1
|
Esteva-Font C, Zheng X, Rudolphi CF, Hansen TPK, Hoorn EJ, Fenton RA. Deletion of the prostaglandin EP4 receptor in the kidney tubule of mature mice alters kidney water handling only in males. Am J Physiol Renal Physiol 2025; 328:F800-F814. [PMID: 40261807 DOI: 10.1152/ajprenal.00071.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/20/2025] [Accepted: 04/16/2025] [Indexed: 04/24/2025] Open
Abstract
Prostaglandin E2 (PGE2) is a lipid mediator modulating several aspects of kidney function. PGE2 acts via four prostanoid receptors, EP1 to EP4. In renal tubules, EP4 has very low expression, yet a role for EP4 in maintaining water balance has been proposed. The major aim of this study was to clarify the role of the EP4 receptor in the kidney tubule of adult mice for body water homeostasis. To examine this, a mouse model with doxycycline-dependent deletion of EP4 along the renal tubule (Pax8Cre system) was developed and phenotyped with respect to water handling. Two weeks after doxycycline treatment, EP4 mRNA expression (RT-qPCR) was reduced by >80% in the medulla of male and female knockout mice (EP4-/-) compared with controls (EP4+/+). With free access to water, there were no detectable differences between genotypes in food intake, body weight, or plasma biochemistries. Male, but not female, EP4-/- mice had a small but significantly higher basal urinary output with decreased osmolality, concomitant with lower urinary Na+, K+, Cl-, urea, and creatinine concentrations. The urea channel UT-A1 was reduced in the medulla, but otherwise no major differences in the levels of proteins involved in water balance were observed between genotypes. There were no differences between genotypes in their ability to concentrate urine during a 14-h water restriction or after treatment with the vasopressin V2 receptor agonist 1-deamino-8-d-arginine-vasopressin. EP4-/- mice of both sexes excreted an acute water load similarly to control mice. In conclusion, EP4 in the renal tubule has a mild role in renal water handling, but only in male mice. This new model provides a novel tool for assessing the role of EP4 in kidney tubule function in various (patho)physiological conditions independently of developmental abnormalities or systemic effects.NEW & NOTEWORTHY Acting through the EP1-EP4 receptors, prostaglandin E2 (PGE2) modulates various aspects of kidney function. Despite extremely low expression in the collecting duct, EP4 has been proposed to play a key role in regulation of Aquaporin-2 and water balance. In this study, we show that EP4 only plays a mild role in kidney water handling in male mice, calling into question the standard concepts presented in physiology textbooks regarding PGE2 actions on the collecting duct.
Collapse
Affiliation(s)
| | - Xiang Zheng
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Crissy F Rudolphi
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Toke P K Hansen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Ewout J Hoorn
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Robert A Fenton
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
2
|
Luo Y, Sheikh TMM, Li X, Yuan Y, Yao F, Wang M, Guo X, Wu J, Shafiq M, Xie Q, Jiao X. Exploring the dynamics of gut microbiota, antibiotic resistance, and chemotherapy impact in acute leukemia patients: A comprehensive metagenomic analysis. Virulence 2024; 15:2428843. [PMID: 39620486 PMCID: PMC11622590 DOI: 10.1080/21505594.2024.2428843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 08/24/2024] [Accepted: 11/06/2024] [Indexed: 12/08/2024] Open
Abstract
Leukemia poses significant challenges to its treatment, and understanding its complex pathogenesis is crucial. This study used metagenomic sequencing to investigate the interplay between chemotherapy, gut microbiota, and antibiotic resistance in patients with acute leukemia (AL). Pre- and post-chemotherapy stool samples from patients revealed alterations in microbial richness, taxa, and antibiotic resistance genes (ARGs). The analysis revealed a decreased alpha diversity, increased dispersion in post-chemotherapy samples, and changes in the abundance of specific bacteria. Key bacteria such as Enterococcus, Klebsiella, and Escherichia coli have been identified as prevalent ARG carriers. Correlation analysis between gut microbiota and blood indicators revealed potential links between microbial species and inflammatory biomarkers, including C-reactive protein (CRP) and adenosine deaminase (ADA). This study investigated the impact of antibiotic dosage on microbiota and ARGs, revealing networks connecting co-occurring ARGs with microbial species (179 nodes, 206 edges), and networks associated with ARGs and antibiotic dosages (50 nodes, 50 edges). Antibiotics such as cephamycin and sulfonamide led to multidrug-resistant Klebsiella colonization. Our analyses revealed distinct microbial profiles with Salmonella enterica elevated post-chemotherapy in NF patients and Akkermansia muciniphila elevated pre-chemotherapy. These microbial signatures could inform strategies to modulate the gut microbiome, potentially mitigating the risk of neutropenic fever in patients undergoing chemotherapy. Finally, a comprehensive analysis of KEGG modules shed light on disrupted metabolic pathways after chemotherapy, providing insights into potential targets for managing side effects. Overall, this study revealed intricate relationships between gut microbiota, chemotherapy, and antibiotic resistance, providing new insights into improving therapy and enhancing patient outcomes.
