101
|
Stewart G. The emerging physiological roles of the SLC14A family of urea transporters. Br J Pharmacol 2012; 164:1780-92. [PMID: 21449978 DOI: 10.1111/j.1476-5381.2011.01377.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In mammals, urea is the main nitrogenous breakdown product of protein catabolism and is produced in the liver. In certain tissues, the movement of urea across cell membranes is specifically mediated by a group of proteins known as the SLC14A family of facilitative urea transporters. These proteins are derived from two distinct genes, UT-A (SLC14A2) and UT-B (SLC14A1). Facilitative urea transporters play an important role in two major physiological processes - urinary concentration and urea nitrogen salvaging. Although UT-A and UT-B transporters both have a similar basic structure and mediate the transport of urea in a facilitative manner, there are a number of significant differences between them. UT-A transporters are mainly found in the kidney, are highly specific for urea, have relatively lower transport rates and are highly regulated at both gene expression and cellular localization levels. In contrast, UT-B transporters are more widespread in their tissue location, transport both urea and water, have a relatively high transport rate, are inhibited by mercurial compounds and currently appear to be less acutely regulated. This review details the fundamental research that has so far been performed to investigate the function and physiological significance of these two types of urea transporters.
Collapse
Affiliation(s)
- Gavin Stewart
- School of Biology & Environmental Science, College of Life Sciences, University College Dublin, Belfield, Dublin, Ireland.
| |
Collapse
|
102
|
Saadat N, IglayReger HB, Myers MG, Bodary P, Gupta SV. Differences in metabolomic profiles of male db/db and s/s, leptin receptor mutant mice. Physiol Genomics 2012; 44:374-81. [PMID: 22318992 DOI: 10.1152/physiolgenomics.00081.2011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Leptin, a protein hormone secreted by adipose tissue, plays an important role in regulating energy metabolism and the immune response. Despite similar extremes of adiposity, mutant mouse models, db/db, carrying spontaneous deletion of the active form of the leptin receptor (LEPR-B) intracellular signaling domain, and the s/s, carrying a specific point mutation leading to a dysfunctional LEPR-B-STAT3 signaling pathway, have been shown to have robust differences in glucose homeostasis. This suggests specific effects of leptin, mediated by non-STAT3 LEPR-B pathways. Differences in the LEPR-B signaling pathways in these two LEPR-B mutant mice models are expected to lead to differences in metabolism. In the current study, the hypothesized differences in metabolism were investigated using the metabolomics approach. Proton nuclear magnetic resonance spectroscopy ((1)HNMR) was conducted on 24 h urine samples in deuterium oxide using a 500 MHz instrument at 25°C. Principle Component Analysis showed clear separation of urine NMR spectra between the groups (P < 0.05). The CHENOMX metabolite database was used to identify several metabolites that differed between the two mouse models. Significant differences (P < 0.05) in metabolites associated with the glycine, serine, and homocysteine metabolism were observed. The results demonstrate that the metabolomic profile of db/db and s/s mice are fundamentally different and provide insight into the unique metabolic effects of leptin exerted through non-STAT3 LEPR-B pathways.
Collapse
Affiliation(s)
- Nadia Saadat
- Nutrition and Food Science, Wayne State University, Detroit, MI, USA
| | | | | | | | | |
Collapse
|
103
|
Zhou L, Meng Y, Lei T, Zhao D, Su J, Zhao X, Yang B. UT-B-deficient mice develop renal dysfunction and structural damage. BMC Nephrol 2012; 13:6. [PMID: 22289137 PMCID: PMC3293738 DOI: 10.1186/1471-2369-13-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 01/30/2012] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Urea transporter UT-B is the major urea transporter in erythrocytes and the descending vasa recta in the kidney. In this study, we investigated the effects of long-term UT-B deficiency on functional and structural defect in the kidney of 16-and 52-week-old UT-B-null mice. METHODS UT-B-knockout mice were generated by targeted gene disruption and lacked UT-B protein expression in all organs. The urinary concentrating ability of mice was studied in terms of daily urine output, urine osmolality, and urine and plasma chemistries. Changes in renal morphology were evaluated by hematoxylin and eosin staining. RESULTS The UT-B-null mice showed defective urine concentrating ability. The daily urine output in UT-B-null mice (2.5 ± 0.1 ml) was 60% higher and urine osmolality (985 ± 151 mosm) was significantly lower than that in wild-type mice (1463 ± 227 mosm). The 52-week-old UT-B-null mice exhibited polyuria after water deprivation, although urine osmolality was increased. At 52 weeks of age, over 31% of UT-B-null mice exhibited renal medullary atrophy because of severe polyuria and hydronephrosis. CONCLUSIONS Long-term UT-B deficiency causes severe renal dysfunction and structural damage. These results demonstrate the important role of UT-B in countercurrent exchange and urine concentration.
Collapse
Affiliation(s)
- Lei Zhou
- Prostate Diseases Prevention and Treatment Research Center, Department of Pathophysiology, Norman Bethune College of Medicine, Jilin University, Changchun, China
- Department of Pathology, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Yan Meng
- Prostate Diseases Prevention and Treatment Research Center, Department of Pathophysiology, Norman Bethune College of Medicine, Jilin University, Changchun, China
| | - Tianluo Lei
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Dan Zhao
- Prostate Diseases Prevention and Treatment Research Center, Department of Pathophysiology, Norman Bethune College of Medicine, Jilin University, Changchun, China
| | - Jing Su
- Prostate Diseases Prevention and Treatment Research Center, Department of Pathophysiology, Norman Bethune College of Medicine, Jilin University, Changchun, China
| | - Xuejian Zhao
- Prostate Diseases Prevention and Treatment Research Center, Department of Pathophysiology, Norman Bethune College of Medicine, Jilin University, Changchun, China
| | - Baoxue Yang
- Prostate Diseases Prevention and Treatment Research Center, Department of Pathophysiology, Norman Bethune College of Medicine, Jilin University, Changchun, China
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| |
Collapse
|
104
|
|
105
|
Lei T, Zhou L, Layton AT, Zhou H, Zhao X, Bankir L, Yang B. Role of thin descending limb urea transport in renal urea handling and the urine concentrating mechanism. Am J Physiol Renal Physiol 2011; 301:F1251-9. [PMID: 21849488 PMCID: PMC3233864 DOI: 10.1152/ajprenal.00404.2011] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 08/15/2011] [Indexed: 11/22/2022] Open
Abstract
Urea transporters UT-A2 and UT-B are expressed in epithelia of thin descending limb of Henle's loop and in descending vasa recta, respectively. To study their role and possible interaction in the context of the urine concentration mechanism, a UT-A2 and UT-B double knockout (UT-A2/B knockout) mouse model was generated by targeted deletion of the UT-A2 promoter in embryonic stem cells with UT-B gene knockout. The UT-A2/B knockout mice lacked detectable UT-A2 and UT-B transcripts and proteins and showed normal survival and growth. Daily urine output was significantly higher in UT-A2/B knockout mice than that in wild-type mice and lower than that in UT-B knockout mice. Urine osmolality in UT-A2/B knockout mice was intermediate between that in UT-B knockout and wild-type mice. The changes in urine osmolality and flow rate, plasma and urine urea concentration, as well as non-urea solute concentration after an acute urea load or chronic changes in protein intake suggested that UT-A2 plays a role in the progressive accumulation of urea in the inner medulla. These results suggest that in wild-type mice UT-A2 facilitates urea absorption by urea efflux from the thin descending limb of short loops of Henle. Moreover, UT-A2 deletion in UT-B knockout mice partially remedies the urine concentrating defect caused by UT-B deletion, by reducing urea loss from the descending limbs to the peripheral circulation; instead, urea is returned to the inner medulla through the loops of Henle and the collecting ducts.
Collapse
Affiliation(s)
- Tianluo Lei
- Dept. of Pharmacology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Lu, Haidian District, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
106
|
Spector DA, Deng J, Stewart KJ. Hydration status affects urea transport across rat urothelia. Am J Physiol Renal Physiol 2011; 301:F1208-17. [PMID: 21900453 DOI: 10.1152/ajprenal.00386.2011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Although mammalian urinary tract epithelium (urothelium) is generally considered impermeable to water and solutes, recent data suggest that urine constituents may be reabsorbed during urinary tract transit and storage. To study water and solute transport across the urothelium in an in vivo rat model, we instilled urine (obtained during various rat hydration conditions) into isolated in situ rat bladders and, after a 1-h dwell, retrieved the urine and measured the differences in urine volume and concentration and total quantity of urine urea nitrogen and creatinine between instilled and retrieved urine in rat groups differing by hydration status. Although urine volume did not change >1.9% in any group, concentration (and quantity) of urine urea nitrogen in retrieved urine fell significantly (indicating reabsorption of urea across bladder urothelia), by a mean of 18% (489 mg/dl, from an instilled 2,658 mg/dl) in rats receiving ad libitum water and by a mean of 39% (2,544 mg/dl, from an instilled 6,204 mg/dl) in water-deprived rats, but did not change (an increase of 15 mg/dl, P = not significant, from an instilled 300 mg/dl) in a water-loaded rat group. Two separate factors affected urea nitrogen reabsorption rates, a urinary factor related to hydration status, likely the concentration of urea nitrogen in the instilled urine, and a bladder factor(s), also dependent on the animal's state of hydration. Urine creatinine was also absorbed during the bladder dwell, and hydration group effects on the concentration and quantity of creatinine reabsorbed were qualitatively similar to the hydration group effect on urea transport. These findings support the notion(s) that urinary constituents may undergo transport across urinary tract epithelia, that such transport may be physiologically regulated, and that urine is modified during transit and storage through the urinary tract.
