1
|
Lv L, Liu Y, Xiong J, Wang S, Li Y, Zhang B, Huang Y, Zhao J. Role of G protein coupled receptors in acute kidney injury. Cell Commun Signal 2024; 22:423. [PMID: 39223553 PMCID: PMC11367933 DOI: 10.1186/s12964-024-01802-8] [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: 06/11/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
Acute kidney injury (AKI) is a clinical condition characterized by a rapid decline in kidney function, which is associated with local inflammation and programmed cell death in the kidney. The G protein-coupled receptors (GPCRs) represent the largest family of signaling transduction proteins in the body, and approximately 40% of drugs on the market target GPCRs. The expressions of various GPCRs, prostaglandin receptors and purinergic receptors, to name a few, are significantly altered in AKI models. And the role of GPCRs in AKI is catching the eyes of researchers due to their distinctive biological functions, such as regulation of hemodynamics, metabolic reprogramming, and inflammation. Therefore, in this review, we aim to discuss the role of GPCRs in the pathogenesis of AKI and summarize the relevant clinical trials involving GPCRs to assess the potential of GPCRs and their ligands as therapeutic targets in AKI and the transition to AKI-CKD.
Collapse
Affiliation(s)
- Liangjing Lv
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University), Chongqing, 400037, China
| | - Yong Liu
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University), Chongqing, 400037, China
| | - Jiachuan Xiong
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University), Chongqing, 400037, China
| | - Shaobo Wang
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University), Chongqing, 400037, China
| | - Yan Li
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University), Chongqing, 400037, China
| | - Bo Zhang
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University), Chongqing, 400037, China
| | - Yinghui Huang
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University), Chongqing, 400037, China
| | - Jinghong Zhao
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University), Chongqing, 400037, China.
| |
Collapse
|
2
|
Esteva-Font C, Geurts F, Hansen TPK, Hoorn EJ, Fenton RA. Inducible deletion of the prostaglandin EP3 receptor in kidney tubules of male and female mice has no major effect on water homeostasis. Am J Physiol Renal Physiol 2024; 327:F504-F518. [PMID: 38961846 DOI: 10.1152/ajprenal.00146.2024] [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: 05/14/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 07/05/2024] Open
Abstract
The prostaglandin E2 (PGE2) receptor EP3 has been detected in the thick ascending limb (TAL) and the collecting duct of the kidney, where its actions are proposed to inhibit water reabsorption. However, EP3 is also expressed in other cell types, including vascular endothelial cells. The aim here was to determine the contribution of EP3 in renal water handling in male and female adult mice by phenotyping a novel mouse model with doxycycline-dependent deletion of EP3 throughout the kidney tubule (EP3-/- mice). RNAscope demonstrated that EP3 was highly expressed in the cortical and medullary TAL of adult mice. Compared with controls EP3 mRNA expression was reduced by >80% in whole kidney (RT-qPCR) and nondetectable (RNAscope) in renal tubules of EP3-/- mice. Under basal conditions, there were no significant differences in control and EP3-/- mice of both sexes in food and water intake, body weight, urinary output, or clinical biochemistries. No differences were detectable between genotypes in handling of an acute water load or in their response to the vasopressin analog 1-deamino-8-d-arginine-vasopressin (dDAVP). No differences in water handling were observed when PGE2 production was enhanced using 1% NaCl load. Expression of proteins involved in kidney water handling was not different between genotypes. This study demonstrates that renal tubular EP3 is not essential for body fluid homeostasis in males or females, even when PGE2 levels are high. The mouse model is a novel tool for examining the role of EP3 in kidney function independently of potential developmental abnormalities or systemic effects.NEW & NOTEWORTHY The prostanoid EP3 receptor is proposed to play a key role in the kidney tubule and antagonize the effects of vasopressin on aquaporin-mediated water reabsorption. Here, we phenotyped a kidney tubule-specific inducible knockout mouse model of the EP3 receptor. Our major finding is that, even under physiological stress, tubular EP3 plays no detectable role in renal water or solute handling. This suggests that other EP receptors must be important for renal salt and water handling.
Collapse
Affiliation(s)
| | - Frank Geurts
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Toke P K Hansen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Ewout J Hoorn
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Robert A Fenton
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
3
|
Delrue C, Speeckaert R, Moresco RN, Speeckaert MM. Cyclic Adenosine Monophosphate Signaling in Chronic Kidney Disease: Molecular Targets and Therapeutic Potentials. Int J Mol Sci 2024; 25:9441. [PMID: 39273390 PMCID: PMC11395066 DOI: 10.3390/ijms25179441] [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: 07/31/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
Chronic kidney disease (CKD) is characterized by a steady decline in kidney function and affects roughly 10% of the world's population. This review focuses on the critical function of cyclic adenosine monophosphate (cAMP) signaling in CKD, specifically how it influences both protective and pathogenic processes in the kidney. cAMP, a critical secondary messenger, controls a variety of cellular functions, including transcription, metabolism, mitochondrial homeostasis, cell proliferation, and apoptosis. Its compartmentalization inside cellular microdomains ensures accurate signaling. In kidney physiology, cAMP is required for hormone-regulated activities, particularly in the collecting duct, where it promotes water reabsorption through vasopressin signaling. Several illnesses, including Fabry disease, renal cell carcinoma, nephrogenic diabetes insipidus, Bartter syndrome, Liddle syndrome, diabetic nephropathy, autosomal dominant polycystic kidney disease, and renal tubular acidosis, have been linked to dysfunction in the cAMP system. Both cAMP analogs and phosphodiesterase inhibitors have the potential to improve kidney function and reduce kidney damage. Future research should focus on developing targeted PDE inhibitors for the treatment of CKD.
Collapse
Affiliation(s)
- Charlotte Delrue
- Department of Nephrology, Ghent University Hospital, 9000 Ghent, Belgium
| | | | - Rafael Noal Moresco
- Graduate Program in Pharmaceutical Sciences, Center of Health Sciences, Federal University of Santa Maria, Santa Maria 97105-900, Brazil
| | - Marijn M Speeckaert
- Department of Nephrology, Ghent University Hospital, 9000 Ghent, Belgium
- Research Foundation-Flanders (FWO), 1000 Brussels, Belgium
| |
Collapse
|
4
|
Li XJ, Suo P, Wang YN, Zou L, Nie XL, Zhao YY, Miao H. Arachidonic acid metabolism as a therapeutic target in AKI-to-CKD transition. Front Pharmacol 2024; 15:1365802. [PMID: 38523633 PMCID: PMC10957658 DOI: 10.3389/fphar.2024.1365802] [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: 01/05/2024] [Accepted: 02/06/2024] [Indexed: 03/26/2024] Open
Abstract
Arachidonic acid (AA) is a main component of cell membrane lipids. AA is mainly metabolized by three enzymes: cyclooxygenase (COX), lipoxygenase (LOX) and cytochrome P450 (CYP450). Esterified AA is hydrolysed by phospholipase A2 into a free form that is further metabolized by COX, LOX and CYP450 to a wide range of bioactive mediators, including prostaglandins, lipoxins, thromboxanes, leukotrienes, hydroxyeicosatetraenoic acids and epoxyeicosatrienoic acids. Increased mitochondrial oxidative stress is considered to be a central mechanism in the pathophysiology of the kidney. Along with increased oxidative stress, apoptosis, inflammation and tissue fibrosis drive the progressive loss of kidney function, affecting the glomerular filtration barrier and the tubulointerstitium. Recent studies have shown that AA and its active derivative eicosanoids play important roles in the regulation of physiological kidney function and the pathogenesis of kidney disease. These factors are potentially novel biomarkers, especially in the context of their involvement in inflammatory processes and oxidative stress. In this review, we introduce the three main metabolic pathways of AA and discuss the molecular mechanisms by which these pathways affect the progression of acute kidney injury (AKI), diabetic nephropathy (DN) and renal cell carcinoma (RCC). This review may provide new therapeutic targets for the identification of AKI to CKD continuum.
Collapse
Affiliation(s)
- Xiao-Jun Li
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Department of Nephrology, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Ping Suo
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yan-Ni Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Liang Zou
- School of Food and Bioengineering, Chengdu University, Chengdu, Sichuan, China
| | - Xiao-Li Nie
- Department of Nephrology, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Ying-Yong Zhao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Hua Miao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
5
|
Guo Y, Luo T, Xie G, Zhang X. Bile acid receptors and renal regulation of water homeostasis. Front Physiol 2023; 14:1322288. [PMID: 38033333 PMCID: PMC10684672 DOI: 10.3389/fphys.2023.1322288] [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: 10/16/2023] [Accepted: 11/07/2023] [Indexed: 12/02/2023] Open
Abstract
The kidney is the key organ responsible for maintaining the body's water and electrolyte homeostasis. About 99% of the primary urine filtered from the Bowman's capsule is reabsorbed along various renal tubules every day, with only 1-2 L of urine excreted. Aquaporins (AQPs) play a vital role in water reabsorption in the kidney. Currently, a variety of molecules are found to be involved in the process of urine concentration by regulating the expression or activity of AQPs, such as antidiuretic hormone, renin-angiotensin-aldosterone system (RAAS), prostaglandin, and several nuclear receptors. As the main bile acid receptors, farnesoid X receptor (FXR) and membrane G protein-coupled bile acid receptor 1 (TGR5) play important roles in bile acid, glucose, lipid, and energy metabolism. In the kidney, FXR and TGR5 exhibit broad expression across all segments of renal tubules, and their activation holds significant therapeutic potential for numerous acute and chronic kidney diseases through alleviating renal lipid accumulation, inflammation, oxidative stress, and fibrosis. Emerging evidence has demonstrated that the genetic deletion of FXR or TGR5 exhibits increased basal urine output, suggesting that bile acid receptors play a critical role in urine concentration. Here, we briefly summarize the function of bile acid receptors in renal water reabsorption and urine concentration.
Collapse
Affiliation(s)
- Yanlin Guo
- Division of Nephrology, Wuhu Hospital, East China Normal University, Wuhu, China
- Health Science Center, East China Normal University, Shanghai, China
| | - Taotao Luo
- Division of Nephrology, Wuhu Hospital, East China Normal University, Wuhu, China
| | - Guixiang Xie
- Division of Nephrology, Wuhu Hospital, East China Normal University, Wuhu, China
| | - Xiaoyan Zhang
- Division of Nephrology, Wuhu Hospital, East China Normal University, Wuhu, China
- Health Science Center, East China Normal University, Shanghai, China
| |
Collapse
|
6
|
Kubota H, Kunisawa K, Wulaer B, Hasegawa M, Kurahashi H, Sakata T, Tezuka H, Kugita M, Nagao S, Nagai T, Furuyashiki T, Narumiya S, Saito K, Nabeshima T, Mouri A. High salt induces cognitive impairment via the interaction of the angiotensin II-AT 1 and prostaglandin E2-EP 1 systems. Br J Pharmacol 2023; 180:2393-2411. [PMID: 37076133 DOI: 10.1111/bph.16093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/05/2023] [Accepted: 04/13/2023] [Indexed: 04/21/2023] Open
Abstract
BACKGROUND AND PURPOSE High salt (HS) intake has been associated with hypertension and cognitive impairment. It is well known that the angiotensin II (Ang II)-AT1 receptor and prostaglandin E2 (PGE2)-EP1 receptor systems are involved in hypertension and neurotoxicity. However, the involvement of these systems in HS-mediated hypertension and emotional and cognitive impairments remains unclear. EXPERIMENTAL APPROACH Mice were loaded with HS solution (2% NaCl drinking water) for 12 weeks, and blood pressure was monitored. Subsequently, effects of HS intake on emotional and cognitive function and tau phosphorylation in the prefrontal cortex (PFC) and hippocampus (HIP) were investigated. The involvement of Ang II-AT1 and PGE2-EP1 systems in HS-induced hypertension and neuronal and behavioural impairments was examined by treatment with losartan, an AT1 receptor blocker (ARB), or EP1 gene knockout. KEY RESULTS We demonstrate that hypertension and impaired social behaviour and object recognition memory following HS intake may be associated with tau hyperphosphorylation, decreased phosphorylation of Ca2+ /calmodulin-dependent protein kinase II (CaMKII), and postsynaptic density protein 95 (PSD95) expression in the PFC and HIP of mice. These changes were blocked by pharmacological treatment with losartan or EP1 receptor gene knockout. CONCLUSIONS AND IMPLICATIONS Our findings suggest that the interaction of Ang II-AT1 receptor and PGE2-EP1 receptor systems could be novel therapeutic targets for hypertension-induced cognitive impairment.
