Next Article in Journal
Intermolecular Interactions in 3-Aminopropyltrimethoxysilane, N-Methyl-3-aminopropyltrimethoxysilane and 3-Aminopropyltriethoxysilane: Insights from Computational Spectroscopy
Previous Article in Journal
Boosting Mitochondrial Biogenesis Diminishes Foam Cell Formation in the Post-Stroke Brain
Previous Article in Special Issue
Role of Tyrosine Nitrosylation in Stress-Induced Major Depressive Disorder: Mechanisms and Implications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Immunolocalization of Sphingolipid Catabolism Enzymes along the Nephron: Novel Early Urinary Biomarkers of Renal Damage

by
Martha Franco
1,†,
Agustina Cano-Martínez
2,†,
María del Pilar Ramos-Godínez
3,
Rebeca López-Marure
2,
Luis Donis-Maturano
4,
José Santamaría Sosa
1 and
Rocio Bautista-Pérez
5,*
1
Department of Cardio-Renal Pathophysiology, Instituto Nacional de Cardiología “Ignacio Chávez”, Mexico City 14080, Mexico
2
Department of Physiology, Instituto Nacional de Cardiología “Ignacio Chávez”, Mexico City 14080, Mexico
3
Department of Electron Microscopy, Instituto Nacional de Cancerología, Mexico City 14080, Mexico
4
Faculty of Higher Studies Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico
5
Department of Molecular Biology, Instituto Nacional de Cardiología “Ignacio Chávez”, Mexico City 14080, Mexico
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(23), 16633; https://doi.org/10.3390/ijms242316633
Submission received: 13 October 2023 / Revised: 14 November 2023 / Accepted: 20 November 2023 / Published: 23 November 2023

Abstract

:
The objective of this study was to investigate whether the activity of enzymes involved in sphingolipid catabolism could be biomarkers to predict early renal damage in streptozotocin (STZ)-induced diabetic rats and Angiotensin II (Ang II)-induced hypertension rats. Diabetic and hypertensive rats had no changes in plasma creatinine concentration. However, transmission electron microscopy (TEM) analysis showed slight ultrastructural changes in the glomeruli and tubular epithelial cells from diabetic and hypertensive rats. Our results show that the acid sphingomyelinase (aSMase) and neutral sphingomyelinase (nSMase) activity increased in the urine of diabetic rats and decreased in hypertensive rats. Only neutral ceramidase (nCDase) activity increased in the urine of diabetic rats. Furthermore, the immunofluorescence demonstrated positive staining for the nSMase, nCDase, and sphingosine kinase (SphK1) in glomerular mesangial cells, proximal tubule, ascending thin limb of the loop of Henle, thick ascending limb of Henle’s loop, and principal cells of the collecting duct in the kidney. In conclusion, our results suggest that aSMase and nCDase activity in urine could be a novel predictor of early slight ultrastructural changes in the nephron, aSMase and nCDase as glomerular injury biomarkers, and nSMase as a tubular injury biomarker in diabetic and hypertensive rats.

Graphical Abstract

1. Introduction

In metabolic, cardiovascular, and renal diseases, the cellular content of the sphingolipids and their metabolites are regulated by enzymes involved in the anabolism (glucosylceramide synthase, sphingomyelin synthase, ceramide synthase) and catabolism (SMase, CDase, SphK, and sphingosine-1-phosphate lyase). In the kidney and other organs, the expression or activity of these enzymes changes in pathophysiological conditions [1,2,3,4,5,6,7,8,9]. These findings are important since the failure of one organ can induce pathological changes in another organ, or ambos may present simultaneous dysfunction (e.g., Kidney–heart, kidney–liver) [10,11].
In the kidney, there is nSMase expression in principal and intercalated cells of the collecting duct, and its activity is stimulated by fluid shear stress [12]. The nCDase is localizing at the apical membrane of tubules [13]. SphK1 is expressed in the cortex, followed by the medulla and papilla [14,15]. In addition, SphK1 and SphK2 are expressed throughout development, predominantly in the metanephric mesenchyme [16].
During development and aging, the nSMase and CDase activity increases in the kidney [17]. In the renal cortical of serum nephritis rats and rats intoxicated with carbon tetrachloride, SMase activity increased. In contrast, after renal injury by ischemia, ischemic/reperfusion, or myohemoglobinuric, SMase activity decreased [18,19]. After the induction of diabetes, nCDase and SphK1 activity increased, and only nCDase activity decreased after 28 days [20]. In the kidneys of diabetic rats, nSMase activity increased, and nCDase activity decreased, whereas in the kidney of hypertensive rats, the activity of both enzymes increased [9].
These enzymes, sphingolipids, and their metabolites can be detected in blood or urine, so they have been proposed as biomarkers of kidney damage. In this regard, in the plasma of patients or preclinical animal models, the enzymatic activity, the concentration of sphingolipids, and their metabolites have already been determined [6,9,21,22,23]. Interestingly, in the urine of diabetic patients, glycosphingolipids, ceramides, and hexose ceramide levels increase [24,25,26]. In patients with severe COVID-19, increased urinary levels of sphingomyelin, ceramides, sphingosine, and glycerophospholipids might result in renal damage [27]. Also, aSMase and nSMase were purified from human urine [28,29].
These findings demonstrate that sphingolipids, their metabolites, and enzymes involved in sphingolipid metabolism can be detected in the urine and are attractive candidates as biomarkers of renal damage.
The objective of this study was to investigate whether the activity of enzymes involved in sphingolipid catabolism could be biomarkers to predict early renal damage in STZ-induced diabetic rats and Ang II-induced hypertension rats.

2. Results

2.1. General Characteristics of Diabetic and Hypertensive Rats

The blood glucose concentration was higher in diabetic rats compared with control rats (589 ± 34 vs. 113 ± 2.5 mg/dL). The systolic blood pressure (SBP) was elevated in hypertensive rats compared with normotensive rats on day 14 (213.4 ± 4 vs. 125.9 ± 2.5 mm Hg). The plasma creatinine concentration showed no changes in diabetic rats compared with control rats (0.56 ± 0.06 mg/dL vs. 0.5059 ± 0.07 mg/dL); the hypertensive rats also showed no changes (0.471 ± 0.1496 mg/dL vs. 0.4664 ± 0.15 mg/dL).