Collapse
Affiliation(s)
- David A Spector
- Division of Renal Medicine, Johns Hopkins Bayview Medical Center, 4940 Eastern Ave., Baltimore, MD 21224, USA.
| | | | | |
Collapse
|
107
|
Devuyst O, Wang X, Serra A. Vasopressin-2 receptor antagonists in autosomal dominant polycystic kidney disease: from man to mouse and back. Nephrol Dial Transplant 2011; 26:2423-5. [DOI: 10.1093/ndt/gfr380] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
108
|
Mice lacking urea transporter UT-B display depression-like behavior. J Mol Neurosci 2011; 46:362-72. [PMID: 21750947 DOI: 10.1007/s12031-011-9594-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 07/01/2011] [Indexed: 12/25/2022]
Abstract
Urea transporter B is one of urea transporters that selectively transport urea driven by urea gradient across membrane and expressed abundantly in brain. To determine the physiological role of UT-B in brain, UT-B localization, urea concentration, tissue morphology of brain, and behavioral phenotypes were studied in UT-B heterozygous mice via UT-B null mice. UT-B mRNA was expressed in olfactory bulb, cortex, caudate nucleus, hippocampus and hypothalamus of UT-B heterozygous mice. UT-B null mice exhibited depression-like behavior, with urea accumulation, nitric oxide reduction, and selective neuronal nitric oxide synthase level increase in hippocampus. After acute urea loading, the urea level increased, NO production decreased in hippocampus from both types of mice. Moreover, urea level was higher, and NO concentration was lower consistently in UT-B null hippocampus than that in heterozygous hippocampus. In vitro, 25 mM urea inhibited NO production too. Furthermore, UT-B knockout induced a long-lasting notable decrease in regional cerebral blood flow and altered morphology, such as loss of neurons in CA3 region, swelling, and membranous myelin-like structure formation within myelinated and unmyelinated fibers in hippocampus. These results suggest that urea accumulation in the hippocampus induced by UT-B deletion can cause depression-like behavior, which possibly attribute to disturbance in NOS/NO system.
Collapse
|
109
|
Collins D, Walpole C, Ryan E, Winter D, Baird A, Stewart G. UT-B1 mediates transepithelial urea flux in the rat gastrointestinal tract. J Membr Biol 2011; 239:123-30. [PMID: 21127847 DOI: 10.1007/s00232-010-9331-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2010] [Accepted: 11/12/2010] [Indexed: 11/26/2022]
Abstract
The process of urea nitrogen salvaging plays a vital role in the symbiotic relationship between mammals and their intestinal bacteria. The first step in this process requires the movement of urea from the mammalian bloodstream into the gastrointestinal tract lumen via specialized proteins known as facilitative urea transporters. In this study, we examined both transepithelial urea fluxes and urea transporter protein abundance along the length of the rat gastrointestinal tract. Urea flux experiments that used rat gastrointestinal tissues showed significantly higher transepithelial urea transport was present in caecum and proximal colon (P < 0.01, n = 8, analysis of variance [ANOVA]). This large urea flux was significantly inhibited by 1,3,dimethylurea (P < 0.001, n = 8, ANOVA) and thiourea (P < 0.05, n = 6, unpaired t-test), both known blockers of facilitative urea transporters. Immunoblotting analysis failed to detect any UT-A protein within rat gastrointestinal tissue protein samples. In contrast, a 30-kDa UT-B1 protein was strongly detected in both caecum and proximal colon samples at significantly higher levels compared to the rest of the gastrointestinal tract (P < 0.01, n = 4, ANOVA). We therefore concluded that UT-B1 mediates the transepithelial movement of urea that occurs in specific distal regions of the rat gastrointestinal tract.
Collapse
Affiliation(s)
- Danielle Collins
- Institute for Clinical Outcomes Research and Education, St. Vincent's University Hospital, Dublin, Ireland
| | | | | | | | | | | |
Collapse
|
110
|
Neerathilingam M, Volk DE, Sarkar S, Alam TM, Alam MK, Ansari GS, Luxon BA. 1H NMR-based metabonomic investigation of tributyl phosphate exposure in rats. Toxicol Lett 2010; 199:10-6. [DOI: 10.1016/j.toxlet.2010.07.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2010] [Revised: 07/24/2010] [Accepted: 07/26/2010] [Indexed: 11/16/2022]
|
111
|
Erythrocyte permeability to urea and water: comparative study in rodents, ruminants, carnivores, humans, and birds. J Comp Physiol B 2010; 181:65-72. [DOI: 10.1007/s00360-010-0515-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Revised: 08/28/2010] [Accepted: 09/07/2010] [Indexed: 10/19/2022]
|
112
|
Hardwicke J, Jones E, Wilson-Jones N. Optimization of silicone urinary catheters for hypospadias repair. J Pediatr Urol 2010; 6:385-8. [PMID: 19897421 DOI: 10.1016/j.jpurol.2009.10.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2009] [Accepted: 10/12/2009] [Indexed: 10/20/2022]
Abstract
OBJECTIVE We have experienced difficulty with the removal of all-silicone Foley catheters after hypospadias repair, relating to the formation of a 'cuff' of residual balloon material that fails to deflate, after aspiration of the instilled volume of water. This could potentially lead to both short- and long-term complications (stenosis, fistula). In all-silicone paediatric catheters, we investigated the production of such 'cuffs', and any other significant deformity which may be associated with deformation of the catheter balloon mechanism, in vitro. MATERIALS AND METHODS Catheters were inflated with 0 (control) to 7 mL of sterile water. The catheter balloon dimensions were measured before and after incubation of the catheters for 168 h in a solution simulating human urine. The aspiration volumes were recorded. RESULTS At volumes greater than 40% of the manufacturer's advised inflation volume, a significant increase in the transverse diameter of the catheter occurred after deflation, compared to controls (P<0.001). CONCLUSION We advise the maximum instillation of 2 mL of water into a 5-mL paediatric catheter balloon to avoid cuff formation. Using this technique allows all of the advantages of a stent, in combination with the security of a catheter, but with a reduced risk of trauma and complications associated with catheter removal.
Collapse
Affiliation(s)
- Joseph Hardwicke
- Welsh Centre for Burns and Plastic Surgery, Abertawe Bro Morgannwg NHS Trust, Morriston Hospital, Swansea, UK.
| | | | | |
Collapse
|
113
|
Kemter E, Rathkolb B, Bankir L, Schrewe A, Hans W, Landbrecht C, Klaften M, Ivandic B, Fuchs H, Gailus-Durner V, Hrabé de Angelis M, Wolf E, Wanke R, Aigner B. Mutation of the Na+-K+-2Cl−cotransporter NKCC2 in mice is associated with severe polyuria and a urea-selective concentrating defect without hyperreninemia. Am J Physiol Renal Physiol 2010; 298:F1405-15. [DOI: 10.1152/ajprenal.00522.2009] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The bumetanide-sensitive Na+-K+-2Cl−cotransporter NKCC2, located in the thick ascending limb of Henle's loop, plays a critical role in the kidney's ability to concentrate urine. In humans, loss-of-function mutations of the solute carrier family 12 member 1 gene ( SLC12A1), coding for NKCC2, cause type I Bartter syndrome, which is characterized by prenatal onset of a severe polyuria, salt-wasting tubulopathy, and hyperreninemia. In this study, we describe a novel chemically induced, recessive mutant mouse line termed Slc12a1I299Fexhibiting late-onset manifestation of type I Bartter syndrome. Homozygous mutant mice are viable and exhibit severe polyuria, metabolic alkalosis, marked increase in plasma urea but close to normal creatininemia, hypermagnesemia, hyperprostaglandinuria, hypotension,, and osteopenia. Fractional excretion of urea is markedly decreased. In addition, calcium and magnesium excretions are more than doubled compared with wild-type mice, while uric acid excretion is twofold lower. In contrast to hyperreninemia present in human disease, plasma renin concentration in homozygotes is not increased. The polyuria observed in homozygotes may be due to the combination of two additive factors, a decrease in activity of mutant NKCC2 and an increase in medullary blood flow, due to prostaglandin-induced vasodilation, that impairs countercurrent exchange of urea in the medulla. In conclusion, this novel viable mouse line with a missense Slc12a1 mutation exhibits most of the features of type I Bartter syndrome and may represent a new model for the study of this human disease.
