• No results found

University of Groningen Exploring Redox Biology in physiology and disease Koning, Anne

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Exploring Redox Biology in physiology and disease Koning, Anne"

Copied!
15
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Exploring Redox Biology in physiology and disease

Koning, Anne

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from

it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date:

2017

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Koning, A. (2017). Exploring Redox Biology in physiology and disease. Rijksuniversiteit Groningen.

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

Chapter 7

H

2

S treatment

in renal ischemia-reperfusion injury

and renal metabolism in rats

Anne M. Koning Harry van Goor Dane H. Hoeksma Andreas Pasch Henri G.D. Leuvenink

(3)

Abstract

Ischemia-reperfusion injury (IRI) is a major cause of renal transplant dysfunction. Hydrogen sulfide (H2S) can attenuate IRI in mice by metabolic suppression. In larger animals applicability

of hypometabolism is unlikely. However, in these species H2S may be protective independent

of hypometabolism, e.g. by its vasodilatory, anti-inflammatory and anti-oxidant properties. This study investigates the effects of sodium hydrosulfide (NaHS) and sodium thiosulfate (STS) in a model of unilateral renal IRI in rats and the potential of NaHS to induce hypometabolism in isolated rat kidneys.

In vivo, rats underwent 35 min of unilateral renal warm ischemia (WI) and received vehicle, NaHS (5.6 mg/kg/day) or STS (1g/kg/day) via intra-peritoneal injections twice daily from 48 h before WI until sacrifice at 24 or 96 h after reperfusion (n=6-7/group). Kidneys were studied for overall structural (PAS staining) and tubular epithelial damage (KIM-1), oxidative stress (MDA), inflammation (macrophages; ED1) and early fibrotic changes (αSMA). Ex vivo, kidneys were exposed to 100 μM or 1 mM NaHS for 30 min and oxygen consumption and renal function parameters, including sodium reabsorption were compared to controls (n=2-5/group).

In kidneys from the in vivo renal IRI experiment, histology (PAS staining) and immunohistochemistry (KIM-1, ED1, αSMA) showed no benefit from NaHS or STS treatment. Also, at the mRNA level no differences were found (KIM-1, αSMA). MDA levels were the same for all treatment groups. Ex vivo, H2S reduced oxygen consumption and sodium reabsorption

both by over 50% (p<0.05).

In conclusion, in vivo, NaHS and STS treatment by twice daily intra-peritoneal injections did not protect against renal IRI in rats. Ex vivo, NaHS-induced metabolic suppression in isolated rat kidneys, indicating a possible role for H2S as a treatment modality for kidney transplant

(4)

1 Introduction

In end-stage renal disease, patients rely on replacement therapy and transplantation is the treatment of choice. Unfortunately, the transplant process is inherently associated with ischemia-reperfusion injury (IRI), which is a major cause of kidney transplant dysfunction.(1)

Gaseous signalling molecule hydrogen sulfide (H2S) can attenuate renal IRI in mice by

metabolic suppression, through reversible inhibition of complex IV of the mitochondrial electron transport chain.(2,3) Although the applicability of H2S-induced hypometabolism in

larger animals is improbable, H2S is also considered to be protective independent of

metabolic suppression, e.g. by exerting vasodilatory, anti-inflammatory and anti-oxidant effects.(4–10) Accordingly, benefit of H2S treatment in the context of renal IRI has not only

been described in mice, but also in rats and pigs.(11–24)

In this study, apart from sulfide salt sodium hydrosulfide (NaHS), sodium thiosulfate is used as a treatment modality. Although thiosulfate (TS) is generally dismissed as a break down product of H2S, there is evidence to support its reconversion into H2S in vivo.(25–28) Also,

previous studies have shown TS to offer protection in various models of disease, including Angiotensin II-induced hypertensive renal damage.(29–33) This form of H2S treatment is of

particular interest as TS can safely be administered to humans. In fact, it is clinically applied, e.g. for the treatment of calciphylaxis in dialysis patients.(34)

The aim of this study is to investigate the effects of H2S treatment, in the form of NaHS and

STS, in a model of unilateral renal IRI in rats and to determine the potential of NaHS to induce a hypometabolic state in isolated rat kidneys. Hereby, we wish to contribute to the expansion of knowledge on the protective properties of H2S and their applicability in renal

transplantation.

