1
|
Elliott J, Oyama MA. Sodium glucose transporter 2 inhibitors: Will these drugs benefit non-diabetic veterinary patients with cardiac and kidney diseases? J Vet Pharmacol Ther 2025; 48 Suppl 1:1-18. [PMID: 39001645 PMCID: PMC11737021 DOI: 10.1111/jvp.13472] [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: 03/24/2024] [Revised: 06/14/2024] [Accepted: 07/03/2024] [Indexed: 01/18/2025]
Abstract
Sodium glucose transporter type 2 (SGLT2) inhibitors have been introduced into human medicine where their beneficial effects go beyond the expected improvement in blood glucose control. These drugs appear to prevent progression of both cardiovascular and kidney diseases, not only in diabetic but also in non-diabetic human patients. As these drugs have received conditional approval for use in diabetic cats and are being used in other veterinary species, the intriguing question as to whether they will have similar cardioprotective and nephroprotective effects in dogs and cats is being asked. The primary mechanism(s) by which SGLT2 inhibitors are cardio- and nephroprotective remain to be fully characterized. This paper reviews these suggested mechanisms in the context of the pathophysiology of progressive cardiovascular and kidney diseases in dogs and cats with the goal of predicting which categories of non-diabetic veterinary patients these drugs might be of most benefit.
Collapse
Affiliation(s)
- Jonathan Elliott
- Department of Comparative Biomedical SciencesRoyal Veterinary College, University of LondonLondonUK
| | - Mark A. Oyama
- Department of Clinical Sciences & Advanced MedicineUniversity of Pennsylvania School of Veterinary MedicinePhiladelphiaPennsylvaniaUSA
| |
Collapse
|
2
|
Chen L, Luo X, Wang X, Wang Y, Yang H, Zhao S, Zhang Q, Liu X, Jiang H. Nanoengineered Nonenzymatic Paper-Based Fluorescent Platform for Pre-Dilution-Free and Visual Real-Time Home Monitoring of Urea for Early Warning of Abnormal Nitrogen-Based Unhealthy Issues. Adv Healthc Mater 2024; 13:e2402009. [PMID: 39113342 DOI: 10.1002/adhm.202402009] [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: 06/01/2024] [Revised: 07/11/2024] [Indexed: 12/18/2024]
Abstract
Distorted urea levels indicate several liver, kidney, or metabolic diseases; however, traditional clinical urea detection relies on urease-based methods enslaved to well-known limitations of high-price, unstable properties, complicated sample pretreatment and analysis procedures, and difficult visual real-time monitoring. Herein, nonenzymatic paper-based fluorescent materials (UFP-BP) are strategically integrated with an on-demand fluorescent-sensor (UFP) self-aggregated nanoparticle on commercial filter paper for pre-dilution-free and visual real-time urea monitoring. The UFP is synthesized and self-aggregated into the fluorescent nanoparticles for selective urea recognition. Then, the nanoparticles are interstitially loaded on filter paper to nanoengineer the UFP-BP, achieving selective quantitative urea detection in the normal concentration range (10-1000 mm). UFP and UFP-BP can successfully monitor urea levels in real rat urine, artificial simulants, and milk. The proposed sensing platform, integrated with smartphones, offers accurate, quantitative, nonenzymatic, noninvasive, pre-dilution-free, on-site, rapid, low-cost, easy-to-operate, real-time visual urea detection in food samples and human body fluids. The designed sensing system can provide early warnings of abnormal nitrogen-based health issues.
Collapse
Affiliation(s)
- Lijuan Chen
- Key Laboratory of Auxiliary Chemistry and Technology for Chemical Industry, Ministry of Education, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
- College of Bioresources Chemistry and Materials Engineering, National Demonstration Center for Experimental Light Chemistry Engineering Education, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| | - Xiaomin Luo
- Key Laboratory of Auxiliary Chemistry and Technology for Chemical Industry, Ministry of Education, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| | - Xuechuan Wang
- Key Laboratory of Auxiliary Chemistry and Technology for Chemical Industry, Ministry of Education, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| | - Yitong Wang
- Key Laboratory of Auxiliary Chemistry and Technology for Chemical Industry, Ministry of Education, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| | - Hao Yang
- Key Laboratory of Auxiliary Chemistry and Technology for Chemical Industry, Ministry of Education, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| | - Suqiu Zhao
- College of Bioresources Chemistry and Materials Engineering, National Demonstration Center for Experimental Light Chemistry Engineering Education, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| | - Qian Zhang
- College of Bioresources Chemistry and Materials Engineering, National Demonstration Center for Experimental Light Chemistry Engineering Education, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| | - Xinhua Liu
- Key Laboratory of Auxiliary Chemistry and Technology for Chemical Industry, Ministry of Education, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| | - Huie Jiang
- Key Laboratory of Auxiliary Chemistry and Technology for Chemical Industry, Ministry of Education, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
- College of Bioresources Chemistry and Materials Engineering, National Demonstration Center for Experimental Light Chemistry Engineering Education, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| |
Collapse
|
3
|
Jordan CZ, Chen Y, Husain I, Dilts M, Fay OK, Privratsky J, Luo X, Tunbridge M. Murine kidney transplant outcome is best measured by transdermal glomerular filtration rate. Am J Transplant 2024; 24:2150-2156. [PMID: 39098449 DOI: 10.1016/j.ajt.2024.07.010] [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: 03/14/2024] [Revised: 06/22/2024] [Accepted: 07/10/2024] [Indexed: 08/06/2024]
Abstract
Mouse kidney transplantation provides a powerful preclinical model for the study of kidney transplant alloimmunity. However, accurate measurement of graft function is difficult because of the inaccuracy of traditional surrogate markers serum creatinine and urea. We report the use of transdermal glomerular filtration rate measurement under the experimental conditions of unilateral nephrectomy and allogeneic kidney transplantation. Our findings demonstrate that transdermal glomerular filtration rate measurement is easy to perform, reproducible, and has more interexperimental consistency than serum creatinine or urea measurements. Most importantly, it significantly reduces the numbers of experimental animals required to detect subtle and yet clinically relevant differences in kidney function as often is the case in experimental murine kidney transplantation models.
Collapse
Affiliation(s)
- Collin Z Jordan
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA; Duke Transplant Center, Duke University School of Medicine, Durham, North Carolina, USA
| | - Yanting Chen
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Irma Husain
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA; Duke Transplant Center, Duke University School of Medicine, Durham, North Carolina, USA
| | - Miriam Dilts
- Department of Pathology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Olivia K Fay
- Department of Pathology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Jamie Privratsky
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Xunrong Luo
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA; Duke Transplant Center, Duke University School of Medicine, Durham, North Carolina, USA
| | - Matthew Tunbridge
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA; Duke Transplant Center, Duke University School of Medicine, Durham, North Carolina, USA; Adelaide Medical School, University of Adelaide, Adelaide, Australia.
| |
Collapse
|
4
|
Lin X, Huang Z, Huang H, Fang Y, Weng Y, Wang Z, Zhao H, Liu H. A tough Janus poly(vinyl alcohol)-based hydrogel for wound closure and anti postoperative adhesion. Acta Biomater 2024; 188:103-116. [PMID: 39243837 DOI: 10.1016/j.actbio.2024.08.049] [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: 07/06/2024] [Revised: 08/10/2024] [Accepted: 08/27/2024] [Indexed: 09/09/2024]
Abstract
Traditional adhesive hydrogels perform well in tissue adhesion but they fail to prevent postoperative tissue adhesion. To address this challenge, a biodegradable Janus adhesive hydrogel (J-AH) was designed and fabricated by the assembly of three different functional layers including anti-adhesive layer, reinforceable layer, and wet tissue adhesive layer. Each layer of J-AH serves a specific function: the top zwitterionic polymeric anti-adhesive layer shows superior resistance to cell/protein and tissue adhesion; the middle poly(vinyl alcohol)/tannic acid reinforceable matrix layer endows the hydrogel with good mechanical toughness of ∼2.700 MJ/m3; the bottom poly(acrylic acid)/polyethyleneimine adhesive layer imparts tough adhesion (∼382.93 J/m2 of interfacial toughness) to wet tissues. In the rat liver and femoral injury models, J-AH could firmly adhere to the bleeding tissues to seal the wounds and exhibit impressive hemostatic efficiency. Moreover, in the in vivo adhesion/anti-adhesion assay of J-AH between the defected cecum and peritoneal walls, the top anti-adhesive layer can effectively inhibit undesired postoperative abdominal adhesion and inflammatory reaction. Therefore, this research may present a new strategy for the design of advanced bio-absorbable Janus adhesive hydrogels with multi-functions including tissue adhesion, anti-postoperative adhesion and biodegradation. STATEMENT OF SIGNIFICANCE: Despite many adhesive hydrogels with tough tissue adhesion capability have been reported, their proclivity for undesired postoperative adhesion remains a serious problem. The postoperative adhesion may lead to major complications and even endanger the lives of patients. The injectable hydrogels can cover the irregular wound and suppress the formation of postoperative adhesion. However, due to the lack of adhesive properties with tissue, it is difficult for the hydrogels to maintain on the wound surface, resulting in poor anti-postoperative adhesion effect. Herein, we design a Janus adhesive hydrogel (J-AH). J-AH integrates together robust wet tissue adhesion and anti-postoperative adhesion. Therefore, this research may present a new strategy for the design of advanced bio-absorbable Janus adhesive hydrogels.
Collapse
Affiliation(s)
- Xiaojin Lin
- College of Chemistry and Materials Science, Fujian Normal University, Fujian 350007, China
| | - Zongxuan Huang
- Department of General Surgery, Fuzhou General Teaching Hospital, Fujian University of Traditional Chinese Medicine (900TH Hospital of Joint Logistics Support Force), Fuzhou 35025, China
| | - Hongjian Huang
- College of Chemistry and Materials Science, Fujian Normal University, Fujian 350007, China
| | - Yan Fang
- College of Chemistry and Materials Science, Fujian Normal University, Fujian 350007, China
| | - Yunxiang Weng
- College of Chemistry and Materials Science, Fujian Normal University, Fujian 350007, China
| | - Zhengchao Wang
- College of Life Science, Fujian Normal University, Fujian 350007, China
| | - Hu Zhao
- Department of General Surgery, Fuzhou General Teaching Hospital, Fujian University of Traditional Chinese Medicine (900TH Hospital of Joint Logistics Support Force), Fuzhou 35025, China
| | - Haiqing Liu
- College of Chemistry and Materials Science, Fujian Normal University, Fujian 350007, China; Fujian-Taiwan Science and Technology Cooperation Base of Biomedical, Materials and Tissue Engineering, Fujian 350007, China; Engineering Research Center of Industrial Biocatalysis, Fujian 350007, China.
| |
Collapse
|
5
|
Tang S, Tang D, Zhou H, Li Y, Zhou D, Peng X, Ren C, Su Y, Zhang S, Zheng H, Wan F, Yoo J, Han H, Ma X, Gao W, Wu S. Bacterial outer membrane vesicle nanorobot. Proc Natl Acad Sci U S A 2024; 121:e2403460121. [PMID: 39008666 PMCID: PMC11287275 DOI: 10.1073/pnas.2403460121] [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: 02/19/2024] [Accepted: 06/05/2024] [Indexed: 07/17/2024] Open
Abstract
Autonomous nanorobots represent an advanced tool for precision therapy to improve therapeutic efficacy. However, current nanorobotic designs primarily rely on inorganic materials with compromised biocompatibility and limited biological functions. Here, we introduce enzyme-powered bacterial outer membrane vesicle (OMV) nanorobots. The immobilized urease on the OMV membrane catalyzes the decomposition of bioavailable urea, generating effective propulsion for nanorobots. This OMV nanorobot preserves the unique features of OMVs, including intrinsic biocompatibility, immunogenicity, versatile surface bioengineering for desired biofunctionalities, capability of cargo loading and protection. We present OMV-based nanorobots designed for effective tumor therapy by leveraging the membrane properties of OMVs. These involve surface bioengineering of robotic body with cell-penetrating peptide for tumor targeting and penetration, which is further enhanced by active propulsion of nanorobots. Additionally, OMV nanorobots can effectively safeguard the loaded gene silencing tool, small interfering RNA (siRNA), from enzymatic degradation. Through systematic in vitro and in vivo studies using a rodent model, we demonstrate that these OMV nanorobots substantially enhanced siRNA delivery and immune stimulation, resulting in the utmost effectiveness in tumor suppression when juxtaposed with static groups, particularly evident in the orthotopic bladder tumor model. This OMV nanorobot opens an inspiring avenue to design advanced medical robots with expanded versatility and adaptability, broadening their operation scope in practical biomedical domains.
