1
|
Kessler EC, Bruckmaier RM, Gross JJ. Kidney function, but not nitrogen excretion differs between Brown Swiss and Holstein dairy cows. J Dairy Sci 2024:S0022-0302(24)00958-5. [PMID: 38908706 DOI: 10.3168/jds.2024-24997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 05/21/2024] [Indexed: 06/24/2024]
Abstract
Brown Swiss (BS) cows have greater urea concentrations in milk and blood compared with Holstein (HO) cows. We tested the hypothesis that BS and HO cows differ in kidney function and nitrogen excretion. Blood, saliva, urine, and feces were sampled in 31 multiparous BS and 46 HO cows kept under identical feeding and management conditions. Samples were collected at different lactational stages after the monthly DHIA control test-day. To test the glomerular filtration rate (GFR) and urea excretion, concentrations of creatinine and urea were measured in serum, urine, and saliva. As an additional marker to estimate GFR, we determined symmetric dimethylarginine (SDMA) in serum. Feces were analyzed for dry matter content and nitrogen concentration. Data on milk urea and protein concentrations, and daily milk yield were obtained from the monthly DHIA test-day records. The effects of breed, time, and parity number on blood, saliva, urine, feces, and milk parameters were evaluated with the GLM procedure with breed, time, and parity number as fixed effects. Differences between BS and HO were assessed by the Tukey-corrected t-test at P < 0.05. Concentrations of urea, creatinine, and SDMA in serum, were greater in BS than in HO cows (P < 0.01): 5.46 ± 0.19 vs 4.72 ± 0.13 mmol/L (urea), 105.96 ± 2.23 vs 93.07 ± 1.50 mmol/l (creatinine), and 16.78 ± 0.69 vs 13.39 ± 0.44 µg/dL (SDMA). We observed a greater urea concentration in BS cows (25.8 ± 0.7 vs 21.8 ± 0.7 mg/dL) and protein content in milk (3.70 ± 0.08 vs 3.45 ± 0.07%) than in HO cows (P < 0.01). Urea and creatinine concentrations in urine and saliva did not differ among breeds. No differences between BS and HO were observed for milk yield, fecal DM, and fecal nitrogen content. Dry matter intake and body weight were similar in BS and HO cows (P > 0.05). Despite greater urea, creatinine, and SDMA concentrations in blood as well as a higher milk urea content in BS compared with HO, respective concentrations in urine did not differ between breeds. In conclusion, our results demonstrate a lower renal GFR in BS compared with HO cows, thereby contributing to the greater plasma urea concentration in BS cows. However, estimation of nitrogen excretion via milk, urine, and feces does not entirely reflect nitrogen turnover within the animal.
Collapse
Affiliation(s)
- E C Kessler
- Veterinary Physiology, Vetsuisse Faculty, University of Bern, CH-3012 Bern, Switzerland
| | - R M Bruckmaier
- Veterinary Physiology, Vetsuisse Faculty, University of Bern, CH-3012 Bern, Switzerland
| | - J J Gross
- Veterinary Physiology, Vetsuisse Faculty, University of Bern, CH-3012 Bern, Switzerland.
| |
Collapse
|
2
|
Purkerson JM, Everett CA, Schwartz GJ. Ammonium chloride-induced acidosis exacerbates cystitis and pyelonephritis caused by uropathogenic E. coli. Physiol Rep 2022; 10:e15471. [PMID: 36151614 PMCID: PMC9508385 DOI: 10.14814/phy2.15471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 06/16/2023] Open
Abstract
Acute pyelonephritis caused by uropathogenic E. coli (UPEC) can cause renal scarring and lead to development of chronic kidney disease. Prevention of kidney injury requires an understanding of host factors and/or UPEC adaptive responses that are permissive for UPEC colonization of the urinary tract. Although some studies have suggested urine acidification limits UPEC growth in culture, other studies have described acid-resistance mechanisms (AR) in E. coli such as the CadC/CadBA module that promotes adaptation to acid and nitrosative stress. Herein we confirm and extend our previous study by demonstrating that despite urine acidification, metabolic acidosis induced by dietary ammonium chloride (NH4 Cl-A) exacerbates cystitis and pyelonephritis in innate immune competent (C3H-HeN) mice characterized by: (1) markedly elevated UPEC burden and increased chemokine/cytokine and NOS2 mRNA expression, (2) accumulation of intravesicular debris noninvasively detected by Power Doppler Ultrasound (PDUS), and (3) collecting duct (CD) dysfunction that manifests as a urine concentration defect. Bladder debris and CD dysfunction were due to the inflammatory response, as neither was observed in Tlr4-deficient (C3H-HeJ) mice. The effect of NH4 Cl-A was unrelated to acidosis as dietary administration of hydrochloric acid (HCl-A) yielded a comparable acid-base status yet did not increase UPEC burden. NH4 Cl-A increased polyamines and decreased nitric oxide (NO) metabolites in urine indicating that excess dietary ammonium shifts arginine metabolism toward polyamines at the expense of NO synthesis. Furthermore, despite increased expression of NOS2, NO production post UPEC infection was attenuated in NH4 Cl-A mice compared to controls. Thus, in addition to induction of metabolic acidosis and urine acidification, excess dietary ammonium alters the polyamine:NO balance and thereby compromises NOS2-mediated innate immune defense.
Collapse
Affiliation(s)
- Jeffrey M. Purkerson
- Pediatric NephrologyUniversity of Rochester Medical CenterRochesterNew YorkUSA
- Strong Children's Research CenterUniversity of Rochester Medical CenterRochesterNew YorkUSA
| | - Coralee A. Everett
- Pediatric NephrologyUniversity of Rochester Medical CenterRochesterNew YorkUSA
- Strong Children's Research CenterUniversity of Rochester Medical CenterRochesterNew YorkUSA
| | - George J. Schwartz
- Pediatric NephrologyUniversity of Rochester Medical CenterRochesterNew YorkUSA
- Strong Children's Research CenterUniversity of Rochester Medical CenterRochesterNew YorkUSA
| |
Collapse
|
3
|
Li J, Wang X, Lan T, Lu Y, Hong M, Ding L, Wang L. CDK5/NFAT5-Regulated Transporters Involved in Osmoregulation in Fejervarya cancrivora. BIOLOGY 2022; 11:biology11060858. [PMID: 35741379 PMCID: PMC9220195 DOI: 10.3390/biology11060858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 11/19/2022]
Abstract
Crab-eating frogs (Fejervarya cancrivora) can live in brackish water with a salinity of up to 18‱, although most amphibians are not able to tolerate such high saline environments. To investigate its potential osmoregulation, we conducted experiments in F. cancrivora and F. multistriata. The results showed that F. cancrivora made use of ions (such as Na+ and Cl−) to increase intracellular concentrations via the Na+/K+-ATPase (NKA) enzyme. The mRNA expression of aldose reductase (AR) was significantly higher in F. cancrivora (p < 0.05), indicating that more organic osmolytes were produced and transported to maintain cellular homeosis. The mRNA expressions of Aquaporin 1 (AQP1) and AQP3 in kidney were significantly higher in F. cancrivora, while AQP expression in skin was higher in F. multistriata (p < 0.05). The mRNA level in activating the transcription of the nuclear factor of activated T cells-5 (NFAT5) which is one of the target genes of regulating the cellular response to hypertonicity, was higher in F. cancrivora. The protein expression of CDK5, the upstream protein of the NFAT5 pathway, was 2 times higher in F. cancrivora. Therefore, we can conclude that CDK5/NFAT5-regulated transporters might be involved in osmoregulation in F. cancrivora.