2 Methods

2.1 Animals

Male Sprague Dawley (200-220 gram, in vivo experiment) and Fisher 344 (240-270 gram, ex vivo experiment) rats (Harlan, Zeist, the Netherlands) were housed at the animal research facility under standard conditions with a 12 h light-dark cycle and access to food and water ad libitum. Experimental procedures were in agreement with institutional and legislator regulations and approved by the local ethics committee for animal experiments.

2.2 Ischemia-reperfusion protocol and NaHS and STS treatment

Under general anesthesia (2% isoflurane in O2), a midline abdominal incision was made and

rats underwent 35 min of unilateral renal warm ischemia (WI) by clamping of the left renal artery and vein using non-traumatic clamps.

From 48 h before WI until sacrifice at 24 of 96 h after reperfusion rats received vehicle (0.9% NaCl, n=6/group), NaHS (5.6 mg/kg/day, Sigma, Zwijndrecht, the Netherlands, n=7/group) or STS (1 g/kg/day, Sigma, Zwijndrecht, the Netherlands, n=7/group) twice daily via intra-peritoneal injections.

(5)

When rats were sacrificed, the left kidney was perfused with 0.9% NaCl and harvested. Transverse slices were fixed in 4% paraformaldehyde and paraffin embedded for histological and immunohistochemical analysis or immediately snap frozen in liquid nitrogen and stored at -80 °C for molecular analysis.

2.3 Histology

Periodic acid-Schiff (PAS) staining was performed according to the standard protocol. Stained sections were subjected to blinded evaluation by light microscopy, resulting in a tubular damage score ranging from 1 to 6 (<5% to 75-100% damage).

2.4 Immunohistochemistry

Deparaffinised kidney sections were incubated overnight with 0.1 M Tris/HCl buffer (pH 9.0) at 80 °C for antigen retrieval. Endogenous peroxidase was blocked with 0.075% H2O2 in

phosphate buffered saline (PBS, pH 7.4) for 30 min. Section were incubated with primary antibodies for KIM-1 (rabbit anti-KIM-1 peptide 9, 1:400, gift V. Baily), macrophages (mouse anti-CD68 ED1, MCA341R AbD, 1:750, Serotec Ltd, Oxford, UK) or αSMA (mouse anti-SMA, clone 1A4 A2547, 1:10.000, Sigma, Zwijndrecht, the Netherlands) for 60 min at room temperature. Binding was detected by sequential incubation with peroxidase-labelled secondary and tertiary antibodies (Dakopatts, Glostrup, Denmark) for 30 min at room temperature. All antibodies were diluted with PBS supplemented with 1% bovine serum albumin. At the secondary and tertiary antibody dilutions 1% normal rat serum was added. Peroxidase activity was developed for 10 min using 3,3’-diaminobenzidine tetrachloride containing 0.03% H2O2. Counterstaining was performed using Mayer’s hematoxylin.

Kidney sections were scanned using an Aperio Scanscope slide scanner (Aperio Technologies, Vista, CA, USA). Tubular epithelial damage (KIM-1), macrophages (ED1) and early fibrotic changes (αSMA) were determined using the Aperio positive pixel analysis v9.1 algorithm and expressed as the number of positive pixels per μm of the cortical and corticomedullary border surface area. Analysis was performed in a blinded fashion.

2.5 Qualitative real-time polymerase chain reaction

Kidney tissue, consisting of cortex and medulla, was homogenized in lysis buffer and total RNA was extracted using the TRIZOL method (Invitrogen, Carlsbad, USA). RNA concentrations were measured by means of a nanodrop UV-detector (Nanodrop Technologies, Wilminton, DE). cDNA was synthesized using Superscript II with random hexamer primers (Invitrogen, Carlsbad, USA). Gene expression (Applied Biosystems, Foster City, CA, USA) was determined by qualitative real-time PCR (qRT-PCR) based on the Taqman methodology. HPRT was used as a housekeeping gene with the following primers (Integrated DNA Technologies) and probe (Eurogentec): Forward: 5’- GCC CTT GAC TAT AAT GAG CAC TTC A-3’, Reverse: 5’-TCT TTT AGG CTT TGT ACT TGG CTT TT-3’ and Probe: 6-FAM 5’-ATT TGA ATC ATG TTT GTG TCA TCA GCG AAA GTG-3’ TAMRA. Other primers were obtained from Applied Biosystems as Assays-on-Demand (AOD) gene expression products. The AOD IDs used were: Acta2 (αSMA) Rn01759928_g1 and Havcr1 (KIM-1) Rn00597703_m1. The