Collapse
Affiliation(s)
- Songsong Tang
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen518000, People’s Republic of China
- Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA91125
| | - Daitian Tang
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen518000, People’s Republic of China
- South China Hospital, Health Science Center, Shenzhen University, Shenzhen518116, People’s Republic of China
- Luohu Clinical Institute of Shantou University Medical College, Shantou University Medical College, Shantou515000, People’s Republic of China
| | - Houhong Zhou
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen518000, People’s Republic of China
- Department of General Surgery, Shenzhen Samii Medical Center, Shenzhen518118, People’s Republic of China
| | - Yangyang Li
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen518000, People’s Republic of China
| | - Dewang Zhou
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen518000, People’s Republic of China
| | - Xiqi Peng
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen518000, People’s Republic of China
- South China Hospital, Health Science Center, Shenzhen University, Shenzhen518116, People’s Republic of China
- Luohu Clinical Institute of Shantou University Medical College, Shantou University Medical College, Shantou515000, People’s Republic of China
| | - Chunyu Ren
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen518000, People’s Republic of China
| | - Yilin Su
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen518000, People’s Republic of China
- Luohu Clinical Institute of Shantou University Medical College, Shantou University Medical College, Shantou515000, People’s Republic of China
| | - Shaohua Zhang
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen518000, People’s Republic of China
- South China Hospital, Health Science Center, Shenzhen University, Shenzhen518116, People’s Republic of China
| | - Haoxiang Zheng
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen518000, People’s Republic of China
- South China Hospital, Health Science Center, Shenzhen University, Shenzhen518116, People’s Republic of China
| | - Fangchen Wan
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen518000, People’s Republic of China
| | - Jounghyun Yoo
- Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA91125
| | - Hong Han
- Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA91125
| | - Xiaotian Ma
- Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA91125
| | - Wei Gao
- Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA91125
| | - Song Wu
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen518000, People’s Republic of China
- South China Hospital, Health Science Center, Shenzhen University, Shenzhen518116, People’s Republic of China
- Luohu Clinical Institute of Shantou University Medical College, Shantou University Medical College, Shantou515000, People’s Republic of China
| |
Collapse
|
6
|
Li N, Zhang H, Wang S, Xu Y, Ying Y, Li J, Li X, Li M, Yang B. Urea transporter UT-A1 as a novel drug target for hyponatremia. FASEB J 2024; 38:e23760. [PMID: 38924449 DOI: 10.1096/fj.202400555rr] [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: 03/11/2024] [Revised: 06/06/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024]
Abstract
Hyponatremia is the most common disorder of electrolyte imbalances. It is necessary to develop new type of diuretics to treat hyponatremia without losing electrolytes. Urea transporters (UT) play an important role in the urine concentrating process and have been proved as a novel diuretic target. In this study, rat and mouse syndromes of inappropriate antidiuretic hormone secretion (SIADH) models were constructed and analyzed to determine if UTs are a promising drug target for treating hyponatremia. Experimental results showed that 100 mg/kg UT inhibitor 25a significantly increased serum osmolality (from 249.83 ± 5.95 to 294.33 ± 3.90 mOsm/kg) and serum sodium (from 114 ± 2.07 to 136.67 ± 3.82 mmol/L) respectively in hyponatremia rats by diuresis. Serum chemical examination showed that 25a neither caused another electrolyte imbalance nor influenced the lipid metabolism. Using UT-A1 and UT-B knockout mouse SIADH model, it was found that serum osmolality and serum sodium were lowered much less in UT-A1 knockout mice than in UT-B knockout mice, which suggest UT-A1 is a better therapeutic target than UT-B to treat hyponatremia. This study provides a proof of concept that UT-A1 is a diuretic target for SIADH-induced hyponatremia and UT-A1 inhibitors might be developed into new diuretics to treat hyponatremia.
Collapse
Affiliation(s)
- Nannan Li
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Hang Zhang
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Shuyuan Wang
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yue Xu
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Yi Ying
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jing Li
- The State Key Laboratory of Anti-Infective Drug Development, Sunshine Lake Pharma Co., Ltd., Dongguan, China
| | - Xiaowei Li
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Min Li
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Baoxue Yang
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| |
Collapse
|
7
|
Bankir L, Crambert G, Vargas-Poussou R. The SLC6A18 Transporter Is Most Likely a Na-Dependent Glycine/Urea Antiporter Responsible for Urea Secretion in the Proximal Straight Tubule: Influence of This Urea Secretion on Glomerular Filtration Rate. Nephron Clin Pract 2024; 148:796-822. [PMID: 38824912 PMCID: PMC11651341 DOI: 10.1159/000539602] [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: 02/13/2024] [Accepted: 05/25/2024] [Indexed: 06/04/2024] Open
Abstract
BACKGROUND Urea is the major end-product of protein metabolism in mammals. In carnivores and omnivores, a large load of urea is excreted daily in urine, with a concentration that is 30-100 times above that in plasma. This is important for the sake of water economy. Too little attention has been given to the existence of energy-dependent urea transport that plays an important role in this concentrating activity. SUMMARY This review first presents functional evidence for an energy-dependent urea secretion that occurs exclusively in the straight part of the proximal tubule (PST). Second, it proposes a candidate transmembrane transporter responsible for this urea secretion in the PST. SLC6A18 is expressed exclusively in the PST and has been identified as a glycine transporter, based on findings in SLC6A18 knockout mice. We propose that it is actually a glycine/urea antiport, secreting urea into the lumen in exchange for glycine and Na. Glycine is most likely recycled back into the cell via a transporter located in the brush border. Urea secretion in the PST modifies the composition of the tubular fluid in the thick ascending limb and, thus, contributes, indirectly, to influence the "signal" at the macula densa that plays a crucial role in the regulation of the glomerular filtration rate (GFR) by the tubulo-glomerular feedback. KEY MESSAGES Taking into account this secondary active secretion of urea in the mammalian kidney provides a new understanding of the influence of protein intake on GFR, of the regulation of urea excretion, and of the urine-concentrating mechanism.
Collapse
Affiliation(s)
- Lise Bankir
- Laboratoire de Physiologie Rénale et Tubulopathies, Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Université Paris Cité, Paris, France
- CNRS EMR 8228, Unité Métabolisme et Physiologie Rénale, Centre de Recherche des Cordeliers, Paris, France
| | - Gilles Crambert
- Laboratoire de Physiologie Rénale et Tubulopathies, Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Université Paris Cité, Paris, France
- CNRS EMR 8228, Unité Métabolisme et Physiologie Rénale, Centre de Recherche des Cordeliers, Paris, France
| | - Rosa Vargas-Poussou
- CNRS EMR 8228, Unité Métabolisme et Physiologie Rénale, Centre de Recherche des Cordeliers, Paris, France
- Service de Médecine Génomique des Maladies Rares, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Paris, France
- Centre de Référence des Maladies Rénales Héréditaires de l’Enfant et de l’Adulte, MARHEA, Hôpital Européen Georges Pompidou, Paris, France
| |
Collapse
|
8
|
Noble KA, Chan HKY, Kavanagh ON. Meta-analysis guided development of a standard artificial urine. Eur J Pharm Biopharm 2024; 198:114264. [PMID: 38492868 DOI: 10.1016/j.ejpb.2024.114264] [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: 01/19/2024] [Revised: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 03/18/2024]
Abstract
In this study, we present the first meta-analysis of human urine reported in the literature, drawing data from a total of 35 articles with a combined participant count of 14,021. Through this analysis, we have developed an artificial urine (AU) composition that can be adjusted within typical physiological parameters for in vitro applications. Our findings demonstrate the utility of this AU in determining the solubility of nitrofurantoin, particularly in the context of crystalluria. Notably, we observe that in saline, nitrofurantoin solubility, within the framework of its urinary pharmacokinetics, suggests a risk of crystalluria. However, in AU, this risk is mitigated due to complexation with urea. More broadly, we anticipate that our developed formulation will serve as a foundation for translational studies across biomedical and pharmaceutical sciences.
Collapse
Affiliation(s)
| | - Hayley K Y Chan
- School of Pharmacy, Newcastle University, Newcastle upon Tyne, UK
| | - Oisín N Kavanagh
- School of Pharmacy, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
9
|
Zheng Y, Baidya A, Annabi N. Molecular design of an ultra-strong tissue adhesive hydrogel with tunable multifunctionality. Bioact Mater 2023; 29:214-229. [PMID: 37520304 PMCID: PMC10372327 DOI: 10.1016/j.bioactmat.2023.06.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 05/15/2023] [Accepted: 06/08/2023] [Indexed: 08/01/2023] Open
Abstract
Designing adhesive hydrogels with optimal properties for the treatment of injured tissues is challenging due to the tradeoff between material stiffness and toughness while maintaining adherence to wet tissue surfaces. In most cases, bioadhesives with improved mechanical strength often lack an appropriate elastic compliance, hindering their application for sealing soft, elastic, and dynamic tissues. Here, we present a novel strategy for engineering tissue adhesives in which molecular building blocks are manipulated to allow for precise control and optimization of the various aforementioned properties without any tradeoffs. To introduce tunable mechanical properties and robust tissue adhesion, the hydrogel network presents different modes of covalent and noncovalent interactions using N-hydroxysuccinimide ester (NHS) conjugated alginate (Alg-NHS), poly (ethylene glycol) diacrylate (PEGDA), tannic acid (TA), and Fe3+ ions. Through combining and tuning different molecular interactions and a variety of crosslinking mechanisms, we were able to design an extremely elastic (924%) and tough (4697 kJ/m3) multifunctional hydrogel that could quickly adhere to wet tissue surfaces within 5 s of gentle pressing and deform to support physiological tissue function over time under wet conditions. While Alg-NHS provides covalent bonding with the tissue surfaces, the catechol moieties of TA molecules synergistically adopt a mussel-inspired adhesive mechanism to establish robust adherence to the wet tissue. The strong adhesion of the engineered bioadhesive patch is showcased by its application to rabbit conjunctiva and porcine cornea. Meanwhile, the engineered bioadhesive demonstrated painless detachable characteristics and in vitro biocompatibility. Additionally, due to the molecular interactions between TA and Fe3+, antioxidant and antibacterial properties required to support the wound healing pathways were also highlighted. Overall, by tuning various molecular interactions, we were able to develop a single-hydrogel platform with an "all-in-one" multifunctionality that can address current challenges of engineering hydrogel-based bioadhesives for tissue repair and sealing.
Collapse
Affiliation(s)
- Yuting Zheng
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, 90095, United States
| | - Avijit Baidya
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, 90095, United States
| | - Nasim Annabi
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, 90095, United States
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, United States
| |
Collapse
|
10
|
Muresan AR, Rahaman KA, Son J, Kang MJ, Kwon OS. Metabolites identification of anabolic steroid bolasterone in vitro and in rats by high resolution liquid chromatography mass spectrometry. Drug Test Anal 2023; 15:1329-1343. [PMID: 36700373 DOI: 10.1002/dta.3447] [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: 11/28/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/27/2023]
Abstract
Bolasterone (7α,17α-dimethyltestosterone) and anabolic androgenic steroids are included in the World Anti-Doping Agency's Prohibited list of substances. This study aimed to evaluate the metabolism of bolasterone through in vitro experiments using rat liver microsomes and in vivo experiments using rat urine after oral administration. Urine samples were collected over a 168-h period. Bolasterone and its metabolites were detected by liquid chromatography coupled with a Q-Exactive Obitrap mass spectrometry (LC-HRMS). Ultimately 16 hydroxylated metabolites (M1-M16), one metabolite from the reduction of the 3-keto function and 4-ene (M17), and one glucuronic acid conjugated metabolite (M18) were detected. Metabolites M17 and M18 were confirmed by comparison with available reference or authentic standards. Metabolic modifications in the structure of the parent bolasterone result in different fragmentation patterns. Based on the sensitivity of the HRMS data, characteristic ions such as m/z 121.064 (C8 H9 O) generated from ring A of the mono-hydroxylated metabolites and 121.101 (C9 H13 ) generated from ring D of the di-hydroxylated metabolites were observed that helped differentiate between the obtained metabolites. The structures of fragment ions were tentatively proposed based on their fragmentation pathways, where the significant ions were correlated to the possible structural fragments. In conclusion, new metabolites of bolasterone were detected and characterized by the use of the full-scan and dd-MS/MS using LC-HRMS, and this data can be useful for providing metabolite information for the interpretation of mass spectra of anabolic bolasterone analogues for doping screening tests.
Collapse
Affiliation(s)
- Anca Raluca Muresan
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, South Korea
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology, Seoul, South Korea
| | - Khandoker Asiqur Rahaman
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, South Korea
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology, Seoul, South Korea
| | - Junghyun Son
- Doping Control Center, Korea Institute of Science and Technology, Seoul, South Korea
| | - Min-Jung Kang
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, South Korea
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology, Seoul, South Korea
| | - Oh-Seung Kwon
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, South Korea
- Doping Control Center, Korea Institute of Science and Technology, Seoul, South Korea
| |
Collapse
|
11
|
Zheng Y, Shariati K, Ghovvati M, Vo S, Origer N, Imahori T, Kaneko N, Annabi N. Hemostatic patch with ultra-strengthened mechanical properties for efficient adhesion to wet surfaces. Biomaterials 2023; 301:122240. [PMID: 37480758 DOI: 10.1016/j.biomaterials.2023.122240] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 06/15/2023] [Accepted: 07/06/2023] [Indexed: 07/24/2023]
Abstract
Controlling traumatic bleeding from damaged internal organs while effectively sealing the wound is critical for saving the lives of patients. Existing bioadhesives suffer from blood incompatibility, insufficient adhesion to wet surfaces, weak mechanical properties, and complex application procedures. Here, we engineered a ready-to-use hemostatic bioadhesive with ultra-strengthened mechanical properties and fatigue resistance, robust adhesion to wet tissues within a few seconds of gentle pressing, deformability to accommodate physiological function and action, and the ability to stop bleeding efficiently. The engineered hydrogel, which demonstrated high elasticity (>900%) and toughness (>4600 kJ/m3), was formed by fine-tuning a series of molecular interactions and crosslinking mechanisms involving N-hydroxysuccinimide (NHS) conjugated alginate (Alg-NHS), poly (ethylene glycol) diacrylate (PEGDA), tannic acid (TA), and Fe3+ ions. Dual adhesive moieties including mussel-inspired pyrogallol/catechol and NHS synergistically enhanced wet tissue adhesion (>400 kPa in a wound closure test). In conjunction with physical sealing, the high affinity of TA/Fe3+ for blood could further augment hemostasis. The engineered bioadhesive demonstrated excellent in vitro and in vivo biocompatibility as well as improved hemostatic efficacy as compared to commercial Surgicel®. Overall, the hydrogel design strategy described herein holds great promise for overcoming existing obstacles impeding clinical translation of engineered hemostatic bioadhesives.