Collapse
Affiliation(s)
- Hisayoshi Kubota
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Toyoake, Aichi, Japan
| | - Kazuo Kunisawa
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Toyoake, Aichi, Japan
| | - Bolati Wulaer
- Laboratory of Health and Medical Science Innovation (HMSI), Fujita Health University Graduate School of Health Science, Toyoake, Aichi, Japan
| | - Masaya Hasegawa
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Toyoake, Aichi, Japan
| | - Hitomi Kurahashi
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Toyoake, Aichi, Japan
| | - Takatoshi Sakata
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Toyoake, Aichi, Japan
| | - Hiroyuki Tezuka
- Department of Cellular Function Analysis, Research Promotion and Support Headquarters, Fujita Health University, Toyoake, Aichi, Japan
| | - Masanori Kugita
- Education and Research Facility of Animal Models for Human Diseases, Center for Research Promotion and Support, Fujita Health University, Toyoake, Aichi, Japan
| | - Shizuko Nagao
- Education and Research Facility of Animal Models for Human Diseases, Center for Research Promotion and Support, Fujita Health University, Toyoake, Aichi, Japan
| | - Taku Nagai
- Division of Behavioral Neuropharmacology International Center for Brain Science (ICBS), Fujita Health University, Toyoake, Aichi, Japan
| | - Tomoyuki Furuyashiki
- Division of Pharmacology, Graduate School of Medicine, Kobe University, Kobe, Hyogo, Japan
| | - Shuh Narumiya
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto, Kyoto, Japan
| | - Kuniaki Saito
- Laboratory of Health and Medical Science Innovation (HMSI), Fujita Health University Graduate School of Health Science, Toyoake, Aichi, Japan
- Department of Disease Control and Prevention, Fujita Health University Graduate School of Health Science, Toyoake, Aichi, Japan
- Japanese Drug Organization of Appropriate Use and Research, Nagoya, Aichi, Japan
| | - Toshitaka Nabeshima
- Laboratory of Health and Medical Science Innovation (HMSI), Fujita Health University Graduate School of Health Science, Toyoake, Aichi, Japan
- Japanese Drug Organization of Appropriate Use and Research, Nagoya, Aichi, Japan
| | - Akihiro Mouri
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Toyoake, Aichi, Japan
- Japanese Drug Organization of Appropriate Use and Research, Nagoya, Aichi, Japan
| |
Collapse
|
7
|
Yang X, Zeng H, Wang L, Luo S, Zhou Y. Activation of Piezo1 downregulates renin in juxtaglomerular cells and contributes to blood pressure homeostasis. Cell Biosci 2022; 12:197. [PMID: 36471394 PMCID: PMC9720979 DOI: 10.1186/s13578-022-00931-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 11/17/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The synthesis and secretion of renin in juxtaglomerular (JG) cells are closely regulated by the blood pressure. To date, however, the molecular identity through which JG cells respond to the blood pressure remains unclear. RESULTS Here we discovered that Piezo1, a mechanosensitive ion channel, was colocalized with renin in mouse kidney as well as As4.1 cells, a commonly used JG cell line. Activation of Piezo1 by its agonist Yoda1 induced an intracellular calcium increase and downregulated the expression of renin in these cells, while knockout of Piezo1 in JG cells abolished the effect of Yoda1. Meanwhile, mechanical stress using microfluidics also induced an intracellular calcium increase in wildtype but not Piezo1 knockout JG cells. Mechanistically, we demonstrated that activation of Piezo1 upregulated the Ptgs2 expression via the calcineurin-NFAT pathway and increased the production of Ptgs2 downstream molecule PGE2 in JG cells. Surprisingly, we discovered that increased PGE2 could decreased the renin expression through the PGE2 receptor EP1 and EP3, which inhibited the cAMP production in JG cells. In mice, we found that activation of Piezo1 significantly downregulated the renin expression and blood pressure in wildtype but not adeno-associated virus (AAV)-mediated kidney specific Piezo1 knockdown mice. CONCLUSIONS In summary, these results revealed that activation of Piezo1 could downregulate the renin expression in JG cells and mice, subsequently a reduction of blood pressure, highlighting its therapeutic potential as a drug target of the renin-angiotensin system.
Collapse
Affiliation(s)
- Xiaoqiang Yang
- grid.412536.70000 0004 1791 7851Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China ,grid.412536.70000 0004 1791 7851Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 Guangdong China
| | - Honghui Zeng
- grid.412536.70000 0004 1791 7851Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China ,grid.412536.70000 0004 1791 7851Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 Guangdong China
| | - Le Wang
- grid.412536.70000 0004 1791 7851Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China ,grid.412536.70000 0004 1791 7851Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 Guangdong China
| | - Siweier Luo
- grid.412536.70000 0004 1791 7851Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China ,grid.412536.70000 0004 1791 7851Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 Guangdong China
| | - Yiming Zhou
- grid.412536.70000 0004 1791 7851Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China ,grid.412536.70000 0004 1791 7851Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 Guangdong China
| |
Collapse
|
8
|
Yang T, Song C, Ralph DL, Andrews P, Sparks MA, Koller BH, McDonough AA, Coffman TM. Cell-Specific Actions of the Prostaglandin E-Prostanoid Receptor 4 Attenuating Hypertension: A Dominant Role for Kidney Epithelial Cells Compared With Macrophages. J Am Heart Assoc 2022; 11:e026581. [PMID: 36172956 PMCID: PMC9673718 DOI: 10.1161/jaha.122.026581] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Background A beneficial role for prostanoids in hypertension is suggested by clinical studies showing nonsteroidal anti-inflammatory drugs, which block the production of all prostanoids, cause sodium retention and exacerbate hypertension. Among prostanoids, prostaglandin E2 and its E-prostanoid receptor 4 receptor (EP4R) have been implicated in blood pressure control. Our previous study found that conditional deletion of EP4R from all tissues in adult mice exacerbates angiotensin II-dependent hypertension, suggesting a powerful effect of EP4R to resist blood pressure elevation. We also found that elimination of EP4R from vascular smooth muscle cells did not affect the severity of hypertension, suggesting nonvascular targets of prostaglandin E mediate this antihypertensive effect. Methods and Results Here we generated mice with cell-specific deletion of EP4R from macrophage-specific EP4 receptor knockouts or kidney epithelial cells (KEKO) to assess the contributions of EP4R in these cells to hypertension pathogenesis. Macrophage-specific EP4 receptor knockouts showed similar blood pressure responses to alterations in dietary sodium or chronic angiotensin II infusion as Controls. By contrast, angiotensin II-dependent hypertension was significantly augmented in KEKOs (mean arterial pressure: 146±3 mm Hg) compared with Controls (137±4 mm Hg; P=0.02), which was accompanied by impaired natriuresis in KEKOs. Because EP4R expression in the kidney is enriched in the collecting duct, we compared responses to amiloride in angiotensin II-infused KEKOs and Controls. Blockade of the epithelial sodium channel with amiloride caused exaggerated natriuresis in KEKOs compared with Controls (0.21±0.01 versus 0.15±0.02 mmol/24 hour per 20 g; P=0.015). Conclusions Our data suggest EP4R in kidney epithelia attenuates hypertension. This antihypertension effect of EP4R may be mediated by reducing the activity of the epithelial sodium channel, thereby promoting natriuresis.
Collapse
Affiliation(s)
- Ting Yang
- Division of Nephrology‐Department of MedicineDuke UniversityDurhamNC
| | - Chengcheng Song
- Division of Nephrology‐Department of MedicineDuke UniversityDurhamNC,Department of AnesthesiologyTianjin Medical University General HospitalTianjinChina
| | - Donna L. Ralph
- Department of Physiology and NeuroscienceKeck School of Medicine of the University of Southern CaliforniaLos AngelesCA
| | - Portia Andrews
- Division of Nephrology‐Department of MedicineDuke UniversityDurhamNC
| | - Matthew A. Sparks
- Division of Nephrology‐Department of MedicineDuke UniversityDurhamNC
| | | | - Alicia A. McDonough
- Department of Physiology and NeuroscienceKeck School of Medicine of the University of Southern CaliforniaLos AngelesCA
| | - Thomas M. Coffman
- Division of Nephrology‐Department of MedicineDuke UniversityDurhamNC,Cardiovascular and Metabolic Disorders Research ProgramDuke‐National University of Singapore Graduate Medical SchoolSingapore
| |
Collapse
|
9
|
Hansen JN, Kaiser F, Leyendecker P, Stüven B, Krause J, Derakhshandeh F, Irfan J, Sroka TJ, Preval KM, Desai PB, Kraut M, Theis H, Drews A, De‐Domenico E, Händler K, Pazour GJ, Henderson DJP, Mick DU, Wachten D. A cAMP signalosome in primary cilia drives gene expression and kidney cyst formation. EMBO Rep 2022; 23:e54315. [PMID: 35695071 PMCID: PMC9346484 DOI: 10.15252/embr.202154315] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 05/11/2022] [Accepted: 05/19/2022] [Indexed: 12/22/2022] Open
Abstract
The primary cilium constitutes an organelle that orchestrates signal transduction independently from the cell body. Dysregulation of this intricate molecular architecture leads to severe human diseases, commonly referred to as ciliopathies. However, the molecular underpinnings how ciliary signaling orchestrates a specific cellular output remain elusive. By combining spatially resolved optogenetics with RNA sequencing and imaging, we reveal a novel cAMP signalosome that is functionally distinct from the cytoplasm. We identify the genes and pathways targeted by the ciliary cAMP signalosome and shed light on the underlying mechanisms and downstream signaling. We reveal that chronic stimulation of the ciliary cAMP signalosome transforms kidney epithelia from tubules into cysts. Counteracting this chronic cAMP elevation in the cilium by small molecules targeting activation of phosphodiesterase-4 long isoforms inhibits cyst growth. Thereby, we identify a novel concept of how the primary cilium controls cellular functions and maintains tissue integrity in a specific and spatially distinct manner and reveal novel molecular components that might be involved in the development of one of the most common genetic diseases, polycystic kidney disease.
Collapse
Affiliation(s)
- Jan N Hansen
- Institute of Innate ImmunityMedical FacultyUniversity of BonnBonnGermany
| | - Fabian Kaiser
- Institute of Innate ImmunityMedical FacultyUniversity of BonnBonnGermany
| | | | - Birthe Stüven
- Institute of Innate ImmunityMedical FacultyUniversity of BonnBonnGermany
| | | | | | | | - Tommy J Sroka
- Center for Molecular Signaling (PZMS)Center of Human and Molecular Biology (ZHMB)Saarland University, School of MedicineHomburgGermany
| | - Kenley M Preval
- Program in Molecular MedicineUniversity of Massachusetts Chan Medical School, Biotech IIWorcesterMAUSA
| | - Paurav B Desai
- Program in Molecular MedicineUniversity of Massachusetts Chan Medical School, Biotech IIWorcesterMAUSA
| | - Michael Kraut
- Precise Platform for Single Cell Genomics and EpigenomicsDepartment of Systems MedicineGerman Center for Neurogenerative DiseasesBonnGermany
| | - Heidi Theis
- Precise Platform for Single Cell Genomics and EpigenomicsDepartment of Systems MedicineGerman Center for Neurogenerative DiseasesBonnGermany
| | - Anna‐Dorothee Drews
- Precise Platform for Single Cell Genomics and EpigenomicsDepartment of Systems MedicineGerman Center for Neurogenerative DiseasesBonnGermany
| | - Elena De‐Domenico
- Precise Platform for Single Cell Genomics and EpigenomicsDepartment of Systems MedicineGerman Center for Neurogenerative DiseasesBonnGermany
| | - Kristian Händler
- Precise Platform for Single Cell Genomics and EpigenomicsDepartment of Systems MedicineGerman Center for Neurogenerative DiseasesBonnGermany
| | - Gregory J Pazour
- Program in Molecular MedicineUniversity of Massachusetts Chan Medical School, Biotech IIWorcesterMAUSA
| | | | - David U Mick
- Center for Molecular Signaling (PZMS)Center of Human and Molecular Biology (ZHMB)Saarland University, School of MedicineHomburgGermany
| | - Dagmar Wachten
- Institute of Innate ImmunityMedical FacultyUniversity of BonnBonnGermany
| |
Collapse
|
10
|
Boi R, Ebefors K, Henricsson M, Borén J, Nyström J. Modified lipid metabolism and cytosolic phospholipase A2 activation in mesangial cells under pro-inflammatory conditions. Sci Rep 2022; 12:7322. [PMID: 35513427 PMCID: PMC9072365 DOI: 10.1038/s41598-022-10907-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/08/2022] [Indexed: 02/07/2023] Open
Abstract
Diabetic kidney disease is a consequence of hyperglycemia and other complex events driven by early glomerular hemodynamic changes and a progressive expansion of the mesangium. The molecular mechanisms behind the pathophysiological alterations of the mesangium are yet to be elucidated. This study aimed at investigating whether lipid signaling might be the missing link. Stimulation of human mesangial cells with high glucose primed the inflammasome-driven interleukin 1 beta (IL-1β) secretion, which in turn stimulated platelet-derived growth factor (PDGF-BB) release. Finally, PDGF-BB increased IL-1β secretion synergistically. Both IL-1β and PDGF-BB stimulation triggered the formation of phosphorylated sphingoid bases, as shown by lipidomics, and activated cytosolic phospholipase cPLA2, sphingosine kinase 1, cyclooxygenase 2, and autotaxin. This led to the release of arachidonic acid and lysophosphatidylcholine, activating the secretion of vasodilatory prostaglandins and proliferative lysophosphatidic acids. Blocking cPLA2 release of arachidonic acid reduced mesangial cells proliferation and prostaglandin secretion. Validation was performed in silico using the Nephroseq database and a glomerular transcriptomic database. In conclusion, hyperglycemia primes glomerular inflammatory and proliferative stimuli triggering lipid metabolism modifications in human mesangial cells. The upregulation of cPLA2 was critical in this setting. Its inhibition reduced mesangial secretion of prostaglandins and proliferation, making it a potential therapeutical target.
Collapse
Affiliation(s)
- Roberto Boi
- Institute of Neuroscience and Physiology, Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 40530, Gothenburg, Sweden
| | - Kerstin Ebefors
- Institute of Neuroscience and Physiology, Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 40530, Gothenburg, Sweden
| | - Marcus Henricsson
- Institute of Medicine, Department of Molecular and Clinical Medicine, Wallenberg Laboratory, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jan Borén
- Institute of Medicine, Department of Molecular and Clinical Medicine, Wallenberg Laboratory, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jenny Nyström
- Institute of Neuroscience and Physiology, Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 40530, Gothenburg, Sweden.
| |
Collapse
|
11
|
Xu C, Yang G, Fu Z, Chen Y, Xie S, Wang F, Yang T. Na +-Retaining Action of COX-2 (Cyclooxygenase-2)/EP 1 Pathway in the Collecting Duct via Activation of Intrarenal Renin-Angiotensin-Aldosterone System and Epithelial Sodium Channel. Hypertension 2022; 79:1190-1202. [PMID: 35296155 DOI: 10.1161/hypertensionaha.121.17245] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The collecting duct (CD) is a major site of both biosynthesis and action of prostaglandin E2 as highlighted by the predominant expression of COX-2 (cyclooxygenase-2) and some E-prostanoid (EP) subtypes at this nephron site. The purpose of this study was to determine the relevance and mechanism of CD COX-2/prostaglandin E2/EP1 signaling for the regulation of Na+ hemostasis during Na+ depletion. METHODS Mice with Aqp2Cre-driven deletion of COX-2 (COX-2fl/flAqp2Cre+) or the EP1 subtype (EP1fl/flAqp2Cre+) were generated and the Na+-wasting phenotype of these mice during low-salt (LS) intake was examined. EP subtypes responsible for prostaglandin E2-induced local renin response were analyzed in primary cultured mouse inner medullary CD cells. RESULTS Following 28-day LS intake, COX-2fl/flAqp2Cre+ mice exhibited a higher urinary Na+ excretion and lower cumulative Na+ balance, accompanied with suppressed intrarenal renin, AngII (angiotensin II), and aldosterone, expression of CYP11B2 (cytochrome P450 family 11 subfamily B member 2), and blunted expression of epithelial sodium channel subunits compared to floxed controls (COX-2fl/flAqp2Cre-), whereas no differences were observed for indices of systemic renin-angiotensin-aldosterone system. In cultured CD cells, exposure to prostaglandin E2 stimulated release of soluble (pro)renin receptor, prorenin/renin and aldosterone and the stimulation was more sensitive to antagonism of EP1 as compared other EP subtypes. Subsequently, EP1fl/flAqp2Cre+ mice largely recapitulated Na+-wasting phenotype seen in COX-2fl/flAqp2Cre+ mice. CONCLUSIONS The study for the first time reports that CD COX-2/EP1 pathway might play a key role in maintenance of Na+ homeostasis in the face of Na+ depletion, at least in part, through activation of intrarenal renin-angiotensin-aldosterone-system and epithelial sodium channel.
