1
|
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
|
2
|
Chen X, Delić D, Cao Y, Zhang Z, Wu H, Hasan AA, Gaballa MMS, Yin L, Krämer BK, Klein T, Shi X, He B, Shen L, Hocher B. Renal and cardiac effects of the PDE9 inhibitor BAY 73-6691 in 5/6 nephrectomized rats. Pflugers Arch 2024; 476:755-767. [PMID: 38305876 DOI: 10.1007/s00424-024-02915-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/15/2024] [Accepted: 01/23/2024] [Indexed: 02/03/2024]
Abstract
It has been suggested that the novel selective phosphodiesterase 9 (PDE9) inhibitor may improve cardiac and renal function by blocking 3',5'-cyclic guanosine monophosphate (cGMP) degradation. 5/6 nephrectomized (5/6Nx) rats were used to investigate the effects of the PDE9 inhibitor (BAY 73-6691) on the heart and kidney. Two doses of BAY 73-6691 (1 mg/kg/day and 5 mg/kg/day) were given for 95 days. The 5/6Nx rats developed albuminuria, a decrease in serum creatinine clearance (Ccr), and elevated serum troponin T levels. Echocardiographic data showed that 5/6 nephrectomy resulted in increased fractional shortening (FS), stroke volume (SV), and left ventricular ejection fraction (EF). However, 95 days of PDE9 inhibitor treatment did not improve any cardiac and renal functional parameter. Histopathologically, 5/6 nephrectomy resulted in severe kidney and heart damage, such as renal interstitial fibrosis, glomerulosclerosis, and enlarged cardiomyocytes. Telmisartan attenuated renal interstitial fibrosis and glomerulosclerosis as well as improved cardiomyocyte size. However, except for cardiomyocyte size and renal perivascular fibrosis, BAY 73-6691 had no effect on other cardiac and renal histologic parameters. Pathway enrichment analysis using RNA sequencing data of kidney and heart tissue identified chronic kidney disease pathways, such as phosphatidylinositol 3-kinase (PI3K)-protein kinase B (Akt) signaling pathway, complement and coagulation cascades, and nuclear factor kappa B (NF-κB) signaling pathway. PDE9i did not affect any of these disease-related pathways. Two dosages of the PDE9 inhibitor BAY 73-6691 known to be effective in other rat models have only limited cardio-renal protective effects in 5/6 nephrectomized rats.
Collapse
Affiliation(s)
- Xin Chen
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
- Department of Nephrology, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
- The First Clinical Medical College of Jinan University, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Denis Delić
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
- Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorferstr.65, 88397, Biberach, Germany
| | - Yaochen Cao
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
- Department of Nephrology, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
| | - Zeyu Zhang
- The First Clinical Medical College of Jinan University, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Hongwei Wu
- The First Clinical Medical College of Jinan University, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Ahmed A Hasan
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | | | - Lianghong Yin
- The First Clinical Medical College of Jinan University, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Bernhard K Krämer
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
- European Center for Angioscience, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Thomas Klein
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88397, Biberach, Germany
| | - Xin Shi
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Linghong Shen
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Berthold Hocher
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany.
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, China.
- IMD Institut Für Medizinische Diagnostik Berlin-Potsdam GbR, Berlin, Germany.
| |
Collapse
|
3
|
Kubohara Y, Fukunaga Y, Shigenaga A, Kikuchi H. Dictyostelium Differentiation-Inducing Factor 1 Promotes Glucose Uptake via Direct Inhibition of Mitochondrial Malate Dehydrogenase in Mouse 3T3-L1 Cells. Int J Mol Sci 2024; 25:1889. [PMID: 38339168 PMCID: PMC10855897 DOI: 10.3390/ijms25031889] [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: 12/14/2023] [Revised: 01/25/2024] [Accepted: 02/02/2024] [Indexed: 02/12/2024] Open
Abstract
Differentiation-inducing factor 1 (DIF-1), found in Dictyostelium discoideum, has antiproliferative and glucose-uptake-promoting activities in mammalian cells. DIF-1 is a potential lead for the development of antitumor and/or antiobesity/antidiabetes drugs, but the mechanisms underlying its actions have not been fully elucidated. In this study, we searched for target molecules of DIF-1 that mediate the actions of DIF-1 in mammalian cells by identifying DIF-1-binding proteins in human cervical cancer HeLa cells and mouse 3T3-L1 fibroblast cells using affinity chromatography and liquid chromatography-tandem mass spectrometry and found mitochondrial malate dehydrogenase (MDH2) to be a DIF-1-binding protein in both cell lines. Since DIF-1 has been shown to directly inhibit MDH2 activity, we compared the effects of DIF-1 and the MDH2 inhibitor LW6 on the growth of HeLa and 3T3-L1 cells and on glucose uptake in confluent 3T3-L1 cells in vitro. In both HeLa and 3T3-L1 cells, DIF-1 at 10-40 μM dose-dependently suppressed growth, whereas LW6 at 20 μM, but not at 2-10 μM, significantly suppressed growth in these cells. In confluent 3T3-L1 cells, DIF-1 at 10-40 μM significantly promoted glucose uptake, with the strongest effect at 20 μM DIF-1, whereas LW6 at 2-20 μM significantly promoted glucose uptake, with the strongest effect at 10 μM LW6. Western blot analyses showed that LW6 (10 μM) and DIF-1 (20 μM) phosphorylated and, thus, activated AMP kinase in 3T3-L1 cells. Our results suggest that MDH2 inhibition can suppress cell growth and promote glucose uptake in the cells, but appears to promote glucose uptake more strongly than it suppresses cell growth. Thus, DIF-1 may promote glucose uptake, at least in part, via direct inhibition of MDH2 and a subsequent activation of AMP kinase in 3T3-L1 cells.
Collapse
Affiliation(s)
- Yuzuru Kubohara
- Laboratory of Health and Life Science, Graduate School of Health and Sports Science, Juntendo University, Inzai 270-1695, Japan
| | - Yuko Fukunaga
- Department of Animal Risk Management, Faculty of Risk and Crisis Management, Chiba Institute of Science, Choshi 288-0025, Japan;
| | - Ayako Shigenaga
- Institute of Health and Sports Science & Medicine, Juntendo University, Inzai 270-1695, Japan;
| | - Haruhisa Kikuchi
- Division of Natural Medicines, Faculty of Pharmacy, Keio University, Tokyo 105-8512, Japan;
| |
Collapse
|
4
|
Rademaker MT, Scott NJA, Charles CJ, Richards AM. Combined Inhibition of Phosphodiesterase-5 and -9 in Experimental Heart Failure. JACC. HEART FAILURE 2024; 12:100-113. [PMID: 37921801 DOI: 10.1016/j.jchf.2023.08.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 08/08/2023] [Accepted: 08/31/2023] [Indexed: 11/04/2023]
Abstract
BACKGROUND Intracellular second messenger cyclic guanosine monophosphate (cGMP) mediates bioactivity of the natriuretic peptides and nitric oxide, and is key to circulatory homeostasis and protection against cardiovascular disease. Inhibition of cGMP-degrading phosphodiesterases (PDEs) PDE5 and PDE9 are emerging as pharmacological targets in heart failure (HF). OBJECTIVES The present study investigated dual enhancement of cGMP in experimental HF by combining inhibition of PDE-5 (P5-I) and PDE-9 (P9-I). METHODS Eight sheep with pacing-induced HF received on separate days intravenous P5-I (sildenafil), P9-I (PF-04749982), P5-I+P9-I, and vehicle control, in counterbalanced order. RESULTS Compared with control, separate P5-I and P9-I significantly increased circulating cGMP concentrations in association with reductions in mean arterial pressure (MAP), left atrial pressure (LAP), and pulmonary arterial pressure (PAP), with effects of P5-I on cGMP, MAP, and PAP greater than those of P9-I. Only P5-I decreased pulmonary vascular resistance. Combination P5-I+P9-I further reduced MAP, LAP, and PAP relative to inhibition of either phosphodiesterase alone. P9-I and, especially, P5-I elevated urinary cGMP levels relative to control. However, whereas inhibition of either enzyme increased urine creatinine excretion and clearance, only P9-I induced a significant diuresis and natriuresis. Combined P5-I+P9-I further elevated urine cGMP with concomitant increases in urine volume, sodium and creatinine excretion, and clearance similar to P9-I alone, despite the greater MAP reductions induced by combination treatment. CONCLUSIONS Combined P5-I+P9-I amalgamated the superior renal effects of P9-I and pulmonary effects of P5-1, while concurrently further reducing cardiac preload and afterload. These findings support combination P5-I+P9-I as a therapeutic strategy in HF.
Collapse
Affiliation(s)
- Miriam T Rademaker
- Christchurch Heart Institute, Department of Medicine, University of Otago-Christchurch, Christchurch, New Zealand.
| | - Nicola J A Scott
- Christchurch Heart Institute, Department of Medicine, University of Otago-Christchurch, Christchurch, New Zealand
| | - Christopher J Charles
- Christchurch Heart Institute, Department of Medicine, University of Otago-Christchurch, Christchurch, New Zealand
| | - A Mark Richards
- Christchurch Heart Institute, Department of Medicine, University of Otago-Christchurch, Christchurch, New Zealand; Cardiovascular Research Institute, National University of Singapore, Singapore
| |
Collapse
|
5
|
Xu Y, Qiu Y, Zhang Y, Li X. A cAMP phosphodiesterase is essential for sclerotia formation and virulence in Sclerotinia sclerotiorum. FRONTIERS IN PLANT SCIENCE 2023; 14:1175552. [PMID: 37324679 PMCID: PMC10264682 DOI: 10.3389/fpls.2023.1175552] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/12/2023] [Indexed: 06/17/2023]
Abstract
Sclerotinia sclerotiorum is a plant pathogenic fungus that causes white mold or stem rot diseases. It affects mostly dicotyledonous crops, resulting in significant economic losses worldwide. Sclerotia formation is a special feature of S. sclerotiorum, allowing its survival in soil for extended periods and facilitates the spread of the pathogen. However, the detailed molecular mechanisms of how sclerotia are formed and how virulence is achieved in S. sclerotiorum are not fully understood. Here, we report the identification of a mutant that cannot form sclerotia using a forward genetics approach. Next-generation sequencing of the mutant's whole genome revealed candidate genes. Through knockout experiments, the causal gene was found to encode a cAMP phosphodiesterase (SsPDE2). From mutant phenotypic examinations, we found that SsPDE2 plays essential roles not only in sclerotia formation, but also in the regulation of oxalic acid accumulation, infection cushion functionality and virulence. Downregulation of SsSMK1 transcripts in Sspde2 mutants revealed that these morphological defects are likely caused by cAMP-dependent inhibition of MAPK signaling. Moreover, when we introduced HIGS construct targeting SsPDE2 in Nicotiana benthamiana, largely compromised virulence was observed against S. sclerotiorum. Taken together, SsPDE2 is indispensable for key biological processes of S. sclerotiorum and can potentially serve as a HIGS target to control stem rot in the field.
Collapse
Affiliation(s)
- Yan Xu
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Botany, University of British Columbia, Vancouver, BC, Canada
| | - Yilan Qiu
- Department of Life Science, Hunan Normal University, Changsha, China
| | - Yuelin Zhang
- Department of Botany, University of British Columbia, Vancouver, BC, Canada
| | - Xin Li
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Botany, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
6
|
Exogenous H 2S Attenuates Hypertension by Regulating Renin Exocytosis under Hyperglycaemic and Hyperlipidaemic Conditions. Int J Mol Sci 2023; 24:ijms24021690. [PMID: 36675205 PMCID: PMC9860892 DOI: 10.3390/ijms24021690] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Obesity, along with type 2 diabetes mellitus (T2DM), is a major contributor to hypertension. The renin-angiotensin-aldosterone system is involved in the occurrence of diabetes and hypertension. However, the mechanism by which obesity is related to T2DM induced hypertension is unclear. In this study, we observed that blood pressure and serum renin content were increased in patients with diabetes and hypertension. Hydrogen sulfide (H2S), as an endogenous bioactive molecule, has been shown to be a vasodilator. Db/db mice, characterized by obesity and T2DM, and juxtaglomerular (JG) cells, which line the afferent arterioles at the entrance of the glomeruli to produce renin, treated with glucose, palmitic acid (PA) and oleic acid (OA), were used as animal and cellular models. NaHS, the H2S donor, was administered to db/db mice through intraperitoneal injection. NaHS significantly alleviated blood pressure in db/db mice, decreased the renin content in the serum of db/db mice and reduced renin secretion from JG cells. NaHS modulated renin release via cAMP and soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs), including synaptosome-associated protein 23 (SNAP23) and vesicle-associated membrane protein 2 (VAMP2), which mediate renin exocytosis. Furthermore, NaHS increased the levels of autophagy-related proteins and colocalization with EGFP-LC3 puncta with renin-containing granules and VAMP2 to consume excessive renin to maintain intracellular homeostasis. Therefore, exogenous H2S attenuates renin release and promotes renin-vesicular autophagy to relieve diabetes-induced hypertension.
