JM, Zonderman AB, Becker DM, Boehnke M, Bowden DW, Chambers JC, Chen YDI, de Faire U, Deary IJ, Esko T, Farrall M, Forrester T, Franks PW, Freedman BI, Froguel P, Gasparini P, Gieger C, Horta BL, Hung YJ, Jonas JB, Kato N, Kooner JS, Laakso M, Lehtimäki T, Liang KW, Magnusson PKE, Newman AB, Oldehinkel AJ, Pereira AC, Redline S, Rettig R, Samani NJ, Scott J, Shu XO, van der Harst P, Wagenknecht LE, Wareham NJ, Watkins H, Weir DR, Wickremasinghe AR, Wu T, Zheng W, Kamatani Y, Laurie CC, Bouchard C, Cooper RS, Evans MK, Gudnason V, Kardia SLR, Kritchevsky SB, Levy D, O'Connell JR, Psaty BM, van Dam RM, Sims M, Arnett DK, Mook-Kanamori DO, Kelly TN, Fox ER, Hayward C, Fornage M, Rotimi CN, Province MA, van Duijn CM, Tai ES, Wong TY, Loos RJF, Reiner AP, Rotter JI, Zhu X, Bierut LJ, Gauderman WJ, Caulfield MJ, Elliott P, Rice K, Munroe PB, Morrison AC, Cupples LA, Rao DC, Chasman DI. A Large-Scale Multi-ancestry Genome-wide Study Accounting for Smoking Behavior Identifies Multiple Significant Loci for Blood Pressure. Am J Hum Genet 2018; 102:375-400. [PMID: 29455858 PMCID: PMC5985266 DOI: 10.1016/j.ajhg.2018.01.015] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 01/18/2018] [Indexed: 12/18/2022] Open
Abstract
Genome-wide association analysis advanced understanding of blood pressure (BP), a major risk factor for vascular conditions such as coronary heart disease and stroke. Accounting for smoking behavior may help identify BP loci and extend our knowledge of its genetic architecture. We performed genome-wide association meta-analyses of systolic and diastolic BP incorporating gene-smoking interactions in 610,091 individuals. Stage 1 analysis examined ∼18.8 million SNPs and small insertion/deletion variants in 129,913 individuals from four ancestries (European, African, Asian, and Hispanic) with follow-up analysis of promising variants in 480,178 additional individuals from five ancestries. We identified 15 loci that were genome-wide significant (p < 5 × 10-8) in stage 1 and formally replicated in stage 2. A combined stage 1 and 2 meta-analysis identified 66 additional genome-wide significant loci (13, 35, and 18 loci in European, African, and trans-ancestry, respectively). A total of 56 known BP loci were also identified by our results (p < 5 × 10-8). Of the newly identified loci, ten showed significant interaction with smoking status, but none of them were replicated in stage 2. Several loci were identified in African ancestry, highlighting the importance of genetic studies in diverse populations. The identified loci show strong evidence for regulatory features and support shared pathophysiology with cardiometabolic and addiction traits. They also highlight a role in BP regulation for biological candidates such as modulators of vascular structure and function (CDKN1B, BCAR1-CFDP1, PXDN, EEA1), ciliopathies (SDCCAG8, RPGRIP1L), telomere maintenance (TNKS, PINX1, AKTIP), and central dopaminergic signaling (MSRA, EBF2).
Collapse
Affiliation(s)
- Yun J Sung
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Thomas W Winkler
- Department of Genetic Epidemiology, University of Regensburg, Regensburg 93051, Germany
| | - Lisa de las Fuentes
- Cardiovascular Division, Department of Medicine, Washington University, St. Louis, MO 63110, USA
| | - Amy R Bentley
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Michael R Brown
- Department of Epidemiology, Human Genetics, and Environmental Sciences, The University of Texas School of Public Health, Houston, TX 77030, USA
| | - Aldi T Kraja
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Karen Schwander
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ioanna Ntalla
- William Harvey Research Institute, Clinical Pharmacology, Queen Mary University of London, London EC1M 6BQ, UK
| | - Xiuqing Guo
- Genomic Outcomes, Department of Pediatrics, LABioMed at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Nora Franceschini
- Department of Epidemiology, University of North Carolina Gillings School of Global Public Health, Chapel Hill, NC 27514, USA
| | - Yingchang Lu
- Icahn School of Medicine at Mount Sinai, The Charles Bronfman Institute for Personalized Medicine, New York, NY 10029, USA
| | - Ching-Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore 169856, Singapore; Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Singapore 169857, Singapore; Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore 117597, Singapore
| | - Xueling Sim
- Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore, Singapore 117549, Singapore
| | - Dina Vojinovic
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jonathan Marten
- Medical Research Council Human Genetics Unit, Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Solomon K Musani
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39213, USA
| | - Changwei Li
- Department of Epidemiology and Biostatistics, University of Giorgia at Athens College of Public Health, Athens, GA 30602, USA
| | - Mary F Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Tuomas O Kilpeläinen
- Section of Metabolic Genetics, Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark; Department of Environmental Medicine and Public Health, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Melissa A Richard
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden 2300RC, the Netherlands
| | - Stella Aslibekyan
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Hugues Aschard
- Department of Epidemiology, Harvard School of Public Health, Boston, MA 02115, USA; Centre de Bioinformatique Biostatistique et Biologie Integrative (C3BI), Institut Pasteur, Paris 75015, France
| | - Traci M Bartz
- Cardiovascular Health Research Unit, Biostatistics and Medicine, University of Washington, Seattle, WA 98101, USA
| | - Rajkumar Dorajoo
- Genome Institute of Singapore, Agency for Science Technology and Research, Singapore 138672, Singapore
| | - Yongmei Liu
- Division of Biostatistical Sciences, Department of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Alisa K Manning
- Center for Human Genetics Research, Massachusetts General Hospital, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Tuomo Rankinen
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Albert Vernon Smith
- Icelandic Heart Association, Kopavogur 201, Iceland; Faculty of Medicine, University of Iceland, Reykjavik 101, Iceland
| | - Salman M Tajuddin
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Bamidele O Tayo
- Department of Public Health Sciences, Loyola University Chicago, Maywood, IL 60153, USA