2.2. Transmission Electron Microscopy (TEM)

TEM analysis showed slight ultrastructural changes in the kidney from diabetic and hypertension rats: disrupted glomerular ultrastructure (detached endothelial layer in the glomerular capillary, glomerular basement membrane thickening, effacement of podocytes, and fusion of podocytes foot processes; the tubular epithelial cells show decreased and irregular microvilli, and the mitochondria are round with loss of matrix density (Figure 1).

2.3. Sphingomielinase and Ceramidase Activity in Urine of Diabetic and Hypertensive Rats

In the urine of diabetic rats, the aSMase and nSMase activity was increased, while the aCDase activity did not show changes, and nCDase activity also increased compared with the control group (Figure 2).
In the urine of hypertensive rats, the aSMase and nSMase activity decreased, while the aCDase and nCDase activity did not show changes compared with the normotensive group (Figure 3). Moreover, the nSMase activity was lower compared to aSMase activity in both groups.

2.4. Expression and Localization of Enzymes Involved in Sphingolipid Catabolism along the Nephron

The functional unit of the kidney is the nephron; each nephron consists of glomerulus, proximal tubule (convoluted and straight segments), loop of Henle, distal tubule, the connecting tubule, and collecting duct. The glomerulus is formed of three types of cells: endothelial cells, podocytes, and mesangial cells. In the kidney of control, diabetic, and hypertensive rats, we observed expression of all markers. However, we only show the photographs of the control group. As shown in Figure 4, Thy-1 is a mesangial cell marker, and these cells were also positive for nSMase, nCDase, and SphK1.
The proximal tubule is responsible for 70% of water reabsorption and descending thin limb of Henle’s loop by 20%; the aquaporin-1 (AQP1) facilitates water transport across the membranes’ apical and basolateral membranes in these two nephron segments. In this study, AQP1 was used as a marker of the proximal tubule. As shown in Figure 5, nSMase, nCDase, and SphK1 were also immunolocalized to proximal tubule cells.
The ascending thin limb expressed the Cl channel ClCK1 along the entire lengths and was used as a marker of this nephron segment. As shown in Figure 6, nSMase, nCDase, and SphK1 were also immunolocalized to the ascending thin limb of the loop of Henle.
The Tamm–Horsfall (THP) or uromodulin protein is expressed in the ascending thick limb of the loop of Henle, extending through the whole outer medulla and cortex. As shown in Figure 7, THP colocalized with nSMase, nCDase, and SphK1 in the ascending thick limb of the loop of Henle.
The distal convoluted tubule, the connective tubule, and the collecting duct form the distal part of the nephron and oversee the final tuning of renal excretion.
In the collecting duct, two functionally and morphologically distinct cells are present: principal cells and intercalated cells. The primary mechanism by which water is reabsorbed in the principal cell is through aquaporin 2 (AQP2). As shown in Figure 8, AQP2 colocalized with nSMase, nCDase, and SphK1 in the principal cells.

3. Discussion

In this study, we evaluated whether the activity of enzymes involved in sphingolipid catabolism could be biomarkers to predict early renal damage in diabetic and hypertensive rats. Diabetic and hypertensive rats had no changes in plasma creatinine concentration. However, TEM analysis showed slight ultrastructural changes in the glomeruli and tubular epithelial cells.
Our results agree with studies that using serum creatinine concentration-based criteria does not correlate well with the severity of renal injury. The serum creatinine can increase in the absence of glomerular or tubular injury and can be unchanged under conditions of glomerular or tubular injury [30,31]. Thus, it is necessary to find specific and sensitive biomarkers that reflect a pathophysiological process, and these can easily be detected in the plasma or urine.
Urine is an ideal biomarker source for clinical metabolomics or proteomics studies [32]. The concentration of some sphingolipid metabolites increases in urine from diabetic patients and patients with severe COVID-19 [24,25,26,27]. Interestingly, aSMase and nSMase were purified from human urine [28,29]. Thus, the determination of urinary enzymes may be valuable for the diagnosis of renal damage [33].
We recently reported that the aSMase and nCDase activity increased in the plasma of diabetic and hypertensive rats [9]. This increase is possibly due to fact that interleukin 1β (IL-1β) and interferon-gamma (IFN-gamma) stimulate the secretion of aSMase in endothelial cells [34,35,36]. Thus, our results suggest that aSMase and nCDase are plasma proteins that cross the slightly damaged glomerular barrier and increase their elimination in the urine of diabetic rats, which explains their increase in activity.
On the other hand, some studies reported the localization of murine and human nSMase in the nucleus, endoplasmic reticulum, Golgi apparatus, lysosomes, mitochondria, and plasma membrane [37,38,39]. In this study, we show nSMase expression along the nephron, proximal tubule cell with decreased and irregular microvilli, and round mitochondria with a loss of matrix density, which may contribute to the release of nSMase in the urine of diabetic rats.
In addition, an imbalance in the levels of sphingolipids or their metabolites results in changes in the structure and function of mitochondria, leading to the overproduction of reactive oxygen and nitrogen species [40]. The reactive oxygen/nitrogen species can activate aSMase and nSMase [41,42]. Also, the depletion of reduced glutathione and the concentration of magnesium (Mg2+) contribute to the regulation of nSMase activity [43,44]. Fluid shear stress also induces nSMase activity in principal cells and intercalated cells of the kidney [12]. These mechanisms explain why it increases the nSMase activity in the urine of diabetic rats. In contrast, aSMase activity decreased, while nCDase activity showed no changes in the urine of hypertensive rats. As mentioned above, the aSMase and nCDase are plasma proteins but do not cross the glomerular barrier because Ang II induces vasoconstriction and decreases the kidney glomerular filtration rate, as we have reported in previous studies [45,46].
On the other hand, the nSMase present in the urine of hypertensive rats may originate from kidney tissue; however, its activity decreases depending on the concentration of Mg [44]. Also, in renal mesangial cell cultures, the inflammatory cytokines, growth factors, and vasoconstrictor peptides regulate the SMase and CDase activity [47,48,49]. The bradykinin inhibits SMase activity in the cortical-collecting duct cells line [50].
In addition, it has been reported that dysregulation of sphingolipid metabolism in podocytes can compromise the proper functioning of the glomerular basement membrane, leading to proteinuria, glomerular fibrosis, and glomerulosclerosis [51]. Sphingosine-1-phosphate induces tubule interstitial renal inflammation and fibrosis in diabetic nephropathy [52]. Interestingly, we reported an S1P content increase in the kidney of diabetic and hypertensive rats [9].
Thus, sphingolipid catabolic enzymes could be biomarkers to predict early kidney damage in pathology such as diabetes and hypertension. The aSMase and nCDase can be considered as glomerular injury biomarkers and nSMase as a tubular injury biomarker.