Collapse
Affiliation(s)
- Ying Luo
- Department of Laboratory Medicine, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | | | - Xin Li
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - YuMeng Yuan
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Fen Yao
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Meimei Wang
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Xiaoling Guo
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Jilong Wu
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Muhammad Shafiq
- Research Institute of Clinical Pharmacy, Shantou University Medical College, Shantou, China
| | - Qingdong Xie
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Xiaoyang Jiao
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| |
Collapse
|
3
|
Yin H, Zhong Y, Wang H, Hu J, Xia S, Xiao Y, Nie S, Xie M. Short-term exposure to high relative humidity increases blood urea and influences colonic urea-nitrogen metabolism by altering the gut microbiota. J Adv Res 2022; 35:153-168. [PMID: 35003799 PMCID: PMC8721250 DOI: 10.1016/j.jare.2021.03.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 03/09/2021] [Indexed: 01/20/2023] Open
Abstract
Plasma urea was increased along with erythrocyte Na+/K+ -ATPase activity reduced and abnormal erythrocyte morphologies appeared during 14-day high relative humidity (90 ± 2%) exposure. Shortly after 12-h and 24-h exposures, urea influx and ammonia level were increased in the colon of mice, respectively. Colonic urea-nitrogen metabolism was influenced by the increased levels of ammonia, amino acids and short-chain fatty acids during 14-day exposure. Gut bacteria related to urease production, amino acids metabolism and SCFAs production was enriched during the exposure.
Introduction Colonic urea-nitrogen metabolites have been implicated in the pathogenesis of certain diseases which can be affected by environmental factors. Objectives We aimed to explore the influence of ambient humidity on colonic urea-nitrogen metabolism. Methods Blood biochemical indexes, metabolites of intestinal tract, and gut microbiota composition of mice (n = 10/group) exposed to high relative humidity (RH, 90 ± 2%) were analyzed during the 14-day exposure. Results After 12-h exposure, plasma blood urea nitrogen (BUN) level increased along with a decrease in the activity of erythrocyte Na+/K+ -ATPase. Moreover, abnormal erythrocyte morphologies appeared after 3 days of exposure. The colonic BUN and ammonia levels increased significantly after the 12-h and 24-h exposure, respectively. The colonic level of amino acids, partly synthesized by gut microbiota using ammonia as the nitrogen source, was significantly higher on the 7th day. Furthermore, the level of fecal short-chain fatty acids was significantly higher after 3-day exposure and the level of branched-chain fatty acids increased on the 14th day. Overall, gut microbiota composition was continuously altered during exposure, facilitating the preferential proliferation of urea-nitrogen metabolism bacteria. Conclusion Our findings suggest that short-term high RH exposure influences colonic urea-nitrogen metabolism by increasing the influx of colonic urea and altering gut microbiota, which might further impact the host health outcomes.
Collapse
Affiliation(s)
- Hongmei Yin
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| | - Yadong Zhong
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| | - Hui Wang
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| | - Jielun Hu
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| | - Shengkun Xia
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| | - Yuandong Xiao
- The College of National Land Resource and Environment, Jiangxi Agriculture University, Nanchang, Jiangxi 330045, China
| | - Shaoping Nie
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| | - Mingyong Xie
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.,National R&D Center for Freshwater Fish Processing, Jiangxi Normal University, Nanchang, Jiangxi 330022, China
| |
Collapse
|
4
|
Souza VC, Aguilar M, Van Amburgh M, Nayananjalie WAD, Hanigan MD. Milk urea nitrogen variation explained by differences in urea transport into the gastrointestinal tract in lactating dairy cows. J Dairy Sci 2021; 104:6715-6726. [PMID: 33773787 DOI: 10.3168/jds.2020-19787] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 02/13/2021] [Indexed: 01/30/2023]
Abstract
Milk urea nitrogen (MUN) and blood urea nitrogen are correlated with nitrogen balance and nitrogen excretion; however, there is also a genetic component to MUN concentrations that could be associated with differences in urea transport. It was hypothesized that a portion of the variation in MUN concentrations among cows is caused by variation in gastrointestinal and kidney urea clearance rates. Eight lactating cows with varying MUN concentrations while fed a common diet were infused with [15N15N]urea to determine urea N entry rate (UER), gastrointestinal entry rate, returned to ornithine cycle, urea N used for anabolism, urea N excretion in feces and urine. Urea clearance rates by the kidneys and gastrointestinal tract were calculated from isotopic enrichment of urea excretion in urine and gut entry rate, respectively, and plasma urea N concentrations (PUN). Over the course of the experiment, animals weighed an average of 506 ± 62 kg and produced 26.3 ± 4.39 kg of milk/d, with MUN concentrations ranging from 11.6 to 17.3 mg/dL (average of 14.9 ± 2.1 mg/dL). Plasma urea N was positively correlated with UER, urea N excretion in urine, and urea N used for anabolism. Plasma urea N and MUN were negatively correlated with gut clearance rates and ratio of gastrointestinal entry rate to UER. This relationship supports the hypothesis that differences in gut urea transport activity among animals causes variation in PUN and MUN concentrations, and that cows with high PUN and MUN are less efficient at recycling PUN to the gastrointestinal tract and thus may be more susceptible to ruminal N deficiencies when fed low RDP diets. Such biological variation in urea metabolism necessitates an adequate safety margin when setting regulations for maximal MUN levels as an indicator of herd N efficiency.