| | - Helen R Warren
- William Harvey Research Institute, Clinical Pharmacology, Queen Mary University of London, London EC1M 6BQ, UK; NIHR Cardiovascular Biomedical Research Unit, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Wei Zhao
- School of Public Health, Department of Epidemiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yanhua Zhou
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Nana Matoba
- Laboratory for Statistical Analysis, Center for Integrative Medical Sciences, RIKEN, Yokohama 230-0045, Japan
| | - Tamar Sofer
- Department of Biostatistics, University of Washington, Seattle, WA 98105, USA
| | - Maris Alver
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Marzyeh Amini
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen 9700 RB, the Netherlands
| | - Mathilde Boissel
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, Lille 59000, France
| | - Jin Fang Chai
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore 117549, Singapore
| | - Xu Chen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm 17177, Sweden
| | - Jasmin Divers
- Division of Biostatistical Sciences, Department of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Ilaria Gandin
- Department of Medical Sciences, University of Trieste, Trieste 34137, Italy
| | - Chuan Gao
- Department of Molecular Genetics and Genomics Program, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Franco Giulianini
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02215, USA
| | - Anuj Goel
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, Oxfordshire OX3 9DU, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Sarah E Harris
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh EH8 9JZ, UK; Medical Genetics Section, University of Edinburgh Centre for Genomic and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Fernando Pires Hartwig
- Postgraduate Program in Epidemiology, Federal University of Pelotas, Pelotas, RS 96020220, Brazil
| | - Andrea R V R Horimoto
- Lab Genetics and Molecular Cardiology, Department of Cardiology, Heart Institute, University of Sao Paulo, Sao Paulo, Brazil
| | - Fang-Chi Hsu
- Division of Biostatistical Sciences, Department of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Anne U Jackson
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mika Kähönen
- Department of Clinical Physiology, Faculty of Medicine and Life Sciences, University of Tampere, Tampere 33014, Finland; Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland
| | | | - Brigitte Kühnel
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany; Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Karin Leander
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm 17177, Sweden
| | - Wen-Jane Lee
- Department of Medical Research, Taichung Veterans General Hospital, Department of Social Work, Tunghai University, Taichung 40705, Taiwan
| | - Keng-Hung Lin
- Department of Opthalmology, Taichung Veterans General Hospital, Taichung 40705, Taiwan
| | - Jian 'an Luan
- MRC Epidemiology Unit, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Colin A McKenzie
- Tropical Metabolism Research Unit, Tropical Medicine Research Institute, University of the West Indies, Mona JMAAW15, Jamaica
| | - He Meian
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| | - Christopher P Nelson
- Department of Cardiovascular Sciences, University of Leicester, Leicester LE3 9QP, UK; NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester LE3 9QP, UK
| | - Rainer Rauramaa
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio 70100, Finland
| | - Nicole Schupf
- Taub Institute for Research on Alzheimer disease and the Aging Brain, Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY 10032, USA
| | - Robert A Scott
- MRC Epidemiology Unit, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Wayne H H Sheu
- Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung 40705, Taiwan; School of Medicine, National Yang-ming University, Taipei, Taiwan; School of Medicine, National Defense Medical Center, Taipei, Taiwan; Institute of Medical Technology, National Chung-Hsing University, Taichung 40705, Taiwan
| | - Alena Stančáková
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio 70210, Finland
| | - Fumihiko Takeuchi
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo 1628655, Japan
| | - Peter J van der Most
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen 9700 RB, the Netherlands
| | - Tibor V Varga
- Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University, Malmö, Skåne 205 02, Sweden
| | - Heming Wang
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Yajuan Wang
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Erin B Ware
- School of Public Health, Department of Epidemiology, University of Michigan, Ann Arbor, MI 48109, USA; Institute for Social Research, Research Center for Group Dynamics, University of Michigan, Ann Arbor, MI 48104, USA
| | - Stefan Weiss
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine and Ernst-Moritz Arndt University Greifswald, Greifswald 17487, Germany; DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald 17475, Germany
| | - Wanqing Wen
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Lisa R Yanek
- General Internal Medicine, GeneSTAR Research Program, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Weihua Zhang
- Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK; Department of Cardiology, Ealing Hospital, Middlesex UB1 3HW, UK
| | - Jing Hua Zhao
- MRC Epidemiology Unit, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Saima Afaq
- Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK
| | - Tamuno Alfred
- Icahn School of Medicine at Mount Sinai, The Charles Bronfman Institute for Personalized Medicine, New York, NY 10029, USA
| | - Najaf Amin
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Dan Arking
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Tin Aung
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore 169856, Singapore; Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Singapore 169857, Singapore; Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore 117597, Singapore
| | - R Graham Barr
- Departments of Medicine and Epidemiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Lawrence F Bielak
- School of Public Health, Department of Epidemiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Eric Boerwinkle
- Human Genetics Center, The University of Texas School of Public Health, Houston, TX 77030, USA; Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX 77030, USA