4. Materials and Methods

4.1. Animal Models

Animal procedures were performed in accordance with the Mexican Federal Regulation for Animal Experimentation and Care (NOM-062-ZOO-2001), and protocols were approved by the Investigation Committee of the Instituto Nacional de Cardiología “Ignacio Chávez” (INC-CICUAL/012/2019, INC-CICUAL/005/2020). All rats had free access to water and standard chow diet.

4.1.1. Streptozotocin-Induced Diabetic Rats

Male Wistar rats (300–350 g) were divided into two groups (n = 10 each): (1) control (injected with citrate buffer) and (2) streptozotocin (STZ)-induced diabetic rats (STZ, 65 mg/kg body weight). Blood glucose levels were measured at 24 h after STZ administration. The animals with blood glucose 300 mg/dL (18 mmol/L) were used as diabetic rats and maintained for 30 days [9].

4.1.2. Angiotensin II-Induced Hypertension Rats

Male Wistar rats weighing 350–360 g were separated into two groups: normotensive (Sham, n = 10) and hypertensive (Ang II, n = 10) rats. Ang II (Sigma, St. Louis, MO, USA) was infused via subcutaneous osmotic minipumps (Alzet 2002; Alza, Palo Alto, CA, USA), implanted under isoflurane anesthesia. Minipumps delivered Ang II at a rate of 435 ng/kg/min. Experiments were performed 13–14 days after the beginning of Ang II infusion, as previously described [9,46].

4.1.3. Sample Collection and Tissue Preparation

At 30 days after STZ administration or 2 weeks after the infusion of Ang II, urine samples were collected over a 24 h period and stored at −20 °C to assay for proteinuria by Bradford assay (Bio-Rad, Hercules, CA, USA), as well as to determine the activity of nSMase, acid SMase (aSMase), nCDase, and acid CDase (aCDase).
The animals were anesthetized with sodium pentobarbital (30 mg/kg, i.p.); blood samples were taken and collected in heparinized tubes, and after centrifugation (10,000× g for 5 min), plasma was obtained. Plasma samples were stored at −20 °C and used for creatinine determination (Quanti Chrom™ creatinine assay kit, BioAssay Systems, Hayward, CA, USA) [53].
After decapsulation, the kidneys were perfused with ice-cold phosphate-buffered solution (PBS) pH 7.4, to remove red blood cells and clots. After perfusion with PBS, left kidney was sectioned in small pieces and embedded in Epon 812 for transmission electron microscopy (TEM). The right kidney was fixed in 4% paraformaldehyde-PBS, embedded in paraffin, and mounted in coverslips for immunostaining.

4.1.4. Transmission Electron Microscopy (TEM)

The kidneys were excised and fixed in PBS with 2.5% glutaraldehyde plus and 2.5% paraformaldehyde. The tissues were post-fixed with 1% osmium tetroxide (OsO4). Then, the fixed renal tissue fragments were dehydrated in an ascending series of ethanol and embedded in Epon 812 epoxy resin (sigma). Subsequently, the tissue was treated with uranyl acetate and incubated in a lead citrate solution. Samples were observed in a JEOL 10-10 transmission electron microscope (JEOL, Tokyo, Japan) [54].

4.2. Sphingomielinase Activity

The aSMase and nSMase activity was measured as previously described [9]. Fluorescence was measured at ƛex = 545 nm and λem = 590 nm using a microplate reader (Synergy® HTX multimode).

4.3. Ceramidase Activity

aCDase and nCDase activity was measured as previously described [9]. Fluorescence was measured at λex 355 nm and λem 460 nm using a microplate fluorescence reader (Synergy® HTX multimode, BioTek Instruments, Inc., Winooski, VT, USA). The same reaction mixture without enzyme was used as a blank.

4.4. Immunofluorescence

The kidneys were excised, fixed in 4% paraformaldehyde-PBS, and dehydrated in ascending grades of ethanol, cleaned in xylene, and embedded in paraffin. The kidney sections were cut at 3 μm thickness and mounted in coverslips for immunostaining. The sections were incubated with blocking solution for 1 h at room temperature followed by incubation at 4 °C overnight with the following primary antibodies (1:500): nSMase (mouse monoclonal antibody, sc-166637, Santa Cruz Biotechnology, Dallas, TX, USA), nCDase—FITC (mouse monoclonal antibody, sc-374634 FITC, Santa Cruz Biotechnology), SphK1—FITC (mouse monoclonal antibody, sc-365401 FITC, Santa Cruz Biotechnology) [55]. Only the sections with nSMase were incubated with the secondary antibody (m-IgGκ BP-FITC (sc-516140, Santa Cruz Biotechnology) at 4 °C overnight.
To identify the presence of nSMase, nCDase, and SphK1 in the glomerulus and subsegments of the renal tubule, the tissues were co-stained with anti-thymocyte-1 (Thy-1 PE) (mouse monoclonal antibody, sc-53116 PE, Santa Cruz Biotechnology) as a marker of mesangial cells [56], anti-aquaporin-1 (AQP1) (mouse monoclonal antibody, sc-25287 PE, Santa Cruz Biotechnology) as a marker of proximal tubule and thin descending limb of Henle’s loop and the vasa recta in the outer medulla [57], anti-ClC-K1 (Santa Cruz Biotechnology) as a marker of thin ascending limb of Henle’s loop in inner medulla (tAL) [58], anti-Tamm–Horsfall protein (THP) (mouse monoclonal antibody, sc-271022 PE, Santa Cruz Biotechnology) as a marker of distal and thick ascending limb of Henle’s loop (TAL) [59], and anti-aquqporin-2 (AQP2) (mouse monoclonal antibody, sc-47710 AF594, Santa Cruz Biotechnology) as a marker of principal cells of the collecting duct [60,61]. Specificity of immunostaining was assessed by incubation in the absence of primary antibody and to reduce autofluorescence, we used TrueBlack Lipofuscin Autofluorescence Quencher (#23007; Biotium, TermoFisher, Waltham, MA, USA) [62]. Fluorescence images were obtained using a Cell Imaging Station (Life Technologies, Carlsbad, CA, USA).