Collapse
Affiliation(s)
| | | | | | | | | | - Li Ding
- Correspondence: (L.D.); (L.W.)
| | | |
Collapse
|
4
|
Lee S, Sirich TL, Meyer TW. Improving Solute Clearances by Hemodialysis. Blood Purif 2022; 51:1-12. [PMID: 35613554 PMCID: PMC9691790 DOI: 10.1159/000524512] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 04/04/2022] [Indexed: 11/19/2022]
Abstract
The adequacy of hemodialysis is now assessed by measuring the removal of the single-solute urea. The urea clearance provided by contemporary dialysis is a large fraction of the blood flow through the dialyzer and therefore cannot be increased much further. Other solutes however likely contribute more than urea to the residual uremic illness suffered by hemodialysis patients. We here review methods which could be employed to increase the clearance of nonurea solutes. We will separately consider the clearances of free low-molecular-mass solutes, free larger solutes, and protein-bound solutes. New clinical studies will be required to test the extent to which increasing the clearance on nonurea solutes with these various characteristics can improve patients' health.
Collapse
Affiliation(s)
- Seolhyun Lee
- The Department of Medicine, Stanford University, Palo Alto, California, USA
- The Department of Medicine, VA Palo Alto Healthcare System, Palo Alto, California, USA
| | - Tammy L. Sirich
- The Department of Medicine, Stanford University, Palo Alto, California, USA
- The Department of Medicine, VA Palo Alto Healthcare System, Palo Alto, California, USA
| | - Timothy W. Meyer
- The Department of Medicine, Stanford University, Palo Alto, California, USA
- The Department of Medicine, VA Palo Alto Healthcare System, Palo Alto, California, USA
| |
Collapse
|
5
|
Yahouédéhou SCMA, Neres JSDS, da Guarda CC, Carvalho SP, Santiago RP, Figueiredo CVB, Fiuza LM, Ndidi US, de Oliveira RM, Fonseca CA, Nascimento VML, Rocha LC, Adanho CSA, da Rocha TSC, Adorno EV, Goncalves MS. Sickle Cell Anemia: Variants in the CYP2D6, CAT, and SLC14A1 Genes Are Associated With Improved Hydroxyurea Response. Front Pharmacol 2020; 11:553064. [PMID: 33013391 PMCID: PMC7510454 DOI: 10.3389/fphar.2020.553064] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 08/18/2020] [Indexed: 11/13/2022] Open
Abstract
Differences in hydroxyurea response in sickle cell anemia may arise due to a series of factors with genetic factors appearing to be predominant. This study aims to investigate the effects of single nucleotide polymorphisms in genes encoding drug-metabolizing enzymes and solute carriers on hydroxyurea response, in patients with sickle cell anemia. For that purpose, a total number of 90 patients with sickle cell anemia were recruited, 45 were undergoing hydroxyurea treatment, while 45 were not under the treatment. Association analyses were performed between CYP3A4 (rs2740574), CYP2D6 (rs3892097), CAT (rs7943316 and rs1001179), and SLC14A1 (rs2298720) variants and laboratory parameters. According to our findings, patients with hydroxyurea treatment demonstrated higher HbF levels and a significant improvement in hemolytic, hepatic, inflammatory, and lipid parameters in comparison to those without the treatment. We also found significant associations between the CYP2D6 (rs3892097), CAT (rs7943316 and rs1001179), and SLC14A1 (rs2298720) variants and an improvement of the therapeutic effects, specifically the hemolytic, hepatic, inflammatory, lipid, and renal parameters. In conclusion, our results highlight the importance of the investigated variants, and their strong association with hydroxyurea efficacy in patients with sickle cell anemia, which may be considered in the future as genetic markers.
Collapse
Affiliation(s)
- Sètondji Cocou Modeste Alexandre Yahouédéhou
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, Salvador, Brazil.,Laboratório de Pesquisa em Anemia, Departamento de Análises Clínicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Brazil
| | | | - Caroline Conceição da Guarda
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, Salvador, Brazil.,Laboratório de Pesquisa em Anemia, Departamento de Análises Clínicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Brazil
| | - Suellen Pinheiro Carvalho
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, Salvador, Brazil.,Laboratório de Pesquisa em Anemia, Departamento de Análises Clínicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Brazil
| | - Rayra Pereira Santiago
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, Salvador, Brazil.,Laboratório de Pesquisa em Anemia, Departamento de Análises Clínicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Brazil
| | - Camylla Vilas Boas Figueiredo
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, Salvador, Brazil.,Laboratório de Pesquisa em Anemia, Departamento de Análises Clínicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Brazil
| | - Luciana Magalhães Fiuza
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, Salvador, Brazil.,Laboratório de Pesquisa em Anemia, Departamento de Análises Clínicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Brazil
| | | | - Rodrigo Mota de Oliveira
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, Salvador, Brazil.,Laboratório de Pesquisa em Anemia, Departamento de Análises Clínicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Brazil
| | - Cleverson Alves Fonseca
- Laboratório de Pesquisa em Anemia, Departamento de Análises Clínicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Brazil
| | | | | | | | | | - Elisângela Vitória Adorno
- Laboratório de Pesquisa em Anemia, Departamento de Análises Clínicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Brazil
| | - Marilda Souza Goncalves
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, Salvador, Brazil.,Laboratório de Pesquisa em Anemia, Departamento de Análises Clínicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Brazil
| |
Collapse
|
6
|
Chan CYL, Hiong KC, Boo MV, Choo CYL, Wong WP, Chew SF, Ip YK. Light exposure enhances urea absorption in the fluted giant clam, Tridacna squamosa, and up-regulates the protein abundance of a light-dependent urea active transporter, DUR3-like, in its ctenidium. J Exp Biol 2018; 221:jeb176313. [PMID: 29540461 DOI: 10.1242/jeb.176313] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 03/12/2018] [Indexed: 12/26/2022]
Abstract
Giant clams live in nutrient-poor reef waters of the Indo-Pacific and rely on symbiotic dinoflagellates (Symbiodinium spp., also known as zooxanthellae) for nutrients. As the symbionts are nitrogen deficient, the host clam has to absorb exogenous nitrogen and supply it to them. This study aimed to demonstrate light-enhanced urea absorption in the fluted giant clam, Tridacna squamosa, and to clone and characterize the urea active transporter DUR3-like from its ctenidium (gill). The results indicate that T. squamosa absorbs exogenous urea, and the rate of urea uptake in the light was significantly higher than that in darkness. The DUR3-like coding sequence obtained from its ctenidium comprised 2346 bp, encoding a protein of 782 amino acids and 87.0 kDa. DUR3-like was expressed strongly in the ctenidium, outer mantle and kidney. Twelve hours of exposure to light had no significant effect on the transcript level of ctenidial DUR3-like However, between 3 and 12 h of light exposure, DUR3-like protein abundance increased progressively in the ctenidium, and became significantly greater than that in the control at 12 h. DUR3-like had an apical localization in the epithelia of the ctenidial filaments and tertiary water channels. Taken together, these results indicate that DUR3-like might participate in light-enhanced urea absorption in the ctenidium of T. squamosa When made available to the symbiotic zooxanthellae that are known to possess urease, the absorbed urea can be metabolized to NH3 and CO2 to support amino acid synthesis and photosynthesis, respectively, during insolation.
