51
|
Otterpohl KL, Hart RG, Evans C, Surendran K, Chandrasekar I. Nonmuscle myosin 2 proteins encoded by Myh9, Myh10, and Myh14 are uniquely distributed in the tubular segments of murine kidney. Physiol Rep 2018; 5. [PMID: 29208685 PMCID: PMC5727274 DOI: 10.14814/phy2.13513] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 10/27/2017] [Accepted: 10/30/2017] [Indexed: 11/24/2022] Open
Abstract
The diverse epithelial cell types of the kidneys are segregated into nephron segments and the collecting ducts in order to endow each tubular segment with unique functions. The rich diversity of the epithelial cell types is highlighted by the unique membrane channels and receptors expressed within each nephron segment. Our previous work identified a critical role for Myh9 and Myh10 in mammalian endocytosis. Here, we examined the expression patterns of Nonmuscle myosin 2 (NM2) heavy chains encoded by Myh9, Myh10, and Myh14 in mouse kidneys as these genes may confer unique nephron segment‐specific membrane transport properties. Interestingly, we found that each segment of the renal tubules predominately expressed only two of the three NM2 isoforms, with isoform‐specific subcellular localization, and different levels of expression within a nephron segment. Additionally, we identify Myh14 to be restricted to the intercalated cells and Myh10 to be restricted to the principal cells within the collecting ducts and connecting segments. We speculate that the distinct expression pattern of the NM2 proteins likely reflects the diversity of the intracellular trafficking machinery present within the different renal tubular epithelial segments.
Collapse
Affiliation(s)
- Karla L Otterpohl
- Enabling Technologies Group - Sanford Research, Sioux Falls, South Dakota, USA
| | - Ryan G Hart
- Enabling Technologies Group - Sanford Research, Sioux Falls, South Dakota, USA
| | - Claire Evans
- Molecular Pathology Core, Sanford Research, Sioux Falls, South Dakota, USA
| | - Kameswaran Surendran
- Pediatrics and Rare Diseases Group - Sanford Research, Sioux Falls, South Dakota, USA.,Department of Pediatrics, USD Sanford School of Medicine, Sioux Falls, South Dakota, USA
| | - Indra Chandrasekar
- Enabling Technologies Group - Sanford Research, Sioux Falls, South Dakota, USA.,Department of Pediatrics, USD Sanford School of Medicine, Sioux Falls, South Dakota, USA
| |
Collapse
|
52
|
Baturina GS, Palchikova IG, Konev AA, Smirnov ES, Katkova LE, Solenov EI, Iskakov IА. STUDY OF THE EFFECT OF HYPOTHERMIC CONSERVATION ON THE INTRACELLULAR SODIUM CONCENTRATION IN THE ENDOTHELIUM OF CORNEAL TRANSPLANTS. Vavilovskii Zhurnal Genet Selektsii 2018. [DOI: 10.18699/vj18.379] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Endothelial keratoplasty has become the treatment of choice for corneal endothelial dysfunction. Advancements in the surgical treatment of corneal endothelial diseases depend on progress in graft conservation and its related advantages in assessing the suitability of grafts for transplantation. Transport of water and ions by cornea endothelium is important for the optic properties of cornea. In this work, we study the intracellular sodium concentration in cornea endothelial cells in samples of pig cornea that underwent hypothermic conservation for 1 and 10 days and endothelial cells of human cornea grafts after 10-day conservation. The concentration of intracellular sodium in preparations of endothelial cells was assayed using fluorescent dye SodiumGreen. The fluorescent images were analyzed with the custom-made computer program CytoDynamics. An increased level of intracellular sodium was shown in the endothelium after 10-day conservation in comparison with one-day conservation (pig samples). Sodium permeability of pig endothelial cell plasma membranes significantly decreased in these samples. Assessment of intracellular sodium in human cornea endothelium showed a higher level – as was in analogues pig samples of the corneal endothelium. The assay of the intracellular sodium balance concentration established in endothelial cells after hypothermic conservation in mediums L-15 and Optisol-GS showed a significant advantage of specialized me dium Optisol-GS. The balanced intracellular concentration after 10 days of hypothermic conservation was significantly lower in cells incubated at 4 °C in Optisol-GS (L-15, 128 ± 14, n = 15; Optisol-GS, 108 ± 14, n = 11; mM, p < 0.001). Intracellular sodium concentration could be a useful parameter for assessing cornea endothelium cell viability.
Collapse
|
53
|
Gonsalez SR, Ferrão FM, Souza AMD, Lowe J, Morcillo LDSL. Inappropriate activity of local renin-angiotensin-aldosterone system during high salt intake: impact on the cardio-renal axis. ACTA ACUST UNITED AC 2018; 40:170-178. [PMID: 29944159 PMCID: PMC6533978 DOI: 10.1590/2175-8239-jbn-3661] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Accepted: 01/11/2017] [Indexed: 12/12/2022]
Abstract
Although there is a general agreement on the recommendation for reduced salt
intake as a public health issue, the mechanism by which high salt intake
triggers pathological effects on the cardio-renal axis is not completely
understood. Emerging evidence indicates that the renin-angiotensin-aldosterone
system (RAAS) is the main target of high Na+ intake. An inappropriate
activation of tissue RAAS may lead to hypertension and organ damage. We reviewed
the impact of high salt intake on the RAAS on the cardio-renal axis highlighting
the molecular pathways that leads to injury effects. We also provide an
assessment of recent observational studies related to the consequences of
non-osmotically active Na+ accumulation, breaking the paradigm that
high salt intake necessarily increases plasma Na+ concentration
promoting water retention
Collapse
Affiliation(s)
- Sabrina Ribeiro Gonsalez
- Universidade Federal do Rio de Janeiro, Instituto de Ciências Biomédicas, Rio de Janeiro, RJ, Brasil
| | - Fernanda Magalhães Ferrão
- Universidade do Estado do Rio de Janeiro, Instituto de Biologia Roberto Alcântara Gomes, Rio de Janeiro, RJ, Brasil
| | | | - Jennifer Lowe
- Universidade Federal do Rio de Janeiro, Instituto de Biofísica Carlos Chagas Filho, Rio de Janeiro, Brasil
| | | |
Collapse
|
54
|
Sampaio LS, Iannotti FA, Veneziani L, Borelli-Tôrres RT, De Maio F, Piscitelli F, Reis RAM, Di Marzo V, Einicker-Lamas M. Experimental ischemia/reperfusion model impairs endocannabinoid signaling and Na +/K + ATPase expression and activity in kidney proximal tubule cells. Biochem Pharmacol 2018; 154:482-491. [PMID: 29890144 DOI: 10.1016/j.bcp.2018.06.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 06/04/2018] [Indexed: 12/19/2022]
Abstract
LLC-PK1 cells, an immortalized epithelial cell line derived from pig renal proximal tubules, express all the major players of the endocannabinoid system (ECS) such as CB1, CB2 and TRPV1 receptors, as well as the main enzymes involved in the biosynthesis and degradation of the major endocannabinoids named 2-arachidonoylglycerol, 2-AG and anandamide, AEA. Here we investigated whether the damages caused by ischemic insults either in vitro using LLC-PK1 cells exposed to antimycin A (an inductor of ATP-depletion) or in vivo using Wistar rats in a classic renal ischemia and reperfusion (IR) protocol, lead to changes in AEA and 2-AG levels, as well as altered expression of genes from the main enzymes involved in the regulation of the ECS. Our data show that the mRNA levels of the CB1 receptor gene were downregulated, while the transcript levels of monoacylglycerol lipase (MAGL), the main 2-AG degradative enzyme, were upregulated in LLC-PK1 cells after IR model. Accordingly, IR was accompanied by a significant reduction in the levels of 2-AG and AEA, as well as of the two endocannabinoid related molecules, oleoylethanolamide (OEA) and palmitoylethanolamide (PEA) in LLC-PK1 cells. In kidney cortex homogenates, only AEA levels were significantly decreased. In addition, we found that in both the in vitro and in vivo model IR caused a reduction in the expression and activity of the Na+/K+ ATPase. These changes were reversed by the CB1/CB2 agonist WIN55,212, in a CB1-receptor dependent manner in the LLC-PK1 IR model. In conclusion, the ECS and Na+/K+ ATPase are down-regulated following IR in LLC-PK1 cells and rat kidney. We suggest that CB1 agonists might represent a potential strategy to reverse the consequences of IR injury in kidney tissues.
Collapse
Affiliation(s)
- Luzia S Sampaio
- Instituto de Biofísica Carlos Chagas Filho, UFRJ, Rio de Janeiro, Brazil; Lab. Neuroquímica, Instituto Nacional de Ciência e Tecnologia em Neurociência Translacional, Brazil; CAPES Foundation, Ministry of Education of Brazil, Brasilia DF 70.040-020, Brazil
| | - Fabio A Iannotti
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare, CNR, Pozzuoli (Naples), Italy
| | - Luciana Veneziani
- Instituto de Biofísica Carlos Chagas Filho, UFRJ, Rio de Janeiro, Brazil
| | - Rosa T Borelli-Tôrres
- Instituto de Biofísica Carlos Chagas Filho, UFRJ, Rio de Janeiro, Brazil; Lab. Neuroquímica, Instituto Nacional de Ciência e Tecnologia em Neurociência Translacional, Brazil
| | - Fabrizia De Maio
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare, CNR, Pozzuoli (Naples), Italy
| | - Fabiana Piscitelli
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare, CNR, Pozzuoli (Naples), Italy
| | - Ricardo A M Reis
- Instituto de Biofísica Carlos Chagas Filho, UFRJ, Rio de Janeiro, Brazil; Lab. Neuroquímica, Instituto Nacional de Ciência e Tecnologia em Neurociência Translacional, Brazil
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare, CNR, Pozzuoli (Naples), Italy
| | | |
Collapse
|
55
|
Some Biological Consequences of the Inhibition of Na,K-ATPase by Translationally Controlled Tumor Protein (TCTP). Int J Mol Sci 2018; 19:ijms19061657. [PMID: 29867020 PMCID: PMC6032315 DOI: 10.3390/ijms19061657] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/29/2018] [Accepted: 05/30/2018] [Indexed: 12/17/2022] Open
Abstract
Na,K-ATPase is an ionic pump that regulates the osmotic equilibrium and membrane potential of cells and also functions as a signal transducer. The interaction of Na,K-ATPase with translationally controlled tumor protein (TCTP) results, among others, in the inhibition of the former's pump activity and in the initiation of manifold biological and pathological phenomena. These phenomena include hypertension and cataract development in TCTP-overexpressing transgenic mice, as well as the induction of tumorigenesis signaling pathways and the activation of Src that ultimately leads to cell proliferation and migration. This review attempts to collate the biological effects of Na,K-ATPase and TCTP interaction and suggests that this interaction has the potential to serve as a possible therapeutic target for selected diseases.
Collapse
|
56
|
Kawasaki Y, Saito M, Won J, Bae JY, Sato H, Toyoda H, Kuramoto E, Kogo M, Tanaka T, Kaneko T, Oh SB, Bae YC, Kang Y. Inhibition of GluR Current in Microvilli of Sensory Neurons via Na +-Microdomain Coupling Among GluR, HCN Channel, and Na +/K + Pump. Front Cell Neurosci 2018; 12:113. [PMID: 29740287 PMCID: PMC5928758 DOI: 10.3389/fncel.2018.00113] [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: 10/25/2017] [Accepted: 04/06/2018] [Indexed: 11/13/2022] Open
Abstract
Glutamatergic dendritic EPSPs evoked in cortical pyramidal neurons are depressed by activation of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels expressed in dendritic spines. This depression has been attributed to shunting effects of HCN current (Ih) on input resistance or Ih deactivation. Primary sensory neurons in the rat mesencephalic trigeminal nucleus (MTN) have the somata covered by spine-like microvilli that express HCN channels. In rat MTN neurons, we demonstrated that Ih enhancement apparently diminished the glutamate receptor (GluR) current (IGluR) evoked by puff application of glutamate/AMPA and enhanced a transient outward current following IGluR (OT-IGluR). This suggests that some outward current opposes inward IGluR. The IGluR inhibition displayed a U-shaped voltage-dependence with a minimal inhibition around the resting membrane potential, suggesting that simple shunting effects or deactivation of Ih cannot explain the U-shaped voltage-dependence. Confocal imaging of Na+ revealed that GluR activation caused an accumulation of Na+ in the microvilli, which can cause a negative shift of the reversal potential for Ih (Eh). Taken together, it was suggested that IGluR evoked in MTN neurons is opposed by a transient decrease or increase in standing inward or outward Ih, respectively, both of which can be caused by negative shifts of Eh, as consistent with the U-shaped voltage-dependence of the IGluR inhibition and the OT-IGluR generation. An electron-microscopic immunohistochemical study revealed the colocalization of HCN channels and glutamatergic synapses in microvilli of MTN neurons, which would provide a morphological basis for the functional interaction between HCN and GluR channels. Mathematical modeling eliminated the possibilities of the involvements of Ih deactivation and/or shunting effect and supported the negative shift of Eh which causes the U-shaped voltage-dependent inhibition of IGluR.