| | - Erwin P Bottinger
- Icahn School of Medicine at Mount Sinai, The Charles Bronfman Institute for Personalized Medicine, New York, NY 10029, USA
| | - Peter S Braund
- Department of Cardiovascular Sciences, University of Leicester, Leicester LE3 9QP, UK; NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester LE3 9QP, UK
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Medicine, University of Washington, Seattle, WA 98101, USA
| | - Ulrich Broeckel
- Section of Genomic Pediatrics, Department of Pediatrics, Medicine and Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Claudia P Cabrera
- William Harvey Research Institute, Clinical Pharmacology, Queen Mary University of London, London EC1M 6BQ, UK; NIHR Cardiovascular Biomedical Research Unit, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Brian Cade
- Sleep Medicine and Circadian Disorders, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Yu Caizheng
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| | - Archie Campbell
- Centre for Genomic & Experimental Medicine, Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Mickaël Canouil
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, Lille 59000, France
| | - Aravinda Chakravarti
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ganesh Chauhan
- Centre for Brain Research, Indian Institute of Schience, Bangalore 560012, India
| | - Kaare Christensen
- The Danish Aging Research Center, Institute of Public Health, University of Southern Denmark, Odense, Denmark
| | - Massimiliano Cocca
- Department of Medical Sciences, University of Trieste, Trieste 34137, Italy
| | - Francis S Collins
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - John M Connell
- Ninewells Hospital & Medical School, University of Dundee, Dundee DD1 9SY, UK
| | - Renée de Mutsert
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden 2300RC, the Netherlands
| | | | - Stephanie Debette
- Inserm U1219 Neuroepidemiology, University of Bordeaux, Bordeaux, France; Department of Neurology, University Hospital, Bordeaux, France; Boston University School of Medicine, Boston, MA 02118, USA
| | - Marcus Dörr
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald 17475, Germany; Department of Internal Medicine B, University Medicine Greifswald, Greifswald 17475, Germany
| | - Qing Duan
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Charles B Eaton
- Department of Family Medicine and Epidemiology, Alpert Medical School of Brown University, Providence, RI 02860, USA
| | - Georg Ehret
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Division of Cardiology, Department of Specialties of Medicine, Geneva University Hospital, Geneva 1211, Switzerland
| | - Evangelos Evangelou
- Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK; Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina 45110, Greece
| | - Jessica D Faul
- Institute for Social Research, Survey Research Center, University of Michigan, Ann Arbor, MI 48104, USA
| | - Virginia A Fisher
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Nita G Forouhi
- MRC Epidemiology Unit, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Oscar H Franco
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Yechiel Friedlander
- Braun School of Public Health, Hebrew University-Hadassah Medical Center, Jerusalem 91120, Israel
| | - He Gao
- Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK; MRC-PHE Centre for Environment and Health, Department of Epidemiology & Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Bruna Gigante
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm 17177, Sweden
| | - Misa Graff
- Department of Epidemiology, University of North Carolina Gillings School of Global Public Health, Chapel Hill, NC 27514, USA
| | - C Charles Gu
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Dongfeng Gu
- Department of Epidemiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Preeti Gupta
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore 169856, Singapore
| | - Saskia P Hagenaars
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh EH8 9JZ, UK; Department of Psychology, The University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIH, Bethesda, MD 20892, USA
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA; Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Sami Heikkinen
- University of Eastern Finland, Institute of Biomedicine, Kuopio 70211, Finland
| | - Chew-Kiat Heng
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; Khoo Teck Puat - National University Children's Medical Institute, National University Health System, Singapore 119228, Singapore
| | - Makoto Hirata
- Laboratory of Genome Technology, Human Genome Center, Institute of Medical Science, The University of Tokyo, Minato-ku 108-8639, Japan
| | - Albert Hofman
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Barbara V Howard
- MedStar Health Research Institute, Hyattsville, MD 20782, USA; Center for Clinical and Translational Sciences and Department of Medicine, Georgetown-Howard Universities, Washington, DC 20057, USA
| | - Steven Hunt
- Cardiovascular Genetics, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84108, USA; Weill Cornell Medicine in Qatar, Doha, Qatar
| | - Marguerite R Irvin
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Yucheng Jia
- Genomic Outcomes, Department of Pediatrics, LABioMed at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Roby Joehanes
- Hebrew SeniorLife, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02131, USA; Framingham Heart Study, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20982, USA
| | - Anne E Justice
- Department of Epidemiology, University of North Carolina Gillings School of Global Public Health, Chapel Hill, NC 27514, USA
| | - Tomohiro Katsuya
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita 5650871, Japan; Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita 5650871, Japan
| | - Joel Kaufman
- Epidemiology, Department of Occupational and Environmental Medicine Program, University of Washington, Seattle, WA 98105, USA
| | - Nicola D Kerrison
- MRC Epidemiology Unit, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Chiea Chuen Khor
- Genome Institute of Singapore, Agency for Science Technology and Research, Singapore 138672, Singapore; Department of Biochemistry, National University of Singapore, Singapore 117596, Singapore
| | - Woon-Puay Koh
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore 117549, Singapore; Duke-NUS Medical School, Singapore 169857, Singapore