Collapse
Affiliation(s)
- Elisabeth Kemter
- Chair for Molecular Animal Breeding and Biotechnology, and Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, and
- Institute of Veterinary Pathology, Center for Clinical Veterinary Medicine, LMU Munich, Munich
| | - Birgit Rathkolb
- Chair for Molecular Animal Breeding and Biotechnology, and Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, and
| | - Lise Bankir
- INSERM Unité 872, Centre de Recherche des Cordeliers, Paris, France
| | - Anja Schrewe
- Department of Medicine III, Division of Cardiology, University of Heidelberg, Heidelberg; and
- Institute of Experimental Genetics, Helmholtz Zentrum München, Neuherberg, and Chair for Experimental Genetics, Technische Universität München, Munich, Germany
| | - Wolfgang Hans
- Institute of Experimental Genetics, Helmholtz Zentrum München, Neuherberg, and Chair for Experimental Genetics, Technische Universität München, Munich, Germany
| | - Christina Landbrecht
- Chair for Molecular Animal Breeding and Biotechnology, and Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, and
| | - Matthias Klaften
- Institute of Experimental Genetics, Helmholtz Zentrum München, Neuherberg, and Chair for Experimental Genetics, Technische Universität München, Munich, Germany
| | - Boris Ivandic
- Department of Medicine III, Division of Cardiology, University of Heidelberg, Heidelberg; and
| | - Helmut Fuchs
- Institute of Experimental Genetics, Helmholtz Zentrum München, Neuherberg, and Chair for Experimental Genetics, Technische Universität München, Munich, Germany
| | - Valérie Gailus-Durner
- Institute of Experimental Genetics, Helmholtz Zentrum München, Neuherberg, and Chair for Experimental Genetics, Technische Universität München, Munich, Germany
| | - Martin Hrabé de Angelis
- Institute of Experimental Genetics, Helmholtz Zentrum München, Neuherberg, and Chair for Experimental Genetics, Technische Universität München, Munich, Germany
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology, and Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, and
| | - Ruediger Wanke
- Institute of Veterinary Pathology, Center for Clinical Veterinary Medicine, LMU Munich, Munich
| | - Bernhard Aigner
- Chair for Molecular Animal Breeding and Biotechnology, and Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, and
| |
Collapse
|
114
|
Ashok PC, Singh GP, Tan KM, Dholakia K. Fiber probe based microfluidic raman spectroscopy. OPTICS EXPRESS 2010; 18:7642-7649. [PMID: 20588604 DOI: 10.1364/oe.18.007642] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
We report a novel fiber probe based Raman detection system on a microfluidic platform where a split Raman probe is directly embedded into a polydimethylsiloxane (PDMS) chip. In contrast to previous Raman detection schemes in microfluidics, probe based detection offers reduced background and portability. Compared to conventional backscattering probe designs, the split fiber probe we used in this system, results in a reduced size and offers flexibility to modify the collection geometry to minimize the background generated by the fibers. Also our microfluidic chip design enables us to obtain an alignment free system. As a proof of concept we demonstrate the sensitivity of the device for urea detection at relevant human physiological levels with a low acquisition time. The development of this system on a microfluidic platform means portable, lab on a chip devices for biological analyte detection and environmental sensing using Raman spectroscopy are now within reach.
Collapse
Affiliation(s)
- P C Ashok
- SUPA, School of Physics and Astronomy, University of St Andrews, North Haugh, Fife, KY16 9SS, UK
| | | | | | | |
Collapse
|
115
|
Poirrier A, Van den Ackerveken P, Kim T, Vandenbosch R, Nguyen L, Lefebvre P, Malgrange B. Ototoxic drugs: Difference in sensitivity between mice and guinea pigs. Toxicol Lett 2010; 193:41-9. [DOI: 10.1016/j.toxlet.2009.12.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2009] [Revised: 12/02/2009] [Accepted: 12/06/2009] [Indexed: 01/18/2023]
|
116
|
Abdoun K, Stumpff F, Rabbani I, Martens H. Modulation of urea transport across sheep rumen epithelium in vitro by SCFA and CO2. Am J Physiol Gastrointest Liver Physiol 2010; 298:G190-202. [PMID: 19926818 DOI: 10.1152/ajpgi.00216.2009] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Urea transport across the gastrointestinal tract involves transporters of the urea transporter-B group, the regulation of which is poorly understood. The classical stimulatory effect of CO(2) and the effect of short-chain fatty acids (SCFA) on the ruminal recycling of urea were investigated by using Ussing chamber and microelectrode techniques with isolated ruminal epithelium of sheep. The flux of urea was found to be phloretin sensitive and passive. At a luminal pH of 6.4, but not at 7.4, the addition of SCFA (40 mmol/l) or CO(2)/HCO3- (10% and 25 mmol/l) led to a fourfold increase in urea flux. The stepwise reduction of luminal pH in the presence of SCFA from 7.4 to 5.4 led to a bell-shaped modification of urea transport, with a maximum at pH 6.2. Lowering the pH in the absence of SCFA or CO(2) had no effect. Inhibition of Na(+)/H(+) exchange increased urea flux at pH 7.4, with a decrease being seen at pH 6.4. In experiments with double-barreled, pH-sensitive microelectrodes, we confirmed the presence of an apical pH microclimate and demonstrated the acidifying effects of SCFA on the underlying epithelium. We confirm that the permeability of the ruminal epithelium to urea involves a phloretin-sensitive pathway. We present clear evidence for the regulation of urea transport by strategies that alter intracellular pH, with permeability being highest after a moderate decrease. The well-known postprandial stimulation of urea transport to the rumen in vivo may involve acute pH-dependent effects of intraruminal SCFA and CO(2) on the function of existing urea transporters.
Collapse
Affiliation(s)
- Khalid Abdoun
- Dept. of Veterinary Physiology, Free University of Berlin, Oertzenweg 19b, Berlin, Germany
| | | | | | | |
Collapse
|
117
|
Torres VE. Vasopressin in chronic kidney disease: an elephant in the room? Kidney Int 2009; 76:925-8. [PMID: 19829311 DOI: 10.1038/ki.2009.325] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Perico et al. report that a dual arginine vasopressin (AVP) V(2) and V(1a) receptor antagonist lowers blood pressure, proteinuria, and glomerulosclerosis in 5/6 nephrectomized rats, pointing to its potential value in the treatment of chronic kidney disease (CKD). AVP likely contributes to CKD progression by its effects on renal hemodynamics, blood pressure, and mesangial and/or epithelial cells, but the relative contributions of V(2) and V(1a) receptors and potential usefulness of V(2) and V(1a) receptor antagonists remain ill defined.
Collapse
|
118
|
Kemter E, Rathkolb B, Rozman J, Hans W, Schrewe A, Landbrecht C, Klaften M, Ivandic B, Fuchs H, Gailus-Durner V, Klingenspor M, de Angelis MH, Wolf E, Wanke R, Aigner B. Novel missense mutation of uromodulin in mice causes renal dysfunction with alterations in urea handling, energy, and bone metabolism. Am J Physiol Renal Physiol 2009; 297:F1391-8. [DOI: 10.1152/ajprenal.00261.2009] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Uromodulin-associated kidney disease is a heritable renal disease in humans caused by mutations in the uromodulin ( UMOD) gene. The pathogenesis of the disease is mostly unknown. In this study, we describe a novel chemically induced mutant mouse line termed UmodA227T exhibiting impaired renal function. The A227T amino acid exchange may impair uromodulin trafficking, leading to dysfunction of thick ascending limb cells of Henle's loop of the kidney. As a consequence, homozygous mutant mice display azotemia, impaired urine concentration ability, reduced fractional excretion of uric acid, and a selective defect in concentrating urea. Osteopenia in mutant mice is presumably a result of chronic hypercalciuria. In addition, body composition, lipid, and energy metabolism are indirectly affected in heterozygous and homozygous mutant UmodA227T mice, manifesting in reduced body weight, fat mass, and metabolic rate as well as reduced blood cholesterol, triglycerides, and nonesterified fatty acids. In conclusion, UmodA227T might act as a gain-of-toxic-function mutation. Therefore, the UmodA227T mouse line provides novel insights into consequences of disturbed uromodulin excretion regarding renal dysfunction as well as bone, energy, and lipid metabolism.
Collapse
Affiliation(s)
- Elisabeth Kemter
- Chair for Molecular Animal Breeding and Biotechnology and Laboratory for Functional Genome Analysis, Gene Center, and
| | - Birgit Rathkolb
- Chair for Molecular Animal Breeding and Biotechnology and Laboratory for Functional Genome Analysis, Gene Center, and
| | - Jan Rozman
- Molecular Nutricial Medicine, Else-Kröner-Fresenius Center, Technische Universität München, Freising-Weihenstephan
| | - Wolfgang Hans
- Institute of Experimental Genetics, Helmholtz Zentrum München, Neuherberg, and Chair for Experimental Genetics, Technische Universität München, Munich; and
| | - Anja Schrewe
- Department of Medicine III, Division of Cardiology, University of Heidelberg, Heidelberg, Germany
| | - Christina Landbrecht
- Chair for Molecular Animal Breeding and Biotechnology and Laboratory for Functional Genome Analysis, Gene Center, and
| | - Matthias Klaften
- Institute of Experimental Genetics, Helmholtz Zentrum München, Neuherberg, and Chair for Experimental Genetics, Technische Universität München, Munich; and
| | - Boris Ivandic
- Department of Medicine III, Division of Cardiology, University of Heidelberg, Heidelberg, Germany
| | - Helmut Fuchs
- Institute of Experimental Genetics, Helmholtz Zentrum München, Neuherberg, and Chair for Experimental Genetics, Technische Universität München, Munich; and
| | - Valérie Gailus-Durner
- Institute of Experimental Genetics, Helmholtz Zentrum München, Neuherberg, and Chair for Experimental Genetics, Technische Universität München, Munich; and
| | - Martin Klingenspor
- Molecular Nutricial Medicine, Else-Kröner-Fresenius Center, Technische Universität München, Freising-Weihenstephan
| | - Martin Hrabé de Angelis
- Institute of Experimental Genetics, Helmholtz Zentrum München, Neuherberg, and Chair for Experimental Genetics, Technische Universität München, Munich; and
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology and Laboratory for Functional Genome Analysis, Gene Center, and
| | - Ruediger Wanke
- Institute of Veterinary Pathology, Center for Clinical Veterinary Medicine, Ludwig-Maximilians-Universität Munich, Munich
| | - Bernhard Aigner
- Chair for Molecular Animal Breeding and Biotechnology and Laboratory for Functional Genome Analysis, Gene Center, and
| |
Collapse
|
119
|
Song MF, Li YS, Ootsuyama Y, Kasai H, Kawai K, Ohta M, Eguchi Y, Yamato H, Matsumoto Y, Yoshida R, Ogawa Y. Urea, the most abundant component in urine, cross-reacts with a commercial 8-OH-dG ELISA kit and contributes to overestimation of urinary 8-OH-dG. Free Radic Biol Med 2009; 47:41-6. [PMID: 19264122 DOI: 10.1016/j.freeradbiomed.2009.02.017] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2008] [Revised: 02/16/2009] [Accepted: 02/20/2009] [Indexed: 11/29/2022]
Abstract
Urinary 8-OH-dG is commonly analyzed as a marker of oxidative stress. For its analysis, ELISA and HPLC methods are generally used, although discrepancies in the data obtained by these methods have often been discussed. To clarify this problem, we fractionated human urine by reverse-phase HPLC and assayed each fraction by the ELISA method. In addition to the 8-OH-dG fraction, a positive reaction was observed in the first eluted fraction. The components in this fraction were examined by the ELISA. Urea was found to be the responsible component in this fraction. Urea is present in high concentrations in the urine of mice, rats, and humans, and its level is influenced by many factors. Therefore, certain improvements, such as a correction based on urea content or urease treatment, are required for the accurate analysis of urinary 8-OH-dG by the ELISA method. In addition, performance of the ELISA at 4 degrees C reduced the recognition of urea considerably and improved the 8-OH-dG analysis.
