Next Article in Journal
Salivary Gland Dysfunction, Protein Glycooxidation and Nitrosative Stress in Children with Chronic Kidney Disease
Next Article in Special Issue
Liver Transplant Patients with High Levels of Preoperative Serum Ammonia Are at Increased Risk for Postoperative Acute Kidney Injury: A Retrospective Study
Previous Article in Journal
Group IIA Secretory Phospholipase A2 Predicts Graft Failure and Mortality in Renal Transplant Recipients by Mediating Decreased Kidney Function
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current Concepts on the Reno-Protective Effects of Phosphodiesterase 5 Inhibitors in Acute Kidney Injury: Systematic Search and Review

by
Georgios Georgiadis
1,†,
Ioannis-Erineos Zisis
1,2,†,
Anca Oana Docea
3,
Konstantinos Tsarouhas
4,
Irene Fragkiadoulaki
1,2,
Charalampos Mavridis
1,
Markos Karavitakis
1,
Stavros Stratakis
5,
Kostas Stylianou
5,
Christina Tsitsimpikou
6,
Daniela Calina
7,
Nikolaos Sofikitis
8,
Aristidis Tsatsakis
2 and
Charalampos Mamoulakis
1,*
1
Department of Urology, University General Hospital of Heraklion, University of Crete, Medical School, Heraklion, Crete, Greece
2
Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, Heraklion, Crete 71003, Greece
3
Department of Toxicology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
4
Department of Cardiology, University General Hospital of Larissa, Larissa, Greece
5
Department of Nephrology, University General Hospital of Heraklion, University of Crete, Medical School, Heraklion, Crete, Greece
6
Department of Hazardous Substances, Mixtures and Articles, General Chemical State Laboratory of Greece, Ampelokipi, Athens, Greece
7
Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
8
Department of Urology, School of Medicine, Ioannina University, Ioannina, Greece
*
Author to whom correspondence should be addressed.
Equal contribution of authors.
J. Clin. Med. 2020, 9(5), 1284; https://doi.org/10.3390/jcm9051284
Submission received: 3 March 2020 / Revised: 16 April 2020 / Accepted: 24 April 2020 / Published: 29 April 2020

Abstract

:
Acute kidney injury (AKI) is associated with increased morbidity, prolonged hospitalization, and mortality, especially in high risk patients. Phosphodiesterase 5 inhibitors (PDE5Is), currently available as first-line therapy of erectile dysfunction in humans, have shown a beneficial potential of reno-protection through various reno-protective mechanisms. The aim of this work is to provide a comprehensive overview of the available literature on the reno-protective properties of PDE5Is in the various forms of AKI. Medline was systematically searched from 1946 to November 2019 to detect all relevant animal and human studies in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) statement. In total, 83 studies were included for qualitative synthesis. Sildenafil is the most widely investigated compound (42 studies), followed by tadalafil (20 studies), icariin (10 studies), vardenafil (7 studies), zaprinast (4 studies), and udenafil (2 studies). Even though data are limited, especially in humans with inconclusive or negative results of only two clinically relevant studies available at present, the results of animal studies are promising. The reno-protective action of PDE5Is was evident in the vast majority of studies, independently of the AKI type and the agent applied. PDE5Is appear to improve the renal functional/histopathological alternations of AKI through various mechanisms, mainly by affecting regional hemodynamics, cell expression, and mitochondrial response to oxidative stress and inflammation.

1. Introduction

AKI is considered a complex disorder with increased morbidity, prolonged hospitalization and mortality especially in high risk patients that may be attributed to various causes (pre-renal; renal, i.e., intrinsic to the renal parenchyma; and post-renal), including the use of nephrotoxic medications such as contrast media (CM), dehydration, sepsis, renal surgery, renal ischemia, ischemia–reperfusion (IR) renal injury, and urinary tract obstruction [1]. Criteria used for the diagnosis of AKI vary widely among studies in humans [2], including percent change in the baseline serum creatinine (sCr) levels (e.g., an increase of variously 25–50%) and absolute elevation from baseline sCr level (e.g., an increase of variously 0.5–2.0 mg/dL) [3]. These variable definitions have been addressed by two consensus groups, namely the Acute Dialysis Quality Initiative (ADQI) proposing the RIFLE (Risk, Injury, Failure, Loss and End-stage kidney disease) system [4] and more recently the Acute Kidney Injury Network (AKIN), which have attempted to standardize the diagnosis of AKI irrespective of etiology. According to the AKIN diagnostic criteria [5], AKI is an abrupt (within 48 h) reduction in human kidney function defined as occurrence of any of the following after a reno-toxic event: (a) absolute increase in sCr ≥ 0.3 mg/dL (≥ 26.4 μmol/L) or a percentage increase in sCr ≥ 50% (1.5-fold from baseline), which is known or presumed to have occurred within the prior seven days [6]; or (b) a reduction in urine output (documented oliguria of < 0.5 mL/kg/h for more than 6 h). This definition is in accordance with the current Clinical Practice Guideline for AKI by “Kidney Disease: Improving Global Outcomes” (KDIGO) [6]. Nevertheless, a recent systematic review evaluating the methods used to investigate AKI biomarkers showed that results are difficult to interpret, not comparable, and not consistently reproducible due to the impact of the variable AKI definitions still used to determine the outcome of interest in human studies (38.0% of the studies used the AKIN; 21.4% used the RIFLE; 20.3% used the KDIGO; and 20.3% used another definition) [2]. Similarly, variable definitions of AKI have been used in animal studies, a fact that has been recognized as an important limitation in translating preclinical findings in clinical studies [7,8] among others [9]. Several reviews of available animal models, including their advantages and disadvantages, have been discussed [10]; however, the types of models are often incomplete and many details, such as model techniques and modeling time, are not mentioned. Currently proposed AKI models include, among others: IR renal injury, including shock wave lithotripsy (SWL); injection of drugs, toxins, or endogenous toxins; ureteral obstruction, contrast-induced nephropathy (CIN); trauma such as burn; etc. [10,11,12,13,14,15,16].
Depending on the insult type, there are various mechanisms leading to renal damage such as renal vasoconstriction [17], vascular endothelial damage, cytokine expression [18], increase of IL-18, mediating acute tubular necrosis, caspase activity stimulation, p53 up-regulation [19], accumulation of toxic metabolites [20], mast cells/neutrophils activation, reactive oxygen species (ROS) generation causing lipid peroxidation that leads to cellular membrane destruction, excessive intracellular DNA breakdown, energy depletion, intracellular Ca2+ elevation, higher inducible nitric oxide (NO) synthase (iNOS) expression, NO deficiency, intra-parenchymal hemorrhage [21], fibrosis, direct cellular toxicity, tubular obstruction, vascular congestion, activation of angiotensin II axis [22], mitochondrial dysfunction [23], cell cycle arrest in G2 phase, ATPase activity inhibition, and cellular transport modification. ROS activate pro-apoptotic proteins eventually promoting Bax translocation (regulated by PI3K/Akt pathway) to the outer mitochondrial membrane, causing the release of cytochrome c in the cytosol [24]. Bax is also responsible for caspase 9 activation that activates caspase 3, triggering apoptosis. The tubular component of AKI consists of injured, necrotic/apoptotic cells falling into the lumen that cause obstruction/back leak of the filtrate to the interstitial space, inducing inflammation.
CIN is a real, albeit rare, entity in current clinical medical practice that represents a serious iatrogenic AKI form, occurring 24–72 h after administration of iodinated contrast media (CM) during angiographic or other procedures, such as urography [3,25]. The exact pathophysiology of CIN is not fully elucidated but oxidative stress is considered a major mechanism in CIN [26], and the identification of novel biomarkers that may more accurately detect renal function changes, reflect kidney damage, assist monitoring, and elucidate pathophysiology have attracted considerable scientific attention nowadays [27]. CM passing through the kidney results in an intense tubular transport that increases the activity in the thick ascending limb of Henle’s loop. This process increases oxygen consumption/metabolic activity of outer renal medulla, exacerbating the marginal hypoxic conditions. Prostanoids and NO are mainly responsible for the medullary vasodilatory response [28]. Therefore, any NO deficit may contribute to an additional hypoxic renal insult. CIN and IR renal injury share common pathways regarding the vasodilatory potential of NO. IR renal injury is a common complication during renal transplantation/artery angioplasty, partial nephrectomy, cardiopulmonary/aortic bypass surgery, and others [29]. In the IR renal injury setting, however, there are conflicting results reported, with some studies suggesting that NO induces cytotoxicity, and others showing that increased NOS activity is linked to increased renal blood flow in the ischemic region [30]. NOSs are a family of enzymes catalyzing the production of NO from L-arginine. There are three isoforms: the endothelial NOS (eNOS), the neuronal NOS (nNOS), and the iNOS involved in immune response. In the IR renal injury, endogenous NO is synthesized by eNOS and iNOS [31], while it is found that eNOS-mediated NO production plays a pivotal protective role in IR-induced AKI [1]. IR renal injury is also closely linked to ROS generation/apoptosis.
Prevention and/or management of the various AKI forms, such as CIN, is mainly supportive at present, consisting of intravenous hydration [32]. Even though the potential beneficial effects of many agents with antioxidant properties have been tested, the value of such substances other than sodium bicarbonate remains controversial [32,33]. Phosphodiesterase 5 (PDE5) inhibitors (PDE5Is) are currently recommended as first-line therapy of erectile dysfunction (ED) by enhancing the vasodilatory effects of NO [34]. Acting via the selective inhibition of cyclic guanosine monophosphate (cGMP)-specific PDE5 that metabolizes cGMP, the principal mediator of NO-induced smooth muscle relaxation, PDE5Is cause vasodilatation in the corpora cavernosa promoting erection (Figure 1). This class of drugs has shown beneficial potential through various mechanisms in some CIN animal models [33]. The aim of this paper is to provide a comprehensive overview of the available literature on the potential reno-protective properties of PDE5Is in the various forms of AKI.

2. Experimental Section

Medline (Ovid Medline Epub Ahead of Print, In-Process & Other Non-Indexed Citations, Ovid MEDLINE(R) Daily, and Ovid MEDLINE(R) 1946 to November 2019) was systematically searched to detect all relevant animal and human studies in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) statement [37], using the following keyword combinations (Medical Subject Headings; MeSH): PDE5i or avanafil or benzamidenafil or dasantafil or icariin or lodenafil or mirodenafil or sildenafil or tadalafil or udenafil or vardenafil or zaprinast combined with renal or kidney or nephrotoxicity or contrast or CIN or AKI or nephrotoxic or cisplatin or aminoglycoside or trauma or acute kidney injury or NSAIDS or non-steroidal or shock or sepsis or hypoperfusion or hypovolaemia or hypovolemia or renal artery stenosis or obstruction or acute tubular necrosis or glomerulonephritis or nephritis or renal failure or adenine or cyclosporine. The specific literature search strategy used is available in Appendix A. The reference lists of selected studies were screened for other potentially eligible studies. After excluding duplicates, citations in abstract form, and non-English citations, the titles/abstracts of full papers were screened for relevance, defined as original research focusing on the topic “nephropathy AND effects of phosphodiesterase 5 inhibitors”. Studies focusing on alterations of renal function and/or structure for >3 months (conventionally considered as following the KDIGO definition of chronic kidney disease (CKD) were excluded [6]). Two review authors (G.G. and IE.Z.) independently scanned the title and the abstract content, or both, of every record retrieved to determine which studies should be assessed further evaluated and extracted all data. Disagreements were resolved through consensus or by consultation with a third author (C.M.). A final draft of the manuscript was prepared after several revisions and approved by all authors.

3. Results

In total, 83 studies were included for qualitative synthesis (Figure 2). Among the 11 natural/synthetic agents currently available (avanafil, benzamidenafil, dasantafil, icariin, lodenafil, mirodenafil, sildenafil, tadalafil, udenafil, vardenafil, and zaprinast), sildenafil is the most widely investigated (n = 42 studies), followed by tadalafil (n = 20 studies), icariin (n = 10 studies), vardenafil (n = 7 studies), zaprinast (n = 4 studies), and udenafil (n = 2 studies). No studies on lodenafil, benzamidenafil, mirodenafil, avanafil, or dasantafil were detected. Most of the studies (n = 79) used animal models, including among others currently proposed AKI models (IR renal injury, including SWL; injection of drugs, toxins, or endogenous toxins; ureteral obstruction; CIN; trauma such as burn; etc.) [10,11,12,13,14,15,16] and variable definitions of AKI in line with the situation observed in human studies [2]. Only four human studies were detected: two preclinical studies utilizing human tissue [24,38] and two clinical trials [17,39].
The reno-protective action of PDE5Is was evident in the vast majority of studies (n = 81), independently of the AKI type and the agent applied. Only one human study on sildenafil [39] and one animal study on zaprinast [40] failed to reveal any reno-protective action of PDE5Is, showing a neutral effect. PDE5Is appeared to be beneficial in AKI of various etiologies by improving renal functional/histopathological alternations through various mechanisms, such as affecting regional hemodynamics, cell expression, and mitochondrial response to oxidative stress and inflammation.
The main characteristics and results of the human studies evaluating the potential reno-protective effects of PDE5Is are summarized in Table 1 [17,24,38,39]. The main characteristics and results of the animal studies on currently proposed AKI models evaluating the potential reno-protective effects of sildenafil, tadalafil, icariin, vardenafil, zaprinast–udenafil are summarized in Table 2 [23,30,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61], Table 3 [29,35,45,49,62,63,64,65,66,67,68,69,70,71,72,73,74], Table 4 [18,75,76], Table 5 [45,77,78], and Table 6 [21,40,79,80], respectively. The main characteristics and results of the animal studies in the AKI-CKD transition spectrum (focusing on renal function and/or structure alterations for up to three months, not fulfilling the KDIGO definition for CKD [6]) evaluating the potential reno-protective effects of sildenafil, tadalafil, icariin, vardenafil, zaprinast–udenafil are summarized in Table A1 [19,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98], Table A2 [99,100], Table A3 [22,101,102,103,104], Table A4 [105,106,107,108], and Table A5 [109,110], respectively (Appendix B).