| | - Heikki A Koistinen
- Department of Health, National Institute for Health and Welfare, Helsinki 00271, Finland; Department of Medicine and Abdominal Center: Endocrinology, University of Helsinki and Helsinki University Central Hospital, Helsinki 00029, Finland
| | - Pirjo Komulainen
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio 70100, Finland
| | - Charles Kooperberg
- Fred Hutchinson Cancer Research Center, University of Washington School of Public Health, Seattle, WA 98109, USA
| | - Jose E Krieger
- Lab Genetics and Molecular Cardiology, Department of Cardiology, Heart Institute, University of Sao Paulo, Sao Paulo, Brazil
| | - Michiaki Kubo
- Center for Integrative Medical Sciences, RIKEN, Yokohama 230-0045, Japan
| | - Johanna Kuusisto
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio 70210, Finland
| | - Carl D Langefeld
- Division of Biostatistical Sciences, Department of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | | | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIH, Bethesda, MD 20892, USA
| | - Benjamin Lehne
- Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK
| | - Cora E Lewis
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35205, USA
| | - Yize Li
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sing Hui Lim
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore 169856, Singapore
| | - Shiow Lin
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Jianjun Liu
- Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore, Singapore 117549, Singapore; Genome Institute of Singapore, Agency for Science Technology and Research, Singapore 138672, Singapore
| | - Jingmin Liu
- WHI CCC, Fred Hutchinson Cancer Research Center, Seattle, WA 98115, USA
| | - Kiang Liu
- Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yeheng Liu
- Genomic Outcomes, Department of Pediatrics, LABioMed at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Marie Loh
- Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK; Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research 138648, Singapore
| | - Kurt K Lohman
- Division of Biostatistical Sciences, Department of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Jirong Long
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Tin Louie
- Department of Biostatistics, University of Washington, Seattle, WA 98105, USA
| | - Reedik Mägi
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Anubha Mahajan
- Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany; Institute of Human Genetics, Technische Universität München, Munich 80333, Germany
| | - Andres Metspalu
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Lili Milani
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Yukihide Momozawa
- Laboratory for Genotyping Development, Center for Integrative Medical Sciences, RIKEN, Yokohama 230-0045, Japan
| | - Andrew P Morris
- Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Department of Biostatistics, University of Liverpool, Liverpool L69 3GL, UK
| | - Thomas H Mosley
- Geriatrics, Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Peter Munson
- Mathematical and Statistical Computing Laboratory, Center for Information Technology, NIH, Bethesda, MD 20892, USA
| | - Alison D Murray
- The Institute of Medical Sciences, Aberdeen Biomedical Imaging Centre, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Mike A Nalls
- Data Tecnica International, Glen Echo, MD 20812, USA; Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD 20892, USA
| | - Ubaydah Nasri
- Genomic Outcomes, Department of Pediatrics, LABioMed at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Jill M Norris
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO 80045, USA
| | - Kari North
- Department of Epidemiology, University of North Carolina Gillings School of Global Public Health, Chapel Hill, NC 27514, USA
| | | | - Sandosh Padmanabhan
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Walter R Palmas
- Internal Medicine, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Nicholette D Palmer
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - James S Pankow
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN 55454, USA
| | - Nancy L Pedersen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm 17177, Sweden
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Neuherberg 85764, Germany
| | - Patricia A Peyser
- School of Public Health, Department of Epidemiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ozren Polasek
- Faculty of Medicine, University of Split, Split, Croatia
| | - Olli T Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku 20521, Finland; Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku 20520, Finland
| | - Frida Renström
- Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University, Malmö, Skåne 205 02, Sweden; Department of Biobank Research, Umeå University, Umeå, Västerbotten 901 87, Sweden
| | - Treva K Rice
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Paul M Ridker
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02215, USA
| | | | - Jennifer G Robinson
- Department of Epidemiology and Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Lynda M Rose
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02215, USA
| | - Igor Rudan
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh EH8 9AG, UK
| | - Charumathi Sabanayagam
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore 169856, Singapore; Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Singapore 169857, Singapore
| | | | - Kevin Sandow
- Genomic Outcomes, Department of Pediatrics, LABioMed at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Carsten O Schmidt
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald 17475, Germany; Institute for Community Medicine, University Medicine Greifswald, Greifswald 17475, Germany
| | - Pamela J Schreiner
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN 55454, USA
| | - William R Scott
- Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK; National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Sudha Seshadri
- Framingham Heart Study, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20982, USA; Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Peter Sever
- International Centre for Circulatory Health, Imperial College London, London W2 1PG, UK
| | - Colleen M Sitlani
- Cardiovascular Health Research Unit, Medicine, University of Washington, Seattle, WA 98101, USA
| | - Jennifer A Smith