Collapse
Affiliation(s)
- Ming-Fen Song
- Department of Environmental Oncology, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
120
|
Torres VE, Bankir L, Grantham JJ. A case for water in the treatment of polycystic kidney disease. Clin J Am Soc Nephrol 2009; 4:1140-50. [PMID: 19443627 DOI: 10.2215/cjn.00790209] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Autosomal dominant polycystic disease (ADPKD) is an inherited disorder characterized by the development within renal tubules of innumerable cysts that progressively expand to cause renal insufficiency. Tubule cell proliferation and transepithelial fluid secretion combine to enlarge renal cysts, and 3'-5'-cyclic adenosine monophosphate (cAMP) stimulates that growth. The antidiuretic hormone, arginine vasopressin (AVP), operates continuously in ADPKD patients to stimulate the formation of cAMP, thereby contributing to cyst and kidney enlargement and renal dysfunction. Studies in animal models of ADPKD provide convincing evidence that blocking the action of AVP dramatically ameliorates the disease process. In the current analysis, the authors reason that increasing the amount of solute-free water drunk evenly throughout the day in patients with ADPKD and normal renal function will decrease plasma AVP concentrations and mitigate the action of cAMP on the renal cysts. Potential pitfalls of increasing fluid intake in ADPKD patients are considered, and suggestions for how physicians may prudently implement this therapy are offered.
Collapse
Affiliation(s)
- Vicente E Torres
- Mayo Clinic College of Medicine, Rochester, Minnesota 55905, USA.
| | | | | |
Collapse
|
121
|
Fenton RA. Essential role of vasopressin-regulated urea transport processes in the mammalian kidney. Pflugers Arch 2009; 458:169-77. [PMID: 19011892 DOI: 10.1007/s00424-008-0612-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2008] [Revised: 10/17/2008] [Accepted: 10/23/2008] [Indexed: 10/21/2022]
Abstract
Movement of urea across plasma membranes is modulated by specialized urea transporter proteins. Two urea-transporter genes have been cloned: UT-A (Slc14a2) and UT-B (Slc14a1). In the mammalian kidney, urea transporters are essential for the urinary concentrating mechanism and maintaining body fluid homeostasis. In this article, we discuss (1) an overview of historic discoveries in urea transport mechanisms; (2) an overview of recent discoveries in the regulation of urea transporters; (3) physiological studies in UT-A1/3 (-/-) mice highlighting the essential role of urea transporters in the urinary concentrating mechanism; and (4) physiological studies in UT-A2 and UT-B knockout mice examining the role of countercurrent exchange in the production of a maximally concentrated urine.
Collapse
Affiliation(s)
- Robert A Fenton
- The Water and Salt Research Center, Institute of Anatomy, University of Aarhus, Denmark.
| |
Collapse
|
122
|
Meng Y, Zhao C, Zhang X, Zhao H, Guo L, Lü B, Zhao X, Yang B. Surface electrocardiogram and action potential in mice lacking urea transporter UT-B. SCIENCE IN CHINA. SERIES C, LIFE SCIENCES 2009; 52:474-478. [PMID: 19471871 DOI: 10.1007/s11427-009-0047-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2008] [Accepted: 07/29/2008] [Indexed: 11/26/2022]
Abstract
UT-B is a urea transporter protein expressed in the kidney and in many non-renal tissues including erythrocytes, brain, heart, bladder and the testis. The objective of this study was to determine the phenotype of UT-B deletion in the heart. UT-B expression in the heart was studied in wild-type mice vs UT-B null mice by utilizing RT-PCR and Western blot. A surface electrocardiogram (ECG) recording (lead II) was measured in wild-type mice and UT-B null mice at the ages of 6, 16 and 52 weeks. For the action potential recording, the ventricular myocytes of 16 w mice were isolated and recorded by floating microelectrode method. The sodium current was recorded by the patch clamp technique. RT-PCR and Western blot showed the UT-B expression in the heart of wild-type mice. No UT-B transcript and protein was found in UT-B null mice. The ECG recording showed that the P-R interval was significantly prolonged in UT-B null mice ((43.5 +/- 4.2), (45.5 +/- 6.9) and (43.8 +/- 7.6) ms at ages of 6, 16 and 52 weeks) vs wild-type mice ((38.6 +/- 2.9), (38.7 +/- 5.6) and (38.2 +/- 7.3) ms, P<0.05). The atrial ventricular heart block type II and III only appeared in the aging UT-B null mice (52 w old). The amplitude of action potential and V (max) decreased significantly in UT-B null mice ((92.17 +/- 10.56) and (101.89 +/- 9.54) mV/s) vs those in wild-type mice (vs (110.51 +/- 10.38) and (109.53 +/- 10.64) mV/s, P<0.05). The action potential duration at 50% and 90% (APD(50) and APD(90)) was significantly prolonged in UT-B null mice ((123.83 +/- 11.17) and (195.43 +/- 16.41) ms) vs that in wild-type mice ((108.27 +/- 10.85) and (171.00 +/- 15.53) ms, P<0.05). The maximal sodium current decreased significantly in UT-B null mice (-8.80 +/- 0.92) nA vs that in wild-type mice ((-5.98 +/- 1.07) nA, P<0.05). These results provide the first evidence that UT-B deletion causes progressive heart block in mice.
Collapse
Affiliation(s)
- Yan Meng
- Department of Pathophysiology, School of Basic Medicine, Jilin University, Changchun, 130021, China
| | | | | | | | | | | | | | | |
Collapse
|
123
|
Yan H, Gu W, Yang J, Bi V, Shen Y, Lee E, Winters KA, Komorowski R, Zhang C, Patel JJ, Caughey D, Elliott GS, Lau YY, Wang J, Li YS, Boone T, Lindberg RA, Hu S, Véniant MM. Fully human monoclonal antibodies antagonizing the glucagon receptor improve glucose homeostasis in mice and monkeys. J Pharmacol Exp Ther 2009; 329:102-11. [PMID: 19129372 DOI: 10.1124/jpet.108.147009] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Antagonizing the glucagon signaling pathway represents an attractive therapeutic approach for reducing excess hepatic glucose production in patients with type 2 diabetes. Despite extensive efforts, there is currently no human therapeutic that directly inhibits the glucagon/glucagon receptor pathway. We undertook a novel approach by generating high-affinity human monoclonal antibodies (mAbs) to the human glucagon receptor (GCGR) that display potent antagonistic activity in vitro and in vivo. A single injection of a lead antibody, mAb B, at 3 mg/kg, normalized blood glucose levels in ob/ob mice for 8 days. In addition, a single injection of mAb B dose-dependently lowered fasting blood glucose levels without inducing hypoglycemia and improved glucose tolerance in normal C57BL/6 mice. In normal cynomolgus monkeys, a single injection improved glucose tolerance while increasing glucagon and active glucagon-like peptide-1 levels. Thus, the anti-GCGR mAb could represent an effective new therapeutic for the treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Hai Yan
- Department of Protein Sciences, Amgen Inc., Thousand Oaks, CA 91320, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
124
|
Bhasin KKS, van Nas A, Martin LJ, Davis RC, Devaskar SU, Lusis AJ. Maternal low-protein diet or hypercholesterolemia reduces circulating essential amino acids and leads to intrauterine growth restriction. Diabetes 2009; 58:559-66. [PMID: 19073773 PMCID: PMC2646054 DOI: 10.2337/db07-1530] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
OBJECTIVE We have examined maternal mechanisms for adult-onset glucose intolerance, increased adiposity, and atherosclerosis using two mouse models for intrauterine growth restriction (IUGR): maternal protein restriction and hypercholesterolemia. RESEARCH DESIGN AND METHODS For these studies, we measured the amino acid levels in dams from two mouse models for IUGR: 1) feeding C57BL/6J dams a protein-restricted diet and 2) feeding C57BL/6J LDL receptor-null (LDLR(-/-)) dams a high-fat (Western) diet. RESULTS Both protein-restricted and hypercholesterolemic dams exhibited significantly decreased concentrations of the essential amino acid phenylalanine and the essential branched chain amino acids leucine, isoleucine, and valine. The protein-restricted diet for pregnant dams resulted in litters with significant IUGR. Protein-restricted male offspring exhibited catch-up growth by 8 weeks of age and developed increased adiposity and glucose intolerance by 32 weeks of age. LDLR(-/-) pregnant dams on a Western diet also had litters with significant IUGR. Male and female LDLR(-/-) Western-diet offspring developed significantly larger atherosclerotic lesions by 90 days compared with chow-diet offspring. CONCLUSIONS In two mouse models of IUGR, we found reduced concentrations of essential amino acids in the experimental dams. This indicated that shared mechanisms may underlie the phenotypic effects of maternal hypercholesterolemia and maternal protein restriction on the offspring.