Collapse
Affiliation(s)
- Yasuhiro Kawasaki
- Department of Neuroscience and Oral Physiology, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Mitsuru Saito
- Department of Neuroscience and Oral Physiology, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Jonghwa Won
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Dental Research Institute and Department of Neurobiology and Physiology, School of Dentistry, Seoul National University, Seoul, South Korea
| | - Jin Young Bae
- Department of Oral Anatomy, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Hajime Sato
- Department of Neuroscience and Oral Physiology, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Hiroki Toyoda
- Department of Neuroscience and Oral Physiology, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Eriko Kuramoto
- Department of Morphological Brain Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Mikihiko Kogo
- Department of Neuroscience and Oral Physiology, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Takuma Tanaka
- Department of Computational Intelligence and Systems Science, Interdisciplinary Graduate School of Science and Engineering, Tokyo Institute of Technology, Yokohama, Japan
| | - Takeshi Kaneko
- Department of Morphological Brain Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Seog Bae Oh
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Dental Research Institute and Department of Neurobiology and Physiology, School of Dentistry, Seoul National University, Seoul, South Korea
| | - Yong Chul Bae
- Department of Oral Anatomy, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Youngnam Kang
- Department of Neuroscience and Oral Physiology, Graduate School of Dentistry, Osaka University, Osaka, Japan
| |
Collapse
|
57
|
Isobe-Sasaki Y, Fukuda M, Ogiyama Y, Sato R, Miura T, Fuwa D, Mizuno M, Matsuoka T, Shibata H, Ito H, Ono M, Abe-Dohmae S, Kiyono K, Yamamoto Y, Kobori H, Michikawa M, Hayano J, Ohte N. Sodium balance, circadian BP rhythm, heart rate variability, and intrarenal renin-angiotensin-aldosterone and dopaminergic systems in acute phase of ARB therapy. Physiol Rep 2018; 5:5/11/e13309. [PMID: 28576855 PMCID: PMC5471446 DOI: 10.14814/phy2.13309] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 04/29/2017] [Accepted: 05/10/2017] [Indexed: 01/11/2023] Open
Abstract
We have revealed that even in humans, activated intrarenal renin–angiotensin–aldosterone system (RAAS) enhances tubular sodium reabsorption to facilitate salt sensitivity and nondipper rhythm of blood pressure (BP), and that angiotensin receptor blocker (ARB) could increase daytime urinary sodium excretion rate (UNaV) to produce lower sodium balance and restore nondipper rhythm. However, the sympathetic nervous system and intrarenal dopaminergic system can also contribute to renal sodium handling. A total of 20 patients with chronic kidney disease (61 ± 15 years) underwent 24‐h ambulatory BP monitoring before and during two‐day treatment with ARB, azilsartan. Urinary angiotensinogen excretion rate (UAGTV, μg/gCre) was measured as intrarenal RAAS; urinary dopamine excretion rate (UDAV, pg/gCre) as intrarenal dopaminergic system; heart rate variabilities (HRV, calculated from 24‐h Holter‐ECG) of non‐Gaussianity index λ25s as sympathetic nerve activity; and power of high‐frequency (HF) component or deceleration capacity (DC) as parasympathetic nerve activity. At baseline, glomerular filtration rate correlated inversely with UAGTV (r = −0.47, P = 0.04) and positively with UDAV (r = 0.58, P = 0.009). HF was a determinant of night/day BP ratio (β = −0.50, F = 5.8), rather than DC or λ25s. During the acute phase of ARB treatment, a lower steady sodium balance was not achieved. Increase in daytime UNaV preceded restoration of BP rhythm, accompanied by decreased UAGTV (r = −0.88, P = 0.05) and increased UDAV (r = 0.87, P = 0.05), but with no changes in HRVs. Diminished sodium excretion can cause nondipper BP rhythm. This was attributable to intrarenal RAAS and dopaminergic system and impaired parasympathetic nerve activity. During the acute phase of ARB treatment, cooperative effects of ARB and intrarenal dopaminergic system exert natriuresis to restore circadian BP rhythm.
Collapse
Affiliation(s)
- Yukako Isobe-Sasaki
- Department of Cardio-Renal Medicine and Hypertension, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Michio Fukuda
- Department of Cardio-Renal Medicine and Hypertension, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yoshiaki Ogiyama
- Department of Cardio-Renal Medicine and Hypertension, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Ryo Sato
- Department of Cardio-Renal Medicine and Hypertension, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Toshiyuki Miura
- Department of Cardio-Renal Medicine and Hypertension, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Daisuke Fuwa
- Department of Cardio-Renal Medicine and Hypertension, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Masashi Mizuno
- Department of Cardio-Renal Medicine and Hypertension, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Tetsuhei Matsuoka
- Department of Cardio-Renal Medicine and Hypertension, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hiroko Shibata
- Department of Cardio-Renal Medicine and Hypertension, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hiroyuki Ito
- Department of Cardio-Renal Medicine and Hypertension, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Minamo Ono
- Department of Cardio-Renal Medicine and Hypertension, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Sumiko Abe-Dohmae
- Department of Biochemistry, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Ken Kiyono
- Department of Mechanical Science and Bioengineering, Osaka University, Osaka, Japan
| | - Yoshiharu Yamamoto
- Department of Physical and Health Education, University of Tokyo Graduate School of Education, Tokyo, Japan
| | - Hiroyuki Kobori
- International University of Health and Welfare, Tokyo, Japan
| | - Makoto Michikawa
- Department of Biochemistry, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Junichiro Hayano
- Department of Medical Education, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Nobuyuki Ohte
- Department of Cardio-Renal Medicine and Hypertension, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
58
|
Xiao J, Zhang X, Fu C, Yang Q, Xie Y, Zhang Z, Ye Z. Impaired Na +-K +-ATPase signaling in renal proximal tubule contributes to hyperuricemia-induced renal tubular injury. Exp Mol Med 2018; 50:e452. [PMID: 29497172 PMCID: PMC5898891 DOI: 10.1038/emm.2017.287] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Revised: 09/11/2017] [Accepted: 09/26/2017] [Indexed: 12/27/2022] Open
Abstract
Hyperuricemia contributes to renal inflammation. We aimed to investigate the role of Na+–K+–ATPase (NKA) in hyperuricemia-induced renal tubular injury. Human primary proximal tubular epithelial cells (PTECs) were incubated with uric acid (UA) at increasing doses or for increasing lengths of time. PTECs were then stimulated by pre-incubation with an NKA α1 expression vector or small interfering RNA before UA (100 μg ml−1, 48 h) stimulation. Hyperuricemic rats were induced by gastric oxonic acid and treated with febuxostat (Feb). ATP levels, the activity of NKA and expression of its α1 subunit, Src, NOD-like receptor pyrin domain-containing protein 3 (NLRP3) and interleukin 1β (IL-1β) were measured both in vitro and in vivo. Beginning at concentrations of 100 μg ml−1, UA started to dose-dependently reduce NKA activity. UA at a concentration of 100 μg ml−1 time-dependently affected the NKA activity, with the maximal increased NKA activity at 24 h, but the activity started to decrease after 48 h. This inhibitory effect of UA on NKA activity at 48 h was in addition to a decrease in NKA α1 expression in the cell membrane, but an increase in lysosomes. This process also involved the subsequent activation of Src kinase and NLRP3, promoting IL-1β processing. In hyperuricemic rats, renal cortex NKA activity and its α1 expression were upregulated at the 7th week and both decreased at the 10th week, accompanied with increased renal cortex expression of Src, NLRP3 and IL-1β. The UA levels were reduced and renal tubular injuries in hyperuricemic rats were alleviated in the Feb group. Our data suggested that the impairment of NKA and its consequent regulation of Src, NLRP3 and IL-1β in the renal proximal tubule contributed to hyperuricemia-induced renal tubular injury.
Collapse
Affiliation(s)
- Jing Xiao
- Department of Nephrology, Huadong Hospital affiliated with Fudan University, Shanghai, China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong hospital affiliated with Fudan University, Shanghai, China
| | - Xiaoli Zhang
- Department of Nephrology, Huadong Hospital affiliated with Fudan University, Shanghai, China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong hospital affiliated with Fudan University, Shanghai, China
| | - Chensheng Fu
- Department of Nephrology, Huadong Hospital affiliated with Fudan University, Shanghai, China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong hospital affiliated with Fudan University, Shanghai, China
| | - Qingmei Yang
- Department of Nephrology, Huadong Hospital affiliated with Fudan University, Shanghai, China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong hospital affiliated with Fudan University, Shanghai, China
| | - Ying Xie
- Department of Nephrology, Huadong Hospital affiliated with Fudan University, Shanghai, China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong hospital affiliated with Fudan University, Shanghai, China
| | - Zhenxing Zhang
- Department of Nephrology, Huadong Hospital affiliated with Fudan University, Shanghai, China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong hospital affiliated with Fudan University, Shanghai, China
| | - Zhibin Ye
- Department of Nephrology, Huadong Hospital affiliated with Fudan University, Shanghai, China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong hospital affiliated with Fudan University, Shanghai, China
| |
Collapse
|
59
|
Sampaio LS, da Silva PA, Ribeiro VS, Castro-Chaves C, Lara LS, Vieyra A, Einicker-Lamas M. Bioactive lipids are altered in the kidney of chronic undernourished rats: is there any correlation with the progression of prevalent nephropathies? Lipids Health Dis 2017; 16:245. [PMID: 29246161 PMCID: PMC5732436 DOI: 10.1186/s12944-017-0634-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 12/03/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Undernutrition during childhood leads to chronic diseases in adult life including hypertension, diabetes and chronic kidney disease. Here we explore the hypothesis that physiological alterations in the bioactive lipids pattern within kidney tissue might be involved in the progression of chronic kidney disease. METHODS Membrane fractions from kidney homogenates of undernourished rats (RBD) were submitted to lipid extraction and analysis by thin layer chromatography and cholesterol determination. RESULTS Kidneys from RBD rats had 25% lower cholesterol content, which disturb membrane microdomains, affecting Ca2+ homeostasis and the enzymes responsible for important lipid mediators such as phosphatidylinositol-4 kinase, sphingosine kinase, diacylglicerol kinase and phospholipase A2. We observed a decrease in phosphatidylinositol(4)-phosphate (8.8 ± 0.9 vs. 3.6 ± 0.7 pmol.mg-1.mim-1), and an increase in phosphatidic acid (2.2 ± 0.8 vs. 3.8 ± 1.3 pmol.mg-1.mim-1), being these lipid mediators involved in the regulation of key renal functions. Ceramide levels are augmented in kidney tissue from RBD rats (18.7 ± 1.4 vs. 21.7 ± 1.5 fmol.mg-1.min-1) indicating an ongoing renal lesion. CONCLUSION Results point to an imbalance in the bioactive lipid generation with further consequences to key events related to kidney function, thus contributing to the establishment of chronic kidney disease.
Collapse
Affiliation(s)
- Luzia S Sampaio
- Instituto de Biofísica Carlos Chagas Filho - CCS, UFRJ, Rio de Janeiro, RJ, Brazil
| | - Paulo A da Silva
- Instituto de Biofísica Carlos Chagas Filho - CCS, UFRJ, Rio de Janeiro, RJ, Brazil
- Programa de Pós-Graduação em Biomedicina Translacional, Universidade do Grande Rio, Duque de Caxias, RJ, Brazil
| | | | | | - Lucienne S Lara
- Instituto de Ciências Biomédicas, UFRJ, Rio de Janeiro, RJ, Brazil
| | - Adalberto Vieyra
- Instituto de Biofísica Carlos Chagas Filho - CCS, UFRJ, Rio de Janeiro, RJ, Brazil
- Programa de Pós-Graduação em Biomedicina Translacional, Universidade do Grande Rio, Duque de Caxias, RJ, Brazil
- Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), UFRJ, Rio de Janeiro, Brazil
| | - M Einicker-Lamas
- Instituto de Biofísica Carlos Chagas Filho - CCS, UFRJ, Rio de Janeiro, RJ, Brazil.
- Present Address: Laboratório de Biomembranas, Sala G1-037, Bloco G, Instituto de Biofísica Carlos Chagas Filho - CCS, UFRJ, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.
| |
Collapse
|
60
|
Udwan K, Abed A, Roth I, Dizin E, Maillard M, Bettoni C, Loffing J, Wagner CA, Edwards A, Feraille E. Dietary sodium induces a redistribution of the tubular metabolic workload. J Physiol 2017; 595:6905-6922. [PMID: 28940314 PMCID: PMC5685825 DOI: 10.1113/jp274927] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 09/11/2017] [Indexed: 01/11/2023] Open
Abstract
Key points Body Na+ content is tightly controlled by regulated urinary Na+ excretion. The intrarenal mechanisms mediating adaptation to variations in dietary Na+ intake are incompletely characterized. We confirmed and expanded observations in mice that variations in dietary Na+ intake do not alter the glomerular filtration rate but alter the total and cell‐surface expression of major Na+ transporters all along the kidney tubule. Low dietary Na+ intake increased Na+ reabsorption in the proximal tubule and decreased it in more distal kidney tubule segments. High dietary Na+ intake decreased Na+ reabsorption in the proximal tubule and increased it in distal segments with lower energetic efficiency. The abundance of apical transporters and Na+ delivery are the main determinants of Na+ reabsorption along the kidney tubule. Tubular O2 consumption and the efficiency of sodium reabsorption are dependent on sodium diet.