- School of Public Health, Department of Epidemiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen 9700 RB, the Netherlands
| | - John M Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh EH8 9JZ, UK; Alzheimer Scotland Dementia Research Centre, The University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany; Chair of Genetic Epidemiology, IBE, Faculty of Medicine, LMU Munich, Munich 81377, Germany
| | - Hua Tang
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Kent D Taylor
- Genomic Outcomes, Department of Pediatrics, LABioMed at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Yik Ying Teo
- Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore, Singapore 117549, Singapore; Genome Institute of Singapore, Agency for Science Technology and Research, Singapore 138672, Singapore; Life Sciences Institute, National University of Singapore, Singapore, Singapore 117456, Singapore; NUS Graduate School for Integrative Science and Engineering, National University of Singapore, Singapore 117456, Singapore; Department of Statistics and Applied Probability, National University of Singapore, Singapore 117546, Singapore
| | - Yih Chung Tham
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore 169856, Singapore
| | - André G Uitterlinden
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany; Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Lihua Wang
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Ya X Wang
- Beijing Institute of Ophthalmology, Beijing Ophthalmology and Visual Science Key Lab, Beijing Tongren Eye Center, Capital Medical University, Beijing, China 100730, China
| | - Wen Bin Wei
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Christine Williams
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Gregory Wilson
- Jackson Heart Study, Department of Public Health, Jackson State University, Jackson, MS 39213, USA
| | - Mary K Wojczynski
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Jie Yao
- Genomic Outcomes, Department of Pediatrics, LABioMed at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Jian-Min Yuan
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA; Division of Cancer Control and Population Sciences, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15232, USA
| | - Alan B Zonderman
- Behavioral Epidemiology Section, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Diane M Becker
- General Internal Medicine, GeneSTAR Research Program, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Donald W Bowden
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - John C Chambers
- Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK; Department of Cardiology, Ealing Hospital, Middlesex UB1 3HW, UK
| | - Yii-Der Ida Chen
- Genomic Outcomes, Department of Pediatrics, LABioMed at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Ulf de Faire
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm 17177, Sweden
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh EH8 9JZ, UK; Department of Psychology, The University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Tõnu Esko
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia; Broad Institute of the Massachusetts Institute of Technology and Harvard University, Boston, MA 02142, USA
| | - Martin Farrall
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, Oxfordshire OX3 9DU, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Terrence Forrester
- Tropical Metabolism Research Unit, Tropical Medicine Research Institute, University of the West Indies, Mona JMAAW15, Jamaica
| | - Paul W Franks
- Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University, Malmö, Skåne 205 02, Sweden; Harvard T.H. Chan School of Public Health, Department of Nutrition, Harvard University, Boston, MA 02115, USA; Department of Public Health & Clinical Medicine, Umeå University, Umeå, Västerbotten 901 85, Sweden
| | - Barry I Freedman
- Division of Nephrology, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Philippe Froguel
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, Lille 59000, France; Department of Genomics of Common Disease, Imperial College London, London W12 0NN, UK
| | - Paolo Gasparini
- Department of Medical Sciences, University of Trieste, Trieste 34137, Italy; Division Experimental Genetics, Sidra, Doha 26999, Qatar
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg 85764, Germany
| | - Bernardo Lessa Horta
- Postgraduate Program in Epidemiology, Federal University of Pelotas, Pelotas, RS 96020220, Brazil
| | - Yi-Jen Hung
- Endocrinology and Metabolism, Tri-Service General Hospital, National Defense Medical Center, Taipei City, Taipei 11490, Taiwan
| | - Jost B Jonas
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China; Department of Ophthalmology, Medical Faculty Mannheim, University Heidelberg, Mannheim 68167, Germany
| | - Norihiro Kato
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo 1628655, Japan
| | - Jaspal S Kooner
- Department of Cardiology, Ealing Hospital, Middlesex UB1 3HW, UK; National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio 70210, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere 33520, Finland; Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Lifes Sciences, University of Tampere, Tampere 33014, Finland
| | - Kae-Woei Liang
- School of Medicine, National Yang-ming University, Taipei, Taiwan; Cardiovascular Center, Taichung Veterans General Hospital, Taichung 40705, Taiwan; Department of Medicine, China Medical University, Taichung 40705, Taiwan
| | - Patrik K E Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm 17177, Sweden
| | - Anne B Newman
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Albertine J Oldehinkel
- Department of Psychiatry, University of Groningen, University Medical Center Groningen, Groningen 9700 RB, the Netherlands
| | - Alexandre C Pereira
- Lab Genetics and Molecular Cardiology, Department of Cardiology, Heart Institute, University of Sao Paulo, Sao Paulo, Brazil; Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Susan Redline
- Sleep Medicine and Circadian Disorders, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Rainer Rettig
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald 17475, Germany; Institute of Physiology, University Medicine Greifswald, Greifswald 17495, Germany
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, Leicester LE3 9QP, UK; NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester LE3 9QP, UK
| | - James Scott
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Pim van der Harst