4. Discussion

PDE5Is have received a lot of attention since the first drugs were launched in the market. Four potent selective agents (avanafil, sildenafil, tadalafil, and vardenafil) have been approved by the European Medicines Agency (EMA) and the Food and Drug Administration (FDA) for the treatment of ED [111,112]. ED can be managed successfully with currently available treatment options, but it cannot be cured and most patients will be treated without cause-specific options, such as the use of PDE5Is [34]. Exceptions are psychogenic, post-traumatic arteriogenic in young patients, and hormonal causes (e.g., hypogonadism) of ED, which are potentially curable with specific treatments that might be employed first, when such causes are detected [34]. Consequently, treatment strategy of ED should be tailored depending on invasiveness, efficacy, safety, cost, and patient preference of the currently available options; in the context of this strategy, PDE5Is are currently recommended strongly as first-line treatment option given that lifestyle changes are initiated/risk factors are modified prior to or at the same time as initiating ED treatment [34].
Other EMA/FDA approved indications of PDE5Is include pulmonary arterial hypertension (PAH) (sildenafil and tadalafil) and management of men with moderate to severe LUTS secondary to benign prostatic obstruction with or without ED (tadalafil) [34,113,114,115]. Besides the aforementioned agents, there are other non-EMA/FDA approved PDE5Is including benzamidenafil, dasantafil, lodenafil, mirodenafil, and udenafil, some of which are commercially available in a few countries (lodenafil in Brazil; mirodenafil in South Korea; and udenafil in South Korea, Russia, and Philippines) [113]. Other agents with weak PDE5I properties include icariin and zaprinast [116]. Icariin, a prenylated flavonol glycoside extracted from plants of the Epimedium genus, has demonstrated PDE5I activity in vitro, enhancement of NO, and antioxidant activity [116]. It has been widely used in Chinese traditional medicine. It shows peak concentration levels at 1 h and should be avoided in patients with bleeding disorders, hypotension, arrhythmias, and hormone-sensitive cancers (breast, ovarian, or prostate). Zaprinast is an inhibitor of PDE5, PDE6, PDE9, and PDE11. In the past, it has been used for the treatment of PAH and inhibition of malaria parasites. Zaprinast activates the G-protein coupled receptor, GPR35, that plays a crucial role in cardiovascular disease, pain, regulation of inflammation, hypertension, diabetes, and irritable bowel disease [117,118]. The main characteristics of PDE5Is are summarized in Table 7 [34,112,113,119,120,121,122,123,124,125].
PDE5Is interfere selectively with cGMP hydrolysis by PDE5, increasing intracellular cGMP, which results in smooth muscle relaxation/raised arterial blood flow improving penile erection. PDE5 belongs to a superfamily of enzymes that convert intracellular cAMP/cGMP into the consonant nucleotides. It is a cytosolic protein with three isoforms expressed in various organs apart from the penis (corpora cavernosa), including kidney (vessels, glomeruli, inner medullary collecting ducts, and cortical tubules) that specifically degrades cGMP [66]. In particular, PDE5A1 and PDE5A2 are widely expressed in tubular epithelial cells of the renal proximal tubule and medullary collecting duct, as well as in vascular smooth muscle cells, platelets, brain, and lung, while PDE5A3 is only expressed in vascular smooth muscle cells [126].
Cyclic nucleotide signal transduction pathways represent an emerging research field in kidney disease, with selective PDE5 inhibition attracting ongoing interest nowadays [127]. Current evidence supports the notion that regulation of the cGMP -dependent protein kinase 1-PDE signaling pathway may be reno-protective and that its regulation might provide novel, therapeutic strategies for chronic kidney disease with selective PDE5Is having shown potential in treating kidney fibrosis, while possessing antithrombotic and anticancer activity [128]. In this respect, PDE5Is represent a potential but still understudied/controversial option against various forms of AKI such as CIN [28], given that NO/cGMP are crucial mediators in renal vasculature and NO is an essential endogenous vasodilator for medullary oxygenation [33].
The mechanism of action of PDE5Is in the prevention and management of AKI is still not fully elucidated. Multiple mechanisms have been proposed to play a role in counteracting the cascade of changes caused by the renal injury. Stimulation of NO production through NOS, medullary blood flow improvement, protection against vascular endothelial damage, Bcl2/Bax ratio reversal, ERK phosphorylation, mitochondrial biogenesis activation, renal progenitor cell upregulation, and the regulation of multiple signaling pathways such as insulin/IGF1, T17/Treg, PI3K/Akt, and NF-kB [75] are the most well-described mechanisms through which PDE5Is offer protection. The increased ERK phosphorylation boosts NOS activity and subsequent rapid NO release [30]. The repairing process following any renal injury requires energy provided by the cellular mitochondria. Mitochondria are continuously regenerated but cellular injury such as sepsis and hypoxia induce rapid biogenesis. This process is mediated by a transcriptional co-activator, peroxisome proliferator-activated receptor γ co-activator 1a (PGC-1a). PGC-1a activates the nuclear respiratory factors 1 and 2, which eventually activate mitochondrial transcription factor A that is responsible for the transcription of mitochondrial DNA [23,67]. An alternative process that PDE5Is activate to promote recovery from renal injury is the renal progenitor cell stimulation. PDE5Is, more specifically icariin, upregulates HGF, WT-1, and BMP-7, which lead to an increased number of CD133+ and CD24+, cells that are capable of self-renew and also differentiate into podocytes or tubular cells [57,103]. In addition to the aforementioned actions, PDE5Is are likely to exert their protective effect through an alternative pathway. PDE5Is increase cGMP, which activates protein kinase G that opens mitochondrial KATP channels that induce depolarization of the mitochondrial inner membrane and Mg2+ release. The depolarized membrane results in reduced Ca2+ influx; therefore, suppressed cellular death and increased Mg2+ concentration reduces ROS and lessens p38 MAPK activation, which is responsible for apoptosis [30,88,129]. The most common reno-protective mechanisms of PDE5Is are summarized in Figure 3.
To the best of our knowledge, this is the first review that attempts in a systematic way to define the reno-protective potential of PDE5Is in the various forms of AKI. Based on our results, it appears that sildenafil is the most widely PDE5I studied in AKI among the 11 natural/synthetic agents currently available (avanafil, benzamidenafil, dasantafil, icariin, iodenafil, mirodenafil, sildenafil, tadalafil, udenafil, vardenafil, and zaprinast).
The reno-protective effects of PDE5Is have been evaluated in four human studies to date (preclinical studies using human cells: n = 2 [24,38]; clinical studies: n = 2 [17,39]) (Table 1). In one study, human umbilical cord mesenchymal stem cells (huMSC), which have a high self-renewal/multi-directional differentiation potential, were treated with icariin and administered in an animal model of renal injury induced by adenine [38]. Blood urea nitrogen/sCr analysis showed amelioration of functional parameters. Icariin-treated huMSC increased the number of cells in injured renal tissues, reduced fibrosis, oxidative damage, inflammatory responses, and promoted expression of growth factors protecting injured renal tissue. In another study, cisplatin was added to human embryonic kidney (HEK)-293 renal cell cultures pre-treated with icariin [24]. The authors concluded that icariin prevents cisplatin-induced HEK-293 cell injury by inhibiting oxidative stress, inflammatory response, and cellular apoptosis partly via regulating nuclear factor kappa-like chain-enhancer of activated B cells (NF-κB) and PI3K/Akt signaling pathways. In a non-randomized clinical trial, 49 patients with renal tumors were submitted to open nephron-sparing surgery after renal artery clamping [17]. Twenty-two patients were pre-treated with tadalafil one day pre- and two days post-operatively and 27 patients underwent the same surgery without receiving tadalafil. Renal artery clamping induced kidney dysfunction reflected by increases in urinary NGAL and KIM-1 (two novel biomarkers for AKI) in all participants. Tadalafil reduced the urinary excretion of KIM-1, but not of NGAL. The incidence of AKI was comparable between groups but sCr elevation was significantly attenuated in the tadalafil-treated group compared to controls. It was concluded that tadalafil exerts reno-protective effects in AKI following nephron-sparing surgery. In a randomized placebo-controlled trial, 40 patients were submitted to robot-assisted partial nephrectomy after hilar clamping. The reno-protective effect of a single 100 mg oral dose of sildenafil immediately prior to clamping was evaluated [39]. GFR was similarly decreased between arms during the immediate postoperative period and at an intermediate-term follow-up of six months; the reno-protective effect of sildenafil was not evident in this study (neutral effect).
All animal studies investigating the potential reno-protective effect of sildenafil (n = 41) manifested a beneficial effect, irrespectively of the mechanism of AKI; almost all parameters evaluated (biochemical or morphological) were reported to improve (Table 2 and Table A1). Similarly, all animal studies investigating the potential reno-protective effect of tadalafil (n = 19) revealed beneficial outcomes (attenuated histopathological changes/improved biochemical profile; Table 3 and Table A2). Two studies provided comparative results for sildenafil and tadalafil, demonstrating the superiority of the former against tubular cell apoptosis, oxidative status, lipid peroxidation and NOS alterations [45,49]. Unique proteins, cells, and genes have been utilized to investigate the aftermath following icariin’s administration as a reno-protective agent, such as connective tissue growth factor, TUNEL positive cells, nephrin, α-smooth muscle actin, E-cadherin, LY6G, F4/80, NLRP3, NF-κΒ, etc. All available animal studies evaluating icariin (n = 8) showed a beneficial effect (oxidative injury reversal, obliteration of renal function impairment, and improvement of renal hemodynamics/NO sensitivity; Table 4 and Table A3). Similar to sildenafil/tadalafil, icariin suspends the inflammatory response initiation as well as the alteration of the cellular phase and preserves renal morphology. Finally, vardenafil, zaprinast, and udenafil have been investigated in a limited number of studies (n = 7, n = 4, and n = 2, respectively), almost all of which show antioxidant, anti-inflammatory, and reno-protective effects (Table 5, Table 6, Table A4, and Table A5). In one study, vardenafil was compared to sildenafil and tadalafil in an animal model of partial unilateral ureteric obstruction, reporting that all agents were beneficial with sildenafil showing best results [45]. Only one study failed to demonstrate any beneficial effect from zaprinast pre-treatment in an animal model of nephrectomy and concomitant contra-lateral renal hilar occlusion [40]. Even though data are still limited, especially in humans with inconclusive or negative results of only two clinically relevant studies available at present, the results of animal studies are promising and have already fueled clinical research, which is on-going with results expected to come out in the near future [122]. Nevertheless, the potential reno-protective capacity of PDE5Is in AKI warrants further investigation in clinical trials.

5. Conclusions

PDE5Is appear to be beneficial in AKI of various etiologies by improving renal functional/histopathological alternations through various mechanisms, such as by affecting regional hemodynamics, cell expression, and mitochondrial response to oxidative stress and inflammation. The reno-protective action of PDE5Is was evident in the vast majority of the studies, independently of the AKI type and the agent applied. Even though data are still limited, especially in humans with inconclusive or negative results of only two clinically relevant studies available at present, the results of animal studies are promising. The potential reno-protective capacity of PDE5Is in AKI warrants further investigation in clinical trials.

Author Contributions

Conceptualization, C.M. (Charalampos Mamoulakis); methodology, G.G., I.-E.Z., and C.M. (Charalampos Mamoulakis); validation, A.O.D., K.T., C.T., D.C., and A.T.; formal analysis, G.G. and I.-E.Z.; investigation, G.G. and I.-E.Z.; data curation, A.O.D., K.T., I.F., C.M. (Charalampos Mavridis), M.K., S.S., K.S., C.T., D.C., N.S., A.T., and C.M. (Charalampos Mamoulakis); writing—original draft preparation, G.G. and I.-E.Z.; writing—review and editing, A.O.D., K.T., I.F., C.M. (Charalampos Mamoulakis), M.K., S.S., K.S., C.T., D.C., N.S., A.T., and C.M. (Charalampos Mamoulakis); visualization, G.G., I.-E.Z., A.O.D., K.T., C.T., and D.C.; supervision, C.M. (Charalampos Mamoulakis); and funding acquisition, C.M. (Charalampos Mamoulakis). All authors have read and agreed to the published version of the manuscript.

Funding

The authors would like to thank the Special Research Account of the University of Crete for supporting the publication of this study (ELKE No 3550).

Acknowledgments

The authors would like to thank D. Pantartzi, Scientific Secretary of the Clinical Trial Office of the Department of Urology, University of Crete, Medical School, Heraklion, Crete, Greece, for the administrative and technical support.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ADQI: Acute Dialysis Quality Initiative; AKI, acute kidney injury; AKIN, Acute Kidney Injury Network; Anti-dsDNA, antibody to double stranded DNA; Bax, proapoptotic protein; Bcl-2, antiapoptotic gene; BP, blood pressure; bFGF, basic fibroblast growth factor; Cmax, serum maximum concentration; BMP-7, bone morphogenetic protein-7; cAMP, cyclic adenosine monophosphate; cGMP, cyclic guanosine monophosphate; CHF, chronic heart failure; CIN, contrast-induced nephropathy; CKD, chronic kidney disease; CM, contrast media; COX, cyclo-oxygenase; CVD, cardiovascular disease; CYP3A4, cytochrome P450 3A4; DNA, deoxyribonucleic acid; DOCA, deoxycorticosterone acetate; ED, erectile dysfunction; EMA, European Medicines Agency; eNOS, endothelial nitric oxide synthase; ERK, extracellular signal-regulated kinases; FDA, Food and Drug Administration; GTP, guanosine-5′-triphosphate; GC, guanylyl cyclase; GFR, glomerular filtration rate; GSH, glutathione; HEK, human embryonic kidney cells; HGF, hepatocyte growth factor; HIF-2a, hypoxia induced factor 2a; HO-1, heme oxygenase 1; HSP-70, Heat shock protein 70, huMSCs, human umbilical cord mesenchymal stem cells; ICAM-1, intercellular adhesion molecule-1; ID, insufficient data; IgG, immunoglobulin G; IL, interleukin; iNOS, inducible nitric oxide synthase; IR, ischemia reperfusion; KDIGO, Kidney Disease, Improving Global Outcomes; KIM-1, kidney injury molecule-1; KO, knockout; LNAME, N(ω)-nitro-L-arginine methyl ester; MeSH, medical subject headings; Mac387, macrophage antibody; MDA, malondialdehyde; NAC, N-acetylcysteine; NF-Kb, nuclear factor kappa light-chain-enhancer of activated B cells; NGAL, neutrophil gelatinase-associated lipocalin; NO, nitric oxide; NOS, nitric oxide synthase; Nrf-2, nuclear factor erythroid 2-related factor-2; NQO1, NADPH quinine oxidoreductase 1; NYHA, New York Heart Association; OD, once daily; PAH, pulmonary arterial hypertension; PDE5, phosphodiesterase 5; PDE5Is, phosphodiesterase 5 inhibitors; PGC-1a, peroxisome proliferator-activated receptor γ coactivator 1a; PKG, protein kinase G; PRISMA, preferred reporting items for systematic reviews and meta-analyses; RIFLE, risk, injury, failure, loss and end-stage kidney disease; ROS, reactive oxygen species; SBP, systolic blood pressure; sCr, serum creatinine; Tfam, mitochondrial transcription factor; TGF-β1, transforming growth factor beta; Tmax, transport maximum; TSP, thrombospondin; TUNEL, terminal deoxynucleotidyl transferase dUTP nick end labeling; TNF-a, tumor necrosis factor -a; VEGF, vascular endothelial growth factor; WT-1, Wilms’ tumor 1 gene.

Appendix A. Literature Search Strategy

Database: Ovid Medline Epub Ahead of Print, In-Process & Other Non-Indexed Citations, Ovid MEDLINE(R) Daily and Ovid MEDLINE(R) 1946 to November 2019.
Search Strategy:
((((((((((((PDE5i[Title/Abstract]) OR avanafil[Title/Abstract]) OR benzamidenafil[Title/Abstract]) OR dasantafil[Title/Abstract]) OR icariin[Title/Abstract]) OR lodenafil[Title/Abstract]) OR mirodenafil[Title/Abstract]) OR sildenafil[Title/Abstract]) OR tadalafil[Title/Abstract]) OR vardenafil[Title/Abstract]) OR udenafil[Title/Abstract]) OR zaprinast[Title/Abstract]) AND (((((((((((((((((((((((((renal[Title/Abstract]) OR kidney[Title/Abstract]) OR nephrotoxicity[Title/Abstract]) OR contrast[Title/Abstract]) OR CIN[Title/Abstract]) OR AKI[Title/Abstract]) OR nephrotoxic[Title/Abstract]) OR cisplatin[Title/Abstract]) OR aminoglycoside[Title/Abstract]) OR trauma[Title/Abstract]) OR acute kidney injury[Title/Abstract]) OR nsaids[Title/Abstract]) OR non steroidal[Title/Abstract]) OR shock[Title/Abstract]) OR sepsis[Title/Abstract]) OR hypoperfusion[Title/Abstract]) OR hypovolaemia[Title/Abstract]) OR renal artery stenosis[Title/Abstract]) OR obstruction[Title/Abstract]) OR acute tubular necrosis[Title/Abstract]) OR glomerulonephritis[Title/Abstract]) OR nephritis[Title/Abstract]) OR renal failure[Title/Abstract]) OR cyclosporine[Title/Abstract]) OR adenine[Title/Abstract]).

Appendix B. Animal Studies in the AKI-CKD Transition Spectrum (Focusing on Renal Function and/or Structure Alterations for up to Three Months, Not Fulfilling the KDIGO Definition for CKD [6])

Table A1. Animal studies evaluating the potential reno-protective effects of sildenafil.
Table A1. Animal studies evaluating the potential reno-protective effects of sildenafil.
Reference
/Country/Year
Studied AnimalModelPDE5I RouteTimingSampleRenal Injury EffectsPDE5I Renal EffectsOutcome
[81]/
Venezuela/2005
Male Sprague Dawley rats5/6 nephrectomySildenafil
Orally
2.5 mg/kg/day
Either immediately after nephrectomy for 8 weeks
Or 4 weeks after nephrectomy for 4 weeks
POST8 weeks
↑sCr, ↑SBP, ↑proteinuria
↓urinary NOX, ↓cGMP
Glomerulosclerosis
Tubulo-interstitial damage
Macrophage accumulation
Increased number of apoptotic cells
↓sCr, ↓SBP, ↓proteinuria
↑ urinary NOX, ↑cGMP
All changes improved especially if PDE5i was given early
POS
[82]/
Spain/2007
Laboratory large-white pigsRight single nephrectomy after 45 min of vascular clamping
+ Auto-transplantation
+ Left nephrectomy
Sildenafil
Orally
100 mg, 2 h pre-op
PRE0/15/30/45/60
min after unclamping
↓RVF, ↑RVR, ↓NO
Minimal differences in tubular and endothelial structure
↑↓RVF, ↓RVR, ↑NO
Minimal differences in tubular and endothelial structure
POS
[83]/
Egypt/2008
Adult male Wistar albino ratsL-NAME
Orally
50 mg/kg/day for 4 weeks
Sildenafil
Orally
5 mg/kg/day
2 weeks after L-NAME for 2 weeks
POSTAt 4 weeks
↓NO, ↓cGMP
Glomerular collapse/mesangial matrix expansion with minimal cellular proliferation
↑NO, ↑cGMP
Improvement in histological alterations
POS
[84]/
Spain/2009
Laboratory minipigsRight single nephrectomy after 45 min of vascular clamping
+Auto-transplantation
+Left nephrectomy
Sildenafil
Orally
100 mg, 1.5 h pre-op
PRE0/15/30/45/60
min after unclamping
↓RVF, ↓RVR
↑RVF, ↓ RVR, ↑NO
Reduced tubular edema, Improved endothelial cell integrity and mitochondrial ultrastructure
POS
[85]/
Korea/2009
Male Sprague DawleyStreptozotocin
Single intravenous dose 60 mg/kg
Sildenafil
Orally
3 mg/kg/day
For 8 weeks
POSTAt 8 weeks↑glucose, ↑urine output
↑urine 8-OH dG
↑urine albumin
↑Kidney/BW ratio, ↑iNOS
↑Nitrotyrosine, ↑MCP-1 ↑ED-1
↑glucose, ↓urine output
↓urine 8-OH dG
↓urine albumin
↓Kidney/BW ratio, ↓iNOS
↓Nitrotyrosine, ↓MCP-1, ↓ED-1
POS
[86]/
Korea/2012
Male Sprague Dawley ratsUnilateral Nephrectomy and 1 week later DOCA strip 200 mg/kg implantationSildenafil
Orally
50 mg/kg/day
2 weeks after DOCA for 2 weeks
POSTAt 4 weeks↑mortality, ↑SBP, ↑ Kidney weight
↓CrCl, ↑sCr, ↑FeNa, ↑ACR
↑ED-1, ↑BAX, ↓Bcl2, ↑aSMA, ↑TGF-b1
↑TUNEL +ve, ↑fibronectin
↑mRNA TGF-β1/MCP-1
↑ICAM 1t
Tubular casts/Tubular obstruction/Vessel dilatation/Glomerulosclerosis/interstitial expansion
↓mortality, ⇔ SBP, ↓Kindey weight
↑CrCl, ↓sCr, ↓FeNa, ↓ACR ↓aSMA
↓ED-1, ↓BAX, ↑Bcl2, ↓TGF-b1
↓TUNEL +ve, ↓fibronectin
↓mRNA TGF-β1/MCP-1/↓ mRNA ICAM1
Reversed all renal injuries
POS
[87]/
Venezuela/2012
Male Wistar rats5/6 nephrectomySildenafil
Orally
5 mg/kg/day for 60 days
24 h after nephrectomy
POSTEvery 2 weeks↑Kidney hypertrophy
↑Proteinuria, ↓NO2/NO3 ↓GMP (urine) ↑Nitrotyrosine
↓Kidney hypertrophy
↓Proteinuria,
↑NO2/NO3
↑cGMP (urine) ↓Nitrotyrosine
POS
[19]/
Egypt/2013
Male Wistar ratsCyclosporine A
Subcutaneously
20 mg/kg/day
21 days
Sildenafil
Orally
5 mg/kg/day
21 days
POSTAt 21st day–urine sample/blood sample/renal tissue excision↑BUN, ↑sCr, ↑MDA levels
↑Urine Albumin/Cr
↓GSH/NO/catalase activity
↑iNOS, TNF-a, Caspase 3 activity
Tubular degeneration and dilation and necrosis/Glomerulat damage/Congestion
Dilated Bowman’s space/Hemorrhage
↓BUN, ↓sCr, ↓MDA levels
↓Urine Albumin/Cr, ↑eNOS
↑GSH/NO/catalase activity
↓iNOS, TNF-a, Caspase 3 activity
Improved all histological changes
POS
[88]/
United Kingdom/2014
Porcine kidneys20 min warm ischemia followed by 2 or 18 h of cold storageSildenafil
Intravenously
1.4 mg/kg
10 min prior to injury and 20min after reperfusion
PRE and POSTSamples during reperfusion↓RBF, ↑intrarenal resistance
↓Urine cGMP ↑ sCr
Steady increase of K+
↑Tubular injury
No difference in groups

Tubular dilatation and debris and interstitial edema/Ischemic changes
↑RBF, ↓intrarenal resistance
↑Urine cGMP, ↓sCr