Abstract Na+ excretion by the kidney varies according to dietary Na+ intake. We undertook a systematic study of the effects of dietary salt intake on glomerular filtration rate (GFR) and tubular Na+ reabsorption. We examined the renal adaptive response in mice subjected to 7 days of a low sodium diet (LSD) containing 0.01% Na+, a normal sodium diet (NSD) containing 0.18% Na+ and a moderately high sodium diet (HSD) containing 1.25% Na+. As expected, LSD did not alter measured GFR and increased the abundance of total and cell‐surface NHE3, NKCC2, NCC, α‐ENaC and cleaved γ‐ENaC compared to NSD. Mathematical modelling predicted that tubular Na+ reabsorption increased in the proximal tubule but decreased in the distal nephron because of diminished Na+ delivery. This prediction was confirmed by the natriuretic response to diuretics targeting the thick ascending limb, the distal convoluted tubule or the collecting system. On the other hand, HSD did not alter measured GFR but decreased the abundance of the aforementioned transporters compared to NSD. Mathematical modelling predicted that tubular Na+ reabsorption decreased in the proximal tubule but increased in distal segments with lower transport efficiency with respect to O2 consumption. This prediction was confirmed by the natriuretic response to diuretics. The activity of the metabolic sensor adenosine monophosphate‐activated protein kinase (AMPK) was related to the changes in tubular Na+ reabsorption. Our data show that fractional Na+ reabsorption is distributed differently according to dietary Na+ intake and induces changes in tubular O2 consumption and sodium transport efficiency. Body Na+ content is tightly controlled by regulated urinary Na+ excretion. The intrarenal mechanisms mediating adaptation to variations in dietary Na+ intake are incompletely characterized. We confirmed and expanded observations in mice that variations in dietary Na+ intake do not alter the glomerular filtration rate but alter the total and cell‐surface expression of major Na+ transporters all along the kidney tubule. Low dietary Na+ intake increased Na+ reabsorption in the proximal tubule and decreased it in more distal kidney tubule segments. High dietary Na+ intake decreased Na+ reabsorption in the proximal tubule and increased it in distal segments with lower energetic efficiency. The abundance of apical transporters and Na+ delivery are the main determinants of Na+ reabsorption along the kidney tubule. Tubular O2 consumption and the efficiency of sodium reabsorption are dependent on sodium diet.
Collapse
Affiliation(s)
- Khalil Udwan
- Department of Cellular Physiology and Metabolism, University of Geneva, CMU, 1 Rue Michel-Servet, CH-1211, Geneva 4, Switzerland.,National Centre of Competence in Research, NCCRKidney, CH, Switzerland
| | - Ahmed Abed
- Department of Cellular Physiology and Metabolism, University of Geneva, CMU, 1 Rue Michel-Servet, CH-1211, Geneva 4, Switzerland.,National Centre of Competence in Research, NCCRKidney, CH, Switzerland
| | - Isabelle Roth
- Department of Cellular Physiology and Metabolism, University of Geneva, CMU, 1 Rue Michel-Servet, CH-1211, Geneva 4, Switzerland
| | - Eva Dizin
- Department of Cellular Physiology and Metabolism, University of Geneva, CMU, 1 Rue Michel-Servet, CH-1211, Geneva 4, Switzerland.,National Centre of Competence in Research, NCCRKidney, CH, Switzerland
| | - Marc Maillard
- Centre hospitalier universitaire Vaudois, Service de néphrologie, CH-1011, Lausanne, Switzerland
| | - Carla Bettoni
- Institute of Physiology, University of Zürich, Winterthurerstrasse 190, CH-8057, Zürich, Switzerland
| | - Johannes Loffing
- Institute of Anatomy, University of Zürich, Winterthurerstrasse 190, CH-8057, Zürich, Switzerland.,National Centre of Competence in Research, NCCRKidney, CH, Switzerland
| | - Carsten A Wagner
- Institute of Physiology, University of Zürich, Winterthurerstrasse 190, CH-8057, Zürich, Switzerland.,National Centre of Competence in Research, NCCRKidney, CH, Switzerland
| | - Aurélie Edwards
- Centre de Recherche des Cordeliers, INSERM UMRS1138 and CNRS ERL8228, 15 rue de l'Ecole de Médecine, F-75006, Paris, France.,Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Eric Feraille
- Department of Cellular Physiology and Metabolism, University of Geneva, CMU, 1 Rue Michel-Servet, CH-1211, Geneva 4, Switzerland.,National Centre of Competence in Research, NCCRKidney, CH, Switzerland
| |
Collapse
|
61
|
Mun EG, Sohn HS, Kim MS, Cha YS. Antihypertensive effect of Ganjang (traditional Korean soy sauce) on Sprague-Dawley Rats. Nutr Res Pract 2017; 11:388-395. [PMID: 28989575 PMCID: PMC5621361 DOI: 10.4162/nrp.2017.11.5.388] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 05/17/2017] [Accepted: 09/15/2017] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND/OBJECTIVES Although Korean fermented foods contain large amounts of salt, which is known to exacerbate health problems, these foods still have beneficial effects such as anti-hypertension, anti-cancer, and anti-colitis properties. We hypothesized that ganjang may have different effects on blood pressure compared to same concentrations of salt. MATERIALS/METHODS Sprague-Dawley rats were divided into control (CT), NaCl (NC), and ganjang (GJ) groups and orally administered with 8% NaCl concentration for 9 weeks. The systolic blood pressure (SBP), serum chemistry, Na+ and K+ concentrations and renal gene expressions were measured. RESULTS The SBP was significantly increased in the NC group compared to the GJ and CT groups. In addition, the Na+ concentration in urine was higher in the GJ and NC groups than the CT group, but the urine volume was increased in the GJ group compared to the other groups. The serum renin levels were decreased in the GJ group compared to the CT group, while the serum aldosterone level was decreased in the GJ group relative to the NC group. The mRNA expression of the renin, angiotensin II type I receptor, and mineralocorticoid receptor were significantly lower in the GJ group compared to other groups. Furthermore, GJ group showed the lowest levels of genes for Na+ transporter in kidney cortex such as Na+/K+ ATPaseα1 (NKAα1), Na+/H+ exchanger 3 (NHE3), Na+/HCO3- co-exchanger (NBC), and carbonic anhydrases II (CAII). CONCLUSIONS The decreased SBP in the GJ could be due to decreased renin and aldosterone levels in serum and increased urinary volume and excretion of Na+ with its transporter gene alteration. Therefore, ganjang may have antihypertensive effect despite its high contents of salt.
Collapse
Affiliation(s)
- Eun-Gyung Mun
- Department of Food Science and Human Nutrition, Chonbuk National University, 567, Baekje-daero, Duckjin-gu, Jeonju, Jeonbuk 54896, Korea
| | - Hee-Sook Sohn
- Department of Food Science and Human Nutrition, Chonbuk National University, 567, Baekje-daero, Duckjin-gu, Jeonju, Jeonbuk 54896, Korea
| | - Mi-Sun Kim
- Department of Food Science and Human Nutrition, Chonbuk National University, 567, Baekje-daero, Duckjin-gu, Jeonju, Jeonbuk 54896, Korea
| | - Youn-Soo Cha
- Department of Food Science and Human Nutrition, Chonbuk National University, 567, Baekje-daero, Duckjin-gu, Jeonju, Jeonbuk 54896, Korea
| |
Collapse
|
62
|
Lestari SW, Miati DN, Seoharso P, Sugiyanto R, Pujianto DA. Sperm Na+, K+-ATPase α4 and plasma membrane Ca2+-ATPase (PMCA) 4 regulation in asthenozoospermia. Syst Biol Reprod Med 2017; 63:294-302. [DOI: 10.1080/19396368.2017.1348565] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Silvia W. Lestari
- Department of Medical Biology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Dessy Noor Miati
- Master Program for Biomedical Sciences, Faculty of Medicine, Universitas Indonesi, Jakara, Indonesia
| | - P. Seoharso
- Department of Medical Biology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - R. Sugiyanto
- Department of Medical Biochemistry, Faculty of Medicine, Universitas Kristen Indonesia, Jakarta, Indonesia
| | - Dwi A. Pujianto
- Department of Medical Biology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| |
Collapse
|
63
|
Hu MC, Bobulescu IA, Quiñones H, Gisler SM, Moe OW. Dopamine reduces cell surface Na +/H + exchanger-3 protein by decreasing NHE3 exocytosis and cell membrane recycling. Am J Physiol Renal Physiol 2017; 313:F1018-F1025. [PMID: 28768665 DOI: 10.1152/ajprenal.00251.2017] [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] [Received: 05/12/2017] [Revised: 07/14/2017] [Accepted: 07/24/2017] [Indexed: 01/06/2023] Open
Abstract
The intrarenal autocrine-paracrine dopamine (DA) system mediates a significant fraction of the natriuresis in response to a salt load. DA inhibits a number of Na+ transporters to effect sodium excretion, including the proximal tubule Na+/H+ exchanger-3 (NHE3). DA represent a single hormone that regulates NHE3 at multiple levels, including translation, degradation, endocytosis, and protein phosphorylation. Because cell surface NHE3 protein is determined by the balance between exocytotic insertion and endocytotic retrieval, we examined whether DA acutely affects the rate of NHE3 exocytosis in a cell culture model. DA inhibited NHE3 exocytosis at a dose-dependent manner with a half maximal around 10-6 M. The DA effect on NHE3 exocytosis was blocked by inhibition of protein kinase A and by brefeldin A, which inhibits endoplasmic reticulum-to-Golgi transport. NHE3 directly interacts with the ε-subunit of coatomer protein based on yeast-two-hybrid and coimmunoprecipitation. Because NHE3 has been shown to be recycled back to the cell membrane after endocytosis, we measured NHE3 recycling using a biochemical reinsertion assay and showed that reinsertion of NHE3 back to the membrane is also inhibited by DA. In conclusion, among the many mechanisms by which DA reduces apical membrane NHE3 and induces proximal tubule natriuresis, one additional mechanism is inhibition of exocytotic insertion and reinsertion of NHE3 in the apical cell surface.
Collapse
Affiliation(s)
- Ming Chang Hu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas; .,Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - I Alexandru Bobulescu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas.,Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Henry Quiñones
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Serge M Gisler
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Orson W Moe
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas.,Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas; and.,Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
64
|
Takahashi-Iwanaga H, Kimura S, Konno K, Watanabe M, Iwanaga T. Intrarenal signaling mediated by CCK plays a role in salt intake-induced natriuresis. Am J Physiol Renal Physiol 2017; 313:F20-F29. [DOI: 10.1152/ajprenal.00539.2016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 03/07/2017] [Accepted: 03/10/2017] [Indexed: 11/22/2022] Open
Abstract
The natriuretic hormone CCK exhibits its gene transcripts in total kidney extracts. To test the possibility of CCK acting as an intrarenal mediator of sodium excretion, we examined mouse kidneys by 1) an in situ hybridization technique for CCK mRNA in animals fed a normal- or a high-sodium diet; 2) immuno-electron microscopy for the CCK peptide, 3) an in situ hybridization method and immunohistochemistry for the CCK-specific receptor CCKAR; 4) confocal image analysis of receptor-mediated Ca2+ responses in isolated renal tubules; and 5) metabolic cage experiments for the measurement of urinary sodium excretion in high-salt-fed mice either treated or untreated with the CCKAR antagonist lorglumide. Results showed the CCK gene to be expressed intensely in the inner medulla and moderately in the inner stripe of the outer medulla, with the expression in the latter being enhanced by high sodium intake. Immunoreactivity for the CCK peptide was localized to the rough endoplasmic reticulum of the medullary interstitial cells in corresponding renal regions, confirming it to be a secretory protein. Gene transcripts, protein products, and the functional activity for CCKAR were consistently localized to the late proximal tubule segments (S2 and S3) in the medullary rays, and the outer stripe of the outer medulla. Lorglumide significantly diminished natriuretic responses of mice to a dietary sodium load without altering the glomerular filtration rate. These findings suggest that the medullary interstitial cells respond to body fluid expansion by CCK release for feedback regulation of the late proximal tubular reabsorption.
Collapse
Affiliation(s)
| | - Shunsuke Kimura
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Kohtarou Konno
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Toshihiko Iwanaga
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
65
|
Abramicheva PA, Balakina TA, Bulaeva OA, Guseva AA, Lopina OD, Smirnova OV. Role of Na+/K +-ATPase in Natriuretic Effect of Prolactin in a Model of Cholestasis of Pregnancy. BIOCHEMISTRY (MOSCOW) 2017; 82:632-641. [PMID: 28601073 DOI: 10.1134/s000629791705011x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Participation of Na+/K+-ATPase in the natriuretic effect of prolactin in a cholestasis of pregnancy model was investigated. The Na+/K+-ATPase activity in rat kidney medulla, where active sodium reabsorption occurs, decreased in the model of cholestasis of pregnancy and other hyperprolactinemia types compared with intact animals. This effect was not connected with the protein level of α1- and β-subunits of Na+/K+-ATPase measured by Western blotting in the kidney medulla. Decrease in Na+/K+-ATPase activity in the kidney cortex was not significant, as well as decrease in the quantity of mRNA and proteins of the α1- and β-subunits of Na+/K+-ATPase. There were no correlations between the Na+/K+-ATPase activity and sodium clearance, although sodium clearance increased significantly in the model of cholestasis of pregnancy and other hyperprolactinemia groups under conditions of stable glomerular filtration rate measured by creatinine clearance. We conclude that the Na+/K+-ATPase is not the only mediator of the natriuretic effect of prolactin in the model of cholestasis of pregnancy.