Collapse
Affiliation(s)
- Christabel Y L Chan
- Department of Biological Sciences, National University of Singapore, Kent Ridge, Singapore 117543, Republic of Singapore
| | - Kum C Hiong
- Department of Biological Sciences, National University of Singapore, Kent Ridge, Singapore 117543, Republic of Singapore
| | - Mel V Boo
- Department of Biological Sciences, National University of Singapore, Kent Ridge, Singapore 117543, Republic of Singapore
| | - Celine Y L Choo
- Department of Biological Sciences, National University of Singapore, Kent Ridge, Singapore 117543, Republic of Singapore
| | - Wai P Wong
- Department of Biological Sciences, National University of Singapore, Kent Ridge, Singapore 117543, Republic of Singapore
| | - Shit F Chew
- Natural Sciences and Science Education, National Institute of Education, Nanyang Technological University, 1 Nanyang Walk, Singapore 637616, Republic of Singapore
| | - Yuen K Ip
- Department of Biological Sciences, National University of Singapore, Kent Ridge, Singapore 117543, Republic of Singapore
- The Tropical Marine Science Institute, National University of Singapore, Kent Ridge, Singapore 119227, Republic of Singapore
| |
Collapse
|
7
|
Jiang T, Li Y, Layton AT, Wang W, Sun Y, Li M, Zhou H, Yang B. Generation and phenotypic analysis of mice lacking all urea transporters. Kidney Int 2017; 91:338-351. [PMID: 27914708 PMCID: PMC5423716 DOI: 10.1016/j.kint.2016.09.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 08/30/2016] [Accepted: 09/08/2016] [Indexed: 01/22/2023]
Abstract
Urea transporters (UT) are a family of transmembrane urea-selective channel proteins expressed in multiple tissues and play an important role in the urine concentrating mechanism of the mammalian kidney. UT inhibitors have diuretic activity and could be developed as novel diuretics. To determine if functional deficiency of all UTs in all tissues causes physiological abnormality, we established a novel mouse model in which all UTs were knocked out by deleting an 87 kb of DNA fragment containing most parts of Slc14a1 and Slc14a2 genes. Western blot analysis and immunofluorescence confirmed that there is no expression of urea transporter in these all-UT-knockout mice. Daily urine output was nearly 3.5-fold higher, with significantly lower urine osmolality in all-UT-knockout mice than that in wild-type mice. All-UT-knockout mice were not able to increase urinary urea concentration and osmolality after water deprivation, acute urea loading, or high protein intake. A computational model that simulated UT-knockout mouse models identified the individual contribution of each UT in urine concentrating mechanism. Knocking out all UTs also decreased the blood pressure and promoted the maturation of the male reproductive system. Thus, functional deficiency of all UTs caused a urea-selective urine-concentrating defect with little physiological abnormality in extrarenal organs.
Collapse
Affiliation(s)
- Tao Jiang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yingjie Li
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Anita T Layton
- Department of Mathematics, Duke University, Durham, North Carolina, USA
| | - Weiling Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yi Sun
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Min Li
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Hong Zhou
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Baoxue Yang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China.
| |
Collapse
|
8
|
Abstract
The urea transporter UT-B is expressed in multiple tissues including erythrocytes, kidney, brain, heart, liver, colon, bone marrow, spleen, lung, skeletal muscle, bladder, prostate, and testis in mammals. Phenotype analysis of UT-B-null mice has confirmed that UT-B deletion results in a urea-selective urine-concentrating defect (see Chap. 9 ). The functional significance of UT-B in extrarenal tissues studied in the UT-B-null mouse is discussed in this chapter. UT-B-null mice present depression-like behavior with urea accumulation and nitric oxide reduction in the hippocampus. UT-B deletion causes a cardiac conduction defect, and TNNT2 and ANP expression changes in the aged UT-B-null heart. UT-B also plays a very important role in protecting bladder urothelium from DNA damage and apoptosis by regulating the urea concentration in urothelial cells. UT-B functional deficiency results in urea accumulation in the testis and early maturation of the male reproductive system. These results show that UT-B is an indispensable transporter involved in maintaining physiological functions in different tissues.
Collapse
|
9
|
Guo A, Hao Y, Wang J, Zhao Q, Wang D. Concentrations of osmotically related constituents in plasma and urine of finless porpoise (Neophocaena asiaeorientalis): implications for osmoregulatory strategies for marine mammals living in freshwater. Zool Stud 2014. [DOI: 10.1186/1810-522x-53-10] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Abstract
Background
Most cetaceans inhabit the hyperosmotic marine environment with only a few species living in freshwater habitats. The Yangtze finless porpoise (Neophocaena asiaeorientalis asiaeorientalis) is the only freshwater subspecies of the genus. Our aim was to study whether the osmoregulation mechanism of the Yangtze finless porpoise is different from the marine subspecies, the East Asian finless porpoise (Neophocaena asiaeorientalis sunameri). We assayed and compared the concentrations of the constituents involved in osmoregulation in the blood and urine in the Yangtze finless porpoise and the East Asian finless porpoise. We also compared the corresponding urine constituents of the porpoises with existing data on fin whales (Balaenoptera physalus) and bottlenose dolphins (Tursiops truncatus).
Results
The mean plasma osmolality of Yangtze finless porpoise was significantly lower than that of the marine subspecies (P < 0.01). Similarly, the urine osmolality of Yangtze finless porpoise was also significantly lower than that of its marine counterpart (P < 0.05). However, the urine sodium concentration of freshwater finless porpoise was significantly lower than that in the marine subspecies (P < 0.01), even though their serum sodium has no significant difference. Moreover, the freshwater porpoise has significantly lower urine urea concentration but much higher serum urea than in the marine finless porpoise (P < 0.05).
Conclusions
These results suggest that the freshwater finless porpoise does have different osmoregulatory mechanism from marine cetaceans. Conserving sodium by excreting urine with low ion levels may be an essential strategy to maintain the serum electrolyte balance for the freshwater subspecies that also appears to be more susceptible to hyponatremia.
Collapse
|
10
|
Li X, Chen G, Yang B. Urea transporter physiology studied in knockout mice. Front Physiol 2012; 3:217. [PMID: 22745630 PMCID: PMC3383189 DOI: 10.3389/fphys.2012.00217] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Accepted: 05/31/2012] [Indexed: 01/09/2023] Open
Abstract
In mammals, there are two types of urea transporters; urea transporter (UT)-A and UT-B. The UT-A transporters are mainly expressed in kidney epithelial cells while UT-B demonstrates a broader distribution in kidney, heart, brain, testis, urinary tract, and other tissues. Over the past few years, multiple urea transporter knockout mouse models have been generated enabling us to explore the physiological roles of the different urea transporters. In the kidney, deletion of UT-A1/UT-A3 results in polyuria and a severe urine concentrating defect, indicating that intrarenal recycling of urea plays a crucial role in the overall capacity to concentrate urine. Since UT-B has a wide tissue distribution, multiple phenotypic abnormalities have been found in UT-B null mice, such as defective urine concentration, exacerbated heart blockage with aging, depression-like behavior, and earlier male sexual maturation. This review summarizes the new insights of urea transporter functions in different organs, gleaned from studies of urea transporter knockout mice, and explores some of the potential pharmacological prospects of urea transporters.
Collapse
Affiliation(s)
- Xuechen Li
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education Beijing, China
| | | | | |
Collapse
|
11
|
Grether-Beck S, Felsner I, Brenden H, Kohne Z, Majora M, Marini A, Jaenicke T, Rodriguez-Martin M, Trullas C, Hupe M, Elias PM, Krutmann J. Urea uptake enhances barrier function and antimicrobial defense in humans by regulating epidermal gene expression. J Invest Dermatol 2012; 132:1561-72. [PMID: 22418868 PMCID: PMC3352965 DOI: 10.1038/jid.2012.42] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Urea is an endogenous metabolite, known to enhance stratum corneum hydration. Yet, topical urea anecdotally also improves permeability barrier function, and it appears to exhibit antimicrobial activity. Hence, we hypothesized that urea is not merely a passive metabolite, but a small-molecule regulator of epidermal structure and function. In 21 human volunteers, topical urea improved barrier function in parallel with enhanced antimicrobial peptide (AMP; LL-37 and β-defensin-2) expression. Urea stimulates the expression of, and is transported into, keratinocytes by two urea transporters (UTs), UT-A1 and UT-A2, and by aquaporins 3, 7, and 9. Inhibitors of these UTs block the downstream biological effects of urea, which include increased mRNA and protein levels of (i) transglutaminase-1, involucrin, loricrin, and filaggrin, (ii) epidermal lipid synthetic enzymes, and (iii) cathelicidin/LL-37 and β-defensin-2. Finally, we explored the potential clinical utility of urea, showing that topical urea applications normalized both barrier function and AMP expression in a murine model of atopic dermatitis. Together, these results show that urea is a small-molecule regulator of epidermal permeability barrier function and AMP expression after transporter uptake, followed by gene regulatory activity in normal epidermis, with potential therapeutic applications in diseased skin.