Collapse
|
7
|
Augmentation of Natriuretic Peptide Bioactivity via Combined Inhibition of Neprilysin and Phosphodiesterase-9 in Heart Failure. JACC. HEART FAILURE 2022; 11:227-239. [PMID: 36752488 DOI: 10.1016/j.jchf.2022.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/25/2022] [Accepted: 11/10/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND The natriuretic peptides (NPs) are potent natriuretic/diuretic and vasodilatory factors, and augmentation of their levels or signaling via inhibition of the enzymes neprilysin (NEP) and phosphodiesterase 9 (PDE9), respectively, has beneficial actions in heart failure (HF). OBJECTIVES The authors investigated dual enhancement of NP bioactivity by combining PDE9 inhibition and NEP inhibition in HF using an ovine model. METHODS Eight sheep with pacing-induced HF received on 4 separate days intravenous PDE9 inhibition (PF-04749982), NEP inhibition (SCH-32615), PDE9 inhibition + NEP inhibition (PI+NI), and vehicle control treatment. RESULTS Compared with the control treatment, NEP inhibition significantly increased plasma NP concentrations with a corresponding rise in second messenger cyclic guanosine monophosphate (cGMP), whereas PDE9 inhibition increased circulating cGMP with a negligible effect on NP levels. Combined PI+NI elevated plasma NPs to an extent comparable to that seen with NEP inhibition alone but further increased cGMP, resulting in a rise in the cGMP-to-NP ratio. All active treatments reduced mean arterial pressure, left atrial pressure, pulmonary arterial pressure, and peripheral resistance, with combined PI+NI further reducing mean arterial pressure and left atrial pressure relative to either inhibitor separately. Active treatments increased urine volume and sodium, potassium and creatinine excretion, and creatinine clearance, in association with rises in urine cGMP levels. PI+NI induced a significantly greater natriuresis and increase in urinary cGMP relative to either inhibitor singly. CONCLUSIONS The present study demonstrates for the first time that combined PI+NI has additional beneficial hemodynamic and renal effects when compared with either PDE9 inhibition or NEP inhibition alone. The superior efficacy of this 2-pronged augmentation of NP bioactivity supports PI+NI as a potential therapeutic strategy for HF.
Collapse
|
8
|
Marinho AD, Coelho Jorge AR, Nogueira Junior FA, Alison de Moraes Silveira J, Rocha DG, Negreiros Nunes Alves AP, Ferreira RS, Bezerra Jorge RJ, Azul Monteiro HS. Effects of cilostazol, a Phosphodiesterase-3 inhibitor, on kidney function and redox imbalance in acute kidney injury caused by Bothrops alternatus venom. Toxicon 2022; 220:106922. [PMID: 36167141 DOI: 10.1016/j.toxicon.2022.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/28/2022]
Abstract
The mechanisms of pathogenesis of acute kidney injury (AKI) in snakebites is multifactorial and involves hemodynamic disturbances, with release of free radical causing cytotoxic effects. The phosphodiesterase-3 (PDE3) inhibitor, Cilostazol, has been reported to provide protection against renal oxidative stress. OBJECTIVE We evaluated the protective effects of cilostazol against Bothrops alternatus snake venom (BaV)-induced nephrotoxicity. METHODS Wistar rat kidneys (n = 6, 260-300 g) were isolated and perfused with Krebs-Henseleit solution containing 6 g/100 mL of bovine serum albumin. After 30 min, the kidneys were perfused with BaV to a final concentration of 1 and 3 μg/mL, and subsequently evaluated for perfusion pressure (PP), renal vascular resistance (RVR), urinary flow (UF), glomerular filtration rate (GFR), and percentage of electrolyte tubular sodium and chloride transport (%TNa+, %TCl-). Oxidative stress and renal histological analyses were performed. RESULTS BaV caused a reduction in all the evaluated renal parameters (PP, RVR, GFR, UF, %TNa+, and %TCl-). Although only the effects on PP and UF were reversed with cilostazol treatment, the decrease in the malondialdehyde levels, without changes in glutathione levels, further reduced the venom-induced renal tissue changes. CONCLUSION Our data suggest that PDE3 is involved in BaV-induced nephrotoxicity, as cilostazol administration significantly ameliorated these effects.
Collapse
Affiliation(s)
- Aline Diogo Marinho
- Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceara, Coronel Nunes de Melo St., 1127, 60.430-275, Fortaleza, CE, Brazil; Drug Research and Development Center (NPDM), Federal University of Ceara, Coronel Nunes de Melo St., 1000, 60.430-275, Fortaleza, CE, Brazil.
| | - Antônio Rafael Coelho Jorge
- Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceara, Coronel Nunes de Melo St., 1127, 60.430-275, Fortaleza, CE, Brazil; Drug Research and Development Center (NPDM), Federal University of Ceara, Coronel Nunes de Melo St., 1000, 60.430-275, Fortaleza, CE, Brazil
| | - Francisco Assis Nogueira Junior
- Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceara, Coronel Nunes de Melo St., 1127, 60.430-275, Fortaleza, CE, Brazil; Drug Research and Development Center (NPDM), Federal University of Ceara, Coronel Nunes de Melo St., 1000, 60.430-275, Fortaleza, CE, Brazil
| | - João Alison de Moraes Silveira
- Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceara, Coronel Nunes de Melo St., 1127, 60.430-275, Fortaleza, CE, Brazil; Drug Research and Development Center (NPDM), Federal University of Ceara, Coronel Nunes de Melo St., 1000, 60.430-275, Fortaleza, CE, Brazil
| | - Danilo Galvão Rocha
- Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceara, Coronel Nunes de Melo St., 1127, 60.430-275, Fortaleza, CE, Brazil; Drug Research and Development Center (NPDM), Federal University of Ceara, Coronel Nunes de Melo St., 1000, 60.430-275, Fortaleza, CE, Brazil
| | - Ana Paula Negreiros Nunes Alves
- Drug Research and Development Center (NPDM), Federal University of Ceara, Coronel Nunes de Melo St., 1000, 60.430-275, Fortaleza, CE, Brazil; Department of Dental Clinic, School of Pharmacy, Dentistry and Nursing, Federal University of Ceara, Fortaleza, Monsenhor Furtado St., 60.430-350, Fortaleza, Brazil
| | - Rui Seabra Ferreira
- Center for the Study of Venoms and Venomous Animals, Fazenda Experimental Lageado, São Paulo State University, José Barbosa de Barros St., 1780, 18610-307, Botucatu, SP, Brazil
| | - Roberta Jeane Bezerra Jorge
- Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceara, Coronel Nunes de Melo St., 1127, 60.430-275, Fortaleza, CE, Brazil; Drug Research and Development Center (NPDM), Federal University of Ceara, Coronel Nunes de Melo St., 1000, 60.430-275, Fortaleza, CE, Brazil
| | - Helena Serra Azul Monteiro
- Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceara, Coronel Nunes de Melo St., 1127, 60.430-275, Fortaleza, CE, Brazil; Drug Research and Development Center (NPDM), Federal University of Ceara, Coronel Nunes de Melo St., 1000, 60.430-275, Fortaleza, CE, Brazil
| |
Collapse
|
9
|
Rhynchosia volubilis Promotes Cell Survival via cAMP-PKA/ERK-CREB Pathway. Pharmaceuticals (Basel) 2022; 15:ph15010073. [PMID: 35056130 PMCID: PMC8778899 DOI: 10.3390/ph15010073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/29/2021] [Accepted: 01/03/2022] [Indexed: 02/01/2023] Open
Abstract
Rhynchosia volubilis, a small black bean, has been used as a traditional remedy to treat diseases and maintain health in East Asia, but its cellular effects and molecular mechanisms are not fully understood. The purpose of this study was to investigate the effect of ethanol extract from Rhynchosia volubilis (EERV) on cell survival and to elucidate the biochemical signaling pathways. Our results showed that EERV stimulated the cyclic AMP (cAMP) signal revealed by a fluorescent protein (FP)-based intensiometric sensor. Using a Förster resonance energy transfer (FRET)-based sensor, we further revealed that EERV could activate PKA and ERK signals, which are downstream effectors of cAMP. In addition, we reported that EERV could induce the phosphorylation of CREB, a key signal for cell survival. Thus, our results suggested that EERV protects against apoptosis by activating the cell survival pathway through the cAMP-PKA/ERK-CREB pathway.
Collapse
|
10
|
Phosphodiesterase 4 inhibitors in diabetic nephropathy. Cell Signal 2021; 90:110185. [PMID: 34785349 DOI: 10.1016/j.cellsig.2021.110185] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/31/2021] [Accepted: 11/01/2021] [Indexed: 12/18/2022]
Abstract
Phosphodiesterase subtype 4 (PDE4) hydrolyzes cyclic AMP, a secondary messenger that mediates intracellular signaling, and plays key roles in inflammatory and fibrotic responses. Based on these significant anti-inflammatory effects, oral administration of PDE4 inhibitor is approved for the treatment of chronic obstructive pulmonary disease, atopic dermatitis, and psoriasis. However, PDE4 inhibition also has adverse effects, such as diarrhea, vomiting, dyspepsia, and headache. Therefore, the application of PDE4 inhibitors for chronic diseases, such as diabetes and its complications, has not yet been approved. Recent studies have reported the clinical benefits of pentoxifylline, a non-selective PDE inhibitor, in patients with kidney disease. The PDE4 inhibitor, roflumilast, also clearly ameliorates the symptoms of diabetes mellitus by improving hyperglycemia and insulin resistance. However, the beneficial effects of PDE4 inhibition on diabetic nephropathy have not yet been evaluated, and its potential mechanisms of action remain unknown. In this review, we discuss the beneficial effects of PDE4 inhibitors and their mechanisms of action using diabetes and DN models.
Collapse
|
11
|
Wang L, Tang Y, Buckley AF, Spurney RF. Blockade of the natriuretic peptide clearance receptor attenuates proteinuria in a mouse model of focal segmental glomerulosclerosis. Physiol Rep 2021; 9:e15095. [PMID: 34755480 PMCID: PMC8578888 DOI: 10.14814/phy2.15095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/23/2021] [Accepted: 09/23/2021] [Indexed: 12/31/2022] Open
Abstract
Glomerular podocytes play a key role in proteinuric diseases. Accumulating evidence suggests that cGMP signaling has podocyte protective effects. The major source of cGMP generation in podocytes is natriuretic peptides. The natriuretic peptide clearance receptor (NPRC) binds and degrades natriuretic peptides. As a result, NPRC inhibits natriuretic peptide-induced cGMP generation. To enhance cGMP generation in podocytes, we blocked natriuretic peptide clearance using the specific NPRC ligand ANP(4-23). We then studied the effects of NPRC blockade in both cultured podocytes and in a mouse transgenic (TG) model of focal segmental glomerulosclerosis (FSGS) created in our laboratory. In this model, a single dose of the podocyte toxin puromycin aminonucleoside (PAN) causes robust albuminuria in TG mice, but only mild disease in non-TG animals. We found that natriuretic peptides protected cultured podocytes from PAN-induced apoptosis, and that ANP(4-23) enhanced natriuretic peptide-induced cGMP generation in vivo. PAN-induced heavy proteinuria in vehicle-treated TG mice, and this increase in albuminuria was reduced by treatment with ANP(4-23). Treatment with ANP(4-23) also reduced the number of mice with glomerular injury and enhanced urinary cGMP excretion, but these differences were not statistically significant. Systolic BP was similar in vehicle and ANP(4-23)-treated mice. These data suggest that: 1. Pharmacologic blockade of NPRC may be useful for treating glomerular diseases such as FSGS, and 2. Treatment outcomes might be improved by optimizing NPRC blockade to inhibit natriuretic peptide clearance more effectively.
Collapse
Affiliation(s)
- Liming Wang
- Division of NephrologyDepartment of MedicineDuke University and Durham VA Medical CentersDurhamNorth CarolinaUSA
| | - Yuping Tang
- Division of NephrologyDepartment of MedicineDuke University and Durham VA Medical CentersDurhamNorth CarolinaUSA
| | - Anne F. Buckley
- Department of PathologyDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Robert F. Spurney
- Division of NephrologyDepartment of MedicineDuke University and Durham VA Medical CentersDurhamNorth CarolinaUSA
| |
Collapse
|
12
|
Thapa K, Singh TG, Kaur A. Cyclic nucleotide phosphodiesterase inhibition as a potential therapeutic target in renal ischemia reperfusion injury. Life Sci 2021; 282:119843. [PMID: 34298037 DOI: 10.1016/j.lfs.2021.119843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/10/2021] [Accepted: 07/13/2021] [Indexed: 12/19/2022]
Abstract
AIMS Ischemia/reperfusion (I/R) occurs in renal artery stenosis, partial nephrectomy and most commonly during kidney transplantation. It brings serious consequences such as DGF (Delayed Graft Function) or organ dysfunction leading to renal failure and ultimate death. There is no effective therapy to handle the consequences of Renal Ischemia/Reperfusion (I/R) injury. Cyclic nucleotides, cAMP and cGMP are the important second messengers that stimulate intracellular signal transduction for cell survival in response to growth factors and peptide hormones in normal tissues and in kidneys plays significant role that involves vascular tone regulation, inflammation and proliferation of parenchymal cells. Renal ischemia and subsequent reperfusion injury stimulate signal transduction pathways involved in oxidative stress, inflammation, alteration in renal blood flow leading to necrosis and apoptosis of renal cell. MATERIALS AND METHODS An extensive literature review of various search engines like PubMed, Medline, Bentham, Scopus, and EMBASE (Elsevier) databases was carried out. To understand the functioning of Phosphodiesterases (PDEs) and its pharmacological modulation in Renal Ischemia-Reperfusion Injury. KEY FINDINGS Current therapeutic options may not be enough to treat renal I/R injury in group of patients and therefore, the current review has discussed the general characteristics and physiology of PDEs and preclinical-studies defining the relationship between PDEs expression in renal injury due to I/R and its outcome on renal function. SIGNIFICANCE The role of PDE inhibitors in renal I/R injury and the clinical status of drugs for various renal diseases have been summarized in this review.