Collapse
Affiliation(s)
- P A Abramicheva
- Lomonosov Moscow State University, Faculty of Biology, Moscow, 119991, Russia.
| | | | | | | | | | | |
Collapse
|
66
|
Graham LA, Dominiczak AF, Ferreri NR. Role of renal transporters and novel regulatory interactions in the TAL that control blood pressure. Physiol Genomics 2017; 49:261-276. [PMID: 28389525 PMCID: PMC5451551 DOI: 10.1152/physiolgenomics.00017.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 03/27/2017] [Accepted: 04/05/2017] [Indexed: 12/31/2022] Open
Abstract
Hypertension (HTN), a major public health issue is currently the leading factor in the global burden of disease, where associated complications account for 9.4 million deaths worldwide every year. Excessive dietary salt intake is among the environmental factors that contribute to HTN, known as salt sensitivity. The heterogeneity of salt sensitivity and the multiple mechanisms that link high salt intake to increases in blood pressure are of upmost importance for therapeutic application. A continual increase in the kidney's reabsorption of sodium (Na+) relies on sequential actions at various segments along the nephron. When the distal segments of the nephron fail to regulate Na+, the effects on Na+ homeostasis are unfavorable. We propose that the specific nephron region where increased active uptake occurs as a result of variations in Na+ reabsorption is at the thick ascending limb of the loop of Henle (TAL). The purpose of this review is to urge the consideration of the TAL as contributing to the pathophysiology of salt-sensitive HTN. Further research in this area will enable development of a therapeutic application for targeted treatment.
Collapse
Affiliation(s)
- Lesley A Graham
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow Cardiovascular and Medical Sciences, Glasgow, United Kingdom; and
| | - Anna F Dominiczak
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow Cardiovascular and Medical Sciences, Glasgow, United Kingdom; and
| | - Nicholas R Ferreri
- Department of Pharmacology, New York Medical College, Valhalla, New York
| |
Collapse
|
67
|
Klein K, Rübenacker S, Schaefer SM, Kihm LP, Schwenger V, Macher-Goeppinger S, Schemmer P, Süsald C, Zeier M, Morath C, Becker LE. Tissue Expression of Aquaporin 2 Is Correlated to Urine Output and Allograft Function in Sensitized Kidney Transplant Patients. Transplant Proc 2017; 48:2629-2636. [PMID: 27788793 DOI: 10.1016/j.transproceed.2016.06.056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 05/05/2016] [Accepted: 06/06/2016] [Indexed: 01/23/2023]
Abstract
BACKGROUND Salt and water disturbances often occur during acute kidney allograft dysfunction that contribute to graft failure, but this condition has been poorly investigated in the alloreactivity setting. We evaluated the tissue expression of aquaporins (AQP1 and AQP2) and the epithelial sodium channel (ENAC) in kidney biopsy specimens from sensitized kidney transplant recipients. METHODS Eighty-six biopsy specimens from 33 sensitized patients were divided into 3 groups according to clinical context: time-zero (n = 9), protocol (n = 9), and indication (n = 68). The indication biopsy specimens were further divided into 3 subgroups according to the presence of acute tubular necrosis or rejection. Normal kidney tissue samples (n = 6) served as the control specimens. Immmunohistochemical expression of AQP1, AQP2, and ENAC was determined by using image analyzing software. RESULTS Significantly lower AQP1 expression was observed in the time-zero and indication biopsy specimens with rejection compared with control specimens (P = .03 and P = .04, respectively). AQP2 expression was significantly lower in patients with an indication biopsy specimen compared with control and protocol biopsy specimens (P = .05 and P = .005). For ENAC, a lower expression was noted in the indication biopsy specimens compared with the control specimens (P = .04). Both AQP1 and AQP2 tissue expressions were significantly correlated to urine output (r = 0.45 and r = 0.32; P = .001 and P = .02), and AQP2 was correlated with the glomerular filtration rate estimated by using the Modification of Diet in Renal Disease Study equation at biopsy (r = 0.23; P = .05). CONCLUSIONS These findings partially confirm previous experimental data showing downregulation of AQP1 expression after ischemia/reperfusion injury and during rejection. AQP2 downregulation seems to be rejection-independent, occurring during deteriorating or poor kidney graft function.
Collapse
Affiliation(s)
- K Klein
- Division of Nephrology, University of Heidelberg, Heidelberg, Germany
| | - S Rübenacker
- Division of Nephrology, University of Heidelberg, Heidelberg, Germany
| | - S M Schaefer
- Division of Nephrology, University of Heidelberg, Heidelberg, Germany
| | - L P Kihm
- Division of Nephrology, University of Heidelberg, Heidelberg, Germany
| | - V Schwenger
- Division of Nephrology, University of Heidelberg, Heidelberg, Germany
| | | | - P Schemmer
- Department of Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - C Süsald
- Department of Transplant Immunology, University of Heidelberg, Heidelberg, Germany
| | - M Zeier
- Division of Nephrology, University of Heidelberg, Heidelberg, Germany
| | - C Morath
- Division of Nephrology, University of Heidelberg, Heidelberg, Germany
| | - L E Becker
- Division of Nephrology, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
68
|
Dias J, Axelband F, Lara LS, Muzi-Filho H, Vieyra A. Is angiotensin-(3-4) (Val-Tyr), the shortest angiotensin II-derived peptide, opening new vistas on the renin-angiotensin system? J Renin Angiotensin Aldosterone Syst 2017; 18:1470320316689338. [PMID: 28097883 PMCID: PMC5843854 DOI: 10.1177/1470320316689338] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Angiotensin-(3−4) (Ang-(3−4) or Val-Tyr) is the shorter angiotensin (Ang) II-derived peptide, formed through successive hydrolysis that culminates with the release of Val-Tyr as a dipeptide. It is formed both in plasma and in kidney from Ang II and Ang III, and can be considered a component of the systemic and organ-based renin–angiotensin system. It is potently antihypertensive in humans and rats, and its concerted actions on proximal tubule cells culminate in the inhibition of fluid reabsorption, hyperosmotic urinary excretion of Na+. At the renal cell signaling level, Ang-(3−4) counteracts Ang II-type 1 receptor-mediated responses by acting as an allosteric enhancer in Ang II-type 2 receptor populations that target adenosine triphosphate-dependent Ca2+ and Na+ transporters through a cyclic adenosine monophosphate-activated protein kinase pathway.
Collapse
Affiliation(s)
- Juliana Dias
- 1 National Institute of Cancer, Rio de Janeiro, Brazil.,2 Carlos Chagas Institute of Biophysics, Federal University of Rio de Janeiro, Brazil.,3 National Center of Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Brazil
| | | | - Lucienne S Lara
- 3 National Center of Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Brazil.,4 Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Brazil
| | - Humberto Muzi-Filho
- 2 Carlos Chagas Institute of Biophysics, Federal University of Rio de Janeiro, Brazil.,3 National Center of Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Brazil
| | - Adalberto Vieyra
- 2 Carlos Chagas Institute of Biophysics, Federal University of Rio de Janeiro, Brazil.,3 National Center of Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Brazil.,5 Translational Biomedicine Graduate Program, Grande Rio University, Brazil
| |
Collapse
|
69
|
Liu J, Yan Y, Nie Y, Shapiro JI. Na/K-ATPase Signaling and Salt Sensitivity: The Role of Oxidative Stress. Antioxidants (Basel) 2017; 6:E18. [PMID: 28257114 PMCID: PMC5384181 DOI: 10.3390/antiox6010018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 02/10/2017] [Accepted: 02/22/2017] [Indexed: 02/07/2023] Open
Abstract
Other than genetic regulation of salt sensitivity of blood pressure, many factors have been shown to regulate renal sodium handling which contributes to long-term blood pressure regulation and have been extensively reviewed. Here we present our progress on the Na/K-ATPase signaling mediated sodium reabsorption in renal proximal tubules, from cardiotonic steroids-mediated to reactive oxygen species (ROS)-mediated Na/K-ATPase signaling that contributes to experimental salt sensitivity.
Collapse
Affiliation(s)
- Jiang Liu
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA.
| | - Yanling Yan
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA.
| | - Ying Nie
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA.
| | - Joseph I Shapiro
- Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
| |
Collapse
|
70
|
Tang L, Zheng S, Ren H, He D, Zeng C, Wang WE. Activation of angiotensin II type 1 receptors increases D 4 dopamine receptor expression in rat renal proximal tubule cells. Hypertens Res 2017; 40:652-657. [PMID: 28230199 DOI: 10.1038/hr.2017.13] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 12/27/2016] [Accepted: 12/28/2016] [Indexed: 12/22/2022]
Abstract
Both the dopaminergic and renin-angiotensin systems play important roles in the regulation of blood pressure. Our previous study showed that the stimulation of dopaminergic D4 receptors reduced angiotensin II type 1 (AT1) receptor expression in renal proximal tubule (RPT) cells. In this study, we tested whether AT1 receptors, in return, would regulate D4 receptor expression and function in RPT cells. Expression of the D4 receptor from Wistar-Kyoto (WKY) or spontaneously hypertensive rats (SHRs) RPT cells and renal cortex tissues were determined by western blot, and Na+-K+ ATPase activity was determined using an enzyme assay. Urine volume and urine sodium of WKY rats and SHRs treated with or without D4 receptor stimulation were measured. Thus, activation of AT1 receptors with angiotensin II (Ang II) increased D4 receptor protein expression in RPT cells, and this increase was blocked by nicardipine, a calcium influx blocker. The D4 receptor agonist PD168077 inhibited Na+-K+ ATPase activity in WKY RPT cells but not in SHR RPT cells. Ang II pre-treatment promoted D4 receptor-mediated inhibition of Na+-K+ ATPase in RPT cells in WKY rats but not in SHRs. Meanwhile, Ang II pre-treatment augmented the natriuretic effect of PD168077 in WKY rats but not in SHRs. In conclusion, AT1 stimulation can regulate the expression and natriuretic function of dopaminergic D4 receptors in RPT cells and might be involved in the pathogenesis of essential hypertension.
Collapse
Affiliation(s)
- Luxun Tang
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Cardiovascular Clinical Research Center, Daping Hospital, Third Military Medical University, Chongqing, PR China
| | - Shuo Zheng
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Cardiovascular Clinical Research Center, Daping Hospital, Third Military Medical University, Chongqing, PR China
| | - Hongmei Ren
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Cardiovascular Clinical Research Center, Daping Hospital, Third Military Medical University, Chongqing, PR China
| | - Duofen He
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Cardiovascular Clinical Research Center, Daping Hospital, Third Military Medical University, Chongqing, PR China
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Cardiovascular Clinical Research Center, Daping Hospital, Third Military Medical University, Chongqing, PR China
| | - Wei Eric Wang
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Cardiovascular Clinical Research Center, Daping Hospital, Third Military Medical University, Chongqing, PR China
| |
Collapse
|
71
|
Polyamines regulate phosphorylation–dephosphorylation kinetics in a crustacean gill (Na+, K+)-ATPase. Mol Cell Biochem 2017; 429:187-198. [DOI: 10.1007/s11010-017-2946-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 01/17/2017] [Indexed: 12/15/2022]
|
72
|
Hosseinzadeh Z, Singh Y, Shimshek DR, van der Putten H, Wagner CA, Lang F. Leucine-Rich Repeat Kinase 2 (Lrrk2)-Sensitive Na +/K + ATPase Activity in Dendritic Cells. Sci Rep 2017; 7:41117. [PMID: 28120865 PMCID: PMC5264149 DOI: 10.1038/srep41117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 12/14/2016] [Indexed: 12/18/2022] Open
Abstract
Leucine-rich repeat kinase 2 (Lrrk2) has been implicated in the pathophysiology of Parkinson's disease. Lrrk2 is expressed in diverse cells including neurons and dendritic cells (DCs). In DCs Lrrk2 was shown to up-regulate Na+/Ca2+-exchanger activity. The elimination of Ca2+ by Na+/Ca2+ -exchangers requires maintenance of the Na+ gradient by the Na+/K+ -ATPase. The present study thus explored whether Lrrk2 impacts on Na+/K+ -ATPase expression and function. To this end DCs were isolated from gene-targeted mice lacking Lrrk2 (Lrrk2-/-) and their wild-type littermates (Lrrk2+/+). Na+/K+ -ATPase activity was estimated from K+ induced, ouabain sensitive, current determined by whole cell patch clamp. Na+/K+ -ATPase α1 subunit transcript and protein levels were determined by RT-qPCR and flow cytometry. As a result, the K+ induced current was significantly smaller in Lrrk2-/- than in Lrrk2+/+ DCs and was completely abolished by ouabain (100 μM) in both genotypes. The K+ induced, ouabain sensitive, current in Lrrk2+/+ DCs was significantly blunted by Lrrk2 inhibitor GSK2578215A (1 μM, 24 hours). The Na+/K+ -ATPase α1 subunit transcript and protein levels were significantly lower in Lrrk2-/- than in Lrrk2+/+ DCs and significantly decreased by Lrrk2 inhibitor GSK2578215A (1 μM, 24 hours). In conclusion, Lrrk2 is a powerful regulator of Na+/K+ -ATPase expression and activity in dendritic cells.