Collapse
Affiliation(s)
- Chuanming Xu
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City
| | - Guangrui Yang
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City
| | - Ziwei Fu
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City
| | - Yanting Chen
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City
| | - Shiying Xie
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City
| | - Fei Wang
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City
| | - Tianxin Yang
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City
| |
Collapse
|
12
|
Fuchs MAA, Schrankl J, Leupold C, Wagner C, Kurtz A, Broeker KAE. Intact prostaglandin signaling through EP2 and EP4 receptors in stromal progenitor cells is required for normal development of the renal cortex in mice. Am J Physiol Renal Physiol 2022; 322:F295-F307. [PMID: 35037469 DOI: 10.1152/ajprenal.00414.2021] [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: 11/11/2021] [Accepted: 01/10/2022] [Indexed: 01/20/2023] Open
Abstract
Cyclooxygenase (Cox) inhibitors are known to have severe side effects during renal development. These consist of reduced renal function, underdeveloped subcapsular glomeruli, interstitial fibrosis, and thinner cortical tissue. Global genetic deletion of Cox-2 mimics the phenotype observed after application of Cox inhibitors. This study aimed to investigate which cell types express Cox-2 and prostaglandin E2 receptors and what functions are mediated through this pathway during renal development. Expression of EP2 and EP4 mRNA was detected by RNAscope mainly in descendants of FoxD1+ stromal progenitors; EP1 and EP3, on the other hand, were expressed in tubules. Cox-2 mRNA was detected in medullary interstitial cells and macula densa cells. Functional investigations were performed with a cell-specific approach to delete Cox-2, EP2, and EP4 in FoxD1+ stromal progenitor cells. Our data show that Cox-2 expression in macula densa cells is sufficient to drive renal development. Deletion of EP2 or EP4 in FoxD1+ cells had no functional effect on renal development. Codeletion of EP2 and EP4 in FoxD1+ stromal cells, however, led to severe glomerular defects and a strong decline of glomerular filtration rate (1.316 ± 69.7 µL/min/100 g body wt in controls vs. 644.1 ± 64.58 µL/min/100 g body wt in FoxD1+/Cre EP2-/- EP4ff mice), similar to global deletion of Cox-2. Furthermore, EP2/EP4-deficient mice showed a significant increase in collagen production with a strong downregulation of renal renin expression. This study shows the distinct localization of EP receptors in mice. Functionally, we could identify EP2 and EP4 receptors in stromal FoxD1+ progenitor cells as essential receptor subtypes for normal renal development.NEW & NOTEWORTHY Cyclooxygenase-2 (Cox-2) produces prostaglandins that are essential for normal renal development. It is unclear in which cells Cox-2 and the receptors for prostaglandin E2 (EP receptors) are expressed during late nephrogenesis. This study identified the expression sites for EP subtypes and Cox-2 in neonatal mouse kidneys. Furthermore, it shows that stromal progenitor cells may require intact prostaglandin E2 signaling through EP2 and EP4 receptors for normal renal development.
Collapse
MESH Headings
- Animals
- Cyclooxygenase 2/genetics
- Cyclooxygenase 2/metabolism
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/metabolism
- Gene Expression Regulation, Developmental
- Kidney Cortex/cytology
- Kidney Cortex/enzymology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Organogenesis
- Prostaglandins/metabolism
- Receptors, Prostaglandin E, EP2 Subtype/genetics
- Receptors, Prostaglandin E, EP2 Subtype/metabolism
- Receptors, Prostaglandin E, EP4 Subtype/genetics
- Receptors, Prostaglandin E, EP4 Subtype/metabolism
- Signal Transduction
- Stem Cells/metabolism
- Stromal Cells/enzymology
- Mice
Collapse
Affiliation(s)
| | - Julia Schrankl
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Christina Leupold
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Charlotte Wagner
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Armin Kurtz
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | | |
Collapse
|
13
|
Melnik LI, Garry RF. Enterotoxigenic Escherichia coli Heat-Stable Toxin and Ebola Virus Delta Peptide: Similarities and Differences. Pathogens 2022; 11:pathogens11020170. [PMID: 35215114 PMCID: PMC8878840 DOI: 10.3390/pathogens11020170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 01/27/2023] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) STb toxin exhibits striking structural similarity to Ebola virus (EBOV) delta peptide. Both ETEC and EBOV delta peptide are enterotoxins. Comparison of the structural and functional similarities and differences of these two toxins illuminates features that are important in induction of pathogenesis by a bacterial and viral pathogen.
Collapse
Affiliation(s)
- Lilia I. Melnik
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA;
- Viral Hemorrhagic Fever Consortium, New Orleans, LA 70112, USA
- Correspondence: ; Tel.: +1-(504)988-3818
| | - Robert F. Garry
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA;
- Viral Hemorrhagic Fever Consortium, New Orleans, LA 70112, USA
| |
Collapse
|
14
|
Mutsaers HA, Nørregaard R. Prostaglandin E2 receptors as therapeutic targets in renal fibrosis. Kidney Res Clin Pract 2022; 41:4-13. [PMID: 35108767 PMCID: PMC8816406 DOI: 10.23876/j.krcp.21.222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 10/17/2021] [Indexed: 11/04/2022] Open
Affiliation(s)
| | - Rikke Nørregaard
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Correspondence: Rikke Nørregaard Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, DK-8200 Aarhus N, Denmark E-mail:
| |
Collapse
|
15
|
Ziemanski JF, Wilson L, Barnes S, Nichols KK. Prostaglandin E2 and F2α Alter Expression of Select Cholesteryl Esters and Triacylglycerols Produced by Human Meibomian Gland Epithelial Cells. Cornea 2022; 41:95-105. [PMID: 34483274 PMCID: PMC8648972 DOI: 10.1097/ico.0000000000002835] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 06/10/2021] [Indexed: 01/03/2023]
Abstract
PURPOSE PGF2α analogs are commonly used to treat glaucoma and are associated with higher rates of meibomian gland dysfunction (MGD). The purpose of this study was to evaluate the physiological effects of PGF2α and PGE2 on immortalized human meibomian gland epithelial cells (HMGECs). METHODS HMGECs were immunostained for the 4 PGE2 receptors (EP1, EP2, EP3, and EP4) and 1 PGF2α receptor (FP) and imaged. Rosiglitazone-differentiated HMGECs were exposed to PGF2α and PGE2 (10-9 to 10-6 M) for 3 hours. Cell viability was assessed by an adenosine triphosphate-based luminescent assay, and lipid extracts were analyzed for cholesteryl esters (CEs), wax esters (WEs), and triacylglycerols (TAGs) by ESI-MSMSALL in positive ion mode by a Triple TOF 5600 Mass Spectrometer using SCIEX LipidView 1.3. RESULTS HMGECs expressed 3 PGE2 receptors (EP1, EP2, and EP4) and the 1 PGF2α receptor (FP). Neither PGE2 nor PGF2α showed signs of cytotoxicity at any of the concentrations tested. WEs were not detected from any of the samples, but both CEs and TAGs exhibited a diverse and dynamic profile. PGE2 suppressed select CEs (CE 22:1, CE 26:0, CE 28:1, and CE 30:1). PGF2α dose dependently increased several CEs (CE 20:2, CE 20:1, CE 22:1, and CE 24:0) yet decreased others. Both prostaglandins led to nonspecific TAG remodeling. CONCLUSIONS PGE2 and PGF2α showed minimal effect on HMGEC viability. PGF2α influences lipid expression greater than PGE2 and may do so by interfering with meibocyte differentiation. This work may provide insight into the mechanism of MGD development in patients with glaucoma treated with PGF2α analogs.
Collapse
Affiliation(s)
- Jillian F. Ziemanski
- University of Alabama at Birmingham, School of Optometry, Department of Optometry Vision Science, Birmingham, AL, USA
| | - Landon Wilson
- University of Alabama at Birmingham, School of Medicine, Department of Pharmacology Toxicology, Birmingham, AL, USA
| | - Stephen Barnes
- University of Alabama at Birmingham, School of Medicine, Department of Pharmacology Toxicology, Birmingham, AL, USA
| | - Kelly K. Nichols
- University of Alabama at Birmingham, School of Optometry, Department of Optometry Vision Science, Birmingham, AL, USA
| |
Collapse
|
16
|
Kim S, Jo CH, Kim GH. The Role of Vasopressin V2 Receptor in Drug-Induced Hyponatremia. Front Physiol 2021; 12:797039. [PMID: 34955900 PMCID: PMC8703040 DOI: 10.3389/fphys.2021.797039] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/19/2021] [Indexed: 12/15/2022] Open
Abstract
Hyponatremia is frequently encountered in clinical practice and usually induced by renal water retention. Many medications are considered to be among the various causes of hyponatremia, because they either stimulate the release of arginine vasopressin (AVP) or potentiate its action in the kidney. Antidepressants, anticonvulsants, antipsychotics, diuretics, and cytotoxic agents are the major causes of drug-induced hyponatremia. However, studies addressing the potential of these drugs to increase AVP release from the posterior pituitary gland or enhance urine concentration through intrarenal mechanisms are lacking. We previously showed that in the absence of AVP, sertraline, carbamazepine, haloperidol, and cyclophosphamide each increased vasopressin V2 receptor (V2R) mRNA and aquaporin-2 (AQP2) protein and mRNA expression in primary cultured inner medullary collecting duct cells. The upregulation of AQP2 was blocked by the V2R antagonist tolvaptan or protein kinase A (PKA) inhibitors. These findings led us to conclude that the nephrogenic syndrome of inappropriate antidiuresis (NSIAD) is the main mechanism of drug-induced hyponatremia. Previous studies have also shown that the V2R has a role in chlorpropamide-induced hyponatremia. Several other agents, including metformin and statins, have been found to induce antidiuresis and AQP2 upregulation through various V2R-independent pathways in animal experiments but are not associated with hyponatremia despite being frequently used clinically. In brief, drug-induced hyponatremia can be largely explained by AQP2 upregulation from V2R-cAMP-PKA signaling in the absence of AVP stimulation. This paper reviews the central and nephrogenic mechanisms of drug-induced hyponatremia and discusses the importance of the canonical pathway of AQP2 upregulation in drug-induced NSIAD.
Collapse
Affiliation(s)
- Sua Kim
- Institute of Biomedical Science, Hanyang University College of Medicine, Seoul, South Korea
| | - Chor Ho Jo
- Institute of Biomedical Science, Hanyang University College of Medicine, Seoul, South Korea
| | - Gheun-Ho Kim
- Institute of Biomedical Science, Hanyang University College of Medicine, Seoul, South Korea.,Department of Internal Medicine, Hanyang University College of Medicine, Seoul, South Korea
| |
Collapse
|
17
|
Mansley MK, Niklas C, Nacken R, Mandery K, Glaeser H, Fromm MF, Korbmacher C, Bertog M. Prostaglandin E2 stimulates the epithelial sodium channel (ENaC) in cultured mouse cortical collecting duct cells in an autocrine manner. J Gen Physiol 2021; 152:151804. [PMID: 32442241 PMCID: PMC7398144 DOI: 10.1085/jgp.201912525] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 03/30/2020] [Accepted: 04/21/2020] [Indexed: 12/26/2022] Open
Abstract
Prostaglandin E2 (PGE2) is the most abundant prostanoid in the kidney, affecting a wide range of renal functions. Conflicting data have been reported regarding the effects of PGE2 on tubular water and ion transport. The amiloride-sensitive epithelial sodium channel (ENaC) is rate limiting for transepithelial sodium transport in the aldosterone-sensitive distal nephron. The aim of the present study was to explore a potential role of PGE2 in regulating ENaC in cortical collecting duct (CCD) cells. Short-circuit current (ISC) measurements were performed using the murine mCCDcl1 cell line known to express characteristic properties of CCD principal cells and to be responsive to physiological concentrations of aldosterone and vasopressin. PGE2 stimulated amiloride-sensitive ISC via basolateral prostaglandin E receptors type 4 (EP4) with an EC50 of ∼7.1 nM. The rapid stimulatory effect of PGE2 on ISC resembled that of vasopressin. A maximum response was reached within minutes, coinciding with an increased abundance of β-ENaC at the apical plasma membrane and elevated cytosolic cAMP levels. The effects of PGE2 and vasopressin were nonadditive, indicating similar signaling cascades. Exposing mCCDcl1 cells to aldosterone caused a much slower (∼2 h) increase of the amiloride-sensitive ISC. Interestingly, the rapid effect of PGE2 was preserved even after aldosterone stimulation. Furthermore, application of arachidonic acid also increased the amiloride-sensitive ISC involving basolateral EP4 receptors. Exposure to arachidonic acid resulted in elevated PGE2 in the basolateral medium in a cyclooxygenase 1 (COX-1)–dependent manner. These data suggest that in the cortical collecting duct, locally produced and secreted PGE2 can stimulate ENaC-mediated transepithelial sodium transport.