No significant difference in K+
No effect on tubular injury
regarding GAL/Endothelin1 Slight improvement of histological
POS
[89]/
Brazil/2014
[90]/
Brazil/2014
C57BL/6 miceLeft renal artery clamping for 2 weeksSildenafil
Orally
40 mg/kg/day
2 weeks post op for 2 weeks
POST4 weeksLeft kidney atrophy (clipped)
Right kidney hypertrophy

↓BW, ↑SBP, ↑HR
↑Intrarenal angiotensin I/II
⇔Plasma angiotensin I/II/1-7
↓NO, ↑ONOO-, ↑O2-
Impaired vasodilation response to Ach
↓Left kidney atrophy
↓Right kidney hypertrophy

Normal BW, ↓SBP, ↓HR
↓Intrarenal angiotensin I/II
↑Plasma angiotensin 1-7
↑NO, ↓ONOO-, ↓O2-
Normal vasodilation response to Ach
POS
[91]/
Egypt/2016
White albino male ratsStreptozocin
Single intraperitoneal
dose 55 mg/kg
Sildenafil
Orally
3 mg/kg/day
For 8 weeks after Diabetic nephropathy
POST8 weeks
↓SOD, ↑TGF-β1, ↓NO, ↑sCr
↑BUN, ↑proteinuria
↑Kidney IL-1β
↑Advanced glycation end products (AGEPs)
↑SOD, ↓TGF-β1, ↑NO, ↓sCr
↓BUN, ↓proteinuria
↓Kidney IL-1β
↓Advanced glycation end products (AGEPs)
POS
[92]/
India/2016
Sprague-Dawley ratsStreptozocin
Single intraperitoneal
dose 60 mg/kg
Sildenafil
Orally
2.5 mg/kg/day for 6 weeks after 28 days
POSTAt 28th and 70th day↑sCr, ↑BUN, ↓CrCl
↑Total protein excretion ↑albumin (urine)
Bowman’s capsule thickening, glomerular sclerosis
↓sCr, ↓BUN, ↑CrCl
↓Total protein excretion
↓albumin (urine)
Histopathology improvement
POS
[93]/
Italy/2017
Male CD-1 miceStreptozotocin
Single intraperitoneal dose 150 mg/kg
Sildenafil
Intraperitoneally
1.6 mg/kg
3 days after STZ, for 4 weeks
POST ↑Glucose (urine), ↑MAP, ↓GFR
↑urinary ACR, ↑NGAL, ↑RRI
↓Renal volume, ↓BMP7
↑suPAR, ↑Vascular leakage
↑FITC-dextran extravasation
Reduced glomerular diameter/focal and segmental hyperplasia with diffuse mesangial proliferation/increased mesangial matrix deposition/acute tubular degeneration/eosinophilia/proximal tubule basal membrane thickening
↓Urine glucose, ↓MAP, ↑GFR
↓urinary ACR, ↓NGAL, ↓RRI
↑Renal volume, ↑BMP7, ↓miR-22
↓suPAR, ↓Vascular leakage
↓FITC-dextran extravasation
Reduced mesangial matrix deposition
POS
[94]/
Egypt/2017
Adult male Sprague-Dawley ratsDoxorubicin
Intraperitoneally
3.5 mg/kg
Twice weekly for 3 weeks
Sildenafil
Orally
5 mg/kg/day for 21 days
POST ↑Urea, ↑sCr, ↑uric acid ↑MDA, ↓GSH, ↑TNF-a ↑caspase-3
Eosinophilic casts, tubule degeneration, vacuolization, endothelial cell edema
↓Urea, ↓sCr,
↓uric acid ↓MDA, ↑GSH, ↓TNF-a ↓caspase-3
Histological improvement
POS
[95]/
South Africa/2017
Nulliparous pregnant female Sprague-Dawley ratsL-NAME
Orally
0.3 g/L (drinking water)
4-8 days for EOPE
+
8-14 days for LOPE
Sildenafil
Orally
10 mg/kg
4-8 days for EOPE
8-14 days for LOPE
POSTGestational Day 19↑BP, ↑Urine excretion
↑Urinary nephrin mRNA
↑Podocin (urine), ↑sFlt-1(mRNA)
↓VEGF (mRNA), ↓PIGF
Glomerular and tubular damage and mononuclear cell infiltration
↓BP
↓Urinary nephrin mRNA
↓Podocin (urine)
↓sFlt-1 (mRNA)
↑VEGF (mRNA), ↑PIGF levels
Attenuated histopathological changes
POS
[96]/
Netherlands/2017
RatsAdriamycin
Or
Streptozocin
Sildenafil
Orally
5 mg/kg/day for 6 weeks
POSTImmortalized mouse podocytes
+
Mouse kidney cortex
↑TRPC6 expression
↓Nephrin, ↑Glomerular desmin
↑Urinary albumin
↑Glomerular lesions
↓TRPC6 expression, ↓Ca2+ influx
↑Nephrin ↓Glomerular desmin
↓Urinary albumin
POS
[97]/
Oman/2018
Male
Sprague-Dawley rats
Adenine (0.25% w/w) orally
Daily for 5 weeks
Sildenafil
Orally
(0.1, 0.5 or 2.5 mg/kg)
Daily for 5 weeks (alone or concomitantly with adenine)
SIMAt Day 5
↑BUN, sCr, uric acid, P, NGAL,
↑total NO, IS, Caspase 3 +ve cells
↑Albumin, NAG activity
↓Osmolality, CrCl in urine
↓CAT, glutathione reductase, SOD
↓TAC, ↑MAPK
↑Fibrosis
↑Adiponectin, cystatin-C, TNF-a
↓Sclerostin, ↑MDA
Tubular necrosis, tubular dilatation, tubular cast formation, necrotic nuclei, tubular cells apoptosis, cellular shedding, mononuclear infiltration
↓BUN, sCr, uric acid, ↓P, NGAL,
↓total NO, IS,
↓Caspase 3 +ve cells
↓Albumin,
↓ NAG activity
↑Osmolality,
↓CrCl in urine
↑CAT, SOD
↓glutathione reductase,
↑TAC, ↓MAPK, ↓Fibrosis
↓Adiponectin, cystatin-C, TNF-a
↑Sclerostin (not 0.1 mg/kg) ↓MDA
Improved tubular necrosis, tubular dilatation, tubular cast formation, mononuclear infiltration
POS
[98]/
Egypt/2018
Male albino rats Sprague-DawleyStreptozotocin
Single intraperitoneal dose 45 mg/kg
Sildenafil
Orally: 3 mg/kg/Day 3 weeks after STZ for 15 days
POSTDay 16 after initiation of Sildenafil↑sCr, ↑BUN
↑fasting and post prandial glucose
↓insulin levels
↑insulin resistance
↑MDA, ↓GSH, ↓CAT, ↓GPx,
↓SOD, ↓TAC
↓sCr, ↓BUN
↓fasting and post prandial glucose
↑insulin levels
↓insulin resistance (insignificant)
↓MDA, ↑GSH, ↑CAT, ↑GPx, ↑SOD, ↑TAC
POS
Abbreviations: AKI, acute kidney injury; ACR, albumin-creatinine ratio; aSMA, α-smooth muscle actin; Bax, proapoptotic protein; Bcl-2, antiapoptotic gene; BP, blood pressure; BUN, blood urea nitrogen; BW, body weight; Ca2+, calcium; CAT, catalase; cGMP, cyclic guanosine monophosphate; CrCl, creatinine clearance, ED-1, monoclonal antibody, eNOS, endothelial NOS, FeNa, fractional excretion of sodium, FITC, fluorescein isothiocyanate, GFR, glomerular filtration rate; GPx, glutathione peroxidase; GSH, glutathione; HR, heart rate; ICAM-1, intercellular adhesion molecule 1; IL, interleukin; IS, indoxyl sulfate; iNOS, inducible NOS; K, potassium; MAP, mean arterial pressure; MAPK, mitogen-activated protein kinase; MCP-1, monocyte chemoattractant protein 1; MDA, malondialdehyde; Na, sodium; NAG, N-acetyl-beta-D-glucosaminidase; NGAL, neutrophil gelatinase-associated lipocalin; NO, nitric oxide; NOX, NADPH oxidase; P, phosphorus; PDE5I, phosphodiesterase 5 inhibitor; PIGF, placenta growth factor; RRI, renal resistive index; RVF, renal vascular flow; RVR, renal vascular resistance; sCr, serum creatinine; sFlt1, soluble fms-like tyrosine kinase-1; SOD, superoxide dismutase; SBP, systolic blood pressure; suPAR, soluble urokinase-type plasminogen activator receptor; TAC, total antioxidant capacity; TGF-β1, transforming growth factor beta 1; TRPC6, transient receptor potential cation channel 6; TNF-a, tumor necrosis factor a; TUNEL, terminal deoxynucleotidyl transferase dUTP nick end labeling; VEGF, vascular endothelia growth factor; 8-OH dG, 8-hydroxydeoxyguanosine; ↓, reduced; ↑, increased.
Table A2. Animal studies evaluating the potential reno-protective effects of tadalafil.
Table A2. Animal studies evaluating the potential reno-protective effects of tadalafil.
Reference/
Country/Year
Studied AnimalModelPDE5I RouteTimingSampleRenal Injury EffectsPDE5I Renal EffectsOutcome
[99]/
Turkey/2013
Male
Sprague Dawley rats
SWL modelTadalafil
Orally: 1 mg/kg
Single dose
150 min prior to SWL
PRENephrectomy at Day 1/3/7Loss of micro-villi
Tubular degeneration and necrosis
Interstitial edema and fibrosis
↑ HSP-70
Reduced all histological damage

↓HSP-70
POS
[100]/
Turkey/2017
Male Sprague DawleySWL modelTadalafil
Orally: 1 mg/kg
60 min prior to SWL
PREBilateral nephrectomy 7 days post SWLRenal tubular damage
Peritubular fibrosis/Loss of microvilli
↑HSP-70
Significantly less tissue damage
⇔HSP-70 (glomerular)
↓HSP-70 (medullar/cortical)
POS
Abbreviation: AKI, acute kidney injury; HSP-70, heat shock protein 70; PDE5I, phosphodiesterase 5 inhibitor; SWL, shock wave lithotripsy; ↓, reduced; ↑, increased ⇔, no change.
Table A3. Animal studies evaluating the potential reno-protective effects of icariin.
Table A3. Animal studies evaluating the potential reno-protective effects of icariin.
Reference/
Country/Year
Studied AnimalModelPDE5I RouteTimingSampleRenal Injury EffectsPDE5I Renal EffectsOutcome
[101]/
China/2011
Male Sprague–Dawley ratsStreptozotocin
Single Dose
Intravenously
40 mg/kg
Icariin
Orally
80 mg/kg
For 8 weeks
From 5th to 20th week post streptozotocin
POSTDay 7
and
Week 13
↑sCr, ↑BUN, ↑Glucose, ↑MDA, ↑Hyp, ↓SOD, ↑Collagen IV, ↑TGF-β1
Glomerular Hypertrophy
Expansion of mesangial area and ECM
↓sCr, ↓BUN, ↓MDA, ↓Hyp ↑SOD, ↓Collagen IV, ↓TGF-β1
Inhibited these changes
POS
[102]/
Chiana/2014
Male Sprague-Dawley rats5/6 right nephrectomy
model
Icariin
Orally
20 + 40 mg/kg/day
1 week after AKI for 12 weeks
POST24 h before AKI
and at Week 12
↑BUN,↑sCr, ↑ urinary protein
↑Apoptotic rate, ↑Bcl-2, ↑Bax ↓G0/G1 phase cells
↑S phase cells
↓BUN, ↓sCr, ↓urinary protein
↓Apoptotic rate, ↓Bcl-2, ↓Bax
⇔G0/G1 phase cells, ↑G2/M phase
↓ S phase cells
POS
[103]/
China/2015
Male Sprague Dawley rats1st stage: Partial nephrectomy
2nd stage: Right renal ligation
Icariin
Orally
40 mg/kg/day
8 weeks
POSTAt 8 weeks↑BUN, ↑sCr, ↑uric acid
Mesangial expansion/Edema
Basement membrane thickening and capillary compression/occlusion.
Glomerular sclerosis/fibrosis
Inflammatory cell infiltration
↓BUN, ↓sCr, ↓uric acid
↑Renal progenitor cell proliferation
↓TGF-β1
Significantly improved glomerular lesions and blunted rest of the changes
POS
[22]/
China/2017
Female Wistar ratsPregnancy induced hypertension
L-NAME 0.5 g/L
from Day 12 of gestation
Icariin
Orally
10/50/100 mg/kg
18 days of gestation
POSTBP: Days 1 and 18
Kidney tissue: Day 18
↑SBP (Day 18), ↑ BUN, ↑sCr
↑Proteinuria, ↓Pup weight
↓Nephrinm, ↑Ang II, ↑AGT
Mesangial expansion
Basement membrane thickening
↓SBP (high dose), ↓BUN, ↓sCr
↓Proteinuria (medium/high dose)
No difference in pup weight
↑Nephrin, ↓Ang II, ↓AGT
Markdely reduced severity of lesions
POS
[104]/
China/2018
MRL/lpr miceK/O miceIcariin
Orally
10 mg/kg/day
8 weeks
POSTEvery 2 weeks
↑Urine protein,↑IgG deposit
↑sCr, ↑BUN, ↑TNF-a
↑Serum anti-dsDNA
↑Translocation and phosphorylation of NF-kBp65
↑F4/80, ↑NLRP3, ↑caspase 1p20
Increased glomerular proliferation/sclerosis/peripheral inflammation
↓Urine protein, ↓IgG deposit
↓sCr, ↓BUN, ↓TNF-a
↓Serum anti-dsDNA
↓Translocation and phosphorylation of NF-kBp65
↓F4/80, ↓NLRP3, ↓caspase 1p20
Improved all changes
POS
Abbreviations: AKI, acute kidney injury; Ang II, angiotensin II; Anti-dsDNA, antibody to double stranded DNA; AGT, angiotensinogen; Bax, proapoptotic protein; Bcl-2, antiapoptotic gene; BP, blood pressure; BUN, blood urea nitrogen; F4/80, macrophage marker; Hyp, hydroxyproline; IgG, immunoglobulin G; MDA, malondialdehyde; PDE5I, phosphodiesterase 5 inhibitor; SBP, systolic blood pressure; sCr, serum creatinine; SOD, superoxide dismutase; TGF-β1, transforming growth factor beta 1; TNF-a, tumor necrosis factor a; ↓, reduced; ↑, increased.
Table A4. Animal studies evaluating the potential reno-protective effects of vardenafil.
Table A4. Animal studies evaluating the potential reno-protective effects of vardenafil.
Reference/
Country/Year
Studied AnimalModelPDE5I RouteTimingSampleRenal Injury EffectsPDE5I Renal EffectsOutcome
[105]/
Germany/2008
Sprague Dawley ratsMouse monoclonal anti-Thy 1 antibody ER-4
Single injection, 1 mg/kg
Vardenafil
Orally
20 mg/kg within 18 h and
10 mg/kg/day for 48 h
PRE and POST24-h urine collection on Days 2 and 6
Blood sample: Day 6
↑PDE5-A, ⇔sCr
↑proteinuria
Mesangial proliferation
↑cGMP, ↓TSP-1, ⇔sCr
↓proliferation/cell number(glomerular)
↓collagen IV/fibronectin (glomerular)
↓TGF-β activation ⇔proteinuria
POS
[106]/
China/2009
New Zealand RabbitsInvagination of ureter in renal pelvisVardenafil
Orally
0.3 mg/kg/day
For 8 weeks post op
POST8 weeksDilated renal pelvises
Fibrotic PUJ
↑TGF-β1 ↓nNOS
Dilated renal pelvises
Less fibrotic PUJ
↓TGF-β1 ↑nNOS
POS
[107]/
Hungary/2013
Sprague Dawley male ratsStreptozotocin
Single intraperitoneal dose
60/mg/kg
Vardenafil
Orally
10 mg/kg/day for 8 weeks
72 h post STZ
POST8 weeks after AKI↓cGMP,
NCS elevated Urea levels
Decreased body weight
No difference in MAP
↑Urine protein/creatinine ratio
↑Fibronectin, ↑TGF-β1, ↑desmin, ↓nephrin, ↑Nitrotyrosine, ↑NOS
Glomerular hypertrophy
Mesangial expansion
Adhesions to Bowman’s capsule
Tubular dilatation and atrophy
Mononuclear cell infiltration
↑cGMP
Developed kidney hypertrophy
No difference in MAP
↓Urine protein/creatinine ratio
↓Fibronectin, ↓TGF-β1
↓desmin, ↑nephrin
No difference
Attenuated all changes
POS
[108]/
Turkey/2015
Male Swiss albino miceCyclosporine A
30 mg/kg
Subcutaneously
Daily for 28 days
Vardenafil
Orally
30 mg/kg/day
For 28 days
PREAt 28 days
↓Kidney weight
↑BUN, ↑sCr, ↑TOS levels
↓TAS levels,↓tissue NO
↓COX-1, ↓COX-2, ↓TGF-β1
↓Pgp levels, ↓PDGF-A, ↓PDGF-C
Histological changes: cortex/outer medulla
No change in kidney weight
↓BUN, ↓sCr, ↓TOS levels
↑TAS levels, ↑tissue NO
↑COX-1, ↑COX-2, ⇔TGF-β1
↑Pgp levels, ↑PDGF-A, ↑PDGF-C
Normal histopathological appearances
POS
Abbreviations: AKI, acute kidney injury; BUN, blood urea nitrogen; cGMP, cyclic guanosine monophosphate; COX, cyclo-oxygenase; FeNa, fractional excretion of sodium; MAP, mean arterial pressure; NCS, not clinically significant; NO, nitric oxide, nNOS, neuronal NOS; PDE5I, phosphodiesterase 5 inhibitor; PDGF, platelet-derived growth factor; Pgp, P glycoprotein; PUJ, pelvic ureteric junction; sCr, serum creatinine; TAS, total antioxidant status; TGF-β1, transforming growth factor beta 1; TOS, total oxidant status; TSP-1, thrombospondin -1; ↓, reduced; ↑, increased ⇔, no change.
Table A5. Animal studies evaluating the potential reno-protective effects of zaprinast and udenafil.
Table A5. Animal studies evaluating the potential reno-protective effects of zaprinast and udenafil.
Reference/
Country/Year
Studied AnimalModelPDE5I RouteTimingSampleRenal Injury EffectsPDE5I Renal EffectsOutcome
[109]/
Japan/1998
Mongrel dogsCut left renal nerves and electrostimulation of left renal bundle (distal end)Zaprinast
Intra-renal arterial infusion
10 or 100 μg/kg/min
SIMSimultaneously↓Urine flow, ↓UNaV, ↓FeNa
⇔RBF, ⇔GFR
↑Urine flow, ↑UNaV, ↑FeNa
⇔ RBF, ⇔GFR, ↓RVR
↑Renal venous cGMP
POS
[110]/
Korea/2010
10-week-old male Sprague-DawleyRight nephrectomy
+ Left renal artery clamping for 45 min
and
Cyclosporine A
15 mg/kg subcutaneously
Udenafil
Orally: 10 mg/kg
For 28 days after the procedure
SIM and POSTOn Day 28 blood samples and left nephrectomy↑BUN, ↑sCr, ↓eNOS, ⇔VEGF
Decreased thickness of the proximal tubules and nuclei, vacuolization of the cytoplasm, altered cellular shape, fewer nuclei
↓BUN, ↓sCr, ↑eNOS, ⇔VEGF
↓VEGF mRNA
POS
Abbreviations: AKI, acute kidney injury; BUN, blood urea nitrogen; cGMP, cyclic guanosine monophosphate; eNOS, endothelial NOS; FeNa, fractional excretion of sodium; GFR, glomerular filtration rate; PDE5I, phosphodiesterase 5 inhibitor; RBF, renal blood flow; RVR, renal vascular resistance; sCr, serum creatinine; UNaV, urinary sodium excretion; VEGF, vascular endothelia growth factor; ↓, reduced; ↑, increased ⇔, no change.