Collapse
Affiliation(s)
- Kum Kum S Bhasin
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | | | | | | | | | | |
Collapse
|
125
|
Yu H, Meng Y, Wang LS, Jin X, Gao LF, Zhou L, Ji K, Li Y, Zhao LJ, Chen GQ, Zhao XJ, Yang B. Differential protein expression in heart in UT-B null mice with cardiac conduction defects. Proteomics 2009; 9:504-11. [PMID: 19132680 DOI: 10.1002/pmic.200701079] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Cardiac conduction defects were found in transgenic mice deficient in urea transporter UT-B. To investigate the molecular mechanisms of the conduction defects caused by UT-B deletion, we studied the protein expression profiles of heart tissue (comprising most conduction system) in wild-type versus UT-B null mice at different ages. By two-dimensional electrophoresis-based comparative analysis, we found that more than dozen proteins were modulated (>two-fold) in the myocardium of UT-B null mice. Out of these modulated proteins, troponin T (TNNT2) presented significant changes in UT-B null mice at early stage prior to the development of P-R interval elongation, while the change of atrial natriuretic peptide (ANP) occurred only at late stage in UT-B null mice that had the AV block. These data indicate that UT-B deletion caused the dynamic expression regulation of TNNT2 and ANP, and these proteins may provide new clues to investigate the molecular events involved in cardiac conduction.
Collapse
Affiliation(s)
- Hao Yu
- Department of Pathophysiology, Research Center of Prostate Diseases, School of Basic Medicine, Jilin University, Changchun, P. R. China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
126
|
Abstract
Urea transporters (UTs) encoded by the Slc14a1 (UT-B) and Slc14a2 (UT-A) genes mediate urea flux across cellular membranes. Considerable research has accrued detailing the function and distribution of members of both subfamilies. Much research effort has focused on the kidney, where UTs are highly expressed and function to promote urine concentration. Interestingly, UTs are also expressed in several other tissues that are historically not primarily associated with urea metabolism. In this review, I describe the phenotypes of UT knockout and transgenic mice and highlight the major advances made possible by use of these animal models. Where pertinent, I contrast these findings with known human phenotypes associated with UT mutations.
Collapse
|
127
|
Sohara E, Uchida S, Sasaki S. Function of aquaporin-7 in the kidney and the male reproductive system. Handb Exp Pharmacol 2008:219-31. [PMID: 19096780 DOI: 10.1007/978-3-540-79885-9_11] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
The aquaporin-7 (AQP7) water channel is known to be a member of the aquaglyceroporins, which allow the rapid transport of glycerol and water. In this chapter, we review the physiological functions of AQP7 in the kidney and the male reproductive system.In the kidney, AQP7 is abundantly present at the apical membrane of the proximal straight tubules. Although the contribution of AQP7 to the water permeability of proximal straight tubules was found to be minimal compared with that of AQP1, we identified a novel glycerol reabsorption pathway that may be important for preventing glycerol from being excreted into urine.In the male reproductive system, AQP7 is present particularly in the spermatids, as well as in the testicular and epididymal spermatozoa, suggesting that AQP7 has some role in late spermatogenesis. However, male AQP7 knockout mice were not sterile, and their sperm did not show any morphological or functional abnormalities.
Collapse
Affiliation(s)
- Eisei Sohara
- Department of Nephrology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | | | | |
Collapse
|
128
|
Navar LG, Arendshorst WJ, Pallone TL, Inscho EW, Imig JD, Bell PD. The Renal Microcirculation. Compr Physiol 2008. [DOI: 10.1002/cphy.cp020413] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
129
|
Abstract
The purpose of this review is first to describe the importance of early detection of vasopressin receptor mutations responsible for X-linked nephrogenic diabetes insipidus (NDI). We have proposed that all families with hereditary diabetes insipidus should have their molecular defect identified because early diagnosis and treatment of affected infants can avert the physical and mental retardation that results from repeated episodes of dehydration. Secondly, 95 published missense mutations responsible for X-linked NDI are likely to result in misfolded arginine-vasopressin V(2) receptors that are trapped in the endoplasmic reticulum. These misfolded receptors are unable to reach the plasma membrane in principal collecting duct cells and to engage the circulating antidiuretic hormone, arginine-vasopressin. These misfolded proteins potentially could be rescued with pharmacologic chaperones, an active area of research pertinent to other hereditary protein misfolding diseases such as cystic fibrosis, phenylketonuria, and Anderson-Fabry disease among many others. Finally, a long-term careful surveillance of all patients with hereditary NDI should be performed to prevent chronic renal failure likely caused by the long-term functional tract obstruction with reflux.
Collapse
|
130
|
Pannabecker TL, Dantzler WH, Layton HE, Layton AT. Role of three-dimensional architecture in the urine concentrating mechanism of the rat renal inner medulla. Am J Physiol Renal Physiol 2008; 295:F1271-85. [PMID: 18495796 PMCID: PMC2584911 DOI: 10.1152/ajprenal.90252.2008] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2008] [Accepted: 05/19/2008] [Indexed: 11/22/2022] Open
Abstract
Recent studies of three-dimensional architecture of rat renal inner medulla (IM) and expression of membrane proteins associated with fluid and solute transport in nephrons and vasculature have revealed structural and transport properties that likely impact the IM urine concentrating mechanism. These studies have shown that 1) IM descending thin limbs (DTLs) have at least two or three functionally distinct subsegments; 2) most ascending thin limbs (ATLs) and about half the ascending vasa recta (AVR) are arranged among clusters of collecting ducts (CDs), which form the organizing motif through the first 3-3.5 mm of the IM, whereas other ATLs and AVR, along with aquaporin-1-positive DTLs and urea transporter B-positive descending vasa recta (DVR), are external to the CD clusters; 3) ATLs, AVR, CDs, and interstitial cells delimit interstitial microdomains within the CD clusters; and 4) many of the longest loops of Henle form bends that include subsegments that run transversely along CDs that lie in the terminal 500 microm of the papilla tip. Based on a more comprehensive understanding of three-dimensional IM architecture, we distinguish two distinct countercurrent systems in the first 3-3.5 mm of the IM (an intra-CD cluster system and an inter-CD cluster system) and a third countercurrent system in the final 1.5-2 mm. Spatial arrangements of loop of Henle subsegments and multiple countercurrent systems throughout four distinct axial IM zones, as well as our initial mathematical model, are consistent with a solute-separation, solute-mixing mechanism for concentrating urine in the IM.
Collapse
Affiliation(s)
- Thomas L Pannabecker
- Department of Physiology, College of Medicine, University of Arizona, Tucson, AZ, USA.
| | | | | | | |
Collapse
|
131
|
Lahm T, Crisostomo PR, Markel TA, Wang M, Wang Y, Tan J, Meldrum DR. Selective estrogen receptor-alpha and estrogen receptor-beta agonists rapidly decrease pulmonary artery vasoconstriction by a nitric oxide-dependent mechanism. Am J Physiol Regul Integr Comp Physiol 2008; 295:R1486-93. [PMID: 18832085 DOI: 10.1152/ajpregu.90667.2008] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Both endogenous and exogenous estrogen decrease pulmonary artery (PA) vasoconstriction. Whether these effects are mediated via estrogen receptor (ER)-alpha or ER-beta, and whether the contribution of ERs is stimulus-dependent, remains unknown. We hypothesized that administration of the selective ER-alpha agonist propylpyrazole triol (PPT) and/or the selective ER-beta agonist diarylpropiolnitrile (DPN) rapidly decreases PA vasoconstriction induced by pharmacologic and hypoxic stimuli via a nitric oxide (NO)-dependent mechanism. PA rings (n = 3-10/group) from adult male Sprague-Dawley rats were suspended in physiologic organ baths. Force displacement was measured. Vasoconstrictor responses to phenylephrine (10(-8)M - 10(-5)M) and hypoxia (Po(2) 35-45 mmHg) were determined. Endothelium-dependent and -independent vasorelaxation were measured by generating dose-response curves to acetylcholine (10(-8)M - 10(-4)M) and sodium nitroprusside (10(-9)M - 10(-5)M). PPT or DPN (10(-9)M - 5 x 10(-5)M) were added to the organ bath in the presence and absence of the NO-synthase inhibitor N(omega)-nitro-l-arginine methyl ester (l-NAME) (10(-4)M). Selective ER-alpha activation (PPT, 5 x 10(-5)M) rapidly (<20 min) decreased phenylephrine-induced vasoconstriction. This effect, as well as PPT's effects on endothelium-dependent vasorelaxation, were neutralized by l-NAME. In contrast, selective ER-beta activation (DPN, 5 x 10(-5)M) rapidly decreased phase II of hypoxic pulmonary vasoconstriction (HPV). l-NAME eliminated this phenomenon. Lower PPT or DPN concentrations were less effective. We conclude that both ER-alpha and ER-beta decrease PA vasoconstriction. The immediate onset of effect suggests a nongenomic mechanism. The contribution of specific ERs appears to be stimulus specific, with ER-alpha primarily modulating phenylephrine-induced vasoconstriction, and ER-beta inhibiting HPV. NO inhibition eliminates these effects, suggesting a central role for NO in mediating the pulmonary vascular effects of both ER-alpha and ER-beta.