Collapse
Affiliation(s)
- Morag K Mansley
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Niklas
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Regina Nacken
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Kathrin Mandery
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Hartmut Glaeser
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Martin F Fromm
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Christoph Korbmacher
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Marko Bertog
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
18
|
Aringer I, Artinger K, Schabhüttl C, Bärnthaler T, Mooslechner AA, Kirsch A, Pollheimer M, Eller P, Rosenkranz AR, Heinemann A, Eller K. Agonism of Prostaglandin E2 Receptor 4 Ameliorates Tubulointerstitial Injury in Nephrotoxic Serum Nephritis in Mice. J Clin Med 2021; 10:832. [PMID: 33670614 PMCID: PMC7922874 DOI: 10.3390/jcm10040832] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 12/21/2022] Open
Abstract
Selectively targeting the E-type prostanoid receptor 4 (EP4) might be a new therapeutic option in the treatment of glomerulonephritis (GN), since the EP4 receptor is expressed on different immune cells, resident kidney cells, and endothelial cells, which are all involved in the pathogenesis of immune-complex GN. This study aimed to evaluate the therapeutic potential and to understand the mode of action of EP4 agonist in immune-complex GN using the murine model of nephrotoxic serum nephritis (NTS). In vivo, NTS mice were treated two times daily with two different doses of an EP4 agonist ONO AE1-329 or vehicle for 14 days total. The effect of PGE2 and EP4 agonism and antagonism was tested on murine distal convoluted tubular epithelial cells (DCT) in vitro. In vivo, the higher dose of the EP4 agonist led to an improved NTS phenotype, including a reduced tubular injury score and reduced neutrophil gelatinase-associated lipocalin (NGAL) and blood urea nitrogen (BUN) levels. EP4 agonist treatment caused decreased CD4+ T cell infiltration into the kidney and increased proliferative capacity of tubular cells. Injection of the EP4 agonist resulted in dose-dependent vasodilation and hypotensive episodes. The low-dose EP4 agonist treatment resulted in less pronounced episodes of hypotension. In vitro, EP4 agonism resulted in cAMP production and increased distal convoluted tubular (DCT) proliferation. Taken together, EP4 agonism improved the NTS phenotype by various mechanisms, including reduced blood pressure, decreased CD4+ T cell infiltration, and a direct effect on tubular cells leading to increased proliferation probably by increasing cAMP levels.
Collapse
Affiliation(s)
- Ida Aringer
- Clinical Division of Nephrology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria; (I.A.); (K.A.); (C.S.); (A.A.M.); (A.R.R.)
- Clinical Department of Internal Medicine 1, University Hospital St. Poelten, 3100 St. Poelten, Austria
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, BioTechMed Graz, 8036 Graz, Austria; (T.B.); (A.H.)
| | - Katharina Artinger
- Clinical Division of Nephrology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria; (I.A.); (K.A.); (C.S.); (A.A.M.); (A.R.R.)
| | - Corinna Schabhüttl
- Clinical Division of Nephrology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria; (I.A.); (K.A.); (C.S.); (A.A.M.); (A.R.R.)
| | - Thomas Bärnthaler
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, BioTechMed Graz, 8036 Graz, Austria; (T.B.); (A.H.)
| | - Agnes A. Mooslechner
- Clinical Division of Nephrology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria; (I.A.); (K.A.); (C.S.); (A.A.M.); (A.R.R.)
| | - Andrijana Kirsch
- Clinical Department of Phoniatrics, Medical University of Graz, 8036 Graz, Austria;
| | - Marion Pollheimer
- Institute of Pathology, Medical University of Graz, 8036 Graz, Austria;
| | - Philipp Eller
- Intensive Care Unit, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria;
| | - Alexander R. Rosenkranz
- Clinical Division of Nephrology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria; (I.A.); (K.A.); (C.S.); (A.A.M.); (A.R.R.)
| | - Akos Heinemann
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, BioTechMed Graz, 8036 Graz, Austria; (T.B.); (A.H.)
| | - Kathrin Eller
- Clinical Division of Nephrology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria; (I.A.); (K.A.); (C.S.); (A.A.M.); (A.R.R.)
| |
Collapse
|
19
|
Aukema HM. Prostaglandins as potential targets for the treatment of polycystic kidney disease. Prostaglandins Leukot Essent Fatty Acids 2021; 164:102220. [PMID: 33285393 DOI: 10.1016/j.plefa.2020.102220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/24/2020] [Accepted: 11/24/2020] [Indexed: 12/15/2022]
Abstract
Polycystic kidney disease (PKD) is characterized by the proliferation of fluid-filled kidney cysts that enlarge over time, causing damage to the surrounding kidney and ultimately resulting in kidney failure. Both increased cell proliferation and fluid secretion are stimulated by increased cyclic adenosine monophosphate (cAMP) in PKD kidneys, so many treatments for the disease target cAMP lowering. Prostaglandins (PG) levels are elevated in multiple animal models of PKD and mediate many of their effects by elevating cAMP levels. Inhibiting the production of PG with cyclooxygenase 2 (COX2) inhibitors reduces PG levels and reduces disease progression. However, COX inhibitors also block beneficial PG and can cause nephrotoxicity. In an orthologous model of the main form of PKD, PGD2 and PGI2 were the two PG highest in kidneys and most affected by a COX2 inhibitor. Future studies are needed to determine whether specific blockage of PGD2 and/or PGI2 activity would lead to more targeted and effective treatments with fewer undesirable side-effects.
Collapse
Affiliation(s)
- Harold M Aukema
- Department of Food and Human Nutritional Sciences, University of Manitoba, MB R3T 2N2, Canada; Canadian Centre for Agri-Food Research in Health and Medicine, St Boniface Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada.
| |
Collapse
|
20
|
Himmel NJ, Rogers RT, Redd SK, Wang Y, Blount MA. Purinergic signaling is enhanced in the absence of UT-A1 and UT-A3. Physiol Rep 2021; 9:e14636. [PMID: 33369887 PMCID: PMC7769175 DOI: 10.14814/phy2.14636] [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: 02/26/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 11/24/2022] Open
Abstract
ATP is an important paracrine regulator of renal tubular water and urea transport. The activity of P2Y2 , the predominant P2Y receptor of the medullary collecting duct, is mediated by ATP, and modulates urinary concentration. To investigate the role of purinergic signaling in the absence of urea transport in the collecting duct, we studied wild-type (WT) and UT-A1/A3 null (UT-A1/A3 KO) mice in metabolic cages to monitor urine output, and collected tissue samples for analysis. We confirmed that UT-A1/A3 KO mice are polyuric, and concurrently observed lower levels of urinary cAMP as compared to WT, despite elevated serum vasopressin (AVP) levels. Because P2Y2 inhibits AVP-stimulated transport by dampening cAMP synthesis, we suspected that, similar to other models of AVP-resistant polyuria, purinergic signaling is increased in UT-A1/A3 KO mice. In fact, we observed that both urinary ATP and purinergic-mediated prostanoid (PGE2 ) levels were elevated. Collectively, our data suggest that the reduction of medullary osmolality due to the lack of UT-A1 and UT-A3 induces an AVP-resistant polyuria that is possibly exacerbated by, or at least correlated with, enhanced purinergic signaling.
Collapse
Affiliation(s)
- Nathaniel J. Himmel
- Renal DivisionDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
| | - Richard T. Rogers
- Renal DivisionDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
| | - Sara K. Redd
- Renal DivisionDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
| | - Yirong Wang
- Renal DivisionDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
| | - Mitsi A. Blount
- Renal DivisionDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
- Department of PhysiologyEmory University School of MedicineAtlantaGAUSA
| |
Collapse
|
21
|
Zhang L, Li P, Zhang BL, Yu ML, Xu RL, Wu H, Chen SP. Dysregulation of HULC promotes contrast-induced nephropathy (CIN) via regulating signaling pathway of miRNA-512 and prostaglandin E1 (PGE 1). Sci Rep 2020; 10:11691. [PMID: 32678165 PMCID: PMC7366906 DOI: 10.1038/s41598-020-68634-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 06/04/2020] [Indexed: 01/11/2023] Open
Abstract
It has been shown that contrast-induced nephropathy (CIN) can be attenuated by the administration of PGE1. As an enzyme responsible for the production of PGE1, PTGS1 was confirmed in this study as a miR-512 target. Meanwhile, HULC has been identified as a competing endogenous RNA of miR-512. Therefore, in this study, we tested the diagnostic value of HULC and miR-512 in subjects with or without CIN. In addition, we evaluated the regulatory relationship among HULC, miR-512, PTGS1 and PGE1 in vitro. We enrolled 320 patients with coronary heart disease and divided them into a CIN group and a non-CIN group. Subsequently, we detected the differential expression of miR-512, HULC and PGE1 in the two groups. We also used a dual luciferase reporter assay to evaluate the regulatory relationship among HULC, miR-512, PTGS1 and PGE1 in THP-1 cells. In patients with CIN, the expression levels of HULC and PGE1 were lower, but the expression level of miR-512 was higher. MiR-512 could directly bind to and negatively regulate the expression of PTGS1 and HULC. The expression of HULC was positively correlated with the expression of PTGS1 and PGE1, while negatively correlated with the expression of miR-512. The findings of this study demonstrated that deregulation of lncRNA-HULC/miR-512/PTGS1/PGE1 might be involved in the pathogenesis of CIN.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Cardiology, The First Affiliated Hospital of the Second Military Medical University, No. 168 Changhai Road, Yangpu District, Shanghai, People's Republic of China
| | - Pan Li
- Department of Cardiology, The First Affiliated Hospital of the Second Military Medical University, No. 168 Changhai Road, Yangpu District, Shanghai, People's Republic of China
| | - Bi-Li Zhang
- Department of Cardiology, The First Affiliated Hospital of the Second Military Medical University, No. 168 Changhai Road, Yangpu District, Shanghai, People's Republic of China.
| | - Man-Li Yu
- Department of Cardiology, The First Affiliated Hospital of the Second Military Medical University, No. 168 Changhai Road, Yangpu District, Shanghai, People's Republic of China
| | - Rong-Liang Xu
- Department of Cardiology, The First Affiliated Hospital of the Second Military Medical University, No. 168 Changhai Road, Yangpu District, Shanghai, People's Republic of China
| | - Hong Wu
- Department of Cardiology, The First Affiliated Hospital of the Second Military Medical University, No. 168 Changhai Road, Yangpu District, Shanghai, People's Republic of China
| | - Shao-Ping Chen
- Department of Cardiology, The First Affiliated Hospital of the Second Military Medical University, No. 168 Changhai Road, Yangpu District, Shanghai, People's Republic of China
| |
Collapse
|
22
|
Microbiota and metabolome responses in the cecum and serum of broiler chickens fed with plant essential oils or virginiamycin. Sci Rep 2020; 10:5382. [PMID: 32214106 PMCID: PMC7096418 DOI: 10.1038/s41598-020-60135-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 02/06/2020] [Indexed: 12/29/2022] Open
Abstract
This study investigated the cecal microbiota and serum metabolite profile of chickens fed with plant essential oils (PEO) or virginiamycin (VIRG) using high-throughput 16S rRNA gene sequencing and untargeted metabolomics approach. The main aim of this work was to explore the biochemical mechanisms involved in the improved growth performance of antibiotics and their alternatives in animal production. The results showed that both PEO and VIRG treatment significantly increased the relative abundance of phyla Bacteroidetes and decreased the abundance of phyla Firmicutes and genus of Lactobacillus in cecal microbiota of chickens. Compared to the control group (CT group), the relative abundance of genus of Alistipes, unclassified Rikenellaceae, Roseburia, and Anaeroplasma was enriched in the PEO group; that of genus Bacteroides, Lachnospiraceae, and unclassified Enterobacteriaceae was enriched in the cecal microbiota of the VIRG group. Untargeted metabolomics analyses revealed that the PEO treatment modified 102 metabolites and 3 KEGG pathways (primary bile acid biosynthesis and phenylalanine metabolism) in the cecal microbiota, and 81 metabolites and relevant KEGG pathways (fructose and mannose metabolism, biosynthesis of unsaturated fatty acids, and linoleic acid.) in the serum of the chicken. Compared to the CT group, VIRG treatment group differed 217 metabolites and 10 KEGG pathways in cecal contents and 142 metabolites and 7 KEGG pathways in serum of chickens. Pearson’s correlation analysis showed that phyla Bacteroidetes and genus of Bacteroides, Alistipes, and unclassified Rikenellaceae (in the VIRG and PE group) were positively correlated with many lipid metabolites. However, phyla Firmicutes and genera Lactobacillus (higher in the CT group) were negatively correlated with the lipid and thymine metabolism, and positively correlated with hydroxyisocaproic acid, cytosine, and taurine. This study shows that dietary supplementation with PEO and VIRG altered the composition and metabolism profile of the cecal microbiota, modified the serum metabolism profile.
Collapse
|
23
|
The positive effect of selective prostaglandin E2 receptor EP2 and EP4 blockade on cystogenesis in vitro is counteracted by increased kidney inflammation in vivo. Kidney Int 2020; 98:404-419. [PMID: 32622526 DOI: 10.1016/j.kint.2020.02.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 01/16/2020] [Accepted: 02/07/2020] [Indexed: 01/15/2023]
Abstract
Autosomal Dominant Polycystic Kidney Disease (ADPKD) is a major cause of end-stage kidney disease in man. The central role of cyclic adenosine monophosphate (cAMP) in ADPKD pathogenesis has been confirmed by numerous studies including positive clinical trial data. Here, we investigated the potential role of another major regulator of renal cAMP, prostaglandin E2 (PGE2), in modifying disease progression in ADPKD models using selective receptor modulators to all four PGE2 receptor subtypes (EP1-4). In 3D-culture model systems utilizing dog (MDCK) and patient-derived (UCL93, OX161-C1) kidney cell lines, PGE2 strikingly promoted cystogenesis and inhibited tubulogenesis by stimulating proliferation while reducing apoptosis. The effect of PGE2 on tubulogenesis and cystogenesis in 3D-culture was mimicked or abolished by selective EP2 and EP4 agonists or antagonists but not those specific to EP1 or EP3. In a Pkd1 mouse model (Pkd1nl/nl), kidney PGE2 and COX-2 expression were increased by two-fold at the peak of disease (week four). However, Pkd1nl/nl mice treated with selective EP2 (PF-04418948) or EP4 (ONO-AE3-208) antagonists from birth for three weeks had more severe cystic disease and fibrosis associated with increased cell proliferation and macrophage infiltration. A similar effect was observed for the EP4 antagonist ONO-AE3-208 in a second Pkd1 model (Pax8rtTA-TetO-Cre-Pkd1f/f). Thus, despite the positive effects of slowing cyst growth in vitro, the more complex effects of inhibiting EP2 or EP4 in vivo resulted in a worse outcome, possibly related to unexpected pro-inflammatory effects.