(6)

qRT-PCR reaction mixture contained 20 ng cDNA template and 5 μl qRT-PCR-master mix. Nuclease free water was added to a total volume of 10 μl. All assays were performed in triplicate. The thermal profile was 15 min at 95 °C, followed by 40 cycles of 15 sec at 95 °C and 1 min at 60 °C. The average Ct values for target genes were subtracted from the average housekeeping gene Ct values to yield the delta Ct. Results were expressed as 2-ΔCt.

2.6 Tissue malondialdehyde measurement

Malondialdehyde (MDA) was measured in kidney tissue after binding to thiobarbituric acid (TBA). Twenty μl of tissue homogenate was mixed with 2% sodium dodecyl sulfate and 5 mM butylated hydroxytoluene, followed by 400 μl 0.1 N HCL, 50 μl 10% phosphotungstic acid and 200 μl 0.7% TBA. The mixture was incubated for 1 h at 97 °C. Subsequently, 800 μl 1-butanol was added and the sample was centrifuged at 960 g. Fluorescence of 200 μl of the 1-butanol supernatant was measured at 480 nm excitation and 590 nm emission wavelengths. Results were expressed as μmol/g protein.

2.7 Isolated perfused kidney setup

Under general anesthesia (2% isoflurane in O2), through a midline abdominal incision, kidneys

of rats were isolated. Depending on the vascular anatomy, either the left or right kidney was chosen to be used for the experiment. Of this kidney, the artery, vein and ureter were cannulated. Subsequently, the kidney was placed in the isolated perfused kidney (IPK) setup and continuously perfused at a constant pressure of 100 mmHg with a warmed (37 °C) and oxygenated (95% O2 and 5% CO2 gas mixture) Krebs-Ringer Bicarbonate solution

complemented with albumin and creatinine, at a pH of 7.5 ± 0.05 and a PO2 ≈ 60 kPa. The

perfusion solution was composed of 118.6 mM NaCl, 4.7 mM KCl, 2.5 mM CaCl2, 1.2 mM

KH2PO4, 1.2 mM MgSO4, 25 mM NaHCO3, 6.1 mM glucose, 50 g/l albumin and 7.1 mM

creatinine. The pressure was regulated by means of a roller pump (Ismatec mv-ca/04, Ismatec, Glattbrugg, Switzerland) in combination with an electromechanical pressure transducer (Cobe; Arvada, CO), connected to a computer interface (LabView, National Instruments, Austin, TX).

Control kidneys (n=4) did not receive NaHS and were perfused for 60 min. Other kidneys were exposed to 100 μM (n=4) or 1 mM (n=5) NaHS for 30 min - from 30 min after the start of perfusion until the end of the experiment at 60 min. To assess reversibility of the effect of NaHS, a final set of kidneys (n=2) was also exposed to 1 mM NaHS for 30 min, yet from 15 min after the start of perfusion, followed by a wash out period of 30 min.

Oxygen consumption by the kidney was determined every 15 min, on the basis of the flow and the decline of the oxygen concentration of the perfusate passing through the kidney. The perfusate’s oxygen concentration was measured using flow-through oxygen sensors, incorporated in the afferent and efferent line of the IPK setup, in combination with optical oxygen meter Fibox 4 and PreSens Datamanager software (PreSens - Precision Sensing GmbH, Regensburg, Germany). At the same time points, afferent and efferent perfusate samples and a timed urine sample were taken. Urine samples were weighed and all samples were snap frozen and stored at -20 °C until analysis. Perfusate and urine levels of creatinine,

(7)

sodium and potassium were determined by standard assays from Roche on the Roche Modular (Roche Diagnostics GmbH, Mannheim, Germany) according to routine procedures in our clinical chemical laboratory.

2.6 Statistical analysis

Statistical analysis was performed with and graphs were drawn in GraphPad Prism (version 5.0, GraphPad Software, La Jolla, California, USA).

All data are presented as median (interquartile range (IQR)). Treatment groups were compared by means of the Kruskal-Wallis test for unrelated samples or the Friedman test for repeated measures. In both cases the test was complemented with Dunns post test to compare all pairs of columns. Statistical analysis was performed for groups with n ≥ 4. Values of P<0.05 were considered statistically significant.