Collapse
Affiliation(s)
- V C Souza
- Department of Dairy Science, Virginia Polytechnic Institute and State University, Blacksburg 24061
| | - M Aguilar
- Department of Dairy Science, Virginia Polytechnic Institute and State University, Blacksburg 24061
| | - M Van Amburgh
- Department of Animal Science, Cornell University, Ithaca, NY 14853
| | - W A D Nayananjalie
- Department of Animal and Food Sciences, Faculty of Agriculture, Rajarata University of Sri Lanka, Puliyankulama, Anuradhapura, Sri Lanka 61316
| | - M D Hanigan
- Department of Dairy Science, Virginia Polytechnic Institute and State University, Blacksburg 24061.
| |
Collapse
|
5
|
Xie L, Jin L, Feng J, Lv J. The Expression of AQP5 and UTs in the Sweat Glands of Uremic Patients. BIOMED RESEARCH INTERNATIONAL 2017; 2017:8629783. [PMID: 29279852 PMCID: PMC5723962 DOI: 10.1155/2017/8629783] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 10/17/2017] [Accepted: 11/09/2017] [Indexed: 02/04/2023]
Abstract
PURPOSE To research the distribution and quantitative changes of UT-A1, UT-B1, and AQP5 in uremic skin tissue. METHODS 34 cases of uremic patients (UP) and 11 controls were recruited. Immunohistochemistry, immunofluorescence, RT-PCR, and Western Blot were used to identify the proteins in sweat glands. RESULTS AQP5, UT-A1, and UT-B1 were expressed and localized in human skin basal lines, skin sweat glands, and sweat ducts, both in UP and controls. Compared to controls, AQP5 mRNA abundance was significantly decreased in UP (P < 0.01), and, with the decrease of eGFR, the AQP5 expression was significantly decreased (P < 0.05). By contrast, UT-A1 and UT-B1 mRNA abundance was significantly increased in the skin of UP compared with the control (P < 0.01), and, with the decrease of eGFR, the AQP5 expression was significantly increased (P < 0.05). We found that the gene changes were coincident with the corresponding target proteins. The urea transporter subtypes, UT-A1 and UT-B1, were expressed in the skin basal cell layer and exocrine sweat glands. The abundance of UT-A1 and UT-B1 in uremic sweat glands was significantly increased in UP, while the expression of AQP5 was decreased. CONCLUSION Elimination of urea through the skin by producing sweat is a potential therapeutic strategy for renal failure patients.
Collapse
Affiliation(s)
- Liyi Xie
- Department of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Li Jin
- Department of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jie Feng
- Department of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jing Lv
- Department of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| |
Collapse
|
6
|
Abstract
Some unicellular organisms can take up urea from the surrounding fluids by an uphill pumping mechanism. Several active (energy-dependent) urea transporters (AUTs) have been cloned in these organisms. Functional studies show that active urea transport also occurs in elasmobranchs, amphibians, and mammals. In the two former groups, active urea transport may serve to conserve urea in body fluids in order to balance external high ambient osmolarity or prevent desiccation. In mammals, active urea transport may be associated with the need to either store and/or reuse nitrogen in the case of low nitrogen supply, or to excrete nitrogen efficiently in the case of excess nitrogen intake. There are probably two different families of AUTs, one with a high capacity able to establish only a relatively modest transepithelial concentration difference (renal tubule of some frogs, pars recta of the mammalian kidney, early inner medullary collecting duct in some mammals eating protein-poor diets) and others with a low capacity but able to maintain a high transepithelial concentration difference that has been created by another mechanism or in another organ (elasmobranch gills, ventral skin of some toads, and maybe mammalian urinary bladder). Functional characterization of these transporters shows that some are coupled to sodium (symports or antiports) while others are sodium-independent. In humans, only one genetic anomaly, with a mild phenotype (familial azotemia), is suspected to concern one of these transporters. In spite of abundant functional evidence for such transporters in higher organisms, none have been molecularly identified yet.
Collapse
Affiliation(s)
- Lise Bankir
- INSERM UMRS 1138, Centre de Recherche Des Cordeliers, Paris, France,
| |
Collapse
|
7
|
Davila AM, Blachier F, Gotteland M, Andriamihaja M, Benetti PH, Sanz Y, Tomé D. Intestinal luminal nitrogen metabolism: Role of the gut microbiota and consequences for the host. Pharmacol Res 2013. [DOI: 10.1016/j.phrs.2012.11.005] [Citation(s) in RCA: 344] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
8
|
Davila AM, Blachier F, Gotteland M, Andriamihaja M, Benetti PH, Sanz Y, Tomé D. Re-print of "Intestinal luminal nitrogen metabolism: role of the gut microbiota and consequences for the host". Pharmacol Res 2013; 69:114-26. [PMID: 23318949 DOI: 10.1016/j.phrs.2013.01.003] [Citation(s) in RCA: 155] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Alimentary and endogenous proteins are mixed in the small intestinal lumen with the microbiota. Although experimental evidences suggest that the intestinal microbiota is able to incorporate and degrade some of the available amino acids, it appears that the microbiota is also able to synthesize amino acids raising the view that amino acid exchange between the microbiota and host can proceed in both directions. Although the net result of such exchanges remains to be determined, it is likely that a significant part of the amino acids recovered from the alimentary proteins are used by the microbiota. In the large intestine, where the density of bacteria is much higher than in the small intestine and the transit time much longer, the residual undigested luminal proteins and peptides can be degraded in amino acids by the microbiota. These amino acids cannot be absorbed to a significant extent by the colonic epithelium, but are precursors for the synthesis of numerous metabolic end products in reactions made by the microbiota. Among these products, some like short-chain fatty acids and organic acids are energy substrates for the colonic mucosa and several peripheral tissues while others like sulfide and ammonia can affect the energy metabolism of colonic epithelial cells. More work is needed to clarify the overall effects of the intestinal microbiota on nitrogenous compound metabolism and consequences on gut and more generally host health.