Collapse
Affiliation(s)
- Yuting Zheng
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kaavian Shariati
- David Geffen School of Medicine, University of California - Los Angeles, Los Angeles, CA 90095, USA
| | - Mahsa Ghovvati
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Division of Interventional Neuroradiology, Department of Radiological Sciences, David Geffen School of Medicine, University of California - Los Angeles, Los Angeles, CA 90095, USA
| | - Steven Vo
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Nolan Origer
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Taichiro Imahori
- Division of Interventional Neuroradiology, Department of Radiological Sciences, David Geffen School of Medicine, University of California - Los Angeles, Los Angeles, CA 90095, USA
| | - Naoki Kaneko
- Division of Interventional Neuroradiology, Department of Radiological Sciences, David Geffen School of Medicine, University of California - Los Angeles, Los Angeles, CA 90095, USA
| | - Nasim Annabi
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, United States.
| |
Collapse
|
12
|
Afolabi OB, Olasehinde OR, Olaoye OA, Jaiyesimi KF, Ekakitie IL, Oloyede OI. Nephroprotective Effects of Caffeine, Vanillin, and Their Combination against Experimental AlCl 3-Induced Renal Toxicity in Adult Male Wistar Rats. Biochem Res Int 2023; 2023:6615863. [PMID: 37649570 PMCID: PMC10465259 DOI: 10.1155/2023/6615863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/14/2023] [Accepted: 08/14/2023] [Indexed: 09/01/2023] Open
Abstract
Aluminum (Al) is known to be a nephrotoxic metal that can cause renal toxicity in both humans and animals. The use of functional foods has been reported to have significance in managing the toxic effects associated with such metals. This study aimed to assess the potential protective effects of caffeine, vanillin, and their combination in mitigating AlCl3-induced renal toxicity in adult male Wistar rats. A total of thirty (30) adult male Wistar rats weighing between 150 and 200 g were randomly divided into five groups, each consisting of six rats (n = 6). Group 1 served as the control, while the remaining treatment groups received a daily oral dose of 100 mg/kg AlCl3 for a duration of 21 days. In addition, groups 3-5 were coadministered 50 mg/kg body weight (bw) of caffeine, vanillin, and a combination (50/50 mg/kg bw) of both substances, respectively. In the results, AlCl3-treated showed a significant (p < 0.05) increase in serum biomarkers such as ALT, ALP, urea, and creatinine, and a significant (p < 0.05) decrease in serum total proteins (TPs). The renal tissue's antioxidant system, including SOD, CAT, GPx, and GSH, exhibited a significant (p < 0.05) reduction, accompanied by an elevated MDA level. However, the administration of caffeine, vanillin, and their combination resulted in a significant (p < 0.05) decrease in serum ALT, ALP, urea, and creatinine, and a significant (p < 0.05) increase in serum TP. Furthermore, following the treatment, there was a significant (p < 0.05) increase in renal SOD, CAT, GPx, and GSH levels, along with a reduction in the MDA level. In addition, the treatment for 21 days caused a significant (p < 0.05) reversal to the altered histomorphological architecture. These findings suggest that caffeine, vanillin, and their combination could potentially be an effective regimen in managing AlCl3-induced renal toxicity.
Collapse
Affiliation(s)
- Olakunle Bamikole Afolabi
- Department of Chemical Sciences, Biochemistry Programme, College of Science, Afe Babalola University, P.M.B 5454, Ado-Ekiti, Ekiti State, Nigeria
| | - Oluwaseun Ruth Olasehinde
- Department of Medical Biochemistry, College of Medicine and Health Sciences, Afe Babalola University, P.M.B 5454, Ado-Ekiti, Ekiti State, Nigeria
| | - Oyindamola Adeniyi Olaoye
- Department of Chemical Sciences, Biochemistry Programme, College of Science, Afe Babalola University, P.M.B 5454, Ado-Ekiti, Ekiti State, Nigeria
| | - Kikelomo Folake Jaiyesimi
- Department of Chemical Sciences, Biochemistry Programme, College of Science, Afe Babalola University, P.M.B 5454, Ado-Ekiti, Ekiti State, Nigeria
| | - Ilobekemen Lisa Ekakitie
- Department of Chemical Sciences, Biochemistry Programme, College of Science, Afe Babalola University, P.M.B 5454, Ado-Ekiti, Ekiti State, Nigeria
| | - Omotade Ibidun Oloyede
- Department of Biochemistry, Ekiti State University, P.M.B 5363, Ado-Ekiti, Ekiti State, Nigeria
| |
Collapse
|
13
|
Riskin DK, Carter GG. The evolution of sanguivory in vampire bats: origins and convergences. CAN J ZOOL 2023. [DOI: 10.1139/cjz-2022-0115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Blood-feeding (sanguivory) has evolved more than two dozen times among birds, fishes, insects, arachnids, molluscs, crustaceans, and annelids; however, among mammals, it is restricted to the vampire bats. Here, the authors revisit the question of how it evolved in that group. Evidence to date suggests that the ancestors of phyllostomids were insectivorous, and that carnivory, omnivory, and nectarivory evolved among phyllostomids after vampire bats diverged. Frugivory likely also evolved after vampire bats diverged, but the phylogeny is ambiguous on that point. However, vampire bats lack any genetic evidence of a frugivorous past, and the behavioural progression from frugivory to sanguivory is difficult to envision. Thus, the most parsimonious scenario is that sanguivory evolved in an insectivorous ancestor to vampire bats via ectoparasite-eating, wound-feeding, or some combination of the two—all feeding habits found among blood-feeding birds today. Comparing vampire bats with other sanguivores, the authors find several remarkable examples of convergence. Further, it was found that blood-feeding has been ca. 50 times more likely to evolve in a vertebrate lineage than in an invertebrate one. The authors hypothesize that this difference exists because vertebrates are more likely than invertebrates to have the biochemical necessities required to assimilate the components of vertebrate blood.
Collapse
Affiliation(s)
- Daniel K. Riskin
- Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada
| | - Gerald G. Carter
- Smithsonian Tropical Research Institute, Apartado 0843-03092, Balboa, Ancón, Republic of Panamá
- Department of Ecology, Evolution, and Organismal Biology, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
14
|
Bankir L, Guerrot D, Bichet DG. Vaptans or voluntary increased hydration to protect the kidney: how do they compare? Nephrol Dial Transplant 2023; 38:562-574. [PMID: 34586414 DOI: 10.1093/ndt/gfab278] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Indexed: 11/13/2022] Open
Abstract
The adverse effects of vasopressin (AVP) in diverse forms of chronic kidney disease have been well described. They depend on the antidiuretic action of AVP mediated by V2 receptors (V2R). Tolvaptan, a selective V2R antagonist, is now largely used for the treatment of patients with autosomal dominant polycystic kidney disease. Another way to reduce the adverse effects of AVP is to reduce endogenous AVP secretion by a voluntary increase in fluid intake. These two approaches differ in several ways, including the level of thirst and AVP. With voluntary increased drinking, plasma osmolality will decline and so will AVP secretion. Thus, not only will V2R-mediated effects be reduced, but also those mediated by V1a and V1b receptors (V1aR and V1bR). In contrast, selective V2R antagonism will induce a loss of fluid that will stimulate AVP secretion and thus increase AVP's influence on V1a and V1b receptors. V1aR is expressed in the luminal side of the collecting duct (CD) and in inner medullary interstitial cells, and their activation induces the production of prostaglandins, mostly prostaglandin E2 (PGE2). Intrarenal PGE2 has been shown to reduce sodium and water reabsorption in the CD and increase blood flow in the renal medulla, both effects contributing to increase sodium and water excretion and reduce urine-concentrating activity. Conversely, non-steroidal anti-inflammatory drugs have been shown to induce significant water and sodium retention and potentiate the antidiuretic effects of AVP. Thus, during V2R antagonism, V1aR-mediated actions may be responsible for part of the diuresis observed with this drug. These V1aR-dependent effects do not take place with a voluntary increase in fluid intake. In summary, while both strategies may have beneficial effects, the information reviewed here leads us to assume that pharmacological V2R antagonism, with resulting stimulation of V1aR and increased PGE2 production, may provide greater benefit than voluntary high water intake. The influence of tolvaptan on the PGE2 excretion rate and the possibility to use somewhat lower tolvaptan doses than presently prescribed remain to be evaluated.
Collapse
Affiliation(s)
- Lise Bankir
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France.,CNRS, ERL 8228-Laboratoire de Physiologie Rénale et Tubulopathies, Paris, France
| | - Dominique Guerrot
- Départment de Néphrologie, Hôpital Universitaire de Rouen, Rouen, France.,Université de Normandie, UNIROUEN, INSERM U1096, Rouen, France
| | - Daniel G Bichet
- Université de Montréal, Montréal, Quebec, Canada.,Département de Pharmacologie, Département de Physiologie, and Département de Médecine, Hôpital du Sacré-Coeur de Montréal, Montréal, Quebec, Canada
| |
Collapse
|
15
|
Ying Y, Li N, Wang S, Zhang H, Zuo Y, Tang Y, Qiao P, Quan Y, Li M, Yang B. Urea Transporter Inhibitor 25a Reduces Ascites in Cirrhotic Rats. Biomedicines 2023; 11:biomedicines11020607. [PMID: 36831143 PMCID: PMC9953117 DOI: 10.3390/biomedicines11020607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/03/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Ascites is a typical symptom of liver cirrhosis that is caused by a variety of liver diseases. Ascites severely affects the life quality of patients and needs long-term treatment. 25a is a specific urea transporter inhibitor with a diuretic effect that does not disturb the electrolyte balance. In this study, we aimed to determine the therapeutic effect of 25a on ascites with a dimethylnitrosamine (DMN)-induced cirrhotic rat model. It was found that 100 mg/kg of 25a significantly increased the daily urine output by 60% to 97% and reduced the daily abdominal circumference change by 220% to 260% in cirrhotic rats with a water intake limitation. The 25a treatment kept the serum electrolyte levels within normal ranges in cirrhotic rats. The H&E and Masson staining of liver tissue showed that 25a did not change the cirrhotic degree. A serum biochemical examination showed that 25a did not improve the liver function in cirrhotic rats. A Western blot analysis showed that 25a did not change the expression of fibrosis-related marker protein α-SMA, but significantly decreased the expressions of type I collagen in the liver of cirrhotic rats, indicating that 25a did not reverse cirrhosis, but could slow the cirrhotic progression. These data indicated that 25a significantly reduced ascites via diuresis without an electrolyte imbalance in cirrhotic rats. Our study provides a proof of concept that urea transporter inhibitors might be developed as novel diuretics to treat cirrhotic ascites.
Collapse
Affiliation(s)
- Yi Ying
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Nannan Li
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Shuyuan Wang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Hang Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Yinglin Zuo
- The State Key Laboratory of Anti-Infective Drug Development, Sunshine Lake Pharma Co., Ltd., Dongguan 523871, China
| | - Yiwen Tang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Panshuang Qiao
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Yazhu Quan
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Min Li
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Baoxue Yang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100191, China
- Correspondence:
| |
Collapse
|
16
|
Shin SH, Wendland MF, Wang J, Velasquez M, Vandsburger MH. Noninvasively differentiating acute and chronic nephropathies via multiparametric urea-CEST, nuclear Overhauser enhancement-CEST, and quantitative magnetization transfer MRI. Magn Reson Med 2023; 89:774-786. [PMID: 36226662 PMCID: PMC11027791 DOI: 10.1002/mrm.29477] [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: 06/15/2022] [Revised: 08/26/2022] [Accepted: 09/14/2022] [Indexed: 12/13/2022]
Abstract
PURPOSE Standardized blood tests often lack adequate sensitivity and specificity to capture the gradual progression of renal injuries. We suggest a multiparametric molecular MRI approach as a noninvasive tool for monitoring renal function loss and distinguishing different types of renal injuries. METHODS CEST and quantitative magnetization transfer (qMT) imaging were performed on cisplatin (n = 16) and aristolochic acid (AA)-induced nephropathy (n = 22) mouse models at 7T with an infusion of either saline or urea. Seven-pool Lorentzian fitting was applied for the analysis of CEST Z-spectra, and the T1 -corrected CEST contrast apparent exchange-dependent relaxation (AREX) from urea (+1 ppm) and two nuclear Overhauser enhancement (NOE) pools (-1.6 and -3.5 ppm) were measured. Similarly, qMT spectra were fitted into two-pool Ramani equation and the relative semi-solid macromolecular pool-size ratio was measured. Histology of mouse kidneys was performed to validate the MR findings. RESULTS AA model showed disrupted spatial gradients of urea in the kidney and significantly decreased NOE CEST and qMT contrast. The cisplatin model showed slightly decreased qMT contrast only. The orrelation of MR parameters to histological features showed that NOE CEST and qMT imaging are sensitive to both acute and chronic injuries, whereas urea CEST shows a significant correlation only to acute injuries. CONCLUSION These results indicate that our multiparametric approach allows comprehensive and totally noninvasive monitoring of renal function and histological changes for distinguishing different nephropathies.