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen 9700 RB, the Netherlands
| | - Lynne E Wagenknecht
- Department of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | | | - Hugh Watkins
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, Oxfordshire OX3 9DU, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - David R Weir
- Institute for Social Research, Survey Research Center, University of Michigan, Ann Arbor, MI 48104, USA
| | | | - Tangchun Wu
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Yoichiro Kamatani
- Laboratory for Statistical Analysis, Center for Integrative Medical Sciences, RIKEN, Yokohama 230-0045, Japan
| | - Cathy C Laurie
- Department of Biostatistics, University of Washington, Seattle, WA 98105, USA
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Richard S Cooper
- Department of Public Health Sciences, Loyola University Chicago, Maywood, IL 60153, USA
| | - Michele K Evans
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur 201, Iceland; Faculty of Medicine, University of Iceland, Reykjavik 101, Iceland
| | - Sharon L R Kardia
- School of Public Health, Department of Epidemiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Stephen B Kritchevsky
- Sticht Center for Health Aging and Alzheimer's Prevention, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Daniel Levy
- Framingham Heart Study, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20982, USA; Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Jeff R O'Connell
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Epidemiology, Medicine and Health Services, University of Washington, Seattle, WA 98101, USA; Kaiser Permanente Washington, Health Research Institute, Seattle, WA 98101, USA
| | - Rob M van Dam
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore 117549, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Mario Sims
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39213, USA
| | - Donna K Arnett
- Dean's Office, University of Kentucky College of Public Health, Lexington, KY 40536, USA
| | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden 2300RC, the Netherlands; Department of Public Health and Primary Care, Leiden University Medical Center, Leiden 2300RC, the Netherlands
| | - Tanika N Kelly
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA
| | - Ervin R Fox
- Cardiology, Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit, Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Charles N Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Michael A Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - E Shyong Tai
- Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore, Singapore 117549, Singapore; Duke-NUS Medical School, Singapore 169857, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Tien Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore 169856, Singapore; Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Singapore 169857, Singapore; Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore 117597, Singapore
| | - Ruth J F Loos
- Icahn School of Medicine at Mount Sinai, The Charles Bronfman Institute for Personalized Medicine, New York, NY 10029, USA; Icahn School of Medicine at Mount Sinai, The Mindich Child Health and Development Institute, New York, NY 10029, USA
| | - Alex P Reiner
- Fred Hutchinson Cancer Research Center, University of Washington School of Public Health, Seattle, WA 98109, USA
| | - Jerome I Rotter
- Genomic Outcomes, Department of Pediatrics, LABioMed at Harbor-UCLA Medical Center, Torrance, CA 90502, USA; Genomic Outcomes, Department of Medicine, LABioMed at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Xiaofeng Zhu
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Laura J Bierut
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - W James Gauderman
- Division of Biostatistics, Department of Preventive Medicine, University of Southern California, Los Angeles, CA 90032, USA
| | - Mark J Caulfield
- William Harvey Research Institute, Clinical Pharmacology, Queen Mary University of London, London EC1M 6BQ, UK; NIHR Cardiovascular Biomedical Research Unit, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK; MRC-PHE Centre for Environment and Health, Department of Epidemiology & Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Kenneth Rice
- Department of Biostatistics, University of Washington, Seattle, WA 98105, USA
| | - Patricia B Munroe
- William Harvey Research Institute, Clinical Pharmacology, Queen Mary University of London, London EC1M 6BQ, UK; NIHR Barts Cardiovascular Biomedical Research Unit, Queen Mary University of London, London EC1M 6BQ, UK
| | - Alanna C Morrison
- Department of Epidemiology, Human Genetics, and Environmental Sciences, The University of Texas School of Public Health, Houston, TX 77030, USA
| | - L Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA; Framingham Heart Study, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20982, USA
| | - Dabeeru C Rao
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Daniel I Chasman
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02215, USA.
| |
Collapse
|
95
|
Gaur P, Saini S, Vats P, Kumar B. Regulation, signalling and functions of hormonal peptides in pulmonary vascular remodelling during hypoxia. Endocrine 2018; 59:466-480. [PMID: 29383676 DOI: 10.1007/s12020-018-1529-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 01/10/2018] [Indexed: 01/06/2023]
Abstract
Hypoxic state affects organism primarily by decreasing the amount of oxygen reaching the cells and tissues. To adjust with changing environment organism undergoes mechanisms which are necessary for acclimatization to hypoxic stress. Pulmonary vascular remodelling is one such mechanism controlled by hormonal peptides present in blood circulation for acclimatization. Activation of peptides regulates constriction and relaxation of blood vessels of pulmonary and systemic circulation. Thus, understanding of vascular tone maintenance and hypoxic pulmonary vasoconstriction like pathophysiological condition during hypoxia is of prime importance. Endothelin-1 (ET-1), atrial natriuretic peptide (ANP), and renin angiotensin system (RAS) function, their receptor functioning and signalling during hypoxia in different body parts point them as disease markers. In vivo and in vitro studies have helped understanding the mechanism of hormonal peptides for better acclimatization to hypoxic stress and interventions for better management of vascular remodelling in different models like cell, rat, and human is discussed in this review.