Collapse
Affiliation(s)
- Anne-Marie Davila
- UMR 914 INRA/AgroParisTech, Nutrition Physiology and Ingestive Behavior, Paris, France.
| | | | | | | | | | | | | |
Collapse
|
9
|
Stewart GS, Smith CP. Urea nitrogen salvage mechanisms and their relevance to ruminants, non-ruminants and man. Nutr Res Rev 2012; 18:49-62. [PMID: 19079894 DOI: 10.1079/nrr200498] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Maintaining a correct balance of N is essential for life. In mammals, the major sources of N in the diet are amino acids and peptides derived from ingested proteins. The immediate endproduct of mammalian protein catabolism is ammonia, which is toxic to cells if allowed to accumulate. Therefore, amino acids are broken down in the liver as part of the ornithine-urea cycle, which results in the formation of urea - a highly soluble, biochemically benign molecule. Mammals cannot break down urea, which is traditionally viewed as a simple waste product passed out in the urine. However, urea from the bloodstream can pass into the gastrointestinal tract, where bacteria expressing urease cleave urea into ammonia and carbon dioxide. The bacteria utilise the ammonia as an N source, producing amino acids and peptides necessary for growth. Interestingly, these microbial products can be reabsorbed back into the host mammalian circulation and used for synthetic processes. This entire process is known as 'urea nitrogen salvaging' (UNS). In this review we present evidence supporting a role for this process in mammals - including ruminants, non-ruminants and man. We also explore the possible mechanisms involved in UNS, including the role of specialised urea transporters.
Collapse
Affiliation(s)
- Gavin S Stewart
- School of Biological Sciences, Stopford Building, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | | |
Collapse
|
10
|
Spector DA, Deng J, Stewart KJ. Dietary protein affects urea transport across rat urothelia. Am J Physiol Renal Physiol 2012; 303:F944-53. [DOI: 10.1152/ajprenal.00238.2012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Recent evidence suggests that regulated solute transport occurs across mammalian lower urinary tract epithelia (urothelia). To study the effects of dietary protein on net urothelial transport of urea, creatinine, and water, we used an in vivo rat bladder model designed to mimic physiological conditions. We placed groups of rats on 3-wk diets differing only by protein content (40, 18, 6, and 2%) and instilled 0.3 ml of collected urine in the isolated bladder of anesthetized rats. After 1 h dwell, retrieved urine volumes were unchanged, but mean urea nitrogen (UN) and creatinine concentrations fell 17 and 4%, respectively, indicating transurothelial urea and creatinine reabsorption. The fall in UN (but not creatinine) concentration was greatest in high protein (40%) rats, 584 mg/dl, and progressively less in rats receiving lower protein content: 18% diet, 224 mg/dl; 6% diet, 135 mg/dl; and 2% diet, 87 mg/dl. The quantity of urea reabsorbed was directly related to a urine factor, likely the concentration of urea in the instilled urine. In contrast, the percentage of instilled urea reabsorbed was greater in the two dietary groups receiving the lowest protein (26 and 23%) than in those receiving higher protein (11 and 9%), suggesting the possibility that a bladder/urothelial factor, also affected by dietary protein, may have altered bladder permeability. These findings demonstrate significant regulated urea transport across the urothelium, resulting in alteration of urine excreted by the kidneys, and add to the growing evidence that the lower urinary tract may play an unappreciated role in mammalian solute homeostasis.
Collapse
Affiliation(s)
- David A. Spector
- Division of Renal Medicine, Johns Hopkins Bayview Medical Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Jie Deng
- Division of Renal Medicine, Johns Hopkins Bayview Medical Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Kerry J. Stewart
- Division of Cardiology, Johns Hopkins Bayview Medical Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
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: 115] [Impact Index Per Article: 8.8] [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
|
Abstract
Urea transport proteins were initially proposed to exist in the kidney in the late 1980s when studies of urea permeability revealed values in excess of those predicted by simple lipid-phase diffusion and paracellular transport. Less than a decade later, the first urea transporter was cloned. Currently, the SLC14A family of urea transporters contains two major subgroups: SLC14A1, the UT-B urea transporter originally isolated from erythrocytes; and SLC14A2, the UT-A group with six distinct isoforms described to date. In the kidney, UT-A1 and UT-A3 are found in the inner medullary collecting duct; UT-A2 is located in the thin descending limb, and UT-B is located primarily in the descending vasa recta; all are glycoproteins. These transporters are crucial to the kidney's ability to concentrate urine. UT-A1 and UT-A3 are acutely regulated by vasopressin. UT-A1 has also been shown to be regulated by hypertonicity, angiotensin II, and oxytocin. Acute regulation of these transporters is through phosphorylation. Both UT-A1 and UT-A3 rapidly accumulate in the plasma membrane in response to stimulation by vasopressin or hypertonicity. Long-term regulation involves altering protein abundance in response to changes in hydration status, low protein diets, adrenal steroids, sustained diuresis, or antidiuresis. Urea transporters have been studied using animal models of disease including diabetes mellitus, lithium intoxication, hypertension, and nephrotoxic drug responses. Exciting new animal models are being developed to study these transporters and search for active urea transporters. Here we introduce urea and describe the current knowledge of the urea transporter proteins, their regulation, and their role in the kidney.