Collapse
Affiliation(s)
- Zohreh Hosseinzadeh
- Department of Cardiology, Vascular Medicine and Physiology, University of Tübingen, Gmelinstr. 5, D-72076 Tübingen, Germany
- Experimental Retinal Prosthetics Group, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Yogesh Singh
- Department of Cardiology, Vascular Medicine and Physiology, University of Tübingen, Gmelinstr. 5, D-72076 Tübingen, Germany
| | - Derya R. Shimshek
- Department of Neuroscience, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Herman van der Putten
- Department of Neuroscience, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
- National Contest for Life (NCL) Foundation, 203555 Hamburg, Germany
| | - Carsten A. Wagner
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Florian Lang
- Department of Cardiology, Vascular Medicine and Physiology, University of Tübingen, Gmelinstr. 5, D-72076 Tübingen, Germany
| |
Collapse
|
73
|
Ribeiro NE, Cabral EV, Aires RS, Vieira-Filho LD, Ribeiro VS, Gonçalves DRM, Borges LPNC, Melo IMF, Ferreira CGM, Wanderley-Teixeira V, Teixeira ÁAC, Soares AF, Paixão AD. Maternal Na+intake induces renal function injury in rats prevented by a short-term angiotensin converting enzyme inhibitor. Clin Exp Pharmacol Physiol 2017; 44:275-284. [DOI: 10.1111/1440-1681.12702] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 08/26/2016] [Accepted: 11/10/2016] [Indexed: 01/13/2023]
Affiliation(s)
- Natalie E Ribeiro
- Department of Morphology and Animal Physiology; Federal Rural University of Pernambuco; Recife Pernambuco Brazil
| | - Edjair V Cabral
- Department of Physiology and Pharmacology; Centre of Biological Sciences; Federal University of Pernambuco; Recife Pernambuco Brazil
| | - Regina S Aires
- Department of Physiology and Pharmacology; Centre of Biological Sciences; Federal University of Pernambuco; Recife Pernambuco Brazil
| | - Leucio D Vieira-Filho
- Department of Physiology and Pharmacology; Centre of Biological Sciences; Federal University of Pernambuco; Recife Pernambuco Brazil
| | - Valdilene S Ribeiro
- Department of Physiology and Pharmacology; Centre of Biological Sciences; Federal University of Pernambuco; Recife Pernambuco Brazil
| | - Daianna RM Gonçalves
- Department of Physiology and Pharmacology; Centre of Biological Sciences; Federal University of Pernambuco; Recife Pernambuco Brazil
| | - Luis PNC Borges
- Department of Physiology and Pharmacology; Centre of Biological Sciences; Federal University of Pernambuco; Recife Pernambuco Brazil
| | - Ismaela MF Melo
- Department of Morphology and Animal Physiology; Federal Rural University of Pernambuco; Recife Pernambuco Brazil
| | - Cintia GM Ferreira
- Department of Morphology and Animal Physiology; Federal Rural University of Pernambuco; Recife Pernambuco Brazil
| | - Valeria Wanderley-Teixeira
- Department of Morphology and Animal Physiology; Federal Rural University of Pernambuco; Recife Pernambuco Brazil
| | - Álvaro AC Teixeira
- Department of Morphology and Animal Physiology; Federal Rural University of Pernambuco; Recife Pernambuco Brazil
| | - Anísio F Soares
- Department of Morphology and Animal Physiology; Federal Rural University of Pernambuco; Recife Pernambuco Brazil
| | - Ana D Paixão
- Department of Physiology and Pharmacology; Centre of Biological Sciences; Federal University of Pernambuco; Recife Pernambuco Brazil
| |
Collapse
|
74
|
Ali El-Basyuni Y, Li Y, Anand-Srivastava MB. Knockdown of Inhibitory Guanine Nucleotide Binding Protein Giα-2 by Antisense Oligodeoxynucleotides Attenuates the Development of Hypertension and Tachycardia in Spontaneously Hypertensive Rats. J Am Heart Assoc 2016; 5:e004594. [PMID: 27912212 PMCID: PMC5210347 DOI: 10.1161/jaha.116.004594] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 09/30/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND We previously showed that the levels of both Giα-2 and Giα-3 proteins were augmented in spontaneously hypertensive rats (SHRs) before the onset of hypertension. In addition, intraperitoneal injection of pertussis toxin, which inactivates both Giα proteins, prevented the development of hypertension in SHRs. The aim of the present study was to determine the specific contributions of Giα-2 and Giα-3 proteins to the development of hypertension. METHODS AND RESULTS Antisense oligodeoxynucleotide of Giα-2 and Giα-3 encapsulated in PEG/DOTAP/DOPE cationic liposomes were administrated intravenously into 3-week-old prehypertensive SHRs and Wistar Kyoto rats, whereas the control Wistar Kyoto rats and SHRs received PBS, empty liposomes, or sense. The knockdown of Giα-2 but not Giα-3 protein attenuated tachycardia and prevented the development of hypertension up to age 6 weeks; thereafter, blood pressure started increasing and reached the same level as that of untreated SHRs at 9 weeks. Furthermore, Giα-2 and Giα-3 antisense oligodeoxynucleotide treatments significantly decreased the enhanced levels of Giα-2 and Giα-3 proteins, respectively, and enhanced levels of superoxide anion and NADPH oxidase activity in heart, aorta, and kidney and hyperproliferation of vascular smooth muscle cells from SHRs aged 6 weeks. In addition, antisense oligodeoxynucleotide treatment with Giα-2 but not Giα-3 restored enhanced inhibition of adenylyl cyclase by oxotremorine to WKY levels. CONCLUSIONS These results suggested that the enhanced expression of Giα-2 but not Giα-3 protein plays an important role in the pathogenesis of hypertension and tachycardia in SHRs.
Collapse
MESH Headings
- Adenylyl Cyclase Inhibitors/pharmacology
- Animals
- Aorta/metabolism
- Blood Pressure/physiology
- Cells, Cultured
- Disease Models, Animal
- GTP-Binding Protein alpha Subunit, Gi2/deficiency
- GTP-Binding Protein alpha Subunit, Gi2/physiology
- GTP-Binding Protein alpha Subunits, Gi-Go/deficiency
- GTP-Binding Protein alpha Subunits, Gi-Go/physiology
- Gene Knockdown Techniques
- Heart Rate/physiology
- Hypertension/prevention & control
- Kidney/metabolism
- Liposomes/administration & dosage
- Male
- Muscle, Smooth, Vascular/metabolism
- Myocardium/metabolism
- Oligodeoxyribonucleotides, Antisense/physiology
- Rats, Inbred SHR
- Rats, Inbred WKY
- Signal Transduction/physiology
- Tachycardia/prevention & control
- Transfection/methods
Collapse
Affiliation(s)
- Yousra Ali El-Basyuni
- Department of Molecular and Integrative Physiology, Faculty of Medicine, University of Montreal, Quebec, Canada
| | - Yuan Li
- Department of Molecular and Integrative Physiology, Faculty of Medicine, University of Montreal, Quebec, Canada
| | - Madhu B Anand-Srivastava
- Department of Molecular and Integrative Physiology, Faculty of Medicine, University of Montreal, Quebec, Canada
| |
Collapse
|
75
|
Banday AA. Chronic insulin treatment phosphorylates the renal Na-K-ATPase α1-subunit at serine 16/23 and reduces its activity involving PI3-kinase-dependent PKC activation. Am J Physiol Renal Physiol 2016; 311:F958-F966. [PMID: 27605582 DOI: 10.1152/ajprenal.00355.2016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 08/31/2016] [Indexed: 11/22/2022] Open
Abstract
The regulation of Na-K-ATPase in various tissues is under the control of a number of hormones and peptides that exert both short- and long-term control over its activity. The present study was performed to investigate the effect of chronic insulin treatment on Na-K-ATPase in renal proximal tubular cells. Incubation of opossum kidney (OK) cells, transfected with the rat Na-K-ATPase α1-subunit, with 1 nmol/l insulin for 48 h decreased Na-K-ATPase activity. Insulin decreased α1-protein content and increased α1-serine phosphorylation and α1-adaptor protein 2 (AP2) interaction. Removal of the 26 NH2-terminal (-NT) amino acid from the α1-subunit containing serine/threonine sites abolished the insulin-mediated serine phosphorylation and inhibition of Na-K-ATPase. Substitution of serine 16 and 23 with alanine showed a comparable effect on -NT. Insulin increased the activity of protein kinase C (PKC), which was blocked by the phosphatidylinositol 3-kinase (PI3K) inhibitor wortmannin. Both PI3K and PKC inhibitors abolished the insulin-mediated inhibition of Na-K-ATPase. Insulin increased the expression of PKC-β1, -δ, -ξ, and-λ; however, only PKC-ξ/λ-specific inhibitors blocked insulin-induced phosphorylation and inhibition of Na-K-ATPase. Our data demonstrate that insulin activates the atypical PKC isoforms-ξ/λ via the PI3K pathway. PKC-ξ/λ-induced phosphorylation of the α1-subunit at serine 16 and 23 leads to AP2 recruitment, degradation, and a decrease in Na-K-ATPase activity.
Collapse
|
76
|
Pirkmajer S, Chibalin AV. Na,K-ATPase regulation in skeletal muscle. Am J Physiol Endocrinol Metab 2016; 311:E1-E31. [PMID: 27166285 DOI: 10.1152/ajpendo.00539.2015] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 05/02/2016] [Indexed: 12/17/2022]
Abstract
Skeletal muscle contains one of the largest and the most dynamic pools of Na,K-ATPase (NKA) in the body. Under resting conditions, NKA in skeletal muscle operates at only a fraction of maximal pumping capacity, but it can be markedly activated when demands for ion transport increase, such as during exercise or following food intake. Given the size, capacity, and dynamic range of the NKA pool in skeletal muscle, its tight regulation is essential to maintain whole body homeostasis as well as muscle function. To reconcile functional needs of systemic homeostasis with those of skeletal muscle, NKA is regulated in a coordinated manner by extrinsic stimuli, such as hormones and nerve-derived factors, as well as by local stimuli arising in skeletal muscle fibers, such as contractions and muscle energy status. These stimuli regulate NKA acutely by controlling its enzymatic activity and/or its distribution between the plasma membrane and the intracellular storage compartment. They also regulate NKA chronically by controlling NKA gene expression, thus determining total NKA content in skeletal muscle and its maximal pumping capacity. This review focuses on molecular mechanisms that underlie regulation of NKA in skeletal muscle by major extrinsic and local stimuli. Special emphasis is given to stimuli and mechanisms linking regulation of NKA and energy metabolism in skeletal muscle, such as insulin and the energy-sensing AMP-activated protein kinase. Finally, the recently uncovered roles for glutathionylation, nitric oxide, and extracellular K(+) in the regulation of NKA in skeletal muscle are highlighted.
Collapse
Affiliation(s)
- Sergej Pirkmajer
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia; and
| | - Alexander V Chibalin
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
77
|
Briffa JF, Grinfeld E, Jenkin KA, Mathai ML, Poronnik P, McAinch AJ, Hryciw DH. Diet induced obesity in rats reduces NHE3 and Na(+) /K(+) -ATPase expression in the kidney. Clin Exp Pharmacol Physiol 2016; 42:1118-26. [PMID: 26173747 DOI: 10.1111/1440-1681.12452] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 06/25/2015] [Accepted: 07/01/2015] [Indexed: 01/25/2023]
Abstract
The consumption of a high fat diet (HFD) is associated with proteinuria and altered sodium handling and excretion, which can lead to kidney disease. In the proximal tubule, the Na(+) /H(+) Exchanger 3 (NHE3) is responsible for normal protein reabsorption and the reabsorption of approximately 70% of the renal sodium load. It is the Na(+) /K(+) -ATPase that provides the driving force for the reabsorption of sodium and its exit across the basolateral membrane. This study investigates the effects that consumption of a HFD for 12 weeks has on NHE3 and Na(+) /K(+) -ATPase expression in the kidney. Western blot analysis identified a significant reduction in NHE3 and its modulator, phosphorylated protein kinase B, in renal lysate from obese rats. In the obese rats, a reduction in NHE3 expression in the proximal tubule may impact on the acidification of endosomes which are responsible for albumin uptake, suggesting a key role for the exchanger in protein endocytosis in obesity. Western blot analysis identified a reduction in Na(+) /K(+) -ATPase which could also potentially impact on albumin uptake and sodium reabsorption. This study demonstrates that consumption of a HFD for 12 weeks reduces renal NHE3 and Na(+) /K(+) -ATPase expression, an effect that may contribute to the albuminuria associated with obesity. Furthermore the reduction in these transporters is not likely to contribute to the reduced sodium excretion in obesity. These data highlight a potential link between NHE3 and Na(+) /K(+) -ATPase in the pathophysiological changes in renal protein handling observed in obesity.