Collapse
|
24
|
Gupta A, Puri S, Puri V. Bioinformatics Unmasks the Maneuverers of Pain Pathways in Acute Kidney Injury. Sci Rep 2019; 9:11872. [PMID: 31417109 PMCID: PMC6695489 DOI: 10.1038/s41598-019-48209-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 07/31/2019] [Indexed: 12/30/2022] Open
Abstract
Acute Kidney injury (AKI) is one of the leading health concerns resulting in accumulation of nitrogenous as well as non-nitrogenous wastes in body and characterised by a rapid deterioration in kidney functions. Besides the major toll from the primary insult in the kidney, consequential extra-renal secondary insults endowed with the pathways of inflammatory milieu often complicates the disease outcome. Some of the known symptoms of AKI leading to clinical reporting are fatigue, loss of appetite, headache, nausea, vomiting, and pain in the flanks, wherein proinflammatory cytokines have been strongly implicated in pathogenesis of AKI and neuro-inflammation. Taking in account these clues, we have tried to decode the neuro-inflammation and pain perception phenomenon during the progression of AKI using the pathway integration and biological network strategies. The pathways and networks were generated using bioinformatics software viz. PANTHER, Genomatix and PathVisio to establish the relationship between immune and neuro related pathway in AKI. These observations envisage a neurol-renal axis that is predicted to involve calcium channels in neuro-inflammatory pathway of AKI. These observations, thus, pave a way for a new paradigm in understanding the interplay of neuro-immunological signalling in AKI.
Collapse
Affiliation(s)
- Aprajita Gupta
- Centre for Systems Biology and Bioinformatics, Panjab University, Chandigarh, India
| | - Sanjeev Puri
- Biotechnology Branch UIET, Panjab University, Chandigarh, India
| | - Veena Puri
- Centre for Systems Biology and Bioinformatics, Panjab University, Chandigarh, India.
| |
Collapse
|
25
|
Lin XM, Luo W, Wang H, Li RZ, Huang YS, Chen LK, Wu XP. The Role of Prostaglandin-Endoperoxide Synthase-2 in Chemoresistance of Non-Small Cell Lung Cancer. Front Pharmacol 2019; 10:836. [PMID: 31440159 PMCID: PMC6694719 DOI: 10.3389/fphar.2019.00836] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 07/01/2019] [Indexed: 12/22/2022] Open
Abstract
The prostaglandin-endoperoxide synthase-2 (PTGS2) plays essential roles in diverse pathological process. Although recent studies implied that PTGS2 was closely related with chemoresistance, the precise roles and the underlying mechanisms of PTGS2 in the developing process of chemoresistance in non-small cell lung cancer (NSCLC) remained elusive. In the present study, we revealed a novel molecular mechanism of PTGS2 implicated in the chemoresistance of NSCLC and proposed a model for the positive feedback regulation of PTGS2 in the process of developing resistance phenotype in NSCLC cells. Our results demonstrated that cisplatin induced PTGS2 expression through the ROS-ERK1/2-NF-κB signaling axis. The prostaglandin E2 (PGE2) derived from PTGS2 catalyzation further strengthened PTGS2 expression via the PGE2-EPs-ERK1/2 positive feedback loop, which induced multidrug resistance of NSCLC cells through up-regulation of BCL2 expression and the subsequent attenuation of cell apoptosis. Consistently, high levels of both PTGS2 and BCL2 were closely associated with poor survival in NSCLC patients. Inhibition of PTGS2 significantly reversed the chemoresistance in the resistant NSCLC in vitro and in vivo. Our results suggested that PTGS2 might be employed as an adjunctive therapeutic target for improving the response to the therapeutic agents in a subset of resistant NSCLC.
Collapse
Affiliation(s)
- Xiao-Mian Lin
- Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou, China
| | - Wu Luo
- Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou, China
| | - Heng Wang
- Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou, China
| | - Rong-Zhen Li
- Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou, China
| | - Yi-Shan Huang
- Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou, China
| | - Lian-Kuai Chen
- Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou, China
| | - Xiao-Ping Wu
- Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou, China
| |
Collapse
|
26
|
Herrera M, Yang T, Sparks MA, Manning MW, Koller BH, Coffman TM. Complex Role for E-Prostanoid 4 Receptors in Hypertension. J Am Heart Assoc 2019; 8:e010745. [PMID: 30764697 PMCID: PMC6405651 DOI: 10.1161/jaha.118.010745] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 01/22/2019] [Indexed: 12/24/2022]
Abstract
Background Prostaglandin E2 ( PGE 2) is a major prostanoid with multiple actions that potentially affect blood pressure ( BP ). PGE 2 acts through 4 distinct E-prostanoid ( EP ) receptor isoforms: EP 1 to EP 4. The EP 4 receptor ( EP 4R) promotes PGE 2-dependent vasodilation, but its role in the pathogenesis of hypertension is not clear. Methods and Results To address this issue, we studied mice after temporal- and cell-specific deletion of EP 4R. First, using a mouse line with loss of EP 4 expression induced universally after birth, we confirm that EP 4R mediates a major portion of the acute vasodilatory effects of infused PGE 2. In addition, EP 4 contributes to control of resting BP , which was increased by 5±1 mm Hg in animals with generalized deficiency of this receptor. We also show that EP 4 is critical for limiting elevations in BP caused by high salt feeding and long-term infusion of angiotensin II . To more precisely identify the mechanism for these actions, we generated mice in which EP 4R loss is induced after birth and is limited to smooth muscle. In these mice, acute PGE 2-dependent vasodilation was attenuated, indicating that this response is mediated by EP 4R in vascular smooth muscle cells. However, absence of EP 4R only in this vascular compartment had a paradoxical effect of lowering resting BP , whereas the protective effect of EP 4R on limiting angiotensin II-dependent hypertension was unaffected. Conclusions Taken together, our findings support a complex role for EP 4R in regulation of BP and in hypertension, which appears to involve actions of the EP 4R in tissues beyond vascular smooth muscle cells.
Collapse
Affiliation(s)
- Marcela Herrera
- Division of NephrologyDepartment of MedicineDuke UniversityDurhamNC
| | - Ting Yang
- Division of NephrologyDepartment of MedicineDuke UniversityDurhamNC
| | - Matthew A. Sparks
- Division of NephrologyDepartment of MedicineDuke UniversityDurhamNC
- Renal SectionDurham VA Medical CenterDurhamNC
| | | | | | - Thomas M. Coffman
- Division of NephrologyDepartment of MedicineDuke UniversityDurhamNC
- Renal SectionDurham VA Medical CenterDurhamNC
- Cardiovascular and Metabolic Disorders Research ProgramDuke–National University of Singapore Graduate Medical SchoolSingapore
| |
Collapse
|
27
|
Interpretation of Euphorbia Kansui Stir-Fried with Vinegar Treating Malignant Ascites by a UPLC-Q-TOF/MS Based Rat Serum and Urine Metabolomics Strategy Coupled with Network Pharmacology. Molecules 2018; 23:molecules23123246. [PMID: 30544627 PMCID: PMC6322356 DOI: 10.3390/molecules23123246] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 12/04/2018] [Accepted: 12/05/2018] [Indexed: 12/12/2022] Open
Abstract
Euphorbia kansui stir-fried with vinegar (V-kansui) has promising biological activities toward treating malignant ascites with reduced toxicity compared to crude kansui. But the mechanism concerning promoting the excretion of ascites has not been systematically studied. The purpose of this paper was to investigate the possible mechanism of V-kansui in treating malignant ascites, including metabolic pathways and molecular mechanism using an integrated serum and urine metabolomics coupled with network pharmacology. Serum and urine samples of rats were collected and analyzed by ultra-high-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF/MS). A comparison with crude kansui was also made to demonstrate the feasibility of processing. Principle component analysis (PCA) and orthogonal partial least square discriminate analysis (OPLS-DA) were conducted to discriminate the groups, search important variables and reveal the possible pathways. A compound-target-metabolite network was finally constructed to identify the crucial targets to further understand the molecular mechanism. Sixteen significant metabolites contributing to the discrimination of model and control groups were tentatively screened out. They were mainly involved in the arachidonic acid metabolism, steroid hormone biosynthesis and primary bile acid to possibly reduce inflammatory and modulate the renin-angiotensin-aldosterone system to achieve treating malignant ascites. A bio-network starting from the compounds and ending in the metabolites was constructed to elucidate the molecular mechanism. HSP90AA1, ANXA2, PRDX6, PCNA, SOD2 and ALB were identified as the potential key targets that were responsible for the treatment of malignant ascites by the parameter combining the average shortest path length and betweenness centrality. The correlated 17 compounds were considered as the potential active ingredients in V-kansui. In addition, the metabolomics showed that the effect of V-kansui was almost in accordance with crude kansui. These results systematically revealed the mechanism of V-kansui against malignant ascites for the first time using metabolomics coupled with network pharmacology. V-kansui could be a promising safe and therapeutic medicine for the excretion of ascites.
Collapse
|
28
|
Cao Y, Guan Y, Xu YY, Hao CM. Endothelial prostacyclin protects the kidney from ischemia-reperfusion injury. Pflugers Arch 2018; 471:543-555. [PMID: 30413885 PMCID: PMC6435627 DOI: 10.1007/s00424-018-2229-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 10/11/2018] [Accepted: 10/23/2018] [Indexed: 01/20/2023]
Abstract
Prostacyclin, or PGI2, is a product of PGI synthase (PGIS), down-stream of cyclooxygenase pathway. PGI2 has been demonstrated to play an important role in maintaining renal blood flow. Non-steroidal anti-inflammatory drugs (NSAIDs) that inhibit cyclooxygenase are reported to increase the susceptibility of patients to acute kidney injury (AKI). This study explores the role of endothelium-derived prostacyclin in ischemia-reperfusion injury (I/RI). The renal PGIS expression and PGI2 production markedly increased following I/RI. Loss of one allele of PGIS gene or selective endothelial PGIS deletion (TEK-CRE PGISfl/fl mice) caused more severe renal damage following I/RI than control mice. Iloprost, a PGI2 analog, administered 30 min before the I/R surgery, markedly attenuated the renal damage in both control mice and TEK-CRE PGISfl/fl mice. Renal p-PKA expression significantly increased after I/RI in wild-type mice but not in the PGIS deletion mice, consistent with IP receptor mediating the protective effect. Further studies showed that PGIS deficiency was associated with reduced fluorescence microsphere accumulation in the kidney following I/R. Folic acid also induced marked kidney injury; however, endothelial PGIS deletion did not worsen kidney injury compared with wild-type mice. These studies indicate that PGIS-derived PGI2 can protect the kidney from acute injury caused by ischemia and reperfusion and PGIS/PGI2 is a potential intervention target for AKI.
Collapse
Affiliation(s)
- Yingxue Cao
- Division of Nephrology, Huashan Hospital, Fudan University, 12 Wulumuqi Road (middle), Shanghai, 200040, China
| | - Yi Guan
- Division of Nephrology, Huashan Hospital, Fudan University, 12 Wulumuqi Road (middle), Shanghai, 200040, China
| | - Yun-Yu Xu
- Division of Nephrology, Huashan Hospital, Fudan University, 12 Wulumuqi Road (middle), Shanghai, 200040, China
| | - Chuan-Ming Hao
- Division of Nephrology, Huashan Hospital, Fudan University, 12 Wulumuqi Road (middle), Shanghai, 200040, China.
| |
Collapse
|
29
|
Abstract
Prostaglandins are synthesized through the metabolism of arachidonic acid via the cyclooxygenase pathway. There are five primary prostaglandins, PGD2, PGE2, PGF2, PGI2, and thromboxane B2, that all signal through distinct seven transmembrane, G-protein coupled receptors. The receptors through which the prostaglandins signal determines their immunologic or physiologic effects. For instance, the same prostaglandin may have opposing properties, dependent upon the signaling pathways activated. In this article, we will detail how inhibition of cyclooxygenase metabolism and regulation of prostaglandin signaling regulates allergic airway inflammation and asthma physiology. Possible prostaglandin therapeutic targets for allergic lung inflammation and asthma will also be reviewed, as informed by human studies, basic science, and animal models.
Collapse
Affiliation(s)
- R Stokes Peebles
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States.
| |
Collapse
|
30
|
Abstract
Fever is a common symptom of infectious and inflammatory disease. It is well-established that prostaglandin E2 is the final mediator of fever, which by binding to its EP3 receptor subtype in the preoptic hypothalamus initiates thermogenesis. Here, we review the different hypotheses on how the presence of peripherally released pyrogenic substances can be signaled to the brain to elicit fever. We conclude that there is unequivocal evidence for a humoral signaling pathway by which proinflammatory cytokines, through their binding to receptors on brain endothelial cells, evoke fever by eliciting prostaglandin E2 synthesis in these cells. The evidence for a role for other signaling routes for fever, such as signaling via circumventricular organs and peripheral nerves, as well as transfer into the brain of peripherally synthesized prostaglandin E2 are yet far from conclusive. We also review the efferent limb of the pyrogenic pathways. We conclude that it is well established that prostaglandin E2 binding in the preoptic hypothalamus produces fever by disinhibition of presympathetic neurons in the brain stem, but there is yet little understanding of the mechanisms by which factors such as nutritional status and ambient temperature shape the response to the peripheral immune challenge.
Collapse
Affiliation(s)
- Anders Blomqvist
- Department of Clinical and Experimental Medicine, Faculty of Medicine and Health, Linköping University, Linköping, Sweden
| | - David Engblom
- Department of Clinical and Experimental Medicine, Faculty of Medicine and Health, Linköping University, Linköping, Sweden
| |
Collapse
|
31
|
Mouat MA, Coleman JLJ, Smith NJ. GPCRs in context: sexual dimorphism in the cardiovascular system. Br J Pharmacol 2018; 175:4047-4059. [PMID: 29451687 DOI: 10.1111/bph.14160] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 01/31/2018] [Accepted: 02/09/2018] [Indexed: 12/31/2022] Open
Abstract
Cardiovascular disease (CVD) remains the largest cause of mortality worldwide, and there is a clear gender gap in disease occurrence, with men being predisposed to earlier onset of CVD, including atherosclerosis and hypertension, relative to women. Oestrogen may be a driving factor for female-specific cardioprotection, though androgens and sex chromosomes are also likely to contribute to sexual dimorphism in the cardiovascular system (CVS). Many GPCR-mediated processes are involved in cardiovascular homeostasis, and some exhibit clear sex divergence. Here, we focus on the G protein-coupled oestrogen receptor, endothelin receptors ETA and ETB and the eicosanoid G protein-coupled receptors (GPCRs), discussing the evidence and potential mechanisms leading to gender dimorphic responses in the vasculature. The use of animal models and pharmacological tools has been essential to understanding the role of these receptors in the CVS and will be key to further delineating their sex-specific effects. Ultimately, this may illuminate wider sex differences in cardiovascular pathology and physiology. LINKED ARTICLES This article is part of a themed section on Molecular Pharmacology of GPCRs. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.21/issuetoc.