Collapse
Affiliation(s)
- Komal Thapa
- Chitkara College of Pharmacy, Chitkara University, 140401 Punjab, India; School of Pharmacy, Himachal Pradesh, India
| | | | - Amarjot Kaur
- Chitkara College of Pharmacy, Chitkara University, 140401 Punjab, India
| |
Collapse
|
13
|
Sholokh A, Klussmann E. Local cyclic adenosine monophosphate signalling cascades-Roles and targets in chronic kidney disease. Acta Physiol (Oxf) 2021; 232:e13641. [PMID: 33660401 DOI: 10.1111/apha.13641] [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] [Received: 11/02/2020] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 12/20/2022]
Abstract
The molecular mechanisms underlying chronic kidney disease (CKD) are poorly understood and treatment options are limited, a situation underpinning the need for elucidating the causative molecular mechanisms and for identifying innovative treatment options. It is emerging that cyclic 3',5'-adenosine monophosphate (cAMP) signalling occurs in defined cellular compartments within nanometre dimensions in processes whose dysregulation is associated with CKD. cAMP compartmentalization is tightly controlled by a specific set of proteins, including A-kinase anchoring proteins (AKAPs) and phosphodiesterases (PDEs). AKAPs such as AKAP18, AKAP220, AKAP-Lbc and STUB1, and PDE4 coordinate arginine-vasopressin (AVP)-induced water reabsorption by collecting duct principal cells. However, hyperactivation of the AVP system is associated with kidney damage and CKD. Podocyte injury involves aberrant AKAP signalling. cAMP signalling in immune cells can be local and slow the progression of inflammatory processes typical for CKD. A major risk factor of CKD is hypertension. cAMP directs the release of the blood pressure regulator, renin, from juxtaglomerular cells, and plays a role in Na+ reabsorption through ENaC, NKCC2 and NCC in the kidney. Mutations in the cAMP hydrolysing PDE3A that cause lowering of cAMP lead to hypertension. Another major risk factor of CKD is diabetes mellitus. AKAP18 and AKAP150 and several PDEs are involved in insulin release. Despite the increasing amount of data, an understanding of functions of compartmentalized cAMP signalling with relevance for CKD is fragmentary. Uncovering functions will improve the understanding of physiological processes and identification of disease-relevant aberrations may guide towards new therapeutic concepts for the treatment of CKD.
Collapse
Affiliation(s)
- Anastasiia Sholokh
- Max‐Delbrück‐Center for Molecular Medicine (MDC) Helmholtz Association Berlin Germany
| | - Enno Klussmann
- Max‐Delbrück‐Center for Molecular Medicine (MDC) Helmholtz Association Berlin Germany
- DZHK (German Centre for Cardiovascular Research) Berlin Germany
| |
Collapse
|
14
|
Samidurai A, Xi L, Das A, Iness AN, Vigneshwar NG, Li PL, Singla DK, Muniyan S, Batra SK, Kukreja RC. Role of phosphodiesterase 1 in the pathophysiology of diseases and potential therapeutic opportunities. Pharmacol Ther 2021; 226:107858. [PMID: 33895190 DOI: 10.1016/j.pharmthera.2021.107858] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 03/17/2021] [Accepted: 04/14/2021] [Indexed: 12/15/2022]
Abstract
Cyclic nucleotide phosphodiesterases (PDEs) are superfamily of enzymes that regulate the spatial and temporal relationship of second messenger signaling in the cellular system. Among the 11 different families of PDEs, phosphodiesterase 1 (PDE1) sub-family of enzymes hydrolyze both 3',5'-cyclic adenosine monophosphate (cAMP) and 3',5'-cyclic guanosine monophosphate (cGMP) in a mutually competitive manner. The catalytic activity of PDE1 is stimulated by their binding to Ca2+/calmodulin (CaM), resulting in the integration of Ca2+ and cyclic nucleotide-mediated signaling in various diseases. The PDE1 family includes three subtypes, PDE1A, PDE1B and PDE1C, which differ for their relative affinities for cAMP and cGMP. These isoforms are differentially expressed throughout the body, including the cardiovascular, central nervous system and other organs. Thus, PDE1 enzymes play a critical role in the pathophysiology of diseases through the fundamental regulation of cAMP and cGMP signaling. This comprehensive review provides the current research on PDE1 and its potential utility as a therapeutic target in diseases including the cardiovascular, pulmonary, metabolic, neurocognitive, renal, cancers and possibly others.
Collapse
Affiliation(s)
- Arun Samidurai
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298-0204, USA
| | - Lei Xi
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298-0204, USA
| | - Anindita Das
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298-0204, USA
| | - Audra N Iness
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298-0204, USA
| | - Navin G Vigneshwar
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298-0204, USA
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
| | - Dinender K Singla
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA
| | - Sakthivel Muniyan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Rakesh C Kukreja
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298-0204, USA.
| |
Collapse
|
15
|
Phosphodiesterase (1, 3 & 5) inhibitors attenuate diclofenac-induced acute kidney toxicity in rats. Life Sci 2021; 277:119506. [PMID: 33865881 DOI: 10.1016/j.lfs.2021.119506] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/25/2021] [Accepted: 04/03/2021] [Indexed: 12/24/2022]
Abstract
Diclofenac, one of the most commonly used non-steroidal anti-inflammatory drugs, leads to severe adverse effects on the kidneys. The aim of the present study was to investigate the potential pretreatment effect of phosphodiesterase (1, 3 & 5) inhibitors on diclofenac-induced acute renal failure in rats. Rats orally received pentoxifylline (100 mg/kg), vinpocetine (20 mg/kg), cilostazol (50 mg/kg), or sildenafil (5 mg/kg) once per day for 6 consecutive days. Diclofenac (15 mg/kg) was injected on day-4, -5 and -6 in all groups except normal control group. The used phosphodiesterase inhibitors significantly reduced the diclofenac-induced elevation in the serum levels of blood urea nitrogen, creatinine and cystatin C. Moreover, the renal tissue contents of tumor necrosis factor (TNF)-α, nuclear factor (NF)-κB as well as the protein expression of toll-like receptor (TLR) 4 and high mobility group box (HMGB) 1 were markedly reduced by the used phosphodiesterase inhibitors, as compared to the diclofenac control. This was reflected on the marked improvement in histopathological changes induced by diclofenac. Sildenafil showed the best protection regarding TNF-α and NF-κB, while cilostazol showed the best results regarding TLR4, HMGB1 and histopathological examination. This study revealed the good protective effect of these phosphodiesterase inhibitors against diclofenac-induced acute renal failure.
Collapse
|
16
|
Coskuner ER, Ozkan B. Reno-protective effects of Phosphodiesterase 5 inhibitors. Clin Exp Nephrol 2021; 25:585-597. [PMID: 33754203 DOI: 10.1007/s10157-021-02051-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/09/2021] [Indexed: 12/20/2022]
Abstract
The kidneys are vital organs that play an important role in removing waste materials from the blood, electrolyte balance, blood pressure regulation, and red blood cell genesis. Kidney disease can be caused by various factors, including diabetes, ischemia/reperfusion injury, and nephrotoxic agents. Inflammation and oxidative stress play a key role in the progression and pathogenesis of kidney diseases. Acute kidney injury (AKI) and chronic kidney disease (CKD) are important health problems worldwide, as they are associated with a long-term hospital stay, and increased morbidity and mortality in high-risk patients. Current standard therapeutic options are not sufficient to delay or stop the loss of kidney function. Therefore, it is necessary to develop new therapeutic options. Phosphodiesterase 5 inhibitors (PDE5Is) are a currently available class of drugs that are used to treat erectile dysfunction and pulmonary hypertension in humans. However, recent evidence suggests that PDE5Is have beneficial renoprotective effects via a variety of mechanisms. In this review, the benefits of PDE5 inhibitors in clinical conditions associated with kidney disease, such as diabetic nephropathy, ischemia-reperfusion injury, and acute and chronic kidney injury, are summarized.
Collapse
Affiliation(s)
- Enis Rauf Coskuner
- Department of Urology, Acibadem Mehmet Ali Aydinlar University School of Medicine, Acibadem Bakirkoy Hospital, Halit Ziya Usakligil Cad No:1, Bakirkoy, 34140, Istanbul, Turkey.
| | - Burak Ozkan
- Department of Urology, Acibadem Mehmet Ali Aydinlar University School of Medicine, Acibadem Bakirkoy Hospital, Halit Ziya Usakligil Cad No:1, Bakirkoy, 34140, Istanbul, Turkey
| |
Collapse
|
17
|
Yougbare I, Keravis T, Lugnier C. NCS 613, a PDE4 inhibitor, by increasing cAMP level suppresses systemic inflammation and immune complexes deposition in kidney of MRL/lpr lupus- prone mice. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166019. [PMID: 33227426 DOI: 10.1016/j.bbadis.2020.166019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 11/15/2020] [Accepted: 11/17/2020] [Indexed: 01/06/2023]
Abstract
Nephritis remains the most common severe manifestation of systemic lupus erythematosus in which auto-antibodies mediate chronic inflammation and kidney damage. cAMP-phosphodiesterases regulate sodium excretion and inflammation in various tissues. How cAMP elevation can reduce systemic inflammation and suppress kidney inflammation and damage remains elusive. PDE4 signaling and cAMP metabolism were investigated along immune complex depositions in target tissues and kidney damage (histology). SLE disease progression is associated with changes in kidney PDE4 activity and expression. Moreover, lupus prone mice exhibit low kidney cAMP level which is associated to induction and relocation of nuclear and cytoskeleton PDE4 isoforms. Auto-antibodies-induced kidney damage was attested by mesangial proliferation and cellular infiltration. Interestingly, we reported that NCS 613 treatment decreases systemic auto-antibody secretion and their corresponding immune complex deposition in target tissues. Furthermore, NCS 613 is able to increase cAMP levels in the kidney; hence this compound rescues kidney PDE4 alterations in treated mice. NCS 613 overcomes disease progression in lupus prone mice by improving wellbeing and decreasing inflammation in treated mice. The PDE4 inhibitor, NCS 613, is a new anti-inflammatory compound that is believed to be a leading drug candidate for the treatment of inflammatory diseases such as lupus nephritis.
Collapse
Affiliation(s)
- Issaka Yougbare
- UMR CNRS 7213, Biophotonics and Pharmacology, Faculty of Pharmacy, University of Strasbourg, 67401 Illkirch Cedex, France.
| | - Thérèse Keravis
- UMR CNRS 7213, Biophotonics and Pharmacology, Faculty of Pharmacy, University of Strasbourg, 67401 Illkirch Cedex, France
| | - Claire Lugnier
- UMR CNRS 7213, Biophotonics and Pharmacology, Faculty of Pharmacy, University of Strasbourg, 67401 Illkirch Cedex, France; Institute of Physiology, FMTS-EA 3072, Faculty of Medicine, University of Strasbourg, 11 Humann Street, 67000 Strasbourg, France
| |
Collapse
|
18
|
Xu M, Li S, Wang J, Huang S, Zhang A, Zhang Y, Gu W, Yu X, Jia Z. Cilomilast Ameliorates Renal Tubulointerstitial Fibrosis by Inhibiting the TGF-β1-Smad2/3 Signaling Pathway. Front Med (Lausanne) 2021; 7:626140. [PMID: 33553218 PMCID: PMC7859332 DOI: 10.3389/fmed.2020.626140] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 12/30/2020] [Indexed: 12/17/2022] Open
Abstract
Background: Renal tubulointerstitial fibrosis is the key pathological feature in chronic kidney diseases (CKDs) with no satisfactory therapies in clinic. Cilomilast is a second-generation, selective phosphodiesterase-4 inhibitor, but its role in renal tubulointerstitial fibrosis in CKD remains unclear. Material and Methods: Cilomilast was applied to the mice with unilateral ureteric obstruction (UUO) and renal fibroblast cells (NRK-49F) stimulated by TGF-β1. Renal tubulointerstitial fibrosis and inflammation after UUO or TGF-β1 stimulation were examined by histology, Western blotting, real-time PCR and immunohistochemistry. KIM-1 and NGAL were detected to evaluate tubular injury in UUO mice. Results:In vivo, immunohistochemistry and western blot data demonstrated that cilomilast treatment inhibited extracellular matrix deposition, profibrotic gene expression, and the inflammatory response. Furthermore, cilomilast prevented tubular injury in UUO mice, as manifested by reduced expression of KIM-1 and NGAL in the kidney. In vitro, cilomilast attenuated the activation of fibroblast cells stimulated by TGF-β1, as shown by the reduced expression of fibronectin, α-SMA, collagen I, and collagen III. Cilomilast also inhibited the activation of TGF-β1-Smad2/3 signaling in TGF-β1-treated fibroblast cells. Conclusion: The findings of this study suggest that cilomilast is protective against renal tubulointerstitial fibrosis in CKD, possibly through the inhibition of TGF-β1-Smad2/3 signaling, indicating the translational potential of this drug in treating CKD.