Collapse
Affiliation(s)
- J F Briffa
- Centre for Chronic Disease Prevention and Management, College of Health and Biomedicine, Victoria University, St Albans, Victoria, Australia.,Department of Physiology, The University of Melbourne, Parkville, Victoria, Australia
| | - E Grinfeld
- Centre for Chronic Disease Prevention and Management, College of Health and Biomedicine, Victoria University, St Albans, Victoria, Australia
| | - K A Jenkin
- Centre for Chronic Disease Prevention and Management, College of Health and Biomedicine, Victoria University, St Albans, Victoria, Australia
| | - M L Mathai
- Centre for Chronic Disease Prevention and Management, College of Health and Biomedicine, Victoria University, St Albans, Victoria, Australia
| | - P Poronnik
- Department of Physiology, School of Medical Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - A J McAinch
- Centre for Chronic Disease Prevention and Management, College of Health and Biomedicine, Victoria University, St Albans, Victoria, Australia
| | - D H Hryciw
- Department of Physiology, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
78
|
Feraille E, Dizin E. Coordinated Control of ENaC and Na+,K+-ATPase in Renal Collecting Duct. J Am Soc Nephrol 2016; 27:2554-63. [PMID: 27188842 DOI: 10.1681/asn.2016020124] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Tubular reabsorption of filtered sodium is tightly controlled to maintain body volume homeostasis. The rate of sodium transport by collecting duct (CD) cells varies widely in response to dietary sodium intake, GFR, circulating hormones, neural signals, and local regulatory factors. Reabsorption of filtered sodium by CD cells occurs via a two-step process. First, luminal sodium crosses the apical plasma membrane along its electrochemical gradient through epithelial sodium channels (ENaC). Intracellular sodium is then actively extruded into the interstitial space by the Na(+),K(+)-ATPase located along the basolateral membrane. Mismatch between sodium entry and exit induces variations in sodium intracellular concentration and cell volume that must be maintained within narrow ranges for control of vital cell functions. Therefore, renal epithelial cells display highly coordinated apical and basolateral sodium transport rates. We review evidence from experiments conducted in vivo and in cultured cells that indicates aldosterone and vasopressin, the two major hormones regulating sodium reabsorption by CD, generate a coordinated stimulation of apical ENaC and basolateral Na(+),K(+)-ATPase. Moreover, we discuss evidence suggesting that variations in sodium entry per se induce a coordinated change in Na(+),K(+)-ATPase activity through the signaling of protein kinases such as protein kinase A and p38 mitogen-activated protein kinase.
Collapse
Affiliation(s)
- Eric Feraille
- Department of Cell Biology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Eva Dizin
- Department of Cell Biology and Metabolism, University of Geneva, Geneva, Switzerland
| |
Collapse
|
79
|
Uroguanylin modulates (Na++K+)ATPase in a proximal tubule cell line: Interactions among the cGMP/protein kinase G, cAMP/protein kinase A, and mTOR pathways. Biochim Biophys Acta Gen Subj 2016; 1860:1431-8. [PMID: 27102282 DOI: 10.1016/j.bbagen.2016.04.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 03/31/2016] [Accepted: 04/15/2016] [Indexed: 11/21/2022]
Abstract
BACKGROUND The natriuretic effect of uroguanylin (UGN) involves reduction of proximal tubule (PT) sodium reabsorption. However, the target sodium transporters as well as the molecular mechanisms involved in these processes remain poorly understood. METHODS To address the effects of UGN on PT (Na(+)+K(+))ATPase and the signal transduction pathways involved in this effect, we used LLC-PK1 cells. The effects of UGN were determined through ouabain-sensitive ATP hydrolysis and immunoblotting assays during different experimental conditions. RESULTS We observed that UGN triggers cGMP/PKG and cAMP/PKA pathways in a sequential way. The activation of PKA leads to the inhibition of mTORC2 activity, PKB phosphorylation at S473, PKB activity and, consequently, a decrease in the mTORC1/S6K pathway. The final effects are decreased expression of the α1 subunit of (Na(+)+K(+))ATPase and inhibition of enzyme activity. CONCLUSIONS These results suggest that the molecular mechanism of action of UGN on sodium reabsorption in PT cells is more complex than previously thought. We propose that PKG-dependent activation of PKA leads to the inhibition of the mTORC2/PKB/mTORC1/S6K pathway, an important signaling pathway involved in the maintenance of the PT sodium pump expression and activity. GENERAL SIGNIFICANCE The current results expand our understanding of the signal transduction pathways involved in the overall effect of UGN on renal sodium excretion.
Collapse
|
80
|
Sciancalepore AG, Portone A, Moffa M, Persano L, De Luca M, Paiano A, Sallustio F, Schena FP, Bucci C, Pisignano D. Micropatterning control of tubular commitment in human adult renal stem cells. Biomaterials 2016; 94:57-69. [PMID: 27105437 DOI: 10.1016/j.biomaterials.2016.03.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Revised: 03/19/2016] [Accepted: 03/28/2016] [Indexed: 12/12/2022]
Abstract
The treatment of renal injury by autologous, patient-specific adult stem cells is still an unmet need. Unsolved issues remain the spatial integration of stem cells into damaged areas of the organ, the commitment in the required cell type and the development of improved bioengineered devices. In this respect, biomaterials and architectures have to be specialized to control stem cell differentiation. Here, we perform an extensive study on micropatterned extracellular matrix proteins, which constitute a simple and non-invasive approach to drive the differentiation of adult renal progenitor/stem cells (ARPCs) from human donors. ARPCs are interfaced with fibronectin (FN) micropatterns, in the absence of exogenous chemicals or cellular reprogramming. We obtain the differentiation towards tubular cells of ARPCs cultured in basal medium conditions, the tubular commitment thus being specifically induced by micropatterned substrates. We characterize the stability of the tubular differentiation as well as the induction of a polarized phenotype in micropatterned ARPCs. Thus, the developed cues, driving the functional commitment of ARPCs, offer a route to recreate the microenvironment of the stem cell niche in vitro, that may serve, in perspective, for the development of ARPC-based bioengineered devices.
Collapse
Affiliation(s)
- Anna G Sciancalepore
- Istituto Nanoscienze-CNR, Euromediterranean Center for Nanomaterial Modelling and Technology (ECMT), via Arnesano, 73100 Lecce, Italy; Center for Biomolecular Nanotechnologies @UNILE, Istituto Italiano di Tecnologia, via Barsanti, 73010 Arnesano, Lecce, Italy.
| | - Alberto Portone
- Istituto Nanoscienze-CNR, Euromediterranean Center for Nanomaterial Modelling and Technology (ECMT), via Arnesano, 73100 Lecce, Italy; Dipartimento di Matematica e Fisica "Ennio De Giorgi", Università del Salento, via Arnesano, 73100 Lecce, Italy
| | - Maria Moffa
- Istituto Nanoscienze-CNR, Euromediterranean Center for Nanomaterial Modelling and Technology (ECMT), via Arnesano, 73100 Lecce, Italy; Center for Biomolecular Nanotechnologies @UNILE, Istituto Italiano di Tecnologia, via Barsanti, 73010 Arnesano, Lecce, Italy
| | - Luana Persano
- Istituto Nanoscienze-CNR, Euromediterranean Center for Nanomaterial Modelling and Technology (ECMT), via Arnesano, 73100 Lecce, Italy
| | - Maria De Luca
- Dipartimento di Scienze e Tecnologie Biologiche ed Ambientali (DiSTeBA), Università del Salento, via Provinciale Monteroni, 73100 Lecce, Italy
| | - Aurora Paiano
- Dipartimento di Scienze e Tecnologie Biologiche ed Ambientali (DiSTeBA), Università del Salento, via Provinciale Monteroni, 73100 Lecce, Italy
| | - Fabio Sallustio
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, 70124 Bari, Italy; Centro Addestramento Ricerca Scientifica in Oncologia (C.A.R.S.O.) Consortium, Strada Prov. le Valenzano-Casamassima, 70010 Valenzano, Italy
| | - Francesco P Schena
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, 70124 Bari, Italy; Centro Addestramento Ricerca Scientifica in Oncologia (C.A.R.S.O.) Consortium, Strada Prov. le Valenzano-Casamassima, 70010 Valenzano, Italy
| | - Cecilia Bucci
- Dipartimento di Scienze e Tecnologie Biologiche ed Ambientali (DiSTeBA), Università del Salento, via Provinciale Monteroni, 73100 Lecce, Italy
| | - Dario Pisignano
- Istituto Nanoscienze-CNR, Euromediterranean Center for Nanomaterial Modelling and Technology (ECMT), via Arnesano, 73100 Lecce, Italy; Center for Biomolecular Nanotechnologies @UNILE, Istituto Italiano di Tecnologia, via Barsanti, 73010 Arnesano, Lecce, Italy; Dipartimento di Matematica e Fisica "Ennio De Giorgi", Università del Salento, via Arnesano, 73100 Lecce, Italy.
| |
Collapse
|
81
|
Orellana AM, Kinoshita PF, Leite JA, Kawamoto EM, Scavone C. Cardiotonic Steroids as Modulators of Neuroinflammation. Front Endocrinol (Lausanne) 2016; 7:10. [PMID: 26909067 PMCID: PMC4754428 DOI: 10.3389/fendo.2016.00010] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 01/22/2016] [Indexed: 01/13/2023] Open
Abstract
Cardiotonic steroids (CTS) are a class of specific ligands of the Na(+), K(+)- ATPase (NKA). NKA is a P-type ATPase that is ubiquitously expressed and although well known to be responsible for the maintenance of the cell electrochemical gradient through active transport, NKA can also act as a signal transducer in the presence of CTS. Inflammation, in addition to importantly driving organism defense and survival mechanisms, can also modulate NKA activity and memory formation, as well as being relevant to many chronic illnesses, neurodegenerative diseases, and mood disorders. The aim of the current review is to highlight the recent advances as to the role of CTS and NKA in inflammatory process, with a particular focus in the central nervous system.
Collapse
Affiliation(s)
- Ana Maria Orellana
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Paula Fernanda Kinoshita
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Jacqueline Alves Leite
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Elisa Mitiko Kawamoto
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Cristoforo Scavone
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
- *Correspondence: Cristoforo Scavone,
| |
Collapse
|
82
|
Soares-da-Silva P, Cabral JM, Magalhães D, Fraga S, Magro F. Amine neurotransmitters, inflammation and epithelial sodium transport. Exp Physiol 2015; 101:459-64. [PMID: 26548358 DOI: 10.1113/ep085284] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 11/02/2015] [Indexed: 12/13/2022]
Abstract
NEW FINDINGS What is the topic of this review? The present work reviews the roles of renal and intestinal dopamine and 5-HT in the maintenance of fluid and electrolyte homeostasis. The role of inflammatory agents at the intestinal level that affect fluid and electrolyte homeostasis is also addressed. What advances does it highlight? General mechanisms of epithelial cell ion transport in the gastrointestinal tract and kidney share considerable similarities, particularly with regard to basolateral Na(+) ,K(+-) ATPase as a driving force for the movement of numerous substrates across the cell membrane. The physiological importance of the renal actions of monoamines (dopamine, noradrenaline and 5-HT) mainly depends on the sources of the amines in the kidney and on their availability to activate the amine-specific receptors. Dopamine and 5-HT are also relatively abundant in the mucosal cell layer of the intestine, and recent evidence suggests their physiological relevance in regulating electrolyte transport. The gastrointestinal tract can be an important site for the loss of water and electrolytes, in the presence of intestinal inflammation. General mechanisms of epithelial cell ion transport in the gastrointestinal tract and kidney share considerable similarities with regard to basolateral Na(+) ,K(+) -ATPase as a driving force for the movement of numerous substrates across the cell membrane. The present work reviews the roles of renal and intestinal dopamine and 5-HT in the maintenance of fluid and electrolyte homeostasis. The role of inflammatory agents at the intestinal level that affect fluid and electrolyte homeostasis is also addressed.
Collapse
Affiliation(s)
- Patrício Soares-da-Silva
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto, Porto, Portugal.,MedInUP - Center for Drug Discovery and Innovative Medicines, University of Porto, Porto, Portugal
| | - José Miguel Cabral
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Diogo Magalhães
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Sónia Fraga
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto, Porto, Portugal.,MedInUP - Center for Drug Discovery and Innovative Medicines, University of Porto, Porto, Portugal
| | - Fernando Magro
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto, Porto, Portugal.,MedInUP - Center for Drug Discovery and Innovative Medicines, University of Porto, Porto, Portugal
| |
Collapse
|
83
|
Abstract
From a Cartesian perspective of rational analysis, the electric potential difference across the cell membrane is one of the fundamental concepts for the study of physiology. Unfortunately, undergraduate students often struggle to understand the genesis of this energy gradient, which makes the teaching activity a hard task for the instructor. The topic of bioelectrogenesis encompasses multidisciplinary concepts, involves several mechanisms, and is a dynamic process, i.e., it never turns off during the lifetime of the cell. Therefore, to improve the transmission and acquisition of knowledge in this field, I present an alternative didactic model. The design of the model assumes that it is possible to build, in a series of sequential steps, an assembly of proteins within the membrane of an isolated cell in a simulated electrophysiology experiment. Initially, no proteins are inserted in the membrane and the cell is at a baseline energy state; the extracellular and intracellular fluids are at thermodynamic equilibrium. Students are guided through a sequence of four steps that add key membrane transport proteins to the model cell. The model is simple at the start and becomes progressively more complex, finally producing transmembrane chemical and electrical gradients. I believe that this didactic approach helps instructors with a more efficient tool for the teaching of the mechanisms of resting membrane potential while helping students avoid common difficulties that may be encountered when learning this topic.