References

  1. Makris, K.; Spanou, L. Acute Kidney Injury: Definition, Pathophysiology and Clinical Phenotypes. Clin. Biochem. Rev. 2016, 37, 85–98. [Google Scholar]
  2. Codorniu, A.; Lemasle, L.; Legrand, M.; Blet, A.; Mebazaa, A.; Gayat, E. Methods used to assess the performance of biomarkers for the diagnosis of acute kidney injury: A systematic review and meta-analysis. Biomarkers 2018, 23, 766–772. [Google Scholar] [CrossRef] [PubMed]
  3. ACR Committee on Drugs and Contrast Media. ACR Manual on Contrast Media Version 10.3; ACR: Reston, VA, USA, 2020. [Google Scholar]
  4. Bellomo, R.; Ronco, C.; Kellum, J.A.; Mehta, R.L.; Palevsky, P.; Acute Dialysis Quality Initiative workgroup. Acute renal failure—Definition, outcome measures, animal models, fluid therapy and information technology needs: The Second International Consensus Conference of the Acute Dialysis Quality Initiative (ADQI) Group. Crit. Care 2004, 8, R204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Mehta, R.L.; Kellum, J.A.; Shah, S.V.; Molitoris, B.A.; Ronco, C.; Warnock, D.G.; Levin, A.; Acute Kidney Injury Network. Acute Kidney Injury Network: Report of an initiative to improve outcomes in acute kidney injury. Crit. Care 2007, 11, R31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. KDIGO. KDIGO Clinical Practice Guideline for Acute Kidney Injury. Kidney Int. Suppl. 2012, 2, 1–138. [Google Scholar] [CrossRef] [Green Version]
  7. Fiorentino, M.; Kellum, J.A. Improving Translation from Preclinical Studies to Clinical Trials in Acute Kidney Injury. Nephron 2018, 140, 81–85. [Google Scholar] [CrossRef]
  8. Zarjou, A.; Sanders, P.W.; Mehta, R.L.; Agarwal, A. Enabling innovative translational research in acute kidney injury. Clin. Transl. Sci. 2012, 5, 93–101. [Google Scholar] [CrossRef]
  9. Skrypnyk, N.I.; Siskind, L.J.; Faubel, S.; de Caestecker, M.P. Bridging translation for acute kidney injury with better preclinical modeling of human disease. Am. J. Physiol. Renal. Physiol. 2016, 310, F972–F984. [Google Scholar] [CrossRef] [Green Version]
  10. Ortiz, A.; Sanchez-Nino, M.D.; Izquierdo, M.C.; Martin-Cleary, C.; Garcia-Bermejo, L.; Moreno, J.A.; Ruiz-Ortega, M.; Draibe, J.; Cruzado, J.M.; Garcia-Gonzalez, M.A.; et al. Translational value of animal models of kidney failure. Eur. J. Pharmacol. 2015, 759, 205–220. [Google Scholar] [CrossRef]
  11. Bao, Y.W.; Yuan, Y.; Chen, J.H.; Lin, W.Q. Kidney disease models: Tools to identify mechanisms and potential therapeutic targets. Zool. Res. 2018, 39, 72–86. [Google Scholar] [CrossRef]
  12. Singh, A.P.; Junemann, A.; Muthuraman, A.; Jaggi, A.S.; Singh, N.; Grover, K.; Dhawan, R. Animal models of acute renal failure. Pharmacol. Rep. 2012, 64, 31–44. [Google Scholar] [CrossRef]
  13. Burmeister, D.M.; Gomez, B.I.; Dubick, M.A. Molecular mechanisms of trauma-induced acute kidney injury: Inflammatory and metabolic insights from animal models. Biochim. Biophys. Acta Mol. Basis Dis. 2017, 1863, 2661–2671. [Google Scholar] [CrossRef] [PubMed]
  14. Iordache, A.M.; Docea, A.O.; Buga, A.M.; Zlatian, O.; Ciurea, M.E.; Rogoveanu, O.C.; Burada, F.; Sosoi, S.; Mitrut, R.; Mamoulakis, C.; et al. Sildenafil and tadalafil reduce the risk of contrast-induced nephropathy by modulating the oxidant/antioxidant balance in a murine model. Food Chem. Toxicol. 2020, 135, 111038. [Google Scholar] [CrossRef] [PubMed]
  15. Sanz, A.B.; Sanchez-Nino, M.D.; Martin-Cleary, C.; Ortiz, A.; Ramos, A.M. Progress in the development of animal models of acute kidney injury and its impact on drug discovery. Expert Opin. Drug Discov. 2013, 8, 879–895. [Google Scholar] [CrossRef] [PubMed]
  16. Sarica, K.; Yencilek, F. Prevention of shockwave induced functional and morphological alterations: An overview. Arch. Ital. Urol. Androl. 2008, 80, 27–33. [Google Scholar]
  17. Lahoud, Y.; Hussein, O.; Shalabi, A.; Nativ, O.; Awad, H.; Khamaisi, M.; Matar, I.; Nativ, O.; Abassi, Z. Effects of phosphodiesterase-5 inhibitor on ischemic kidney injury during nephron sparing surgery: Quantitative assessment by NGAL and KIM-1. World J. Urol. 2015, 33, 2053–2062. [Google Scholar] [CrossRef]
  18. Xie, C.; Liu, L.; Wang, Z.; Xie, H.; Feng, Y.; Suo, J.; Wang, M.; Shang, W.; Feng, G. Icariin Improves Sepsis-Induced Mortality and Acute Kidney Injury. Pharmacology 2018, 102, 196–205. [Google Scholar] [CrossRef]
  19. Abdel-latif, R.G.; Morsy, M.A.; El-Moselhy, M.A.; Khalifa, M.A. Sildenafil protects against nitric oxide deficiency-related nephrotoxicity in cyclosporine A treated rats. Eur. J. Pharmacol. 2013, 705, 126–134. [Google Scholar] [CrossRef]
  20. Georgiadis, G.; Mavridis, C.; Belantis, C.; Zisis, I.E.; Skamagkas, I.; Fragkiadoulaki, I.; Heretis, I.; Tzortzis, V.; Psathakis, K.; Tsatsakis, A.; et al. Nephrotoxicity issues of organophosphates. Toxicology 2018, 406–407, 129–136. [Google Scholar] [CrossRef]
  21. Ozlulerden, Y.; Toktas, C.; Aybek, H.; Kucukatay, V.; Sen Turk, N.; Zumrutbas, A.E. The renoprotective effects of mannitol and udenafil in renal ischemia-reperfusion injury model. Investig. Clin. Urol. 2017, 58, 289–295. [Google Scholar] [CrossRef] [Green Version]
  22. Zhang, W.; Yuan, W.; Xu, N.; Li, J.; Chang, W. Icariin improves acute kidney injury and proteinuria in a rat model of pregnancyinduced hypertension. Mol. Med. Rep. 2017, 16, 7398–7404. [Google Scholar] [CrossRef] [Green Version]
  23. Whitaker, R.M.; Wills, L.P.; Stallons, L.J.; Schnellmann, R.G. cGMP-selective phosphodiesterase inhibitors stimulate mitochondrial biogenesis and promote recovery from acute kidney injury. J. Pharmacol. Exp. Ther. 2013, 347, 626–634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Zhou, Y.D.; Hou, J.G.; Yang, G.; Jiang, S.; Chen, C.; Wang, Z.; Liu, Y.Y.; Ren, S.; Li, W. Icariin ameliorates cisplatin-induced cytotoxicity in human embryonic kidney 293 cells by suppressing ROS-mediated PI3K/Akt pathway. Biomed. Pharmacother. 2019, 109, 2309–2317. [Google Scholar] [CrossRef] [PubMed]
  25. van der Molen, A.J.; Reimer, P.; Dekkers, I.A.; Bongartz, G.; Bellin, M.F.; Bertolotto, M.; Clement, O.; Heinz-Peer, G.; Stacul, F.; Webb, J.A.W.; et al. Post-contrast acute kidney injury—Part 1: Definition, clinical features, incidence, role of contrast medium and risk factors: Recommendations for updated ESUR Contrast Medium Safety Committee guidelines. Eur. Radiol. 2018, 28, 2845–2855. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Tsarouhas, K.; Tsitsimpikou, C.; Papantoni, X.; Lazaridou, D.; Koutouzis, M.; Mazzaris, S.; Rezaee, R.; Mamoulakis, C.; Georgoulias, P.; Nepka, C.; et al. Oxidative stress and kidney injury in trans-radial catheterization. Biomed. Rep. 2018, 8, 417–425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Mamoulakis, C.; Fragkiadoulaki, I.; Karkala, P.; Georgiadis, G.; Zisis, I.E.; Stivaktakis, P.; Kalogeraki, A.; Tsiaoussis, I.; Burykina, T.; Lazopoulos, G.; et al. Contrast-induced nephropathy in an animal model: Evaluation of novel biomarkers in blood and tissue samples. Toxicol. Rep. 2019, 6, 395–400. [Google Scholar] [CrossRef]
  28. Morcos, S.K. Can selective inhibitors of cyclic guanosine monophosphate (cGMP)-specific phosphadiesterase type 5 (PDE 5) offer protection against contrast induced nephropathy? Quant. Imaging Med. Surg. 2014, 4, 214–215. [Google Scholar] [CrossRef]
  29. El-Sisi, A.E.; Sokar, S.S.; Abu-Risha, S.E.; Ibrahim, H.A. Combination of tadalafil and diltiazem attenuates renal ischemia reperfusion-induced acute renal failure in rats. Biomed. Pharmacother. 2016, 84, 861–869. [Google Scholar] [CrossRef]
  30. Choi, D.E.; Jeong, J.Y.; Lim, B.J.; Chung, S.; Chang, Y.K.; Lee, S.J.; Na, K.R.; Kim, S.Y.; Shin, Y.T.; Lee, K.W. Pretreatment of sildenafil attenuates ischemia-reperfusion renal injury in rats. Am. J. Physiol. Renal. Physiol. 2009, 297, F362–F370. [Google Scholar] [CrossRef] [Green Version]
  31. Novak, J.E.; Handa, R. Contrast Nephropathy Associated with Percutaneous Coronary Angiography and Intervention. Cardiol. Clin. 2019, 37, 287–296. [Google Scholar] [CrossRef]
  32. van der Molen, A.J.; Reimer, P.; Dekkers, I.A.; Bongartz, G.; Bellin, M.F.; Bertolotto, M.; Clement, O.; Heinz-Peer, G.; Stacul, F.; Webb, J.A.W.; et al. Post-contrast acute kidney injury. Part 2: Risk stratification, role of hydration and other prophylactic measures, patients taking metformin and chronic dialysis patients: Recommendations for updated ESUR Contrast Medium Safety Committee guidelines. Eur. Radiol. 2018, 28, 2856–2869. [Google Scholar] [CrossRef] [Green Version]
  33. Mamoulakis, C.; Tsarouhas, K.; Fragkiadoulaki, I.; Heretis, I.; Wilks, M.F.; Spandidos, D.A.; Tsitsimpikou, C.; Tsatsakis, A. Contrast-induced nephropathy: Basic concepts, pathophysiological implications and prevention strategies. Pharmacol. Ther. 2017, 180, 99–112. [Google Scholar] [CrossRef] [PubMed]
  34. Salonia, A.; Bettocchi, C.; Carvalho, J.; Corona, G.; Jones, T.H.; Kadioglu, A.; Martinez-Salamanca, I.; Minhas, S.; Serefoǧlu, E.C.; Verze, P.; et al. EAU Guidelines on Sexual and Reproductive Health. Edn. Presented at the EAU Annual Congress Amsterdam 2020; EAU Guidelines Office: Arnhem, The Netherlands, 2020. [Google Scholar]
  35. Ozbek, K.; Ceyhan, K.; Koc, F.; Sogut, E.; Altunkas, F.; Karayakali, M.; Celik, A.; Kadi, H.; Koseoglu, R.D.; Onalan, O. The protective effect of single dose tadalafil in contrast-induced nephropathy: An experimental study. Anatol. J. Cardiol. 2015, 15, 306–310. [Google Scholar] [CrossRef] [PubMed]
  36. Lue, T.F. Erectile dysfunction. N. Engl. J. Med. 2000, 342, 1802–1813. [Google Scholar] [CrossRef] [PubMed]
  37. Moher, D.; Liberati, A.; Tetzlaff, J.; Altman, D.G.; Group, P. Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement. Int. J. Surg. 2010, 8, 336–341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Li, W.; Wang, L.; Chu, X.; Cui, H.; Bian, Y. Icariin combined with human umbilical cord mesenchymal stem cells significantly improve the impaired kidney function in chronic renal failure. Mol. Cell. Biochem. 2017, 428, 203–212. [Google Scholar] [CrossRef] [PubMed]
  39. Krane, L.S.; Peyton, C.C.; Olympio, M.A.; Hemal, A.K. A randomized double blinded placebo controlled trial of sildenafil for renoprotection prior to hilar clamping in patients undergoing robotic assisted laparoscopic partial nephrectomy. J. Surg. Oncol. 2016, 114, 785–788. [Google Scholar] [CrossRef] [PubMed]
  40. Polcari, A.J.; Farooq, A.V.; Woods, M.E.; Ripsch, M.S.; Picken, M.; Turk, T.M.; White, F.A. Effect of the phosphodiesterase-5 inhibitor zaprinast on ischemia-reperfusion injury in rats. J. Endourol. 2013, 27, 338–342. [Google Scholar] [CrossRef] [Green Version]
  41. Lee, K.W.; Jeong, J.Y.; Lim, B.J.; Chang, Y.K.; Lee, S.J.; Na, K.R.; Shin, Y.T.; Choi, D.E. Sildenafil attenuates renal injury in an experimental model of rat cisplatin-induced nephrotoxicity. Toxicology 2009, 257, 137–143. [Google Scholar] [CrossRef]
  42. Oruc, O.; Inci, K.; Aki, F.T.; Zeybek, D.; Muftuoglu, S.F.; Kilinc, K.; Ergen, A. Sildenafil attenuates renal ischemia reperfusion injury by decreasing leukocyte infiltration. Acta Histochem. 2010, 112, 337–344. [Google Scholar] [CrossRef]
  43. Medeiros, P.J.; Villarim Neto, A.; Lima, F.P.; Azevedo, I.M.; Leao, L.R.; Medeiros, A.C. Effect of sildenafil in renal ischemia/reperfusion injury in rats. Acta Cir. Bras. 2010, 25, 490–495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Ali, B.H.; Abdelrahman, A.M.; Al-Salam, S.; Sudhadevi, M.; AlMahruqi, A.S.; Al-Husseni, I.S.; Beegam, S.; Dhanasekaran, S.; Nemmar, A.; Al-Moundhri, M. The effect of sildenafil on cisplatin nephrotoxicity in rats. Basic Clin. Pharmacol. Toxicol. 2011, 109, 300–308. [Google Scholar] [CrossRef] [PubMed]
  45. Akgul, T.; Huri, E.; Yagmurdur, H.; Ayyildiz, A.; Ustun, H.; Germiyanoglu, C. Phosphodiesterase 5 inhibitors attenuate renal tubular apoptosis after partial unilateral ureteral obstruction: An experimental study. Kaohsiung J. Med. Sci. 2011, 27, 15–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Lledo-Garcia, E.; Subira-Rios, D.; Ogaya-Pinies, G.; Tejedor-Jorge, A.; Canizo-Lopez, J.F.; Hernandez-Fernandez, C. Intravenous sildenafil as a preconditioning drug against hemodynamic consequences of warm ischemia-reperfusion on the kidney. J. Urol. 2011, 186, 331–333. [Google Scholar] [CrossRef] [PubMed]
  47. Cadirci, E.; Halici, Z.; Odabasoglu, F.; Albayrak, A.; Karakus, E.; Unal, D.; Atalay, F.; Ferah, I.; Unal, B. Sildenafil treatment attenuates lung and kidney injury due to overproduction of oxidant activity in a rat model of sepsis: A biochemical and histopathological study. Clin. Exp. Immunol. 2011, 166, 374–384. [Google Scholar] [CrossRef]
  48. Patel, N.N.; Lin, H.; Toth, T.; Jones, C.; Ray, P.; Welsh, G.I.; Satchell, S.C.; Sleeman, P.; Angelini, G.D.; Murphy, G.J. Phosphodiesterase-5 inhibition prevents postcardiopulmonary bypass acute kidney injury in swine. Ann. Thorac. Surg. 2011, 92, 2168–2176. [Google Scholar] [CrossRef] [PubMed]
  49. Kucuk, A.; Yucel, M.; Erkasap, N.; Tosun, M.; Koken, T.; Ozkurt, M.; Erkasap, S. The effects of PDE5 inhibitory drugs on renal ischemia/reperfusion injury in rats. Mol. Biol. Rep. 2012, 39, 9775–9782. [Google Scholar] [CrossRef]
  50. Stegbauer, J.; Friedrich, S.; Potthoff, S.A.; Broekmans, K.; Cortese-Krott, M.M.; Quack, I.; Rump, L.C.; Koesling, D.; Mergia, E. Phosphodiesterase 5 attenuates the vasodilatory response in renovascular hypertension. PLoS ONE 2013, 8, e80674. [Google Scholar] [CrossRef] [Green Version]
  51. Helmy, M.W.; Helmy, M.M.; Abd Allah, D.M.; Abo Zaid, A.M.; Mohy El-Din, M.M. Role of nitrergic and endothelin pathways modulations in cisplatin-induced nephrotoxicity in male rats. J. Physiol. Pharmacol. 2014, 65, 393–399. [Google Scholar]
  52. Gokakin, A.K.; Atabey, M.; Deveci, K.; Sancakdar, E.; Tuzcu, M.; Duger, C.; Topcu, O. The effects of sildenafil in liver and kidney injury in a rat model of severe scald burn: A biochemical and histopathological study. Ulus Travma Acil Cerrahi Derg 2014, 20, 319–327. [Google Scholar] [CrossRef] [Green Version]
  53. Morsy, M.A.; Ibrahim, S.A.; Amin, E.F.; Kamel, M.Y.; Rifaai, R.A.; Hassan, M.K. Sildenafil Ameliorates Gentamicin-Induced Nephrotoxicity in Rats: Role of iNOS and eNOS. J. Toxicol. 2014, 2014, 489382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Cui, W.; Maimaitiyiming, H.; Qi, X.; Norman, H.; Zhou, Q.; Wang, X.; Fu, J.; Wang, S. Increasing cGMP-dependent protein kinase activity attenuates unilateral ureteral obstruction-induced renal fibrosis. Am. J. Physiol. Renal. Physiol. 2014, 306, F996–F1007. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Lauver, D.A.; Carey, E.G.; Bergin, I.L.; Lucchesi, B.R.; Gurm, H.S. Sildenafil citrate for prophylaxis of nephropathy in an animal model of contrast-induced acute kidney injury. PLoS ONE 2014, 9, e113598. [Google Scholar] [CrossRef] [Green Version]
  56. Zahran, M.H.; Hussein, A.M.; Barakat, N.; Awadalla, A.; Khater, S.; Harraz, A.; Shokeir, A.A. Sildenafil activates antioxidant and antiapoptotic genes and inhibits proinflammatory cytokine genes in a rat model of renal ischemia/reperfusion injury. Int. Urol. Nephrol. 2015, 47, 1907–1915. [Google Scholar] [CrossRef]
  57. de Almeida, L.S.; Barboza, J.R.; Freitas, F.P.; Porto, M.L.; Vasquez, E.C.; Meyrelles, S.S.; Gava, A.L.; Pereira, T.M. Sildenafil prevents renal dysfunction in contrast media-induced nephropathy in Wistar rats. Hum. Exp. Toxicol. 2016, 35, 1194–1202. [Google Scholar] [CrossRef]
  58. Mohey, V.; Singh, M.; Puri, N.; Kaur, T.; Pathak, D.; Singh, A.P. Sildenafil obviates ischemia-reperfusion injury-induced acute kidney injury through peroxisome proliferator-activated receptor gamma agonism in rats. J. Surg. Res. 2016, 201, 69–75. [Google Scholar] [CrossRef]
  59. Altintop, I.; Tatli, M.; Karakukcu, C.; Soyer Sarica, Z.; Hanim Yay, A.; Balcioglu, E.; Ozturk, A. Serum and Tissue HIF-2 Alpha Expression in CIN, N-Acetyl Cysteine, and Sildenafil-Treated Rat Models: An Experimental Study. Medicina 2018, 54, 54. [Google Scholar] [CrossRef] [Green Version]
  60. Behiry, S.; Rabie, A.; Kora, M.; Ismail, W.; Sabry, D.; Zahran, A. Effect of combination sildenafil and gemfibrozil on cisplatin-induced nephrotoxicity; role of heme oxygenase-1. Ren. Fail. 2018, 40, 371–378. [Google Scholar] [CrossRef] [Green Version]
  61. Zahran, M.H.; Barakat, N.; Khater, S.; Awadalla, A.; Mosbah, A.; Nabeeh, A.; Hussein, A.M.; Shokeir, A.A. Renoprotective effect of local sildenafil administration in renal ischaemia-reperfusion injury: A randomised controlled canine study. Arab J. Urol. 2019, 17, 150–159. [Google Scholar] [CrossRef] [Green Version]
  62. Ayyildiz, A.; Kaya, M.; Karaguzel, E.; Bumin, A.; Akgul, T.; Aklan, Z.; Germiyanoglu, C. Effect of tadalafil on renal resistivity and pulsatility index in partial ureteral obstruction. Urol. Int. 2009, 83, 75–79. [Google Scholar] [CrossRef]
  63. Gasanov, F.; Aytac, B.; Vuruskan, H. The effects of tadalafil on renal ischemia reperfusion injury: An experimental study. Bosn. J. Basic Med. Sci. 2011, 11, 158–162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Guzeloglu, M.; Yalcinkaya, F.; Atmaca, S.; Bagriyanik, A.; Oktar, S.; Yuksel, O.; Fansa, I.; Hazan, E. The beneficial effects of tadalafil on renal ischemia-reperfusion injury in rats. Urol. Int. 2011, 86, 197–203. [Google Scholar] [CrossRef] [PubMed]
  65. Faddegon, S.; Best, S.L.; Olweny, E.O.; Tan, Y.K.; Park, S.K.; Mir, S.A.; Cadeddu, J.A. Tadalafil for prevention of renal dysfunction secondary to renal ischemia. Can. J. Urol. 2012, 19, 6274–6279. [Google Scholar] [PubMed]
  66. Sohotnik, R.; Nativ, O.; Abbasi, A.; Awad, H.; Frajewicki, V.; Bishara, B.; Sukhotnik, I.; Armaly, Z.; Aronson, D.; Heyman, S.N.; et al. Phosphodiesterase-5 inhibition attenuates early renal ischemia-reperfusion-induced acute kidney injury: Assessment by quantitative measurement of urinary NGAL and KIM-1. Am. J. Physiol. Renal. Physiol. 2013, 304, F1099–F1104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Zhu, C.Y.; Liu, M.; Liu, Y.Z.; Li, W.; Zhai, W.; Che, J.P.; Yan, Y.; Wang, G.C.; Zheng, J.H. Preventive effect of phosphodiesterase 5 inhibitor tadalafil on experimental post-pyelonephritic renal injury in rats. J. Surg. Res. 2014, 186, 253–261. [Google Scholar] [CrossRef]
  68. Amasyali, A.S.; Akkurt, A.; Kazan, E.; Yilmaz, M.; Erol, B.; Yildiz, Y.; Erol, H. The protective effect of tadalafil on IMA (ischemia modified albumin) levels in experimental renal ischemia-reperfusion injury. Int. J. Clin. Exp. Med. 2015, 8, 15766–15772. [Google Scholar]
  69. Erol, B.; Turker, T.; Tok, A.; Bektas, S.; Mungan, G.; Ozkanli, S.; Karakas, B.; Tokgoz, H.; Akduman, B.; Mungan, A. The protective effects of tadalafil on renal damage following ischemia reperfusion injury in rats. Kaohsiung J. Med. Sci. 2015, 31, 454–462. [Google Scholar] [CrossRef] [Green Version]
  70. Adeneye, A.A.; Benebo, A.S. Chemopreventive Effect of Tadalafil in Cisplatin-Induced Nephrotoxicity in Rats. Niger. J. Physiol. Sci. 2016, 31, 1–10. [Google Scholar]
  71. Benli, E.; Ayyildiz, S.N.; Cirrik, S.; Kokturk, S.; Cirakoglu, A.; Noyan, T.; Ayyildiz, A.; Germiyanoglu, C. The effect of tadalafil therapy on kidney damage caused by sepsis in a polymicrobial septic model induced in rats: A biochemical and histopathological study. Int. Braz. J. Urol. 2017, 43, 345–355. [Google Scholar] [CrossRef] [Green Version]