Collapse
Affiliation(s)
- Tim Lahm
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | | | | | | | | | | |
Collapse
|
132
|
Abstract
PURPOSE OF REVIEW Gene knockout mice have been created for the collecting duct urea transporters UT-A1 and UT-A3, the descending thin-limb urea transporter UT-A2 and the descending vasa recta isoform, UT-B. In this brief review, the new insights in our understanding of the role of urea in the urinary concentrating mechanism and kidney function resulting from studies in these mice are discussed. RECENT FINDINGS The major findings in studies on urea transporter knockout mice are as follows: rapid transport of urea from the inner medulla collecting duct lumen via UT-A1 or UT-A3 is essential for urea accumulation in the inner medullary interstitium; inner medulla collecting duct urea transporters are essential in water conservation by preventing urea-induced osmotic diuresis; an absence of inner medulla collecting duct urea transport does not prevent the concentration of sodium chloride in the inner medulla interstitium; deletion of the vasa recta isoform UT-B has a much greater effect on urinary concentration than deleting the descending limb isoform UT-A2. SUMMARY Multiple urea transport mechanisms within the kidney are essential for producing maximally concentrated urine.
Collapse
Affiliation(s)
- Robert A Fenton
- Water and Salt Research Center, Institute of Anatomy, University of Aarhus, Aarhus, Denmark.
| |
Collapse
|
133
|
Schnöckel U, Reuter S, Stegger L, Schlatter E, Schäfers KP, Hermann S, Schober O, Gabriëls G, Schäfers M. Dynamic 18F-fluoride small animal PET to noninvasively assess renal function in rats. Eur J Nucl Med Mol Imaging 2008; 35:2267-74. [PMID: 18622612 DOI: 10.1007/s00259-008-0878-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2007] [Accepted: 06/23/2008] [Indexed: 12/23/2022]
Abstract
PURPOSE Renal function can be quantified by both laboratory and scintigraphic methods. In the case of small animal diagnostics, scintigraphic image-based methods are ideal since they can assess split renal function, work noninvasively, and can be repeated. The aim of this study is to validate a (18)F-PET-based method to quantify renal function in rats. MATERIALS AND METHODS Fluoride clearance was calculated from a dynamic whole body listmode acquisition of 60 min length in a small animal PET scanner following an i.v. injection of 15 MBq (18)F-fluoride. Volumes of interest (VOIs) were placed in the left ventricle and the bladder as well as traced around the kidney contours. The respective time-activity curves (TAC) were calculated. The renal (18)F-clearance was calculated by the ratio of the total renal excreted activity (bladder VOI) and the integral of the blood TAC. PET-derived renal function was validated by intraindividual measurements of creatinine clearance (n = 23), urea clearance (n = 23), and tubular excretion rate (TER-MAG3). The split renal function was derived from the injection of the clinically available radionuclide (99m)Tc-mercaptotriglycine by blood sampling and planar renography (n = 8). RESULTS In all animals studied, PET revealed high-quality TACs. PET-derived renal fluoride clearance was linearly correlated with intraindividual laboratory measures (PET vs. creatinine: r = 0.78; PET vs. urea: r = 0.73; PET vs. TER-MAG3: r = 0.73). Split function was comparable ((18)F-PET vs. MAG3-renography: r = 0.98). PET-derived measures were highly reproducible. CONCLUSIONS (18)F-PET is able to noninvasively assess renal function in rats and provides a significant potential for serial studies in different experimental scenarios.
Collapse
Affiliation(s)
- Uta Schnöckel
- Klinik und Poliklinik für Nuklearmedizin, 48149 Münster, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
134
|
Pépin MN, Bouchard J, Legault L, Éthier J. In Reply. Am J Kidney Dis 2008. [DOI: 10.1053/j.ajkd.2008.02.311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
135
|
Navar LG, Arendshorst WJ, Pallone TL, Inscho EW, Imig JD, Bell PD. The Renal Microcirculation. Microcirculation 2008. [DOI: 10.1016/b978-0-12-374530-9.00015-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
|
136
|
Zhao D, Sonawane ND, Levin MH, Yang B. Comparative transport efficiencies of urea analogues through urea transporter UT-B. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2007; 1768:1815-21. [PMID: 17506977 DOI: 10.1016/j.bbamem.2007.04.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2006] [Revised: 03/06/2007] [Accepted: 04/10/2007] [Indexed: 11/23/2022]
Abstract
Expression of urea transporter UT-B confers high urea permeability to mammalian erythrocytes. Erythrocyte membranes also permeate various urea analogues, suggesting common transport pathways for urea and structurally similar solutes. In this study, we examined UT-B-facilitated passage of urea analogues and other neutral small solutes by comparing transport properties of wildtype to UT-B-deficient mouse erythrocytes. Stopped-flow light-scattering measurements indicated high UT-B permeability to urea and chemical analogues formamide, acetamide, methylurea, methylformamide, ammonium carbamate, and acrylamide, each with P(s)>5.0 x 10(-6) cm/s at 10 degrees C. UT-B genetic knockout and phloretin treatment of wildtype erythrocytes similarly reduced urea analogue permeabilities. Strong temperature dependencies of formamide, acetamide, acrylamide and butyramide transport across UT-B-null membranes (E(a)>10 kcal/mol) suggested efficient diffusion of these amides across lipid bilayers. Urea analogues dimethylurea, acryalmide, methylurea, thiourea and methylformamide inhibited UT-B-mediated urea transport by >60% in the absence of transmembrane analogue gradients, supporting a pore-blocking mechanism of UT-B inhibition. Differential transport efficiencies of urea and its analogues through UT-B provide insight into chemical interactions between neutral solutes and the UT-B pore.
Collapse
Affiliation(s)
- Dan Zhao
- Department of Medicine, 1246 Health Sciences East Tower, University of California, San Francisco, CA 94143-0521, USA
| | | | | | | |
Collapse
|
137
|
Carey MA, Card JW, Voltz JW, Germolec DR, Korach KS, Zeldin DC. The impact of sex and sex hormones on lung physiology and disease: lessons from animal studies. Am J Physiol Lung Cell Mol Physiol 2007; 293:L272-8. [PMID: 17575008 DOI: 10.1152/ajplung.00174.2007] [Citation(s) in RCA: 162] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Numerous animal studies have revealed significant effects of sex and sex hormones on normal lung development, lung physiology, and various lung diseases. The primary goal of this review is to summarize knowledge to date on the effects of sex and sex hormones on lung development, physiology, and disease in animals. Specific emphasis will be placed on fibrosis, allergic airway disease, acute lung injury models, respiratory infection, and lung toxicology studies.
Collapse
Affiliation(s)
- Michelle A Carey
- National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | | | | | | | | | | |
Collapse
|
138
|
Clark S, Francis PS, Conlan XA, Barnett NW. Determination of urea using high-performance liquid chromatography with fluorescence detection after automated derivatisation with xanthydrol. J Chromatogr A 2007; 1161:207-13. [PMID: 17570379 DOI: 10.1016/j.chroma.2007.05.085] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2007] [Revised: 05/28/2007] [Accepted: 05/29/2007] [Indexed: 11/27/2022]
Abstract
A high-performance liquid chromatography (HPLC) method for the determination of urea that incorporates automated derivatisation with xanthydrol (9H-xanthen-9-ol) is described. Unlike the classic xanthydrol approach for the determination of urea, which involves the precipitation of dixanthylurea (N,N'-di-9H-xanthen-9-ylurea), the derivatisation procedure employed in this method produces N-9H-xanthen-9-ylurea, which remains in solution and can be quantified using fluorescence detection (lambda(ex)=213 nm; lambda(em)=308 nm) after chromatographic separation from interferences. The limit of detection for urea was 5 x 10(-8) M (0.003 mg L(-1)). This method was applied to the determination of urea in human and animal urine and in wine.
Collapse
Affiliation(s)
- Shona Clark
- School of Life and Environmental Sciences, Deakin University, Geelong, Victoria 3217, Australia
| | | | | | | |
Collapse
|
139
|
Layton AT. Role of UTB urea transporters in the urine concentrating mechanism of the rat kidney. Bull Math Biol 2007; 69:887-929. [PMID: 17265123 DOI: 10.1007/s11538-005-9030-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
A mathematical model of the renal medulla of the rat kidney was used to investigate urine concentrating mechanism function in animals lacking the UTB urea transporter. The UTB transporter is believed to mediate countercurrent urea exchange between descending vasa recta (DVR) and ascending vasa recta (AVR) by facilitating urea transport across DVR endothelia. The model represents the outer medulla (OM) and inner medulla (IM), with the actions of the cortex incorporated via boundary conditions. Blood flow in the model vasculature is divided into plasma and red blood cell compartments. In the base-case model configuration tubular dimensions and transport parameters are based on, or estimated from, experimental measurements or immunohistochemical evidence in wild-type rats. The base-case model configuration generated an osmolality gradient along the cortico-medullary axis that is consistent with measurements from rats in a moderately antidiuretic state. When expression of UTB was eliminated in the model, model results indicated that, relative to wild-type, the OM cortico-medullary osmolality gradient and the net urea flow through the OM were little affected by absence of UTB transporter. However, because urea transfer from AVR to DVR was much reduced, urea trapping by countercurrent exchange was significantly compromised. Consequently, urine urea concentration and osmolality were decreased by 12% and 8.9% from base case, respectively, with most of the reduction attributable to the impaired IM concentrating mechanism. These results indicate that the in vivo urine concentrating defect in knockout mouse, reported by Yang et al. (J Biol Chem 277(12), 10633-10637, 2002), is not attributable to an OM concentrating mechanism defect, but that reduced urea trapping by long vasa recta plays a significant role in compromising the concentrating mechanism of the IM. Moreover, model results are in general agreement with the explanation of knockout renal function proposed by Yang et al.