Collapse
Affiliation(s)
- Man Xu
- Department of Endocrinology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Shumin Li
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Jiajia Wang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Songming Huang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Aihua Zhang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yue Zhang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Gu
- Department of Endocrinology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaowen Yu
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Zhanjun Jia
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
19
|
Tomita N, Hotta Y, Naiki‐Ito A, Hirano K, Kataoka T, Maeda Y, Takahashi S, Kimura K. The phosphodiesterase 5 inhibitor tadalafil has renoprotective effects in a rat model of chronic kidney disease. Physiol Rep 2020; 8:e14556. [PMID: 32889777 PMCID: PMC7503090 DOI: 10.14814/phy2.14556] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/26/2020] [Accepted: 07/26/2020] [Indexed: 01/14/2023] Open
Abstract
Phosphodiesterase 5 inhibitors are widely used to treat erectile dysfunction and lower urinary tract symptoms with benign prostatic hyperplasia. Recent studies have indicated the renoprotective effects of this class of compounds. Whether renoprotection depends on blood pressure reduction remains controversial. In this study, we investigated the renoprotective effects of the phosphodiesterase 5 inhibitor, tadalafil, in a rat model of high-salt induced kidney injury with hypertension. Dahl salt-sensitive rats were fed a normal diet, high-salt (8% sodium chloride) diet, or high-salt diet with oral administration of either low- or high-dose tadalafil (1 and 10 mg kg-1 day-1 , respectively). Serum creatinine, urinary protein, and blood pressure were measured at baseline and after 8 weeks, at which point the rats were examined for glomerular injury and fibrosis. PAI1 mRNA levels were also evaluated. After 8 weeks, blood pressure, serum creatinine, and urinary protein levels were significantly higher in the high-salt group than those in the normal-salt group. Serum creatinine and urinary protein were significantly lower in both tadalafil groups than those in the high-salt group, while only high-dose tadalafil affected blood pressure. In addition, glomerulosclerosis and α-smooth muscle actin expression significantly decreased in both tadalafil treatment groups. PAI1 mRNA increased significantly in the high-salt group but decreased in both tadalafil-treated groups. Our results indicated that both low- and high-dose tadalafil prevented fibrosis and glomerular injury in a chronic kidney disease rat model. Mechanistically, these effects may be associated with PAI1 expression and glomerular structure protection.
Collapse
Affiliation(s)
- Natsumi Tomita
- Department of Hospital PharmacyGraduate School of Pharmaceutical SciencesNagoya City UniversityNagoyaJapan
| | - Yuji Hotta
- Department of Hospital PharmacyGraduate School of Pharmaceutical SciencesNagoya City UniversityNagoyaJapan
| | - Aya Naiki‐Ito
- Department of Experimental Pathology and Tumor BiologyGraduate School of Medical SciencesNagoya City UniversityNagoyaJapan
| | - Kana Hirano
- Department of Hospital PharmacyGraduate School of Pharmaceutical SciencesNagoya City UniversityNagoyaJapan
| | - Tomoya Kataoka
- Department of Clinical PharmaceuticsGraduate School of Medical SciencesNagoya City UniversityNagoyaJapan
| | - Yasuhiro Maeda
- Center for Joint Research Facilities SupportFijita Health UniversityToyoakeJapan
| | - Satoru Takahashi
- Department of Experimental Pathology and Tumor BiologyGraduate School of Medical SciencesNagoya City UniversityNagoyaJapan
| | - Kazunori Kimura
- Department of Hospital PharmacyGraduate School of Pharmaceutical SciencesNagoya City UniversityNagoyaJapan
- Department of Clinical PharmaceuticsGraduate School of Medical SciencesNagoya City UniversityNagoyaJapan
| |
Collapse
|
20
|
Liu XQ, Jin J, Li Z, Jiang L, Dong YH, Cai YT, Wu MF, Wang JN, Ma TT, Wen JG, Liu MM, Li J, Wu YG, Meng XM. Rutaecarpine derivative Cpd-6c alleviates acute kidney injury by targeting PDE4B, a key enzyme mediating inflammation in cisplatin nephropathy. Biochem Pharmacol 2020; 180:114132. [PMID: 32622666 DOI: 10.1016/j.bcp.2020.114132] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/27/2020] [Accepted: 06/30/2020] [Indexed: 12/18/2022]
Abstract
Acute kidney injury (AKI), characterized by a rapid decline in renal function, is triggered by an acute inflammatory response that leads to kidney damage. An effective treatment for AKI is lacking. Using in vitro and in vivo AKI models, our laboratory has identified a series of anti-inflammatory molecules and their derivatives. In the current study, we identified the protective role of rutaecarpine (Ru) on renal tubules. We obtained a series of 3-aromatic sulphonamide-substituted Ru derivatives exhibiting enhanced renoprotective and anti-inflammatory function. We identified Compound-6c(Cpd-6c) as having the best activity and examined its protective effect against cisplatin nephropathy both in vivo and in vitro in cisplatin-stimulated tubular epithelial cells (TECs). Our results showed that Cpd-6c restored renal function more effectively than Ru, as evidenced by reduced blood urea nitrogen and serum creatinine levels in mice. Cpd-6c alleviated tubular injury, as shown by PAS staining and molecular analysis of kidney injury molecule-1 (KIM-1), with both prevention and treatment protocols in cisplatin-treated mice. Moreover, Cpd-6c decreased kidney inflammation, oxidative stress and programmed cell death. These results have also been confirmed in cisplatin-treated TECs. Using web-prediction algorithms, molecular docking, and cellular thermal shift assay (CETSA), we identified phosphodiesterase 4B (PDE4B) as a Cpd-6c target. In addition, we firstly found that PDE4B was up-regulated significantly in the serum of AKI patients. After identifying the function of PDE4B in cisplatin-treated tubular epithelial cells by siRNA transfection or PDE4 inhibitor rolipram, we showed that Cpd-6c treatment did not protect against cisplatin-induced injury in PDE4B knockdown TECs, thus indicating that Cpd-6c exerts its renoprotective and anti-oxidative effects via the PDE4B-dependent pathway. Collectively, Cpd-6c might serve as a potential therapeutic agent for AKI and PDE4B may be highly involved in the initiation and progression of AKI.
Collapse
Affiliation(s)
- Xue-Qi Liu
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei 230032, China
| | - Juan Jin
- School of Basic Medical Sciences, Anhui Medical University, Anhui, China
| | - Zeng Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei 230032, China
| | - Ling Jiang
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei 230032, China
| | - Yu-Hang Dong
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei 230032, China
| | - Yu-Ting Cai
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei 230032, China
| | - Ming-Fei Wu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei 230032, China
| | - Jia-Nan Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei 230032, China
| | - Tao-Tao Ma
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei 230032, China
| | - Jia-Gen Wen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei 230032, China
| | - Ming-Ming Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei 230032, China
| | - Jun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei 230032, China
| | - Yong-Gui Wu
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, China; The Center for Scientific Research of Anhui Medical University, Hefei, China.
| | - Xiao-Ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei 230032, China.
| |
Collapse
|
21
|
Chaiyabutr N, Chanhome L, Vasaruchapong T, Laoungbua P, Khow O, Rungsipipat A, Sitprija V. The pathophysiological effects of Russell's viper ( Daboia siamensis) venom and its fractions in the isolated perfused rabbit kidney model: A potential role for platelet activating factor. Toxicon X 2020; 7:100046. [PMID: 32875291 PMCID: PMC7452022 DOI: 10.1016/j.toxcx.2020.100046] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 05/21/2020] [Accepted: 06/04/2020] [Indexed: 12/20/2022] Open
Abstract
The pathophysiological effects of Russell's viper venom (RVV) and its fractions, including phospholipase A2 (RvPLA2), metalloprotease (RvMP), L-amino acid oxidase (RvLAAO), and phosphodiesterase (RvPDE) on renal functions were investigated using the isolated perfused rabbit kidney (IPK) model. Moreover, whether their effects on renal alterations were promoted by platelet activating factor (PAF) was tested using the PAF receptor antagonist, WEB 2086. There was a marked reduction in the perfusion pressure (PP) and renal vascular resistance (RVR) 10 min after RVV administration (1.0 mg/100 ml of perfusate), thereafter both PP and RVR gradually increased and approached the control level within 90 min. These effects were abolished by pretreatment with WEB2086 (2 μg/μl). Administration with RvPLA2 (280 μg/ml), RvMP (280 μg/ml), or RvLAAO (135 μg/ml) alone increased both the PP and RVR, whereas RvPDE (100 μg/ml) reduced both the PP and RVR. Pretreatment with WEB 2086 completely abolished the effects induced by RvMP, but not the other fractions. The RVV also caused a marked decrease in the glomerular filtration rate (GFR), urinary flow rate (UF), and osmolar clearance (Cosm), and these effects were not inhibited by pretreatment with WEB2086. Each RVV fraction also increased, to varying extents, the GFR, UF, and Cosm, and these effects induced by RvPLA2 or RvMP, but not the other fractions, were completely blocked by WEB 2086. Changes in percent filtered Na+ and K+ excreted in the IPK by RVV, RvPDE, and RvMP were abolished by pretreatment with WEB 2086. Histological evaluation profiled mainly tubulonephrosis in the treated kidney. These results reveal that the alterations in renal functions induced by RVV and its fractions are due to the synergistic action of the different components of snake venom, instead of the action of a single component. The effects of RVV and its fractions in rabbit IPK are mediated at least in part by PAF.
Collapse
Affiliation(s)
- Narongsak Chaiyabutr
- Queen Saovabha Memorial Institute, The Thai Red Cross Society, Bangkok, 10330, Thailand
| | - Lawan Chanhome
- Queen Saovabha Memorial Institute, The Thai Red Cross Society, Bangkok, 10330, Thailand
| | - Taksa Vasaruchapong
- Queen Saovabha Memorial Institute, The Thai Red Cross Society, Bangkok, 10330, Thailand
| | - Panithi Laoungbua
- Queen Saovabha Memorial Institute, The Thai Red Cross Society, Bangkok, 10330, Thailand
| | - Orawan Khow
- Queen Saovabha Memorial Institute, The Thai Red Cross Society, Bangkok, 10330, Thailand
| | - Anudep Rungsipipat
- Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Visith Sitprija
- Queen Saovabha Memorial Institute, The Thai Red Cross Society, Bangkok, 10330, Thailand
| |
Collapse
|
22
|
Scott NJA, Rademaker MT, Charles CJ, Espiner EA, Richards AM. Hemodynamic, Hormonal, and Renal Actions of Phosphodiesterase-9 Inhibition in Experimental Heart Failure. J Am Coll Cardiol 2020; 74:889-901. [PMID: 31416533 DOI: 10.1016/j.jacc.2019.05.067] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 04/18/2019] [Accepted: 05/23/2019] [Indexed: 11/27/2022]
Abstract
BACKGROUND Phosphodiesterase-9 (PDE9) reduces natriuretic peptide (NP) signaling and may be involved in the pathophysiology of heart failure (HF). OBJECTIVES This study investigated for the first time the integrated hemodynamic, endocrine, and renal effects of phosphodiesterase-9 inhibition (PDE9-I). METHODS A total of 8 normal sheep and 8 sheep with pacing-induced HF received incremental intravenous boluses of PDE9-I (30, 100, and 300 mg PF-04749982 at 1-h intervals). RESULTS PDE9-I dose-dependently increased plasma cyclic guanosine monophosphate (cGMP) in normal sheep (p < 0.05) while concurrently reducing circulating atrial natriuretic peptide levels (p < 0.01). Similar trends were evident in HF, resulting in significant elevations in the cGMP/NP ratio in both states (p < 0.001 and p < 0.05, respectively). PDE9-I also produced progressive falls in arterial pressure (HF: p < 0.001), atrial pressure (Normal: p < 0.001; HF: p < 0.001), and peripheral resistance (HF: p < 0.001), and transiently increased cardiac output at the top dose (Normal: p < 0.05; HF: p < 0.001). Inhibition of PDE9 had a negligible effect on circulating hormones at the lower doses, but post-high dose, acutely increased renin activity (Normal: p < 0.001; HF: p < 0.05), vasopressin (Normal: p < 0.001; HF: p < 0.01), and cyclic adenosine monophosphate (HF: p < 0.001). Plasma aldosterone increased briefly after high-dose PDE9-I in normal sheep, and fell following the top dose in HF. PDE9-I dose-dependently increased urinary cGMP in both states (both p < 0.001). In HF, this was associated with increases in urine volume (p < 0.01), sodium excretion (p < 0.01), and creatinine clearance (p < 0.001). CONCLUSIONS PDE9-I improves NP efficacy in conjunction with beneficial hemodynamic and renal effects in experimental HF. These results support a role for PDE9 in HF pathophysiology and suggest its inhibition may constitute a novel therapeutic approach to this disease.