Collapse
Affiliation(s)
- Janet D Klein
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia, USA
| | | | | |
Collapse
|
13
|
Tickle P, Thistlethwaite A, Smith CP, Stewart GS. Novel bUT-B2 urea transporter isoform is constitutively activated. Am J Physiol Regul Integr Comp Physiol 2009; 297:R323-9. [PMID: 19474392 DOI: 10.1152/ajpregu.00199.2009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Our previous studies have detailed a novel facilitative UT-B urea transporter isoform, bUT-B2. Despite the existence of mouse and human orthologs, the functional characteristics of UT-B2 remain undefined. In this report, we produced a stable MDCK cell line that expressed bUT-B2 protein and investigated the transepithelial urea flux across cultured cell monolayers. We observed a large basal urea flux that was significantly reduced by known inhibitors of facilitative urea transporters; 1,3 dimethylurea (P < 0.001, n = 17), thionicotinamide (P < 0.05, n = 11), and phloretin (P < 0.05, n = 9). Pre-exposure for 1 h to the antidiuretic hormone vasopressin had no effect on bUT-B2-mediated urea transport (NS, n = 3). Acute vasopressin exposure for up to 30 min also failed to elicit any transient response (NS, n = 9). Further investigation confirmed that bUT-B2 function was not affected by alteration of intracellular cAMP (NS, n = 4), intracellular calcium (NS, n = 3), or protein kinase activity (NS, n = 4). Finally, immunoblot data suggested a possible role for glycosylation in regulating bUT-B2 function. In conclusion, this study showed that bUT-B2-mediated transepithelial urea transport was constitutively activated and unaffected by known regulators of renal UT-A urea transporters.
Collapse
Affiliation(s)
- P Tickle
- Faculty of Life Sciences, The University of Manchester, Manchester, UK
| | | | | | | |
Collapse
|
14
|
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
|
15
|
Kim YM, Kim WY, Lee HW, Kim J, Kwon HM, Klein JD, Sands JM, Kim D. Urea and NaCl regulate UT-A1 urea transporter in opposing directions via TonEBP pathway during osmotic diuresis. Am J Physiol Renal Physiol 2009; 296:F67-77. [PMID: 18945830 PMCID: PMC2636911 DOI: 10.1152/ajprenal.00143.2008] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2008] [Accepted: 10/08/2008] [Indexed: 01/23/2023] Open
Abstract
In our previous studies of varying osmotic diuresis, UT-A1 urea transporter increased when urine and inner medullary (IM) interstitial urea concentration decreased. The purposes of this study were to examine 1) whether IM interstitial tonicity changes with different urine urea concentrations during osmotic dieresis and 2) whether the same result occurs even if the total urinary solute is decreased. Rats were fed a 4% high-salt diet (HSD) or a 5% high-urea diet (HUD) for 2 wk and compared with the control rats fed a regular diet containing 1% NaCl. The urine urea concentration decreased in HSD but increased in HUD. In the IM, UT-A1 and UT-A3 urea transporters, CLC-K1 chloride channel, and tonicity-enhanced binding protein (TonEBP) transcription factor were all increased in HSD and decreased in HUD. Next, rats were fed an 8% low-protein diet (LPD) or a 0.4% low-salt diet (LSD) to decrease the total urinary solute. Urine urea concentration significantly decreased in LPD but significantly increased in LSD. Rats fed the LPD had increased UT-A1 and UT-A3 in the IM base but decreased in the IM tip, resulting in impaired urine concentrating ability. The LSD rats had decreased UT-A1 and UT-A3 in both portions of the IM. CLC-K1 and TonEBP were unchanged by LPD or LSD. We conclude that changes in CLC-K1, UT-A1, UT-A3, and TonEBP play important roles in the renal response to osmotic diuresis in an attempt to minimize changes in plasma osmolality and maintain water homeostasis.
Collapse
Affiliation(s)
- Yu-Mi Kim
- Dept. of Pediatrics, The Catholic Univ. of Korea, Uijeongbu St. Mary's Hospital, 65-1 Kumoh-Dong, Uijeongbu 480-717, Republic of Korea.
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Colonic luminal ammonia and portal blood l-glutamine and l-arginine concentrations: a possible link between colon mucosa and liver ureagenesis. Amino Acids 2008; 37:751-60. [DOI: 10.1007/s00726-008-0218-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2008] [Accepted: 11/18/2008] [Indexed: 11/27/2022]
|
17
|
Kane S. Just a spoonful of sugar helps the medicine go down... If only it was that simple! nonadherence in inflammatory bowel disease. Am J Gastroenterol 2007; 102:1427-8. [PMID: 17593159 DOI: 10.1111/j.1572-0241.2007.01213.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Those factors that are associated with medication nonadherence are complex and multifaceted. Previous work has been derived from tertiary referral centers and has focused on ulcerative colitis. Ediger et al. have studied a population-based cohort of IBD patients, with information on disease activity, medications, as well as psychological factors that are associated with medication-taking behavior. They have demonstrated some new gender-based differences for adherence, and it is clear that disease activity and diagnosis are also important factors.
Collapse
|
18
|
Smith CP, Fenton RA. Genomic organization of the mammalian SLC14a2 urea transporter genes. J Membr Biol 2007; 212:109-17. [PMID: 17264986 DOI: 10.1007/s00232-006-0870-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2006] [Indexed: 11/28/2022]
Abstract
Urea transporters encoded by the UT-A gene play fundamental roles in the kidney and possibly other tissues. Knowledge of the genomic organization of the mouse, rat and human UT-A genes has enabled the engineering of transgenic and knockout animals and these have helped refine our understanding of the role of UT-A proteins. This review summarizes the published work that has accrued on the structure and regulation of these genes. It also documents a novel cDNA, human UT-A3, which has enabled a major refinement of the human UT-A gene structure. This and other information contained in this review should prove useful for future comparative genomic analysis, studies addressing gene regulation and for the engineering of transgenic and knockout animal strains.
Collapse
Affiliation(s)
- C P Smith
- Faculty of Life Sciences, The University of Manchester, Oxford Road, Manchester, M13 9NT, UK.
| | | |
Collapse
|
19
|
Blachier F, Mariotti F, Huneau JF, Tomé D. Effects of amino acid-derived luminal metabolites on the colonic epithelium and physiopathological consequences. Amino Acids 2006; 33:547-62. [PMID: 17146590 DOI: 10.1007/s00726-006-0477-9] [Citation(s) in RCA: 320] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2006] [Accepted: 11/09/2006] [Indexed: 02/07/2023]
Abstract
Depending on the amount of alimentary proteins, between 6 and 18 g nitrogenous material per day enter the large intestine lumen through the ileocaecal junction. This material is used as substrates by the flora resulting eventually in the presence of a complex mixture of metabolites including ammonia, hydrogen sulfide, short and branched-chain fatty acids, amines; phenolic, indolic and N-nitroso compounds. The beneficial versus deleterious effects of these compounds on the colonic epithelium depend on parameters such as their luminal concentrations, the duration of the colonic stasis, the detoxication capacity of epithelial cells in response to increase of metabolite concentrations, the cellular metabolic utilization of these metabolites as well as their effects on colonocyte intermediary and oxidative metabolism. Furthermore, the effects of metabolites on electrolyte movements through the colonic epithelium must as well be taken into consideration for such an evaluation. The situation is further complicated by the fact that other non-nitrogenous compounds are believed to interfere with these various phenomenons. Finally, the pathological consequences of the presence of excessive concentrations of these compounds are related to the short- and, most important, long-term effects of these compounds on the rapid colonic epithelium renewing and homeostasis.
Collapse
Affiliation(s)
- F Blachier
- Unité Mixte de Recherche de Physiologie de la Nutrition et du Comportement Alimentaire, Institut National de la Recherche Agronomique - Institut National Agronomique Paris-Grignon, Paris, France.
| | | | | | | |
Collapse
|
20
|
Doran JJ, Klein JD, Kim YH, Smith TD, Kozlowski SD, Gunn RB, Sands JM. Tissue distribution of UT-A and UT-B mRNA and protein in rat. Am J Physiol Regul Integr Comp Physiol 2006; 290:R1446-59. [PMID: 16373440 DOI: 10.1152/ajpregu.00352.2004] [Citation(s) in RCA: 31] [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]
Abstract
Mammalian urea transporters are facilitated membrane transport proteins belonging to two families, UT-A and UT-B. They are best known for their role of maintaining the renal inner medullary urinary concentrating gradient. Urea transporters have also been identified in tissues not typically associated with urea metabolism. The purpose of this study was to survey the major organs in rat to determine the distribution of UT-A and UT-B mRNA transcripts and protein forms and determine their cellular localization. Five kidney subregions and 17 extrarenal tissues were screened by Northern blot analysis using two UT-A and three UT-B probes and by Western blot analysis using polyclonal COOH-terminal UT-A and UT-B antibodies. Immunohistochemistry was performed on 16 extrarenal tissues using the same antibodies. In kidney, we detected mRNA transcripts and protein bands consistent with previously-identified UT-A and UT-B isoforms, as well as novel forms. We found that UT-A mRNA and protein are widely expressed in extrarenal tissues in various forms that are different from the known isoforms. We determined the cellular localization of UT-A and UT-B in these tissues. We found that both UT-A and UT-B are ubiquitously expressed as numerous tissue-specific mRNA transcripts and protein forms that are localized to cell membranes, cytoplasm, or nuclei.
