1
|
Martínez González Á, González Nunes M, Rodeiro Escobar P, Llópiz Castedo J, Cabaleiro Loureiro A, Martínez Espinosa RP, Ruades Patiño R, Lorenzo Canda G, Aguayo Arjona J, Rodríguez Zorrilla S. Comparative study of the effectiveness of tolvaptan versus urea in patients with hyponatremia caused by SIADH. Rev Clin Esp 2024:S2254-8874(24)00155-3. [PMID: 39638091 DOI: 10.1016/j.rceng.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 08/09/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND AND OBJECTIVES Hyponatraemia is common in elderly and hospitalised patients, often caused by the syndrome of inappropriate antidiuretic hormone secretion (SIADH). This study evaluates the efficacy and safety of tolvaptan and urea in patients with hyponatraemia and SIADH. MATERIALS AND METHODS An observational cohort study was conducted on 198 patients with SIADH and hyponatraemia (Na+ <135 mmol/L) at the Complejo Hospitalario Universitario de Pontevedra from January 2015 to May 2022. Of these, 86 were treated with tolvaptan (average dose of 7.5 mg) and 112 with urea (average dose of 15 g). The primary outcome was the normalization of sodium levels (Na ≥ 135 mmol/L). RESULTS The tolvaptan group showed higher sodium concentrations at the end of therapy compared to the urea group (ME = 136, IQR = 135-137 vs. ME = 134, IQR = 132-137; p < 0.001). The time to normalise sodium was shorter with tolvaptan (4 ± 3.4 days) compared to urea (6 ± 3.6 days; p = 0.03). A higher percentage of patients achieved sodium normalization with tolvaptan (83.72% vs. 59.82%; p = 0.005). Tolvaptan had more adverse effects, such as dry mouth, thirst, and sodium overcorrection, while urea caused dysgeusia, abdominal pain, and diarrhea. There were no significant differences in mortality between the groups. CONCLUSIONS Tolvaptan was more effective and quicker than urea in normalising sodium levels, though it showed a higher percentage of adverse effects, which did not require discontinuation of the drug.
Collapse
Affiliation(s)
- Á Martínez González
- Servicio de Endocrinología y Nutrición, Hospital Universitario Montecelo, Pontevedra, Spain.
| | - M González Nunes
- Servicio de Endocrinología y Nutrición, Hospital Universitario Montecelo, Pontevedra, Spain
| | - P Rodeiro Escobar
- Servicio de Medicina Familiar y Comunitaria, Área Sanitaria de Vigo, Pontevedra, Spain
| | - J Llópiz Castedo
- Servicio de Medicina Familiar y Comunitaria, Área Sanitaria de Vigo, Pontevedra, Spain
| | - A Cabaleiro Loureiro
- Servicio de Medicina Interna, Hospital Universitario Montecelo, Pontevedra, Spain
| | - R P Martínez Espinosa
- Servicio de Medicina Familiar y Comunitaria, Área Sanitaria de Pontevedra y el Salnés, Pontevedra, Spain
| | - R Ruades Patiño
- Servicio de Endocrinología y Nutrición, Hospital Universitario Montecelo, Pontevedra, Spain
| | - G Lorenzo Canda
- Servicio de Medicina Familiar y Comunitaria, Área Sanitaria de Vigo, Pontevedra, Spain
| | - J Aguayo Arjona
- Unidad de Metodología y Estadística (UME), Instituto de Investigación Sanitaria Galicia Sur (IISGS), Hospital Álvaro Cunqueiro, Vigo, Pontevedra, Spain
| | - S Rodríguez Zorrilla
- Unidad de Medicina Oral, Cirugía Oral e Implantología, Facultad de Medicina y Odontología, Universidad de Santiago de Compostela, Santiago de Compostela, A Coruña, Spain
| |
Collapse
|
2
|
Martínez González Á, González Nunes M, Llópiz Castedo J, Rodeiro Escobar P, Silva Sousa JI, Rodríguez Pulian J, Sieiro Peña C, Rodríguez Castiñeira TC. [Effectiveness of the administration of a dietary supplement (oral urea) for the treatment of hyponatremia in SIADH]. NUTR HOSP 2024. [PMID: 39512013 DOI: 10.20960/nh.05516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND AND OBJECTIVES the most common cause of hyponatraemia in hospital settings is the syndrome of inappropriate antidiuretic hormone secretion (SIADH). This study compares the efficacy and safety of urea versus fluid restriction in the treatment of hyponatraemia caused by SIADH. MATERIAL AND METHODS an observational cohort study was conducted with 212 patients suffering from hyponatremia (Na+ < 135 mmol/L) due to SIADH at the Complejo Hospitalario Universitario de Pontevedra between January 2015 and May 2022. Of these, 112 patients received urea (15 g/day) and 100 were treated with fluid restriction (1 liter/day). The primary objective was to normalize sodium levels (Na ≥ 135 mmol/L). RESULTS urea was significantly more effective than fluid restriction. Sodium levels increased from 126.35 to 133.9 mmol/L with urea, compared to an increase from 126.44 to 130.5 mmol/L with fluid restriction (p < 0.001). Sodium normalization was achieved in an average of 6 days with urea, compared to 8 days with fluid restriction (p = 0.04). At discharge, 59.8 % of patients treated with urea reached normal sodium levels, compared to 42 % in the fluid restriction group (p = 0.007). The 60-day mortality rate was lower in the urea group (16.1 %) compared to the fluid restriction group (32.8 %) (p < 0.007). CONCLUSION urea is more effective than fluid restriction in normalizing sodium levels, with a better safety profile and lower 60-day mortality.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Camila Sieiro Peña
- Servicio de Medicina Familiar y Comunitaria. Área Sanitaria Pontevedra e O Salnés
| | | |
Collapse
|
3
|
Zhang Y, Yang Y, Qi X, Cui P, Kang Y, Liu H, Wei Z, Wang H. SLC14A1 and TGF-β signaling: a feedback loop driving EMT and colorectal cancer metachronous liver metastasis. J Exp Clin Cancer Res 2024; 43:208. [PMID: 39061061 PMCID: PMC11282742 DOI: 10.1186/s13046-024-03114-8] [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: 12/01/2023] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) metachronous liver metastasis is a significant clinical challenge, largely attributable to the late detection and the intricate molecular mechanisms that remain poorly understood. This study aims to elucidate the role of Solute Carrier Family 14 Member 1 (SLC14A1) in the pathogenesis and progression of CRC metachronous liver metastasis. METHODS We conducted a comprehensive analysis of CRC patient data from The Cancer Genome Atlas and GSE40967 databases, focusing on the differential expression of genes associated with non-metachronous liver metastasis and metachronous liver metastasis. Functional assays, both in vitro and in vivo, were performed to assess the biological impact of SLC14A1 modulation in CRC cells. Gene set enrichment analysis, molecular assays and immunohistochemical analyses on clinical specimens were employed to unravel the underlying mechanisms through which SLC14A1 exerts its effects. RESULTS SLC14A1 was identified as a differentially expressed gene, with its overexpression significantly correlating with poor relapse-free and overall survival. Mechanistically, elevated SLC14A1 levels enhanced CRC cell invasiveness and migratory abilities, corroborated by upregulated TGF-β/Smad signaling and Epithelial-Mesenchymal Transition. SLC14A1 interacted with TβRII and stabilized TβRII protein, impeding its Smurf1-mediated K48-linked ubiquitination and degradation, amplifying TGF-β/Smad signaling. Furthermore, TGF-β1 reciprocally elevated SLC14A1 mRNA expression, with Snail identified as a transcriptional regulator, binding downstream of SLC14A1's transcription start site, establishing a positive feedback loop. Clinically, SLC14A1, phosphorylated Smad2, and Snail were markedly upregulated in CRC patients with metachronous liver metastasis, underscoring their potential as prognostic markers. CONCLUSIONS Our findings unveil SLC14A1 as a critical regulator in CRC metachronous liver metastasis, providing novel insights into the molecular crosstalk between SLC14A1 and TGF-β/Smad signaling. These discoveries not only enhance our understanding of CRC metachronous liver metastasis pathogenesis, but also highlight SLC14A1 as a promising target for therapeutic intervention and predictive marker.
Collapse
Affiliation(s)
- Yixun Zhang
- Department of Colorectal Surgery, Cancer Hospital Affiliated to Shanxi Medical University, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, China
| | - Yumeng Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing, 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing, China
| | - Xuan Qi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing, 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing, China
| | - Peng Cui
- Department of General Surgery, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yi Kang
- Department of Gastroenterology, Cancer Hospital Affiliated to Shanxi Medical University, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, China
| | - Haiyi Liu
- Department of Colorectal Surgery, Cancer Hospital Affiliated to Shanxi Medical University, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, China
| | - Zhigang Wei
- Hepatobiliary and Pancreatic Surgery and Liver Transplantation Center, First Hospital of Shanxi Medical University, 85 Jiefang Nan Lu, Taiyuan, 030001, Shanxi, China.
| | - Haibo Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing, 100069, China.
- Laboratory for Clinical Medicine, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing, China.
- Beijing Laboratory of Oral Health, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing, China.
| |
Collapse
|
4
|
Dmitrieva NI, Boehm M, Yancey PH, Enhörning S. Long-term health outcomes associated with hydration status. Nat Rev Nephrol 2024; 20:275-294. [PMID: 38409366 DOI: 10.1038/s41581-024-00817-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2024] [Indexed: 02/28/2024]
Abstract
Body water balance is determined by fundamental homeostatic mechanisms that maintain stable volume, osmolality and the composition of extracellular and intracellular fluids. Water balance is maintained by multiple mechanisms that continuously match water losses through urine, the skin, the gastrointestinal tract and respiration with water gains achieved through drinking, eating and metabolic water production. Hydration status is determined by the state of the water balance. Underhydration occurs when a decrease in body water availability, due to high losses or low gains, stimulates adaptive responses within the water balance network that are aimed at decreasing losses and increasing gains. This stimulation is also accompanied by cardiovascular adjustments. Epidemiological and experimental studies have linked markers of low fluid intake and underhydration - such as increased plasma concentration of vasopressin and sodium, as well as elevated urine osmolality - with an increased risk of new-onset chronic diseases, accelerated aging and premature mortality, suggesting that persistent activation of adaptive responses may be detrimental to long-term health outcomes. The causative nature of these associations is currently being tested in interventional trials. Understanding of the physiological responses to underhydration may help to identify possible mechanisms that underlie potential adverse, long-term effects of underhydration and inform future research to develop preventative and treatment approaches to the optimization of hydration status.
Collapse
Affiliation(s)
- Natalia I Dmitrieva
- Laboratory of Cardiovascular Regenerative Medicine, National Heart Lung and Blood Institute, NIH, Bethesda, Maryland, USA.
| | - Manfred Boehm
- Laboratory of Cardiovascular Regenerative Medicine, National Heart Lung and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Paul H Yancey
- Biology Department, Whitman College, Walla Walla, Washington, USA
| | - Sofia Enhörning
- Perinatal and Cardiovascular Epidemiology, Lund University Diabetes Centre, Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
- Department of Internal Medicine, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
5
|
Prahl MC, Müller CBM, Wimmers K, Kuhla B. Mammary gland, kidney and rumen urea and uric acid transporters of dairy cows differing in milk urea concentration. Sci Rep 2023; 13:17231. [PMID: 37821556 PMCID: PMC10567808 DOI: 10.1038/s41598-023-44416-9] [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: 05/09/2023] [Accepted: 10/08/2023] [Indexed: 10/13/2023] Open
Abstract
The milk urea concentration (MUC) serves as indicator of urinary nitrogen emissions, but at comparable crude protein (CP) intake, cows with high (HMU) and low (LMU) MUC excrete equal urea amounts. We hypothesized that urea and uric acid transporters and sizes of the kidney, mammary gland, and rumen account for these phenotypes. Eighteen HMU and 18 LMU Holstein dairy cows fed a low (LP) and normal (NP) CP diet were studied. Milk, plasma and urinary urea concentrations were greater with NP feeding, while plasma and urinary urea concentrations were comparable between phenotypes. Milk and plasma uric acid concentrations were higher with LP feeding but not affected by phenotype. The milk-urine uric acid ratio was greater in HMU cows. The mRNA expressions of the ruminal urea transporter SLC14A1 and AQP10, the mammary gland and rumen AQP3, and the mammary gland uric acid transporter ABCG2 were not affected by group or diet. Renal AQP10, but not AQP3, AQP7, and SLC14A2 expressions, and the kidney weights were lower in HMU cows. These data indicate that renal size and AQP10 limit the urea transfer from blood to urine, and that MUC determines if uric acid is more released with milk or urine.
Collapse
Affiliation(s)
- Marie C Prahl
- Research Institute for Farm Animal Biology (FBN), Institute of Nutritional Physiology 'Oskar Kellner', Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Carolin B M Müller
- Research Institute for Farm Animal Biology (FBN), Institute of Nutritional Physiology 'Oskar Kellner', Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Klaus Wimmers
- Research Institute for Farm Animal Biology (FBN), Institute of Genome Biology, Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Björn Kuhla
- Research Institute for Farm Animal Biology (FBN), Institute of Nutritional Physiology 'Oskar Kellner', Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany.
| |
Collapse
|
6
|
Hailemariam S, Zhao S, He Y, Wang J. Urea transport and hydrolysis in the rumen: A review. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2021; 7:989-996. [PMID: 34738029 PMCID: PMC8529027 DOI: 10.1016/j.aninu.2021.07.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 07/02/2021] [Accepted: 07/08/2021] [Indexed: 11/29/2022]
Abstract
Inefficient dietary nitrogen (N) conversion to microbial proteins, and the subsequent use by ruminants, is a major research focus across different fields. Excess bacterial ammonia (NH3) produced due to degradation or hydrolyses of N containing compounds, such as urea, leads to an inefficiency in a host's ability to utilize nitrogen. Urea is a non-protein N containing compound used by ruminants as an ammonia source, obtained from feed and endogenous sources. It is hydrolyzed by ureases from rumen bacteria to produce NH3 which is used for microbial protein synthesis. However, lack of information exists regarding urea hydrolysis in ruminal bacteria, and how urea gets to hydrolysis sites. Therefore, this review describes research on sites of urea hydrolysis, urea transport routes towards these sites, the role and structure of urea transporters in rumen epithelium and bacteria, the composition of ruminal ureolytic bacteria, mechanisms behind urea hydrolysis by bacterial ureases, and factors influencing urea hydrolysis. This review explores the current knowledge on the structure and physiological role of urea transport and ureolytic bacteria, for the regulation of urea hydrolysis and recycling in ruminants. Lastly, underlying mechanisms of urea transportation in rumen bacteria and their physiological importance are currently unknown, and therefore future research should be directed to this subject.
Collapse
Affiliation(s)
- Samson Hailemariam
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
- Dilla University, College of Agriculture and Natural Resource, Dilla P. O. Box 419, Ethiopia
| | - Shengguo Zhao
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Yue He
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Jiaqi Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| |
Collapse
|
7
|
Nandi S, Sanyal S, Amin SA, Kashaw SK, Jha T, Gayen S. Urea transporter and its specific and nonspecific inhibitors: State of the art and pharmacological perspective. Eur J Pharmacol 2021; 911:174508. [PMID: 34536365 DOI: 10.1016/j.ejphar.2021.174508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 09/10/2021] [Accepted: 09/14/2021] [Indexed: 01/11/2023]
Abstract
Hypertension is a major concern for a wide array of patients. The traditional drugs are commonly referred as 'water pills' and these molecules have been successful in alleviating hypertension. However, this comes at the high expense of precious electrolytes in our body. To dissipate this major adverse effect, the urea transporter inhibitors play especially important roles in maintaining the fluid balance by maintaining the concentration of urea in the inner medullary collecting duct. The purpose of this communication is to provide insights into the structural feature of these target proteins and inhibition of both urea transporter types A (UT-A) and B (UT-B) selectively and non-selectively with a special focus on the UT-A inhibitors as they are the primary target for diuresis. It was observed that a wide class of drugs such as thiourea analogues, 2,7-disubstituted fluorenones can inhibit both the protein non-selectively whereas 8-hydroxyquinoline, aminothiazolone, 1,3,5-triazine, triazolothienopyrimidine, thienoquinoline, arylthiazole, γ-sultambenzosulfonamide and 1,2,4-triazoloquinoxaline classes of compounds inhibit UT-A. The goal of this study is to highlight the important aspects that may be useful to understanding the perspectives of urea transporter inhibitors in rational drug discovery.
Collapse
Affiliation(s)
- Sudipta Nandi
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University, Sagar, MP, India
| | - Saptarshi Sanyal
- School of Pharmaceutical Technology, Adamas University, Kolkata, India; Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Sk Abdul Amin
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Sushil Kumar Kashaw
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University, Sagar, MP, India
| | - Tarun Jha
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India.
| | - Shovanlal Gayen
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University, Sagar, MP, India; Laboratory of Drug Design and Discovery, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India.
| |
Collapse
|
8
|
Nielsen MMK, Aryal E, Safari E, Mojsoska B, Jenssen H, Prabhala BK. Current State of SLC and ABC Transporters in the Skin and Their Relation to Sweat Metabolites and Skin Diseases. Proteomes 2021; 9:proteomes9020023. [PMID: 34065737 PMCID: PMC8163169 DOI: 10.3390/proteomes9020023] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 01/02/2023] Open
Abstract
With a relatively large surface area (2 m2) and 15% of total body mass, the skin forms the largest organ of the human body. The main functions of the skin include regulation of body temperature by insulation or sweating, regulation of the nervous system, regulation of water content, and protection against external injury. To perform these critical functions, the skin encodes genes for transporters responsible for the cellular trafficking of essential nutrients and metabolites to maintain cellular hemostasis. However, the knowledge on the expression, regulation, and function of these transporters is very limited and needs more work to elucidate how these transporters play a role both in disease progression and in healing. Furthermore, SLC and ABC transporters are understudied, and even less studied in skin. There are sparse reports on relation between transporters in skin and sweat metabolites. This mini review focuses on the current state of SLC and ABC transporters in the skin and their relation to sweat metabolites and skin diseases.
Collapse
Affiliation(s)
- Marcus M. K. Nielsen
- Institute of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark; (M.M.K.N.); (E.A.)
| | - Eva Aryal
- Institute of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark; (M.M.K.N.); (E.A.)
| | - Elnaz Safari
- Department of Organic Chemistry, Faculty of Chemistry, Urmia University, Urmia 5756151818, Iran;
| | - Biljana Mojsoska
- Institute of Science and Environment, Roskilde University, Universitetsvej 1, 4000 Roskilde, Denmark; (B.M.); (H.J.)
| | - Håvard Jenssen
- Institute of Science and Environment, Roskilde University, Universitetsvej 1, 4000 Roskilde, Denmark; (B.M.); (H.J.)
| | - Bala Krishna Prabhala
- Institute of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark; (M.M.K.N.); (E.A.)
- Correspondence:
| |
Collapse
|
9
|
Patil VS, Yadavalli SR, Merugu R, S J S, Devunuri N. One-pot, two-step synthesis of substituted triazoloquinoxalinone starting from 3-hydrazineylquinoxalin-2(1H)-one. SYNTHETIC COMMUN 2021. [DOI: 10.1080/00397911.2021.1918171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Vikas S Patil
- Division of Chemistry, Department of Sciences & Humanities, Vignan’s Foundation for Science, Technology & Research (VFSTR) Deemed University, Guntur, India
| | - Subba Rao Yadavalli
- Division of Chemistry, Department of Sciences & Humanities, Vignan’s Foundation for Science, Technology & Research (VFSTR) Deemed University, Guntur, India
| | - Ramchander Merugu
- Department of Bio Chemistry, Mahatma Gandhi University, Nalgonda, India
| | - Swamy S J
- Department of Chemistry, Kakatiya University, Warangal, India
| | - Nagaraju Devunuri
- Division of Chemistry, Department of Sciences & Humanities, Vignan’s Foundation for Science, Technology & Research (VFSTR) Deemed University, Guntur, India
| |
Collapse
|
10
|
Zhao J, Feng WG, Wei Z, Zhou J, Chen XY, Zhang ZL. Follow-Up of Adefovir Dipivoxil Induced Osteomalacia: Clinical Characteristics and Genetic Predictors. Front Pharmacol 2021; 12:636352. [PMID: 33995038 PMCID: PMC8113870 DOI: 10.3389/fphar.2021.636352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 04/16/2021] [Indexed: 11/13/2022] Open
Abstract
Adefovir dipivoxil (ADV) is widely used for chronic hepatitis B therapy in China. To explore the clinical features and prognosis of ADV-induced osteomalacia and to analyze the association between osteomalacia and genetic variants in 51 drug transporters genes. Clinical and follow-up data of the ADV-treated patients were collected. Target capture sequencing was used to identify genetic variations of 51 drug transporter genes. A total of 193 hepatitis B patients treated with ADV were enrolled, of whom 140 had osteomalacia. The other 53 without osteomalacia were included in the control group. The median duration of ADV treatment before the onset of osteomalacia was 6.5 years (range:1.5–7 years). We found that most patients with osteomalacia had hypophosphatemia, high serum alkaline phosphatase levels, hypouricemia, nondiabetic glycosuria, proteinuria. Stopping ADV administration, supplementing calcitriol and calcium were effective treatments. During 3–6 months of follow-up, the clinical symptoms and biochemical indicators of patients with osteomalacia have been significantly improved. There was no significant difference in duration of adefovir treatment in patients with or without osteomalacia (p = 0.791). Through regression analysis, we found that age was a risk factor for osteomalacia [per 1 year, odds ratio (OR), 1.053; 95% confidence interval (95% CI), 1.020–1.087; p = 0.015]. 1992 single nucleotide variants were found using target capture sequencing. However, the associations of genetic variants of 51 drug transporter genes and the risk of osteomalacia were negligible. Osteomalacia is prone to occur in patients with chronic hepatitis B treated with long-term ADV at a therapeutic dose. After standard treatment, the prognosis is mostly good. We failed to find genetic variants that can predict the risk of ADV-induced osteomalacia.
Collapse
Affiliation(s)
- Jiao Zhao
- Shanghai Clinical Research Center of Bone Disease, Department of Osteoporosis and Bone Disease, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Wei-Guang Feng
- Department of Hepatology, The Fourth People's Hospital of Huai'an, Huai'an, China
| | - Zhe Wei
- Shanghai Clinical Research Center of Bone Disease, Department of Osteoporosis and Bone Disease, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jian Zhou
- Department of Hepatology, The Fourth People's Hospital of Huai'an, Huai'an, China
| | - Xiao-Yun Chen
- Department of Rheumatology, Shanghai University of Traditional Chinese Medicine Affiliated LongHua Hospital, Shanghai, China
| | - Zhen-Lin Zhang
- Shanghai Clinical Research Center of Bone Disease, Department of Osteoporosis and Bone Disease, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
11
|
Kell DB. A protet-based, protonic charge transfer model of energy coupling in oxidative and photosynthetic phosphorylation. Adv Microb Physiol 2021; 78:1-177. [PMID: 34147184 DOI: 10.1016/bs.ampbs.2021.01.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Textbooks of biochemistry will explain that the otherwise endergonic reactions of ATP synthesis can be driven by the exergonic reactions of respiratory electron transport, and that these two half-reactions are catalyzed by protein complexes embedded in the same, closed membrane. These views are correct. The textbooks also state that, according to the chemiosmotic coupling hypothesis, a (or the) kinetically and thermodynamically competent intermediate linking the two half-reactions is the electrochemical difference of protons that is in equilibrium with that between the two bulk phases that the coupling membrane serves to separate. This gradient consists of a membrane potential term Δψ and a pH gradient term ΔpH, and is known colloquially as the protonmotive force or pmf. Artificial imposition of a pmf can drive phosphorylation, but only if the pmf exceeds some 150-170mV; to achieve in vivo rates the imposed pmf must reach 200mV. The key question then is 'does the pmf generated by electron transport exceed 200mV, or even 170mV?' The possibly surprising answer, from a great many kinds of experiment and sources of evidence, including direct measurements with microelectrodes, indicates it that it does not. Observable pH changes driven by electron transport are real, and they control various processes; however, compensating ion movements restrict the Δψ component to low values. A protet-based model, that I outline here, can account for all the necessary observations, including all of those inconsistent with chemiosmotic coupling, and provides for a variety of testable hypotheses by which it might be refined.
Collapse
Affiliation(s)
- Douglas B Kell
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative, Biology, University of Liverpool, Liverpool, United Kingdom; The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark.
| |
Collapse
|
12
|
Narasimhan B, Aravinthkumar R, Correa A, Aronow WS. Pharmacotherapeutic principles of fluid management in heart failure. Expert Opin Pharmacother 2021; 22:595-610. [PMID: 33560159 DOI: 10.1080/14656566.2020.1850694] [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: 10/22/2022]
Abstract
Introduction: Heart failure is a major public health concern that is expected to increase over the decades to come. Despite significant advances, fluid overload and congestion remain a major therapeutic challenge. Vascular congestion and neurohormonal activation are intricately linked and the goal of therapy fundamentally aims to reduce both.Areas covered: The authors briefly review a number of core concepts that elucidate the link between fluid overload and neuro-hormonal activation. This is followed by a review of heart-kidney interactions and the impact of diuresis in this setting. Following an in-depth review of currently available pharmacological agents, the rationale and evidence behind their use, the authors end with a brief note on novel agents/approaches to aid volume management in HF.Expert opinion: A number of non-pharmacological advances in the management of volume overload in heart failure, though promising - are associated with a number of shortcomings. Pharmacological therapy remains the cornerstone of volume management. A number of novel approaches, utilizing existing therapies as well as the emergence of new agents over the past decade bode well for the vulnerable HF population.
Collapse
Affiliation(s)
- Bharat Narasimhan
- Department of Medicine, Mount Sinai Morningside, Mount Sinai West, New York, NY
| | | | - Ashish Correa
- Department of Cardiology, Mount Sinai Morningside, Mount Sinai West, Icahn School of Medicine at Mount Sinai
| | - Wilbert S Aronow
- Department of Cardiology, Westchester Medical center/New York Medical College, Valhalla, NY
| |
Collapse
|
13
|
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
|
14
|
Rianto F, Kuma A, Ellis CL, Hassounah F, Rodriguez EL, Wang XH, Sands JM, Klein JD. UT-A1/A3 knockout mice show reduced fibrosis following unilateral ureteral obstruction. Am J Physiol Renal Physiol 2020; 318:F1160-F1166. [PMID: 32174141 PMCID: PMC7294340 DOI: 10.1152/ajprenal.00008.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/24/2020] [Accepted: 03/12/2020] [Indexed: 12/23/2022] Open
Abstract
Renal fibrosis is a major contributor to the development and progression of chronic kidney disease. A low-protein diet can reduce the progression of chronic kidney disease and reduce the development of renal fibrosis, although the mechanism is not well understood. Urea reabsorption into the inner medulla is regulated by inner medullary urea transporter (UT)-A1 and UT-A3. Inhibition or knockout of UT-A1/A3 will reduce interstitial urea accumulation, which may be beneficial in reducing renal fibrosis. To test this hypothesis, the effect of unilateral ureteral obstruction (UUO) was compared in wild-type (WT) and UT-A1/A3 knockout mice. UUO causes increased extracellular matrix associated with increases in transforming growth factor-β, vimentin, and α-smooth muscle actin (α-SMA). In WT mice, UUO increased the abundance of three markers of fibrosis: transforming growth factor-β, vimentin, and α-SMA. In contrast, in UT-A1/A3 knockout mice, the increase following UUO was significantly reduced. Consistent with the Western blot results, immunohistochemical staining showed that the levels of vimentin and α-SMA were increased in WT mice with UUO and that the increase was reduced in UT-A1/A3 knockout mice with UUO. Masson's trichrome staining showed increased collagen in WT mice with UUO, which was reduced in UT-A1/A3 knockout mice with UUO. We conclude that reduced UT activity reduces the severity of renal fibrosis following UUO.
Collapse
Affiliation(s)
- Fitra Rianto
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Akihiro Kuma
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
- Second Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Carla L Ellis
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia
| | - Faten Hassounah
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Eva L Rodriguez
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Xiaonan H Wang
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Jeff M Sands
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Janet D Klein
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
15
|
Yu L, Liu T, Fu S, Li L, Meng X, Su X, Xie Z, Ren J, Meng Y, Lv X, Du Y. Physiological functions of urea transporter B. Pflugers Arch 2019; 471:1359-1368. [PMID: 31734718 PMCID: PMC6882768 DOI: 10.1007/s00424-019-02323-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 10/16/2019] [Accepted: 10/17/2019] [Indexed: 11/04/2022]
Abstract
Urea transporters (UTs) are membrane proteins in the urea transporter protein A (UT-A) and urea transporter protein B (UT-B) families. UT-B is mainly expressed in endothelial cell membrane of the renal medulla and in other tissues, including the brain, heart, pancreas, colon, bladder, bone marrow, and cochlea. UT-B is responsible for the maintenance of urea concentration, male reproductive function, blood pressure, bone metabolism, and brain astrocyte and cardiac functions. Its deficiency and dysfunction contribute to the pathogenesis of many diseases. Actually, UT-B deficiency increases the sensitivity of bladder epithelial cells to apoptosis triggers in mice and UT-B-null mice develop II-III atrioventricular block and depression. The expression of UT-B in the rumen of cow and sheep may participate in digestive function. However, there is no systemic review to discuss the UT-B functions. Here, we update research approaches to understanding the functions of UT-B.
Collapse
Affiliation(s)
- Lanying Yu
- Changchun University of Chinese Medicine, Changchun, 130117, People's Republic of China
| | - Tiantian Liu
- Changchun University of Chinese Medicine, Changchun, 130117, People's Republic of China
| | - Shuang Fu
- Changchun University of Chinese Medicine, Changchun, 130117, People's Republic of China
| | - Li Li
- Changchun University of Chinese Medicine, Changchun, 130117, People's Republic of China
| | - Xiaoping Meng
- Changchun University of Chinese Medicine, Changchun, 130117, People's Republic of China
| | - Xin Su
- Changchun University of Chinese Medicine, Changchun, 130117, People's Republic of China
| | - Zhanfeng Xie
- Changchun University of Chinese Medicine, Changchun, 130117, People's Republic of China
| | - Jiayan Ren
- Changchun University of Chinese Medicine, Changchun, 130117, People's Republic of China
| | - Yan Meng
- Department of Pathophysiology, College of Basic Medicine, Jilin University, Changchun, 130021, Jilin, People's Republic of China.
| | - Xuejiao Lv
- Department of Respiratory Medicine, the Second Affiliated Hospital of Jilin University, Changchun, 130041, Jilin, People's Republic of China.
| | - Yanwei Du
- Changchun University of Chinese Medicine, Changchun, 130117, People's Republic of China.
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
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: 15] [Impact Index Per Article: 2.1] [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
|
19
|
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
|
20
|
Xie L, Jin L, Feng J, Lv J. The Expression of AQP5 and UTs in the Sweat Glands of Uremic Patients. BIOMED RESEARCH INTERNATIONAL 2017; 2017:8629783. [PMID: 29279852 PMCID: PMC5723962 DOI: 10.1155/2017/8629783] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 10/17/2017] [Accepted: 11/09/2017] [Indexed: 02/04/2023]
Abstract
PURPOSE To research the distribution and quantitative changes of UT-A1, UT-B1, and AQP5 in uremic skin tissue. METHODS 34 cases of uremic patients (UP) and 11 controls were recruited. Immunohistochemistry, immunofluorescence, RT-PCR, and Western Blot were used to identify the proteins in sweat glands. RESULTS AQP5, UT-A1, and UT-B1 were expressed and localized in human skin basal lines, skin sweat glands, and sweat ducts, both in UP and controls. Compared to controls, AQP5 mRNA abundance was significantly decreased in UP (P < 0.01), and, with the decrease of eGFR, the AQP5 expression was significantly decreased (P < 0.05). By contrast, UT-A1 and UT-B1 mRNA abundance was significantly increased in the skin of UP compared with the control (P < 0.01), and, with the decrease of eGFR, the AQP5 expression was significantly increased (P < 0.05). We found that the gene changes were coincident with the corresponding target proteins. The urea transporter subtypes, UT-A1 and UT-B1, were expressed in the skin basal cell layer and exocrine sweat glands. The abundance of UT-A1 and UT-B1 in uremic sweat glands was significantly increased in UP, while the expression of AQP5 was decreased. CONCLUSION Elimination of urea through the skin by producing sweat is a potential therapeutic strategy for renal failure patients.
Collapse
Affiliation(s)
- Liyi Xie
- Department of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Li Jin
- Department of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jie Feng
- Department of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jing Lv
- Department of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| |
Collapse
|
21
|
Jones R, Watson K, Bruce A, Nersesian S, Kitz J, Moorehead R. Re-expression of miR-200c suppresses proliferation, colony formation and in vivo tumor growth of murine claudin-low mammary tumor cells. Oncotarget 2017; 8:23727-23749. [PMID: 28423599 PMCID: PMC5410340 DOI: 10.18632/oncotarget.15829] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 02/06/2017] [Indexed: 12/17/2022] Open
Abstract
Claudin-low breast cancer is a relatively rare breast cancer subtype. These cancers are typically ER-/PR-/HER2- and express high levels of mesenchymal genes as well as genes associated with inflammation, angiogenesis and stem cell function. In addition to alterations in gene expression, it was recently demonstrated that claudin-low breast cancers express very low levels of the miR-200 family of miRNAs. Given that each miRNA can regulate tens, hundreds or even thousands of genes, miRNAs are being evaluated as therapeutic targets. In this study we show that mammary tumors from MTB-IGFIR transgenic mice and cell lines derived from these tumors represent a model of human claudin-low breast cancer and murine claudin-low mammary tumors and cell lines express only very low levels of all five members of the miR-200 family. Reduced miR-200 family expression appears to be regulated via methylation as cells and tumors expressing low levels of miR-200 family members had higher levels of CpG methylation in a putative promoter region than tumors and cells expressing high levels of miR-200 family members. Re-expression of miR-200c in murine claudin-low mammary tumor cells inhibited tumor cell proliferation and colony formation in vitro and tumor growth in vivo. With respect to tumor growth in vivo, re-expression of miR-200c was associated with a reduction in tumor vasculature and expression of Flt1 and Vegfc. Therefore, miR-200c is an important regulator of mesenchymal tumor cell growth.
Collapse
Affiliation(s)
- Robert Jones
- Department of Biomedical Science, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Katrina Watson
- Department of Biomedical Science, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Anthony Bruce
- Department of Biomedical Science, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Sarah Nersesian
- Department of Biomedical Science, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Jenna Kitz
- Department of Biomedical Science, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Roger Moorehead
- Department of Biomedical Science, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|