Collapse
Affiliation(s)
- Susanne Grether-Beck
- Institut fuer Umweltmedizinische Forschung, Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
von Morze C, Larson PEZ, Hu S, Keshari K, Wilson DM, Ardenkjaer-Larsen JH, Goga A, Bok R, Kurhanewicz J, Vigneron DB. Imaging of blood flow using hyperpolarized [(13)C]urea in preclinical cancer models. J Magn Reson Imaging 2011; 33:692-7. [PMID: 21563254 DOI: 10.1002/jmri.22484] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
PURPOSE To demonstrate dynamic imaging of a diffusible perfusion tracer, hyperpolarized [(13)C]urea, for regional measurement of blood flow in preclinical cancer models. MATERIALS AND METHODS A pulse sequence using balanced steady state free precession (bSSFP) was developed, with progressively increasing flip angles for efficient sampling of the hyperpolarized magnetization. This allowed temporal and volumetric imaging of the [(13)C]urea signal. Regional signal dynamics were quantified for kidneys and liver, and estimates of relative blood flows were derived from the data. Detailed perfusion simulations were performed to validate the methodology. RESULTS Significant differences were observed in the signal patterns between normal and cancerous murine hepatic tissues. In particular, a 19% reduction in mean blood flow was observed in tumors, with 26% elevation in the tumor rim. The blood flow maps were also compared with metabolic imaging results with hyperpolarized [1-(13)C]pyruvate. CONCLUSION Regional assessment of perfusion is possible by imaging of hyperpolarized [(13)C]urea, which is significant for the imaging of cancer.
Collapse
Affiliation(s)
- Cornelius von Morze
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California 94158-2512, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Wester ES, Storry JR, Olsson ML. Characterization of Jk(a+(weak)): a new blood group phenotype associated with an altered JK*01 allele. Transfusion 2011; 51:380-92. [PMID: 21309779 DOI: 10.1111/j.1537-2995.2010.02795.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND The clinically important Kidd (JK) blood group system is considered to be relatively uncomplicated, both serologically and genetically. The JK*01 and JK*02 alleles give rise to Jk(a) and Jk(b) antigens, respectively, and silenced alleles result in Jk(a-b-). Other inherited variants analogous to Fy(x) and weak D phenotypes have not been characterized for JK, although recent abstracts indicate their presence. STUDY DESIGN AND METHODS Six index samples from individuals whose RBCs reacted variably or weakly with different sources of anti-Jk(a) and 300 controls of the four known JK phenotypes were investigated by standard serology, flow cytometry, Western blotting, and the urea hemolysis test. Molecular analysis, including allele-specific polymerase chain reaction (PCR), DNA sequencing, and transcript analysis by real-time PCR, was performed. RESULTS All Jk(a+(w)b-) and Jk(a+(w)b+) index samples were homo- or heterozygous for an altered JK*01 allele carrying 130G>A (Glu44Lys) and the JK*02-associated silent SNPs 588G and Intron 9 -46g. Blood donor screening indicated an allele frequency of 0.042. Titration and flow cytometry with anti-Jk(a) gave lower values in index samples compared to controls, as did anti-Jk3 titers. Donors with 130A also showed significantly decreased Jk(a) density by flow cytometry versus 130G. Western blotting with anti-UT-B demonstrated weaker reactivity with Jk(a+(w)) membranes while JK mRNA levels could not discriminate index samples from controls. The urea hemolysis test was only moderately affected in two Jk(a+(w)b-) samples. CONCLUSIONS A new phenotype with weakened Jk(a) expression on RBCs is associated with a JK*01-like allele, which may constitute a risk for hemolytic transfusion reactions if antigen-positive units are missed by routine serology.
Collapse
Affiliation(s)
- Elisabet S Wester
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | | | | |
Collapse
|
14
|
Hwang S, Gunaratne R, Rinschen MM, Yu MJ, Pisitkun T, Hoffert JD, Fenton RA, Knepper MA, Chou CL. Vasopressin increases phosphorylation of Ser84 and Ser486 in Slc14a2 collecting duct urea transporters. Am J Physiol Renal Physiol 2010; 299:F559-67. [PMID: 20576681 PMCID: PMC2944290 DOI: 10.1152/ajprenal.00617.2009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2009] [Accepted: 06/21/2010] [Indexed: 11/22/2022] Open
Abstract
Vasopressin-regulated urea transport in the renal inner medullary collecting duct (IMCD) is mediated by two urea channel proteins, UT-A1 and UT-A3, derived from the same gene (Slc14a2) by alternative splicing. The NH(2)-terminal 459 amino acids are the same in both proteins. To study UT-A1/3 phosphorylation, we made phospho-specific antibodies to UT-A sequences targeting phospho-serines at positions 84 and 486, sites identified previously by protein mass spectrometry. Both antibodies proved specific, recognizing only the phosphorylated forms of UT-A1 and -A3. Immunoblotting of rat IMCD suspensions or whole inner medullas showed that the V2R-selective vasopressin analog 1-deamino-8-d-arginine vasopressin (dDAVP) increases phosphorylation at Ser84 (in UT-A1 and UT-A3) and Ser486 (in UT-A1) by about eightfold. Time course studies in rat IMCD suspensions showed maximum phosphorylation within 1 min of dDAVP exposure, consistent with the time course of vasopressin-stimulated phosphorylation of the vasopressin-sensitive water channel aquaporin-2 at Ser256. Confocal immunofluorescence in Brattleboro rat medullary tissue showed labeling limited to the IMCD, which increased markedly in response to dDAVP. Immuno-electron microscopy studies showed that both phosphorylated forms were present mainly in intracellular compartments in the presence of vasopressin. These studies demonstrate regulated phosphorylation of both UT-A1 and UT-A3 in response to vasopressin in a manner consistent with coordinate regulation of UT-A and aquaporin-2 in the renal IMCD. The findings add to prior evidence for vasopressin-induced phosphorylation of UT-A1, providing evidence that UT-A3 may be regulated by phosphorylation as well.
Collapse
Affiliation(s)
- Shelly Hwang
- Laboratory of Kidney and Electrolyte Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
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: 3.1] [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
|
16
|
Abstract
Cells in the renal inner medulla are normally exposed to extraordinarily high levels of NaCl and urea. The osmotic stress causes numerous perturbations because of the hypertonic effect of high NaCl and the direct denaturation of cellular macromolecules by high urea. High NaCl and urea elevate reactive oxygen species, cause cytoskeletal rearrangement, inhibit DNA replication and transcription, inhibit translation, depolarize mitochondria, and damage DNA and proteins. Nevertheless, cells can accommodate by changes that include accumulation of organic osmolytes and increased expression of heat shock proteins. Failure to accommodate results in cell death by apoptosis. Although the adapted cells survive and function, many of the original perturbations persist, and even contribute to signaling the adaptive responses. This review addresses both the perturbing effects of high NaCl and urea and the adaptive responses. We speculate on the sensors of osmolality and document the multiple pathways that signal activation of the transcription factor TonEBP/OREBP, which directs many aspects of adaptation. The facts that numerous cellular functions are altered by hyperosmolality and remain so, even after adaptation, indicate that both the effects of hyperosmolality and adaptation to it involve profound alterations of the state of the cells.