Collapse
Affiliation(s)
- Soo Hyun Shin
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA
| | - Michael F. Wendland
- Berkeley Preclinical Imaging Core (BPIC), University of California, Berkeley, Berkeley, CA
| | - Jingshen Wang
- Department of Biostatistics, University of California, Berkeley, Berkeley, CA
| | - Mark Velasquez
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA
| | | |
Collapse
|
17
|
Zhong C, Long R, Stewart GS. The role of rumen epithelial urea transport proteins in urea nitrogen salvage: A review. ANIMAL NUTRITION 2022; 9:304-313. [PMID: 35600543 PMCID: PMC9097623 DOI: 10.1016/j.aninu.2022.01.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/26/2021] [Accepted: 01/24/2022] [Indexed: 11/27/2022]
|
18
|
Tolvaptan induces body fluid loss and subsequent water conservation in normal rats. J Pharmacol Sci 2022; 149:115-123. [DOI: 10.1016/j.jphs.2022.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 04/13/2022] [Accepted: 04/19/2022] [Indexed: 01/12/2023] Open
|
19
|
He M, Yao J, Zhang Z, Zhang Y, Chen R, Gu Z, Huang X, Deng C, Zhou R, Fan J, Zhang B, Xie Y, Gao G, Sun T. Gold nanoclusters eliminate obesity induced by antipsychotics. Sci Rep 2022; 12:5502. [PMID: 35365730 PMCID: PMC8975852 DOI: 10.1038/s41598-022-09541-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/24/2022] [Indexed: 12/18/2022] Open
Abstract
Obesity induced by antipsychotics have plagued more than 20 million people worldwide. However, no drug is available to eliminate the obesity induced by antipsychotics. Here we examined the effect and potential mechanisms of a gold nanoclusters (AuNCs) modified by N-isobutyryl-L-cysteine on the obesity induced by olanzapine, the most prescribed but obesogenic antipsychotics, in a rat model. Our results showed that AuNCs completely prevented and reversed the obesity induced by olanzapine and improved glucose metabolism profile in rats. Further mechanism investigations revealed that AuNCs exert its anti-obesity function through inhibition of olanzapine-induced dysfunction of histamine H1 receptor and proopiomelanocortin signaling therefore reducing hyperphagia, and reversing olanzapine-induced inhibition of uncoupling-protein-1 signaling which increases thermogenesis. Together with AuNCs' good biocompatibility, these findings not only provide AuNCs as a promising nanodrug candidate for treating obesity induced by antipsychotics, but also open an avenue for the potential application of AuNCs-based nanodrugs in treating general obesity.
Collapse
Affiliation(s)
- Meng He
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China
| | - Jing Yao
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China
| | - Zijun Zhang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, 430070, China
| | - Ying Zhang
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China
| | - Rui Chen
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, 430070, China
| | - Zhenhua Gu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, 430070, China
| | - XuFeng Huang
- School of Medicine and Molecular Horizons, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Chao Deng
- School of Medicine and Molecular Horizons, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Ruqin Zhou
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China
| | - Jun Fan
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China
| | - Baohua Zhang
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Capital Medical University, Beijing, 100191, China
| | - Yanqian Xie
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China
| | - Guanbin Gao
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, 430070, China.
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China.
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, 430070, China.
| |
Collapse
|
20
|
Akihisa T, Kataoka H, Makabe S, Manabe S, Yoshida R, Ushio Y, Sato M, Tsuchiya K, Mochizuki T, Nitta K. Initial decline in eGFR to predict tolvaptan response in autosomal-dominant polycystic kidney disease. Clin Exp Nephrol 2022; 26:540-551. [PMID: 35165806 DOI: 10.1007/s10157-022-02192-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 01/29/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND Tolvaptan, a vasopressin V2 receptor antagonist, is used to treat autosomal-dominant polycystic kidney disease (ADPKD). Although tolvaptan curbs disease progression, a few reports have examined factors related to treatment response. The estimated glomerular filtration rate (eGFR) decreases soon after tolvaptan is initiated. We investigated whether initial eGFR decline affects renal prognosis of patients. METHODS This was a single-center, retrospective observational cohort study. Eighty-three patients with ADPKD who initiated tolvaptan were selected. We analyzed the relationship of the initial eGFR change with clinical parameters and analyzed the annual eGFR change in terms of renal prognostic value using univariable and multivariable linear regression analyses. RESULTS The initial eGFR change was - 4.6 ± 8.0%/month. The initial eGFR change correlated significantly with the annual eGFR change in multivariable analysis, suggesting that the larger decline in the initial eGFR change, the better the renal prognosis. Furthermore, the change in fractional excretion (FE) of free water (FEH2O) correlated positively with initial eGFR change. FEH2O and urea nitrogen FE (FEUN) increased significantly; however, sodium FE (FENa) level remained unchanged. In approximately half of the patients, FENa unexpectedly decreased. CONCLUSIONS The initial eGFR decline might be caused by suppressing glomerular hyperfiltration, due to the pharmacological effect of tolvaptan, and/or by reducing renal plasma flow, due to potential volume depletion. The initial eGFR change reflects the tolvaptan effect, can be easily evaluated in clinical practice, and may be useful as one of the clinical indicator for predicting renal prognosis in patients under tolvaptan.
Collapse
Affiliation(s)
- Taro Akihisa
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Hiroshi Kataoka
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Shiho Makabe
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Shun Manabe
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Rie Yoshida
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Yusuke Ushio
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Masayo Sato
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Ken Tsuchiya
- Department of Blood Purification, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Toshio Mochizuki
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan.
| | - Kosaku Nitta
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| |
Collapse
|
21
|
Lee AS, Lamanna OK, Ishida K, Hill E, Nguyen A, Hsieh MH. A Novel Propidium Monoazide-Based PCR Assay Can Measure Viable Uropathogenic E. coli In Vitro and In Vivo. Front Cell Infect Microbiol 2022; 12:794323. [PMID: 35178354 PMCID: PMC8844370 DOI: 10.3389/fcimb.2022.794323] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/11/2022] [Indexed: 11/13/2022] Open
Abstract
Background Polymerase chain reaction (PCR) is an important means by which to study the urine microbiome and is emerging as possible alternative to urine cultures to identify pathogens that cause urinary tract infection (UTI). However, PCR is limited by its inability to differentiate DNA originating from viable, metabolically active versus non-viable, inactive bacteria. This drawback has led to concerns that urobiome studies and PCR-based diagnosis of UTI are confounded by the presence of relic DNA from non-viable bacteria in urine. Propidium monoazide (PMA) dye can penetrate cells with compromised cell membranes and covalently bind to DNA, rendering it inaccessible to amplification by PCR. Although PMA has been shown to differentiate between non-viable and viable bacteria in various settings, its effectiveness in urine has not been previously studied. We sought to investigate the ability of PMA to differentiate between viable and non-viable bacteria in urine. Methods Varying amounts of viable or non-viable uropathogenic E. coli (UTI89) or buffer control were titrated with mouse urine. The samples were centrifuged to collect urine sediment or not centrifuged. Urine samples were incubated with PMA and DNA cross-linked using blue LED light. DNA was isolated and uidA gene-specific PCR was performed. For in vivo studies, mice were inoculated with UTI89, followed by ciprofloxacin treatment or no treatment. After the completion of ciprofloxacin treatment, an aliquot of urine was plated on non-selective LB agar and another aliquot was treated with PMA and subjected to uidA-specific PCR. Results PMA’s efficiency in excluding DNA signal from non-viable bacteria was significantly higher in bacterial samples in phosphate-buffered saline (PBS, dCT=13.69) versus bacterial samples in unspun urine (dCT=1.58). This discrepancy was diminished by spinning down urine-based bacterial samples to collect sediment and resuspending it in PBS prior to PMA treatment. In 3 of 5 replicate groups of UTI89-infected mice, no bacteria grew in culture; however, there was PCR amplification of E. coli after PMA treatment in 2 of those 3 groups. Conclusion We have successfully developed PMA-based PCR methods for amplifying DNA from live bacteria in urine. Our results suggest that non-PMA bound DNA from live bacteria can be present in urine, even after antibiotic treatment. This indicates that viable but non-culturable E. coli can be present following treatment of UTI, and may explain why some patients have persistent symptoms but negative urine cultures following UTI treatment.
Collapse
Affiliation(s)
- Albert S. Lee
- Division of Pediatric Urology, Children’s National Hospital, Washington, DC, United States
| | - Olivia K. Lamanna
- Sheikh Zayed Institute, Children’s National Hospital, Washington, DC, United States
| | - Kenji Ishida
- Sheikh Zayed Institute, Children’s National Hospital, Washington, DC, United States
| | - Elaise Hill
- School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
- Center for Cancer and Immunology Research, Children’s National Hospital, Washington, DC, United States
| | - Andrew Nguyen
- School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
| | - Michael H. Hsieh
- Division of Pediatric Urology, Children’s National Hospital, Washington, DC, United States
- Sheikh Zayed Institute, Children’s National Hospital, Washington, DC, United States
- School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
- *Correspondence: Michael H. Hsieh,
| |
Collapse
|
22
|
Hus-Citharel A, Bouby N, Corbani M, Mion J, Mendre C, Darusi J, Tomboly C, Trueba M, Serradeil-Le Gal C, Llorens-Cortes C, Guillon G. Characterization of a functional V 1B vasopressin receptor in the male rat kidney: evidence for cross talk between V 1B and V 2 receptor signaling pathways. Am J Physiol Renal Physiol 2021; 321:F305-F321. [PMID: 34282956 DOI: 10.1152/ajprenal.00081.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Although vasopressin V1B receptor (V1BR) mRNA has been detected in the kidney, the precise renal localization as well as pharmacological and physiological properties of this receptor remain unknown. Using the selective V1B agonist d[Leu4, Lys8]VP, either fluorescent or radioactive, we showed that V1BR is mainly present in principal cells of the inner medullary collecting duct (IMCD) in the male rat kidney. Protein and mRNA expression of V1BR were very low compared with the V2 receptor (V2R). On the microdissected IMCD, d[Leu4, Lys8]VP had no effect on cAMP production but induced a dose-dependent and saturable intracellular Ca2+ concentration increase mobilization with an EC50 value in the nanomolar range. This effect involved both intracellular Ca2+ mobilization and extracellular Ca2+ influx. The selective V1B antagonist SSR149415 strongly reduced the ability of vasopressin to increase intracellular Ca2+ concentration but also cAMP, suggesting a cooperation between V1BR and V2R in IMCD cells expressing both receptors. This cooperation arises from a cross talk between second messenger cascade involving PKC rather than receptor heterodimerization, as supported by potentiation of arginine vasopressin-stimulated cAMP production in human embryonic kidney-293 cells coexpressing the two receptor isoforms and negative results obtained by bioluminescence resonance energy transfer experiments. In vivo, only acute administration of high doses of V1B agonist triggered significant diuretic effects, in contrast with injection of selective V2 agonist. This study brings new data on the localization and signaling pathways of V1BR in the kidney, highlights a cross talk between V1BR and V2R in the IMCD, and suggests that V1BR may counterbalance in some pathophysiological conditions the antidiuretic effect triggered by V2R activation.NEW & NOTEWORTHY Although V1BR mRNA has been detected in the kidney, the precise renal localization as well as pharmacological and physiological properties of this receptor remain unknown. Using original pharmaceutical tools, this study brings new data on the localization and signaling pathways of V1BR, highlights a cross talk between V1BR and V2 receptor (V2R) in the inner medullary collecting duct, and suggests that V1BR may counterbalance in some pathophysiological conditions the antidiuretic effect triggered by V2R activation.
Collapse
Affiliation(s)
- Annette Hus-Citharel
- Collège de France, Neuropeptides Centraux et Régulations Hydrique et Cardiovasculaire, Centre Interdisciplinaire de Recherche en Biologie, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Paris, France
| | - Nadine Bouby
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France
| | - Maithé Corbani
- Institut de Génomique Fonctionnelle, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Université de Montpellier, Montpellier, France
| | - Julie Mion
- Institut de Génomique Fonctionnelle, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Université de Montpellier, Montpellier, France
| | - Christiane Mendre
- Institut de Génomique Fonctionnelle, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Université de Montpellier, Montpellier, France
| | - Judit Darusi
- Biological Research Center of the Hungarian Academy of Sciences, Szeged, Hungary
| | - Csaba Tomboly
- Biological Research Center of the Hungarian Academy of Sciences, Szeged, Hungary
| | - Miguel Trueba
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, Basque Country University, Leioa, Spain
| | | | - Catherine Llorens-Cortes
- Collège de France, Neuropeptides Centraux et Régulations Hydrique et Cardiovasculaire, Centre Interdisciplinaire de Recherche en Biologie, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Paris, France
| | - Gilles Guillon
- Institut de Génomique Fonctionnelle, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Université de Montpellier, Montpellier, France
| |
Collapse
|
23
|
Urinary Carnosinase-1 Excretion is Associated with Urinary Carnosine Depletion and Risk of Graft Failure in Kidney Transplant Recipients: Results of the TransplantLines Cohort Study. Antioxidants (Basel) 2021; 10:antiox10071102. [PMID: 34356335 PMCID: PMC8301129 DOI: 10.3390/antiox10071102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/26/2021] [Accepted: 07/06/2021] [Indexed: 11/16/2022] Open
Abstract
Carnosine affords protection against oxidative and carbonyl stress, yet high concentrations of the carnosinase-1 enzyme may limit this. We recently reported that high urinary carnosinase-1 is associated with kidney function decline and albuminuria in patients with chronic kidney disease. We prospectively investigated whether urinary carnosinase-1 is associated with a high risk for development of late graft failure in kidney transplant recipients (KTRs). Carnosine and carnosinase-1 were measured in 24 h urine in a longitudinal cohort of 703 stable KTRs and 257 healthy controls. Cox regression was used to analyze the prospective data. Urinary carnosine excretions were significantly decreased in KTRs (26.5 [IQR 21.4–33.3] µmol/24 h versus 34.8 [IQR 25.6–46.8] µmol/24 h; p < 0.001). In KTRs, high urinary carnosinase-1 concentrations were associated with increased risk of undetectable urinary carnosine (OR 1.24, 95%CI [1.06–1.45]; p = 0.007). During median follow-up for 5.3 [4.5–6.0] years, 84 (12%) KTRs developed graft failure. In Cox regression analyses, high urinary carnosinase-1 excretions were associated with increased risk of graft failure (HR 1.73, 95%CI [1.44–2.08]; p < 0.001) independent of potential confounders. Since urinary carnosine is depleted and urinary carnosinase-1 imparts a higher risk for graft failure in KTRs, future studies determining the potential of carnosine supplementation in these patients are warranted.