  72. Wietzikoski, E.G.G.; Foiatto, J.C.; Czeczko, N.G.; Malafaia, O.; Koleski, F.C.; Mierzwa, T.C.; Gomes, R.P.X. Tadalafil protector effect during ischemia-reperfusion in rats. Acta Cir. Bras. 2017, 32, 973–983. [Google Scholar] [CrossRef] [Green Version]
  73. Medeiros, V.F.; Azevedo, I.M.; Carvalho, M.D.; Oliveira, C.N.; Egito, E.S.; Medeiros, A.C. The renoprotective effect of oral Tadalafil pretreatment on ischemia/reperfusion injury in rats. Acta Cir. Bras. 2017, 32, 90–97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Kokturk, S.; Benli, E.; Ayyildiz, A.; Cirrik, S.; Cetinkol, Y.; Ayyildiz, S.N.; Noyan, T. Positive outcomes of phosphodiesterase type 5 inhibitor on histopathologic and biochemical changes induced by ureteral obstruction. Rev. Assoc. Med. Bras. (1992) 2019, 65, 388–393. [Google Scholar] [CrossRef] [PubMed]
  75. Ma, P.; Zhang, S.; Su, X.; Qiu, G.; Wu, Z. Protective effects of icariin on cisplatin-induced acute renal injury in mice. Am. J. Transl. Res. 2015, 7, 2105–2114. [Google Scholar] [PubMed]
  76. Chen, H.A.; Chen, C.M.; Guan, S.S.; Chiang, C.K.; Wu, C.T.; Liu, S.H. The antifibrotic and anti-inflammatory effects of icariin on the kidney in a unilateral ureteral obstruction mouse model. Phytomedicine 2019, 59, 152917. [Google Scholar] [CrossRef]
  77. Kyriazis, I.; Kagadis, G.C.; Kallidonis, P.; Georgiopoulos, I.; Marazioti, A.; Geronasiou, A.; Liourdi, D.; Loudos, G.; Schinas, V.; Apostolopoulos, D.; et al. PDE5 inhibition against acute renal ischemia reperfusion injury in rats: Does vardenafil offer protection? World J. Urol. 2013, 31, 597–602. [Google Scholar] [CrossRef]
  78. Sousa, R.C.; Moreira Neto, A.A.; Capelozzi, V.L.; Ab’Saber, A.M.; Rodrigues, O.R. Effects of vardenafil on the kidney of Wistar rats submitted to acute ischemia and reperfusion. Acta Cir. Bras. 2015, 30, 339–344. [Google Scholar] [CrossRef] [Green Version]
  79. Guan, Z.; Miller, S.B.; Greenwald, J.E. Zaprinast accelerates recovery from established acute renal failure in the rat. Kidney Int. 1995, 47, 1569–1575. [Google Scholar] [CrossRef] [Green Version]
  80. Wetzl, V.; Schinner, E.; Kees, F.; Faerber, L.; Schlossmann, J. Differences in the renal antifibrotic cGMP/cGKI-dependent signaling of serelaxin, zaprinast, and their combination. Naunyn. Schmiedebergs. Arch. Pharmacol. 2017, 390, 939–948. [Google Scholar] [CrossRef]
  81. Rodriguez-Iturbe, B.; Ferrebuz, A.; Vanegas, V.; Quiroz, Y.; Espinoza, F.; Pons, H.; Vaziri, N.D. Early treatment with cGMP phosphodiesterase inhibitor ameliorates progression of renal damage. Kidney Int. 2005, 68, 2131–2142. [Google Scholar] [CrossRef] [Green Version]
  82. Lledo-Garcia, E.; Rodriguez-Martinez, D.; Cabello-Benavente, R.; Moncada-Iribarren, I.; Tejedor-Jorge, A.; Dulin, E.; Hernandez-Fernandez, C.; Del Canizo-Lopez, J.F. Sildenafil improves immediate posttransplant parameters in warm-ischemic kidney transplants: Experimental study. Transplant. Proc. 2007, 39, 1354–1356. [Google Scholar] [CrossRef]
  83. Aboutabl, M.E.; Raafat, M.; Maklad, Y.A.; Kenawy, S.A.; El Din, A.G. Sildenafil augments the beneficial hemodynamic and histopathological effects of amlodipine in nitric oxide-deficient hypertensive rats: Role of nitric oxide-cyclic GMP pathway. Pharmacol. Res. 2008, 57, 456–463. [Google Scholar] [CrossRef] [PubMed]
  84. Lledo-Garcia, E.; Subira-Rios, D.; Rodriguez-Martinez, D.; Dulin, E.; Alvarez-Fernandez, E.; Hernandez-Fernandez, C.; del Canizo-Lopez, J.F. Sildenafil as a protecting drug for warm ischemic kidney transplants: Experimental results. J. Urol. 2009, 182, 1222–1225. [Google Scholar] [CrossRef] [PubMed]
  85. Jeong, K.H.; Lee, T.W.; Ihm, C.G.; Lee, S.H.; Moon, J.Y.; Lim, S.J. Effects of sildenafil on oxidative and inflammatory injuries of the kidney in streptozotocin-induced diabetic rats. Am. J. Nephrol. 2009, 29, 274–282. [Google Scholar] [CrossRef] [PubMed]
  86. Bae, E.H.; Kim, I.J.; Joo, S.Y.; Kim, E.Y.; Kim, C.S.; Choi, J.S.; Ma, S.K.; Kim, S.H.; Lee, J.U.; Kim, S.W. Renoprotective effects of sildenafil in DOCA-salt hypertensive rats. Kidney Blood Press. Res. 2012, 36, 248–257. [Google Scholar] [CrossRef] [PubMed]
  87. Tapia, E.; Sanchez-Lozada, L.G.; Soto, V.; Manrique, A.M.; Ortiz-Vega, K.M.; Santamaria, J.; Medina-Campos, O.N.; Cristobal, M.; Avila-Casado, C.; Pedraza-Chaverri, J.; et al. Sildenafil treatment prevents glomerular hypertension and hyperfiltration in rats with renal ablation. Kidney Blood Press. Res. 2012, 35, 273–280. [Google Scholar] [CrossRef] [PubMed]
  88. Hosgood, S.A.; Randle, L.V.; Patel, M.; Watson, C.J.; Bradley, J.A.; Nicholson, M.L. Sildenafil citrate in a donation after circulatory death experimental model of renal ischemia-reperfusion injury. Transplantation 2014, 98, 612–617. [Google Scholar] [CrossRef]
  89. Dias, A.T.; Rodrigues, B.P.; Porto, M.L.; Gava, A.L.; Balarini, C.M.; Freitas, F.P.; Palomino, Z.; Casarini, D.E.; Campagnaro, B.P.; Pereira, T.M.; et al. Sildenafil ameliorates oxidative stress and DNA damage in the stenotic kidneys in mice with renovascular hypertension. J. Transl. Med. 2014, 12, 35. [Google Scholar] [CrossRef] [Green Version]
  90. Dias, A.T.; Cintra, A.S.; Frossard, J.C.; Palomino, Z.; Casarini, D.E.; Gomes, I.B.; Balarini, C.M.; Gava, A.L.; Campagnaro, B.P.; Pereira, T.M.; et al. Inhibition of phosphodiesterase 5 restores endothelial function in renovascular hypertension. J. Transl. Med. 2014, 12, 250. [Google Scholar] [CrossRef] [Green Version]
  91. El-Mahdy, N.A.; El-Sayad, M.E.; El-Kadem, A.H. Combination of telmisartan with sildenafil ameliorate progression of diabetic nephropathy in streptozotocin-induced diabetic model. Biomed. Pharmacother. 2016, 81, 136–144. [Google Scholar] [CrossRef]
  92. Tripathi, A.S.; Mazumder, P.M.; Chandewar, A.V. Sildenafil, a phosphodiesterase type 5 inhibitor, attenuates diabetic nephropathy in STZ-induced diabetic rats. J. Basic Clin. Physiol. Pharmacol. 2016, 27, 57–62. [Google Scholar] [CrossRef]
  93. Pofi, R.; Fiore, D.; De Gaetano, R.; Panio, G.; Gianfrilli, D.; Pozza, C.; Barbagallo, F.; Xiang, Y.K.; Giannakakis, K.; Morano, S.; et al. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Khames, A.; Khalaf, M.M.; Gad, A.M.; Abd El-Raouf, O.M. Ameliorative effects of sildenafil and/or febuxostat on doxorubicin-induced nephrotoxicity in rats. Eur. J. Pharmacol. 2017, 805, 118–124. [Google Scholar] [CrossRef] [PubMed]
  95. Baijnath, S.; Murugesan, S.; Mackraj, I.; Gathiram, P.; Moodley, J. The effects of sildenafil citrate on urinary podocin and nephrin mRNA expression in an L-NAME model of pre-eclampsia. Mol. Cell. Biochem. 2017, 427, 59–67. [Google Scholar] [CrossRef] [PubMed]
  96. Sonneveld, R.; Hoenderop, J.G.; Isidori, A.M.; Henique, C.; Dijkman, H.B.; Berden, J.H.; Tharaux, P.L.; van der Vlag, J.; Nijenhuis, T. Sildenafil Prevents Podocyte Injury via PPAR-gamma-Mediated TRPC6 Inhibition. J. Am. Soc. Nephrol. 2017, 28, 1491–1505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Ali, B.H.; Al Za’abi, M.; Adham, S.A.; Al Suleimani, Y.; Karaca, T.; Manoj, P.; Al Kalbani, J.; Yasin, J.; Nemmar, A. The effect of sildenafil on rats with adenine-Induced chronic kidney disease. Biomed. Pharmacother. 2018, 108, 391–402. [Google Scholar] [CrossRef] [PubMed]
  98. Mehanna, O.M.; El Askary, A.; Al-Shehri, S.; El-Esawy, B. Effect of phosphodiesterase inhibitors on renal functions and oxidant/antioxidant parameters in streptozocin-induced diabetic rats. Arch. Physiol. Biochem. 2018, 124, 424–429. [Google Scholar] [CrossRef]
  99. Danisoglu, M.E.; Aytac, B.; Kilicaslan, H.; Dogan, S.; Vuruskan, H. Reduction of shock wave lithotripsy-induced renal tubular injury by tadalafil. Bratisl. Lek. Listy 2013, 114, 616–620. [Google Scholar] [CrossRef] [Green Version]
  100. Ozmerdiven, G.; Vuruskan, B.A.; Kaygisiz, O.; Vuruskan, H. Protective effects of diltiazem and tadalafil on shock wave-induced kidney injury in rats. Bratisl. Lek. Listy 2017, 118, 228–232. [Google Scholar] [CrossRef] [Green Version]
  101. Qi, M.Y.; Kai, C.; Liu, H.R.; Su, Y.H.; Yu, S.Q. Protective effect of Icariin on the early stage of experimental diabetic nephropathy induced by streptozotocin via modulating transforming growth factor beta1 and type IV collagen expression in rats. J. Ethnopharmacol. 2011, 138, 731–736. [Google Scholar] [CrossRef]
  102. Liang, S.R.; Bi, J.W.; Guo, Z.L.; Bai, Y.; Hu, Z. Protective effect of icariin on kidney in 5/6 nephrectomized rats and its mechanism. Genet. Mol. Res. 2014, 13, 6466–6471. [Google Scholar] [CrossRef]
  103. Huang, Z.; He, L.; Huang, D.; Lei, S.; Gao, J. Icariin protects rats against 5/6 nephrectomy-induced chronic kidney failure by increasing the number of renal stem cells. BMC Complement. Altern. Med. 2015, 15, 378. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Su, B.; Ye, H.; You, X.; Ni, H.; Chen, X.; Li, L. Icariin alleviates murine lupus nephritis via inhibiting NF-kappaB activation pathway and NLRP3 inflammasome. Life Sci. 2018, 208, 26–32. [Google Scholar] [CrossRef] [PubMed]
  105. Hohenstein, B.; Daniel, C.; Wittmann, S.; Hugo, C. PDE-5 inhibition impedes TSP-1 expression, TGF-beta activation and matrix accumulation in experimental glomerulonephritis. Nephrol. Dial. Transplant. 2008, 23, 3427–3436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Wang, H.G.; Yan, X.Y.; Cai, Y.M.; Wang, H.G.; Chen, J.Y. The effect of phosphodiesterase isoenzyme 5 inhibitors on ureteropelvic junction obstruction in rabbits. Urol. Int. 2009, 83, 316–322. [Google Scholar] [CrossRef]
  107. Fang, L.; Radovits, T.; Szabo, G.; Mozes, M.M.; Rosivall, L.; Kokeny, G. Selective phosphodiesterase-5 (PDE-5) inhibitor vardenafil ameliorates renal damage in type 1 diabetic rats by restoring cyclic 3′,5′ guanosine monophosphate (cGMP) level in podocytes. Nephrol. Dial. Transplant. 2013, 28, 1751–1761. [Google Scholar] [CrossRef] [Green Version]
  108. Essiz, D.; Sozmen, M.; Sudagidan, M.; Devrim, A.K. Phosphodiesterase type 5 inhibition attenuates cyclosporine A induced nephrotoxicity in mice. Biotech. Histochem. 2015, 90, 167–178. [Google Scholar] [CrossRef]
  109. Sekizawa, T.; Shima, Y.; Yoshida, K.; Tanahashi, M.; Yoshida, M.; Suzuki-Kusaba, M.; Hisa, H.; Satoh, S. Effects of zaprinast on renal nerve stimulation-induced anti-natriuresis in anaesthetized dogs. Clin Exp Pharmacol. Physiol. 1998, 25, 1008–1012. [Google Scholar] [CrossRef]
  110. Yang, J.W.; Han, S.T.; Kim, Y.S.; Song, S.H.; Kim, B.R.; Eom, M.S.; Jung, S.H.; Choi, S.O.; Han, B.G. Effects of a cGMP-specific phosphodiesterase inhibitor on expression of endothelial nitric oxide synthase and vascular endothelial growth factor in rats with cyclosporine-induced nephrotoxicity. Transplant. Proc. 2010, 42, 4625–4632. [Google Scholar] [CrossRef]
  111. Yuan, J.; Zhang, R.; Yang, Z.; Lee, J.; Liu, Y.; Tian, J.; Qin, X.; Ren, Z.; Ding, H.; Chen, Q.; et al. Comparative effectiveness and safety of oral phosphodiesterase type 5 inhibitors for erectile dysfunction: A systematic review and network meta-analysis. Eur. Urol. 2013, 63, 902–912. [Google Scholar] [CrossRef]
  112. Dhaliwal, A.; Gupta, M. PDE5 Inhibitor; StatPearls: Treasure Island, FL, USA, 2020. [Google Scholar]
  113. Tzoumas, N.; Farrah, T.E.; Dhaun, N.; Webb, D.J. Established and emerging therapeutic uses of PDE type 5 inhibitors in cardiovascular disease. Br. J. Pharmacol. 2019. [Google Scholar] [CrossRef] [Green Version]
  114. Oelke, M.; Weiss, J.P.; Mamoulakis, C.; Cox, D.; Ruff, D.; Viktrup, L. Effects of tadalafil on nighttime voiding (nocturia) in men with lower urinary tract symptoms suggestive of benign prostatic hyperplasia: A post hoc analysis of pooled data from four randomized, placebo-controlled clinical studies. World J. Urol. 2014, 32, 1127–1132. [Google Scholar] [CrossRef] [PubMed]
  115. Sakalis, V.I.; Karavitakis, M.; Bedretdinova, D.; Bach, T.; Bosch, J.; Gacci, M.; Gratzke, C.; Herrmann, T.R.; Madersbacher, S.; Mamoulakis, C.; et al. Medical Treatment of Nocturia in Men with Lower Urinary Tract Symptoms: Systematic Review by the European Association of Urology Guidelines Panel for Male Lower Urinary Tract Symptoms. Eur. Urol. 2017, 72, 757–769. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Dell’Agli, M.; Galli, G.V.; Dal Cero, E.; Belluti, F.; Matera, R.; Zironi, E.; Pagliuca, G.; Bosisio, E. Potent inhibition of human phosphodiesterase-5 by icariin derivatives. J. Nat. Prod. 2008, 71, 1513–1517. [Google Scholar] [CrossRef] [PubMed]
  117. Mackenzie, A.E.; Lappin, J.E.; Taylor, D.L.; Nicklin, S.A.; Milligan, G. GPR35 as a Novel Therapeutic Target. Front. Endocrinol. (Lausanne) 2011, 2, 68. [Google Scholar] [CrossRef] [Green Version]
  118. Keswani, A.N.; Peyton, K.J.; Durante, W.; Schafer, A.I.; Tulis, D.A. The cyclic GMP modulators YC-1 and zaprinast reduce vessel remodeling through antiproliferative and proapoptotic effects. J. Cardiovasc. Pharmacol. Ther. 2009, 14, 116–124. [Google Scholar] [CrossRef] [Green Version]
  119. Zhang, W.H.; Zhang, X.H. Clinical and preclinical treatment of urologic diseases with phosphodiesterase isoenzymes 5 inhibitors: An update. Asian J. Androl. 2016, 18, 723–731. [Google Scholar] [CrossRef]
  120. Kang, S.G.; Kim, J.J. Udenafil: Efficacy and tolerability in the management of erectile dysfunction. Ther. Adv. Urol. 2013, 5, 101–110. [Google Scholar] [CrossRef]
  121. Aujla, H.; Kumar, T.; Wozniak, M.; Dott, W.; Sullo, N.; Joel-David, L.; Morris, T.; Brookes, C.; Barber, S.; Murphy, G.J. Effect of sildenafil (Revatio) on postcardiac surgery acute kidney injury: A randomised, placebo-controlled clinical trial: The REVAKI-2 trial protocol. Open Heart 2018, 5, e000838. [Google Scholar] [CrossRef]
  122. Paick, J.S.; Ahn, T.Y.; Choi, H.K.; Chung, W.S.; Kim, J.J.; Kim, S.C.; Kim, S.W.; Lee, S.W.; Min, K.S.; Moon, K.H.; et al. Efficacy and safety of mirodenafil, a new oral phosphodiesterase type 5 inhibitor, for treatment of erectile dysfunction. J. Sex. Med. 2008, 5, 2672–2680. [Google Scholar] [CrossRef]
  123. Glina, S.; Fonseca, G.N.; Bertero, E.B.; Damiao, R.; Rocha, L.C.; Jardim, C.R.; Cairoli, C.E.; Teloken, C.; Torres, L.O.; Faria, G.E.; et al. Efficacy and tolerability of lodenafil carbonate for oral therapy of erectile dysfunction: A phase III clinical trial. J. Sex. Med. 2010, 7, 1928–1936. [Google Scholar] [CrossRef]
  124. Tzortzis, V.; Mitrakas, L.; Gravas, S.; Mamoulakis, C.; Meissner, A.; Kyriakou, D.; Melekos, M.D. Oral phosphodiesterase type 5 inhibitors alleviate recurrent priapism complicating thalassemia intermedia: A case report. J. Sex. Med. 2009, 6, 2068–2071. [Google Scholar] [CrossRef] [PubMed]
  125. Sofikitis, N.; Kaltsas, A.; Dimitriadis, F.; Rassweiler, J.; Grivas, N.; Zachariou, A.; Kaponis, A.; Tsounapi, P.; Paterakis, N.; Karagiannis, A.; et al. The Effect of PDE5 Inhibitors on the Male Reproductive Tract. Curr. Pharm. Des. 2020. [Google Scholar] [CrossRef] [PubMed]
  126. Bender, A.T.; Beavo, J.A. Cyclic nucleotide phosphodiesterases: Molecular regulation to clinical use. Pharmacol. Rev. 2006, 58, 488–520. [Google Scholar] [CrossRef] [PubMed]
  127. Afsar, B.; Ortiz, A.; Covic, A.; Gaipov, A.; Esen, T.; Goldsmith, D.; Kanbay, M. Phosphodiesterase type 5 inhibitors and kidney disease. Int. Urol. Nephrol. 2015, 47, 1521–1528. [Google Scholar] [CrossRef] [PubMed]
  128. Shen, K.; Johnson, D.W.; Gobe, G.C. The role of cGMP and its signaling pathways in kidney disease. Am. J. Physiol. Renal. Physiol. 2016, 311, F671–F681. [Google Scholar] [CrossRef] [PubMed]
  129. Yamanaka, R.; Shindo, Y.; Hotta, K.; Suzuki, K.; Oka, K. NO/cGMP/PKG signaling pathway induces magnesium release mediated by mitoKATP channel opening in rat hippocampal neurons. FEBS Lett. 2013, 587, 2643–2648. [Google Scholar] [CrossRef] [Green Version]
Figure 1. PDE5I-induced smooth muscle relaxation in the corpora cavernosa. cGMP is the principal mediator of NO-induced smooth muscle relaxation/vasodilation [35]. cGMP propels a series of intracellular changes including inhibition of Ca2+ entry into the cell, Ca2+ shift into the endoplasmic reticulum, activation of K+ channels leading to membrane hyperpolarization, and stimulation of a cGMP-dependent protein kinase that activates a myosin light chain phosphatase. All these actions promote smooth muscle relaxation. NO penetrates the cytoplasm of smooth muscle cells binding to guanylyl cyclase (sGC), which catalyzes the enzymatic conversion of GTP to cGMP. Elevation of cGMP stimulates cGMP-dependent protein kinase G leading to PDE5 phosphorylation/activation. PDE5 hydrolyzes cGMP in the cavernosal tissue. Inhibition of PDE5 results in smooth muscle relaxation with increased arterial blood flow, leading to compression of the sub-tunical venous plexus followed by penile erection [36].
Figure 1. PDE5I-induced smooth muscle relaxation in the corpora cavernosa. cGMP is the principal mediator of NO-induced smooth muscle relaxation/vasodilation [35]. cGMP propels a series of intracellular changes including inhibition of Ca2+ entry into the cell, Ca2+ shift into the endoplasmic reticulum, activation of K+ channels leading to membrane hyperpolarization, and stimulation of a cGMP-dependent protein kinase that activates a myosin light chain phosphatase. All these actions promote smooth muscle relaxation. NO penetrates the cytoplasm of smooth muscle cells binding to guanylyl cyclase (sGC), which catalyzes the enzymatic conversion of GTP to cGMP. Elevation of cGMP stimulates cGMP-dependent protein kinase G leading to PDE5 phosphorylation/activation. PDE5 hydrolyzes cGMP in the cavernosal tissue. Inhibition of PDE5 results in smooth muscle relaxation with increased arterial blood flow, leading to compression of the sub-tunical venous plexus followed by penile erection [36].
Jcm 09 01284 g001
Figure 2. PRISMA flow chart showing the study selection procedure.
Figure 2. PRISMA flow chart showing the study selection procedure.
Jcm 09 01284 g002
Figure 3. Reno-protective mechanisms of PDE5Is.
Figure 3. Reno-protective mechanisms of PDE5Is.
Jcm 09 01284 g003
Table 1. Human studies evaluating the potential reno-protective effects of phosphodiesterase 5 inhibitors.
Table 1. Human studies evaluating the potential reno-protective effects of phosphodiesterase 5 inhibitors.
Reference
Country/Year
Type of StudyAKI ModelPDE5I RouteTimingSampleAKI Renal EffectsPDE5I Renal EffectsOutcome
[24]/
China/2019
Preclinical study on HEK-293 cell cultureCisplatin
Various doses
Finally chosen 20 μΜ dose
24 h
Icariin
Various doses
(0.25–2.0 μΜ)
24 h prior to cisplatin
PRECentrifuged at 4 °C, 10,000 g, for 20 minReduced viability, ↑p-NF-Kb
↓GSH concentration
↑MDA levels,↑Bax, ↓Bcl-2
↑ROS generation, ↑Caspace 3
↑iNOS/TNF-a/IL-1β
Nuclear fragmentation and cellular condensation
Improved viability, ↓p-NF-kB
↑GSH concentration
↓MDA levels, ↓Bax, ↑Bcl-2
↓ ROS generation, ↓Caspace 3
↓iNOS/TNF-a/IL-1β
Blunted apoptotic changes
Antiapoptotic action (PI3K/Akt pathway)
POS
[38]/
China/2017
Preclinical study using huMSCs in adult male Wistar rats2.5% Adenine
Orally
4 weeks
+4th generation huMSCs
Icariin
huMSCs were pretreated with 100 uM ICA for 1 week
PRE3, 7, 14 days after treatment↑Urine outputm, ↑Urea, ↑Cr
↑Damage renal tissue, ↑TNF-a
↓SOD, ↑MDA, ↑IL-6, ↑IL-10
↓Urine output,↓Urea, ↓Cr
↓Damage renal tissue, ↓TNF-a
↑SOD, ↓MDA, ↓IL-6, ↓IL-10
↑BMP-7, ↑bFGF
POS
[17]/
Israel/2015
Clinical trial
(non-RCT)