Collapse
Affiliation(s)
- John J Doran
- Emory Univ. School of Medicine, Renal Div., 1639 Pierce Dr. NE, WMB Rm. 338, Atlanta, GA 30322, USA.
| | | | | | | | | | | | | |
Collapse
|
21
|
Fenton RA, Shodeinde A, Knepper MA. UT-A urea transporter promoter, UT-Aalpha, targets principal cells of the renal inner medullary collecting duct. Am J Physiol Renal Physiol 2006; 290:F188-95. [PMID: 16091580 PMCID: PMC1435687 DOI: 10.1152/ajprenal.00285.2005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The urea transporters, UT-A1 and UT-A3, two members of the UT-A gene family, are localized to the terminal portion of the inner medullary collecting duct (IMCD). In this manuscript, we demonstrate that 4.2 kb of the 5'-flanking region of the UT-A gene (UT-Aalpha promoter) is sufficient to drive the IMCD-specific expression of a heterologous reporter gene, beta-galactosidase (beta-Gal), in transgenic mice. RT-PCR, immunoblotting, and immunohistochemistry demonstrate that within the kidney, transgene expression is confined to the terminal portion of the IMCD. Colocalization studies with aquaporin-2 show that expression is localized to the principal cells of the IMCD2 and IMCD3 regions. Utilizing beta-Gal activity assays, we further show that within the kidney, the beta-Gal transgene can be regulated by both water restriction and glucocorticoids, similar to the regulation of the endogenous UT-A gene. These results demonstrate that 4.2 kb of the UT-Aalpha promoter is sufficient to drive expression of a heterologous reporter gene in a tissue-specific and cell-specific fashion in transgenic mice.
Collapse
Affiliation(s)
- Robert A Fenton
- Laboratory of Kidney and Electrolyte Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA.
| | | | | |
Collapse
|
22
|
Stewart GS, Graham C, Cattell S, Smith TPL, Simmons NL, Smith CP. UT-B is expressed in bovine rumen: potential role in ruminal urea transport. Am J Physiol Regul Integr Comp Physiol 2005; 289:R605-R612. [PMID: 15845882 DOI: 10.1152/ajpregu.00127.2005] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The UT-A (SLC14a2) and UT-B (SLC14a1) genes encode a family of specialized urea transporter proteins that regulate urea movement across plasma membranes. In this report, we describe the structure of the bovine UT-B (bUT-B) gene and characterize UT-B expression in bovine rumen. Northern analysis using a full-length bUT-B probe detected a 3.7-kb UT-B signal in rumen. RT-PCR of bovine mRNA revealed the presence of two UT-B splice variants, bUT-B1 and bUT-B2, with bUT-B2 the predominant variant in rumen. Immunoblotting studies of bovine rumen tissue, using an antibody targeted to the NH2-terminus of mouse UT-B, confirmed the presence of 43- to 54-kDa UT-B proteins. Immunolocalization studies showed that UT-B was mainly located on cell plasma membranes in epithelial layers of the bovine rumen. Ussing chamber measurements of ruminal transepithelial transport of (14)C-labeled urea indicated that urea flux was characteristically inhibited by phloretin. We conclude that bUT-B is expressed in the bovine rumen and may function to transport urea into the rumen as part of the ruminant urea nitrogen salvaging process.
Collapse
Affiliation(s)
- G S Stewart
- Faculty of Life Sciences, Medical School, The University of Manchester, Manchester M13 9PT, UK
| | | | | | | | | | | |
Collapse
|
23
|
Inoue H, Kozlowski SD, Klein JD, Bailey JL, Sands JM, Bagnasco SM. Regulated expression of renal and intestinal UT-B urea transporter in response to varying urea load. Am J Physiol Renal Physiol 2005; 289:F451-8. [PMID: 15798087 DOI: 10.1152/ajprenal.00376.2004] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Production, recycling, and elimination of urea are important to maintain nitrogen balance. Adaptation to varying loads of urea due to different protein intake or in renal failure may involve changes in urea transport and may possibly affect urea transporters. In this study, we examined the expression of the UT-B urea transporter in rats fed a low-protein diet (LPD), a high-protein diet (HPD), and a 20% urea-supplemented diet. In the kidney, UT-B protein abundance increased in the outer medulla of both LPD-fed rats and 20% urea-fed rats, without changes in the inner medulla of either group compared with controls. In HPD-fed rats, UT-B protein decreased significantly in both the outer and inner medulla. We identified expression of UT-B in the rat colon, as a 2-kb mRNA transcript and as an approximately 45-kDa protein, with apical localization in superficial colon epithelial cells. UT-B also is expressed in rat small intestine. In rat colon, UT-B protein abundance was mildly, but significantly, decreased in LPD-fed and 20% urea-fed rats. UT-B abundance also was examined in the colon of 7/8 nephrectomized, uremic rats and in HPD-fed rats and was not significantly different from that in control rats. These findings indicate that UT-B expression is regulated in response to different loads of urea, with a pattern that suggests involvement of tissue-specific regulatory mechanism in kidney and colon.