Collapse
|
17
|
Jung JY, Kwon HM, Kim J. Regulation of Urea Transporters by Tonicity-responsive Enhancer Binding Protein. Electrolyte Blood Press 2007; 5:28-33. [PMID: 24459497 PMCID: PMC3894503 DOI: 10.5049/ebp.2007.5.1.28] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2007] [Accepted: 04/29/2007] [Indexed: 11/29/2022] Open
Abstract
Urea accumulation in the renal inner medulla plays a key role in the maintenance of maximal urinary concentrating ability. Urea transport in the kidney is mediated by transporter proteins that include renal urea transporter (UT-A) and erythrocyte urea transporter (UT-B). UT-A1 and UT-A2 are produced from the same gene. There is an active tonicity-responsive enhancer (TonE) in the promoter of UT-A1, and the UT-A1 promoter is stimulated by hypertonicity via tonicity-responsive enhancer binding protein (TonEBP). The downregulation of UT-A2 raises the possibility that TonEBP also regulates its promoter. There is some evidence that TonEBP regulates expression of UT-A in vivo; (1) during the renal development of the urinary concentrating ability, expression of TonEBP precedes that of UT-A1; (2) in transgenic mice expressing a dominant negative form of TonEBP, expression of UT-A1 and UT-A2 is severely impaired; (3) in treatment with cyclosporine A, TonEBP was significantly downregulated after 28 days. This downregulation involves mRNA levels of UT-A2; (4) in hypokalemic animals, downregulation of TonEBP contributed to the down regulation of UT-A in the inner medulla. These data support that TonEBP directly contributes to the urinary concentration and renal urea recycling by the regulation of urea transporters.
Collapse
Affiliation(s)
- Ju-Young Jung
- Department of Veterinary Medicine, Chungnam National University, Daejeon, Korea
| | - H Moo Kwon
- Department of Medicine, University of Maryland, Baltimore, MD, USA
| | - Jim Kim
- Department of Anatomy, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
18
|
|
19
|
Kim DU. Pathophysiology and management of disorders in water metabolism. KOREAN JOURNAL OF PEDIATRICS 2007. [DOI: 10.3345/kjp.2007.50.5.430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Dong Un Kim
- Department of Pediatrics, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
20
|
Chen G, Fröhlich O, Yang Y, Klein JD, Sands JM. Loss of N-linked glycosylation reduces urea transporter UT-A1 response to vasopressin. J Biol Chem 2006; 281:27436-42. [PMID: 16849333 DOI: 10.1074/jbc.m605525200] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The vasopressin-regulated urea transporter (UT)-A1 is a transmembrane protein with two glycosylated forms of 97 and 117 kDa; both are derived from a single 88-kDa core protein. However, the precise molecular sites and the function for UT-A1 N-glycosylation are not known. In this study, we compared Madin-Darby canine kidney cells stably expressing wild-type (WT) UT-A1 to Madin-Darby canine kidney cell lines stably expressing mutant UT-A1 lacking one (A1m1, A1m2) or both glycosylation sites (m1m2). Site-directed mutagenesis revealed that UT-A1 has two glycosylation sites at Asn-279 and -742. Urea flux is stimulated by 10 nM vasopressin (AVP) or 10 microM forskolin (FSK) in WT cells. In contrast, m1m2 cells have a delayed and significantly reduced maximal urea flux. A 15-min treatment with AVP and FSK significantly increased UT-A1 cell surface expression in WT but not in m1m2 cells, as measured by biotinylation. We confirmed this finding using immunostaining. Membrane fractionation of the plasma membrane, Golgi, and endoplasmic reticulum revealed that AVP or FSK treatment increases UT-A1 abundance in both Golgi and plasma membrane compartments in WT but not in m1m2 cells. Pulse-chase experiments showed that UT-A1 half-life is reduced in m1m2 cells compared with WT cells. Our results suggest that mutation of the N-linked glycosylation sites reduces urea flux by reducing UT-A1 half-life and decreasing its accumulation in the apical plasma membrane. In vivo, inner medullary collecting duct cells may regulate urea uptake by altering UT-A1 glycosylation in response to AVP stimulation.
Collapse
Affiliation(s)
- Guangping Chen
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia 30322, USA.
| | | | | | | | | |
Collapse
|
21
|
Jeon US, Kim JA, Sheen MR, Kwon HM. How tonicity regulates genes: story of TonEBP transcriptional activator. Acta Physiol (Oxf) 2006; 187:241-7. [PMID: 16734761 DOI: 10.1111/j.1748-1716.2006.01551.x] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
TonEBP stimulates genes whose products drive cellular accumulation of organic osmolytes and HSP70, which protect cells from the deleterious effects of hypertonicity and urea, respectively. Mice deficient in the TonEBP gene display severe atrophy of the renal medulla because cells failed to adapt to the hyperosmolality. Emerging data suggest that TonEBP plays a key role in the urinary concentrating mechanism by stimulating the UT-A urea transporters and possibly AQP2 water channel. Thus, TonEBP is an essential regulator in the urinary concentrating mechanism. Studies on structural basis of TonEBP function have revealed the structure of the DNA binding domain, and defined the transactivation domains. Molecular mechanisms underlying the nucleocytoplasmic trafficking, transactivation, and phosphorylation in response to changes in tonicity need to be understood in molecular detail. Such knowledge is needed for the identification of the sensor that detects changes in ambient tonicity and signals to TonEBP.
Collapse
Affiliation(s)
- U S Jeon
- Department of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | | | | | | |
Collapse
|
22
|
Abstract
To produce a concentrated urine, the renal medulla needs hypertonicity for the reabsorption of free water from collecting duct. The single effect that increases interstitial tonicity in the outer medulla is the active NaCl reabsorption in the thick ascending limb, while the single effect in the inner medulla is the passive efflux of NaCl through the thin ascending limb. The passive mechanism in the inner medulla requires high interstitial urea concentration. Two main groups of urea transporters (UT-A, UT-B) are present in the kidney, which maintains the high concentration of urea in the deepest portion of the inner medulla by intra-renal urea recycling. Recent studies suggest that UT-A1 in the terminal inner medullary collecting duct is up-regulated when urine or inner medullary interstitial urea is depleted in order to enhance the reabsorption of urea, while UT-A2 in the descending thin limb of loops of Henle and UT-B in the descending vasa recta are increased when outer medullary interstitial urea concentration is high, in order to prevent the loss of urea from the medulla to the systemic circulation, thereby increasing intra-renal urea recycling. This review will summarize the functions of the renal urea transporters in urine concentration mechanism and the recent knowledge about their long-term regulation.
Collapse
Affiliation(s)
- Dong-Un Kim
- Department of Pediatrics, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
23
|
Affiliation(s)
- Jeff M Sands
- Renal Division, Emory University School of Medicine, Atlanta, Georgia 30322, USA.
| | | |
Collapse
|
24
|
Lucien N, Bruneval P, Lasbennes F, Belair MF, Mandet C, Cartron JP, Bailly P, Trinh-Trang-Tan MM. UT-B1 urea transporter is expressed along the urinary and gastrointestinal tracts of the mouse. Am J Physiol Regul Integr Comp Physiol 2005; 288:R1046-56. [PMID: 15563580 DOI: 10.1152/ajpregu.00286.2004] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Selective transporters account for rapid urea transport across plasma membranes of several cell types. UT-B1 urea transporter is widely distributed in rat and human tissues. Because mice exhibit high urea turnover and are the preferred species for gene engineering, we have delineated UT-B1 tissue expression in murine tissues. A cDNA was cloned from BALB/c mouse kidney, encoding a polypeptide that differed from C57BL/6 mouse UT-B1 by one residue (Val-8-Ala). UT-B1 mRNA was detected by RT-PCR in brain, kidney, bladder, testis, lung, spleen, and digestive tract (liver, stomach, jejunum, colon). Northern blotting revealed seven UT-B1 transcripts in mouse tissues. Immunoblots identified a nonglycosylated UT-B1 protein of 29 kDa in most tissues and of 36 and 32 kDa in testis and liver, respectively. UT-B1 protein of gastrointestinal tract did not undergo N-glycosylation. Immunohistochemistry and in situ hybridization localized UT-B1 in urinary tract urothelium (papillary surface, ureter, bladder, and urethra), prominently on plasma membranes and restricted to the basolateral area in umbrella cells. UT-B1 was found in endothelial cells of descending vasa recta in kidney medulla and in astrocyte processes in brain. Dehydration induced by water deprivation for 2 days caused a tissue-specific decrease in UT-B1 abundance in the urinary bladder and the ureter.