Collapse
Affiliation(s)
- Vander Baptista
- Department of Physiological Sciences, Centre of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| |
Collapse
|
84
|
Gattineni J, Baum M. Developmental changes in renal tubular transport-an overview. Pediatr Nephrol 2015; 30:2085-98. [PMID: 24253590 PMCID: PMC4028442 DOI: 10.1007/s00467-013-2666-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 10/01/2013] [Accepted: 10/11/2013] [Indexed: 10/26/2022]
Abstract
The adult kidney maintains a constant volume and composition of extracellular fluid despite changes in water and salt intake. The neonate is born with a kidney that has a small fraction of the glomerular filtration rate of the adult and immature tubules that function at a lower capacity than that of the mature animal. Nonetheless, the neonate is also able to maintain a constant extracellular fluid volume and composition. Postnatal renal tubular development was once thought to be due to an increase in the transporter abundance to meet the developmental increase in glomerular filtration rate. However, postnatal renal development of each nephron segment is quite complex. There are isoform changes of several transporters as well as developmental changes in signal transduction that affect the capacity of renal tubules to reabsorb solutes and water. This review will discuss neonatal tubular function with an emphasis on the differences that have been found between the neonate and adult. We will also discuss some of the factors that are responsible for the maturational changes in tubular transport that occur during postnatal renal development.
Collapse
Affiliation(s)
- Jyothsna Gattineni
- Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-9061, USA
| | - Michel Baum
- Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-9061, USA.
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
85
|
Jansson K, Venugopal J, Sánchez G, Magenheimer BS, Reif GA, Wallace DP, Calvet JP, Blanco G. Ouabain Regulates CFTR-Mediated Anion Secretion and Na,K-ATPase Transport in ADPKD Cells. J Membr Biol 2015; 248:1145-57. [PMID: 26289599 DOI: 10.1007/s00232-015-9832-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 08/06/2015] [Indexed: 01/16/2023]
Abstract
Cyst enlargement in autosomal dominant polycystic kidney disease (ADPKD) requires the transepithelial secretion of fluid into the cyst lumen. We previously showed that physiological amounts of ouabain enhance cAMP-dependent fluid secretion and cyst growth of human ADPKD cyst epithelial cells in culture and formation of cyst-like dilations in metanephric kidneys from Pkd1 mutant mice. Here, we investigated the mechanisms by which ouabain promotes cAMP-dependent fluid secretion and cystogenesis. Ouabain (3 nM) enhanced cAMP-induced cyst-like dilations in embryonic kidneys from Pkd1 (m1Bei) mice, but had no effect on metanephroi from Pkd1 (m1Bei) mice that lack expression of the cystic fibrosis transmembrane conductance regulator (CFTR). Similarly, ouabain stimulation of cAMP-induced fluid secretion and in vitro cyst growth of ADPKD cells were abrogated by CFTR inhibition, showing that CFTR is required for ouabain effects on ADPKD fluid secretion. Moreover, ouabain directly enhanced the cAMP-dependent Cl(-) efflux mediated by CFTR in ADPKD monolayers. Ouabain increased the trafficking of CFTR to the plasma membrane and up-regulated the expression of the CFTR activator PDZK1. Finally, ouabain decreased plasma membrane expression and activity of the Na,K-ATPase in ADPKD cells. Altogether, these results show that ouabain enhances net fluid secretion and cyst formation by activating apical anion secretion via CFTR and decreasing basolateral Na(+) transport via Na,K-ATPase. These results provide new information on the mechanisms by which ouabain affects ADPKD cells and further highlight the importance of ouabain as a non-genomic stimulator of cystogenesis in ADPKD.
Collapse
Affiliation(s)
- Kyle Jansson
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA.,The Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jessica Venugopal
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA.,The Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Gladis Sánchez
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA.,The Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Brenda S Magenheimer
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA.,The Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Gail A Reif
- Department of Medicine, University of Kansas Medical Center, Kansas City, KS, USA.,The Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Darren P Wallace
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA.,Department of Medicine, University of Kansas Medical Center, Kansas City, KS, USA.,The Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - James P Calvet
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA.,The Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Gustavo Blanco
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA. .,The Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
86
|
Gomez-Sanchez CE, Kuppusamy M, Gomez-Sanchez EP. Somatic mutations of the ATP1A1 gene and aldosterone-producing adenomas. Mol Cell Endocrinol 2015; 408:213-9. [PMID: 25496839 PMCID: PMC4417446 DOI: 10.1016/j.mce.2014.12.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 12/05/2014] [Accepted: 12/05/2014] [Indexed: 01/01/2023]
Abstract
Primary aldosteronism is the most common form of secondary hypertension. It affects approximately 10% of patients with hypertension and causes greater cardiovascular morbidity and mortality compared to essential hypertension of similar severity and duration. The cause of primary aldosteronism in about half of these patients is an aldosterone-producing adenoma; over half of these adenomas have mutations in one of several ion channels and pumps, including the potassium channel KCNJ5, calcium channel Cav1.3, α1 subunit of the sodium potassium ATPase, and membrane calcium ATPase 3. This review concentrates on the molecular and physiological mechanisms by which mutations of the ATP1A1 gene increase aldosterone production.
Collapse
Affiliation(s)
- Celso E Gomez-Sanchez
- Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS, USA; Department of Medicine-Endocrinology, University of Mississippi Medical Center, Jackson, MS, USA.
| | - Maniselvan Kuppusamy
- Department of Medicine-Endocrinology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Elise P Gomez-Sanchez
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
87
|
Abstract
The amiloride-sensitive epithelial Na(+) channel (ENaC) is a key player in the regulation of Na(+) homeostasis. Its functional activity is under continuous control by a variety of signaling molecules, including bioactive peptides of endothelin family. Since ENaC dysfunction is causative for disturbances in total body Na(+) levels associated with the abnormal regulation of blood volume, blood pressure, and lung fluid balance, uncovering the molecular mechanisms of inhibitory modulation or inappropriate activation of ENaC is crucial for the successful treatment of a variety of human diseases including hypertension. The precise regulation of ENaC is particularly important for normal Na(+) and fluid homeostasis in organs where endothelins are known to act: the kidneys, lung, and colon. Inhibition of ENaC by endothelin-1 (ET-1) has been established in renal cells, and several molecular mechanisms of inhibition of ENaC by ET-1 are proposed and will be reviewed in this chapter.
Collapse
Affiliation(s)
- Andrey Sorokin
- Division of Nephrology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
| | | |
Collapse
|
88
|
Maturana-Teixeira S, Braga LEG, Carpi Santos R, Calaza KDC, Giestal-de-Araujo E, Leão-Ferreira LR. The (Na(+)/K (+))-ATPase activity in the developing rat retina: the role of insulin-like growth factor-I (IGF-I). Cell Mol Neurobiol 2015; 35:243-54. [PMID: 25274047 DOI: 10.1007/s10571-014-0119-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 09/23/2014] [Indexed: 02/04/2023]
Abstract
In this work, the (Na(+)/K(+))-ATPase activity was evaluated during the early stages of the postnatal development of rat retina and showed an almost three-time increase from P0 to P14. Expression of the three catalytic subunit isoforms (α1, α2, and α3) of the (Na(+)/K(+))-ATPase was also evaluated by immunoblot in the same period, but no correlation to the catalytic activity increment was observed. On the other hand, immunolocalization of these three α-catalytic isoforms in the developing retina showed an age-related pattern. Involvement of IGF-I in the stimulation of the (Na(+)/K(+))-ATPase was investigated. Our results demonstrate that the exogenous IGF-I (10 ng/mL) stimulates enzyme activity at the age of P7 only. Incubation of retinas with 10 μM I-OMe-AG 538 (inhibitor of the IGF-I receptor) indicates that the basal (Na(+)/K(+))-ATPase activity is sustained by endogenous IGF-I in P7 animals. These data were corroborated by an age-dependent decrease in the immunodetection of endogenous IGF-I as well as in the phosphorylation level of its cognate receptor in rat retina homogenates. The signaling pathway involved in IGF-I-induced modulation of the (Na(+)/K(+))-ATPase was also investigated. Our data show that the inhibitory effects induced by I-OMe-AG 538 and the PI 3-kinase inhibitor Ly 294002 on the basal (Na(+)/K(+))-ATPase activity were non-cumulative. Furthermore, IGF-I induced phosphorylation of PKB in a Ly 294002-sensitive manner. Together, these data demonstrate that the PI 3-kinase/PKB signaling pathway is involved in the IGF-I-sustained basal (Na(+)/K(+))-ATPase activity during the first 7 days of the postnatal development of rat retina.
Collapse
Affiliation(s)
- Sheila Maturana-Teixeira
- Programa de Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Outeiro de São João Batista s/n, Niterói, Rio De Janeiro, CEP 24020-140, Brazil
| | | | | | | | | | | |
Collapse
|
89
|
Sepúlveda FV, Pablo Cid L, Teulon J, Niemeyer MI. Molecular aspects of structure, gating, and physiology of pH-sensitive background K2P and Kir K+-transport channels. Physiol Rev 2015; 95:179-217. [PMID: 25540142 DOI: 10.1152/physrev.00016.2014] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
K(+) channels fulfill roles spanning from the control of excitability to the regulation of transepithelial transport. Here we review two groups of K(+) channels, pH-regulated K2P channels and the transport group of Kir channels. After considering advances in the molecular aspects of their gating based on structural and functional studies, we examine their participation in certain chosen physiological and pathophysiological scenarios. Crystal structures of K2P and Kir channels reveal rather unique features with important consequences for the gating mechanisms. Important tasks of these channels are discussed in kidney physiology and disease, K(+) homeostasis in the brain by Kir channel-equipped glia, and central functions in the hearing mechanism in the inner ear and in acid secretion by parietal cells in the stomach. K2P channels fulfill a crucial part in central chemoreception probably by virtue of their pH sensitivity and are central to adrenal secretion of aldosterone. Finally, some unorthodox behaviors of the selectivity filters of K2P channels might explain their normal and pathological functions. Although a great deal has been learned about structure, molecular details of gating, and physiological functions of K2P and Kir K(+)-transport channels, this has been only scratching at the surface. More molecular and animal studies are clearly needed to deepen our knowledge.
Collapse
Affiliation(s)
- Francisco V Sepúlveda
- Centro de Estudios Científicos, Valdivia, Chile; UPMC Université Paris 06, Team 3, Paris, France; and Institut National de la Santé et de la Recherche Médicale, UMR_S 1138, Paris, France
| | - L Pablo Cid
- Centro de Estudios Científicos, Valdivia, Chile; UPMC Université Paris 06, Team 3, Paris, France; and Institut National de la Santé et de la Recherche Médicale, UMR_S 1138, Paris, France
| | - Jacques Teulon
- Centro de Estudios Científicos, Valdivia, Chile; UPMC Université Paris 06, Team 3, Paris, France; and Institut National de la Santé et de la Recherche Médicale, UMR_S 1138, Paris, France
| | - María Isabel Niemeyer
- Centro de Estudios Científicos, Valdivia, Chile; UPMC Université Paris 06, Team 3, Paris, France; and Institut National de la Santé et de la Recherche Médicale, UMR_S 1138, Paris, France
| |
Collapse
|
90
|
Wang F, Cai B, Li KC, Hu XY, Lu YJ, Wang Q, Bao L, Zhang X. FXYD2, a γ subunit of Na⁺, K⁺-ATPase, maintains persistent mechanical allodynia induced by inflammation. Cell Res 2015; 25:318-34. [PMID: 25633594 DOI: 10.1038/cr.2015.12] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Revised: 10/09/2014] [Accepted: 12/15/2014] [Indexed: 01/08/2023] Open
Abstract
Na⁺, K⁺-ATPase (NKA) is required to generate the resting membrane potential in neurons. Nociceptive afferent neurons express not only the α and β subunits of NKA but also the γ subunit FXYD2. However, the neural function of FXYD2 is unknown. The present study shows that FXYD2 in nociceptive neurons is necessary for maintaining the mechanical allodynia induced by peripheral inflammation. FXYD2 interacted with α1NKA and negatively regulated the NKA activity, depolarizing the membrane potential of nociceptive neurons. Mechanical allodynia initiated in FXYD2-deficient mice was abolished 4 days after inflammation, whereas it persisted for at least 3 weeks in wild-type mice. Importantly, the FXYD2/α1NKA interaction gradually increased after inflammation and peaked on day 4 post inflammation, resulting in reduction of NKA activity, depolarization of neuron membrane and facilitation of excitatory afferent neurotransmission. Thus, the increased FXYD2 activity may be a fundamental mechanism underlying the persistent hypersensitivity to pain induced by inflammation.