PN with 20 min cold ischemiaTadalafil
Orally: 20 mg/day
1 day pre-operatively and 2 days postoperatively
PRE and POSTPre-op and at 1,3,8, 24, 48, 72 h post op↑NGAL, ↑KIM-1,↑sCr, ↓GFRAttenuated all studied parametersPOS
[39]/
USA/2016
Clinical trial
(RCT)
RAPNSildenafil
Orally
100 mg prior to RAPN
PRE ↓GFR↓GFR (No improvement)NEUT
Abbreviations: AKI, acute kidney injury; Bax, proapoptotic protein; Bcl-2, antiapoptotic gene; bFGF, basic fibroblast growth factor; BMP-7, bone morphogenetic protein-7; GSH, glutathione; HEK, human embryonic kidney cells; huMSCs, human umbilical cord mesenchymal stem cells; iNOS, inducible NOS; IL, interleukin; LY6G, MDA, malondialdehyde; NOX-4, NADPH oxidase 4; PDE5I, phosphodiesterase 5 inhibitor; p-NF-Kb, phosphorylation nuclear factor kappa-light-chain-enhancer of activated B cells; PN, partial nephrectomy; RAPN, Robot assisted partial nephrectomy; RCT, randomized controlled trial; ROS, reactive oxygen species; sCr, serum creatinine; SOD, superoxide dismutase; TNF-a, tumor necrosis factor a; ↓, reduced; ↑, increased.
Table 2. Animal studies evaluating the potential reno-protective effects of sildenafil.
Table 2. Animal studies evaluating the potential reno-protective effects of sildenafil.
Reference
/Country/Year
Studied AnimalAKI ModelPDE5I RouteTimingSampleAKI Renal EffectsPDE5I Renal EffectsOutcome
[41]/
South Korea/2009
Male
Sprague Dawley rats
Cisplatin
Single intraperitoneal injection 5 mg/kg
Sildenafil
Intraperitoneal
0.4 mg/kg
Just after the injection of cisplatin
POSTLeft nephrectomy 96 h post cisplatin
↑BUN, ↑sCr, ↑Bax/Bcl-2 ratio
↑Caspase 3 expression
↑TUNEL positive cells
Loss of brush border
Vacuolation/Desquamation
↓sCr, ↓Bax/Bcl-2 ratio
↓Caspase 3 expression
↓TUNEL positive cells
↑eNOS and iNOS
Significantly attenuated renal changes
POS
[30]/
Korea/2009
Male
Sprague Dawley rats
IR renal injury modelSildenafil
Intraperitoneal
0.5 mg/kg
1 h prior to ischemia
PREDepending on the group 0-168 h after reperfusion↑BUN, ↑sCr, ↑cGMP
↑Bax/Bcl-2 ratio, ↑Caspase 3 activity
↑TUNEL positive cells
Loss of brush border
Vacuolation/Desquamation
↓BUN, ↓sCr, ↑↑ cGMP
↓Bax/Bcl-2 ratio, ↓Caspase 3 activity
↓TUNEL positive cells
↑↑ ERK activity
Attenuated all histological changes
POS
[42]/
Turkey/2010
Male Wistar albino ratsIR renal injury modelSildenafil
Orally
60 min pre-operatively
PRELeft nephrectomy either at 45 min post occlusion or at 105 min post occlusion and reperfusion injury↑MPO enzyme level and activity
↑TBARS
Sclerosis of glomeruli
Enlargement of Bowman space
Loss of microvilli/Pyknotic nuclei
Tubular necrosis/Interstitial edema
Leucocyte infiltration
Glomerular and tubular degeneration
↓MPO enzyme level and activity
⇔TBARS
Attenuated tubular damage
Preserved normal morphology
Significantly decreased neutrophil infiltration
POS
[43]/
Brazil/2010
Wistar ratsIR renal injury modelSildenafil
Orally
1 mg/kg
60 min prior to ischemia
PREAt 24 h and 7 days scintigraphy and nephrectomyScintigraphy: functional deficit representing ATN No PDE5i: ↑ cellular necrosis Vacuolation
Intratubular cast formation
Reversed effect to normal split function
PDE5i: just dilatation of tubular lumen
No significant change in histology
POS
[44]/
Oman/2011
Male Wistar ratsCisplatin
Single intraperitoneal injection
5 mg/kg
Sildenafil
Intraperitoneally
0.4 mg/kg for 5 days
or
Sildenafil
Subcutaneously
10 mg/kg for 5 days
POSTBlood samples and bilateral nephrectomy 5 days post treatment↓RBF, ↓BP, ↓Body weight ↑Urine output
↑BUN, ↑sCr, ↓CrCl
↑N-acetyl-β-D-glycosaminidase
↑TNF-a (plasma and renal)
↑Renal platinum concentration
Acute Tubular Necrosis/Apoptotic cells
↑RBF, ↑BP (i.p.)
No improvement in b.w. and u.o.
↓BUN, ↓sCr, ↑CrCl (i.p.)
↓N-acetyl-β-D-glycosaminidase
Minimal improvement in TNF-a
No change in platinum concentration
Improvement of histological changes
POS
[45]/
Turkey/2011
Wistar albino ratsUUO model
Sildenafil-orally-1 mg/day
Vardenafil-orally-0.5 mg/day
Tadalafil-orally-10 mg/72 h
For 30 days
POST30 days↑Tubular cell apoptosis
↑ eNOS
↑ iNOS
↓ Tubular cell apoptosis
↓ eNOS
↓ iNOS
Sildenafil better results
POS
[46]/
Spain/2011
MinipigsIR renal injury modelSildenafil
Intravenously
0.7 or 1.4 mg/kg 30 min prior to or during warm ischemia
PRE OR SIMMonitoring of hemodynamics up to 45 min following unclamping ↓Systemic MAP (especially 1.4 mg/kg)
↑RVF (0.7 mg/kg)
POS
[47]/
Turkey/2011
Male Wistar ratsCLP model
Sildenafil
Orally
10 or 20 mg/kg
After the procedure
POST16 h after CLP↓SOD, ↓GSH, ↑MPO, ↑LPO
↑Mean inflammation score
↑TNF-a
↑SOD, ↑GSH, ↓MPO, ↓LPO
↓Mean inflammation score
↓TNF-a
POS
[48]/
United Kingdom
Female Large White Landrace crossbred pigsCardiopulmonary bypass 2.5 hSildenafil
Intravenously
10 mg in 50 mL saline 0.9%
SIM90 min pre-op
90 min post-op
24 h post-op
↓CCl, ↑Proteinuria, ↑IL-18
↓ NO
Pseudodilation of proximal tubules
↑iNOS
↑ cortical expression endothelin-1
Inflammatory cell infiltration
↑CCl ↓Proteinuria ↓IL-18
Significantly increased RBF (24 h)
↑NO
Prevented phenotypic changes in proximal tubular cells
↓cortical expression endothelin-1
Preserved eNOS
↓iNOS
↓ inflammatory cell infiltration
POS
[49]/
Turkey/2012
Male
Sprague Dawley rats
IR renal injury modelSildenafil
Orally: 1 mg/kg
60 min pre-operatively
Tadalafil
Orally: 1 mg/kg
60 min pre-operatively
PRENephrectomy post procedure↑MPO levels
↑MDA levels
↑iNOS gen, ↑eNOS expression
↑ apoptotic cells
↑p53 positive cells
Leucocyte migration
Edema/Tubular dilatation
MPO: no significant improvement
↓MDA (Sdf), ⇔ MDA (Tdf) levels
↓iNOS gen, ↓eNOS expression
↓apoptotic cell death ( Sdf > Tdf)
↓p53 positive cells
All changes were attenuated
POS
[50]/
Germany/2013
NO-GC1 KO mice
C57Bl/6Rj
UUO modelSildenafil
Orally
100 mg/kg
In the 4th week post op
POST4 weeks post op↓cGMP
↓NO-stimulated guanyle cyclase activity (KO mice)
↑cGMP
↑NO sensitivity
↓SBP (more efficient in operated group rather than KO group)
POS
[23]/
USA/2013
Female New Zealand white rabbitsFolic Acid
Intraperitoneally
Single dose
250 mg/kg
Sildenafil
Intraperitoneally
24 h after injury
0.3 mg/kg/day
For 6 days
POSTBlood samples and kidneys were harvested 24 h post treatment↓mRNA expression COX1 and Tfam
↓mtDNA copy number
↑KIM-1
↑mRNA expression COX1 and Tfam
↑mtDNA copy number
↓KIM-1
POS
[51]/
Egypt/2014
Sprague Dawley male ratsCisplatin
Intraperitoneally
6 mg/kg
Sildenafil
Intraperitoneally
2 mg/kg
1 h before and 24 h after cisplatin injection
PRE and POST96 h after cisplatin injection↑BUN, ↑sCr, ↑MDA, ↑TNF-a
↑Caspase-3, ↓SOD
↓Nitrite/nitrate level
Acute tubular necrosis
↓BUN, ↓sCr, ↓MDA, ↓TNF-a
↓Caspase-3, ↑SOD ↑Nitrite/nitrate
POS
[52]/
Turkey/2014
Adult female Wistar albino ratsBurn modelSildenafil
Orally
10 or 20 mg/kg just after burn
POST24 h after the scald burnRenal: ↑MDA, ↓Gpx, ↑VEGF
⇔ Flt-1, ⇔TAC, ⇔OSI, ⇔TOS Serum: ↑MDA, ↓Gpx, ⇔VEGF, ⇔Flt-1, ↓TAC, ⇔OSI, ↑TOS, ⇔Flt-1/VEGF ratio
Renal: ↓MDA, ↑Gpx, ↓VEGF
⇔Flt-1 (T10), ⇔TAC, ⇔OSI, ⇔TOS(T20)
Serum: ↓MDA, ↑Gpx, ⇔VEGF
⇔Flt-1, ↑TAC, ↓OSI (T10)
↑Flt-1/VEGF ratio (T10) ↓TOS (T10)
↓Histopathological scores (no significant difference in T20)
POS
[53]/
Egypt/2014
Male Wistar ratsGentamicin
Intraperitoneally
100 mg/kg/day for 6 days
Sildenafil
Orally
5 mg/kg/day for 6 days
1 h before gentamycin
PRE24 h after last gentamycin injection↑Cr, ↑Urea, ↑urinary albumin
↑MDA, ↑nitrite/nitrate levels
↓CAT (renal), ↓SOD, ↑iNOS, ↓eNOS
Degeneration and necrobiosis in epithelial cells
↓Cr, ↓Urea, ↓urinary albumin
↓MDA, ↓nitrite/nitrate levels
↑CAT (renal), ↑SOD
↓iNOS, ↑eNOS
Reversed histological alterations
POS
[54]/
USA/2014
Male wild-type (WT) littermates or PKG Tg miceUUO model
Sildenafil
Subcutaneously
12 mg/kg twice daily for 14 days
POST14 days↓Renal PKG activity
Increase (↑) at Ang II, Collagen type I, III mRNA, α-SMA, E-cadherin, TNF-a, TGF-β1, pSmad2, ICAM-1
↑Macrophage infiltration
↑Renal PKG activity
Decrease (↓) at Ang II, Collagen type I, III mRNA, α-SMA, E-cadherin, TNF-a, TGF-β1, pSmad2, ICAM-1
↓Macrophage infiltration
POS
[55]/
Brazil/2014
New Zealand white rabbitsCIN modelSildenafil
Orally
6 mg/kg before CM
or
6 mg/kg before CM and 8 hourly for 48 h
PRE and POST1/2/24/48
h
No changes in kidney to body weight ratio
↑sCr
↓Na, ↑K
Multifocal tubular necrosis
Tubular degeneration
Luminal protein casts
No significant changes in kidney to body weight ratio
↓↓sCr (continuous)
↑Na, ↓K
Continuous treatment blunted all changes
POS
[56]/
Egypt/2015
Male Sprague-Dawley ratsIR renal injury modelSildenafil
Orally
(1 mg/kg)
60 min before anesthesia
PREBlood + urine samples (basal, at 2, 24, 48 h and 7 days)
+
Kidney tissue
↑sCr, ↑BUN, ↓Bcl-2
↓Nrf2/HO-1/NQO-1 (genes)
↑ Proinflammatory cytokine genes (TNF-a, ICAM-1, IL-β)
↓Nrf 2 protein expression
Acute tubular necrosis, detachment of epithelial cells from basement membrane, intracellular cast formation, loss of brush border, neutrophil infiltration
No improvement in BUN/sCr, ↑Bcl-2
↑Nrf2/HO-1/NQO-1 (genes)
↓ Proinflamamtory cytokine genes (TNF-a, ICAM-1, IL-β)
↑ Nrf 2 protein expression
Improved histological features of renal injury (mild tubular necrosis)
POS
[57]/
Brazil/2016
Male Wistar ratsCIN modelSildenafil
Orally
50 mg/kg/d
7 days (started 5 days before CM)
PRE and POST48 h after CM administration↑BUN, ↑sCr, ↑urine protein
↓GFR, ↓RPF, ↑RVR
↑superoxide anions production
↑H2O2 production
↑peroxynitrite and hydroxyl production
⇔ NO
Reduced body weight
Renal hypertrophy
↓BUN, ↓sCr, ↓urine protein
↑GFR, ↑RPF, ↓RVR
⇔superoxide anions production
↓ H2O2 production
↓peroxynitriteand hydroxyl production
⇔ NO
No effect of PDE5 on histological changes
POS
[58]/
Egypt/2016
Male Wistar albino ratsIR renal injury modelSildenafil
Intraperitoneally
(0.5 + 1.0 mg/kg)
1 h before ischemia
PREBlood/kidney tissue samples 24 h after reperfusion↓CrCl, ↑ BUN, ↑Uric acid, ↑FeNa
↑Plasma potassium
↓GSH levels,↑TBARS, ↑SAG levels
Glomerular damage, detachment of basement membrane, loss of brush border, tubular dilation, atroprhy, neutrophil accumulation
↑CrCl, ↓BUN, ↓Uric acid
↓FeNa
↓Plasma potassium ↑GSH levels, ↓TBARS ↓ SAG levels
↓Renal tissue damage
POS
[59]/
Turkey/2018
Female
Wistar albino rats
CIN modelSildenafil
Orally
50 mg/kg 48 h prior to CM
PRE48 h after CM administration↑HIF-2a (serum and tissue)
↑ BUN, ↑Cr (serum and urine)
Hemorrhage, shedding of brush border, tubular vacuolization, degeneration, inflammatory cell infiltration, intratubulat cast obstruction
↓HIF-2a (serum and tissue) ↓ sCr
Sildenafil improved all histological changes
POS
[60]/
Egypt/2018
Male albino ratsCisplatin
5 mg/kg
Single dose intraperitoneally
Combination
Sildenafil, Orally
40 mg/kg
Gemfibrozil-Orally
100 mg/kg
14 days prior or after
PRE OR POSTDay 17↑sCr, ↓HO-1, ↓GSH
↓eNOS, ↓TNF-a
↑Tubular injury/tubular necrosis
All changes improved with sildenafil and gemfibrozil especially in the group given after cisplatinPOS
[61]/
Egypt/2019
Mongrel dogs (aged 2-3 years)IR renal injury modelSildenafil
Orally
1 mg/kg 1 h prior to operation
or
In the perfusion fluid
0.5 mg/kg during the operation
PRE OR SIMPrior and at the end of the experiment
(Day 1,3,7,14)
↑sCr, ↑BUN, ↓GFR
↑caspase 3, ↑Nrf2
↑TNF-a, ↑ IL-1Β, ↑ICAM -1
↓eNOS
Renal degeneration
Cortical and medullary interstitial fibrosis
↓sCr, ↓BUN, ↑GFR
↓caspase 3, ↑↑Nrf2
↓TNF-a, ↓IL-1Β, ↓ICAM -1
↑eNOS
Significantly improved all histological changes
POS
Abbreviations: AKI, acute kidney injury; Ang II, angiotensin II; Bax, proapoptotic protein; Bcl-2, antiapoptotic gene; BP, blood pressure; BUN, blood urea nitrogen; Ca2+, calcium; CAT, catalase; cGMP, cyclic guanosine monophosphate; CIN, contrast induced nephropathy; CLP, caecal ligation and puncture; COX, cyclo-oxygenase CrCl, creatinine clearance, eNOS, endothelial NOS, FeNa, fractional excretion of sodium, GFR, glomerular filtration rate; GPx, glutathione peroxidase; GSH, glutathione; HIF-2a, heterodimeric nuclear transcription factor-2 alpha; HO-1, heme oxygenase 1; IR, ischemia reperfusion; ICAM-1, intercellular adhesion molecule 1; IL, interleukin; iNOS, inducible NOS; K, potassium; KIM-1, kidney injury molecule-1; LPO, lipid peroxidation; MAP, mean arterial pressure; MDA, malondialdehyde; MPO, myeloperoxidase; Na, sodium; NO, nitric oxide; NRF2, nuclear erythroid related factor 2; OSI, oxidative stress index; P, phosphorus; PDE5I, phosphodiesterase 5 inhibitor; PKG, protein kinase G; pSmad2, antibody; RBF, renal blood flow; RPF, renal plasma flow; RRI, renal resistive index; RVF, renal vascular flow; RVR, renal vascular resistance; SAG, superoxide anion generation; sCr, serum creatinine; sFlt1, soluble fms-like tyrosine kinase-1; SOD, superoxide dismutase; SBP, systolic blood pressure; TAC, total antioxidant capacity; Tfam, mitochondrial transcription factor; TGF-β1, transforming growth factor beta 1; TBARS, thiobarbituric acid reactive substances; TNF-a, tumor necrosis factor a; TOS, total oxidant status; TUNEL, terminal deoxynucleotidyl transferase dUTP nick end labeling; UUO, unilateral ureteral obstruction; VEGF, vascular endothelia growth factor; ↓, reduced; ↑, increased ⇔, no change.
Table 3. Animal studies evaluating the potential reno-protective effects of tadalafil.
Table 3. Animal studies evaluating the potential reno-protective effects of tadalafil.
Reference/
Country/Year
Studied AnimalAKI ModelPDE5I RouteTimingSampleAKI Renal EffectsPDE5I Renal EffectsOutcome
[62]/
Turkey/2019
New Zealand rabbitsUUO model
Tadalafil
Orally
10 mg/72 h for 30 days prior to obstruction
PRE4th hour and 1st and 3rd day↑Resistivity index
↑Pulsatility index
↓Resistivity index
↓Pulsatility index
In the non-obstructed kidney reduced resistivity index at 4th hour then normal
POS
[63]/
Turkey/2011
Male
Sprague Dawley rats
IR renal injury modelTadalafil
Orally
1 mg/kg
60 min pre-operatively
PREAt 45 min post occlusion or at 105 min post occlusion and reperfusion injurySclerosis of glomeruli
Enlargement of Bowman space
Loss of microvilli/Tubular necrosis Interstitial edema/Leucocyte infiltration
Hyaline degeneration
Attenuated histological changes and decreased neutrophil infiltrationPOS
[45]/
Turkey/2011
Wistar albino ratsUUO model
Sildenafil-orally-1 mg/day
Vardenafil-orally-0.5 mg/day
Tadalafil-orally-10 mg/72 h
For 30 days
POST30 days↑Tubular cell apoptosis
↑eNOS
↑iNOS
↓Tubular cell apoptosis
↓eNOS
↓iNOS
Sildenafil better results
POS
[64]/
Turkey/2011
Male Wistar albino ratsIR renal injury modelTadalafil
Orally
10 mg/kg
60 min pre-operatively
PRELeft nephrectomy at 120 min post-operatively↑Total oxidant status
Tubular necrosis/Vacuolization Congestion/Mononuclear cell infiltration
↑ Total antioxidant status
Reduced all injuries to the renal tissue.
POS
[65]/
USA/2012
Adult female pigsIR renal injury modelTadalafil
40 mg
Two doses (12 h before and just prior to surgery)
PREInduction
and
Days 1, 3, 7 post occlusion
↑Creatinine after nephrectomy
↑↑ Creatinine Day 1 post ischemia
↓Creatinine after nephrectomy
No significant change in creatinine Day 1 post ischemia
POS
[49]/
Turkey/2012
Male
Sprague Dawley rats
IR renal injury modelSildenafil
Orally: 1 mg/kg
60 min pre-operatively
Tadalafil
Orally: 1 mg/kg
60 min pre-operatively
PRENephrectomy post procedure↑ MPO levels
↑MDA levels
↑iNOS gen, ↑eNOS expression
↑apoptotic cells
↑p53 positive cells
Leucocyte migration
Edema/Tubular dilatation
MPO: no significant improvement
↓MDA (Sdf), ⇔MDA (Tdf) levels
↓iNOS gen, ↓eNOS expression
↓apoptotic cell death (Sdf > Tdf)
↓p53 positive cells
All changes were attenuated
POS
[66]/
Israel/2013
Male
Sprague Dawley rats
IR renal injury modelTadalafil
Orally
10 mg/kg
24-hr prior to ischemia
PRE30/60 min after nephrectomy
60/120/180/240
min after clamping
↑V, ↑UNaV, ↑FeNa, ↓GFR, ⇔RPF, ↑NGAL, ↑KIM-1
Tubular dilatation/Loss of brush border
Necrosis and cast formation
↓V, ↓UNaV, ↓FeNa, ↑GFR, ↑RPF, ↓NGAL, ↓KIM-1
Blunted all changes
POS
[67]/
China/2014
Male Wistar ratsSepsis modelTadalafil
Orally
10 mg/kg
24 h prior to procedure for 28 days
PRE and POSTNephrectomy and samples at: 8 days post treatment and 6 weeks post treatment↑Systolic and diastolic BP, ↑NO
↑BUN, ↑sCr, ↑MDA levels ↓SOD, ↑TGF-β
↓Systolic and diastolic BP, ↓NO, ↓BUN, ↓sCr, ↓MDA levels, ↑SOD
↑IL-10, ↓TNF-a, ↓IL-1β, ↓TGF-β
↓RANTES, ↓MIP-1β, ↓MCP-1
POS
[68]/
Turkey/2015
Female Wistar albino ratsIR renal injury modelTadalafil
Orally
10 mg/kg
24 h prior to procedure
PRECardiac blood samples and nephrectomy after reperfusion injuryNo significant difference
Severe tubular dilatation degeneration and necrosis/Enlargement of Bowman capsule
in IMA/NO/MDA levels
Blunted all changes
POS
[69]/Turkey/2015Wistar albino ratsIR renal injury modelTadalafil
Intraperitoneally
10 mg/kg
Immediately prior to procedure
PREBlood samples and nephrectomy following 60 min of reperfusion injury↑MDA levels (serum/renal)
↓TAC levels (serum/renal)
↑APAF-1, ↑iNOS, ↑eNOS
Loss of nucleus/Cellular edema
Vacuolization/Brush border loss
Tubular dilatation/edema
Interstitial congestion
⇔MDA (renal), ↓MDA (serum)
⇔TAC (renal), ↑TAC (serum)
↓APAF-1, ↓iNOS, ↓eNOS
Damage was significantly less after tadalafil treatment
POS
[35]/Turkey/2015Female Wistar albino ratsCIN modelTadalafil
Orally
10 mg/kg
immediately after contrast
POST48 h after CM administrationSignificant weight loss after dehydration
↑Serum cystatin C
↑BUN, ↑sCr, ↑MDA
Medullary congestion
Significant weight loss after dehydration
↓Serum cystatin C
↓BUN, ↓sCr, ↓MDA
Similar histological findings
POS
[29]/
Egypt/2016
Adult male albino ratsIR renal injury modelTadalafil
Orally
(5 mg/kg)
Pre-treatment
PREBlood/kidney tissue samples 6 h after reperfusion↑sCr, ↑BUΝ, ↑MDA levels
↓SOD activity, ↑MPO activity
↑ICAM-1, ↑TNF-a, ↑IL-1β
↑Caspase-3 activity
Congestion and interstitial hemorrhage, proximal and tubular necrosis
↓sCr, ↓BUΝ, ↓MDA levels
↑SOD activity, ↓MPO activity
↓ICAM-1, ↓TNF-a, ↓IL-1β
↓Caspase-3 activity
Dilated proximal, distal, and collecting tubules and interstitial connection
POS
[70]/
Nigeria/2016
Male Wistar ratsCisplatin
Intraperitoneal
5 mg/kg
Tadalafil
Orally: 2 or 5 mg/kg for 7 days pretreatment
PREBlood samples and renal tissue obtained 3 days post cisplatin↓Na/K/HCO3/Ca2+/P
↑BUN, ↑sCr, ↑MDA/GPx
↓GSH/SOD/CAT (renal)
Significant attenuation of all histological and biochemical alterationsPOS
[71]/
Israel/2017
Male albino Wistar ratsCLP modelTadalafil
Orally
5 or 10 mg/kg
End of the procedure
POSTLeft nephrectomy
+
Blood samples
16 h postoperatively
↓CAT, ↓SOD, ↑IL-6, ↑sCr, ↑MPO, ↑MDA, ↑Cystatin C
↑Mac387 antibody
↑Tubular injury, glomerulus deformities
↑Inflammatory cell infiltration
↑CAT, ↑SOD, ↓IL-6, ↓sCr, ↓MPO, ↓MDA, ↓Cystatin C
↓Mac387 antibody
↓Tubular injury, glomerulus deformities
↓Inflammatory cell infiltration
POS
[72]/
Brazil/2017
Male Wistar ratsIR renal injury modelTadalafil
Orally
10 mg/kg
1 h pre-procedure
PREAfter nephrectomyInterstitial Leucocyte accumulationSuccessful reversal by tadalafilPOS
[73]/
Brazil/2017
Male Wistar ratsIR renal injury modelTadalafil
Orally: 10 mg/kg
1 h before ischemia
PREFluorescence imaging (ICG)
Blood samples
↓ICG signal, ↑TNF-a, ↑IL-1β
↑IL-6 ↑BUN, ↑sCr, ↑CRP
↑ICG signal, ↓TNF-a, ↓IL-1β
↓IL-6 ↓BUN, ↓sCr, ↓CRP
POS
[74]/
Turkey/2019
Male Sprague Dawley ratsUUO modelTadalafil
Orally
10 mg/72 h
---15 days post ligation↑aSMA, ↑TGF-β
Partial: inflammatory cell infiltration/severe epithelial atrophy/edema of epithelial cells/vacuolation
Complete: macrophage infiltration/hemorrhage/irregular dark nuclei/thinner epithelium/denuded epithelial cells
↓aSMA, ↓TGF-β