Collapse
Affiliation(s)
- N Lucien
- INSERM U76, Institut National de Transfusion Sanguine, 6, rue Alexandre Cabanel, F-75015 Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Mistry AC, Chen G, Kato A, Nag K, Sands JM, Hirose S. A novel type of urea transporter, UT-C, is highly expressed in proximal tubule of seawater eel kidney. Am J Physiol Renal Physiol 2005; 288:F455-65. [PMID: 15383403 DOI: 10.1152/ajprenal.00296.2004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
A new type of urea transporter was identified by a database search and shown to be highly expressed in the renal proximal tubule cells of teleosts; proximal tubule-type urea transporters have not been describe previously. We first identified urea transporter-like sequences in the fugu genome and in an EST database of rainbow trout. Based on these pieces of sequence information, we obtained a full-length cDNA for the eel ortholog, consisting of 378 amino acid residues, and named it eUT-C. Although its sequence similarity to the known urea transporters is low (approximately 35%), its heterologous expression in Xenopus laevis oocytes indicated that it is a facilitative urea transporter sensitive to phloretin. Its activity is not dependent on Na+. Northern blot analysis showed that expression of eUT-C is highly restricted to the kidney, with weak expression in the stomach. In both tissues, eUT-C mRNA was strongly induced when eels were transferred from freshwater to seawater. Immunohistochemistry and in situ hybridization histochemistry revealed proximal tubule cell localization of eUT-C. Taking into account that 1) urea is mainly secreted from the gill where another type of urea transporter (eUT) has been identified and 2) fish excrete a very small volume of urine in seawater, we propose that eUT-C cloned here is a key component working in combination with the gill transporter to achieve an efficient urea excretory system in fish, namely, eUT-C reabsorbs urea from glomerular filtrate and sends it to the gill, through the circulation, for excretion.
Collapse
Affiliation(s)
- Abinash Chandra Mistry
- Dept. of Biological Sciences, Tokyo Institute of Technology, 4259-B-19 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | | | | | | | | | | |
Collapse
|
26
|
Han KH, Woo SK, Kim WY, Park SH, Cha JH, Kim J, Kwon HM. Maturation of TonEBP expression in developing rat kidney. Am J Physiol Renal Physiol 2004; 287:F878-85. [PMID: 15226152 DOI: 10.1152/ajprenal.00047.2004] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Tonicity-responsive enhancer binding protein (TonEBP) is a transcriptional activator of the Rel family. In the renal medulla, TonEBP stimulates genes encoding proteins involved in cellular accumulation of organic osmolytes, the vasopressin-regulated urea transporters (UT-A), and heat shock protein 70. To understand the role of TonEBP in the development of urinary concentrating ability, TonEBP expression during rat kidney development was investigated. In embryonic kidneys, TonEBP immunoreactivity was detected 16 days postcoitus in the cytoplasm of the endothelial cells surrounding the medullary collecting ducts (MCD). By 20 days, TonEBP was detected in most tubular profiles in the medulla, including the loop of Henle and MCD, and interstitial cells. The intensity of TonEBP immunoreactivity was much higher in the vasa recta than the tubules. In addition, immunoreactivity was localized predominantly to the cytoplasm. On postnatal day 1, two major changes were observed. TonEBP immunoreactivity shifted to the nucleus, and the intensity of TonEBP immunoreactivity of the tubules increased dramatically. These changes were associated with an increase in TonEBP and sodium-myo-inositol cotransporter mRNA abundance. Thereafter, TonEBP expression in tubular profiles increased moderately. The adult pattern of TonEBP expression was established at postnatal day 21 coincident with full maturation of the renal medulla. Thus expression of TonEBP in developing kidneys occurred predominantly in the medulla and preceded expression of its target genes, including UT-A. These data suggest that TonEBP contributes to the development of urine-concentrating ability.
Collapse
Affiliation(s)
- Ki-Hwan Han
- Departmrnt of Anatomy and Cell Death Disease Research Center, Catholic University Medical College, Seoul 137-701, South Korea
| | | | | | | | | | | | | |
Collapse
|
27
|
Li C, Klein JD, Wang W, Knepper MA, Nielsen S, Sands JM, Frøkiaer J. Altered expression of urea transporters in response to ureteral obstruction. Am J Physiol Renal Physiol 2004; 286:F1154-62. [PMID: 14982816 DOI: 10.1152/ajprenal.00453.2003] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Urea plays an important role in the urinary concentrating capacity. Renal inner medullary (IM) urea transporter expression was examined in rats with bilateral (BUO) or unilateral ureteral obstruction (UUO). BUO (24 h) was associated with markedly increased plasma urea (42.4 +/- 1.0 vs. 5.2 +/- 0.2 mmol/l) and a significant decrease in expression of UT-A1 (28 +/- 8% of sham levels), UT-A3 (45 +/- 11%), and UT-B (70 +/- 8%). Immunocytochemistry confirmed downregulation of UT-A1 and UT-A3 in IM collecting duct and UT-B in the descending vasa recta. Three days after release of BUO, UT-A1, UT-A3, and UT-B remained significantly downregulated (UT-A1: 37 +/- 6%; UT-A3: 25 +/- 6%; and UT-B: 10 +/- 5% of sham levels; P < 0.05) concurrent with a persistent polyuria and a marked reduction in solute-free water reabsorption (115 +/- 11 vs. 196 +/- 8 microl.min(-1).kg(-1), P < 0.05). Moreover, 14 days after release of BUO, total UT-A1, UT-A3, and UT-B remained significantly decreased compared with sham-operated controls and urine urea remained reduced (588 +/- 43 vs. 1,150 +/- 94 mmol/l). Consistent with increased levels of plasma urea 24 h after onset of UUO (7.4 +/- 0.3 vs. 4.8 +/- 0.3 mmol/l), the protein abundance of UT-A1, UT-A3, and UT-B in IM was markedly reduced in the obstructed kidney, which was confirmed by immunocytochemistry. In the nonobstructed kidney, the expression of urea transporters did not change. In conclusion, reduced expression of UT-A1, UT-A3, and UT-B levels in both BUO and UUO rats suggests that urea transporters play important roles in the impaired urinary concentrating capacity in response to urinary tract obstruction.
Collapse
Affiliation(s)
- Chunling Li
- The Water and Salt Research Center/Institute of Experimental Clinical Research, Aarhus Univ. Hospital-Skejby, DK-8200 Aarhus, Denmark
| | | | | | | | | | | | | |
Collapse
|
28
|
Trinh-Trang-Tan MM, Geelen G, Teillet L, Corman B. Urea transporter expression in aging kidney and brain during dehydration. Am J Physiol Regul Integr Comp Physiol 2003; 285:R1355-65. [PMID: 12933359 DOI: 10.1152/ajpregu.00207.2003] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Aging is commonly associated with defective urine-concentrating ability. The present study examined how the kidney and the brain of senescent (30-mo-old) female WAG/Rij rats respond to dehydration induced by 2 days of water deprivation in terms of urea transporter (UT) regulation. In euhydrated situation, senescent rats exhibited similar vasopressin plasma level but lower urine osmolality and papillary urea concentration and markedly reduced kidney UT-A1, UT-A3, and UT-B1 abundances compared with adult (10-mo-old) rats. Senescent rats responded to dehydration similarly to adult rats by a sixfold increase in vasopressin plasma level. Their papillary urea concentration was doubled, without, however, attaining that of dehydrated adult rats. Such an enhanced papillary urea sequestration occurred with a great fall of both UT-A1 and UT-A3 abundances in the tip of inner medulla and an increased UT-A1 abundance in the base of inner medulla. UT-A2 and UT-B1 were unchanged. These data suggest that the inability of control and thirsted senescent rats to concentrate urine as much as their younger counterparts derives from lower papillary urea concentration. In aging brain, UT-B1 abundance was increased twofold together with a fourfold increase in aquaporin-4 abundance. Dehydration did not alter the abundance of these transporters.
Collapse
Affiliation(s)
- M-M Trinh-Trang-Tan
- Institut National de la Santé et de la Recherche Médicale U76, Institut National de Transfusion Sanguine, 6, rue Alexandre Cabanel, F-75015 Paris, France.
| | | | | | | |
Collapse
|
29
|
Zimpelmann J, Li N, Burns KD. Nitric oxide inhibits superoxide-stimulated urea permeability in the rat inner medullary collecting duct. Am J Physiol Renal Physiol 2003; 285:F1160-7. [PMID: 12965888 DOI: 10.1152/ajprenal.00077.2003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The inner medullary collecting duct (IMCD) contains relatively high nitric oxide (NO) synthetic capacity, but the effect of NO on IMCD transport remains unclear. We determined the effect of NO on basal and vasopressin (AVP)-stimulated urea (Purea) and water (Pf) permeabilities in isolated, perfused rat IMCD. The NO donor S-nitroso-N-acetylpenicillamine (SNAP) increased cGMP production in IMCD, but neither SNAP (10(-4) M) nor 8-BrcGMP (10(-4) M), the cell-permeable analog of cGMP, affected basal or AVP-stimulated Purea. The free radical superoxide is produced by oxidases in the kidney and can interact with NO. To determine the effect of superoxide generation on transport, IMCDs were incubated with diethyldithiocarbamate (DETC; 10(-3) M), the inhibitor of superoxide dismutase (SOD). DETC significantly increased basal and AVP-stimulated Purea (control: 28.7 +/- 4.5 vs. DETC: 40.9 +/- 6.2 x 10(-5) cm/s; P < 0.001; n = 9). Preincubation of IMCD with SNAP or the SOD mimetic tempol completely inhibited DETC-stimulated Purea. DETC caused a significant increase in superoxide generation by IMCD, and this was blocked by SNAP. Incubation of IMCD with the NO synthase (NOS) substrate l-arginine blocked the stimulatory effect of DETC on Purea, and this was reversed by the neuronal NOS inhibitor 7-nitroindazole. In contrast, neither basal nor AVP-stimulated Pf was affected by NO donors or DETC. In summary, exogenous or endogenously produced NO does not affect basal urea transport in the IMCD but inhibits superoxide-stimulated Purea. In the inner medulla, superoxide generation by local oxidases may stimulate urea transport, and the role of endogenous NO may be to dampen this effect by decreasing superoxide levels.
Collapse
Affiliation(s)
- Joseph Zimpelmann
- Division of Nephrology, The Ottawa Hospital and University of Ottawa, 1967 Riverside Drive, Ottawa, Ontario, Canada K1H 7W9
| | | | | |
Collapse
|
30
|
Jung JY, Madsen KM, Han KH, Yang CW, Knepper MA, Sands JM, Kim J. Expression of urea transporters in potassium-depleted mouse kidney. Am J Physiol Renal Physiol 2003; 285:F1210-24. [PMID: 12952854 DOI: 10.1152/ajprenal.00111.2003] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Urea transport in the kidney is mediated by a family of transporter proteins that include the renal urea transporter (UT-A) and the erythrocyte urea transporter (UT-B). The purpose of this study was to determine the location of the urea transporter isoforms in the mouse kidney and to examine the effects of prolonged potassium depletion on the expression and distribution of these transporters by ultrastructural immunocytochemistry. C57BL6 mice were fed a low-potassium diet for 2 wk, and control animals received normal chow. After 2 wk on a low-potassium diet, urinary volume increased and urinary osmolality decreased (833 +/- 30 vs. 1,919 +/- 174 mosmol/kgH2O), as previously demonstrated. Kidneys were processed for immunocytochemistry with antibodies against UT-A1 (L446), UT-A1 and UT-A2 (L194), UT-A3 (Q2), and UT-B. In normal mice, UT-A1 and UT-A3 were expressed mainly in the cytoplasm of the terminal inner medullary collecting duct (IMCD). UT-A2 immunoreactivity was observed mainly on the basolateral membrane of the type 1 epithelium of the descending thin limb (DTL) of short-looped nephrons. The intensity of UT-A1 and UT-A3 immunoreactivity in the IMCD was markedly reduced in potassium-depleted mice. In contrast, there was a significant increase in UT-A2 immunoreactivity in the DTL. The intensity of UT-B immunoreactivity in the descending vasa recta (DVR) was reduced in potassium-depleted animals compared with controls. In control animals, UT-B immunoreactivity was predominantly observed in the plasma membrane, whereas in potassium-depleted mice, it was mainly observed in cytoplasmic granules in endothelial cells of the DVR. In summary, potassium depletion is associated with reduced expression of UT-A1, UT-A3, and UT-B but increased expression of UT-A2. We conclude that reduced expression of urea transporters may play a role in the impaired urine-concentrating ability associated with potassium deprivation.
Collapse
Affiliation(s)
- Ju-Young Jung
- Department of Anatomy, College of Medicine, The Catholic University of Korea, 505 Banpo-Dong, Socho-Gu, Seoul 137-701, Korea
| | | | | | | | | | | | | |
Collapse
|
31
|
Schumacher K, Strehl R, Minuth WW. Urea Restrains Aldosterone-Induced Development of Peanut Agglutinin–Binding on Embryonic Renal Collecting Duct Epithelia. J Am Soc Nephrol 2003; 14:2758-66. [PMID: 14569085 DOI: 10.1097/01.asn.0000090744.88722.ff] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
ABSTRACT. Peanut agglutinin (PNA) represents a commonly used marker for β-type intercalated (IC) cells and their distribution in the corticomedullary course of the collecting duct (CD) in the mature rabbit kidney. It has been shown that aldosterone is able to generate >90% of PNA-binding cells in an embryonic CD epithelium in vitro. In adult kidney, a maximum of only 25% PNA-positive cells is found in the cortical segment of the CD, and PNA-binding completely disappears in the inner-medullary CD. Molecules that regulate the gradual development of CD-specific cells during organ growth are unknown. In the present experiments, it was found that addition of physiologic concentrations of urea to the culture medium is able to restrain the action of aldosterone in embryonic CD epithelia. Urea antagonizes in a concentration-dependent manner the action of aldosterone finally leading to only 10% of PNA-binding cells. The data point to a urea-specific effect, because osmolytes such as NaCl and mannitol did not affect PNA binding. In addition, urea did not influence expression of principal-cell typical markers such as AQP2 and 3. The findings may explain that a higher number of PNA-positive cells is found in the cortical region of the kidney correlated with a low concentration of urea as compared with only few PNA-binding cells in the medullary CD, where a high concentration of urea occurs. Thus, an increasing concentration of urea may trigger the number of PNA-positive cells in the cortical-medullary course of the CD during organ development. E-mail: karl.schumacher@vkl.uni-regensburg.de
Collapse
Affiliation(s)
- Karl Schumacher
- Department of Molecular and Cellular Anatomy, University of Regensburg, Regensburg, Germany.
| | | | | |
Collapse
|
32
|
Kwun YS, Yeo SW, Ahn YH, Lim SW, Jung JY, Kim WY, Sands JM, Kim J. Immunohistochemical localization of urea transporters A and B in the rat cochlea. Hear Res 2003; 183:84-96. [PMID: 13679141 DOI: 10.1016/s0378-5955(03)00218-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Urea is present in the inner ear, and when it is administered, induces rapid changes in the volume and osmolality of the inner ear fluid. However, the regulating mechanisms are unknown. Two groups of urea transporters (UTs), the renal urea transporter (UT-A) and the erythrocyte urea transporter (UT-B) have been cloned recently. The aims of the current study were to investigate the cellular localization of UTs in the cochlea of male Sprague-Dawley rats by immunohistochemistry. Both UT-A1 and UT-B were expressed in the inner and outer pillar cells, inner and outer hair cells, Boettcher's cells, and Deiters' cells in the organ of Corti. Immunoreactivity for UT-A3 was localized only in the mesothelial cells underlying the basilar membrane. In the stria vascularis, UT-A1 was expressed only in the marginal cells, whereas UT-B was expressed only in the basal cells. In the spiral ganglion, most cells had strong UT-A1 immunoreactivity whereas UT-B was not expressed. In the spiral limbus, UT-B was expressed in the interdental cells whereas UT-A was not expressed. In the crista ampullaris, UT-A1 was expressed in the dark cells, and UT-B expressed in the apical membrane of supporting cells in the neuroepithelium. The distribution of UT-A and UT-B in the inner ear suggests that the cells that surround the inner ear fluids may be involved in urea transport and thus play an important role in fluid homeostasis in the inner ear. The expression of UT-A and UT-B in the hair cells raises the possibility that UTs may be involved in volume regulation in these cells and mediate hair cell turgor.