3 Results

3.1 NaHS and STS treatment had no effect on renal ischemia-reperfusion injury in vivo Assessment of structural damage (PAS staining), tubular epithelial damage (KIM-1 mRNA and protein) and oxidative stress (lipid peroxidation, measured as tissue MDA) 24 hours after reperfusion, as well as influx of macrophages (ED1 protein) and early fibrotic changes (αSMA mRNA and protein) 96 hours after reperfusion revealed no differences between controls and NaHS and STS treated rats (Fig. 1-4).

Figure 1: IRI-induced structural damage

24 hours after reperfusion of the kidney Periodic acid-Schiff staining (A), KIM-1 protein (B), and KIM-1 mRNA (C) levels showed no differences in tubular damage between controls (n=6) and NaHS (n=7) or STS (n=7) treated rats.

Data are presented as median (IQR)

(8)

Figure 4: IRI-induced early fibrotic changes

7

96 hours after reperfusion αSMA mRNA (A) and protein (B) levels showed no differences in early fibrotic changes between controls (n=6) and NaHS (n=7) or STS (n=7) treated rats.

Data are presented as median (IQR).

NaHS; sodium hydrosulfide, STS; sodium thiosulfate, IRI; ischemia-reperfusion injury

24 hours after reperfusion of the kidney there were no differences in lipid peroxidation - as determined by tissue MDA levels - between controls (n=6) and rats treated with NaHS (n=7) or STS (n=7).

Data are presented as median (IQR).

NaHS; sodium hydrosulfide, STS; sodium thiosulfate, IRI; ischemia-reperfusion injury, MDA; malondialdehyde

96 hours after reperfusion of the kidney ED1 staining revealed no differences in influx of macrophages between controls (n=6) and NaHS (n=7) or STS (n=7) treated rats. Data are presented as median (IQR).

IRI; ischemia-reperfusion injury, NaHS; sodium hydrosulfide, STS; sodium thiosulfate

(9)

3.2 NaHS-induced hypometabolism ex vivo

In the IPK setup NaHS was found to decrease renal oxygen consumption in a dose dependent manner (Fig. 5, A-C). Data from two kidneys suggest reversibility of this effect (Fig. 5, D). Furthermore, 1 mM NaHS was found to significantly decrease the fractional reabsorption of sodium by the kidney (P<0.01), whereas other renal function parameters (urine production, creatinine clearance and excretion of potassium) were unaffected (Fig. 6).

Figure 5: Oxygen consumption by the isolated perfused kidney

NaHS decreased renal oxygen consumption in a dose dependent manner (A-C, n=4 for controls, n=4 for kidneys exposed to 100 μM NaHS and n=5 for kidneys exposed to 1 mM NaHS). Data from two kidneys suggest this effect to be reversible (D). Data are presented as median (IQR), **P<0.01. NaHS; sodium hydrosulfide, STS; sodium thiosulfate

(10)

Figure 6: Function of the isolated perfused kidney

After 30 min NaHS did not significantly change urine production (A) or creatinine clearance (B). 1 mM NaHS significantly decreased sodium reabsorption (A). Potassium excretion was not changed by NaHS (B). n=4 for controls, n=4 for kidneys exposed to 100 μM NaHS, n=5 for kidneys exposed to 1 mM NaHS, data are presented as median (IQR), **P<0.01.

NaHS; sodium hydrosulfide, STS; sodium thiosulfate

4 Discussion

In the context of renal IRI in rats, this study shows no benefit of NaHS and STS treatment through twice daily intra-peritoneal injections. Ex vivo, however, NaHS did significantly supress metabolism in the isolated rat kidney, as evidenced by the decrease of renal oxygen consumption and tubular sodium reabsorption.

The treatment regimens applied in our in vivo IRI experiment have previously been shown by our group to protect against Angiotensin II-induced hypertensive renal damage.(30) The absence of any sign of protection in our model was unexpected and, at first glance, does not comply with existing evidence on H2S treatment in renal IRI and transplantation. Indeed, NaHS

has repeatedly been shown to protect the kidney from IRI, even in rats and when administered through intra-peritoneal injections.(11,13,17,18,21,24) However, notably, in all of these studies H2S treatment was applied close to or during reperfusion, whereas in our case the

time between restoration of blood flow and administration of treatment was much longer.(11,13,17,18,21,24) Thus, although in the context of cardiac IRI positive results of H2S

pre-treatment have been described, application of therapy close to or even during the reperfusion phase may be the key to protection of rat kidneys in vivo.(35,36) This may be

(11)

related to the unique way in which renal blood flow is regulated. Indeed, while in most organs vascular tone is adapted in response to tissue oxygenation, in the kidney blood flow is relatively unresponsive to hypoxia or hyperoxia and controlled by mechanisms that regulate renal function, i.e. glomerular filtration and tubular reabsorption.(37) Under normal conditions, arterial-to-venous (AV) oxygen shunting helps to retain renal tissue oxygenation within a physiological range. However, the same mechanism causes the kidney to be vulnerable in case of hypoxia. Conversely, during reperfusion AV oxygen shunting is likely to be inadequate. As a result, the kidney is susceptible to hyperoxia and consequent oxidation.(37)

Aside from the warm ischemia and reperfusion that occur in vivo, ex vivo preservation of the kidney is another phase of the transplant process in which renal damage arises. Traditionally, during this phase the evident mismatch between oxygen supply and demand is confined by suppression of the kidney’s metabolism through the induction of hypothermia. However, hypothermia itself is a cause of renal damage and, accordingly, prolonged cold storage significantly contributes to kidney transplant dysfunction.(38) H2S therapy possibly

represents an opportunity to accomplish metabolic suppression in the kidney without the detrimental effects associated with hypothermia. While in mice H2S can safely induce a

reversible hypometabolic state, studies in larger animals, including pigs and sheep, have questioned the applicability of this phenomenon in large mammals, including humans. As previously described by Haouzi et al., differences in mechanisms controlling resting metabolic rate in relation to body mass may explain discrepancies in the response to H2S, dosed

according to body mass. Possibly, much higher concentrations of H2S would induce metabolic

suppression in larger animals, however, considering H2S toxicity at higher levels, this would

not be without side-effects.(39) Even though a therapeutic dose of H2S may not affect whole

body metabolism in larger animals as it does in mice, our data show that NaHS is certainly able to decrease metabolism in isolated rat kidneys. STS was not applied in the IPK setup because previous research suggests that the reconversion of TS into H2S predominantly

occurs in the liver and under hypoxic circumstances.(28) The unlikeliness of TS conversion in the IPKis supported by an earlier IPK experiment by our group in which STS, in contrast to NaHS, was found to have no effect on intra-renal pressure.(30) One might argue that 1 mM NaHS - the dose that resulted in the most significant suppression of renal metabolism - is high and potentially toxic. However, cell damage can not have caused the observed decrease of renal oxygen consumption as this was found to be reversible. Our finding indicates a possible role for H2S therapy in the kidney transplant preservation phase and warrants confirmation in

porcine and eventually human kidneys. While our ex vivo experiment was not followed by transplantation of the kidneys and, therefore, outcome could not be assessed, previous studies by Lobb et al. have shown beneficial effects of adding 150 μM NaHS to University of Wisconsin preservation solution.(18,23) NaHS treatment was shown to increase graft function, attenuate necrosis, apoptosis and inflammation and increase recipient survival. However, it is unclear from these studies whether the improved outcome is the result of H2S-induced

hypometabolism, other protective properties of H2S or both. Notably, all kidneys were stored

at 4 °C.(18,23) Thus, whether and at which dose H2S therapy actually permits normothermic

(12)

Apart from the caveats mentioned elsewhere in this discussion, the limitations of our study include the use of different strains of rats for the in vivo and the ex vivo experiment. We can not be sure NaHS would similarly have induced a hypometabolic state in the kidneys from Sprague Dawley rats. At the same time, NaHS and STS treatment, as applied in this study, may have shown protection against renal IRI in Fisher 344 rats. Furthermore, the unilateral IRI model, chosen to be used for animal welfare reasons, did not permit assessment of renal function following IR. Therefore, any functional benefit of treatment that may have been present could not be detected.

In conclusion, NaHS and STS therapy by twice daily intra-peritoneal injections did not protect against renal IRI in rats. Possibly, application of treatment close to or even during the reperfusion phase is essential to in vivo protection of the kidney. Ex vivo, NaHS induced metabolic suppression in isolated rat kidneys. This points towards a possible role for H2S as a

treatment modality for kidney transplant protection during the preservation stage.

Funding

This work was supported by grants from the Dutch Kidney Foundation (IP13-114, to HvG) and the Groningen University Institute for Drug Exploration (to AMK).

Acknowledgements

The authors would like to express their gratitude to Pieter Klok, Annemiek Schaafsma, Sander Twickler, Petra Ottens, Susanne Veldhuis, Sippie Huitema and Marian Bulthuis for their excellent technical assistance.

Conflicts of interest None declared.

(13)

References

1. Land W, Messmer K, Events E. The Impact of Ischemia/Reperfusion Injury on Specific and Non-Specific, Early and Late Chronic Events After Organ Transplantation. Transplant Rev. 1996 Apr;10(2):108–27.

2. Blackstone E, Morrison M, Roth MB. H2S Induces a Suspended Animation–Like State in Mice. Science. 2005 Apr 22;308(5721):518–518.

3. Bos EM, Leuvenink HGD, Snijder PM, Kloosterhuis NJ, Hillebrands J-L, Leemans JC, et al. Hydrogen Sulfide-Induced Hypometabolism Prevents Renal Ischemia/Reperfusion Injury. J Am Soc Nephrol. 2009 Jul 23;20(9):1901–5.

4. Haouzi P, Bell HJ, Notet V, Bihain B. Comparison of the metabolic and ventilatory response to hypoxia and H2S in unsedated mice and rats. Respir Physiol Neurobiol. 2009 Jul 31;167(3):316–22.

5. Aslami H, Heinen A, Roelofs JJTH, Zuurbier CJ, Schultz MJ, Juffermans NP. Suspended animation inducer hydrogen sulfide is protective in an in vivo model of ventilator-induced lung injury. Intensive Care Med. 2010 Nov;36(11):1946–52.

6. Seitz DH, Fröba JS, Niesler U, Palmer A, Veltkamp HA, Braumüller ST, et al. Inhaled hydrogen sulfide induces suspended animation, but does not alter the inflammatory response after blunt chest trauma. Shock Augusta Ga. 2012 Feb;37(2):197–204.

7. Aslami H, Beurskens CJP, de Beer FM, Kuipers MT, Roelofs JJTH, Hegeman MA, et al. A short course of infusion of a hydrogen sulfide-donor attenuates endotoxemia induced organ injury via stimulation of anti-inflammatory pathways, with no additional protection from prolonged infusion. Cytokine. 2013 Feb;61(2):614–21.

8. Li J, Zhang G, Cai S, Redington AN. Effect of inhaled hydrogen sulfide on metabolic responses in anesthetized, paralyzed, and mechanically ventilated piglets. Pediatr Crit Care Med J Soc Crit Care Med World Fed Pediatr Intensive Crit Care Soc. 2008 Jan;9(1):110–2.

9. Drabek T, Kochanek PM, Stezoski J, Wu X, Bayir H, Morhard RC, et al. Intravenous hydrogen sulfide does not induce hypothermia or improve survival from hemorrhagic shock in pigs. Shock Augusta Ga. 2011 Jan;35(1):67–73.

10. Szabo C. Medicinal Chemistry and Therapeutic Applications of the Gasotransmitters NO, CO, and H2S and their Prodrugs. In: Burger’s Medicinal Chemistry and Drug Discovery. Seventh Edition. Hoboken, NJ, USA: John Wiley & Sons, Inc.; 2010. p. 265–368.

11. Tripatara P, Patel NSA, Collino M, Gallicchio M, Kieswich J, Castiglia S, et al. Generation of endogenous hydrogen sulfide by cystathionine gamma-lyase limits renal ischemia/reperfusion injury and dysfunction. Lab Investig J Tech Methods Pathol. 2008 Oct;88(10):1038–48.

12. Tripatara P, Patel NSA, Brancaleone V, Renshaw D, Rocha J, Sepodes B, et al. Characterisation of cystathionine gamma-lyase/hydrogen sulphide pathway in ischaemia/reperfusion injury of the mouse kidney: an in vivo study. Eur J Pharmacol. 2009 Mar 15;606(1–3):205–9.

13. Xu Z, Prathapasinghe G, Wu N, Hwang S-Y, Siow YL, O K. Ischemia-reperfusion reduces cystathionine-beta-synthase-mediated hydrogen sulfide generation in the kidney. Am J Physiol Renal Physiol. 2009 Jul;297(1):F27-35.

14. Hosgood SA, Nicholson ML. Hydrogen sulphide ameliorates ischaemia-reperfusion injury in an experimental model of non-heart-beating donor kidney transplantation. Br J Surg. 2010 Feb;97(2):202–9.

15. Simon F, Scheuerle A, Gröger M, Stahl B, Wachter U, Vogt J, et al. Effects of intravenous sulfide during porcine aortic occlusion-induced kidney ischemia/reperfusion injury. Shock Augusta Ga. 2011 Feb;35(2):156– 63.

16. Hunter JP, Hosgood SA, Patel M, Rose R, Read K, Nicholson ML. Effects of hydrogen sulphide in an experimental model of renal ischaemia-reperfusion injury. Br J Surg. 2012 Dec;99(12):1665–71.

17. Zhu JXG, Kalbfleisch M, Yang YX, Bihari R, Lobb I, Davison M, et al. Detrimental effects of prolonged warm renal ischaemia-reperfusion injury are abrogated by supplemental hydrogen sulphide: an analysis using real-time intravital microscopy and polymerase chain reaction. BJU Int. 2012 Dec;110(11 Pt C):E1218-1227. 18. Lobb I, Mok A, Lan Z, Liu W, Garcia B, Sener A. Supplemental hydrogen sulphide protects transplant kidney

function and prolongs recipient survival after prolonged cold ischaemia-reperfusion injury by mitigating renal graft apoptosis and inflammation. BJU Int. 2012 Dec;110(11 Pt C):E1187-1195.

(14)

19. Bos EM, Wang R, Snijder PM, Boersema M, Damman J, Fu M, et al. Cystathionine γ-lyase protects against renal ischemia/reperfusion by modulating oxidative stress. J Am Soc Nephrol JASN. 2013 Apr;24(5):759-70. 20. McCook O, Radermacher P, Volani C, Asfar P, Ignatius A, Kemmler J, et al. H2S during circulatory shock:

Some unresolved questions. Nitric Oxide Biol Chem Off J Nitric Oxide Soc. 2014 Mar 18;

21. Lobb I, Zhu J, Liu W, Haig A, Lan Z, Sener A. Hydrogen sulfide treatment ameliorates long-term renal dysfunction resulting from prolonged warm renal ischemia-reperfusion injury. Can Urol Assoc J J Assoc Urol Can. 2014 May;8(5–6):E413-418.

22. King AL, Polhemus DJ, Bhushan S, Otsuka H, Kondo K, Nicholson CK, et al. Hydrogen sulfide cytoprotective signaling is endothelial nitric oxide synthase-nitric oxide dependent. Proc Natl Acad Sci U S A. 2014 Feb 10; 23. Lobb I, Davison M, Carter D, Liu W, Haig A, Gunaratnam L, et al. Hydrogen Sulfide Treatment Mitigates Renal

Allograft Ischemia-Reperfusion Injury during Cold Storage and Improves Early Transplant Kidney Function and Survival Following Allogeneic Renal Transplantation. J Urol. 2015 Dec;194(6):1806–15.

24. Azizi F, Seifi B, Kadkhodaee M, Ahghari P. Administration of hydrogen sulfide protects ischemia reperfusion-induced acute kidney injury by reducing the oxidative stress. Ir J Med Sci. 2016 Aug;185(3):649–54. 25. Mikami Y, Shibuya N, Kimura Y, Nagahara N, Ogasawara Y, Kimura H. Thioredoxin and dihydrolipoic acid are

required for 3-mercaptopyruvate sulfurtransferase to produce hydrogen sulfide. Biochem J. 2011 Nov 1;439(3):479–85.

26. Villarejo M, Westley J. Mechanism of Rhodanese catalysis of thiosulfate-lipoate oxidation-reduction. J Biol Chem. 1963 Dec;238:4016–20.

27. Koj A, Frendo J, Janik Z. (35S)thiosulphate oxidation by rat liver mitochondria in the presence of glutathione. Biochem J. 1967 Jun;103(3):791–5.

28. Olson KR, Deleon ER, Gao Y, Hurley K, Sadauskas V, Batz C, et al. Thiosulfate: a readily accessible source of hydrogen sulfide in oxygen sensing. Am J Physiol Regul Integr Comp Physiol. 2013 Sep 15;305(6):R592-603. 29. Sen U, Vacek TP, Hughes WM, Kumar M, Moshal KS, Tyagi N, et al. Cardioprotective Role of Sodium Thiosulfate on Chronic Heart Failure by Modulating Endogenous H&lt;sub&gt;2&lt;/sub&gt;S Generation. Pharmacology. 2008;82(3):201–13.