Collapse
Affiliation(s)
- Nicola J A Scott
- Christchurch Heart Institute, Department of Medicine, University of Otago-Christchurch, Christchurch, New Zealand
| | - Miriam T Rademaker
- Christchurch Heart Institute, Department of Medicine, University of Otago-Christchurch, Christchurch, New Zealand.
| | - Christopher J Charles
- Christchurch Heart Institute, Department of Medicine, University of Otago-Christchurch, Christchurch, New Zealand; Cardiovascular Research Institute, Department of Surgery, National University of Singapore, Singapore
| | - Eric A Espiner
- Christchurch Heart Institute, Department of Medicine, University of Otago-Christchurch, Christchurch, New Zealand
| | - A Mark Richards
- Christchurch Heart Institute, Department of Medicine, University of Otago-Christchurch, Christchurch, New Zealand; Cardiovascular Research Institute, National University Health Systems, Centre for Translational Medicine, Singapore
| |
Collapse
|
23
|
Sussman CR, Wang X, Chebib FT, Torres VE. Modulation of polycystic kidney disease by G-protein coupled receptors and cyclic AMP signaling. Cell Signal 2020; 72:109649. [PMID: 32335259 DOI: 10.1016/j.cellsig.2020.109649] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/16/2020] [Accepted: 04/17/2020] [Indexed: 12/11/2022]
Abstract
Autosomal Dominant Polycystic Kidney Disease (ADPKD) is a systemic disorder associated with polycystic liver disease (PLD) and other extrarenal manifestations, the most common monogenic cause of end-stage kidney disease, and a major burden for public health. Many studies have shown that alterations in G-protein and cAMP signaling play a central role in its pathogenesis. As for many other diseases (35% of all approved drugs target G-protein coupled receptors (GPCRs) or proteins functioning upstream or downstream from GPCRs), treatments targeting GPCR have shown effectiveness in slowing the rate of progression of ADPKD. Tolvaptan, a vasopressin V2 receptor antagonist is the first drug approved by regulatory agencies to treat rapidly progressive ADPKD. Long-acting somatostatin analogs have also been effective in slowing the rates of growth of polycystic kidneys and liver. Although no treatment has so far been able to prevent the development or stop the progression of the disease, these encouraging advances point to G-protein and cAMP signaling as a promising avenue of investigation that may lead to more effective and safe treatments. This will require a better understanding of the relevant GPCRs, G-proteins, cAMP effectors, and of the enzymes and A-kinase anchoring proteins controlling the compartmentalization of cAMP signaling. The purpose of this review is to provide an overview of general GPCR signaling; the function of polycystin-1 (PC1) as a putative atypical adhesion GPCR (aGPCR); the roles of PC1, polycystin-2 (PC2) and the PC1-PC2 complex in the regulation of calcium and cAMP signaling; the cross-talk of calcium and cAMP signaling in PKD; and GPCRs, adenylyl cyclases, cyclic nucleotide phosphodiesterases, and protein kinase A as therapeutic targets in ADPKD.
Collapse
Affiliation(s)
- Caroline R Sussman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States of America
| | - Xiaofang Wang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States of America
| | - Fouad T Chebib
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States of America
| | - Vicente E Torres
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States of America.
| |
Collapse
|
24
|
Tasneem S, Saleem M, Saeed SA. Nonsteroidal anti-inflammatory drugs as potential ecto-nucleotide phosphodiesterase inhibitors. BRAZ J PHARM SCI 2020. [DOI: 10.1590/s2175-97902019000318271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Shumaila Tasneem
- University of Karachi, Pakistan; Dow University of Health Sciences, Pakistan
| | - Muhammad Saleem
- University of Karachi, Pakistan; University of Education, Pakistan
| | | |
Collapse
|
25
|
Iordache AM, Docea AO, Buga AM, Zlatian O, Ciurea ME, Rogoveanu OC, Burada F, Sosoi S, Mitrut R, Mamoulakis C, Albulescu D, Vasile RC, Tsatsakis A, Calina D. Sildenafil and tadalafil reduce the risk of contrast-induced nephropathy by modulating the oxidant/antioxidant balance in a murine model. Food Chem Toxicol 2019; 135:111038. [PMID: 31825855 DOI: 10.1016/j.fct.2019.111038] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/03/2019] [Accepted: 12/04/2019] [Indexed: 12/26/2022]
Abstract
The aim of the study was to evaluate the potential protective role of sildenafil and tadalafil in contrast-induced nephropathy (CIN) by modulating oxidative stress. Thirty Wistar male rats were equally assigned into five groups: sham, CIN, CIN + sildenafil (10 mg/kg bw/day), CIN + tadalafil (5 mg/kg bw/day) and CIN + N-Acetyl Cysteine (NAC) (100 mg/kg bw/day) as a positive control. CIN was induced by 12 h dehydration and administration of indomethacin (10 mg/kg bw), N-ω- nitro-L-arginine methyl ester (10 mg/kg bw), and iopromide (3 g/kg bw iodine). Blood was drawn prior to and 24 h after CIN induction for evaluating renal function and oxidative stress. In the CIN group, total antioxidant capacity (TAC), reduced glutathione (GSH) and catalase (CAT) levels were significantly decreased; and protein carbonyl (PROTC) and thiobarbituric reactive species (TBARS) were significantly increased compared to the sham group. Pre- Sildenafil and tadalafil pre-treatment reduced CIN risk and reversed oxidative stress almost to the sham group levels. These results suggest that PDE5Is can be good candidates for preventing CIN based on their ability to modulate the oxidant/antioxidant balance.
Collapse
Affiliation(s)
- Andrei Mihai Iordache
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| | - Anca Oana Docea
- Department of Toxicology, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| | - Ana Maria Buga
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| | - Ovidiu Zlatian
- Department of Microbiology, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| | - Marius Eugen Ciurea
- Department of Plastic Surgery, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| | - Otilia Constantina Rogoveanu
- Department of Physical and Rehabilitation Medicine Department, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| | - Florin Burada
- Department of Human Genomics Laboratory, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| | - Simona Sosoi
- Department of Human Genomics Laboratory, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| | - Radu Mitrut
- Department of Pathology, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania; Department of Cardiology, University and Emergency Hospital, 050098, Bucharest, Romania.
| | - Charalampos Mamoulakis
- Department of Urology, University General Hospital of Heraklion, University of Crete, Medical School, Heraklion, Crete, Greece.
| | - Dana Albulescu
- Department of Radiology, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| | - Ramona Constantina Vasile
- Department of Epidemiology and Primary Healthcare, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| | - Aristides Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, Heraklion, 71003, Greece.
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| |
Collapse
|
26
|
Huang W, Sundquist J, Sundquist K, Ji J. Use of Phosphodiesterase 5 Inhibitors Is Associated With Lower Risk of Colorectal Cancer in Men With Benign Colorectal Neoplasms. Gastroenterology 2019; 157:672-681.e4. [PMID: 31103628 DOI: 10.1053/j.gastro.2019.05.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/29/2019] [Accepted: 05/13/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND & AIMS Phosphodiesterase 5 (PDE5) inhibitors have been proposed to have chemopreventative effects on colorectal cancer (CRC), although data are needed from population-based studies. We performed a nationwide cohort study to investigate the association between the use of PDE5 inhibitors and the risk of CRC in men with benign colorectal neoplasms. METHODS We identified men who received a diagnosis of benign colorectal neoplasm from July 2005 through March 2015 who were listed in the Swedish Hospital Discharge Register. We linked data with those from other national Swedish registers to obtain information about the prescription of PDE5 inhibitors and CRC diagnoses. Cox regression was used to calculate hazard ratios (HRs) and 95% confidence intervals (CIs). RESULTS A total of 4823 patients were prescribed PDE5 inhibitors during the study period; the incidence rate of CRC was 2.64 per 1000 person-years for men prescribed PDE5 inhibitors compared with 4.46 per 1000 person-years for men without a prescription. We found a significant negative association between PDE5 inhibitor use and risk of CRC (adjusted HR, 0.65; 95% CI, 0.49-0.85); the decreased risk of CRC was associated with an increased cumulative dose of PDE5 inhibitors (P = .003). PDE5 prescription was associated with greater reduction in risk of advanced-stage CRC (adjusted HR, 0.61; 95% CI, 0.37-1.00) than early-stage CRC (adjusted HR, 0.70; 95% CI, 0.50-0.98), but the difference was not significant. CONCLUSIONS In a nationwide population-based study of men with a diagnosis of benign colorectal neoplasm in Sweden, we found evidence that use of PDE5 inhibitors is associated with a reduced risk of CRC. Further studies are needed to confirm the observed association.
Collapse
Affiliation(s)
- Wuqing Huang
- Center for Primary Health Care Research, Lund University/Region Skåne, Sweden
| | - Jan Sundquist
- Center for Primary Health Care Research, Lund University/Region Skåne, Sweden; Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Community-Based Healthcare Research and Education, Department of Functional Pathology, School of Medicine, Shimane University, Japan
| | - Kristina Sundquist
- Center for Primary Health Care Research, Lund University/Region Skåne, Sweden; Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Community-Based Healthcare Research and Education, Department of Functional Pathology, School of Medicine, Shimane University, Japan
| | - Jianguang Ji
- Center for Primary Health Care Research, Lund University/Region Skåne, Sweden.
| |
Collapse
|
27
|
Imam F, Al-Harbi NO, Al-Harbi MM, Ansari MA, Al-Asmari AF, Ansari MN, Al-Anazi WA, Bahashwan S, Almutairi MM, Alshammari M, Khan MR, Alsaad AM, Alotaibi MR. Apremilast prevent doxorubicin-induced apoptosis and inflammation in heart through inhibition of oxidative stress mediated activation of NF-κB signaling pathways. Pharmacol Rep 2018; 70:993-1000. [DOI: 10.1016/j.pharep.2018.03.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 01/01/2018] [Accepted: 03/27/2018] [Indexed: 12/25/2022]
|
28
|
Yarla NS, Gali H, Pathuri G, Smriti S, Farooqui M, Panneerselvam J, Kumar G, Madka V, Rao CV. Targeting the paracrine hormone-dependent guanylate cyclase/cGMP/phosphodiesterases signaling pathway for colorectal cancer prevention. Semin Cancer Biol 2018; 56:168-174. [PMID: 30189250 DOI: 10.1016/j.semcancer.2018.08.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/30/2018] [Accepted: 08/31/2018] [Indexed: 02/06/2023]
Abstract
Colorectal cancer (CRC) is one of the leading causes of cancer related-deaths. The risk of development of CRC is complex and multifactorial, and includes disruption of homeostasis of the intestinal epithelial layer mediated though dysregulations of tumor suppressing/promoting signaling pathways. Guanylate cyclase 2C (GUCY2C), a membrane-bound guanylate cyclase receptor, is present in the apical membranes of intestinal epithelial cells and maintains homeostasis. GUCY2C is activated upon binding of paracrine hormones (guanylin and uroguanylin) that lead to formation of cyclic GMP from GTP and activation of downstream signaling pathways that are associated with normal homeostasis. Dysregulation/suppression of the GUCY2C-mediated signaling promotes CRC tumorigenesis. High-calorie diet-induced obesity is associated with deficiency of guanylin expression and silencing of GUCY2C-signaling in colon epithelial cells, leading to tumorigenesis. Thus, GUCY2C agonists, such as linaclotide, exhibit considerable role in preventing CRC tumorigenesis. However, phosphodiesterases (PDEs) are elevated in intestinal epithelial cells during CRC tumorigenesis and block GUCY2C-mediated signaling by degrading cyclic GMP to 5`-GMP. PDE5-specific inhibitors, such as sildenafil, show considerable anti-tumorigenic potential against CRC by amplifying the GUCY2C/cGMP signaling pathway, but cannot achieve complete anti-tumorigenic effects. Hence, dual targeting the elevation of cGMP by providing paracrine hormone stimuli to GUCY2C and by inhibition of PDEs may be a better strategy for CRC prevention than alone. This review delineates the involvement of the GUCY2C/cGMP/PDEs signaling pathway in the homeostasis of intestinal epithelial cells. Further, the events are associated with dysregulation of this pathway during CRC tumorigenesis are also discussed. In addition, current updates on targeting the GUCY2C/cGMP/PDEs pathway with GUCY2C agonists and PDEs inhibitors for CRC prevention and treatment are described in detail.
Collapse
Affiliation(s)
- N S Yarla
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology-Oncology Section, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - H Gali
- Department of Pharmaceutical Sciences, College of Pharmacy, and Stephenson Oklahoma Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - G Pathuri
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology-Oncology Section, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - S Smriti
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology-Oncology Section, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - M Farooqui
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology-Oncology Section, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - J Panneerselvam
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology-Oncology Section, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - G Kumar
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology-Oncology Section, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; VA Medical Center, Oklahoma City, OK, USA
| | - V Madka
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology-Oncology Section, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - C V Rao
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology-Oncology Section, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; VA Medical Center, Oklahoma City, OK, USA.
| |
Collapse
|
29
|
Ding H, Bai F, Cao H, Xu J, Fang L, Wu J, Yuan Q, Zhou Y, Sun Q, He W, Dai C, Zen K, Jiang L, Yang J. PDE/cAMP/Epac/C/EBP-β Signaling Cascade Regulates Mitochondria Biogenesis of Tubular Epithelial Cells in Renal Fibrosis. Antioxid Redox Signal 2018; 29:637-652. [PMID: 29216750 DOI: 10.1089/ars.2017.7041] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
AIMS Cyclic adenosine 3'5'-monophosphate (cAMP) is a universal second messenger that plays an important role in intracellular signal transduction. cAMP is synthesized by adenylate cyclases from adenosine triphosphate and terminated by the phosphodiesterases (PDEs). In the present study, we investigated the role of the cAMP pathway in tubular epithelial cell mitochondrial biogenesis in the pathogenesis of renal fibrosis. RESULTS We found that the cAMP levels were decreased in fibrotic kidney tissues, and replenishing cAMP could ameliorate tubular atrophy and extracellular matrix deposition. The downregulation of cAMP was mainly attributed to the increased PDE4 expression in tubular epithelial cells. The inhibition of PDE4 by PDE4 siRNA or the specific inhibitor, rolipram, attenuated unilateral ureteral obstruction-induced renal interstitial fibrosis and transforming growth factor (TGF)-β1-stimulated primary tubular epithelial cell (PTC) damage. The Epac1/Rap1 pathway contributed to the main effect of cAMP on renal fibrosis. Rolipram could restore C/EBP-β and PGC-1α expression and protect the mitochondrial function and structure of PTCs under TGF-β1 stimulation. The antifibrotic role of rolipram in renal fibrosis relies on C/EBP-β and PGC-1α expression in tubular epithelial cells. Innovation and Conclusion: The results of the present study indicate that cAMP signaling regulates the mitochondrial biogenesis of tubular epithelial cells in renal fibrosis. Restoring cAMP by the PDE4 inhibitor rolipram may ameliorate renal fibrosis by targeting C/EBP-β/PGC1-α and mitochondrial biogenesis. Antioxid. Redox Signal. 29, 637-652.