Collapse
Affiliation(s)
- Margaret A Mouat
- Molecular Pharmacology Laboratory, Division of Molecular Cardiology and Biophysics, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, University of New South Wales, NSW, Australia
| | - James L J Coleman
- Molecular Pharmacology Laboratory, Division of Molecular Cardiology and Biophysics, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, University of New South Wales, NSW, Australia
| | - Nicola J Smith
- Molecular Pharmacology Laboratory, Division of Molecular Cardiology and Biophysics, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, University of New South Wales, NSW, Australia
| |
Collapse
|
32
|
Physiological and pathophysiological implications of PGE2 and the PGE2 synthases in the kidney. Prostaglandins Other Lipid Mediat 2018; 134:1-6. [DOI: 10.1016/j.prostaglandins.2017.10.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 10/09/2017] [Accepted: 10/31/2017] [Indexed: 12/16/2022]
|
33
|
Muthukaman N, Tambe M, Deshmukh S, Pisal D, Tondlekar S, Shaikh M, Sarode N, Kattige VG, Pisat M, Sawant P, Honnegowda S, Karande V, Kulkarni A, Behera D, Jadhav SB, Sangana RR, Gudi GS, Khairatkar-Joshi N, Gharat LA. Discovery of furan and dihydrofuran-fused tricyclic benzo[d]imidazole derivatives as potent and orally efficacious microsomal prostaglandin E synthase-1 (mPGES-1) inhibitors: Part-1. Bioorg Med Chem Lett 2017; 27:5131-5138. [PMID: 29100801 DOI: 10.1016/j.bmcl.2017.10.062] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 10/18/2017] [Accepted: 10/25/2017] [Indexed: 01/28/2023]
Abstract
This letter describes the synthesis and biological evaluation of furan and dihydrofuran-fused tricyclic benzo[d]imidazole derivatives as novel mPGES-1 inhibitors, capable of inhibiting an increased PGE2 production in the disease state. Structure-activity optimization afforded many potent mPGES-1 inhibitors having <50 nM potencies in the A549 cellular assay and adequate metabolic stability in liver microsomes. Lead compounds 8l and 8m demonstrated reasonable in vitro pharmacology and pharmacokinetic properties over other compounds. In particular, 8m revealed satisfactory oral pharmacokinetics and bioavailability in multiple species like rat, guinea pig, dog and cynomolgus monkey. In addition, the representative compound 8m showed in vivo efficacy by inhibiting LPS-induced thermal hyperalgesia with an ED50 of 14.3 mg/kg in guinea pig.
Collapse
Affiliation(s)
- Nagarajan Muthukaman
- Chemical Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Macchindra Tambe
- Chemical Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Sanjay Deshmukh
- Chemical Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Dnyandeo Pisal
- Chemical Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Shital Tondlekar
- Chemical Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Mahamadhanif Shaikh
- Chemical Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Neelam Sarode
- Chemical Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Vidya G Kattige
- Biological Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Monali Pisat
- Biological Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Pooja Sawant
- Biological Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Srinivasa Honnegowda
- Biological Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Vikas Karande
- Biological Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Abhay Kulkarni
- Biological Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Dayanidhi Behera
- Drug Metabolism and Pharmacokinetics, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Satyawan B Jadhav
- Drug Metabolism and Pharmacokinetics, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Ramchandra R Sangana
- Drug Metabolism and Pharmacokinetics, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Girish S Gudi
- Drug Metabolism and Pharmacokinetics, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Neelima Khairatkar-Joshi
- Biological Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Laxmikant A Gharat
- Chemical Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India.
| |
Collapse
|
34
|
Li Y, Wei Y, Zheng F, Guan Y, Zhang X. Prostaglandin E2 in the Regulation of Water Transport in Renal Collecting Ducts. Int J Mol Sci 2017; 18:ijms18122539. [PMID: 29186911 PMCID: PMC5751142 DOI: 10.3390/ijms18122539] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 11/16/2017] [Accepted: 11/18/2017] [Indexed: 01/26/2023] Open
Abstract
The kidney plays a central role in the regulation of the body water balance. The process of targeting the water channel aquaporin-2 (AQP2) on the apical plasma membrane of the collecting duct (CD) principal cells is mainly regulated by the antidiuretic peptide hormone arginine vasopressin (AVP), which is responsible for the maintenance of water homeostasis. Recently, much attention has been focused on the local factors modulating renal water reabsorption by AQP2 in the collecting ducts, especially prostaglandin E2 (PGE₂). PGE₂ is a lipid mediator involved in a variety of physiological and pathophysiological processes in the kidney. The biological function of PGE₂ is mainly mediated by four G-protein-coupled receptors, namely EP1-4, which couple to drive separate intracellular signaling pathways. Increasing evidence demonstrates that PGE₂ is essential for renal water transport regulation via multiple mechanisms. Each EP receptor plays a unique role in regulating water reabsorption in renal collecting ducts. This brief review highlights the role of PGE₂ in the regulation of water reabsorption and discusses the involvement of each EP receptor subtype in renal collecting duct. A better understanding of the role of PGE₂ in renal water transport process may improve disease management strategies for water balance disorders, including nephrogenic diabetes insipidus.
Collapse
Affiliation(s)
- Yuyuan Li
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China.
| | - Yuanyi Wei
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China.
| | - Feng Zheng
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China.
| | - Youfei Guan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China.
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China.
- AstraZeneca-Shenzhen University Joint Institute of Nephrology, Shenzhen University Health Science Center, Shenzhen 518060, China.
| | - Xiaoyan Zhang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China.
- AstraZeneca-Shenzhen University Joint Institute of Nephrology, Shenzhen University Health Science Center, Shenzhen 518060, China.
| |
Collapse
|
35
|
Milano S, Carmosino M, Gerbino A, Svelto M, Procino G. Hereditary Nephrogenic Diabetes Insipidus: Pathophysiology and Possible Treatment. An Update. Int J Mol Sci 2017; 18:ijms18112385. [PMID: 29125546 PMCID: PMC5713354 DOI: 10.3390/ijms18112385] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 11/03/2017] [Accepted: 11/07/2017] [Indexed: 12/17/2022] Open
Abstract
Under physiological conditions, excessive loss of water through the urine is prevented by the release of the antidiuretic hormone arginine-vasopressin (AVP) from the posterior pituitary. In the kidney, AVP elicits a number of cellular responses, which converge on increasing the osmotic reabsorption of water in the collecting duct. One of the key events triggered by the binding of AVP to its type-2 receptor (AVPR2) is the exocytosis of the water channel aquaporin 2 (AQP2) at the apical membrane the principal cells of the collecting duct. Mutations of either AVPR2 or AQP2 result in a genetic disease known as nephrogenic diabetes insipidus, which is characterized by the lack of responsiveness of the collecting duct to the antidiuretic action of AVP. The affected subject, being incapable of concentrating the urine, presents marked polyuria and compensatory polydipsia and is constantly at risk of severe dehydration. The molecular bases of the disease are fully uncovered, as well as the genetic or clinical tests for a prompt diagnosis of the disease in newborns. A real cure for nephrogenic diabetes insipidus (NDI) is still missing, and the main symptoms of the disease are handled with s continuous supply of water, a restrictive diet, and nonspecific drugs. Unfortunately, the current therapeutic options are limited and only partially beneficial. Further investigation in vitro or using the available animal models of the disease, combined with clinical trials, will eventually lead to the identification of one or more targeted strategies that will improve or replace the current conventional therapy and grant NDI patients a better quality of life. Here we provide an updated overview of the genetic defects causing NDI, the most recent strategies under investigation for rescuing the activity of mutated AVPR2 or AQP2, or for bypassing defective AVPR2 signaling and restoring AQP2 plasma membrane expression.
Collapse
Affiliation(s)
- Serena Milano
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy.
| | - Monica Carmosino
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy.
| | - Andrea Gerbino
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy.
| | - Maria Svelto
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy.
| | - Giuseppe Procino
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy.
| |
Collapse
|
36
|
Beharry KD, Cai CL, Soontarapornchai K, Ahmad T, Valencia GB, Aranda JV. Intermittent hypoxia alters dose dependent caffeine effects on renal prostanoids and receptors in neonatal rats. Prostaglandins Other Lipid Mediat 2017; 134:57-65. [PMID: 29107023 DOI: 10.1016/j.prostaglandins.2017.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 10/02/2017] [Accepted: 10/18/2017] [Indexed: 12/20/2022]
Abstract
Caffeine, one of the most commonly prescribed drugs in preterm neonates, is given in standard or suprapharmacologic doses. Although known as a diuretic, its effects in the neonatal kidneys are not well studied. We tested the hypothesis that neonatal intermittent hypoxia (IH) and high caffeine doses (HCD) alter renal regulators of vasomotor tone and water balance. Newborn rats were randomized to room air, hyperoxia, or IH and treated with standard or high caffeine doses; or placebo saline. Renal prostanoids; histopathology; and cyclooxygenase (COX), prostanoid receptor, and aquaporin (AQP) immunoreactivity were determined. HCD in IH caused severe pathological changes in the glomeruli and proximal tubules, consistent with acute kidney injury. This was associated with reductions in anthropometric growth, PGI2, and IP, DP, and AQP-4 immunoreactivity, well as a robust increase in COX-2, suggesting that the use of HCD should be avoided in preterm infants who experience frequent IH episodes.
Collapse
Affiliation(s)
- Kay D Beharry
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, USA; Department of Ophthalmology, State University of New York, Downstate Medical Center, Brooklyn, NY, USA; SUNY Eye Institute, New York, NY, USA.
| | - Charles L Cai
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, USA
| | - Kultida Soontarapornchai
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, USA
| | - Taimur Ahmad
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, USA
| | - Gloria B Valencia
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, USA
| | - Jacob V Aranda
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, USA; Department of Ophthalmology, State University of New York, Downstate Medical Center, Brooklyn, NY, USA; SUNY Eye Institute, New York, NY, USA
| |
Collapse
|
37
|
de Souza P, da Silva LM, Boeing T, Somensi LB, Cechinel-Zanchett CC, Campos A, Krueger CDMA, Bastos JK, Cechinel-Filho V, Andrade SFD. Influence of Prostanoids in the Diuretic and Natriuretic Effects of Extracts and Kaempferitrin fromBauhinia forficataLink Leaves in Rats. Phytother Res 2017; 31:1521-1528. [DOI: 10.1002/ptr.5876] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 06/30/2017] [Accepted: 07/03/2017] [Indexed: 01/24/2023]
Affiliation(s)
- Priscila de Souza
- Programa de Pós-Graduação em Ciências Farmacêuticas, Núcleo de Investigações Químico-Farmacêuticas (NIQFAR); Universidade do Vale do Itajaí (UNIVALI); 88302-901 Itajaí SC Brazil
| | - Luisa Mota da Silva
- Programa de Pós-Graduação em Ciências Farmacêuticas, Núcleo de Investigações Químico-Farmacêuticas (NIQFAR); Universidade do Vale do Itajaí (UNIVALI); 88302-901 Itajaí SC Brazil
| | - Thaise Boeing
- Programa de Pós-Graduação em Ciências Farmacêuticas, Núcleo de Investigações Químico-Farmacêuticas (NIQFAR); Universidade do Vale do Itajaí (UNIVALI); 88302-901 Itajaí SC Brazil
| | - Lincon Bordignon Somensi
- Programa de Pós-Graduação em Ciências Farmacêuticas, Núcleo de Investigações Químico-Farmacêuticas (NIQFAR); Universidade do Vale do Itajaí (UNIVALI); 88302-901 Itajaí SC Brazil
| | - Camile Cecconi Cechinel-Zanchett
- Programa de Pós-Graduação em Ciências Farmacêuticas, Núcleo de Investigações Químico-Farmacêuticas (NIQFAR); Universidade do Vale do Itajaí (UNIVALI); 88302-901 Itajaí SC Brazil
| | - Adriana Campos
- Programa de Pós-Graduação em Ciências Farmacêuticas, Núcleo de Investigações Químico-Farmacêuticas (NIQFAR); Universidade do Vale do Itajaí (UNIVALI); 88302-901 Itajaí SC Brazil
| | - Clarissa de Medeiros Amorim Krueger
- Programa de Pós-Graduação em Ciências Farmacêuticas, Núcleo de Investigações Químico-Farmacêuticas (NIQFAR); Universidade do Vale do Itajaí (UNIVALI); 88302-901 Itajaí SC Brazil
| | - Jairo Kenupp Bastos
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto; Universidade de São Paulo (USP); 14040-903 Ribeirão Preto SP Brazil
| | - Valdir Cechinel-Filho
- Programa de Pós-Graduação em Ciências Farmacêuticas, Núcleo de Investigações Químico-Farmacêuticas (NIQFAR); Universidade do Vale do Itajaí (UNIVALI); 88302-901 Itajaí SC Brazil
| | - Sérgio Faloni de Andrade
- Programa de Pós-Graduação em Ciências Farmacêuticas, Núcleo de Investigações Químico-Farmacêuticas (NIQFAR); Universidade do Vale do Itajaí (UNIVALI); 88302-901 Itajaí SC Brazil
| |
Collapse
|
38
|
Schlickmann F, de Souza P, Boeing T, Mariano LNB, Steimbach VMB, Krueger CDMA, da Silva LM, de Andrade SF, Cechinel-Filho V. Chemical composition and diuretic, natriuretic and kaliuretic effects of extracts of Mimosa bimucronata (DC.) Kuntze leaves and its majority constituent methyl gallate in rats. J Pharm Pharmacol 2017; 69:1615-1624. [DOI: 10.1111/jphp.12785] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 06/18/2017] [Indexed: 01/28/2023]
Abstract
Abstract
Objectives
Some species of the genus Mimosa showed promising results in previous investigations, which include diuretic effect; however, no chemical analyses or animal model has been conducted so far to evaluate the biological properties of M. bimucronata.