4.5. Statistical Analysis

Statistical analysis was performed using Prism software (GraphPad 6, San Diego, CA, USA). Results are presented as means ± SE. The significance of differences between groups was evaluated via ANOVA followed by the Bonferroni post hoc test. Differences with p < 0.05 were considered statistically significant.

5. Conclusions

In conclusion, our results suggest that aSMase and nCDase activity in urine could be a novel predictor of early slight ultrastructural changes in the nephron, aSMase and nCDase as glomerular injury biomarkers, and nSMase as a tubular injury biomarker in diabetic and hypertensive rats.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms242316633/s1.

Author Contributions

Conceptualization, R.B.-P.; performed the experiments, M.F., A.C.-M., M.d.P.R.-G., R.L.-M., L.D.-M., J.S.S. and R.B.-P.; data analysis, R.B.-P. and M.F., and writing—original draft preparation, R.B.-P. review and editing, M.F.; funding acquisition, R.B.-P. All authors have read and agreed to the published version of the manuscript.

Funding

This project was supported by National Council of Humanities, Sciences and Technologies (CONAHCyT) “Project supported by the sectoral research fund for education” Research Grant A1-S-9870 (to Rocio Bautista-Pérez).

Institutional Review Board Statement

The procedures used in this study were performed in accordance with the Mexican Federal Regulation for Animal Experimentation and Care (NOM-062-ZOO-1999, published in 2001) and approved by the Institutional Committee for the Care and Use of Laboratory Animals of the Instituto Nacional de Cardiología “Ignacio Chávez”, under protocols Numbers (INC-CICUAL/012/2019, INC-CICUAL/005/2020).

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article and supplementary materials.

Acknowledgments

This study was partially supported by Fondos de Gasto Directo Autorizado a la Subdirección de Investigación Básica del Instituto Nacional de Cardiología “Ignacio Chávez” to Project number 19-1126 (to Rocío Bautista-Pérez). A special acknowledgement must go to Olga Lidia Pérez-Reyes of Pathology Department, QFB Silvestre Ramírez Fuentes of Molecular Biology Department, Instituto Nacional de Cardiología “Ignacio Chávez” for technical support.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Baranowski, M.; Blachnio-Zabielska, A.; Hirnle, T.; Harasiuk, D.; Matlak, K.; Knapp, M.; Zabielski, P.; Górski, J. Myocardium of type 2 diabetic and obese patients is characterized by alterations in sphingolipid metabolic enzymes but not by accumulation of ceramide. J. Lipid Res. 2010, 51, 74–80. [Google Scholar] [CrossRef]
  2. Blachnio-Zabielska, A.U.; Koutsari, C.; Tchkonia, T.; Jensen, M.D. Sphingolipid Content of Human Adipose Tissue: Relationship to Adiponectin and Insulin Resistance. Obesity 2012, 20, 2341–2347. [Google Scholar] [CrossRef]
  3. Błachnio-Zabielska, A.; Pułka, M.; Baranowski, M.; Nikołajuk, A.; Zabielski, P.; Górska, M.; Górski, J. Ceramide metabolism is affected by obesity and diabetes in human adipose tissue. J. Cell. Physiol. 2012, 227, 550–557. [Google Scholar] [CrossRef] [PubMed]
  4. Kolak, M.; Gertow, J.; Westerbacka, J.; Summers, A.S.; Liska, J.; Franco-Cereceda, A.; Orešič, M.; Yki-Järvinen, H.; Eriksson, P.; Fisher, R.M. Expression of ceramide-metabolising enzymes in subcutaneous and intra-abdominal human adipose tissue. Lipids Health Dis. 2012, 11, 115. [Google Scholar] [CrossRef]
  5. Pepe, G.; Cotugno, M.; Marracino, F.; Giova, S.; Capocci, L.; Forte, M.; Stanzione, R.; Bianchi, F.; Marchitti, S.; Di Pardo, A.; et al. Differential Expression of Sphingolipid Metabolizing Enzymes in Spontaneously Hypertensive Rats: A Possible Substrate for Susceptibility to Brain and Kidney Damage. Int. J. Mol. Sci. 2021, 22, 3796. [Google Scholar] [CrossRef]
  6. Doehner, W.; Bunck, A.C.; Rauchhaus, M.; von Haehling, S.; Brunkhorst, F.M.; Cicoira, M.; Tschope, C.; Ponikowski, P.; Claus, R.A.; Anker, S.D. Secretory sphingomyelinase is upregulated in chronic heart failure: A second messenger system of immune activation relates to body composition, muscular functional capacity, and peripheral blood flow. Eur. Heart J. 2007, 28, 821–828. [Google Scholar] [CrossRef] [PubMed]
  7. Empinado, H.M.; Deevska, G.M.; Nikolova-Karakashian, M.; Yoo, J.-K.; Christou, D.D.; Ferreira, L.F. Diaphragm dysfunction in heart failure is accompanied by increases in neutral sphingomyelinase activity and ceramide content. Eur. J. Heart Fail. 2014, 16, 519–525. [Google Scholar] [CrossRef]
  8. Boini, K.M.; Xia, M.; Koka, S.; Gehr, T.W.; Li, P.-L. Instigation of NLRP3 inflammasome activation and glomerular injury in mice on the high fat diet: Role of acid sphingomyelinase gene. Oncotarget 2016, 7, 19031–19044. [Google Scholar] [CrossRef] [PubMed]
  9. Pérez-Villavicencio, R.; Flores-Estrada, J.; Franco, M.; Escalante, B.; Pérez-Méndez, O.; Mercado, A.; Bautista-Pérez, R. Effect of Empagliflozin on Sphingolipid Catabolism in Diabetic and Hypertensive Rats. Int. J. Mol. Sci. 2022, 23, 2883. [Google Scholar] [CrossRef]
  10. Urbanek, K.; Cappetta, D.; Bellocchio, G.; Coppola, M.A.; Imbrici, P.; Telesca, M.; Donniacuo, M.; Riemma, M.A.; Mele, E.; Cianflone, E.; et al. Dapagliflozin protects the kidney in a non-diabetic model of cardiorenal syndrome. Pharmacol. Res. 2023, 188, 106659. [Google Scholar] [CrossRef]
  11. Mantovani, A.; Petracca, G.; Beatrice, G.; Csermely, A.; Lonardo, A.; Schattenberg, J.M.; Tilg, H.; Byrne, C.D.; Targher, G. Non-alcoholic fatty liver disease and risk of incident chronic kidney disease: An updated meta-analysis. Gut 2022, 71, 156–162. [Google Scholar] [CrossRef]
  12. Liu, Y.; Flores, D.; Carrisoza-Gaytán, R.; Rohatgi, R. Biomechanical regulation of cyclooxygenase-2 in the renal collecting duct. Am. J. Physiol.-Ren. Physiol. 2014, 306, F214–F223. [Google Scholar] [CrossRef] [PubMed]
  13. Mitsutake, S.; Tani, M.; Okino, N.; Mori, K.; Ichinose, S.; Omori, A.; Iida, H.; Nakamura, T.; Ito, M. Purification, characterization, molecular cloning, and subcellular distribution of neutral ceramidase of rat kidney. J. Biol. Chem. 2001, 276, 26249–26259. [Google Scholar] [CrossRef] [PubMed]
  14. Olivera, A.; Kohama, T.; Tu, Z.; Milstien, S.; Spiegel, S. Purification and characterization of rat kidney sphingosine kinase. J. Biol. Chem. 1998, 273, 12576–12583. [Google Scholar] [CrossRef]
  15. Facchinetti, M.M.; Leocata Nieto, F.; Márquez, M.G.; Sterin-Speziale, N. Stratification of sphingosine kinase-1 expression and activity in rat kidney. Cells Tissues Organs 2008, 188, 384–392. [Google Scholar] [CrossRef]
  16. Kirby, R.J.; Jin, Y.; Fu, J.; Cubillos, J.; Swertfeger, D.; Arend, L.J. Dynamic regulation of sphingosine-1-phosphate homeostasis during development of mouse metanephric kidney. Am. J. Physiol.-Ren. Physiol. 2009, 296, F634–F641. [Google Scholar] [CrossRef] [PubMed]
  17. Sacket, S.J.; Chung, H.Y.; Okajima, F.; Im, D.S. Increase in sphingolipid catabolic enzyme activity during aging. Acta Pharmacol. Sin. 2009, 30, 1454–1461. [Google Scholar] [CrossRef]
  18. Zager, R.A.; Conrad, S.; Lochhead, K.; Sweeney, E.A.; Igarashi, Y.; Burkhart, K.M. Altered sphingomyelinase and ceramide expression in the setting of ischemic and nephrotoxic acute renal failure. Kidney Int. 1998, 53, 573–582. [Google Scholar] [CrossRef]
  19. Ichi, I.; Kamikawa, C.; Nakagawa, T.; Kobayashi, K.; Kataoka, R.; Nagata, E.; Kitamura, Y.; Nakazaki, C.; Matsura, T.; Kojo, S. Neutral sphingomyelinase-induced ceramide accumulation by oxidative stress during carbon tetrachloride intoxication. Toxicology 2009, 261, 33–40. [Google Scholar] [CrossRef]
  20. Geoffroy, K.; Troncy, L.; Wiernsperger, N.; Lagarde, M.; El Bawab, S. Glomerular proliferation during early stages of diabetic nephropathy is associated with local increase of sphingosine-1-phosphate levels. FEBS Lett. 2005, 579, 1249–1254. [Google Scholar] [CrossRef]
  21. Gorska, M.; Barańczuk, E.; Dobrzyń, A. Secretory Zn2+-dependent Sphingomyelinase Activity in the Serum of Patients with Type 2 Diabetes is Elevated. Horm. Metab. Res. 2003, 35, 506–507. [Google Scholar]
  22. Pan, W.; Yu, J.; Shi, R.; Yan, L.; Yang, T.; Li, Y.; Zhang, Z.; Yu, G.; Bai, Y.; Schuchman, E.H.; et al. Elevation of ceramide and activation of secretory acid sphingomyelinase in patients with acute coronary syndromes. Coron. Artery Dis. 2014, 25, 230–235. [Google Scholar] [CrossRef]
  23. Spijkers, L.J.; van den Akker, R.F.; Janssen, B.J.; Debets, J.J.; De Mey, J.G.; Stroes, E.S.; van den Born, B.J.; Wijesinghe, D.S.; Chalfant, C.E.; MacAleese, L.; et al. Hypertension is associated with marked alterations in sphingolipid biology: A potential role for ceramide. PLoS ONE 2011, 6, e21817. [Google Scholar] [CrossRef]
  24. Subathra, M.; Korrapati, M.; Howell, L.A.; Arthur, J.M.; Shayman, J.A.; Schnellmann, R.G.; Siskind, L.J. Kidney glycosphingolipids are elevated early in diabetic nephropathy and mediate hypertrophy of mesangial cells. Am. J. Physiol.-Ren. Physiol. 2015, 309, F204–F215. [Google Scholar] [CrossRef]
  25. Magagnotti, C.; Zerbini, G.; Fermo, I.; Carletti, R.M.; Bonfanti, R.; Vallone, F.; Andolfo, A. Identification of nephropathy predictors in urine from children with a recent diagnosis of type 1 diabetes. J. Proteom. 2019, 193, 205–216. [Google Scholar] [CrossRef]
  26. Morita, Y.; Kurano, M.; Sakai, E.; Nishikawa, T.; Nishikawa, M.; Sawabe, M.; Aoki, J.; Yatomi, Y. Analysis of urinary sphingolipids using liquid chromatography-tandem mass spectrometry in diabetic nephropathy. J. Diabetes Investig. 2020, 11, 441–449. [Google Scholar] [CrossRef] [PubMed]
  27. Kurano, M.; Jubishi, D.; Okamoto, K.; Hashimoto, H.; Sakai, E.; Morita, Y.; Saigusa, D.; Kano, K.; Aoki, J.; Harada, S.; et al. Dynamic modulations of urinary sphingolipid and glycerophospholipid levels in COVID-19 and correlations with COVID-19-associated kidney injuries. J. Biomed. Sci. 2022, 29, 94. [Google Scholar] [CrossRef] [PubMed]
  28. Quintern, L.E.; Weitz, G.; Nehrkorn, H.; Tager, J.M.; Schram, A.; Sandhoff, K. Acid sphingomyelinase from human urine: Purification and characterization. Biochim. Et Biophys. Acta (BBA)-Lipids Lipid Metab. 1987, 922, 323–336. [Google Scholar] [CrossRef] [PubMed]
  29. Chatterjee, S.; Ghosh, N. Neutral sphingomyelinase from human urine. Purification and preparation of monospecific antibodies. J. Biol. Chem. 1989, 264, 12554–12561, Erratum in: J. Biol. Chem. 1990, 265, 1231. [Google Scholar] [CrossRef]
  30. Mima, A.; Arai, H.; Matsubara, T.; Abe, H.; Nagai, K.; Tamura, Y.; Torikoshi, K.; Araki, M.; Kanamori, H.; Takahashi, T.; et al. Urinary Smad1 is a novel marker to predict later onset of mesangial matrix expansion in diabetic nephropathy. Diabetes 2008, 57, 1712–1722. [Google Scholar] [CrossRef] [PubMed]
  31. Waikar, S.S.; Betensky, R.A.; Emerson, S.C.; Bonventre, J.V. Imperfect gold standards for biomarker evaluation. Clin. Trials 2013, 10, 696–700. [Google Scholar] [CrossRef] [PubMed]
  32. Klein, J.; Schanstra, J.P. Implementation of Proteomics Biomarkers in Nephrology: From Animal Models to Human Application? PROTEOMICS—Clin. Appl. 2019, 13, e1800089. [Google Scholar] [CrossRef] [PubMed]
  33. Westhuyzen, J.; Endre, Z.H.; Reece, G.; Reith, D.M.; Saltissi, D.; Morgan, T.J. Measurement of tubular enzymuria facilitates early detection of acute renal impairment in the intensive care unit. Nephrol. Dial. Transplant. 2003, 18, 543–551. [Google Scholar] [CrossRef] [PubMed]
  34. Marathe, S.; Schissel, S.L.; Yellin, M.J.; Beatini, N.; Mintzer, R.; Williams, K.J.; Tabas, I. Human vascular endothelial cells are a rich and regulatable source of secretory sphingomyelinase. Implications for early atherogenesis and ceramide-mediated cell signaling. J. Biol. Chem. 1998, 273, 4081–4088. [Google Scholar] [CrossRef] [PubMed]
  35. Romiti, E.; Meacci, E.; Tani, M.; Nuti, F.; Farnararo, M.; Ito, M.; Bruni, P. Neutral/alkaline and acid ceramidase activities are actively released by murine endothelial cells. Biochem. Biophys. Res. Commun. 2000, 275, 746–751. [Google Scholar] [CrossRef] [PubMed]
  36. Romiti, E.; Meacci, E.; Donati, C.; Formigli, L.; Zecchi-Orlandini, S.; Farnararo, M.; Ito, M.; Bruni, P. Neutral ceramidase secreted by endothelial cells is released in part associated with caveolin-1. Arch. Biochem. Biophys. 2003, 417, 27–33. [Google Scholar] [CrossRef] [PubMed]
  37. Tomiuk, S.; Zumbansen, M.; Stoffel, W. Characterization and subcellular localization of murine and human magnesium-dependent neutral sphingomyelinase. J. Biol. Chem. 2000, 275, 5710–5717. [Google Scholar] [CrossRef]
  38. Wu, B.X.; Rajagopalan, V.; Roddy, P.L.; Clarke, C.J.; Hannun, Y.A. Identification and characterization of murine mitochondria-associated neutral sphingomyelinase (MA-nSMase), the mammalian sphingomyelin phosphodiesterase 5. J. Biol. Chem. 2010, 285, 17993–18002. [Google Scholar] [CrossRef]
  39. Gorelik, A.; Illes, K.; Heinz, L.X.; Superti-Furga, G.; Nagar, B. Crystal structure of mammalian acid sphingomyelinase. Nat. Commun. 2016, 7, 12196. [Google Scholar] [CrossRef]
  40. García-Ruiz, C.; Colell, A.; Marí, M.; Morales, A.; Fernández-Checa, J.C. Direct effect of ceramide on the mitochondrial electron transport chain leads to generation of reactive oxygen species. Role of mitochondrial glutathione. J. Biol. Chem. 1997, 272, 11369–11377. [Google Scholar] [CrossRef]
  41. Castillo, S.S.; Levy, M.; Thaikoottathil, J.V.; Goldkorn, T. Reactive nitrogen and oxygen species activate different sphingomyelinases to induce apoptosis in airway epithelial cells. Exp. Cell Res. 2007, 313, 2680–2686. [Google Scholar] [CrossRef]
  42. Birbes, H.; El Bawab, S.; Hannun, Y.A.; Obeid, L.M. Selective hydrolysis of a mitochondrial pool of sphingomyelin induces apoptosis. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2001, 15, 2669–2679. [Google Scholar] [CrossRef]
  43. Rutkute, K.; Asmis, R.H.; Nikolova-Karakashian, M.N. Regulation of neutral sphingomyelinase-2 by GSH: A new insight to the role of oxidative stress in aging-associated inflammation. J. Lipid Res. 2007, 48, 2443–2452. [Google Scholar] [CrossRef]
  44. Altura, B.M.; Shah, N.C.; Shah, G.; Zhang, A.; Li, W.; Zheng, T.; Perez-Albela, J.L.; Altura, B.T. Short-term magnesium deficiency upregulates ceramide synthase in cardiovascular tissues and cells: Cross-talk among cytokines, Mg2+, NF-κB, and de novo ceramide. Am. J. Physiology Heart Circ. Physiol. 2012, 302, H319–H332. [Google Scholar] [CrossRef] [PubMed]
  45. Franco, M.; Tapia, E.; Santamaría, J.; Zafra, I.; García-Torres, R.; Gordon, K.L.; Pons, H.; Rodríguez-Iturbe, B.; Johnson, R.J.; Herrera-Acosta, J. Renal cortical vasoconstriction contributes to development of salt-sensitive hypertension after angiotensin II exposure. J. Am. Soc. Nephrol. JASN 2001, 12, 2263–2271. [Google Scholar] [CrossRef] [PubMed]
  46. Bautista-Pérez, R.; Pérez-Méndez, O.; Cano-Martínez, A.; Pacheco, U.; Santamaría, J.; Rodríguez-Iturbe, F.R.B.; Navar, L.G.; Franco, M. The Role of P2X7 Purinergic Receptors in the Renal Inflammation Associated with Angiotensin II-induced Hypertension. Int. J. Mol. Sci. 2020, 21, 4041. [Google Scholar] [CrossRef] [PubMed]
  47. Coroneos, E.; Martinez, M.; McKenna, S.; Kester, M. Differential regulation of sphingomyelinase and ceramidase activities by growth factors and cytokines. Implications for cellular proliferation and differentiation. J. Biol. Chem. 1995, 270, 23305–23309. [Google Scholar] [CrossRef]
  48. Kaszkin, M.; Huwiler, A.; Scholz, K.; van den Bosch, H.; Pfeilschifter, J. Negative regulation of interleukin-1beta-activated neutral sphingomyelinase by protein kinase C in rat mesangial cells. FEBS Lett. 1998, 440, 163–166. [Google Scholar] [CrossRef]
  49. Franzen, R.; Pautz, A.; Bräutigam, L.; Geisslinger, G.; Pfeilschifter, J.; Huwiler, A. Interleukin-1beta induces chronic activation and de novo synthesis of neutral ceramidase in renal mesangial cells. J. Biol. Chem. 2001, 276, 35382–35389. [Google Scholar] [CrossRef] [PubMed]
  50. Liu, G.; Kleine, L.; Nasrallah, R.; Hébert, R.L. Bradykinin inhibits ceramide production and activates phospholipase D in rabbit cortical collecting duct cells. Am. J. Physiol. 1999, 276, F589–F598. [Google Scholar] [CrossRef]
  51. Hammad, S.M.; Twal, W.O.; Arif, E.; Semler, A.J.; Klein, R.L.; Nihalani, D. Transcriptomics Reveal Altered Metabolic and Signaling Pathways in Podocytes Exposed to C16 Ceramide-Enriched Lipoproteins. Genes 2020, 11, 178. [Google Scholar] [CrossRef] [PubMed]
  52. Yaghobian, D.; Don, A.S.; Yaghobian, S.; Chen, X.; Pollock, C.A.; Saad, S. Increased sphingosine 1-phosphate mediates inflammation and fibrosis in tubular injury in diabetic nephropathy. Clin. Exp. Pharmacol. Physiol. 2016, 43, 56–66. [Google Scholar] [CrossRef] [PubMed]
  53. Zhang, S.X.; Wang, J.J.; Lu, K.; Mott, R.; Longeras, R.; Ma, J.X. Therapeutic potential of angiostatin in diabetic nephropathy. J. Am. Soc. Nephrol. JASN 2006, 17, 475–486. [Google Scholar] [CrossRef]
  54. Tizro, P.; Choi, C.; Khanlou, N. Sample Preparation for Transmission Electron Microscopy. Methods Mol. Biol. 2019, 1897, 417–424. [Google Scholar] [CrossRef] [PubMed]
  55. Arroyo, A.I.; Camoletto, P.G.; Morando, L.; Sassoe-Pognetto, M.; Giustetto, M.; Van Veldhoven, P.P.; Schuchman, E.H.; Ledesma, M.D. Pharmacological reversion of sphingomyelin-induced dendritic spine anomalies in a Niemann Pick disease type A mouse model. EMBO Mol. Med. 2014, 6, 398–413. [Google Scholar] [CrossRef]
  56. Yamamoto, T.; Yamamoto, K.; Kawasaki, K.; Yaoita, E.; Shimizu, F.; Kihara, I. Immunoelectron microscopic demonstration of Thy-1 antigen on the surfaces of mesangial cells in the rat glomerulus. Nephron 1986, 43, 293–298. [Google Scholar] [CrossRef]
  57. Tinning, A.R.; Jensen, B.L.; Schweda, F.; Machura, K.; Hansen, P.B.; Stubbe, J.; Gramsbergen, J.B.; Madsen, K. The water channel aquaporin-1 contributes to renin cell recruitment during chronic stimulation of renin production. Am. J. Physiol.-Ren. Physiol. 2014, 307, F1215–F1226. [Google Scholar] [CrossRef]
  58. Uchida, S.; Sasaki, S.; Nitta, K.; Uchida, K.; Horita, S.; Nihei, H.; Marumo, F. Localization and functional characterization of rat kidney-specific chloride channel, ClC-K1. J. Clin. Investig. 1995, 95, 104–113. [Google Scholar] [CrossRef]
  59. Dong, L.; Zieren, R.C.; Horie, K.; Kim, C.J.; Mallick, E.; Jing, Y.; Feng, M.; Kuczler, M.D.; Green, J.; Amend, S.R.; et al. Comprehensive evaluation of methods for small extracellular vesicles separation from human plasma, urine, and cell culture medium. J. Extracell. Vesicles 2020, 10, e12044. [Google Scholar] [CrossRef]
  60. Disset, A.; Cheval, L.; Soutourina, O.; Duong Van Huyen, J.P.; Li, G.; Genin, C.; Tostain, J.; Loupy, A.; Doucet, A.; Rajerison, R. Tissue compartment analysis for biomarker discovery by gene expression profiling. PLoS ONE 2009, 4, e7779. [Google Scholar] [CrossRef]
  61. Loupy, A.; Ramakrishnan, S.K.; Wootla, B.; Chambrey, R.; de la Faille, R.; Bourgeois, S.; Bruneval, P.; Mandet, C.; Christensen, E.I.; Faure, H.; et al. PTH-independent regulation of blood calcium concentration by the calcium-sensing receptor. J. Clin. Investig. 2012, 122, 3355–3367. [Google Scholar] [CrossRef] [PubMed]
  62. Sorrelle, N.; Ganguly, D.; Dominguez AT, A.; Zhang, Y.; Huang, H.; Dahal, L.N.; Burton, N.; Ziemys, A.; Brekken, R.A. Improved Multiplex Immunohistochemistry for Immune Microenvironment Evaluation of Mouse Formalin-Fixed, Paraffin-Embedded Tissues. J. Immunol. 2019, 202, 292–299. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Representative electron micrographs of kidney from control (a,d,g), diabetic (b,e,h) and hypertension rats (c,f,i). Glomeruli: (a) detached endothelial layer in the glomerular capillary (thick arrow), glomerular basement membrane thickening (GBM) (arrow pairs), and fusion of podocytes foot (stars) (b,c). The proximal tubule shows that decreased and irregular microvilli (e,f), the mitochondria are round with loss of matrix density (h,i), and swelled cristae (h,i). Supplemental material available online with this article.
Figure 1. Representative electron micrographs of kidney from control (a,d,g), diabetic (b,e,h) and hypertension rats (c,f,i). Glomeruli: (a) detached endothelial layer in the glomerular capillary (thick arrow), glomerular basement membrane thickening (GBM) (arrow pairs), and fusion of podocytes foot (stars) (b,c). The proximal tubule shows that decreased and irregular microvilli (e,f), the mitochondria are round with loss of matrix density (h,i), and swelled cristae (h,i). Supplemental material available online with this article.
Ijms 24 16633 g001
Figure 2. Enzymatic activity of aSMase, nSMase, and nCDase in the urine of control, and diabetic rats. Each bar represents the mean ± SE of n = 10. * p < 0.05 diabetic vs. control rats.
Figure 2. Enzymatic activity of aSMase, nSMase, and nCDase in the urine of control, and diabetic rats. Each bar represents the mean ± SE of n = 10. * p < 0.05 diabetic vs. control rats.
Ijms 24 16633 g002
Figure 3. Enzymatic activity of aSMase, nSMase, and nCDase in the urine of normotensive, and hypertensive rats. Each bar represents the mean ± SE of n = 10. * p < 0.05 hypertensive vs. normotensive rats.
Figure 3. Enzymatic activity of aSMase, nSMase, and nCDase in the urine of normotensive, and hypertensive rats. Each bar represents the mean ± SE of n = 10. * p < 0.05 hypertensive vs. normotensive rats.
Ijms 24 16633 g003
Figure 4. Coimmunostaining of nSMase, nCDase or SphK1 with Thy-1 as marker of mesangial cells in kidney.
Figure 4. Coimmunostaining of nSMase, nCDase or SphK1 with Thy-1 as marker of mesangial cells in kidney.
Ijms 24 16633 g004
Figure 5. Coimmunostaining of nSMase, nCDase or SphK1 with AQP1 as a marker of proximal tubule in kidney.
Figure 5. Coimmunostaining of nSMase, nCDase or SphK1 with AQP1 as a marker of proximal tubule in kidney.
Ijms 24 16633 g005
Figure 6. Coimmunostaining of nSMase, nCDase or SphK1 with CIC-1 as a marker of ascending thin limb of the loop of Henle in kidney.
Figure 6. Coimmunostaining of nSMase, nCDase or SphK1 with CIC-1 as a marker of ascending thin limb of the loop of Henle in kidney.
Ijms 24 16633 g006
Figure 7. Coimmunostaining of nSMase, nCDase or SphK1 with THP as a marker of ascending thick limb of Henle’s loop (TAL) in kidney.
Figure 7. Coimmunostaining of nSMase, nCDase or SphK1 with THP as a marker of ascending thick limb of Henle’s loop (TAL) in kidney.
Ijms 24 16633 g007
Figure 8. Coimmunostaining of nSMase, nCDase or SphK1 with AQP2 as a marker of principal cells of the collecting duct (CD) in kidney.
Figure 8. Coimmunostaining of nSMase, nCDase or SphK1 with AQP2 as a marker of principal cells of the collecting duct (CD) in kidney.
Ijms 24 16633 g008
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Franco, M.; Cano-Martínez, A.; Ramos-Godínez, M.d.P.; López-Marure, R.; Donis-Maturano, L.; Sosa, J.S.; Bautista-Pérez, R. Immunolocalization of Sphingolipid Catabolism Enzymes along the Nephron: Novel Early Urinary Biomarkers of Renal Damage. Int. J. Mol. Sci. 2023, 24, 16633. https://doi.org/10.3390/ijms242316633

AMA Style

Franco M, Cano-Martínez A, Ramos-Godínez MdP, López-Marure R, Donis-Maturano L, Sosa JS, Bautista-Pérez R. Immunolocalization of Sphingolipid Catabolism Enzymes along the Nephron: Novel Early Urinary Biomarkers of Renal Damage. International Journal of Molecular Sciences. 2023; 24(23):16633. https://doi.org/10.3390/ijms242316633

Chicago/Turabian Style

Franco, Martha, Agustina Cano-Martínez, María del Pilar Ramos-Godínez, Rebeca López-Marure, Luis Donis-Maturano, José Santamaría Sosa, and Rocio Bautista-Pérez. 2023. "Immunolocalization of Sphingolipid Catabolism Enzymes along the Nephron: Novel Early Urinary Biomarkers of Renal Damage" International Journal of Molecular Sciences 24, no. 23: 16633. https://doi.org/10.3390/ijms242316633

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