Collapse
Affiliation(s)
- Hao Ding
- 1 Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University , Nanjing, China
| | - Feng Bai
- 1 Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University , Nanjing, China .,2 Department of Endocrinology and Metabolism, Huai'an Hospital Affiliated to Xuzhou Medical University and Huai'an Second People's Hospital , Huai'an, China
| | - Hongdi Cao
- 1 Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University , Nanjing, China
| | - Jing Xu
- 1 Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University , Nanjing, China
| | - Li Fang
- 3 Department of Nephrology, Affiliated Hospital of Nantong University , Nantong, China
| | - Jining Wu
- 1 Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University , Nanjing, China
| | - Qi Yuan
- 1 Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University , Nanjing, China
| | - Yang Zhou
- 1 Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University , Nanjing, China
| | - Qi Sun
- 1 Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University , Nanjing, China
| | - Weichun He
- 1 Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University , Nanjing, China
| | - Chunsun Dai
- 1 Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University , Nanjing, China
| | - Ke Zen
- 4 State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University Advanced Institute of Life Sciences , Nanjing, China
| | - Lei Jiang
- 1 Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University , Nanjing, China
| | - Junwei Yang
- 1 Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University , Nanjing, China
| |
Collapse
|
30
|
The Impact of the Nitric Oxide (NO)/Soluble Guanylyl Cyclase (sGC) Signaling Cascade on Kidney Health and Disease: A Preclinical Perspective. Int J Mol Sci 2018; 19:ijms19061712. [PMID: 29890734 PMCID: PMC6032334 DOI: 10.3390/ijms19061712] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/07/2018] [Accepted: 06/08/2018] [Indexed: 12/20/2022] Open
Abstract
Chronic Kidney Disease (CKD) is a highly prevalent disease with a substantial medical need for new and more efficacious treatments. The Nitric Oxide (NO), soluble guanylyl cyclase (sGC), cyclic guanosine monophosphate (cGMP) signaling cascade regulates various kidney functions. cGMP directly influences renal blood flow, renin secretion, glomerular function, and tubular exchange processes. Downregulation of NO/sGC/cGMP signaling results in severe kidney pathologies such as CKD. Therefore, treatment strategies aiming to maintain or increase cGMP might have beneficial effects for the treatment of progressive kidney diseases. Within this article, we review the NO/sGC/cGMP signaling cascade and its major pharmacological intervention sites. We specifically focus on the currently known effects of cGMP on kidney function parameters. Finally, we summarize the preclinical evidence for kidney protective effects of NO-donors, PDE inhibitors, sGC stimulators, and sGC activators.
Collapse
|
31
|
Sharma M, Levenson C, Browning JC, Becker EM, Clements I, Castella P, Cox ME. East Indian Sandalwood Oil Is a Phosphodiesterase Inhibitor: A New Therapeutic Option in the Treatment of Inflammatory Skin Disease. Front Pharmacol 2018; 9:200. [PMID: 29593534 PMCID: PMC5854648 DOI: 10.3389/fphar.2018.00200] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 02/22/2018] [Indexed: 01/01/2023] Open
Abstract
Cyclic adenosine monophosphate phosphodiesterases (PDEs) regulate pro-inflammatory cytokine production. One isoform, PDE4, is overactive in chronic relapsing inflammatory skin diseases: psoriasis and eczema/atopic dermatitis, and in several cancers. East Indian sandalwood oil (EISO) has significant anti-inflammatory properties. Here, we report that 75% of pediatric eczema/atopic dermatitis patients treated with topical EISO formulations achieved a >50% reduction in their Eczema Area and Severity Index score. EISO treatment of a psoriasis model reduced PDE4 expression and reversed histopathology. EISO directly inhibited PDE enzymatic activity in vitro. In lipopolysaccharide-stimulated human dermal fibroblast, BEAS-2B, A549, and THP-1 cells, EISO suppressed total cellular PDE activity, PDE4, and 7 transcript levels, nuclear factor kappa B (NF-κB) activation, and pro-inflammatory cytokines/chemokine production. These results suggest that EISO anti-inflammatory activity is mediated through suppressing PDE activity, thus facilitating cAMP-regulated inhibition of NF-κB and indicate EISO as an attractive natural therapeutic for chronic and acute inflammatory disorders.
Collapse
Affiliation(s)
- Manju Sharma
- The Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
| | - Corey Levenson
- Santalis Pharmaceuticals, Inc., San Antonio, TX, United States
| | - John C Browning
- Texas Dermatology and Laser Specialists, San Antonio, TX, United States
| | - Emily M Becker
- Texas Dermatology and Laser Specialists, San Antonio, TX, United States
| | - Ian Clements
- Santalis Pharmaceuticals, Inc., San Antonio, TX, United States
| | - Paul Castella
- Santalis Pharmaceuticals, Inc., San Antonio, TX, United States
| | - Michael E Cox
- The Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada.,Department of Urologic Sciences, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
32
|
Umejiego EN, Wang Y, Knepper MA, Chou CL. Roflumilast and aquaporin-2 regulation in rat renal inner medullary collecting duct. Physiol Rep 2017; 5:5/2/e13121. [PMID: 28108651 PMCID: PMC5269416 DOI: 10.14814/phy2.13121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 11/29/2016] [Accepted: 12/12/2016] [Indexed: 12/23/2022] Open
Abstract
Roflumilast is a cyclic nucleotide phosphodiesterase inhibitor that is FDA‐approved for treatment of chronic obstructive pulmonary disease. With a view toward possible use for treatment of patients with X‐linked nephrogenic diabetes insipidus (NDI) due to hemizygous mutations in the V2 vasopressin receptor, this study sought to determine the effect of roflumilast on aquaporin‐2 (AQP2) phosphorylation, AQP2 trafficking, and water permeability in the rat inner medullary collecting duct (IMCD). In the presence of the vasopressin analog dDAVP (0.1 nmol/L), both roflumilast and its active metabolite roflumilast N‐oxide (RNO) significantly increased phosphorylation at S256, S264, and S269, and decreased phosphorylation at S261 (immunoblotting) in IMCD suspensions in a dose‐dependent manner (3–3000 nmol/L). Another commonly used phosphodiesterase inhibitor, IBMX, affected phosphorylation only at the highest concentration in this range. However, neither roflumilast nor RNO had an effect on AQP2 phosphorylation in the absence of vasopressin. Furthermore, roflumilast alone did not increase AQP2 trafficking to the plasma membrane (immunofluorescence) or increase water permeability in freshly microdissected perfused IMCD segments. We conclude that roflumilast can be used to enhance vasopressin's action on AQP2 activity in the renal collecting duct, but has no detectable effect in the absence of vasopressin. These findings suggest that roflumilast may not have a beneficial effect in X‐linked NDI, but could find useful application in acquired NDI.
Collapse
Affiliation(s)
- Ezigbobiara N Umejiego
- Epithelial Systems Biology Laboratory, Systems Biology Center NHLBI National Institutes of Health, Bethesda, Maryland, 20892-1603
| | - Yanhua Wang
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia, 30322
| | - Mark A Knepper
- Epithelial Systems Biology Laboratory, Systems Biology Center NHLBI National Institutes of Health, Bethesda, Maryland, 20892-1603
| | - Chung-Lin Chou
- Epithelial Systems Biology Laboratory, Systems Biology Center NHLBI National Institutes of Health, Bethesda, Maryland, 20892-1603
| |
Collapse
|
33
|
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: 53] [Impact Index Per Article: 7.6] [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
|
34
|
Wang X, Yamada S, LaRiviere WB, Ye H, Bakeberg JL, Irazabal MV, Chebib FT, van Deursen J, Harris PC, Sussman CR, Behfar A, Ward CJ, Torres VE. Generation and phenotypic characterization of Pde1a mutant mice. PLoS One 2017; 12:e0181087. [PMID: 28750036 PMCID: PMC5531505 DOI: 10.1371/journal.pone.0181087] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 06/26/2017] [Indexed: 12/15/2022] Open
Abstract
It has been proposed that a reduction in intracellular calcium causes an increase in intracellular cAMP and PKA activity through stimulation of calcium inhibitable adenylyl cyclase 6 and inhibition of phosphodiesterase 1 (PDE1), the main enzymes generating and degrading cAMP in the distal nephron and collecting duct, thus contributing to the development and progression of autosomal dominant polycystic kidney disease (ADPKD). In zebrafish pde1a depletion aggravates and overexpression ameliorates the cystic phenotype. To study the role of PDE1A in a mammalian system, we used a TALEN pair to Pde1a exon 7, targeting the histidine-aspartic acid dipeptide involved in ligating the active site Zn++ ion to generate two Pde1a null mouse lines. Pde1a mutants had a mild renal cystic disease and a urine concentrating defect (associated with upregulation of PDE4 activity and decreased protein kinase A dependent phosphorylation of aquaporin-2) on a wild-type genetic background and aggravated renal cystic disease on a Pkd2WS25/- background. Pde1a mutants additionally had lower aortic blood pressure and increased left ventricular (LV) ejection fraction, without a change in LV mass index, consistent with the high aortic and low cardiac expression of Pde1a in wild-type mice. These results support an important role of PDE1A in the renal pathogenesis of ADPKD and in the regulation of blood pressure.
Collapse
Affiliation(s)
- Xiaofang Wang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Satsuki Yamada
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Wells B. LaRiviere
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Hong Ye
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Jason L. Bakeberg
- Division of Nephrology and Hypertension, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - María V. Irazabal
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Fouad T. Chebib
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Jan van Deursen
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Peter C. Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Caroline R. Sussman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Atta Behfar
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Christopher J. Ward
- Division of Nephrology and Hypertension, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- * E-mail: (VET); (CJW)
| | - Vicente E. Torres
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail: (VET); (CJW)
| |
Collapse
|
35
|
Tetsi L, Charles AL, Paradis S, Lejay A, Talha S, Geny B, Lugnier C. Effects of cyclic nucleotide phosphodiesterases (PDEs) on mitochondrial skeletal muscle functions. Cell Mol Life Sci 2017; 74:1883-1893. [PMID: 28039524 PMCID: PMC11107545 DOI: 10.1007/s00018-016-2446-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 12/12/2016] [Accepted: 12/19/2016] [Indexed: 12/31/2022]
Abstract
Mitochondria play a critical role in skeletal muscle metabolism and function, notably at the level of tissue respiration, which conduct muscle strength as well as muscle survival. Pathological conditions induce mitochondria dysfunctions notably characterized by free oxygen radical production disturbing intracellular signaling. In that way, the second messengers, cyclic AMP and cyclic GMP, control intracellular signaling at the physiological and transcription levels by governing phosphorylation cascades. Both nucleotides are specifically and selectively hydrolyzed in their respective 5'-nucleotide by cyclic nucleotide phosphodiesterases (PDEs), which constitute a multi-genic family differently tissue distributed and subcellularly compartmentalized. These PDEs are presently recognized as therapeutic targets for cardiovascular, pulmonary, and neurologic diseases. However, very few data concerning cyclic nucleotides and PDEs in skeletal muscle, specifically in mitochondria, are reported in the literature. The knowledge of PDE implication in mitochondrial signaling would be helpful for resolving critical mitochondrial dysfunctions in skeletal muscle.