Collapse
Affiliation(s)
- Yong-Sig Kwun
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Kim D, Sands JM, Klein JD. Changes in renal medullary transport proteins during uncontrolled diabetes mellitus in rats. Am J Physiol Renal Physiol 2003; 285:F303-9. [PMID: 12697581 DOI: 10.1152/ajprenal.00438.2002] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We tested whether the abundance of transport proteins involved in the urinary concentrating mechanism was altered in rats with uncontrolled diabetes mellitus (DM). Rats were injected with streptozotocin and killed 5, 10, 14, or 20 days later. Blood glucose in DM rats was 300-450 mg/dl (control: 70-130 mg/dl). Urine volume increased in DM rats from 41 +/- 7 ml/100 g body wt (BW) at 5 days to 69 +/- 3 ml/100 g BW at 20 days (control: 9 +/- 1). Urine osmolality of DM rats decreased at 5 days DM and remained low at 20 days. UT-A1 urea transporter protein in the inner medullary (IM) tip was 55% of control in 5-day DM rats but increased to 170, 220, and 280% at 10, 14, and 20 days DM, respectively, due to an increase in the 117-kDa glycoprotein form. UT-A1 in the IM base was increased to 325% of control at 5 days DM with no further increase at 20 days. Aquaporin-2 (AQP2) increased to 290% in the IM base at 5 days DM and 150% in the IM tip at 10 days; both showed no further increase at 20 days. NKCC2/BSC1 increased to 240% in outer medulla at 20 days DM, but not at 5 or 10 days. UT-B and ROMK were unchanged at any time point. The increases in UT-A1, AQP2, and NKCC2/BSC1 proteins during uncontrolled DM would tend to limit the loss of fluid and solute during uncontrolled diabetes.
Collapse
Affiliation(s)
- Dongun Kim
- Renal Division, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | |
Collapse
|
34
|
Faber P, Johnstone AM, Gibney ER, Elia M, Stubbs RJ, Roger PL, Milne E, Buchan W, Lobley GE. The effect of rate and extent of weight loss on urea salvage in obese male subjects. Br J Nutr 2003; 90:221-31. [PMID: 12844395 DOI: 10.1079/bjn2003859] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
It is well established that in human subjects a proportion of urea production undergoes hydrolysis in the gastrointestinal tract with release of N potentially available for amino acid synthesis. Previous studies have suggested adaptive changes in urea kinetics, with more urea-N retained within the metabolic pool during reduced dietary intakes of energy and protein. We therefore investigated the effect of rate and extent of weight loss on adaptive changes in urea kinetics in two groups (each n 6) of obese men (mean age 43 (sd 12) years, BMI 34.8 (sd 2.9) kg/m(2)) during either total starvation for 6 d or a very-low-energy diet (2.55 MJ/d) for 21 d. Subjects were resident in the Human Nutrition Unit of the Rowett Research Institute (Aberdeen, Scotland, UK) and lost 6 and 9 % initial body weight within the starvation and dieting groups respectively. Changes in urea-N metabolism were assessed by stable isotope tracer kinetics using [(15)N(15)N]urea infused intravenously for 36 h before, during and after weight loss. In response to weight loss, urea production decreased (P<0.01) by 25 % from 278 to 206 micromol urea-N/h per kg within the dieting group only. However, no changes were observed in the proportion of urea being hydrolysed in the gastrointestinal tract (range 20-25 %) or in the proportion of N retained for anabolic purposes (80-85 % urea-N from gastrointestinal hydrolysis) within either group. It was concluded that no adaptive changes in urea kinetics occurred in response to either the different rate or extent of weight loss.
Collapse
Affiliation(s)
- Peter Faber
- Rowett Research Institute, Greenburn Road, Bucksburn, Aberdeen AB21 9SB, Scotland, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Christensen BM, Wang W, Frøkiaer J, Nielsen S. Axial heterogeneity in basolateral AQP2 localization in rat kidney: effect of vasopressin. Am J Physiol Renal Physiol 2003; 284:F701-17. [PMID: 12453871 DOI: 10.1152/ajprenal.00234.2002] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The purpose of the present study was to examine whether there is axial heterogeneity in the basolateral plasma membrane (BLM) localization of AQP2 and whether altered vasopressin action or medullary tonicity affects the BLM localization of AQP2. Immunocytochemistry and immunoelectron microscopy revealed AQP2 labeling of the BLM in connecting tubule (CNT) cells and inner medullary collecting duct (IMCD) principal cells in normal rats and vasopressin-deficient Brattleboro rats. In contrast there was little basolateral AQP2 labeling in cortical (CCD) and outer medullary collecting duct principal cells. Short-term desamino-Cys(1), (D)-Arg(8) vasopressin (dDAVP) treatment (2 h) of Brattleboro rats caused no increase in AQP2 labeling of the BLM. In contrast, long-term dDAVP treatment (6 days) of Brattleboro rats caused an increased BLM labeling in CNT, CCD, and IMCD. Treatment of normal rats with V(2)-receptor antagonist for 60 min caused retrieval of AQP2 from the apical plasma membrane. Moreover, AQP2 labeling of the BLM was unchanged in CNT and IMCD but increased in CCD. In conclusion, there is an axial heterogeneity in the subcellular localization of AQP2 with prominent AQP2 labeling of the BLM in CNT and IMCD. There was no increase in AQP2 labeling of the BLM in response to short-term dDAVP. Moreover, acute V(2)-receptor antagonist treatment did not cause retrieval of AQP2 from the BLM. In contrast, long-term dDAVP treatment caused a major increase in AQP2 expression in the BLM in CCD.
Collapse
|
36
|
Minocha R, Studley K, Saier MH. The urea transporter (UT) family: bioinformatic analyses leading to structural, functional, and evolutionary predictions. RECEPTORS & CHANNELS 2003; 9:345-52. [PMID: 14698962 DOI: 10.3109/714041015] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We have identified all currently sequenced members of the urea transporter (UT) family (TC #1.A.28). Homologues occur exclusively in vertebrate animals and bacteria but not in other eukaryotic kingdoms or archaea. Sequence, structural, and phylogenetic analyses reveal conserved regions and residues and suggest that a primordial 5 transmembrane helical segment (TMS)-encoding genetic element duplicated to give a 10 TMS-encoding element early during evolutionary history, at about the time when eukaryotes diverged from prokaryotes. Two well-conserved, strongly amphipathic, putative alpha-helices that precede both 5 TMS repeat elements are predicted to be of structural, functional, or biogenic significance. A second duplication event (or a gene fusion event) occurred during development of the vertebrate lineage, giving rise to 20 TMS mammalian homologues. The results suggest that vertebrates acquired UT genetic information from bacteria only once and that all current orthologues and paralogues in the animal kingdom arose from this one primordial system.
Collapse
Affiliation(s)
- Ranjeet Minocha
- Division of Biological Sciences, University of California at San Diego, La Jolla, CA 92093-0116, USA
| | | | | |
Collapse
|