Collapse
Affiliation(s)
- Hideki Inoue
- Department of Pathology, Johns Hopkins Hospital, Baltimore, MD 21287, USA
| | | | | | | | | | | |
Collapse
|
24
|
Bagnasco SM. Role and regulation of urea transporters. Pflugers Arch 2005; 450:217-26. [PMID: 15924241 DOI: 10.1007/s00424-005-1403-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2005] [Revised: 02/10/2005] [Accepted: 02/21/2005] [Indexed: 10/25/2022]
Abstract
In the past few years, significant knowledge has been gained about the physiological role and regulation of urea transporters, which have been now cloned in many species. The two major mammalian urea transporters, UT-A and UT-B, have been best studied in the kidney, where they mediate the facilitated diffusion of urea across tubular, interstitial, and vascular compartments, necessary to maintain an osmolar gradient along the renal corticomedullary axis. The genes encoding these transporters, Slc14A2 for UT-A and Slc14A1 for UT-B, have been characterized in rodents and humans, allowing identification of transcriptional mechanisms involved in the regulation of UT-A expression. The crucial role that urea transporters play in renal physiology is underscored by the phenotypic characteristics of UT-A and UT-B knockout mice, in which lack of specific urea transporters impairs the ability to concentrate urine. Expression of the UT-A and UT-B transporters has also been identified in extra-renal sites, where their physiological significance is only beginning to be elucidated. More information on the mechanisms modulating urea transporter expression is becoming available, and the possible involvement of aberrant regulation of these transporters in pathological conditions, or as a result of certain pharmacological treatments, has emerged from recent studies.
Collapse
Affiliation(s)
- Serena M Bagnasco
- Department of Pathology, Johns Hopkins University, 600 North Wolfe Street, Baltimore, MD, 21287, USA.
| |
Collapse
|
25
|
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: 2.1] [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
|
26
|
Smith CP, Potter EA, Fenton RA, Stewart GS. Characterization of a human colonic cDNA encoding a structurally novel urea transporter, hUT-A6. Am J Physiol Cell Physiol 2004; 287:C1087-93. [PMID: 15189812 DOI: 10.1152/ajpcell.00363.2003] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Two closely related genes, UT-A (Slc14a2) and UT-B (Slc14a1), encode specialized transporter proteins that modulate the movement of urea across cell membranes. In this article, we report the characterization of a cDNA isolated from human colonic mucosa encoding a novel UT-A urea transporter, hUT-A6. The encoded protein is 235 amino acids (aa) in length, making it the smallest UT-A member characterized. On the basis of previous structural predictions, hUT-A6 is structurally unique in that it consists of a single hydrophobic core flanked by hydrophilic NH(2)- and COOH-terminal domains. The transcript encoding hUT-A6 contains a novel 129-bp exon, exon 5a, which, as a result of alternative splicing, introduces a unique 19-aa segment and a stop codon. Functionally, the protein transports urea, and this activity is inhibited by phloretin. Interestingly, despite the lack of a protein kinase A (PKA) consensus site [RK](2)-X-[ST], transport of urea by hUT-A6 is stimulated by PKA agonists. Deletion of the two PKA consensus sites from murine UT-A3 (mUT-A3) did not affect the stimulatory response of PKA agonists, which, together with the lack of PKA consensus sites in hUT-A6, indicates that regulation of hUT-A6 and mUT-A3 is not mediated through a classic PKA phosphorylation consensus.
Collapse
Affiliation(s)
- Craig P Smith
- School of Biological Sciences, University of Manchester, G.38, Stopford Bldg., Oxford Road, Manchester M13 9PT, United Kingdom.
| | | | | | | |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW Urea is transported across the kidney inner medullary collecting duct by urea-transporter proteins. Two urea-transporter genes have been cloned from humans and rodents: the UT-A (Slc14A2) gene encodes five protein and eight cDNA isoforms; the UT-B (Slc14A1) gene encodes a single isoform. In the past year, significant progress has been made in understanding the regulation of urea-transporter protein abundance in kidney, studies of genetically engineered mice that lack a urea transporter, identification of urea transporters outside of the kidney, cloning of urea transporters in nonmammalian species, and active urea transport in microorganisms. RECENT FINDINGS UT-A1 protein abundance is increased by 12 days of vasopressin, but not by 5 days. Analysis of the UT-A1 promoter suggests that vasopressin increases UT-A1 indirectly following a direct effect to increase the transcription of other genes, such as the Na(+)-K(+)-2Cl- cotransporter NKCC2/BSC1 and the aquaporin (AQP) 2 water channel, that begin to increase inner medullary osmolality. UT-A1 protein abundance is also increased by adrenalectomy, and is decreased by glucocorticoids or mineralocorticoids. However, each hormone works through its own receptor. Knockout mice that lack UT-A1 and UT-A3, or lack UT-B, have a urine-concentrating defect and a decrease in inner medullary interstitial urea content. SUMMARY Urea transporters play a critical role in the urine-concentrating mechanism. Their abundance is regulated by vasopressin, glucocorticoids, and mineralocorticoids. These regulatory mechanisms may be important in disease states such as diabetes because changes in urea-transporter abundance in diabetic rats require glucocorticoids and vasopressin.
Collapse
Affiliation(s)
- Jeff M Sands
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|