Collapse
Affiliation(s)
- Feng Wang
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Bing Cai
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Kai-Cheng Li
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xu-Ye Hu
- Shanghai Clinical Center, Chinese Academy of Sciences/XuHui Central Hospital, Shanghai, China
| | - Ying-Jin Lu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qiong Wang
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Lan Bao
- 1] State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China [2] School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China
| | - Xu Zhang
- 1] Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China [2] School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China
| |
Collapse
|
91
|
Zhang LN, Sun YJ, Wang LX, Gao ZB. Glutamate Transporters/Na(+), K(+)-ATPase Involving in the Neuroprotective Effect as a Potential Regulatory Target of Glutamate Uptake. Mol Neurobiol 2015; 53:1124-1131. [PMID: 25586061 DOI: 10.1007/s12035-014-9071-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Accepted: 12/29/2014] [Indexed: 02/05/2023]
Abstract
The glutamate (Glu) transporters GLAST and GLT-1, as the two most important transporters in brain tissue, transport Glu from the extracellular space into the cell protecting against Glu toxicity. Furthermore, GLAST and GLT-1 are sodium-dependent Glu transporters (GluTs) that rely on sodium and potassium gradients generated principally by Na(+), K(+)-ATPase to generate ion gradients that drive Glu uptake. There is an interaction between Na(+), K(+)-ATPase and GluTs to modulate Glu uptake, and Na(+), K(+)-ATPase α, β or γ subunit can be directly coupled to GluTs, co-localizing with GLAST or GLT-1 in vivo to form a macromolecular complex and operate as a functional unit to regulate glutamatergic neurotransmission. Therefore, GluTs/Na(+), K(+)-ATPase may be involved in the neuroprotective effect as a potential regulatory target of Glu uptake in neurodegenerative diseases induced by Glu-mediated neurotoxicity as the final common pathway.
Collapse
Affiliation(s)
- Li-Nan Zhang
- Department of Pharmacy, Hebei University of Science and Technology, 70 Yuhua East Road, Shijiazhuang, Hebei, 050018, People's Republic of China
| | - Yong-Jun Sun
- Department of Pharmacy, Hebei University of Science and Technology, 70 Yuhua East Road, Shijiazhuang, Hebei, 050018, People's Republic of China
| | - Li-Xue Wang
- Cadre Ward, Capital Medical University Electric Power Teaching Hospital, Compound A1, Taiping Bridge Xili, Beijing, 100073, People's Republic of China
| | - Zi-Bin Gao
- Department of Pharmacy, Hebei University of Science and Technology, 70 Yuhua East Road, Shijiazhuang, Hebei, 050018, People's Republic of China. .,State Key Laboratory Breeding Base-Hebei Province Key Laboratory of Molecular Chemistry for Drug, 70 Yuhua East Road, Shijiazhuang, 050018, People's Republic of China.
| |
Collapse
|
92
|
Kumar G, Abd-Elfattah A, El-Matbouli M. Identification of differentially expressed genes of brown trout (Salmo trutta) and rainbow trout (Oncorhynchus mykiss) in response to Tetracapsuloides bryosalmonae (Myxozoa). Parasitol Res 2015; 114:929-39. [PMID: 25563603 PMCID: PMC4336411 DOI: 10.1007/s00436-014-4258-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 12/15/2014] [Indexed: 01/08/2023]
Abstract
Tetracapsuloides bryosalmonae Canning et al., 1999 (Myxozoa) is the causative agent of proliferative kidney disease in various species of salmonids in Europe and North America. We have shown previously that the development and distribution of the European strain of T. bryosalmonae differs in the kidney of brown trout (Salmo trutta) Linnaeus, 1758 and rainbow trout (Oncorhynchus mykiss) Walbaum, 1792, and that intra-luminal sporogonic stages were found in brown trout but not in rainbow trout. We have now compared transcriptomes from kidneys of brown trout and rainbow trout infected with T. bryosalmonae using suppressive subtractive hybridization (SSH). The differentially expressed transcripts produced by SSH were cloned, transformed, and tested by colony PCR. Differential expression screening of PCR products was validated using dot blot, and positive clones having different signal intensities were sequenced. Differential screening and a subsequent NCBI-BLAST analysis of expressed sequence tags revealed nine clones expressed differently between both fish species. These differentially expressed genes were validated by quantitative real-time PCR of kidney samples from both fish species at different time points of infection. Expression of anti-inflammatory (TSC22 domain family protein 3) and cell proliferation (Prothymin alpha) genes were upregulated significantly in brown trout but downregulated in rainbow trout. The expression of humoral immune response (immunoglobulin mu) and endocytic pathway (Ras-related protein Rab-11b) genes were significantly upregulated in rainbow trout but downregulated in brown trout. This study suggests that differential expression of host anti-inflammatory, humoral immune and endocytic pathway responses, cell proliferation, and cell growth processes do not inhibit the development of intra-luminal sporogonic stages of the European strain of T. bryosalmonae in brown trout but may suppress it in rainbow trout.
Collapse
Affiliation(s)
- Gokhlesh Kumar
- Clinical Division of Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Veterinärplatz 1, 1210, Vienna, Austria
| | | | | |
Collapse
|
93
|
Abstract
The thick ascending limb occupies a central anatomic and functional position in human renal physiology, with critical roles in the defense of the extracellular fluid volume, the urinary concentrating mechanism, calcium and magnesium homeostasis, bicarbonate and ammonium homeostasis, and urinary protein composition. The last decade has witnessed tremendous progress in the understanding of the molecular physiology and pathophysiology of this nephron segment. These advances are the subject of this review, with emphasis on particularly recent developments.
Collapse
Affiliation(s)
- David B Mount
- Renal Division, Brigham and Women's Hospital, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| |
Collapse
|
94
|
Hosseinzadeh Z, Schmid E, Shumilina E, Laufer S, Borst O, Gawaz M, Lang F. Effect of TGFβ on Na+/K+ ATPase activity in megakaryocytes. Biochem Biophys Res Commun 2014; 452:537-41. [DOI: 10.1016/j.bbrc.2014.08.093] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 08/20/2014] [Indexed: 11/26/2022]
|
95
|
Posttranslational regulation of the cation–chloride symporter Na+–K+–2Cl− cotransporter-2 in spontaneously hypertensive rat kidneys. J Hypertens 2014; 32:1778-9. [DOI: 10.1097/hjh.0000000000000303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
96
|
Abstract
The concept of homeostasis has been inextricably linked to the function of the kidneys for more than a century when it was recognized that the kidneys had the ability to maintain the "internal milieu" and allow organisms the "physiologic freedom" to move into varying environments and take in varying diets and fluids. Early ingenious, albeit rudimentary, experiments unlocked a wealth of secrets on the mechanisms involved in the formation of urine and renal handling of the gamut of electrolytes, as well as that of water, acid, and protein. Recent scientific advances have confirmed these prescient postulates such that the modern clinician is the beneficiary of a rich understanding of the nephron and the kidney's critical role in homeostasis down to the molecular level. This review summarizes those early achievements and provides a framework and introduction for the new CJASN series on renal physiology.
Collapse
Affiliation(s)
- Melanie P Hoenig
- Division of Nephrology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Mark L Zeidel
- Division of Nephrology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
97
|
Clausell M, Fang Z, Chen W. In vivo study of transepithelial potential difference (TEPD) in proximal convoluted tubules of rat kidney by synchronization modulation electric field. J Membr Biol 2014; 247:601-9. [PMID: 24894719 DOI: 10.1007/s00232-014-9676-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 05/09/2014] [Indexed: 11/30/2022]
Abstract
Synchronization modulation (SM) electric field has been shown to effectively activate function of Na(+)/K(+) pumps in various cells and tissues, including skeletal muscle cells, cardiomyocyte, monolayer of cultured cell line, and peripheral blood vessels. We are now reporting the in vivo studies in application of the SM electric field to kidney of living rats. The field-induced changes in the transepithelial potential difference (TEPD) or the lumen potential from the proximal convoluted tubules were monitored. The results showed that a short time (20 s) application of the SM electric field can significantly increase the magnitude of TEPD from 1-2 mV to about 20 mV. The TEPD is an active potential representing the transport current of the Na/K pumps in epithelial wall of renal tubules. This study showed that SM electric field can increase TEPD by activation of the pump molecules. Considering renal tubules, many active transporters are driven by the Na(+) concentration gradient built by the Na(+)/K(+) pumps, activation of the pump functions and increase in the magnitude of TEPD imply that the SM electric field may improve reabsorption functions of the renal tubules.
Collapse
Affiliation(s)
- Mathis Clausell
- Department of Physics, Cellular and Molecular Biophysics Lab, University of South Florida, Tampa, FL, 33620, USA
| | | | | |
Collapse
|
98
|
Ouabain affects the expression of activation markers, cytokine production, and endocytosis of human monocytes. Mediators Inflamm 2014; 2014:760368. [PMID: 24904197 PMCID: PMC4034513 DOI: 10.1155/2014/760368] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Accepted: 03/03/2014] [Indexed: 11/18/2022] Open
Abstract
Ouabain is a steroid capable of binding to and inhibiting Na+,-K+-ATPase. Studies have demonstrated some actions of ouabain on immune cells, which indicated both pro- and anti-inflammatory properties of this molecule. Nevertheless, its effects on human monocytes are still poorly understood. Thus, the present work investigated effects of ouabain in the activation and function of human adherent monocytes. Our results show that there is an increase in intracellular calcium levels already 5 minutes following monocyte treatment with 10−7 M of ouabain. Furthermore, monocytes expressed increased amounts of surface activation markers such as CD69, HLA-DR, CD86, and CD80 and also presented an augmented endocytic activity of dextran-FITC particles after 24 hours of culture in the presence of ouabain. However, monocytes treated with ouabain did not have an increased stimulatory capacity in allogeneic mixed leukocyte reaction. Ouabain-treated monocytes produced higher levels of IL-1β and TNF-α as reported before. A novel observation was the fact that ouabain induced IL-10 and VEGF as well. Collectively, these results suggest that ouabain impacts monocyte activation and modulates monocyte functions, implying that this steroid could act as an immunomodulator of these cells.
Collapse
|
99
|
Mechanisms of contrast-induced nephropathy reduction for saline (NaCl) and sodium bicarbonate (NaHCO3). BIOMED RESEARCH INTERNATIONAL 2014; 2014:510385. [PMID: 24826379 PMCID: PMC4009105 DOI: 10.1155/2014/510385] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Revised: 02/28/2014] [Accepted: 03/19/2014] [Indexed: 11/17/2022]
Abstract
Nephropathy following contrast media (CM) exposure is reduced by administration before, during, and after the contrast procedure of either isotonic sodium chloride solution (Saline) or isotonic sodium bicarbonate solution (IsoBicarb). The reasons for this reduction are not well established for either sodium salt; probable mechanisms are discussed in this paper. For Saline, the mechanism for the decrease in CIN is likely related primarily to the increased tubular flow rates produced by volume expansion and therefore a decreased concentration of the filtered CM during transit through the kidney tubules. Furthermore, increased tubular flow rates produce a slight increase in tubular pH resulting from a fixed acid excretion in an increased tubular volume. The mechanism for the decreased CIN associated with sodium bicarbonate includes the same mechanisms listed for Saline in addition to a renal pH effect. Increased filtered bicarbonate anion raises both tubular pH and tubular bicarbonate anion levels toward blood physiologic levels, thus providing increased buffer for reactive oxygen species (ROS) formed in the tubules as a result of exposure to CM in renal tubular fluid.
Collapse
|
100
|
Kamenický P, Mazziotti G, Lombès M, Giustina A, Chanson P. Growth hormone, insulin-like growth factor-1, and the kidney: pathophysiological and clinical implications. Endocr Rev 2014; 35:234-81. [PMID: 24423979 DOI: 10.1210/er.2013-1071] [Citation(s) in RCA: 142] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Besides their growth-promoting properties, GH and IGF-1 regulate a broad spectrum of biological functions in several organs, including the kidney. This review focuses on the renal actions of GH and IGF-1, taking into account major advances in renal physiology and hormone biology made over the last 20 years, allowing us to move our understanding of GH/IGF-1 regulation of renal functions from a cellular to a molecular level. The main purpose of this review was to analyze how GH and IGF-1 regulate renal development, glomerular functions, and tubular handling of sodium, calcium, phosphate, and glucose. Whenever possible, the relative contributions, the nephronic topology, and the underlying molecular mechanisms of GH and IGF-1 actions were addressed. Beyond the physiological aspects of GH/IGF-1 action on the kidney, the review describes the impact of GH excess and deficiency on renal architecture and functions. It reports in particular new insights into the pathophysiological mechanism of body fluid retention and of changes in phospho-calcium metabolism in acromegaly as well as of the reciprocal changes in sodium, calcium, and phosphate homeostasis observed in GH deficiency. The second aim of this review was to analyze how the GH/IGF-1 axis contributes to major renal diseases such as diabetic nephropathy, renal failure, renal carcinoma, and polycystic renal disease. It summarizes the consequences of chronic renal failure and glucocorticoid therapy after renal transplantation on GH secretion and action and questions the interest of GH therapy in these conditions.
Collapse
Affiliation(s)
- Peter Kamenický
- Assistance Publique-Hôpitaux de Paris (P.K., M.L., P.C.), Hôpital de Bicêtre, Service d'Endocrinologie et des Maladies de la Reproduction, Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Le Kremlin Bicêtre F-94275, France; Univ Paris-Sud (P.K., M.L., P.C.), Faculté de Médecine Paris-Sud, Le Kremlin Bicêtre F-94276, France; Inserm Unité 693 (P.K., M.L., P.C.), Le Kremlin Bicêtre F-94276, France; and Department of Clinical and Experimental Sciences (A.G., G.M.), Chair of Endocrinology, University of Brescia, 25125 Brescia, Italy
| | | | | | | | | |
Collapse
|