Methods
Male Wistar rats received the oral treatment with vehicle; hydrochlorothiazide; methanolic extract from M. bimucronata (MEMB), dichloromethane (DCM) and ethyl acetate (EA) fractions or methyl gallate (MG). The cumulative urine volume, electrolytes excretion, pH and osmolality were determined at the end of the experiment.
Key findings
The chemical studies demonstrated that the phenolic compounds are the majorities in the plant, with the MG being the main substance identified. We showed that MEMB and EA fraction, but not DCM, exhibited diuretic and saluretic effects. Similarly, the MG also revealed diuretic, natriuretic and kaliuretic properties to both normotensive and spontaneously hypertensive rats. Atropine, a muscarinic receptor antagonist, fully prevented MG-induced diuresis and saluresis. In addition, MG did not alter the viability of A7r5 and L929 cell lines and neither stimulated nitric oxide generation.
Conclusions
These findings suggest that M. bimucronata extracts and its majority compound MG present diuretic, natriuretic and kaliuretic properties, which was dependent on the activation of muscarinic acetylcholine receptor.
Collapse
Affiliation(s)
- Fabile Schlickmann
- Programa de Pós-Graduação em Ciências Farmacêuticas, Núcleo de Investigações Químico-Farmacêuticas (NIQFAR), Universidade do Vale do Itajaí (UNIVALI), Itajaí, SC, Brazil
| | - Priscila de Souza
- Programa de Pós-Graduação em Ciências Farmacêuticas, Núcleo de Investigações Químico-Farmacêuticas (NIQFAR), Universidade do Vale do Itajaí (UNIVALI), Itajaí, SC, Brazil
| | - Thaise Boeing
- Programa de Pós-Graduação em Ciências Farmacêuticas, Núcleo de Investigações Químico-Farmacêuticas (NIQFAR), Universidade do Vale do Itajaí (UNIVALI), Itajaí, SC, Brazil
| | - Luisa N B Mariano
- Programa de Pós-Graduação em Ciências Farmacêuticas, Núcleo de Investigações Químico-Farmacêuticas (NIQFAR), Universidade do Vale do Itajaí (UNIVALI), Itajaí, SC, Brazil
| | - Viviane M B Steimbach
- Programa de Pós-Graduação em Ciências Farmacêuticas, Núcleo de Investigações Químico-Farmacêuticas (NIQFAR), Universidade do Vale do Itajaí (UNIVALI), Itajaí, SC, Brazil
| | - Clarissa de M A Krueger
- Programa de Pós-Graduação em Ciências Farmacêuticas, Núcleo de Investigações Químico-Farmacêuticas (NIQFAR), Universidade do Vale do Itajaí (UNIVALI), Itajaí, SC, Brazil
| | - Luísa M da Silva
- Programa de Pós-Graduação em Ciências Farmacêuticas, Núcleo de Investigações Químico-Farmacêuticas (NIQFAR), Universidade do Vale do Itajaí (UNIVALI), Itajaí, SC, Brazil
| | - Sérgio F de Andrade
- Programa de Pós-Graduação em Ciências Farmacêuticas, Núcleo de Investigações Químico-Farmacêuticas (NIQFAR), Universidade do Vale do Itajaí (UNIVALI), Itajaí, SC, Brazil
| | - Valdir Cechinel-Filho
- Programa de Pós-Graduação em Ciências Farmacêuticas, Núcleo de Investigações Químico-Farmacêuticas (NIQFAR), Universidade do Vale do Itajaí (UNIVALI), Itajaí, SC, Brazil
| |
Collapse
|
39
|
Chen D, Tang J, Wan Q, Zhang J, Wang K, Shen Y, Yu Y. E-Prostanoid 3 Receptor Mediates Sprouting Angiogenesis Through Suppression of the Protein Kinase A/β-Catenin/Notch Pathway. Arterioscler Thromb Vasc Biol 2017; 37:856-866. [DOI: 10.1161/atvbaha.116.308587] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 02/16/2017] [Indexed: 01/14/2023]
Abstract
Objective—
Angiogenesis is a hallmark of embryonic development and various ischemic and inflammatory diseases. Prostaglandin E2 receptor subtype 3 (EP3) plays an important role in pathophysiologic angiogenesis; however, the precise mechanisms remain unknown. Here, we investigated the role of EP3 in zebra fish embryo and mouse retina angiogenesis and evaluated the underlying mechanisms.
Approach and Results—
The EP3 receptor was highly expressed in the vasculature in both zebra fish embryos and murine fetal retinas. Pharmacological inhibition or genetic deletion of EP3 significantly reduced vasculature formation in zebra fish embryos and mouse retinas. Further characterization revealed reduced filopodia extension of tip cells in embryonic retinas in EP3-deficient mice. EP3 deletion activated Notch activity by upregulation of delta-like ligand 4 expression in endothelial cells (ECs). Inhibition of Notch signaling rescued the angiogenic defects in EP3-deficient mouse retinas. Moreover, EP3 deficiency led to a significant increase in β-catenin phosphorylation at Ser675 and nuclear accumulation of β-catenin in ECs. Knockdown or inhibition of β-catenin restored the impaired sprouting angiogenesis resulting from EP3 deficiency in ECs. The EP3 receptor depressed protein kinase A activity in ECs by coupling to Gαi. Inhibition of protein kinase A activity significantly reduced Ser675 phosphorylation and nuclear translocation of β-catenin, abolished the increased delta-like ligand 4 expression, and subsequently restored the impaired angiogenic capacity of EP3-deficient ECs both in vitro and in vivo.
Conclusions—
Activation of the EP3 receptor facilitates sprouting angiogenesis through protein kinase A–dependent Notch signaling, suggesting that EP3 and its downstream pathways maybe potential therapeutic targets in the treatment of ischemic diseases.
Collapse
Affiliation(s)
- Di Chen
- From the Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.C., J.Z., Y.S., Y.Y.); and Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China (D.C., J.T., Q.W., K.W., Y.Y.)
| | - Juan Tang
- From the Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.C., J.Z., Y.S., Y.Y.); and Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China (D.C., J.T., Q.W., K.W., Y.Y.)
| | - Qiangyou Wan
- From the Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.C., J.Z., Y.S., Y.Y.); and Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China (D.C., J.T., Q.W., K.W., Y.Y.)
| | - Jian Zhang
- From the Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.C., J.Z., Y.S., Y.Y.); and Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China (D.C., J.T., Q.W., K.W., Y.Y.)
| | - Kai Wang
- From the Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.C., J.Z., Y.S., Y.Y.); and Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China (D.C., J.T., Q.W., K.W., Y.Y.)
| | - Yujun Shen
- From the Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.C., J.Z., Y.S., Y.Y.); and Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China (D.C., J.T., Q.W., K.W., Y.Y.)
| | - Ying Yu
- From the Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.C., J.Z., Y.S., Y.Y.); and Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China (D.C., J.T., Q.W., K.W., Y.Y.)
| |
Collapse
|
40
|
Diuretic, natriuretic and potassium-sparing effect of nothofagin isolated from Leandra dasytricha (A. Gray) Cogn. leaves in normotensive and hypertensive rats. Chem Biol Interact 2017; 268:103-110. [DOI: 10.1016/j.cbi.2017.03.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 02/24/2017] [Accepted: 03/06/2017] [Indexed: 11/19/2022]
|
41
|
Yeung J, Holinstat M. Who is the real 12-HETrE? Prostaglandins Other Lipid Mediat 2017; 132:25-30. [PMID: 28259546 DOI: 10.1016/j.prostaglandins.2017.02.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 02/16/2017] [Accepted: 02/28/2017] [Indexed: 12/24/2022]
Abstract
Oxygenases, including lipoxygenases and cytochrome P450s, generate an array of structurally diverse oxylipins that modulate distinct biological responses in mammals. Depending on the source of tissues and enzymes, distinct oxylipins are generated with inherent cellular function. Here, we report structurally different forms of 12-HETrE, with distinct biological function in tissues as well as their derived enzymatic source.
Collapse
Affiliation(s)
- Jennifer Yeung
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, United States
| | - Michael Holinstat
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, United States; Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|
42
|
Hosono K, Isonaka R, Kawakami T, Narumiya S, Majima M. Signaling of Prostaglandin E Receptors, EP3 and EP4 Facilitates Wound Healing and Lymphangiogenesis with Enhanced Recruitment of M2 Macrophages in Mice. PLoS One 2016; 11:e0162532. [PMID: 27711210 PMCID: PMC5053515 DOI: 10.1371/journal.pone.0162532] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 08/24/2016] [Indexed: 01/09/2023] Open
Abstract
Lymphangiogenesis plays an important role in homeostasis, metabolism, and immunity, and also occurs during wound-healing. Here, we examined the roles of prostaglandin E2 (PGE2) receptor (EP) signaling in enhancement of lymphangiogenesis in wound healing processes. The hole-punch was made in the ears of male C57BL/6 mice using a metal ear punch. Healing process and lymphangiogenesis together with macrophage recruitment were analyzed in EP knockout mice. Lymphangiogenesis was up-regulated in the granulation tissues at the margins of punched-hole wounds in mouse ears, and this increase was accompanied by increased expression levels of COX-2 and microsomal prostaglandin E synthase-1. Administration of celecoxib, a COX-2 inhibitor, suppressed lymphangiogenesis in the granulation tissues and reduced the induction of the pro-lymphangiogenic factors, vascular endothelial growth factor (VEGF) -C and VEGF-D. Topical applications of selective EP receptor agonists enhanced the expressions of lymphatic vessel endothelial hyaluronan receptor-1 and VEGF receptor-3. The wound-healing processes and recruitment of CD11b-positive macrophages, which produced VEGF-C and VEGF-D, were suppressed under COX-2 inhibition. Mice lacking either EP3 or EP4 exhibited reduced wound-healing, lymphangiogenesis and recruitment of M2 macrophages, compared with wild type mice. Proliferation of cultured human lymphatic endothelial cells was not detected under PGE2 stimulation. Lymphangiogenesis and recruitment of M2 macrophages that produced VEGF-C/D were suppressed in mice treated with a COX-2 inhibitor or lacking either EP3 or EP4 during wound healing. COX-2 and EP3/EP4 signaling may be novel targets to control lymphangiogenesis in vivo.
Collapse
MESH Headings
- Animals
- CD11b Antigen/metabolism
- Cyclooxygenase 2/metabolism
- Cyclooxygenase 2 Inhibitors/pharmacology
- Ear/physiology
- Gene Knockout Techniques
- Lymphangiogenesis/drug effects
- Macrophages/cytology
- Macrophages/drug effects
- Macrophages/immunology
- Macrophages/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Prostaglandin-E Synthases/metabolism
- Receptors, Prostaglandin E, EP3 Subtype/deficiency
- Receptors, Prostaglandin E, EP3 Subtype/genetics
- Receptors, Prostaglandin E, EP3 Subtype/metabolism
- Receptors, Prostaglandin E, EP4 Subtype/deficiency
- Receptors, Prostaglandin E, EP4 Subtype/genetics
- Receptors, Prostaglandin E, EP4 Subtype/metabolism
- Signal Transduction/drug effects
- Up-Regulation/drug effects
- Vascular Endothelial Growth Factor C/biosynthesis
- Vascular Endothelial Growth Factor D/biosynthesis
- Wound Healing/drug effects
Collapse
Affiliation(s)
- Kanako Hosono
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Kanagawa, Japan
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
- Department of Physiology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Risa Isonaka
- Department of Physiology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Tadashi Kawakami
- Department of Physiology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Shuh Narumiya
- Center for Innovation in Immunoregulation Technology and Therapeutics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masataka Majima
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Kanagawa, Japan
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
- * E-mail:
| |
Collapse
|
43
|
Role of the prostaglandin E2 receptor agonists in TGF-β1-induced mesangial cell damage. Biosci Rep 2016; 36:BSR20160038. [PMID: 27512093 PMCID: PMC5041160 DOI: 10.1042/bsr20160038] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 08/10/2016] [Indexed: 02/05/2023] Open
Abstract
PGE2 exerts its biological effect through binding to various EP receptors that result inactivation of various signal transduction pathways. It also plays an important role in mice glomerular mesangial cells (MCs) damage induced by transforming growth factor-β1 (TGF-β1); however, the molecular mechanisms remain unknown. In the present study, we tested the efficacy of four selective agonists of PGE2 receptor, EP1A (17-phenyl trinor prostaglandin E2 ethyl amid), EP2A (butaprost), EP3A (sulprostone) and EP4A (cay10580), on mice MCs. Compared with the cAMP produced by TGF-β1, additional pretreatment of EP3A decreased the cAMP level. MCs treated with EP1A and EP3A augmented PGE2, cyclooxygenase-2 (COX-2), membrane-bound PGE synthase 1 (mPGES1), laminin (LN), connective tissue growth factor (CTGF) and cyclin D1 expression stimulated by TGFβ1. EP1A and EP3A increased the number of cells in S+G2/M phase and reduced cells in G0/G1 phase. EP1 and EP3 agonists also strengthened TGFβ1-induced mitogen-activated protein kinase (p38MAPK) and extracellular-signal-regulated kinase 1/2 (ERK1/2) phosphorylation. Whereas MCs treated with EP2A and EP4A weakened PGE2, COX-2, mPGES1, LN, CTGF and cyclin D1 expression stimulated by TGFβ1. EP2A and EP4A decreased the number of cells in S+G2/M phase and increased cells in G0/G1 phase. EP2 and EP4 agonists weakened TGFβ1-induced p38MAPK and ERK1/2 phosphorylation. These findings suggest that PGE2 has an important role in the progression of kidney disease via the EP1/EP3 receptor, whereas EP2 and EP4 receptors are equally important in preserving the progression of chronic kidney failure. Thus, agonists of EP2 and EP4 receptors may provide a basis for treating kidney damage induced by TGF-β1.