Collapse
Affiliation(s)
- Liliane Tetsi
- EA 3072 "Mitochondrie, Stress Oxydant et Protection Musculaire", Fédération de Médecine Translationnelle, Faculté de Médecine, Institut de Physiologie, Université de Strasbourg, 4, Rue Kirschleger, 67085, Strasbourg Cedex, France
| | - Anne-Laure Charles
- EA 3072 "Mitochondrie, Stress Oxydant et Protection Musculaire", Fédération de Médecine Translationnelle, Faculté de Médecine, Institut de Physiologie, Université de Strasbourg, 4, Rue Kirschleger, 67085, Strasbourg Cedex, France
| | - Stéphanie Paradis
- EA 3072 "Mitochondrie, Stress Oxydant et Protection Musculaire", Fédération de Médecine Translationnelle, Faculté de Médecine, Institut de Physiologie, Université de Strasbourg, 4, Rue Kirschleger, 67085, Strasbourg Cedex, France
| | - Anne Lejay
- EA 3072 "Mitochondrie, Stress Oxydant et Protection Musculaire", Fédération de Médecine Translationnelle, Faculté de Médecine, Institut de Physiologie, Université de Strasbourg, 4, Rue Kirschleger, 67085, Strasbourg Cedex, France
| | - Samy Talha
- EA 3072 "Mitochondrie, Stress Oxydant et Protection Musculaire", Fédération de Médecine Translationnelle, Faculté de Médecine, Institut de Physiologie, Université de Strasbourg, 4, Rue Kirschleger, 67085, Strasbourg Cedex, France
| | - Bernard Geny
- EA 3072 "Mitochondrie, Stress Oxydant et Protection Musculaire", Fédération de Médecine Translationnelle, Faculté de Médecine, Institut de Physiologie, Université de Strasbourg, 4, Rue Kirschleger, 67085, Strasbourg Cedex, France
| | - Claire Lugnier
- EA 3072 "Mitochondrie, Stress Oxydant et Protection Musculaire", Fédération de Médecine Translationnelle, Faculté de Médecine, Institut de Physiologie, Université de Strasbourg, 4, Rue Kirschleger, 67085, Strasbourg Cedex, France.
| |
Collapse
|
36
|
Phosphodiesterase-5 inhibition preserves renal hemodynamics and function in mice with diabetic kidney disease by modulating miR-22 and BMP7. Sci Rep 2017; 7:44584. [PMID: 28294194 PMCID: PMC5353686 DOI: 10.1038/srep44584] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 02/09/2017] [Indexed: 01/13/2023] Open
Abstract
Diabetic Nephropathy (DN) is the leading cause of end-stage renal disease. Preclinical and experimental studies show that PDE5 inhibitors (PDE5is) exert protective effects in DN improving perivascular inflammation. Using a mouse model of diabetic kidney injury we investigated the protective proprieties of PDE5is on renal hemodynamics and the molecular mechanisms involved. PDE5i treatment prevented the development of DN-related hypertension (P < 0.001), the increase of urine albumin creatinine ratio (P < 0.01), the fall in glomerular filtration rate (P < 0.001), and improved renal resistive index (P < 0.001) and kidney microcirculation. Moreover PDE5i attenuated the rise of nephropathy biomarkers, soluble urokinase-type plasminogen activator receptor, suPAR and neutrophil gelatinase-associated lipocalin, NGAL. In treated animals, blood vessel perfusion was improved and vascular leakage reduced, suggesting preserved renal endothelium integrity, as confirmed by higher capillary density, number of CD31+ cells and pericyte coverage. Analysis of the mechanisms involved revealed the induction of bone morphogenetic protein-7 (BMP7) expression, a critical regulator of angiogenesis and kidney homeostasis, through a PDE5i-dependent downregulation of miR-22. In conclusion PDE5i slows the progression of DN in mice, improving hemodynamic parameters and vessel integrity. Regulation of miR-22/BMP7, an unknown mechanism of PDE5is in nephrovascular protection, might represent a novel therapeutic option for treatment of diabetic complications.
Collapse
|
37
|
Motte E, Le Stunff C, Briet C, Dumaz N, Silve C. Modulation of signaling through GPCR-cAMP-PKA pathways by PDE4 depends on stimulus intensity: Possible implications for the pathogenesis of acrodysostosis without hormone resistance. Mol Cell Endocrinol 2017; 442:1-11. [PMID: 27908835 DOI: 10.1016/j.mce.2016.11.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 11/25/2016] [Accepted: 11/26/2016] [Indexed: 12/30/2022]
Abstract
In acrodysostosis without hormone resistance, a disease caused by phosphodiesterase (PDE)-4D mutations, increased PDE activity leads to bone developmental defects but with normal renal responses to PTH. To identify potential mechanisms for these disparate responses, we compared the effect of PDE activity on hormone signaling through the GPCR-Gsα-cAMP-PKA pathway in cells from two lineages, HEK-293 cells stably overexpressing PTH1R (HEKpthr) and human dermal fibroblasts, including studies evaluating cAMP levels using an Epac-based BRET-sensor for cAMP (CAMYEL). For ligand-induced responses inducing strong cAMP accumulation, the inhibition of PDE4 activity resulted in relatively small further increases. In contrast, when ligand-induced cAMP accumulation was of lesser intensity, the inhibition of PDE4 had a more pronounced effect. Similar results were obtained evaluating downstream events (cellular CREB phosphorylation and CRE-luciferase activity). Thus, the ability of PDE4 to modulate signaling through GPCR-cAMP-PKA pathways can depend on the cell type and stimulus intensity.
Collapse
Affiliation(s)
- Emmanuelle Motte
- INSERM U1169, Université Paris Sud, Hôpital Bicêtre, Le Kremlin Bicêtre, France
| | - Catherine Le Stunff
- INSERM U1169, Université Paris Sud, Hôpital Bicêtre, Le Kremlin Bicêtre, France
| | - Claire Briet
- INSERM U1169, Université Paris Sud, Hôpital Bicêtre, Le Kremlin Bicêtre, France
| | - Nicolas Dumaz
- INSERM U976, Institut de Recherche sur la Peau, Hôpital Saint Louis, Paris, France
| | - Caroline Silve
- INSERM U1169, Université Paris Sud, Hôpital Bicêtre, Le Kremlin Bicêtre, France; Centre de Référence des Maladies Rares du Métabolisme du Phosphore et du Calcium, Assistance Publique Hôpitaux de Paris, Paris, France; Service de Biochimie et Génétique Moléculaires, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Paris, France.
| |
Collapse
|
38
|
Kotlo K, Samarel AM, Chen HY, Aldstadt J, Danziger RS. Global comparison of phosphoproteins in human and rodent hearts: implications for translational studies of myosin light chain and troponin phosphorylations. SPRINGERPLUS 2016; 5:808. [PMID: 27390648 PMCID: PMC4916117 DOI: 10.1186/s40064-016-2469-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 05/29/2016] [Indexed: 11/10/2022]
Abstract
Cardiac remodeling and failure are regulated by a myriad of cardiac protein phosphorylations. In the present study, cardiac phosphoprotein patterns were examined in rodent and human hearts Left ventricular tissue samples were obtained from human systolic failing (n = 5) and control (n = 5) hearts and from two rat models of hypertensive heart failure, i.e., spontaneously hypertensive heart failure and Dahl salt-sensitive rats and corresponding controls. Phosphoproteins were separated by 2D-DIGE with Cydye staining, phosphoprotein patterns were analyzed using pixel intensity in rectified images. Specific phosphoproteins which were different in human versus rodent hearts were identified by MALDI-TOF/TOF Mass Spectrometry. Targeted pair-wise analyses showed differences (p < 0.05) in 26 % of the pixels, which included pixels containing phosphorylated troponin T, myosin light chain, peroxiredoxin, and haptoglobin. These results show differences in rodent versus human cardiac remodeling which will influence the translation rodent studies to humans in this area.
Collapse
|
39
|
Imam F, Al-Harbi NO, Al-Harbi MM, Ansari MA, Almutairi MM, Alshammari M, Almukhlafi TS, Ansari MN, Aljerian K, Ahmad SF. Apremilast reversed carfilzomib-induced cardiotoxicity through inhibition of oxidative stress, NF-κB and MAPK signaling in rats. Toxicol Mech Methods 2016; 26:700-708. [PMID: 27785949 DOI: 10.1080/15376516.2016.1236425] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Carfilzomib (CFZ), is a potent, selective second generation proteasome inhibitor, used for the treatment of multiple myeloma. The aim of the present study was to investigate the possible protective effect of apremilast (AP) on the CFZ -induced cardiotoxicity. Rats were randomly divided into four groups: Group 1, served as the control group, received normal saline. Group 2, served as the toxic group, received CFZ (4 mg/kg, intraperitoneally [i.p.]). Groups 3 and 4, served as treatment groups, and received CFZ with concomitant oral administration of AP in doses of 10 and 20 mg/kg/day, respectively. In the present study, administration of CFZ resulted in a significant increase in serum aspartate transaminase (AST), lactate dehydrogenase (LDH), creatine kinase (CK) and creatine kinase-MB (CK-MB), which were reversed by treatment with AP. CFZ resulted in a significant increase in heart malondialdehyde (MDA) contents and decrease in cardiac glutathione (GSH) level and catalase (CAT) enzyme activity which were significantly reversed by treatment with AP. Induction of cardiotoxicity by CFZ significantly increased caspase-3 enzyme activity which were reversed by treatment with AP. RT-PCR analysis revealed an increased mRNA expression of NF-κB, ERK and JNK which were reversed by treatment with AP in cardiac tissues. Western blot analysis revealed an increased expression of caspase-3 and NF-κB p65 and a decrease expression of inhibitory kappa B-alpha (Iκbα) with CFZ, which were reversed by treatment with AP. In conclusion, apremilast showed protective effect against CFZ-induced cardiotoxicity.
Collapse
Affiliation(s)
- Faisal Imam
- a Department of Pharmacology and Toxicology, College of Pharmacy , King Saud University , Riyadh , Saudi Arabia
| | - Naif O Al-Harbi
- a Department of Pharmacology and Toxicology, College of Pharmacy , King Saud University , Riyadh , Saudi Arabia
| | - Mohammad Matar Al-Harbi
- a Department of Pharmacology and Toxicology, College of Pharmacy , King Saud University , Riyadh , Saudi Arabia
| | - Mushtaq Ahmad Ansari
- a Department of Pharmacology and Toxicology, College of Pharmacy , King Saud University , Riyadh , Saudi Arabia
| | - Mashal M Almutairi
- a Department of Pharmacology and Toxicology, College of Pharmacy , King Saud University , Riyadh , Saudi Arabia
| | - Musaad Alshammari
- a Department of Pharmacology and Toxicology, College of Pharmacy , King Saud University , Riyadh , Saudi Arabia
| | - Talal Saad Almukhlafi
- b Department of Pharmacology, College of Pharmacy , Prince Sattam Bin Abdulaziz University , Al-Kharj , Saudi Arabia
| | - Mohd Nazam Ansari
- b Department of Pharmacology, College of Pharmacy , Prince Sattam Bin Abdulaziz University , Al-Kharj , Saudi Arabia
| | - Khaldoon Aljerian
- c King Khalid University Hospital, College of Medicine , King Saud University, Forensic Medicine and Toxicology Unit , Riyadh , Saudi Arabia
| | - Sheikh Fayaz Ahmad
- a Department of Pharmacology and Toxicology, College of Pharmacy , King Saud University , Riyadh , Saudi Arabia
| |
Collapse
|
40
|
Tripathi AS, Mazumder PM, Chandewar AV. Sildenafil, a phosphodiesterase type 5 inhibitor, attenuates diabetic nephropathy in STZ-induced diabetic rats. J Basic Clin Physiol Pharmacol 2016; 27:57-62. [PMID: 26295204 DOI: 10.1515/jbcpp-2015-0035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 07/15/2015] [Indexed: 11/15/2022]
Abstract
BACKGROUND The present study evaluates the possible mechanism of sildenafil citrate (SIL) for the attenuation of renal failure in diabetic nephropathic (DN) animals. METHODS Diabetic nephropathy was induced by a single dose of streptozotocin (STZ) (60 mg/kg, i.p.) and confirmed by assessing the blood and urine biochemical parameters on the 28th day of its induction. The selected DN animals were treated with glimepiride (0.5 mg/kg, p.o.) and SIL (2.5 mg/kg, p.o.) for a period of 6 weeks. Biochemical parameters in blood and urine were estimated after the 29th and 70th day of the protocol for the estimation of the effect of SIL. RESULT There were significant alterations in the blood and urine biochemical parameters in STZ-treated groups which confirmed DN. There was a significant decrease in the triglyceride level in the SIL-only-treated group on the 70th day of the protocol. The histopathology study also suggested that SIL treatment results in the improvement in the podocyte count in DN animals. CONCLUSIONS The present study concludes that SIL improves the renal function by decreasing the triglyceride level and improving the podocyte count in DN animals.
Collapse
|
41
|
Brenneman J, Hill J, Pullen S. Emerging therapeutics for the treatment of diabetic nephropathy. Bioorg Med Chem Lett 2016; 26:4394-4402. [PMID: 27520943 DOI: 10.1016/j.bmcl.2016.07.079] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 07/27/2016] [Accepted: 07/29/2016] [Indexed: 02/06/2023]
Abstract
Diabetic nephropathy (DN) is the most common pathology contributing to the development of chronic kidney disease (CKD). DN caused by hypertension and unmitigated inflammation in diabetics, renders the kidneys unable to perform normally, and leads to renal fibrosis and organ failure. The increasing global prevalence of DN has been directly attributed to rising incidences of Type II diabetes, and is now the largest non-communicable cause of death worldwide. Despite the high morbidity, successful new treatments for DN are lacking. This review seeks to provide new insight on emerging clinical candidates under investigation for the treatment of DN.