Attenuation of all changes with tadalafil
POS
Abbreviation: AKI, acute kidney injury; APAF-1, apoptotic protease activating factor 1; aSMA, α-smooth muscle actin; BUN, blood urea nitrogen; Ca2+, calcium; CAT, catalase; CIN, contrast induced nephropathy; CLP, caecal ligation and puncture; CRP, c-reactive protein; eNOS, endothelial NOS; FeNa, fractional excretion of sodium; GFR, glomerular filtration rate; GPx, glutathione peroxidase; GSH, glutathione; HCO3, bicarbonate; IR, ischemia/reperfusion; ICAM-1, intercellular adhesion molecule 1; IL, interleukin; ICG, indocyanine green; IMA, ischemia modified albumin; iNOS, inducible NOS; K, potassium; KIM-1, kidney injury molecule-1; Mac387, Macrophage antibody; MCP-1, monocyte chemoattractant protein 1; MDA, malondialdehyde; MIP-1β, macrophage inflammatory protein-1β; MPO, myeloperoxidase; Na, sodium; NGAL, neutrophil gelatinase-associated lipocalin; NO, nitric oxide; P, phosphorus; PDE5I, phosphodiesterase 5 inhibitor; RANTES, Regulated upon Activation Normal T-cell Expressed, and Secreted; RPF, renal plasma flow; sCr, serum creatinine; Sdf, sildenafil; SOD, superoxide dismutase; TAC, total antioxidant capacity; Tdf, tadalafil; TGF-β1, transforming growth factor beta 1; TNF-a, tumor necrosis factor a; UNaV, urine sodium volume; UUO, unilateral ureteral obstruction; V, urine volume; ↓, reduced; ↑, increased ⇔, no change.
Table 4. Animal studies evaluating the potential reno-protective effects of icariin.
Table 4. Animal studies evaluating the potential reno-protective effects of icariin.
Reference/
Country/Year
Studied AnimalAKI ModelPDE5I RouteTimingSampleAKI Renal EffectsPDE5I Renal EffectsOutcome
[75]/
China/2015
Male BALB/c miceCisplatin
15 mg/kg
Intraperitoneal
Icariin
Orally
30 or 60 mg/kg/day
For 6 days
PREAt 6 days↑BUN, ↑sCr, ↑MDA
↓GSH concentration, ↓Catalase
↓SOD activity, ↑TNF-a, ↑NF-Kb
↑TUNEL positive cells
↑Caspase-3, ↓Bcl-2
Tubular congestion/edema
Loss of brush border/Tubular cell flattening and necrosis/nuclear pyknosis
Severe invasion of inflammatory cells
↓BUN, ↓sCr,↓MDA
↑GSH concentration, ↑Catalase
↑SOD activity, ↓TNF-a, ↓NF-kB
↓TUNEL positive cells
↓Caspase-3, ↑Bcl-2
Partial improvement of the features (dose dependent)
POS
[18]/
China/2018
Male C57BL/6N miceCLP modelIcariin
Orally
30 or 60 mg/kg
3 days prior to surgery
PREObserved for 5 days↑BUN, ↑sCr, ↑MDA levels
↑IL-1β/IL-6/TNF-a ↑ NF-κB
↓ GSH concentration
↓Catalase, ↓SOD activity
↑TUNEL +ve cells
↑Renal vascular permeability
↑Bax,↓Bcl-2, ↑Caspase 3
Extensive tubular necrosis/Loss of brush border
↓BUN, ↓sCr, ↓MDA levels
↓IL-1β/IL-6/TNF-a, ↓ NF-κB
↑GSH concentration
↑Catalase, ↑SOD activity
↓TUNEL +ve cells (60>30)
↓Renal vascular permeability
↓Bax, ↑Bcl-2, ↓Caspase 3
↑Survival (both doses)
Improvement in all histological features
POS
[76]/
Taiwan/2019
Adult C57BL/6JUUO modelIcariin
Orally
20 mg/kg/day
For 3 days prior and 3, 7, or 14 days after
PRE and POST3, 7, or 14 days post ligation↑TGF-β, ↑α-SMA ↑fibronectin
↑NOX-4, ↓E-cadherin, ↓SOD-1
↓Catalase, ↑CTGF, ↑Ly6G
↑F4/80, ↑phosphorylation IL-1β
↑Phosphorylation COX-2/NF-κΒ-65
Tubular dilatation/interstitial cell proliferation/inflammatory cell infiltration/tuft to capsule glomerular adhesions/collagen deposition
↓TGF-β, ↓α-SMA, ↓fibronectin
↓NOX-4,↑E-cadherin, ↑SOD-1
↑Catalase, ↓CTGF, ↓Ly6G
↓F4/80, ↓phosphorylation IL-1β
↓Phosphorylation COX-2/NF-κΒ-65
Non-significant moderate reversal by icariin
POS
Abbreviations: AKI, acute kidney injury; Bcl-2, antiapoptotic gene; BUN, blood urea nitrogen; CLP, caecal ligation and puncture; COX, cyclo-oxygenase; CTGF, connective tissue growth factor; F4/80, macrophage marker; GSH, glutathione; IL, interleukin; LY6G, neutrophil marker; MDA, malondialdehyde; NF-κB, nuclear factor kappa-like chain-enhancer of activated B cells; NOX-4, NADPH oxidase 4; PDE5I, phosphodiesterase 5 inhibitor; sCr, serum creatinine; SOD, superoxide dismutase; TGF-β1, transforming growth factor beta 1; TNF-a, tumor necrosis factor a; TUNEL, Terminal deoxynucleotidyl transferase dUTP nick end labeling; UUO, unilateral ureteral obstruction; ↓, reduced; ↑, increased.
Table 5. Animal studies evaluating the potential reno-protective effects of vardenafil.
Table 5. Animal studies evaluating the potential reno-protective effects of vardenafil.
Reference/
Country/Year
Studied AnimalAKI ModelPDE5I RouteTimingSampleAKI Renal EffectsPDE5I Renal EffectsOutcome
[45]/
Turkey/2011
Wistar albino ratsUUO model
Sildenafil-orally-1 mg/day
Vardenafil-orally-0.5 mg/day
Tadalafil-orally-10 mg/72 h
For 30 days
POST30 days↑Tubular cell apoptosis
↑eNOS
↑ iNOS
↓Tubular cell apoptosis
↓ eNOS
↓iNOS
Sildenafil better results
POS
[77]/
Greece/2013
Male Wistar ratsIR renal injury model
Vardenafil
Intravenously
0.02, 0.2, 2, 20 μg/kg
1 h pre-operatively
or 2μg/kg 45 min post occlusion
PRE or POSTBlood samples and right nephrectomy 4 h post ischemiaEdema
Loss of brush border
Nuclear condensation
↓sCr (0.2, 2, 20 μg/kg)
No change when given post-ischemia
↓FENa, ↑Renal uptake of tracer
↑cGMP, ↑ERK 1/2 phosphorylation
Renoprotection (in scintigraphy)
Significant improvement in all histo-logical changes irrespectively of dose
POS
[78]/
Brazil/2015
Male Wistar ratsIR renal injury modelVardenafil
Solution in a probe (1 mg/mL in 10 mg/kg)
1 h prior the ligation
PRELeft nephrectomy
Cytophotometry
24 h after reperfusion
↑Cleaved caspase-3 ↑sCr
↑Vacuolar degeneration
↓ Cleaved caspase-3
↓ Vacuolar degeneration
POS
Abbreviations: AKI, acute kidney injury; cGMP, cyclic guanosine monophosphate; eNOS, endothelial NOS; ERK, extracellular signal-regulated kinase; FeNa, fractional excretion of sodium; IR, ischemia/reperfusion; iNOS, inducible NOS; PDE5I, phosphodiesterase 5 inhibitor; sCr, serum creatinine; UUO, unilateral ureteral obstruction; ↓, reduced; ↑, increased.
Table 6. Animal studies evaluating the potential reno-protective effects of zaprinast and udenafil.
Table 6. Animal studies evaluating the potential reno-protective effects of zaprinast and udenafil.
Reference/
Country/Year
Studied AnimalAKI ModelPDE5I RouteTimingSampleAKI Renal EffectsPDE5I Renal EffectsOutcome
[79]/
USA/1995
Male Sprague-Dawley ratsIR renal injury modelZaprinast
Intravenously
0.03 and 0.3 mg/kg/min
24 h after ischemia
POSTDuring clamping, PDE5i infusion, up to 6 days following ischemia↑sCr, ↓GFR↓sCr, ↑GFR, ↓Low MAP
↑UNaV, ↑Urinary cGMP
↑Cortical and medullary blood flow
POS
[40]/
USA/2013
Female Sprague-Dawley ratsIR renal injury modelZaprinast
Intraperitoneally
10 mg/kg or 20 mg/kg
Single dose
30 min pre-operatively
PRE24 h post operatively
blood samples and left nephrectomy
No statistically significant differrences in either BUN levels or sCr levels or histologic scores or TUNEL positive cellsNEUT
[80]/
Germany/2017
6-8-week-old miceUUO modelZaprinast,
Intraperitoneally
10 mg/kg/day for 7 days
POSTAfter 7 days↑cGMP, ↑sCr↑↑cGMP, ↑MMP9, ↑TGF-β
⇔sCr, ↓Collagen
POS
[21]/Turkey/2017Female Wistar albino ratsIR renal injury modelUdenafil
Orally: 10 mg/kg
1 h prior to clamping
PRE60 min and
24 h
after reperfusion
↑BUN, ↑sCr
↑MDA, ↑NGAL
↓BUN, ↓sCr
↓MDA, ↓NGAL
Lowest pathological damage rates
POS
Abbreviations: AKI, acute kidney injury; BUN, blood urea nitrogen; cGMP, cyclic guanosine monophosphate; GFR, glomerular filtration rate; IR renal, ischemia/reperfusion; MAP, mean arterial pressure; MDA, malondialdehyde; MMP9, Matrix metallopeptidase 9; NGAL, neutrophil gelatinase-associated lipocalin; PDE5I, phosphodiesterase 5 inhibitor; sCr, serum creatinine; TUNEL, terminal deoxynucleotidyl transferase dUTP nick end labeling; UNaV, urinary sodium excretion; UUO, unilateral ureteral obstruction; ↓, reduced; ↑, increased ⇔, no change.
Table 7. Main characteristics of phosphodiesterase 5 inhibitors.
Table 7. Main characteristics of phosphodiesterase 5 inhibitors.
PDE5iFDA ApprovedLaunch DatePharmacokineticsRecommended DosageIndicationsSide EffectsContraindicationsEmerging and Other Off-Label Therapeutic Applications
SildenafilYes1998Cmax = 560 µg/L
Tmax = 0.8–1 h
T1/2 = 2.6–3.7 h
Affected by heavy/fatty meals
ED: 25–100 mg OD
PAH: 5–20 mg TDS
ED
PAH
Headache: 12.8%
Flushing: 10.4%
Dyspepsia: 4.6%
Nasal congestion: 1.1%
Dizziness: 1.2%
Abnormal vision: 1.9%
Absolute:
  • Any form of organic nitrate or NO donors
  • Myocardial infarction, stroke, or life-threatening arrhythmia within the last 6 months
  • Resting BP <90/50 or >170/100
  • Unstable angina, angina with intercourse, CHF NYHA IV