Collapse
|
24
|
Kamel KS, Halperin ML. Use of Urine Electrolytes and Urine Osmolality in the Clinical Diagnosis of Fluid, Electrolytes, and Acid-Base Disorders. Kidney Int Rep 2021; 6:1211-1224. [PMID: 34013099 PMCID: PMC8116912 DOI: 10.1016/j.ekir.2021.02.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 02/01/2021] [Indexed: 01/16/2023] Open
Abstract
We discuss the use of urine electrolytes and urine osmolality in the clinical diagnosis of patients with fluid, electrolytes, and acid-base disorders, emphasizing their physiological basis, their utility, and the caveats and limitations in their use. While our focus is on information obtained from measurements in the urine, clinical diagnosis in these patients must integrate information obtained from the history, the physical examination, and other laboratory data.
Collapse
Affiliation(s)
- Kamel S. Kamel
- Renal Division, St. Michael’s Hospital and The University of Toronto, Toronto, Ontario, Canada
- Keenan Research Center in the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada
| | - Mitchell L. Halperin
- Renal Division, St. Michael’s Hospital and The University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
25
|
Zhang S, Zhao Y, Wang S, Li M, Xu Y, Ran J, Geng X, He J, Meng J, Shao G, Zhou H, Ge Z, Chen G, Li R, Yang B. Discovery of novel diarylamides as orally active diuretics targeting urea transporters. Acta Pharm Sin B 2021; 11:181-202. [PMID: 33532188 PMCID: PMC7838058 DOI: 10.1016/j.apsb.2020.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/14/2020] [Accepted: 05/26/2020] [Indexed: 12/17/2022] Open
Abstract
Urea transporters (UT) play a vital role in the mechanism of urine concentration and are recognized as novel targets for the development of salt-sparing diuretics. Thus, UT inhibitors are promising for development as novel diuretics. In the present study, a novel UT inhibitor with a diarylamide scaffold was discovered by high-throughput screening. Optimization of the inhibitor led to the identification of a promising preclinical candidate, N-[4-(acetylamino)phenyl]-5-nitrofuran-2-carboxamide (1H), with excellent in vitro UT inhibitory activity at the submicromolar level. The half maximal inhibitory concentrations of 1H against UT-B in mouse, rat, and human erythrocyte were 1.60, 0.64, and 0.13 μmol/L, respectively. Further investigation suggested that 8 μmol/L 1H more powerfully inhibited UT-A1 at a rate of 86.8% than UT-B at a rate of 73.9% in MDCK cell models. Most interestingly, we found for the first time that oral administration of 1H at a dose of 100 mg/kg showed superior diuretic effect in vivo without causing electrolyte imbalance in rats. Additionally, 1H did not exhibit apparent toxicity in vivo and in vitro, and possessed favorable pharmacokinetic characteristics. 1H shows promise as a novel diuretic to treat hyponatremia accompanied with volume expansion and may cause few side effects.
Collapse
Key Words
- AQP1, aquaporin 1
- BCRP, breast cancer resistance protein
- CCK-8, cell counting kit-8
- CMC-Na, carboxymethylcellulose sodium
- DMF, N,N-dimethylformamide
- Diuretic
- Fa, fraction absorbance
- GFR, glomerular filtration rate
- HDL-C and LDL-C, high- and low-density lipoprotein
- IC50, half maximal inhibitory concentration
- IMCD, inner medulla collecting duct
- Oral administration
- P-gp, P-glycoprotein
- PBS, phosphate buffered saline
- Papp, apparent permeability
- Structure optimization
- THF, tetrahydrofuran
- UT, urea transporter
- Urea transporter inhibitor
- r.t., room temperature
Collapse
|
26
|
Abstract
Complex multicellular life in mammals relies on functional cooperation of different organs for the survival of the whole organism. The kidneys play a critical part in this process through the maintenance of fluid volume and composition homeostasis, which enables other organs to fulfil their tasks. The renal endothelium exhibits phenotypic and molecular traits that distinguish it from endothelia of other organs. Moreover, the adult kidney vasculature comprises diverse populations of mostly quiescent, but not metabolically inactive, endothelial cells (ECs) that reside within the kidney glomeruli, cortex and medulla. Each of these populations supports specific functions, for example, in the filtration of blood plasma, the reabsorption and secretion of water and solutes, and the concentration of urine. Transcriptional profiling of these diverse EC populations suggests they have adapted to local microenvironmental conditions (hypoxia, shear stress, hyperosmolarity), enabling them to support kidney functions. Exposure of ECs to microenvironment-derived angiogenic factors affects their metabolism, and sustains kidney development and homeostasis, whereas EC-derived angiocrine factors preserve distinct microenvironment niches. In the context of kidney disease, renal ECs show alteration in their metabolism and phenotype in response to pathological changes in the local microenvironment, further promoting kidney dysfunction. Understanding the diversity and specialization of kidney ECs could provide new avenues for the treatment of kidney diseases and kidney regeneration.
Collapse
|
27
|
Shin SH, Wendland MF, Vandsburger MH. Delayed urea differential enhancement CEST (dudeCEST)-MRI with T 1 correction for monitoring renal urea handling. Magn Reson Med 2020; 85:2791-2804. [PMID: 33180343 DOI: 10.1002/mrm.28583] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE We demonstrate a method of delayed urea differential enhancement CEST for probing urea recycling action of the kidney using expanded multi-pool Lorentzian fitting and apparent exchange-dependent relaxation compensation. METHODS T1 correction of urea CEST contrast by apparent exchange-dependent relaxation was tested in phantoms. Nine mice were scanned at 7 Tesla following intraperitoneal injection of 2M 150 μL urea, and later saline. T1 maps and Z-spectra were acquired before and 20 and 40 min postinjection. Z-spectra were fit to a 7-pool Lorentzian model for CEST quantification and compared to urea assay of kidney homogenate. Renal injury was induced by aristolochic acid in 7 mice, and the same scan protocol was performed. RESULTS Apparent exchange-dependent relaxation corrected for variable T1 times in phantoms. Urea CEST contrast at +1 ppm increased significantly at both time points following urea injection in the inner medulla and papilla. When normalizing the postinjection urea CEST contrast to the corresponding baseline value, both urea and saline injection resulted in identical fold changes in urea CEST contrast. Urea assay of kidney homogenate showed a significant correlation to both apparent exchange-dependent relaxation (R2 = 0.4687, P = .0017) and non-T1 -corrected Lorentzian amplitudes (R2 = 0.4964, P = .0011). Renal injury resulted in increased T1 time in the cortex and reduced CEST contrast change upon urea and saline infusion. CONCLUSION Delayed urea enhancement following infusion can provide insight into renal urea handling. In addition, changes in CEST contrast at 1.0 ppm following saline infusion may provide insight into renal function.
Collapse
Affiliation(s)
- Soo Hyun Shin
- Department of Bioengineering, University of California, Berkeley, Berkeley, California, USA
| | - Michael F Wendland
- Berkeley Preclinical Imaging Core (BPIC), University of California, Berkeley, Berkeley, California, USA
| | - Moriel H Vandsburger
- Department of Bioengineering, University of California, Berkeley, Berkeley, California, USA
| |
Collapse
|
28
|
Dobrek L, Nalik-Iwaniak K, Fic K, Arent Z. The Effect of Acetylcysteine on Renal Function in Experimental Models of Cyclophosphamide-and Ifosfamide-Induced Cystitis. Curr Urol 2020; 14:150-162. [PMID: 33224008 DOI: 10.1159/000499245] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 12/12/2019] [Indexed: 01/17/2023] Open
Abstract
Introduction Urotoxicity is a characteristic attribute of cy-clophosphamide and ifosfamide. Acetylcysteine is perceived as a uroprotective and possible nephroprotective compound. The purpose of the study was to assess the effect of acetylcysteine treatment on the morphology of the kidneys and the urinary bladder, and renal function in rats with cystitis induced by cyclophosphamide or ifosfamide. Methods Cystitis was induced in rats belonging to groups 2 and 3, as well as 4 and 5, by five administrations of cyclophosphamide (75 mg/kg) or ifosfamide (80 mg/kg) respectively. Additionally, groups 3 and 5 received acetylcysteine (200 mg/kg). Group 1 was "sham treated" as a control. Upon conclusion of the experiment, the animals were euthanized and their kidneys and urinary bladders were collected for histopathological analysis. The assessment of renal function was based on classic nitrogen blood parameters (urea, creatinine, and uric acid), as well as proteinuria and cystatin C (CysC) and kidney injury molecule-1 (KIM-1) urinary concentrations, and their 24-hour elimination with urine. Results Reduction of blood urea nitrogen and uric acid, and urinary pH with a significant increase of CysC and KIM-1 urinary concentrations, and their 24-hour elimination with urine were observed in groups 2 and 4. The acetylcysteine treatment did not cause a significant change of blood parameters, but significantly decreased 24-hour elimination of CysC and KIM-1 with urine, and accounted for alleviation of the histopathological abnormalities of urinary bladders, with no significant effects on the structure of the kidneys. Conclusions Acetylcysteine used in the experimental model of cyclophosphamide- and ifosfamide-induced cystitis had a uroprotective effect and also reduced renal dysfunction, which suggests its potential use as a nephroprotective compound in cyclophosphamide/ifosfamide therapy.
Collapse
Affiliation(s)
- Lukasz Dobrek
- Department of Clinical Pharmacology, Wroclaw Medical University, Wroclaw, Poland
| | - Klaudia Nalik-Iwaniak
- Experimental and Innovative Medicine Centre, University Centre of Veterinary Medicine UJ-UR, University of Agriculture in Krakow, Krakow, Poland
| | - Kinga Fic
- Experimental and Innovative Medicine Centre, University Centre of Veterinary Medicine UJ-UR, University of Agriculture in Krakow, Krakow, Poland
| | - Zbigniew Arent
- Experimental and Innovative Medicine Centre, University Centre of Veterinary Medicine UJ-UR, University of Agriculture in Krakow, Krakow, Poland
| |
Collapse
|
29
|
Chapman CL, Johnson BD, Parker MD, Hostler D, Pryor RR, Schlader Z. Kidney physiology and pathophysiology during heat stress and the modification by exercise, dehydration, heat acclimation and aging. Temperature (Austin) 2020; 8:108-159. [PMID: 33997113 PMCID: PMC8098077 DOI: 10.1080/23328940.2020.1826841] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/17/2020] [Accepted: 09/17/2020] [Indexed: 02/06/2023] Open
Abstract
The kidneys' integrative responses to heat stress aid thermoregulation, cardiovascular control, and water and electrolyte regulation. Recent evidence suggests the kidneys are at increased risk of pathological events during heat stress, namely acute kidney injury (AKI), and that this risk is compounded by dehydration and exercise. This heat stress related AKI is believed to contribute to the epidemic of chronic kidney disease (CKD) occurring in occupational settings. It is estimated that AKI and CKD affect upwards of 45 million individuals in the global workforce. Water and electrolyte disturbances and AKI, both of which are representative of kidney-related pathology, are the two leading causes of hospitalizations during heat waves in older adults. Structural and physiological alterations in aging kidneys likely contribute to this increased risk. With this background, this comprehensive narrative review will provide the first aggregation of research into the integrative physiological response of the kidneys to heat stress. While the focus of this review is on the human kidneys, we will utilize both human and animal data to describe these responses to passive and exercise heat stress, and how they are altered with heat acclimation. Additionally, we will discuss recent studies that indicate an increased risk of AKI due to exercise in the heat. Lastly, we will introduce the emerging public health crisis of older adults during extreme heat events and how the aging kidneys may be more susceptible to injury during heat stress.
Collapse
Affiliation(s)
- Christopher L. Chapman
- Center for Research and Education in Special Environments, Department of Exercise and Nutrition Sciences, University at Buffalo, Buffalo, NY, USA
- Department of Human Physiology, University of Oregon, Eugene, OR, USA
| | - Blair D. Johnson
- Department of Kinesiology, School of Public Health, Indiana University, Bloomington, IN, USA
| | - Mark D. Parker
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
- Department of Ophthalmology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - David Hostler
- Center for Research and Education in Special Environments, Department of Exercise and Nutrition Sciences, University at Buffalo, Buffalo, NY, USA
| | - Riana R. Pryor
- Center for Research and Education in Special Environments, Department of Exercise and Nutrition Sciences, University at Buffalo, Buffalo, NY, USA
| | - Zachary Schlader
- Department of Kinesiology, School of Public Health, Indiana University, Bloomington, IN, USA
| |
Collapse
|
30
|
Marton A, Kaneko T, Kovalik JP, Yasui A, Nishiyama A, Kitada K, Titze J. Organ protection by SGLT2 inhibitors: role of metabolic energy and water conservation. Nat Rev Nephrol 2020; 17:65-77. [PMID: 33005037 DOI: 10.1038/s41581-020-00350-x] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/18/2020] [Indexed: 12/17/2022]
Abstract
Therapeutic inhibition of the sodium-glucose co-transporter 2 (SGLT2) leads to substantial loss of energy (in the form of glucose) and additional solutes (in the form of Na+ and its accompanying anions) in urine. However, despite the continuously elevated solute excretion, long-term osmotic diuresis does not occur in humans with SGLT2 inhibition. Rather, patients on SGLT2 inhibitor therapy adjust to the reduction in energy availability and conserve water. The metabolic adaptations that are induced by SGLT2 inhibition are similar to those observed in aestivation - an evolutionarily conserved survival strategy that enables physiological adaptation to energy and water shortage. Aestivators exploit amino acids from muscle to produce glucose and fatty acid fuels. This endogenous energy supply chain is coupled with nitrogen transfer for organic osmolyte production, which allows parallel water conservation. Moreover, this process is often accompanied by a reduction in metabolic rate. By comparing aestivation metabolism with the fuel switches that occur during therapeutic SGLT2 inhibition, we suggest that SGLT2 inhibitors induce aestivation-like metabolic patterns, which may contribute to the improvements in cardiac and renal function observed with this class of therapeutics.
Collapse
Affiliation(s)
- Adriana Marton
- Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Tatsuroh Kaneko
- Medicine Division, Nippon Boehringer Ingelheim Co., Ltd, Tokyo, Japan
| | - Jean-Paul Kovalik
- Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Atsutaka Yasui
- Medicine Division, Nippon Boehringer Ingelheim Co., Ltd, Tokyo, Japan
| | - Akira Nishiyama
- Department of Pharmacology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Kento Kitada
- Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore.,Department of Pharmacology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Jens Titze
- Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore. .,Division of Nephrology and Hypertension, University Clinic Erlangen, Erlangen, Germany. .,Division of Nephrology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
31
|
AQP2: Mutations Associated with Congenital Nephrogenic Diabetes Insipidus and Regulation by Post-Translational Modifications and Protein-Protein Interactions. Cells 2020; 9:cells9102172. [PMID: 32993088 PMCID: PMC7599609 DOI: 10.3390/cells9102172] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 09/22/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023] Open
Abstract
, the molecular defects in the AVPR2 and AQP2 mutants, post-translational modifications (i.e., phosphorylation, ubiquitination, and glycosylation) and various protein-protein interactions that regulate phosphorylation, ubiquitination, tetramerization, trafficking, stability, and degradation of AQP2.
Collapse
|
32
|
Guo Z, Niu X, Fu G, Yang B, Chen G, Sun S. SLC14A1 (UT-B) gene rearrangement in urothelial carcinoma of the bladder: a case report. Diagn Pathol 2020; 15:94. [PMID: 32703295 PMCID: PMC7376696 DOI: 10.1186/s13000-020-01009-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 07/14/2020] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Bladder cancer (BC) is a common and deadly disease. Over the past decade, a number of genetic alterations have been reported in BC. Bladder urothelium expresses abundant urea transporter UT-B encoded by Slc14a1 gene at 18q12.3 locus, which plays an important role in preventing high concentrated urea-caused cell injury. Early genome-wide association studies (GWAS) showed that UT-B gene mutations are genetically linked to the urothelial bladder carcinoma (UBC). In this study, we examined whether Slc14a1 gene has been changed in UBC, which has never been reported. CASE PRESENTATION A 59-year-old male was admitted to a hospital with the complaint of gross hematuria for 6 days. Ultrasonography revealed a size of 2.8 × 1.7 cm mass lesion located on the rear wall and dome of the bladder. In cystoscopic examination, papillary tumoral lesions 3.0-cm in total diameter were seen on the left wall of the bladder and 2 cm to the left ureteric orifice. Transurethral resection of bladder tumor (TURBT) was performed. Histology showed high-grade non-muscle invasive UBC. Immunostaining was negative for Syn, CK7, CK20, Villin, and positive for HER2, BRCA1, GATA3. Using a fluorescence in situ hybridization (FISH), Slc14a1 gene rearrangement was identified by a pair of break-apart DNA probes. CONCLUSIONS We for the first time report a patient diagnosed with urothelial carcinoma accompanied with split Slc14a1 gene abnormality, a crucial gene in bladder.
Collapse
Affiliation(s)
- Zhongying Guo
- Department of Pathology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, 223300, China
| | - Xiaobing Niu
- Department of Urology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, 223300, China
| | - Guangbo Fu
- Department of Urology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, 223300, China
| | - Baoxue Yang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Guangping Chen
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Su'an Sun
- Department of Pathology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, 223300, China.
| |
Collapse
|
33
|
Geng X, Zhang S, He J, Ma A, Li Y, Li M, Zhou H, Chen G, Yang B. The urea transporter UT-A1 plays a predominant role in a urea-dependent urine-concentrating mechanism. J Biol Chem 2020; 295:9893-9900. [PMID: 32461256 PMCID: PMC7380188 DOI: 10.1074/jbc.ra120.013628] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/22/2020] [Indexed: 12/12/2022] Open
Abstract
Urea transporters are a family of urea-selective channel proteins expressed in multiple tissues that play an important role in the urine-concentrating mechanism of the mammalian kidney. Previous studies have shown that knockout of urea transporter (UT)-B, UT-A1/A3, or all UTs leads to urea-selective diuresis, indicating that urea transporters have important roles in urine concentration. Here, we sought to determine the role of UT-A1 in the urine-concentrating mechanism in a newly developed UT-A1-knockout mouse model. Phenotypically, daily urine output in UT-A1-knockout mice was nearly 3-fold that of WT mice and 82% of all-UT-knockout mice, and the UT-A1-knockout mice had significantly lower urine osmolality than WT mice. After 24-h water restriction, acute urea loading, or high-protein (40%) intake, UT-A1-knockout mice were unable to increase urine-concentrating ability. Compared with all-UT-knockout mice, the UT-A1-knockout mice exhibited similarly elevated daily urine output and decreased urine osmolality, indicating impaired urea-selective urine concentration. Our experimental findings reveal that UT-A1 has a predominant role in urea-dependent urine-concentrating mechanisms, suggesting that UT-A1 represents a promising diuretic target.
Collapse
Affiliation(s)
- Xiaoqiang Geng
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Shun Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jinzhao He
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Ang Ma
- 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
| | - 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
| | - Guangping Chen
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - 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
|
34
|
Allen GJP, Kuan PL, Tseng YC, Hwang PP, Quijada-Rodriguez AR, Weihrauch D. Specialized adaptations allow vent-endemic crabs (Xenograpsus testudinatus) to thrive under extreme environmental hypercapnia. Sci Rep 2020; 10:11720. [PMID: 32678186 PMCID: PMC7367285 DOI: 10.1038/s41598-020-68656-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 06/29/2020] [Indexed: 12/13/2022] Open
Abstract
Shallow hydrothermal vent environments are typically very warm and acidic due to the mixing of ambient seawater with volcanic gasses (> 92% CO2) released through the seafloor making them potential ‘natural laboratories’ to study long-term adaptations to extreme hypercapnic conditions. Xenograpsus testudinatus, the shallow hydrothermal vent crab, is the sole metazoan inhabitant endemic to vents surrounding Kueishantao Island, Taiwan, where it inhabits waters that are generally pH 6.50 with maximum acidities reported as pH 5.50. This study assessed the acid–base regulatory capacity and the compensatory response of X. testudinatus to investigate its remarkable physiological adaptations. Hemolymph parameters (pH, [HCO3−], \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$${\text{P}}_{{{\text{CO}}_{2} }}$$\end{document}PCO2, [NH4+], and major ion compositions) and the whole animal’s rates of oxygen consumption and ammonia excretion were measured throughout a 14-day acclimation to pH 6.5 and 5.5. Data revealed that vent crabs are exceptionally strong acid–base regulators capable of maintaining homeostatic pH against extreme hypercapnia (pH 5.50, 24.6 kPa \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$${\text{P}}_{{{\text{CO}}_{2} }}$$\end{document}PCO2) via HCO3−/Cl− exchange, retention and utilization of extracellular ammonia. Intact crabs as well as their isolated perfused gills maintained \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$${\text{P}}_{{{\text{CO}}_{2} }}$$\end{document}PCO2tensions below environmental levels suggesting the gills can excrete CO2 against a hemolymph-directed \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$${\text{P}}_{{{\text{CO}}_{2} }}$$\end{document}PCO2 gradient. These specialized physiological mechanisms may be amongst the adaptations required by vent-endemic animals surviving in extreme conditions.
Collapse
Affiliation(s)
- Garett J P Allen
- Biological Sciences, University of Manitoba, 190 Dysart Rd., Winnipeg, MB, R3T 2M8, Canada
| | - Pou-Long Kuan
- Institute of Cellular and Organismal Biology's Marine Research Station, Academia Sinica, No. 23-10 Dawen Rd., Jiaoxi, 262, Yilan County, Taiwan
| | - Yung-Che Tseng
- Institute of Cellular and Organismal Biology's Marine Research Station, Academia Sinica, No. 23-10 Dawen Rd., Jiaoxi, 262, Yilan County, Taiwan
| | - Pung-Pung Hwang
- Institute of Cellular and Organismal Biology, Academia Sinica, No. 128, Section 2, Academia Rd., Nangang District, Taipei City, 11529, Taiwan
| | | | - Dirk Weihrauch
- Biological Sciences, University of Manitoba, 190 Dysart Rd., Winnipeg, MB, R3T 2M8, Canada.
| |
Collapse
|
35
|
Cody JD. The Consequences of Abnormal Gene Dosage: Lessons from Chromosome 18. Trends Genet 2020; 36:764-776. [PMID: 32660784 DOI: 10.1016/j.tig.2020.06.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 12/18/2022]
Abstract
Accurate interpretation of genomic copy number variation (CNV) remains a challenge and has important consequences for both congenital and late-onset disease. Hemizygosity dosage characterization of the genes on chromosome 18 reveals a spectrum of outcomes ranging from no clinical effect, to risk factors for disease, to both low- and high-penetrance disease. These data are important for accurate and predictive clinical management. Additionally, the potential mechanisms of reduced penetrance due to dosage compensation are discussed as a key to understanding avenues for potential treatment. This review describes the chromosome 18 findings, and discusses the molecular mechanisms that allow haploinsufficiency, reduced penetrance, and dosage compensation.
Collapse
Affiliation(s)
- Jannine DeMars Cody
- Department of Pediatrics, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Chromosome 18 Registry and Research Society, San Antonio, TX 78229, USA.
| |
Collapse
|
36
|
Pallares RM, An DD, Tewari P, Wang ET, Abergel RJ. Rapid Detection of Gadolinium-Based Contrast Agents in Urine with a Chelated Europium Luminescent Probe. ACS Sens 2020; 5:1281-1286. [PMID: 32352783 DOI: 10.1021/acssensors.0c00615] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Gadolinium-based contrast agents are widely used in magnetic resonance imaging procedures to enhance image contrast. Despite their ubiquitous use in clinical settings, gadolinium is not an innocuous element, as suggested by several disorders associated with its use. Therefore, novel analytical technologies capable of tracking contrast agent excretion through urine are necessary for optimizing patient safety after imaging procedures. Here, we describe an assay to detect and quantify contrast agents in urine based on the luminescence quenching of a metal chelate probe, Eu3+-3,4,3-LI(1,2-HOPO), which only requires 10 min incubation before measurement. Gadolinium-based contrast agents prevent the formation of the Eu3+-3,4,3-LI(1,2-HOPO) complex, subsequently decreasing the luminescence of the assay solution. Three commercial contrast agents, Magnevist, Multihance, and Omniscan, were used to demonstrate the analytical concept in synthetic human urine, and subsequent quantification of mouse urine samples. To the best of our knowledge, this is the first assay capable of detecting and quantifying gadolinium-based contrast agents in urine without sample preparation or digestion.
Collapse
Affiliation(s)
- Roger M. Pallares
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Dahlia D. An
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Pariswi Tewari
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Elizabeth T. Wang
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Rebecca J. Abergel
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
- Department of Nuclear Engineering, University of California, Berkeley, California 94720, United States
| |
Collapse
|
37
|
Li M, Zhao Y, Zhang S, Xu Y, Wang SY, Li BW, Ran JH, Li RT, Yang BX. A thienopyridine, CB-20, exerts diuretic activity by inhibiting urea transporters. Acta Pharmacol Sin 2020; 41:65-72. [PMID: 31213671 PMCID: PMC7468274 DOI: 10.1038/s41401-019-0245-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 04/28/2019] [Indexed: 01/08/2023]
Abstract
Urea transporters (UTs) are transmembrane proteins selectively permeable to urea and play an important role in urine concentration. UT-knockout mice exhibit the urea-selective urine-concentrating defect, without affecting electrolyte balance, suggesting that UT-B inhibitors have the potential to be developed as novel diuretics. In this study, we characterized a novel compound 5-ethyl-2-methyl-3-amino-6-methylthieno[2,3-b]pyridine-2,5-dicarboxylate (CB-20) with UT inhibitory activity as novel diuretics with excellent pharmacological properties. This compound was discovered based on high-throughput virtual screening combined with the erythrocyte osmotic lysis assay. Selectivity of UT inhibitors was assayed using transwell chambers. Diuretic activity of the compound was examined in rats and mice using metabolic cages. Pharmacokinetic parameters were detected in rats using LC-MS/MS. Molecular docking was employed to predict the potential binding modes for the CB-20 with human UT-B. This compound dose-dependently inhibited UT-facilitated urea transport with IC50 values at low micromolar levels. It exhibited nearly equal inhibitory activity on both UT-A1 and UT-B. After subcutaneous administration of CB-20, the animals showed polyuria, without electrolyte imbalance and abnormal metabolism. CB-20 possessed a good absorption and rapid clearance in rat plasma. Administration of CB-20 for 5 days did not cause significant morphological abnormality in kidney or liver tissues of rats. Molecular docking showed that CB-20 was positioned near several residues in human UT-B, including Leu364, Val367, and so on. This study provides proof of evidence for the prominent diuretic activity of CB-20 by specifically inhibiting UTs. CB-20 or thienopyridine analogs may be developed as novel diuretics.
Collapse
Affiliation(s)
- Min Li
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Yan Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, School of pharmaceutical Sciences, Peking University, Beijing, 100191, China
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, 010110, China
| | - Shun Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Yue Xu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Shu-Yuan Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Bo-Wen Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Jian-Hua Ran
- Chongqing Medical University, Chongqing, 400016, China
| | - Run-Tao Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| | - Bao-Xue Yang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
| |
Collapse
|
38
|
Ueda Y, Kawamoto K, Konno M, Noguchi K, Kaifuchi S, Satoh T, Eguchi H, Doki Y, Hirotsu T, Mori M, Ishii H. Application of C. elegans cancer screening test for the detection of pancreatic tumor in genetically engineered mice. Oncotarget 2019; 10:5412-5418. [PMID: 31534627 PMCID: PMC6739214 DOI: 10.18632/oncotarget.27124] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 07/17/2019] [Indexed: 12/20/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) exhibits a very early onset of metastasis. Thus, early detection and treatment are pivotal to successful eradication of pancreatic cancers. Economical and non-invasive cancer screening systems is indispensable for this purpose. Previously our group developed a novel method to detect various kinds of human cancer using nematode Caenorhabditis elegans (C. elegans) that respond to cancer odor in urine; however, whether this method is useful for non-human species remains to be understood. In this study, we examined its effectiveness in the detection of murine pancreatic tumor spontaneously generated in genetically-engineered mice. We generated pancreas-specific Kras G12D and/or c-Met deletion mutant mice and measured the probability of spontaneous tumor generation in these mice. The chemotactic indexes of C. elegans to the urine samples of these mutant mice were measured. As previously described, oncogenic KrasG12D was necessary to induce pancreatic intraepithelial neoplasia in this mouse model, while c-Met mutation did not show further effect. The chemotactic analysis indicated that C. elegans avoids urine of healthy recipient mice, while they tended to be attracted to urine of mice with KrasG12D . Our study demonstrated that C. elegans can recognize the odor of pancreatic cancer in urine of KrasG12D model mouse, suggesting the similarity of cancer odor between species. Our result facilitates further studies on mechanism of cancer detection by C. elegans.
Collapse
Affiliation(s)
- Yuji Ueda
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan.,Department of Medical Data Science, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan.,Department of Frontier Science for Cancer and Chemotherapy, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Koichi Kawamoto
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan.,Department of Medical Data Science, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan.,Department of Frontier Science for Cancer and Chemotherapy, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Masamitsu Konno
- Department of Medical Data Science, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan.,Department of Frontier Science for Cancer and Chemotherapy, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Kozo Noguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan.,Department of Medical Data Science, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | | | - Taroh Satoh
- Department of Frontier Science for Cancer and Chemotherapy, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Takaaki Hirotsu
- Hirotsu Bioscience Co., Ltd., Tokyo 107-0062, Japan.,Department of Biology, Graduate School of Science, Kyushu University, Fukuoka 819-0395, Japan
| | - Masaki Mori
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Hideshi Ishii
- Department of Medical Data Science, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan.,Department of Frontier Science for Cancer and Chemotherapy, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
39
|
Suzuki T, Kumagai M, Furusawa M. Effects of Urea on the Reactions of Nucleosides with Hypobromous Acid. Chem Pharm Bull (Tokyo) 2019; 67:707-712. [DOI: 10.1248/cpb.c18-01009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
40
|
Horvath L, Bächinger D, Honegger T, Bodmer D, Naldi AM. Functional and morphological analysis of different aminoglycoside treatment regimens inducing hearing loss in mice. Exp Ther Med 2019; 18:1123-1130. [PMID: 31316607 PMCID: PMC6601143 DOI: 10.3892/etm.2019.7687] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 05/23/2019] [Indexed: 01/05/2023] Open
Abstract
Aminoglycoside ototoxicity is common in clinical practice but reliable protective agents currently do not exist. Aminoglycoside regimens causing ototoxicity in different laboratory animals are under investigation. The assessment method used most commonly to determine auditory effects is the auditory brainstem response (ABR). Distortion product otoacoustic emissions (DPOAE) have been used less frequently. A precise recommendation on the specific method to assess peripheral auditory function before and after aminoglycoside toxicity in mice does not exist. In order to evaluate various mouse models for ototoxic injury caused by various aminoglycoside regimens, there is a need for performing preliminary tests in small cohorts before large experiments. The aim of our study was to investigate different aminoglycoside regimens that cause substantial ototoxic damage in vivo. Aminoglycosides are safe and produce a detectable hearing threshold shift in a small cohort of mice that can be used as a model for preliminary tests. Different ototoxic regimens were assessed by ABR and DPOAE measurements pre- and post-treatment. Further, the sensory cell loss was quantified by counting hair cells in the cochlea. It was revealed that an ototoxic regimen with kanamycin twice daily for 15 consecutive days is safe, well tolerated and produces an early significant hearing threshold shift detected by DPOAE in a small cohort of mice. The study compared ABR and DPOAE in mentioned regimens for the first time and illustrated that DPOAE is well suited for detecting hearing threshold shifts in high frequencies before ABR threshold shifts occur in accordance with predominating outer hair cell damage mainly in the basal turn of the cochlea.
Collapse
Affiliation(s)
- Lukas Horvath
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland.,Department of Biomedicine and The Clinic for Otorhinolaryngology, University Hospital Basel, 4031 Basel, Switzerland
| | - David Bächinger
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Tim Honegger
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Daniel Bodmer
- Department of Biomedicine and The Clinic for Otorhinolaryngology, University Hospital Basel, 4031 Basel, Switzerland
| | - Arianne Monge Naldi
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
41
|
Zhao Y, Li M, Li B, Zhang S, Su A, Xing Y, Ge Z, Li R, Yang B. Discovery and optimization of thienopyridine derivatives as novel urea transporter inhibitors. Eur J Med Chem 2019; 172:131-142. [PMID: 30959323 DOI: 10.1016/j.ejmech.2019.03.060] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/26/2019] [Accepted: 03/28/2019] [Indexed: 10/27/2022]
Abstract
Urea transporters (UTs) play an important role in the urine concentrating mechanism and are recognized as novel targets for developing small molecule inhibitors with salt-sparing diuretic activity. Thienoquinoline derivatives, a class of novel UT-B inhibitors identified by our group, play a significant diuresis in animal model. However, the poor solubility and low bioavailability limited its further development. To overcome these shortcomings, the structure modification of thienoquinoline was carried out in this study, which led to the discovery of novel thienopyridine derivatives as specific urea transporter inhibitors. Further optimization obtained the promising preclinical candidate 8n with not only excellent inhibition effect on urea transporters and diuretic activity on rat model, but also suitable water solubility and Log P value.
Collapse
Affiliation(s)
- Yan Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, School of pharmaceutical Sciences, Peking University, 100191, PR, China; College of Pharmacy, Inner Mongolia Medical University, 010110, PR, China
| | - Min Li
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 100191, PR, China
| | - Bowen Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of pharmaceutical Sciences, Peking University, 100191, PR, China
| | - Shun Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 100191, PR, China
| | - Aoze Su
- State Key Laboratory of Natural and Biomimetic Drugs, School of pharmaceutical Sciences, Peking University, 100191, PR, China
| | - Yongning Xing
- State Key Laboratory of Natural and Biomimetic Drugs, School of pharmaceutical Sciences, Peking University, 100191, PR, China
| | - Zemei Ge
- State Key Laboratory of Natural and Biomimetic Drugs, School of pharmaceutical Sciences, Peking University, 100191, PR, China
| | - Runtao Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of pharmaceutical Sciences, Peking University, 100191, PR, China.
| | - Baoxue Yang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 100191, PR, China.
| |
Collapse
|
42
|
Selim RE, Ahmed HH, Abd-Allah SH, Sabry GM, Hassan RE, Khalil WKB, Abouhashem NS. Mesenchymal Stem Cells: a Promising Therapeutic Tool for Acute Kidney Injury. Appl Biochem Biotechnol 2019; 189:284-304. [PMID: 30976980 DOI: 10.1007/s12010-019-02995-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/26/2019] [Indexed: 12/18/2022]
Abstract
Acute kidney injury (AKI) is a rapid loss of renal function. It has high mortality rates. Still, renal replacement therapy is considered the best solution for recovering AKI. This opens a line of thought to develop an alternative therapy for it without complications. Mesenchymal stem cells are considered a new therapy for treating kidney diseases. The aim of this work was to address the anti-apoptotic, antioxidative, and pro-angiogenic effects of adipose tissue-derived MSCs (AD-MSCs) and bone marrow-MSCs (BM-MSCs) for treating AKI. Adult male Wistar rats were assigned into nine groups (n = 10): (1) the control group; (2) the AKI group, receiving cisplatin; (3) the AKI group treated with AD-MSCs (1 × 106); (4) the AKI group treated with AD-MSCs (2 × 106); (5) the AKI group treated with AD-MSCs (4 × 106); (6) the AKI group treated with losartan; (7) the AKI group treated with BM-MSCs (1 × 106); (8) the AKI group treated with BM-MSCs (2 × 106); and (9) the AKI group treated with BM-MSCs (4 × 106). The results showed a significant rise in creatinine, urea, and cystatin C (cys C) levels and upregulation of p38 mRNA, whereas a significant decline in NAD(P)H quinone oxidoreductase 1 (NQO-1) protein and downregulation of B-cell lymphoma-2 (Bcl-2) mRNA and vascular endothelial growth factor (VEGF) mRNA were recorded in AKI. MSCs could improve renal functions manifested by decreased urea, creatinine, and cys C levels; downregulation of p38; and upregulation of Bcl-2 and VEGF. Moreover, MSC therapy could induce NQO-1 in the treated rats relative to the untreated rats. So, cell-based therapy can reduce AKI through the antioxidative, anti-apoptotic, and pro-angiogenic properties of MSCs. Therefore, the findings received in this attempt create a fertile base for the setup of cell therapy in patients with AKI.
Collapse
Affiliation(s)
- Rehab E Selim
- Hormones Department, National Research Centre, Giza, Egypt. .,Stem Cell Lab., Centre of Excellence for Advanced Science, National Research Centre, Dokki, Giza, Egypt.
| | - Hanaa H Ahmed
- Hormones Department, National Research Centre, Giza, Egypt.,Stem Cell Lab., Centre of Excellence for Advanced Science, National Research Centre, Dokki, Giza, Egypt
| | - Somia H Abd-Allah
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Gilane M Sabry
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Rasha E Hassan
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | | | - Nehal S Abouhashem
- Pathology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
43
|
Armbruster CE, Forsyth VS, Johnson AO, Smith SN, White AN, Brauer AL, Learman BS, Zhao L, Wu W, Anderson MT, Bachman MA, Mobley HLT. Twin arginine translocation, ammonia incorporation, and polyamine biosynthesis are crucial for Proteus mirabilis fitness during bloodstream infection. PLoS Pathog 2019; 15:e1007653. [PMID: 31009518 PMCID: PMC6497324 DOI: 10.1371/journal.ppat.1007653] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 05/02/2019] [Accepted: 02/22/2019] [Indexed: 12/30/2022] Open
Abstract
The Gram-negative bacterium Proteus mirabilis is a common cause of catheter-associated urinary tract infections (CAUTI), which can progress to secondary bacteremia. While numerous studies have investigated experimental infection with P. mirabilis in the urinary tract, little is known about pathogenesis in the bloodstream. This study identifies the genes that are important for survival in the bloodstream using a whole-genome transposon insertion-site sequencing (Tn-Seq) approach. A library of 50,000 transposon mutants was utilized to assess the relative contribution of each non-essential gene in the P. mirabilis HI4320 genome to fitness in the livers and spleens of mice at 24 hours following tail vein inoculation compared to growth in RPMI, heat-inactivated (HI) naïve serum, and HI acute phase serum. 138 genes were identified as ex vivo fitness factors in serum, which were primarily involved in amino acid transport and metabolism, and 143 genes were identified as infection-specific in vivo fitness factors for both spleen and liver colonization. Infection-specific fitness factors included genes involved in twin arginine translocation, ammonia incorporation, and polyamine biosynthesis. Mutants in sixteen genes were constructed to validate both the ex vivo and in vivo results of the transposon screen, and 12/16 (75%) exhibited the predicted phenotype. Our studies indicate a role for the twin arginine translocation (tatAC) system in motility, translocation of potential virulence factors, and fitness within the bloodstream. We also demonstrate the interplay between two nitrogen assimilation pathways in the bloodstream, providing evidence that the GS-GOGAT system may be preferentially utilized. Furthermore, we show that a dual-function arginine decarboxylase (speA) is important for fitness within the bloodstream due to its role in putrescine biosynthesis rather than its contribution to maintenance of membrane potential. This study therefore provides insight into pathways needed for fitness within the bloodstream, which may guide strategies to reduce bacteremia-associated mortality.
Collapse
Affiliation(s)
- Chelsie E. Armbruster
- Department of Microbiology and Immunology; Jacobs School of Medicine and Biomedical Sciences; State University of New York at Buffalo; Buffalo, NY, United States of America
| | - Valerie S. Forsyth
- Department of Microbiology and Immunology; University of Michigan Medical School; Ann Arbor, MI, United States of America
| | - Alexandra O. Johnson
- Department of Microbiology and Immunology; Jacobs School of Medicine and Biomedical Sciences; State University of New York at Buffalo; Buffalo, NY, United States of America
| | - Sara N. Smith
- Department of Microbiology and Immunology; University of Michigan Medical School; Ann Arbor, MI, United States of America
| | - Ashley N. White
- Department of Microbiology and Immunology; Jacobs School of Medicine and Biomedical Sciences; State University of New York at Buffalo; Buffalo, NY, United States of America
| | - Aimee L. Brauer
- Department of Microbiology and Immunology; Jacobs School of Medicine and Biomedical Sciences; State University of New York at Buffalo; Buffalo, NY, United States of America
| | - Brian S. Learman
- Department of Microbiology and Immunology; Jacobs School of Medicine and Biomedical Sciences; State University of New York at Buffalo; Buffalo, NY, United States of America
| | - Lili Zhao
- Department of Biostatistics; University of Michigan School of Public Health; Ann Arbor, MI, United States of America
| | - Weisheng Wu
- Department of Computational Medicine & Bioinformatics; University of Michigan Medical School; Ann Arbor, MI, United States of America
| | - Mark T. Anderson
- Department of Microbiology and Immunology; University of Michigan Medical School; Ann Arbor, MI, United States of America
| | - Michael A. Bachman
- Department of Pathology; University of Michigan Medical School; Ann Arbor, MI, United States of America
| | - Harry L. T. Mobley
- Department of Microbiology and Immunology; University of Michigan Medical School; Ann Arbor, MI, United States of America
| |
Collapse
|
44
|
Kamel KS, Schreiber M, Halperin ML. Renal potassium physiology: integration of the renal response to dietary potassium depletion. Kidney Int 2018; 93:41-53. [PMID: 29102372 DOI: 10.1016/j.kint.2017.08.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 07/31/2017] [Accepted: 08/03/2017] [Indexed: 01/30/2023]
Abstract
We summarize the current understanding of the physiology of the renal handling of potassium (K+), and present an integrative view of the renal response to K+ depletion caused by dietary K+ restriction. This renal response involves contributions from different nephron segments, and aims to diminish the rate of excretion of K+ as a result of: decreasing the rate of electrogenic (and increasing the rate of electroneutral) reabsorption of sodium in the aldosterone-sensitive distal nephron (ASDN), decreasing the abundance of renal outer medullary K+ channels in the luminal membrane of principal cells in the ASDN, decreasing the flow rate in the ASDN, and increasing the reabsorption of K+ in the cortical and medullary collecting ducts. The implications of this physiology for the association between K+ depletion and hypertension, and K+ depletion and formation of calcium kidney stones are discussed.
Collapse
Affiliation(s)
- Kamel S Kamel
- Renal Division, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Keenan Research Center, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada.
| | - Martin Schreiber
- Renal Division, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Mitchell L Halperin
- Renal Division, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Keenan Research Center, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| |
Collapse
|
45
|
Abdulkadir A, Mbajiorgu EF, Nyirenda T. Effects of concurrent chloroquine and ethanol administration on the rat kidney morphology. Pan Afr Med J 2018; 29:49. [PMID: 30402202 PMCID: PMC6211805 DOI: 10.11604/pamj.2018.29.49.12471] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 12/19/2017] [Indexed: 11/22/2022] Open
Abstract
Introduction The use of antimalarial chloroquine in malaria-endemic regions of Africa is rampant and it is not uncommon to find individuals taken the drug concurrent with alcohol. Effects of anti-malarial drug chloroquine (Chq) and ethanol (Et) combination on kidney volume and function using rat model was investigated. Methods 32 adult male rats were randomly distributed into four groups of 8 rats each. Group C serve as control and received vehicle only, while Q is Chq treated only, E is Et treated and QE is Et and Chq treated. Chq was administered intraperitoneally at 1mg/100g body weight weekly and 6% Et in drinking water provided ad libitum. Urine volume was collected before the treatment began and after the treatment. After eight weeks, all animals were euthanized; kidneys were harvested and fixed in 10% neutral formalin. The fixed left kidneys were scanned with computed tomography and the scan slices were used to estimate 3-dimensional kidney volume on ImageJ. Results Total kidney volume was none significantly increased in Q, E and QE treated compared to control groups (p = 0.5150). Also, microscopic analysis showed increased proximal tubule diameter (p = 0.1426) and epithelial hypertrophy (p = 0.2530) and significant urinary space shrinkage (p = 0.00001). The initial urine volume was not significantly different between the control and treated groups (p = 0.9864) however, following treatment urine volume was significantly reduced in QE rats group (p = 0.0029). Conclusion The results suggest chloroquine and ethanol combination as potential cause of kidney injury through structural damage and function derangement.
Collapse
Affiliation(s)
- Abdurrahman Abdulkadir
- Department of Anatomy, College of Health sciences, Federal University Birnin Kebbi, Kebbi State, Nigeria and Division of Histology and Embryology, School of Anatomical Sciences, Faculty of Health Sciences, University of The Witwatersrand, Johannesburg, Gauteng, South Africa
| | - Ejikeme Felix Mbajiorgu
- Division of Histology and Embryology, School of Anatomical Sciences, Faculty of Health Sciences, University of The Witwatersrand, Johannesburg, Gauteng, South Africa
| | - Trust Nyirenda
- Department of Anatomy and Physiology, Faculty of Medicine, National University of Science and Technology, Bulawayo, Zimbabwe and Division of Histology and Embryology, School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
| |
Collapse
|
46
|
Nawata CM, Pannabecker TL. Mammalian urine concentration: a review of renal medullary architecture and membrane transporters. J Comp Physiol B 2018; 188:899-918. [PMID: 29797052 PMCID: PMC6186196 DOI: 10.1007/s00360-018-1164-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 04/23/2018] [Accepted: 05/14/2018] [Indexed: 01/10/2023]
Abstract
Mammalian kidneys play an essential role in balancing internal water and salt concentrations. When water needs to be conserved, the renal medulla produces concentrated urine. Central to this process of urine concentration is an osmotic gradient that increases from the corticomedullary boundary to the inner medullary tip. How this gradient is generated and maintained has been the subject of study since the 1940s. While it is generally accepted that the outer medulla contributes to the gradient by means of an active process involving countercurrent multiplication, the source of the gradient in the inner medulla is unclear. The last two decades have witnessed advances in our understanding of the urine-concentrating mechanism. Details of medullary architecture and permeability properties of the tubules and vessels suggest that the functional and anatomic relationships of these structures may contribute to the osmotic gradient necessary to concentrate urine. Additionally, we are learning more about the membrane transporters involved and their regulatory mechanisms. The role of medullary architecture and membrane transporters in the mammalian urine-concentrating mechanism are the focus of this review.
Collapse
Affiliation(s)
- C Michele Nawata
- Department of Physiology, Banner University Medical Center, University of Arizona, 1501 N. Campbell Avenue, Tucson, AZ, 85724-5051, USA.
| | - Thomas L Pannabecker
- Department of Physiology, Banner University Medical Center, University of Arizona, 1501 N. Campbell Avenue, Tucson, AZ, 85724-5051, USA
| |
Collapse
|
47
|
Chamoun MN, Sullivan MJ, Ulett GC. Quantification of bacteriuria caused by Hemolysin-positive Escherichia coli in human and mouse urine using quantitative polymerase chain reaction (qPCR) targeting hlyD. J Microbiol Methods 2018; 152:173-178. [DOI: 10.1016/j.mimet.2018.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 08/10/2018] [Accepted: 08/12/2018] [Indexed: 12/21/2022]
|
48
|
Pharmacokinetics, Tissue Distribution and Excretion of a Novel Diuretic (PU-48) in Rats. Pharmaceutics 2018; 10:pharmaceutics10030124. [PMID: 30096833 PMCID: PMC6160999 DOI: 10.3390/pharmaceutics10030124] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 07/21/2018] [Accepted: 07/27/2018] [Indexed: 01/02/2023] Open
Abstract
Methyl 3-amino-6-methoxythieno [2,3-b] quinoline-2-carboxylate (PU-48) is a novel diuretic urea transporter inhibitor. The aim of this study is to investigate the profile of plasma pharmacokinetics, tissue distribution, and excretion by oral dosing of PU-48 in rats. Concentrations of PU-48 within biological samples are determined using a validated high performance liquid chromatography-tandem mass spectrometry (LC-MS/MS) method. After oral administration of PU-48 (3, 6, and 12 mg/kg, respectively) in self-nanomicroemulsifying drug delivery system (SNEDDS) formulation, the peak plasma concentrations (Cmax), and the area under the curve (AUC0⁻∞) were increased by the dose-dependent and linear manner, but the marked different of plasma half-life (t1/2) were not observed. This suggests that the pharmacokinetic profile of PU-48 prototype was first-order elimination kinetic characteristics within the oral three doses range in rat plasma. Moreover, the prototype of PU-48 was rapidly and extensively distributed into thirteen tissues, especially higher concentrations were detected in stomach, intestine, liver, kidney, and bladder. The total accumulative excretion of PU-48 in the urine, feces, and bile was less than 2%. This research is the first report on disposition via oral administration of PU-48 in rats, and it provides important information for further development of PU-48 as a diuretic drug candidate.
Collapse
|
49
|
Lee S, Cil O, Diez-Cecilia E, Anderson MO, Verkman AS. Nanomolar-Potency 1,2,4-Triazoloquinoxaline Inhibitors of the Kidney Urea Transporter UT-A1. J Med Chem 2018; 61:3209-3217. [PMID: 29589443 PMCID: PMC5976253 DOI: 10.1021/acs.jmedchem.8b00343] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Urea transporter A (UT-A) isoforms encoded by the Slc14a2 gene are expressed in kidney tubule epithelial cells, where they facilitate urinary concentration. UT-A1 inhibition is predicted to produce a unique salt-sparing diuretic action in edema and hyponatremia. Here we report the discovery of 1,2,4-triazoloquinoxalines and the analysis of 37 synthesized analogues. The most potent compound, 8ay, containing 1,2,4-triazolo[4,3- a]quinoxaline-substituted benzenesulfonamide linked by an aryl ether, rapidly and reversibly inhibited UT-A1 urea transport by a noncompetitive mechanism with IC50 ≈ 150 nM; the IC50 was ∼2 μM for the related urea transporter UT-B encoded by the Slc14a1 gene. Molecular modeling suggested a putative binding site on the UT-A1 cytoplasmic domain. In vitro metabolism showing quinoxaline ring oxidation prompted the synthesis of metabolically stable 7,8-difluoroquinoxaline analogue 8bl, which when administered to rats produced marked diuresis and reduced urinary osmolality. 8bl has substantially improved UT-A1 inhibition potency and metabolic stability compared with prior compounds.
Collapse
Affiliation(s)
- Sujin Lee
- Departments of Medicine and Physiology, University of California, San Francisco, California 94143-0521, United States
| | - Onur Cil
- Departments of Medicine and Physiology, University of California, San Francisco, California 94143-0521, United States
- Department of Pediatrics, Division of Nephrology, University of California, San Francisco, California 94143-0521, United States
| | - Elena Diez-Cecilia
- Department of Chemistry and Biochemistry, San Francisco State University, San Francisco, California 94132-1722, United States
| | - Marc O. Anderson
- Departments of Medicine and Physiology, University of California, San Francisco, California 94143-0521, United States
- Department of Chemistry and Biochemistry, San Francisco State University, San Francisco, California 94132-1722, United States
| | - Alan S. Verkman
- Departments of Medicine and Physiology, University of California, San Francisco, California 94143-0521, United States
| |
Collapse
|
50
|
Zhang ZY, Wang X, Liu D, Zhang H, Zhang Q, Lu YY, Li P, Lou YQ, Yang BX, Lu C, Lou YX, Zhang GL. Development and validation of an LC-MS/MS method for the determination of a novel thienoquinolin urea transporter inhibitor PU-48 in rat plasma and its application to a pharmacokinetic study. Biomed Chromatogr 2018; 32. [PMID: 29193233 DOI: 10.1002/bmc.4157] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 10/05/2017] [Accepted: 11/20/2017] [Indexed: 01/25/2023]
Abstract
A specific, sensitive and stable high-performance liquid chromatographic-tandem mass spectrometry (LC-MS/MS) method was developed and validated for the quantitative determination of methyl 3-amino-6-methoxythieno [2,3-b]quinoline-2-carboxylate (PU-48), a novel diuretic thienoquinolin urea transporter inhibitor in rat plasma. In this method, the chromatographic separation of PU-48 was achieved with a reversed-phase C18 column (100 × 2.1 mm, 3 μm) at 35°C. The mobile phase consisted of acetonitrile and water with 0.05% formic acid added with a gradient elution at flow rate of 0.3 mL/min. Samples were detected with the triple-quadrupole tandem mass spectrometer with multiple reaction monitoring mode via electrospray ionization source in positive mode. The retention time were 6.2 min for PU-48 and 7.2 min for megestrol acetate (internal standard, IS). The monitored ion transitions were mass-to-charge ratio (m/z) 289.1 → 229.2 for PU-48 and m/z 385.3 → 267.1 for the internal standard. The calibration curve for PU-48 was linear over the concentration range of 0.1-1000 ng/mL (r2 > 0.99), and the lower limit of quantitation was 0.1 ng/mL. The precision, accuracy and stability of the method were validated adequately. The developed and validated method was successfully applied to the pharmacokinetic study of PU-48 in rats.
Collapse
Affiliation(s)
- Zhi-Yuan Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xin Wang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Dan Liu
- Proteomics Laboratory, Medical and Health Analysis Center, Peking University, Beijing, China
| | - Hua Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Qiang Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Ying-Yuan Lu
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Pu Li
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Ya-Qing Lou
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Bao-Xue Yang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Chuang Lu
- Department of Drug Metabolism and Pharmacokinetics, Biogen, Cambridge, Massachusetts, USA
| | - Ya-Xin Lou
- Proteomics Laboratory, Medical and Health Analysis Center, Peking University, Beijing, China
| | - Guo-Liang Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| |
Collapse
|