Collapse
Affiliation(s)
- Jehrod Brenneman
- Small Molecule Discovery Research, Boehringer-Ingelheim Pharmaceuticals Inc., 900 Ridgebury Rd., Ridgefield, CT 06877, USA.
| | - Jon Hill
- Research Networking, Boehringer-Ingelheim Pharmaceuticals Inc., 900 Ridgebury Rd., Ridgefield, CT 06877, USA
| | - Steve Pullen
- Cardiometabolic Disease Research, Boehringer-Ingelheim Pharmaceuticals Inc., 900 Ridgebury Rd., Ridgefield, CT 06877, USA
| |
Collapse
|
42
|
Shen K, Johnson DW, Gobe GC. The role of cGMP and its signaling pathways in kidney disease. Am J Physiol Renal Physiol 2016; 311:F671-F681. [PMID: 27413196 DOI: 10.1152/ajprenal.00042.2016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 07/10/2016] [Indexed: 01/20/2023] Open
Abstract
Cyclic nucleotide signal transduction pathways are an emerging research field in kidney disease. Activated cell surface receptors transduce their signals via intracellular second messengers such as cAMP and cGMP. There is increasing evidence that regulation of the cGMP-cGMP-dependent protein kinase 1-phosphodiesterase (cGMP-cGK1-PDE) signaling pathway may be renoprotective. Selective PDE5 inhibitors have shown potential in treating kidney fibrosis in patients with chronic kidney disease (CKD), via their downstream signaling, and these inhibitors also have known activity as antithrombotic and anticancer agents. This review gives an outline of the cGMP-cGK1-PDE signaling pathways and details the downstream signaling and regulatory functions that are modulated by cGK1 and PDE inhibitors with regard to antifibrotic, antithrombotic, and antitumor activity. Current evidence that supports the renoprotective effects of regulating cGMP-cGK1-PDE signaling is also summarized. Finally, the effects of icariin, a natural plant extract with PDE5 inhibitory function, are discussed. We conclude that regulation of cGMP-cGK1-PDE signaling might provide novel, therapeutic strategies for the worsening global public health problem of CKD.
Collapse
Affiliation(s)
- Kunyu Shen
- Centre for Kidney Disease Research, School of Medicine, Translational Research Institute, The University of Queensland, Brisbane, Australia; Second School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China; and
| | - David W Johnson
- Centre for Kidney Disease Research, School of Medicine, Translational Research Institute, The University of Queensland, Brisbane, Australia; Department of Nephrology, Princess Alexandra Hospital, Brisbane, Australia
| | - Glenda C Gobe
- Centre for Kidney Disease Research, School of Medicine, Translational Research Institute, The University of Queensland, Brisbane, Australia;
| |
Collapse
|
43
|
El-Mahdy NA, El-Sayad MES, El-Kadem AH. Combination of telmisartan with sildenafil ameliorate progression of diabetic nephropathy in streptozotocin-induced diabetic model. Biomed Pharmacother 2016; 81:136-144. [DOI: 10.1016/j.biopha.2016.04.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Revised: 03/31/2016] [Accepted: 04/01/2016] [Indexed: 02/07/2023] Open
|
44
|
Spilanthol from Acmella Oleracea Lowers the Intracellular Levels of cAMP Impairing NKCC2 Phosphorylation and Water Channel AQP2 Membrane Expression in Mouse Kidney. PLoS One 2016; 11:e0156021. [PMID: 27213818 PMCID: PMC4877099 DOI: 10.1371/journal.pone.0156021] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 05/09/2016] [Indexed: 11/19/2022] Open
Abstract
Acmella oleracea is well recognized in Brazilian traditional medicine as diuretic, although few scientific data have been published to support this effect. Aim of this study was to determine the molecular effect of Acmella oleracea extract and its main alkylamide spilanthol on two major processes involved in the urine concentrating mechanism: Na-K-2Cl symporter (NKCC2) activity in the thick ascending limb and water channel aquaporin 2 accumulation at the apical plasma membrane of collecting duct cells. Phosphorylation of NKCC2 was evaluated as index of its activation by Western blotting. Rate of aquaporin 2 apical expression was analyzed by confocal laser microscopy. Spilanthol-induced intracellular signalling events were dissected by video-imaging experiments. Exposure to spilanthol reduced the basal phosphorylation level of NKCC2 both in freshly isolated mouse kidney slices and in NKCC2-expresing HEK293 cells. In addition, exposure to spilanthol strongly reduced both desmopressin and low Cl−-dependent increase in NKCC2 phosphorylation in mouse kidney slices and NKCC2-expressing HEK293 cells, respectively. Similarly, spilanthol reduced both desmopressin- and forskolin-stimulated aquaporin 2 accumulation at the apical plasma membrane of collecting duct in mouse kidney slice and MCD4 cells, respectively. Of note, when orally administered, spilanthol induced a significant increase in both urine output and salt urinary excretion associated with a markedly reduced urine osmolality compared with control mice. Finally, at cellular level, spilanthol rapidly reduced or reversed basal and agonist-increased cAMP levels through a mechanism involving increases in intracellular [Ca2+]. In conclusion, spilanthol-induced inhibition of cAMP production negatively modulates urine-concentrating mechanisms thus holding great promise for its use as diuretic.
Collapse
|
45
|
Ye H, Wang X, Sussman CR, Hopp K, Irazabal MV, Bakeberg JL, LaRiviere WB, Manganiello VC, Vorhees CV, Zhao H, Harris PC, van Deursen J, Ward CJ, Torres VE. Modulation of Polycystic Kidney Disease Severity by Phosphodiesterase 1 and 3 Subfamilies. J Am Soc Nephrol 2016; 27:1312-20. [PMID: 26374610 PMCID: PMC4849815 DOI: 10.1681/asn.2015010057] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 08/04/2015] [Indexed: 11/03/2022] Open
Abstract
Aberrant intracellular calcium levels and increased cAMP signaling contribute to the development of polycystic kidney disease (PKD). cAMP can be hydrolyzed by various phosphodiesterases (PDEs). To examine the role of cAMP hydrolysis and the most relevant PDEs in the pathogenesis of PKD, we examined cyst development in Pde1- or Pde3-knockout mice on the Pkd2(-/WS25) background (WS25 is an unstable Pkd2 allele). These PDEs were selected because of their importance in cross-talk between calcium and cyclic nucleotide signaling (PDE1), control of cell proliferation and cystic fibrosis transmembrane conductance regulator (CFTR) -driven fluid secretion (PDE3), and response to vasopressin V2 receptor activation (both). In Pkd2(-/WS25) mice, knockout of Pde1a, Pde1c, or Pde3a but not of Pde1b or Pde3b aggravated the development of PKD and was associated with higher levels of protein kinase A-phosphorylated (Ser133) cAMP-responsive binding protein (P-CREB), activating transcription factor-1, and CREB-induced CRE modulator proteins in kidney nuclear preparations. Immunostaining also revealed higher expression of P-CREB in Pkd2(-/) (WS25);Pde1a(-/-), Pkd2(-) (/WS25);Pde1c(-/-), and Pkd2(-/) (WS25);Pde3a(-/-) kidneys. The cystogenic effect of desmopressin administration was markedly enhanced in Pkd2(-/WS25);Pde3a(-/-) mice, despite PDE3 accounting for only a small fraction of renal cAMP PDE activity. These observations show that calcium- and calmodulin-dependent PDEs (PDE1A and PDE1C) and PDE3A modulate the development of PKD, possibly through the regulation of compartmentalized cAMP pools that control cell proliferation and CFTR-driven fluid secretion. Treatments capable of increasing the expression or activity of these PDEs may, therefore, retard the development of PKD.
Collapse
Affiliation(s)
- Hong Ye
- Division of Nephrology and Hypertension and
| | | | | | | | | | - Jason L Bakeberg
- Division of Nephrology and Hypertension, The Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | | | - Vincent C Manganiello
- Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, US National Institutes of Health, Bethesda, Maryland
| | - Charles V Vorhees
- Department of Pediatrics, Division of Neurology, Cincinnati Children's Research Foundation and University of Cincinnati, Cincinnati, Ohio; and
| | - Haiqing Zhao
- Department of Biology, Johns Hopkins University, Baltimore, Maryland
| | | | - Jan van Deursen
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Christopher J Ward
- Division of Nephrology and Hypertension, The Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | | |
Collapse
|
46
|
Design and Microwave Assisted Synthesis of Coumarin Derivatives as PDE Inhibitors. INTERNATIONAL JOURNAL OF MEDICINAL CHEMISTRY 2016; 2016:9890630. [PMID: 26998358 PMCID: PMC4779523 DOI: 10.1155/2016/9890630] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 12/26/2015] [Accepted: 12/27/2015] [Indexed: 12/04/2022]
Abstract
Coumarins appended to benzimidazole through pyrazole are designed and synthesized using microwave irradiation. These compounds were analyzed for phosphodiesterase (PDE) inhibition indirectly by motility pattern in human spermatozoa. Some of the synthesized compounds, namely, 5d, 5e, 5f, 5g, 5h, and 5k, have exhibited potent inhibitory activity on PDE.
Collapse
|
47
|
Yan K, Gao LN, Cui YL, Zhang Y, Zhou X. The cyclic AMP signaling pathway: Exploring targets for successful drug discovery (Review). Mol Med Rep 2016; 13:3715-23. [PMID: 27035868 PMCID: PMC4838136 DOI: 10.3892/mmr.2016.5005] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 02/08/2016] [Indexed: 12/03/2022] Open
Abstract
During development of disease, complex intracellular signaling pathways regulate an intricate series of events, including resistance to external toxins, the secretion of cytokines and the production of pathological phenomena. Adenosine 3′,5′-cyclic monophosphate (cAMP) is a nucleotide that acts as a key second messenger in numerous signal transduction pathways. cAMP regulates various cellular functions, including cell growth and differentiation, gene transcription and protein expression. This review aimed to provide an understanding of the effects of the cAMP signaling pathway and the associated factors on disease occurrence and development by examining the information from a new perspective. These novel insights aimed to promote the development of novel therapeutic approaches and aid in the development of new drugs.
Collapse
Affiliation(s)
- Kuo Yan
- Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, P.R. China
| | - Li-Na Gao
- Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, P.R. China
| | - Yuan-Lu Cui
- Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, P.R. China
| | - Yi Zhang
- Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, P.R. China
| | - Xin Zhou
- Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, P.R. China
| |
Collapse
|
48
|
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: 5.4] [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
|
49
|
Ramirez CE, Nian H, Yu C, Gamboa JL, Luther JM, Brown NJ, Shibao CA. Treatment with Sildenafil Improves Insulin Sensitivity in Prediabetes: A Randomized, Controlled Trial. J Clin Endocrinol Metab 2015; 100:4533-40. [PMID: 26580240 PMCID: PMC4667163 DOI: 10.1210/jc.2015-3415] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Sildenafil increases insulin sensitivity in mice. In humans, phosphodiesterase 5 inhibition improves disposition index, but the mechanism of this effect has not been elucidated and may depend on duration. In addition, increasing cyclic GMP without increasing nitric oxide could have beneficial effects on fibrinolytic balance. OBJECTIVE The objective was to test the hypothesis that chronic phosphodiesterase 5 inhibition with sildenafil improves insulin sensitivity and secretion without diminishing fibrinolytic function. DESIGN This was a randomized, double-blind, placebo-controlled study. SETTING This trial was conducted at Vanderbilt Clinical Research Center. PARTICIPANTS Participants included overweight individuals with prediabetes. INTERVENTIONS Subjects were randomized to treatment with sildenafil 25 mg three times a day or matching placebo for 3 months. Subjects underwent a hyperglycemic clamp prior to and at the end of treatment. MAIN OUTCOME MEASURES The primary outcomes of the study were insulin sensitivity and glucose-stimulated insulin secretion. RESULT Twenty-one subjects completed each treatment arm. After 3 months, the insulin sensitivity index was significantly greater in the sildenafil group compared to the placebo group by 1.84 mg/kg/min per μU/mL*100 (95% confidence interval, 0.01 to 3.67 mg/kg/min per μU/mL*100; P = .049), after adjusting for baseline insulin sensitivity index and body mass index. In contrast, there was no effect of 3-month treatment with sildenafil on acute- or late-phase glucose-stimulated insulin secretion (P > .30). Sildenafil decreased plasminogen activator inhibitor-1 (P = .01), without altering tissue-plasminogen activator. In contrast to placebo, sildenafil also decreased the urine albumin-to-creatinine ratio from 12.67 ± 14.67 to 6.84 ± 4.86 μg/mg Cr. This effect persisted 3 months after sildenafil discontinuation. CONCLUSIONS Three-month phosphodiesterase 5 inhibition enhances insulin sensitivity and improves markers of endothelial function.
Collapse
Affiliation(s)
- Claudia E Ramirez
- Departments of Medicine (C.E.R., J.L.G., J.M.L., N.J.B., C.A.S.) and Biostatistics (H.N., C.Y.), Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Hui Nian
- Departments of Medicine (C.E.R., J.L.G., J.M.L., N.J.B., C.A.S.) and Biostatistics (H.N., C.Y.), Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Chang Yu
- Departments of Medicine (C.E.R., J.L.G., J.M.L., N.J.B., C.A.S.) and Biostatistics (H.N., C.Y.), Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Jorge L Gamboa
- Departments of Medicine (C.E.R., J.L.G., J.M.L., N.J.B., C.A.S.) and Biostatistics (H.N., C.Y.), Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - James M Luther
- Departments of Medicine (C.E.R., J.L.G., J.M.L., N.J.B., C.A.S.) and Biostatistics (H.N., C.Y.), Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Nancy J Brown
- Departments of Medicine (C.E.R., J.L.G., J.M.L., N.J.B., C.A.S.) and Biostatistics (H.N., C.Y.), Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Cyndya A Shibao
- Departments of Medicine (C.E.R., J.L.G., J.M.L., N.J.B., C.A.S.) and Biostatistics (H.N., C.Y.), Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| |
Collapse
|
50
|
In silico approaches for the identification of virulence candidates amongst hypothetical proteins of Mycoplasma pneumoniae 309. Comput Biol Chem 2015; 59 Pt A:67-80. [DOI: 10.1016/j.compbiolchem.2015.09.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 09/08/2015] [Accepted: 09/14/2015] [Indexed: 01/25/2023]
|