Collapse
Affiliation(s)
- Anita T Layton
- Department of Mathematics, Duke University, Box 90320, Durham, NC 27708-0320, USA.
| |
Collapse
|
140
|
Fenton RA, Knepper MA. Urea and renal function in the 21st century: insights from knockout mice. J Am Soc Nephrol 2007; 18:679-88. [PMID: 17251384 DOI: 10.1681/asn.2006101108] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Since the turn of the 21st century, gene knockout mice have been created for all major urea transporters that are expressed in the kidney: the collecting duct urea transporters UT-A1 and UT-A3, the descending thin limb isoform UT-A2, and the descending vasa recta isoform UT-B. This article discusses the new insights that the results from studies in these mice have produced in the understanding of the role of urea in the urinary concentrating mechanism and kidney function. Following is a summary of the major findings: (1) Urea accumulation in the inner medullary interstitium depends on rapid transport of urea from the inner medullary collecting duct (IMCD) lumen via UT-A1 and/or UT-A3; (2) as proposed by Robert Berliner and colleagues in the 1950s, the role of IMCD urea transporters in water conservation is to prevent a urea-induced osmotic diuresis; (3) the absence of IMCD urea transport does not prevent the concentration of NaCl in the inner medulla, contrary to what would be predicted from the passive countercurrent multiplier mechanism in the form proposed by Kokko and Rector and Stephenson; (4) deletion of UT-B (vasa recta isoform) has a much greater effect on urinary concentration than deletion of UT-A2 (descending limb isoform), suggesting that the recycling of urea between the vasa recta and the renal tubules quantitatively is less important than classic countercurrent exchange; and (5) urea reabsorption from the IMCD and the process of urea recycling are not important elements of the mechanism of protein-induced increases in GFR. In addition, the clinical relevance of these studies is discussed, and it is suggested that inhibitors that specifically target collecting duct urea transporters have the potential for clinical use as potassium-sparing diuretics that function by creation of urea-dependent osmotic diuresis.
Collapse
Affiliation(s)
- Robert A Fenton
- Water and Salt Research Center, Institute of Anatomy, Building 233/234, University of Aarhus, DK-8000 Aarhus, Denmark.
| | | |
Collapse
|
141
|
Abstract
During the past decade significant progress has been made in our understanding of the role played by urea transporters in the production of concentrated urine by the kidney. Urea transporters have been cloned and characterized in a wide range of species. The genomic organization of the two major families of mammalian urea transporters, UT-A and UT-B, has been defined, providing new insight into the mechanisms that regulate their expression and function in physiological and pathological conditions. Beside the kidney, the presence of urea transporters has been documented in a variety of tissues, where their role is not fully known. Recently, mice with targeted deletion of the major urea transporters have been generated, which have shown variable impairment of urine concentrating ability, and have helped to clarify the physiological contribution of individual transporters to this process. This review focuses on the erythrocyte urea transporter UT-B.
Collapse
Affiliation(s)
- Serena M Bagnasco
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 20187, USA.
| |
Collapse
|
142
|
Fenton RA, Smith CP, Knepper MA. Role of collecting duct urea transporters in the kidney--insights from mouse models. J Membr Biol 2007; 212:119-31. [PMID: 17264985 DOI: 10.1007/s00232-006-0871-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2006] [Indexed: 12/22/2022]
Abstract
Urea movement across plasma membranes is modulated by specialized urea transporter proteins. These proteins are proposed to play key roles in the urinary concentrating mechanism and fluid homeostasis. To date, two urea-transporter genes have been cloned; UT-A (Slc14a2), encoding at least five proteins and UT-B (Slc14a1) encoding a single protein isoform. Recently we engineered mice that lack the inner medullary collecting duct (IMCD) urea transporters, UT-A1 and UT-A3 (UT-A1/3 -/- mice). This article includes 1) a historical review of the role of renal urea transporters in renal function; 2) a review of our studies utilizing the UT-A1/3 -/- mice; 3) description of an additional line of transgenic mice in which beta-galactosidase expression is driven by the alpha-promoter of the UT-A gene, which is allowing better physiological definition of control mechanisms for UT-A expression; and 4) a discussion of the implications of the studies in transgenic mice for the teaching of kidney physiology.
Collapse
Affiliation(s)
- R A Fenton
- The Water and Salt Research Center, Institute of Anatomy, Building 1233, University of Aarhus, DK-8000, Aarhus, Denmark.
| | | | | |
Collapse
|
143
|
Ba ZF, Lu A, Shimizu T, Szalay L, Schwacha MG, Rue LW, Bland KI, Chaudry IH. 17β-Estradiol modulates vasoconstriction induced by endothelin-1 following trauma-hemorrhage. Am J Physiol Heart Circ Physiol 2007; 292:H245-50. [PMID: 17213481 DOI: 10.1152/ajpheart.00809.2006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Although endothelin-1 (ET-1) induces vasoconstriction, it remains unknown whether 17β-estradiol (E2) treatment following trauma-hemorrhage alters these ET-1-induced vasoconstrictive effects. In addition, the role of the specific estrogen receptor (ER) subtypes (ER-α and ER-β) and the endothelium-localized downstream mechanisms of actions of E2 remain unclear. We hypothesized that E2 attenuates increased ET-1-induced vasoconstriction following trauma-hemorrhage via an ER-β-mediated pathway. To study this, aortic rings were isolated from male Sprague-Dawley rats following trauma-hemorrhage with or without E2 treatment, and alterations in tension were determined in vitro. Dose-response curves to ET-1 were determined, and the vasoactive properties of E2, propylpyrazole triol (PPT, ER-α agonist), and diarylpropionitrile (DPN, ER-β agonist) were determined. The results showed that trauma-hemorrhage significantly increased ET-1-induced vasoconstriction; however, administration of E2 normalized ET-1-induced vasoconstriction in trauma-hemorrhage vessels to the sham-operated control level. The ER-β agonist DPN counteracted ET-1-induced vasoconstriction, whereas the ER-α agonist PPT was ineffective. Moreover, the vasorelaxing effects of E2 were not observed in endothelium-denuded aortic rings or by pretreatment of the rings with a nitric oxide (NO) synthase inhibitor. Cyclooxygenase inhibition with indomethacin had no effect on the action of E2. Thus, E2 administration attenuates ET-1-induced vasoconstriction following trauma-hemorrhage via an ER-β-mediated pathway that is dependent on endothelium-derived NO synthesis.
Collapse
Affiliation(s)
- Zheng F Ba
- Center for Surgical Research and Dept. of Surgery, Univ. of Alabama at Birmingham, 1670 University Blvd., Volker Hall, Rm. G094, Birmingham, AL 35294-0019, USA
| | | | | | | | | | | | | | | |
Collapse
|
144
|
Frink M, Thobe BM, Hsieh YC, Choudhry MA, Schwacha MG, Bland KI, Chaudry IH. 17beta-Estradiol inhibits keratinocyte-derived chemokine production following trauma-hemorrhage. Am J Physiol Lung Cell Mol Physiol 2006; 292:L585-91. [PMID: 17085520 DOI: 10.1152/ajplung.00364.2006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Neutrophil infiltration is a key step in the development of organ dysfunction following trauma-hemorrhage (T-H). Although we have previously shown that 17beta-estradiol (E2) prevents neutrophil infiltration and organ damage following T-H, the mechanism by which E2 inhibits neutrophil transmigration remains unknown. We hypothesized that E2 prevents neutrophil infiltration via modulation of keratinocyte-derived chemokine (KC), a major attractant for neutrophils. To examine this, male C3H/HeN mice were subjected to T-H or sham operation and thereafter resuscitated with Ringer lactate and E2 (1 mg/kg body wt) or vehicle. Animals were killed 2 h after resuscitation, and Kupffer cells were isolated. Plasma levels and Kupffer cell production capacities of KC, TNF-alpha, and IL-6 were determined by BD Cytometric Bead Arrays; lung mRNA expression of KC was measured with real-time PCR; myeloperoxidase activity assays were performed to determine neutrophil infiltration, and organ damage was assessed by edema formation. Treatment with E2 decreased systemic levels and restored Kupffer cell production of KC, TNF-alpha, and IL-6, as well as KC gene expression and protein in the lung. This was accompanied with a decrease in neutrophil infiltration and edema formation in the lung. These results suggest that E2 prevents lung neutrophil infiltration and organ damage in part by decreasing KC during posttraumatic immune response.
Collapse
Affiliation(s)
- Michael Frink
- Center for Surgical Research and Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294-0019, USA
| | | | | | | | | | | | | |
Collapse
|
145
|
Abstract
The identification, characterization, and mutational analysis of genes coding for key proteins to the mechanisms of urine concentration provide the basis for understanding the 2 types of hereditary nephrogenic diabetes insipidus (NDI): a pure type characterized by loss of water only, and a complex type characterized by loss of water and ions. Patients with hereditary NDI bearing mutations in AVPR2, the gene coding for the arginine vasopressin 2 receptor, or in AQP2, the gene coding for the vasopressin-sensitive water channel, have a pure NDI phenotype with loss of water, but normal conservation of sodium, potassium, chloride, and calcium. Patients bearing inactivating mutations in 1 of the 5 genes (SLC12A1, KCNJ1, CLCNKB, CLCNKA, and CLCNKB in combination, or BSND) that encode the membrane proteins of the thick ascending limb of the loop of Henle have a complex polyuro-polydipsic syndrome with loss of water, sodium, chloride, calcium, magnesium, and potassium. The purpose of this article is to increase the general awareness of these congenital NDI patients to prevent severe episodes of dehydration and provide precise molecular diagnosis and treatment.
Collapse
Affiliation(s)
- Daniel G Bichet
- Groupe d'Etude des Protéines Membranaires; and the Université de Montréal, Research Centre and Nephrology Service, Hôpital du Sacré-Coeur de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
146
|
Abstract
Nephrogenic diabetes insipidus which can be inherited or acquired, is characterized by an inability to concentrate urine despite normal or elevated plasma concentrations of the antidiuretic hormone, arginine-vasopressine (AVP). Polyuria, with hyposthenuria and polydipsia are the cardinal clinical manifestations of the disease. Hypercalcemia, hypokaliemia, lithium administration and chronic renal failure are the principal causes of acquired nephrogenic diabetes insipidus. About 90 percent of patients with congenital nephrogenic diabetes insipidus are males with X-linked recessive nephrogenic diabetes insipidus who have mutations in the arginine-vasopressin receptor 2 (AVPR2) gene that codes for the vasopressin V2 receptor. The gene is located in chromosome region Xq28. In about 10 percent of the families studied, congenital nephrogenic diabetes insipidus has an autosomal recessive or autosomal dominant mode of inheritance. In these cases, mutations have been identified in the aquaporin-2 gene (AQP2), which is located in chromosome region 12q13 and codes for the vasopressin-sensitive water channel. Other inherited disorders with mild, moderate or severe inability to concentrate urine include Bartter's syndrome and Cystinosis. Identification of the molecular defect underlying congenital nephrogenic diabetes insipidus is of immediate clinical significance because early diagnosis and treatment of affected infants can avert the physical and mental retardation associated with episodes of dehydration.
Collapse
Affiliation(s)
- Daniel Georges Bichet
- Génétique des maladies rénales, service de néphrologie, départements de médecine et de physiologie, centre de recherche, hôpital du Sacré-Coeur de Montréal, université de Montréal, Montréal (Québec), Canada.
| |
Collapse
|
147
|
Rick J, Chou TC. Amperometric protein sensor – fabricated as a polypyrrole, poly-aminophenylboronic acid bilayer. Biosens Bioelectron 2006; 22:329-35. [PMID: 16757163 DOI: 10.1016/j.bios.2006.04.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2005] [Revised: 03/03/2006] [Accepted: 04/04/2006] [Indexed: 11/25/2022]
Abstract
An approach to the design of electrodes for the production of sensors, which show significant changes to the passage of current in response to the concentration of target protein molecules, is presented. Screen-printed platinum electrodes, modified with two separately applied conducting polymer layers, have been developed as a potential route to forming cheap disposable protein sensors. To achieve a heightened response for the target molecules, an initial layer of polypyrrole was formed on the electrode's surface by electro-deposition. This composite was then employed as a substrate for the subsequent electro-deposition of a relatively thin 'sensing layer' of poly-aminophenylboronic acid. Cyclic voltammetry (CV) of the prepared films revealed an excursion in the current versus potential curve in the anodic phase at approximately 0.0 to +0.2V. It was clearly shown that the introduction of proteins into the CV cell resulted in a measurable decrease in the passage of current in buffered aqueous media. Measured current reductions observed on introducing lysozyme (10ppm) into the test solution were 2.3x10(-6)A for an electrode formed with a poly-aminophenylboronic acid layer on platinum, and 1.75x10(-5)A for a composite electrode formed with poly-aminophenylboronic acid on a polypyrrole coated platinum substrate. The introduction of the competing analytes, dl adrenaline or dopamine, at concentrations typically found in human urine, had little effect on the sensor's response. Additionally, the sensing system was able to maintain a response to added target proteins with as much as 2vol.% urine in the test solution. Using the electrodes in high concentrations of competing physiological analytes, they were able to respond to protein concentrations as low as 0.5ppm in buffered solutions containing urea at a concentration representative of human urine (17,000ppm), which additionally contained glucose (1000ppm).
Collapse
Affiliation(s)
- John Rick
- Chemical Engineering Department, National Cheng Kung University, Tainan 70101, Taiwan
| | | |
Collapse
|
148
|
Allen IC, Pace AJ, Jania LA, Ledford JG, Latour AM, Snouwaert JN, Bernier V, Stocco R, Therien AG, Koller BH. Expression and function of NPSR1/GPRA in the lung before and after induction of asthma-like disease. Am J Physiol Lung Cell Mol Physiol 2006; 291:L1005-17. [PMID: 16829631 DOI: 10.1152/ajplung.00174.2006] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
A genetic contribution to asthma susceptibility is well recognized, and linkage studies have identified a large number of genes associated with asthma pathogenesis. Recently, a locus encoding a seven-transmembrane protein was shown to be associated with asthma in founder populations. The expression of the protein GPRA (G protein-coupled receptor for asthma susceptibility) in human airway epithelia and smooth muscle, and its increased expression in a mouse model of asthma, suggested that a gain-of-function mutation in this gene increased the disease risk. However, we report here that the development of allergic lung disease in GPRA-deficient mice is unaltered. A possible explanation for this finding became apparent upon reexamination of the expression of this gene. In contrast to initial studies, our analyses failed to detect expression of GPRA in human lung tissue or in mice with allergic lung disease. We identify a single parameter that distinguishes GPRA-deficient and wild-type mice. Whereas the change in airway resistance in response to methacholine was identical in control and GPRA-deficient mice, the mutant animals showed an attenuated response to thromboxane, a cholinergic receptor-dependent bronchoconstricting agent. Together, our studies fail to support a direct contribution of GPRA to asthma pathogenesis. However, our data suggest that GPRA may contribute to the asthmatic phenotype by altering the activity of other pathways, such as neurally mediated mechanisms, that contribute to disease. This interpretation is supported by high levels of GPRA expression in the brain and its recent identification as the neuropeptide S receptor.
Collapse
Affiliation(s)
- Irving C Allen
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, 27599-7264, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
149
|
Musch W, Verfaillie L, Decaux G. Age-Related Increase in Plasma Urea Level and Decrease in Fractional Urea Excretion: Clinical Application in the Syndrome of Inappropriate Secretion of Antidiuretic Hormone. Clin J Am Soc Nephrol 2006; 1:909-14. [PMID: 17699306 DOI: 10.2215/cjn.00320106] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This study confirms in humans an age-related increase in plasma urea levels (r = 0.62; P < 0.001; y = 0.229x + 18.26) and no correlation between plasma creatinine and age (r = 0.06; NS). Fractional urea excretion (FE urea) decreases with age (r = -0.41; P < 0.001; y = -0.226x + 55). Comparing urea and creatinine clearances, measured in 19 young and in 15 old women, a larger decrease of urea clearance (-56%) compared with the creatinine clearance (-43%) was observed as expected, explaining the lower FE urea in the elderly. In old women, the daily urea excretion was 27% and the daily creatinine excretion was 42% lower than in young women. An age-related decrease of same magnitude in both creatinine production and creatinine clearance explains why plasma creatinine remains stable with increasing age. The observation of a more important decrease in urea clearance (56%) than in urea production (27%) in older women led to an expected increase in plasma urea of 29%. These observations incited a comparison of biochemical profiles from younger and older patients with the syndrome of inappropriate secretion of antidiuretic hormone (SIADH). Young patients with SIADH present lower mean plasma urea (18 +/- 8 mg/dl) and higher mean FE urea (58 +/- 14%), compared with both young control subjects (mean plasma urea 27 +/- 7 mg/dl; mean FE urea 46 +/- 10%) and old patients with SIADH (mean plasma urea 29 +/- 8 mg/dl; mean FE urea 44 +/- 15%). Physicians must realize that frankly low plasma urea values and high FE urea values can be expected only in young patients with SIADH, whereas old patients with SIADH will present values of plasma urea and FE urea in the same range than young control subjects. However, old patients with SIADH show still lower mean plasma urea values and higher mean FE urea values, compared with old control subjects (mean plasma urea 39 +/- 8 mg/dl; mean FE urea 36 +/- 9%), in whom plasma urea values between 40 and 50 mg/dl must be considered as usual.
Collapse
Affiliation(s)
- Wim Musch
- Department of Internal Medicine, Iris South Hospitals Site Bracops, Brussels, Belgium
| | | | | |
Collapse
|
150
|
Pinter GG, Shohet JL. Two fluid compartments in the renal inner medulla: a view through the keyhole of the concentrating process. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2006; 364:1551-61. [PMID: 16766360 DOI: 10.1098/rsta.2006.1774] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Approximately four decades ago, the countercurrent theory became influential in studies on the concentrating process in the mammalian kidney. The theory successfully represented the concentrating process in the outer medulla, but the problem of the concentrating mechanism in the inner medulla, as defined by Homer Smith has remained essentially intractable. In a recent comprehensive review by Knepper and coworkers of various theories and models, attention was refocused on the possible role of hyaluronate (HA) in the inner medullary concentrating process. The authors proposed a hypothesis that HA can convert hydrostatic pressure to concentrating work.Here, we briefly survey the earlier ideas on the role imputed to HA and present a new hypothesis which is different from that of Knepper and coworkers. We estimate that the hydrostatic pressures available in the inner medulla can account only for a very small fraction of the concentrating work. We hypothesize that the role of HA is tied up with extravasated plasma albumin and suggest that owing to the property of HA solutions to exclude other macromolecules, extravasated plasma albumin and HA constitute two fluid compartments in the interstitium in the inner medulla. In this proposed two-compartment model, the Gibbs-Donnan distribution influences the movement of ions and water between the HA and the extravasated albumin compartment. To relate the hypothetical role of HA to the concentrating process, we briefly describe new results obtained by other investigators on the accumulation of urea in the inner medulla. This subject has been critically reviewed recently by Yang & Bankir.Many processes have been identified as contributing to the concentrating process in the mammalian inner medulla. We speculate that among these many processes, the primary responsibility for the final concentration of the excreted urine may be portioned out differently in different mammalian species.
Collapse
Affiliation(s)
- G G Pinter
- University of Maryland, Baltimore 21201, USA.
| | | |
Collapse
|