30. Snijder PM, Frenay A-RS, Koning AM, Bachtler M, Pasch A, Kwakernaak AJ, et al. Sodium thiosulfate attenuates angiotensin II-induced hypertension, proteinuria and renal damage. Nitric Oxide Biol Chem Off J Nitric Oxide Soc. 2014 Nov 15;42:87–98.

31. Snijder PM, Frenay AS, de Boer RA, Pasch A, Hillebrands J, Leuvenink HGD, et al. Exogenous administration of thiosulfate, a donor of hydrogen sulfide, attenuates Angiotensin II-induced hypertensive heart disease in rats. Br J Pharmacol. 2015 Mar;172(6):1494-504.

32. Sakaguchi M, Marutani E, Shin H-S, Chen W, Hanaoka K, Xian M, et al. Sodium Thiosulfate Attenuates Acute Lung Injury in Mice. Anesthesiology. 2014 Sep 25;121:1248-1257.

33. Lee M, McGeer EG, McGeer PL. Sodium thiosulfate attenuates glial-mediated neuroinflammation in degenerative neurological diseases. J Neuroinflammation. 2016 Feb 8;13:32.

34. Cicone JS, Petronis JB, Embert CD, Spector DA. Successful treatment of calciphylaxis with intravenous sodium thiosulfate. Am J Kidney Dis Off J Natl Kidney Found. 2004 Jun;43(6):1104–8.

35. Andreadou I, Iliodromitis EK, Rassaf T, Schulz R, Papapetropoulos A, Ferdinandy P. The role of gasotransmitters NO, H2 S, CO in myocardial ischemia/reperfusion injury and cardioprotection by preconditioning, postconditioning, and remote conditioning. Br J Pharmacol. 2015 Mar;172(6):1587-606. 36. Li C, Hu M, Wang Y, Lu H, Deng J, Yan X. Hydrogen sulfide preconditioning protects against myocardial

ischemia/reperfusion injury in rats through inhibition of endo/sarcoplasmic reticulum stress. Int J Clin Exp Pathol. 2015;8(7):7740–51.

37. Evans RG, Gardiner BS, Smith DW, O’Connor PM. Intrarenal oxygenation: unique challenges and the biophysical basis of homeostasis. Am J Physiol Renal Physiol. 2008 Nov;295(5):F1259-1270.

38. Salahudeen AK. Cold ischemic injury of transplanted kidneys: new insights from experimental studies. AJP Ren Physiol. 2004 Apr 27;287(2):F181–7.

39. Haouzi P, Notet V, Chenuel B, Chalon B, Sponne I, Ogier V, et al. H2S induced hypometabolism in mice is missing in sedated sheep. Respir Physiol Neurobiol. 2008 Jan 1;160(1):109–15.

(15)

Referenties

GERELATEERDE DOCUMENTEN

In the following sections, we will therefore discuss major metabolic perturbations in HF and the potential utility of biomarkers and molecular imaging techniques to

Systemic oxidative stress can be measured as the depletion of the free thiol pool in serum.(9) In contrast to the intracellular pool, which mainly consists of low molecular weight

Both treatments were shown to decrease hypertension, proteinuria and renal dysfunction, as well as tubular damage, oxidative stress, influx of inflammatory cells and fibrosis..

Univariable analysis showed that graft survival is improved in kidney transplant recipients that homozygously carry the minor allele of rs11203172.. Furthermore, no PNF was seen

After three weeks of Ang II infusion, renal mRNA levels of (A) CSE, (B) CBS and (C) 3- MST are decreased. Treatment with NaHS and STS partially prevented this down

Therefore, the aim of the present study was to determine the association of urinary thiosulfate and sulfate excretion with risk of CV events and all-cause mortality in a large

In CHF patients, plasma sulfate concentration (A) is significantly higher, whereas 24-h urinary sulfate excretion, fractional excretion of sulfate and sulfate clearance (B-D)

With the exception of 24-h urinary excretion of nitrate, these differences remained significant after additional adjustment for creatinine clearance (all P&lt;0.001), indicating