Collapse
|
44
|
Prieto-García L, Pericacho M, Sancho-Martínez SM, Sánchez Á, Martínez-Salgado C, López-Novoa JM, López-Hernández FJ. Mechanisms of triple whammy acute kidney injury. Pharmacol Ther 2016; 167:132-145. [PMID: 27490717 DOI: 10.1016/j.pharmthera.2016.07.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 07/16/2016] [Indexed: 12/26/2022]
Abstract
Pre-renal acute kidney injury (AKI) results from glomerular haemodynamic alterations leading to reduced glomerular filtration rate (GFR) with no parenchymal compromise. Renin-angiotensin system inhibitors, such as angiotensin-converting enzyme inhibitors (ACEIs), angiotensin receptor antagonists (ARAs), non-steroidal anti-inflammatory drugs (NSAIDs) and diuretics, are highly prescribed drugs that are frequently administered together. Double and triple associations have been correlated with increased pre-renal AKI incidence, termed "double whammy" and "triple whammy", respectively. This article presents an integrative analysis of the complex interplay among the effects of NSAIDs, ACEIs/ARAs and diuretics, acting alone and together in double and triple therapies. In addition, we explore how these drug combinations alter the equilibrium of regulatory mechanisms controlling blood pressure (renal perfusion pressure) and GFR to increase the odds of inducing AKI through the concomitant reduction of blood pressure and distortion of renal autoregulation. Using this knowledge, we propose a more general model of pre-renal AKI based on a multi whammy model, whereby several factors are necessary to effectively reduce net filtration. The triple whammy was the only model associated with pre-renal AKI accompanied by a course of other risk factors, among numerous potential combinations of clinical circumstances causing hypoperfusion in which renal autoregulation is not operative or is deregulated. These factors would uncouple the normal BP-GFR relationship, where lower GFR values are obtained at every BP value.
Collapse
Affiliation(s)
- Laura Prieto-García
- Instituto de Estudios de Ciencias de la Salud de Castilla y León-Instituto de Investigación Biomédica de Salamanca (IECSCYL-IBSAL), Paseo de San Vicente, 58-182 - Hospital Virgen Vega, Planta 10, 37007 Salamanca, Spain; Department of Physiology & Pharmacology, University of Salamanca, Salamanca, Spain; Instituto Reina Sofía de Investigación Nefrológica, Fundación Iñigo Álvarez de Toledo, Madrid, Spain; Group of Biomedical Research in Critical Care Medicine (BioCritic), Hospital Clínico Universitario de Valladolid, Valladolid, Spain; Group of Theranostics for Renal and Cardiovascular Diseases (TERCARD), Edificio Departamental, Campus Miguel de Unamuno, Salamanca, Spain
| | - Miguel Pericacho
- Instituto de Estudios de Ciencias de la Salud de Castilla y León-Instituto de Investigación Biomédica de Salamanca (IECSCYL-IBSAL), Paseo de San Vicente, 58-182 - Hospital Virgen Vega, Planta 10, 37007 Salamanca, Spain; Department of Physiology & Pharmacology, University of Salamanca, Salamanca, Spain; Instituto Reina Sofía de Investigación Nefrológica, Fundación Iñigo Álvarez de Toledo, Madrid, Spain
| | - Sandra M Sancho-Martínez
- Department of Physiology & Pharmacology, University of Salamanca, Salamanca, Spain; Instituto Reina Sofía de Investigación Nefrológica, Fundación Iñigo Álvarez de Toledo, Madrid, Spain; Group of Biomedical Research in Critical Care Medicine (BioCritic), Hospital Clínico Universitario de Valladolid, Valladolid, Spain; Group of Theranostics for Renal and Cardiovascular Diseases (TERCARD), Edificio Departamental, Campus Miguel de Unamuno, Salamanca, Spain
| | - Ángel Sánchez
- Instituto de Estudios de Ciencias de la Salud de Castilla y León-Instituto de Investigación Biomédica de Salamanca (IECSCYL-IBSAL), Paseo de San Vicente, 58-182 - Hospital Virgen Vega, Planta 10, 37007 Salamanca, Spain; Hospital Universitario de Salamanca, Unidad de Hipertensión, Salamanca, Spain
| | - Carlos Martínez-Salgado
- Instituto de Estudios de Ciencias de la Salud de Castilla y León-Instituto de Investigación Biomédica de Salamanca (IECSCYL-IBSAL), Paseo de San Vicente, 58-182 - Hospital Virgen Vega, Planta 10, 37007 Salamanca, Spain; Department of Physiology & Pharmacology, University of Salamanca, Salamanca, Spain; Instituto Reina Sofía de Investigación Nefrológica, Fundación Iñigo Álvarez de Toledo, Madrid, Spain; Group of Biomedical Research in Critical Care Medicine (BioCritic), Hospital Clínico Universitario de Valladolid, Valladolid, Spain; Group of Theranostics for Renal and Cardiovascular Diseases (TERCARD), Edificio Departamental, Campus Miguel de Unamuno, Salamanca, Spain
| | - José Miguel López-Novoa
- Instituto de Estudios de Ciencias de la Salud de Castilla y León-Instituto de Investigación Biomédica de Salamanca (IECSCYL-IBSAL), Paseo de San Vicente, 58-182 - Hospital Virgen Vega, Planta 10, 37007 Salamanca, Spain; Department of Physiology & Pharmacology, University of Salamanca, Salamanca, Spain; Instituto Reina Sofía de Investigación Nefrológica, Fundación Iñigo Álvarez de Toledo, Madrid, Spain; Group of Biomedical Research in Critical Care Medicine (BioCritic), Hospital Clínico Universitario de Valladolid, Valladolid, Spain; Group of Theranostics for Renal and Cardiovascular Diseases (TERCARD), Edificio Departamental, Campus Miguel de Unamuno, Salamanca, Spain
| | - Francisco J López-Hernández
- Instituto de Estudios de Ciencias de la Salud de Castilla y León-Instituto de Investigación Biomédica de Salamanca (IECSCYL-IBSAL), Paseo de San Vicente, 58-182 - Hospital Virgen Vega, Planta 10, 37007 Salamanca, Spain; Department of Physiology & Pharmacology, University of Salamanca, Salamanca, Spain; Instituto Reina Sofía de Investigación Nefrológica, Fundación Iñigo Álvarez de Toledo, Madrid, Spain; Group of Biomedical Research in Critical Care Medicine (BioCritic), Hospital Clínico Universitario de Valladolid, Valladolid, Spain; Group of Theranostics for Renal and Cardiovascular Diseases (TERCARD), Edificio Departamental, Campus Miguel de Unamuno, Salamanca, Spain.
| |
Collapse
|
45
|
Yang T, Li C. Role of COX-2 in unilateral ureteral obstruction: what is new? Am J Physiol Renal Physiol 2016; 310:F746-F747. [PMID: 26661655 DOI: 10.1152/ajprenal.00498.2015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 12/02/2015] [Indexed: 11/22/2022] Open
Affiliation(s)
- Tianxin Yang
- Department of Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah; and .,Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chunling Li
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
46
|
Morla L, Edwards A, Crambert G. New insights into sodium transport regulation in the distal nephron: Role of G-protein coupled receptors. World J Biol Chem 2016; 7:44-63. [PMID: 26981195 PMCID: PMC4768124 DOI: 10.4331/wjbc.v7.i1.44] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 10/02/2015] [Accepted: 11/25/2015] [Indexed: 02/05/2023] Open
Abstract
The renal handling of Na+ balance is a major determinant of the blood pressure (BP) level. The inability of the kidney to excrete the daily load of Na+ represents the primary cause of chronic hypertension. Among the different segments that constitute the nephron, those present in the distal part (i.e., the cortical thick ascending limb, the distal convoluted tubule, the connecting and collecting tubules) play a central role in the fine-tuning of renal Na+ excretion and are the target of many different regulatory processes that modulate Na+ retention more or less efficiently. G-protein coupled receptors (GPCRs) are crucially involved in this regulation and could represent efficient pharmacological targets to control BP levels. In this review, we describe both classical and novel GPCR-dependent regulatory systems that have been shown to modulate renal Na+ absorption in the distal nephron. In addition to the multiplicity of the GPCR that regulate Na+ excretion, this review also highlights the complexity of these different pathways, and the connections between them.
Collapse
|
47
|
Vukićević T, Schulz M, Faust D, Klussmann E. The Trafficking of the Water Channel Aquaporin-2 in Renal Principal Cells-a Potential Target for Pharmacological Intervention in Cardiovascular Diseases. Front Pharmacol 2016; 7:23. [PMID: 26903868 PMCID: PMC4749865 DOI: 10.3389/fphar.2016.00023] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 01/25/2016] [Indexed: 01/13/2023] Open
Abstract
Arginine-vasopressin (AVP) stimulates the redistribution of water channels, aquaporin-2 (AQP2) from intracellular vesicles into the plasma membrane of renal collecting duct principal cells. By this AVP directs 10% of the water reabsorption from the 170 L of primary urine that the human kidneys produce each day. This review discusses molecular mechanisms underlying the AVP-induced redistribution of AQP2; in particular, it provides an overview over the proteins participating in the control of its localization. Defects preventing the insertion of AQP2 into the plasma membrane cause diabetes insipidus. The disease can be acquired or inherited, and is characterized by polyuria and polydipsia. Vice versa, up-regulation of the system causing a predominant localization of AQP2 in the plasma membrane leads to excessive water retention and hyponatremia as in the syndrome of inappropriate antidiuretic hormone secretion (SIADH), late stage heart failure or liver cirrhosis. This article briefly summarizes the currently available pharmacotherapies for the treatment of such water balance disorders, and discusses the value of newly identified mechanisms controlling AQP2 for developing novel pharmacological strategies. Innovative concepts for the therapy of water balance disorders are required as there is a medical need due to the lack of causal treatments.
Collapse
Affiliation(s)
- Tanja Vukićević
- Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association Berlin, Germany
| | - Maike Schulz
- Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association Berlin, Germany
| | - Dörte Faust
- Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association Berlin, Germany
| | - Enno Klussmann
- Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz AssociationBerlin, Germany; German Centre for Cardiovascular ResearchBerlin, Germany
| |
Collapse
|
48
|
Nørregaard R, Kwon TH, Frøkiær J. Physiology and pathophysiology of cyclooxygenase-2 and prostaglandin E2 in the kidney. Kidney Res Clin Pract 2015; 34:194-200. [PMID: 26779421 PMCID: PMC4688592 DOI: 10.1016/j.krcp.2015.10.004] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 10/13/2015] [Indexed: 01/12/2023] Open
Abstract
The cyclooxygenase (COX) enzyme system is the major pathway catalyzing the conversion of arachidonic acid into prostaglandins (PGs). PGs are lipid mediators implicated in a variety of physiological and pathophysiological processes in the kidney, including renal hemodynamics, body water and sodium balance, and the inflammatory injury characteristic in multiple renal diseases. Since the beginning of 1990s, it has been confirmed that COX exists in 2 isoforms, referred to as COX-1 and COX-2. Even though the 2 enzymes are similar in size and structure, COX-1 and COX-2 are regulated by different systems and have different functional roles. This review summarizes the current data on renal expression of the 2 COX isoforms and highlights mainly the role of COX-2 and PGE2 in several physiological and pathophysiological processes in the kidney.
Collapse
Affiliation(s)
- Rikke Nørregaard
- Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Tae-Hwan Kwon
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Korea
| | - Jørgen Frøkiær
- Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
49
|
Vilums M, Heuberger J, Heitman LH, IJzerman AP. Indanes--Properties, Preparation, and Presence in Ligands for G Protein Coupled Receptors. Med Res Rev 2015; 35:1097-126. [PMID: 26018667 DOI: 10.1002/med.21352] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The indane (2,3-dihydro-1H-indene) ring system is an attractive scaffold for biologically active compounds due to the combination of aromatic and aliphatic properties fused together in one rigid system. This bicyclic structure provides a wide range of possibilities to incorporate specific substituents in different directionalities, thus being an attractive scaffold for medicinal chemists. Notably, many indane-based compounds are being used in the clinic to treat various diseases, such as indinavir, an HIV-1 protease inhibitor; indantadol, a potent Monoamine Oxidase (MAO)-inhibitor; the amine uptake inhibitor indatraline; and the ultra-long-acting β-adrenoceptor agonist indacaterol. Given the diversity of targets these drugs act on, one could argue that the indane ring system is a privileged substructure. In the present review, the synthetic and medicinal chemistry of the indane ring system is described. In more detail, it contains a comprehensive overview of compounds bearing the indane substructure with G protein coupled receptor (GPCR) activity, with particular emphasis on their structure-activity relationships (SAR).
Collapse
Affiliation(s)
- Maris Vilums
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - Jules Heuberger
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - Laura H Heitman
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - Adriaan P IJzerman
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| |
Collapse
|
50
|
Nitrooleic acid protects against cisplatin nephropathy: role of COX-2/mPGES-1/PGE2 cascade. Mediators Inflamm 2015; 2015:293474. [PMID: 25861160 PMCID: PMC4377489 DOI: 10.1155/2015/293474] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 01/16/2015] [Accepted: 01/21/2015] [Indexed: 01/20/2023] Open
Abstract
Nitrooleic acid (OA-NO2) is an endogenous lipid product which has novel signaling properties, particularly the activation of peroxisome proliferator-activated receptors. The current study aimed to evaluate the protective effects of OA-NO2 against cisplatin-induced kidney injury in mice. Mice were pretreated with OA-NO2 for 48 h before cisplatin administration, and the cisplatin-caused nephrotoxicity was evaluated. After the cisplatin treatment (72 h), the vehicle-treated mice displayed renal dysfunction, as evidenced by the elevated plasma urea and creatinine, which was consistent with the histological damage, such as tubular necrosis, dilation, protein cast, and desquamation of epithelial cells. In contrast, the severity of the renal dysfunction and histological change were reduced in the OA-NO2 pretreated mice. The renal COX-2 and mPGES-1 mRNAs and their respective proteins expression, together with the renal PGE2 amounts, were induced by the cisplatin treatment, but their initiation was reduced by OA-NO2. Moreover, the circulating TNF-α, renal TNF-α, IL-1β, MCP-1, ICAM-1, and VACAM-1 mRNA levels were higher in the cisplatin-treated mice, compared with the controls, but they were attenuated in the OA-NO2 pretreatment group. In summary, the pretreatment with OA-NO2 remarkably ameliorated the cisplatin-induced kidney injury in mice, possibly via the inhibition of the inflammatory response, associated with the COX-2/mPGES-1/PGE2 cascade.
Collapse
|