Collapse
Affiliation(s)
- Priya Gaur
- Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, India
| | - Supriya Saini
- Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, India
| | - Praveen Vats
- Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, India.
| | - Bhuvnesh Kumar
- Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, India
| |
Collapse
|
96
|
Arginine Vasopressin-Aquaporin-2 Pathway-Mediated Dehydration Effects of Electroacupuncture in Guinea Pig Model of AVP-Induced Eendolymphatic Hydrops. Chin J Integr Med 2018; 25:763-769. [PMID: 29335859 DOI: 10.1007/s11655-017-2411-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2015] [Indexed: 10/18/2022]
Abstract
OBJECTIVE To investigate the effects of electroacupuncture (EA) on endolymphatic hydrops (EH) and the regulation of arginine vasopressin (AVP)-aquaporin-2 (AQP2) pathway in guinea pigs. METHODS EH was induced in male guinea pigs by an intraperitoneal injection of AVP. For the treatment, EA was delivered to Baihui (GV 20) and Tinggong (SI 19) acupoints, once per day for 10 consecutive days. In histomorphological studies, cochlear hydrops degree was evaluated by hematoxylin-eosin (HE) staining, and then the ratio of scala media (SM) area to SM + scala vestibuli (SV) area (R value) was calculated. In mechanical studies, a comparison of plasma AVP (p-AVP) concentrations, cyclic adenosine monophosphate (cAMP) levels, vasopressin type 2 receptor (V2R) and AQP2 mRNA expressions in the cochlea were compared among groups. RESULTS EA significantly reduced cochlear hydrops in guinea pigs (P=0.001). EA significantly attenuated the AVPinduced up-regulation of p-AVP concentrations (P=0.006), cochlear cAMP levels (P=0.003) and AQP2 mRNA expression (P=0.016), and up-regulated the expression of V2R mRNA (P=0.004) in the cochlea. CONCLUSIONS The dehydrating effect of EA might be associated with its inhibition of AVP-AQP2 pathway activation.
Collapse
|
97
|
Hines MT. Clinical Approach to Commonly Encountered Problems. EQUINE INTERNAL MEDICINE 2018. [PMCID: PMC7158300 DOI: 10.1016/b978-0-323-44329-6.00007-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
98
|
Umejiego EN, Wang Y, Knepper MA, Chou CL. Roflumilast and aquaporin-2 regulation in rat renal inner medullary collecting duct. Physiol Rep 2017; 5:5/2/e13121. [PMID: 28108651 PMCID: PMC5269416 DOI: 10.14814/phy2.13121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 11/29/2016] [Accepted: 12/12/2016] [Indexed: 12/23/2022] Open
Abstract
Roflumilast is a cyclic nucleotide phosphodiesterase inhibitor that is FDA‐approved for treatment of chronic obstructive pulmonary disease. With a view toward possible use for treatment of patients with X‐linked nephrogenic diabetes insipidus (NDI) due to hemizygous mutations in the V2 vasopressin receptor, this study sought to determine the effect of roflumilast on aquaporin‐2 (AQP2) phosphorylation, AQP2 trafficking, and water permeability in the rat inner medullary collecting duct (IMCD). In the presence of the vasopressin analog dDAVP (0.1 nmol/L), both roflumilast and its active metabolite roflumilast N‐oxide (RNO) significantly increased phosphorylation at S256, S264, and S269, and decreased phosphorylation at S261 (immunoblotting) in IMCD suspensions in a dose‐dependent manner (3–3000 nmol/L). Another commonly used phosphodiesterase inhibitor, IBMX, affected phosphorylation only at the highest concentration in this range. However, neither roflumilast nor RNO had an effect on AQP2 phosphorylation in the absence of vasopressin. Furthermore, roflumilast alone did not increase AQP2 trafficking to the plasma membrane (immunofluorescence) or increase water permeability in freshly microdissected perfused IMCD segments. We conclude that roflumilast can be used to enhance vasopressin's action on AQP2 activity in the renal collecting duct, but has no detectable effect in the absence of vasopressin. These findings suggest that roflumilast may not have a beneficial effect in X‐linked NDI, but could find useful application in acquired NDI.
Collapse
Affiliation(s)
- Ezigbobiara N Umejiego
- Epithelial Systems Biology Laboratory, Systems Biology Center NHLBI National Institutes of Health, Bethesda, Maryland, 20892-1603
| | - Yanhua Wang
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia, 30322
| | - Mark A Knepper
- Epithelial Systems Biology Laboratory, Systems Biology Center NHLBI National Institutes of Health, Bethesda, Maryland, 20892-1603
| | - Chung-Lin Chou
- Epithelial Systems Biology Laboratory, Systems Biology Center NHLBI National Institutes of Health, Bethesda, Maryland, 20892-1603
| |
Collapse
|
99
|
Pruett WA, Clemmer JS, Hester RL. Validation of an integrative mathematical model of dehydration and rehydration in virtual humans. Physiol Rep 2017; 4:4/22/e13015. [PMID: 27899683 PMCID: PMC5358000 DOI: 10.14814/phy2.13015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 09/15/2016] [Accepted: 09/23/2016] [Indexed: 11/30/2022] Open
Abstract
Water homeostasis is one of the body's most critical tasks. Physical challenges to the body, including exercise and surgery, almost always coordinate with some change in water handling reflecting the changing needs of the body. Vasopressin is the most important hormone that contributes to short‐term water homeostasis. By manipulating vascular tone and regulating water reabsorption in the collecting duct of the kidneys, vasopressin can mediate the retention or loss of fluids quickly. In this study, we validated HumMod, an integrative mathematical model of human physiology, against six different challenges to water homeostasis with special attention to the secretion of vasopressin and maintenance of electrolyte balance. The studies chosen were performed in normal men and women, and represent a broad spectrum of perturbations. HumMod successfully replicated the experimental results, remaining within 1 standard deviation of the experimental means in 138 of 161 measurements. Only three measurements lay outside of the second standard deviation. Observations were made on serum osmolarity, serum vasopressin concentration, serum sodium concentration, urine osmolarity, serum protein concentration, hematocrit, and cumulative water intake following dehydration. This validation suggests that HumMod can be used to understand water homeostasis under a variety of conditions.
Collapse
Affiliation(s)
- W Andrew Pruett
- Department of Physiology, Center for Computational Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - John S Clemmer
- Department of Physiology, Center for Computational Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Robert L Hester
- Department of Physiology, Center for Computational Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
100
|
Trimpert C, Wesche D, de Groot T, Pimentel Rodriguez MM, Wong V, van den Berg DTM, Cheval L, Ariza CA, Doucet A, Stagljar I, Deen PMT. NDFIP allows NEDD4/NEDD4L-induced AQP2 ubiquitination and degradation. PLoS One 2017; 12:e0183774. [PMID: 28931009 PMCID: PMC5606929 DOI: 10.1371/journal.pone.0183774] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 08/10/2017] [Indexed: 12/13/2022] Open
Abstract
Regulation of our water homeostasis is fine-tuned by dynamic translocation of Aquaporin-2 (AQP2)-bearing vesicles to and from the plasma membrane of renal principal cells. Whereas binding of vasopressin to its type-2 receptor initiates a cAMP-protein kinase A cascade and AQP2 translocation to the apical membrane, this is counteracted by protein kinase C-activating hormones, resulting in ubiquitination-dependent internalization of AQP2. The proteins targeting AQP2 for ubiquitin-mediated degradation are unknown. In collecting duct mpkCCD cells, siRNA knockdown of NEDD4 and NEDD4L E3 ligases yielded increased AQP2 abundance, but they did not bind AQP2. Membrane Yeast Two-Hybrid assays using full-length AQP2 as bait, identified NEDD4 family interacting protein 2 (NDFIP2) to bind AQP2. NDFIP2 and its homologue NDFIP1 have PY motifs by which they bind NEDD4 family members and bring them close to target proteins. In HEK293 cells, NDFIP1 and NDFIP2 bound AQP2 and were essential for NEDD4/NEDD4L-mediated ubiquitination and degradation of AQP2, an effect not observed with PY-lacking NDFIP1/2 proteins. In mpkCCD cells, downregulation of NDFIP1, NEDD4 and NEDD4L, but not NDFIP2, increased AQP2 abundance. In mouse kidney, Ndfip1 and Ndfip2 mRNA distribution was similar and high in proximal tubules and collecting ducts, which was also found for NDFIP1 proteins. Our results reveal that NEDD4/NEDD4L mediate ubiquitination and degradation of AQP2, but that NDFIP proteins are needed to connect NEDD4/NEDD4L to AQP2. As NDFIP1/2 bind many NEDD4 family E3 ligases, which are implicated in several cellular processes, NDFIP1/2 may be the missing link for AQP2 ubiquitination and degradation from different subcellular locations.
Collapse
Affiliation(s)
- Christiane Trimpert
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Daniel Wesche
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Theun de Groot
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Martha M. Pimentel Rodriguez
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
- Donnelly Centre for Cellular and Biomolecular Research, Departments of Biochemistry and Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Victoria Wong
- Donnelly Centre for Cellular and Biomolecular Research, Departments of Biochemistry and Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | | | - Lydie Cheval
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Université Paris Descartes, Sorbonne Paris Cité, CNRS, Centre de Recherche des Cordeliers, Paris, France
| | - Carolina A. Ariza
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Alain Doucet
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Université Paris Descartes, Sorbonne Paris Cité, CNRS, Centre de Recherche des Cordeliers, Paris, France
| | - Igor Stagljar
- Donnelly Centre for Cellular and Biomolecular Research, Departments of Biochemistry and Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Peter M. T. Deen
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
- * E-mail:
| |
Collapse
|