Relative:
  • Known serious hypersensitivity reaction
  • Antihypertensive medication
  • a-blockers
  • Drugs that inhibit CYP34A
  • Penile rehabilitation after Radical Prostatectomy
  • Heart Failure/CVD
  • High altitude illness
  • Stroke/Neurodegenerative diseases
  • Peripheral neuropathy
  • Improving fertility
  • Peripheral Arterial Disease
  • Raynaud’s syndrome
  • Diabetic Nephropathy
  • AKI
  • CKD
  • Stuttering priapism
  • Premature ejaculation
  • Ureteral stones
  • Reyronie’s disease
  • Female sexual dysfunction
  • Overactive bladder
  • Diabetes mellitus
TadalafilYes2003Cmax = 378 µg/L
Tmax = 2 h
T1/2 = 17.5 h
Not affected by heavy/fatty meals
ED: 10-20 mg on demand
ED: 5 mg OD
LUTS: 5 mg OD
PAH: 40 mg
ED
PAH
LUTS
Headache: 14.5%
Flushing: 4.1%
Dyspepsia: 12.3%
Nasal congestion: 4.3%
Dizziness: 2.3%
Back pain: 6.5%
Myalgia: 5.7%
VardenafilYes2003Cmax = 18.7 µg/L
Tmax = 0.9 h
T1/2 = 3.9 h
Affected by heavy/fatty meals
ED: 5–20 mg
on demand
EDHeadache: 16%
Flushing: 12%
Dyspepsia: 4%
Nasal congestion: 10%
Dizziness: 2%
Abnormal vision: < 2%
AvanafilYes2013Cmax = 5.2 µg/L
Tmax = 0.5–0.75 h
T1/2 = 6–17 h
Affected by heavy/fatty meals
ED: 50–200 mg
on demand
EDHeadache: 9.3%
Flushing: 3.7%
Dyspepsia: uncommon
Nasal congestion 1.9%
Dizziness: 0.6%
Back pain: < 2%
Myalgia: < 2%
UdenafilNo2005Cmax = 1137 µg/L
Tmax = 0.76 h
T1/2 = 9.88 h
ED: 100 mg
on demand
EDHeadache: 2–9%
Flushing: 11–23%
Dyspepsia: uncommon
Nasal congestion: 4–7%
Red eye: 4–7%
Chest discomfort: 0–5%
LodenafilNo2007Cmax = 157 µg/L
Tmax = 1.2 h
T1/2 = 2.4 h
ED: 80 mg
on demand
EDHeadache: 15–22%
Flushing: 5–6%
Dyspepsia: 5–22%
Nasal congestion: 5–11%
Abnormal vision: 5–6%
MirodenafilNo2011Cmax = 2989 µg/L
Tmax = 1.4 h
T1/2 = 2.5 h
ED: 80 mg
on demand
EDHeadache: 8–11%
Flushing: 10–16%
Dyspepsia: 3%
Red eye: 3–4%
Chest discomfort: 0–3%
BenzamidenafilNo-IDIDIDIDIDID
DasantafilNo-IDIDIDIDIDID
IcariinNo-IDIDIDIDIDID
ZaprinastNo-IDIDIDIDIDID
Abbreviations: AKI, acute kidney injury; BP, blood pressure; Cmax, serum maximum concentration; CHF, chronic heart failure; CKD, chronic kidney disease; CVD, cardiovascular disease; ED, erectile dysfunction; ID, insufficient data; NO, nitric oxide; NYHA, New York Heart Association; OD, once daily; PAH, pulmonary arterial hypertension; PDE5I, phosphodiesterase 5 inhibitor; Tmax, transport maximum.

Share and Cite

MDPI and ACS Style

Georgiadis, G.; Zisis, I.-E.; Docea, A.O.; Tsarouhas, K.; Fragkiadoulaki, I.; Mavridis, C.; Karavitakis, M.; Stratakis, S.; Stylianou, K.; Tsitsimpikou, C.; et al. Current Concepts on the Reno-Protective Effects of Phosphodiesterase 5 Inhibitors in Acute Kidney Injury: Systematic Search and Review. J. Clin. Med. 2020, 9, 1284. https://doi.org/10.3390/jcm9051284

AMA Style

Georgiadis G, Zisis I-E, Docea AO, Tsarouhas K, Fragkiadoulaki I, Mavridis C, Karavitakis M, Stratakis S, Stylianou K, Tsitsimpikou C, et al. Current Concepts on the Reno-Protective Effects of Phosphodiesterase 5 Inhibitors in Acute Kidney Injury: Systematic Search and Review. Journal of Clinical Medicine. 2020; 9(5):1284. https://doi.org/10.3390/jcm9051284

Chicago/Turabian Style

Georgiadis, Georgios, Ioannis-Erineos Zisis, Anca Oana Docea, Konstantinos Tsarouhas, Irene Fragkiadoulaki, Charalampos Mavridis, Markos Karavitakis, Stavros Stratakis, Kostas Stylianou, Christina Tsitsimpikou, and et al. 2020. "Current Concepts on the Reno-Protective Effects of Phosphodiesterase 5 Inhibitors in Acute Kidney Injury: Systematic Search and Review" Journal of Clinical Medicine 9, no. 5: 1284. https://doi.org/10.3390/jcm9051284

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop