1
|
Kaiser-Graf D, Schulz A, Mangelsen E, Rothe M, Bolbrinker J, Kreutz R. Tissue lipidomic profiling supports a mechanistic role of the prostaglandin E2 pathway for albuminuria development in glomerular hyperfiltration. FRONTIERS IN NETWORK PHYSIOLOGY 2023; 3:1271042. [PMID: 38205443 PMCID: PMC10777844 DOI: 10.3389/fnetp.2023.1271042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/21/2023] [Indexed: 01/12/2024]
Abstract
Background: Glomerular hyperfiltration (GH) is an important mechanism in the development of albuminuria in hypertension. The Munich Wistar Frömter (MWF) rat is a non-diabetic model of chronic kidney disease (CKD) with GH due to inherited low nephron number resulting in spontaneous albuminuria and podocyte injury. In MWF rats, we identified prostaglandin (PG) E2 (PGE2) signaling as a potential causative mechanism of albuminuria in GH. Method: For evaluation of the renal PGE2 metabolic pathway, time-course lipidomic analysis of PGE2 and its downstream metabolites 15-keto-PGE2 and 13-14-dihydro-15-keto-PGE2 was conducted in urine, plasma and kidney tissues of MWF rats and albuminuria-resistant spontaneously hypertensive rats (SHR) by liquid chromatography electrospray ionization tandem mass spectrometry (LC/ESI-MS/MS). Results: Lipidomic analysis revealed no dysregulation of plasma PGs over the time course of albuminuria development, while glomerular levels of PGE2 and 15-keto-PGE2 were significantly elevated in MWF compared to albuminuria-resistant SHR. Overall, averaged PGE2 levels in glomeruli were up to ×150 higher than the corresponding 15-keto-PGE2 levels. Glomerular metabolic ratios of 15-hydroxyprostaglandin dehydrogenase (15-PGDH) were significantly lower, while metabolic ratios of prostaglandin reductases (PTGRs) were significantly higher in MWF rats with manifested albuminuria compared to SHR, respectively. Conclusion: Our data reveal glomerular dysregulation of the PGE2 metabolism in the development of albuminuria in GH, resulting at least partly from reduced PGE2 degradation. This study provides first insights into dynamic changes of the PGE2 pathway that support a role of glomerular PGE2 metabolism and signaling for early albuminuria manifestation in GH.
Collapse
Affiliation(s)
- Debora Kaiser-Graf
- Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Clinical Pharmacology and Toxicology, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Angela Schulz
- Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Clinical Pharmacology and Toxicology, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Eva Mangelsen
- Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Clinical Pharmacology and Toxicology, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | | | - Juliane Bolbrinker
- Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Clinical Pharmacology and Toxicology, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Reinhold Kreutz
- Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Clinical Pharmacology and Toxicology, Charité—Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
2
|
Kourpa A, Schulz A, Mangelsen E, Kaiser-Graf D, Koppers N, Stoll M, Rothe M, Bader M, Purfürst B, Kunz S, Gladytz T, Niendorf T, Bachmann S, Mutig K, Bolbrinker J, Panáková D, Kreutz R. Studies in Zebrafish and Rat Models Support Dual Blockade of EP2 and EP4 (Prostaglandin E 2 Receptors Type 2 and 4) for Renoprotection in Glomerular Hyperfiltration and Albuminuria. Hypertension 2023; 80:771-782. [PMID: 36715011 DOI: 10.1161/hypertensionaha.122.20392] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Glomerular hyperfiltration (GH) is an important mechanism in the development of albuminuria in hypertension. Upregulation of COX2 (cyclooxygenase 2) and prostaglandin E2 (PGE2) was linked to podocyte damage in GH. We explored the potential renoprotective effects of either separate or combined pharmacological blockade of EP2 (PGE2 receptor type 2) and EP4 (PGE2 receptor type 4) in GH. METHODS We conducted in vivo studies in a transgenic zebrafish model (Tg[fabp10a:gc-EGFP]) suitable for analysis of glomerular filtration barrier function and a genetic rat model with GH, albuminuria, and upregulation of PGE2. Similar pharmacological interventions and primary outcome analysis on albuminuria phenotype development were conducted in both model systems. RESULTS Stimulation of zebrafish embryos with PGE2 induced an albuminuria-like phenotype, thus mimicking the suggested PGE2 effects on glomerular filtration barrier dysfunction. Both separate and combined blockade of EP2 and EP4 reduced albuminuria phenotypes in zebrafish and rat models. A significant correlation between albuminuria and podocyte damage in electron microscopy imaging was identified in the rat model. Dual blockade of both receptors showed a pronounced synergistic suppression of albuminuria. Importantly, this occurred without changes in arterial blood pressure, glomerular filtration rate, or tissue oxygenation in magnetic resonance imaging, while RNA sequencing analysis implicated a potential role of circadian clock genes. CONCLUSIONS Our findings confirm a role of PGE2 in the development of albuminuria in GH and support the renoprotective potential of combined pharmacological blockade of EP2 and EP4 receptors. These data support further translational research to explore this therapeutic option and a possible role of circadian clock genes.
Collapse
Affiliation(s)
- Aikaterini Kourpa
- Institute of Clinical Pharmacology and Toxicology (A.K., A.S., E.M., D.K.-G., J.B., R.K.), Charité-Universitätsmedizin Berlin, Germany.,Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (A.K., M.B., B.P., S.K., T.G., T.N., D.P.)
| | - Angela Schulz
- Institute of Clinical Pharmacology and Toxicology (A.K., A.S., E.M., D.K.-G., J.B., R.K.), Charité-Universitätsmedizin Berlin, Germany
| | - Eva Mangelsen
- Institute of Clinical Pharmacology and Toxicology (A.K., A.S., E.M., D.K.-G., J.B., R.K.), Charité-Universitätsmedizin Berlin, Germany
| | - Debora Kaiser-Graf
- Institute of Clinical Pharmacology and Toxicology (A.K., A.S., E.M., D.K.-G., J.B., R.K.), Charité-Universitätsmedizin Berlin, Germany
| | - Nils Koppers
- Genetic Epidemiology, Institute for Human Genetics, Westfälische Wilhelms University, Münster, Germany (N.K., M.S.)
| | - Monika Stoll
- Genetic Epidemiology, Institute for Human Genetics, Westfälische Wilhelms University, Münster, Germany (N.K., M.S.)
| | | | - Michael Bader
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (A.K., M.B., B.P., S.K., T.G., T.N., D.P.).,German Center for Cardiovascular Research, Partner Site Berlin, Germany (M.B.).,Charité-Universitätsmedizin Berlin, Germany (M.B.).,Institute for Biology, University of Lübeck, Germany (M.B.)
| | - Bettina Purfürst
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (A.K., M.B., B.P., S.K., T.G., T.N., D.P.)
| | - Severine Kunz
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (A.K., M.B., B.P., S.K., T.G., T.N., D.P.)
| | - Thomas Gladytz
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (A.K., M.B., B.P., S.K., T.G., T.N., D.P.)
| | - Thoralf Niendorf
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (A.K., M.B., B.P., S.K., T.G., T.N., D.P.)
| | - Sebastian Bachmann
- Institute of Vegetative Anatomy (S.B.), Charité-Universitätsmedizin Berlin, Germany
| | - Kerim Mutig
- Institute of Translational Physiology (K.M.), Charité-Universitätsmedizin Berlin, Germany
| | - Juliane Bolbrinker
- Institute of Clinical Pharmacology and Toxicology (A.K., A.S., E.M., D.K.-G., J.B., R.K.), Charité-Universitätsmedizin Berlin, Germany
| | - Daniela Panáková
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (A.K., M.B., B.P., S.K., T.G., T.N., D.P.)
| | - Reinhold Kreutz
- Institute of Clinical Pharmacology and Toxicology (A.K., A.S., E.M., D.K.-G., J.B., R.K.), Charité-Universitätsmedizin Berlin, Germany
| |
Collapse
|
3
|
Srivastava T, Garola RE, Zhou J, Boinpelly VC, Priya L, Ali MF, Rezaiekhaligh MH, Heruth DP, Novak J, Alon US, Joshi T, Jiang Y, McCarthy ET, Savin VJ, Johnson ML, Sharma R, Sharma M. Prostanoid receptors in hyperfiltration-mediated glomerular injury: Novel agonists and antagonists reveal opposing roles for EP2 and EP4 receptors. FASEB J 2022; 36:e22559. [PMID: 36125047 DOI: 10.1096/fj.202200875r] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/23/2022] [Accepted: 09/07/2022] [Indexed: 11/11/2022]
Abstract
Increased fluid-flow shear stress (FFSS) contributes to hyperfiltration-induced podocyte and glomerular injury resulting in progression of chronic kidney disease (CKD). We reported that increased FFSS in vitro and in vivo upregulates PGE2 receptor EP2 (but not EP4 expression), COX2-PGE2 -EP2 axis, and EP2-linked Akt-GSK3β-β-catenin signaling pathway in podocytes. To understand and use the disparities between PGE2 receptors, specific agonists, and antagonists of EP2 and EP4 were used to assess phosphorylation of Akt, GSK3β and β-catenin in podocytes using Western blotting, glomerular filtration barrier function using in vitro albumin permeability (Palb ) assay, and mitigation of hyperfiltration-induced injury in unilaterally nephrectomized (UNX) mice at 1 and 6 months. Results show an increase in Palb by PGE2 , EP2 agonist (EP2AGO ) and EP4 antagonist (EP4ANT ), but not by EP2 antagonist (EP2ANT ) or EP4 agonist (EP4AGO ). Pretreatment with EP2ANT blocked the effect of PGE2 or EP2AGO on Palb . Modulation of EP2 and EP4 also induced opposite effects on phosphorylation of Akt and β-Catenin. Individual agonists or antagonists of EP2 or EP4 did not induce significant improvement in albuminuria in UNX mice. However, treatment with a combination EP2ANT + EP4AGO for 1 or 6 months caused a robust decrease in albuminuria. EP2ANT + EP4AGO combination did not impact adaptive hypertrophy or increased serum creatinine. Observed differences between expression of EP2 and EP4 on the glomerular barrier highlight these receptors as potential targets for intervention. Safe and effective mitigating effect of EP2ANT + EP4AGO presents a novel opportunity to delay the progression of hyperfiltration-associated CKD as seen in transplant donors.
Collapse
Affiliation(s)
- Tarak Srivastava
- Section of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, Missouri, USA.,Midwest Veterans' Biomedical Research Foundation (MVBRF), Kansas City, Missouri, USA.,Department of Oral and Craniofacial Sciences, University of Missouri at Kansas City-School of Dentistry, Kansas City, Missouri, USA
| | - Robert E Garola
- Department of Pathology and Laboratory Medicine, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, Missouri, USA
| | - Jianping Zhou
- Midwest Veterans' Biomedical Research Foundation (MVBRF), Kansas City, Missouri, USA.,Renal Research Laboratory, Kansas City VA Medical Center, Kansas City, Missouri, USA
| | - Varun C Boinpelly
- Midwest Veterans' Biomedical Research Foundation (MVBRF), Kansas City, Missouri, USA.,Renal Research Laboratory, Kansas City VA Medical Center, Kansas City, Missouri, USA
| | - Lakshmi Priya
- Section of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, Missouri, USA
| | - Mohammed Farhan Ali
- Section of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, Missouri, USA
| | - Mohammad H Rezaiekhaligh
- Section of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, Missouri, USA
| | - Daniel P Heruth
- Children's Mercy Research Institute, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, Missouri, USA
| | - Jan Novak
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Uri S Alon
- Section of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, Missouri, USA
| | - Trupti Joshi
- Department of Health Management and Informatics, University of Missouri, Columbia, Missouri, USA.,Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, Missouri, USA.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA.,MU Institute for Data Science and Informatics, University of Missouri, Columbia, Missouri, USA
| | - Yuexu Jiang
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, Missouri, USA.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA
| | - Ellen T McCarthy
- Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Virginia J Savin
- Renal Research Laboratory, Kansas City VA Medical Center, Kansas City, Missouri, USA.,Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Mark L Johnson
- Department of Oral and Craniofacial Sciences, University of Missouri at Kansas City-School of Dentistry, Kansas City, Missouri, USA
| | - Ram Sharma
- Renal Research Laboratory, Kansas City VA Medical Center, Kansas City, Missouri, USA
| | - Mukut Sharma
- Midwest Veterans' Biomedical Research Foundation (MVBRF), Kansas City, Missouri, USA.,Renal Research Laboratory, Kansas City VA Medical Center, Kansas City, Missouri, USA.,Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
4
|
Voggel J, Fink G, Zelck M, Wohlfarth M, Post JM, Bindila L, Rauh M, Amann K, Alejandre Alcázar MA, Dötsch J, Nüsken KD, Nüsken E. Elevated n-3/n-6 PUFA ratio in early life diet reverses adverse intrauterine kidney programming in female rats. J Lipid Res 2022; 63:100283. [PMID: 36152882 PMCID: PMC9619183 DOI: 10.1016/j.jlr.2022.100283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/30/2022] [Accepted: 09/11/2022] [Indexed: 11/27/2022] Open
Abstract
Intrauterine growth restriction (IUGR) predisposes to chronic kidney disease via activation of proinflammatory pathways, and omega-3 PUFAs (n-3 PUFAs) have anti-inflammatory properties. In female rats, we investigated 1) how an elevated dietary n-3/n-6 PUFA ratio (1:1) during postnatal kidney development modifies kidney phospholipid (PL) and arachidonic acid (AA) metabolite content and 2) whether the diet counteracts adverse molecular protein signatures expected in IUGR kidneys. IUGR was induced by bilateral uterine vessel ligation or intrauterine stress through sham operation 3.5 days before term. Control (C) offspring were born after uncompromised pregnancy. On postnatal (P) days P2–P39, rats were fed control (n-3/n-6 PUFA ratio 1:20) or n-3 PUFA intervention diet (N3PUFA; ratio 1:1). Plasma parameters (P33), kidney cortex lipidomics and proteomics, as well as histology (P39) were studied. We found that the intervention diet tripled PL-DHA content (PC 40:6; P < 0.01) and lowered both PL-AA content (PC 38:4 and lyso-phosphatidylcholine 20:4; P < 0.05) and AA metabolites (HETEs, dihydroxyeicosatrienoic acids, and epoxyeicosatrienoic acids) to 25% in all offspring groups. After ligation, our network analysis of differentially expressed proteins identified an adverse molecular signature indicating inflammation and hypercoagulability. N3PUFA diet reversed 61 protein alterations (P < 0.05), thus mitigating adverse IUGR signatures. In conclusion, an elevated n-3/n-6 PUFA ratio in early diet strongly reduces proinflammatory PLs and mediators while increasing DHA-containing PLs regardless of prior intrauterine conditions. Counteracting a proinflammatory hypercoagulable protein signature in young adult IUGR individuals through early diet intervention may be a feasible strategy to prevent developmentally programmed kidney damage in later life.
Collapse
Affiliation(s)
- Jenny Voggel
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Gregor Fink
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany
| | - Magdalena Zelck
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany
| | - Maria Wohlfarth
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany
| | - Julia M Post
- Clinical Lipidomics Unit, Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Laura Bindila
- Clinical Lipidomics Unit, Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Manfred Rauh
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Erlangen, Germany
| | - Kerstin Amann
- Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-University Erlangen, Erlangen, Germany
| | - Miguel A Alejandre Alcázar
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Institute for Lung Health, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Gießen, Germany
| | - Jörg Dötsch
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany
| | - Kai-Dietrich Nüsken
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany
| | - Eva Nüsken
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany.
| |
Collapse
|
5
|
McArdle Z, Singh R, Bielefeldt-Ohmann H, Moritz K, Schreuder M, Denton K. Brief Early Life Angiotensin Converting Enzyme Inhibition Offers Reno-Protection in Sheep with a Solitary Functioning Kidney at 8 Months of Age. J Am Soc Nephrol 2022; 33:1341-1356. [PMID: 35351818 PMCID: PMC9257814 DOI: 10.1681/asn.2021111534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/18/2022] [Indexed: 11/03/2022] Open
Abstract
Background: Children born with a solitary functioning kidney (SFK) are predisposed to develop hypertension and kidney injury. Glomerular hyperfiltration and hypertrophy contribute to the pathophysiology of kidney injury. Angiotensin converting enzyme inhibitors (ACEi) can mitigate hyperfiltration and may be therapeutically beneficial in reducing progression of kidney injury in SFK. Methods: SFK was induced in male sheep fetuses at 100 days gestation (term=150 day). Between 4-8 weeks of age, SFK lambs received enalapril (SFK+ACEi; 0.5mg/kg/day, once daily, orally) or vehicle (SFK). At 8 months we examined whether SFK+ACEi reduced elevation in blood pressure (BP) and improved basal kidney function, renal functional reserve (RFR; glomerular filtration rate (GFR) response to combined amino acid and dopamine infusion), GFR response to nitric oxide synthase (NOS) inhibition and basal nitric oxide (NO) bioavailability (basal urinary total nitrate+nitrite (NOx)). Results: SFK+ACEi prevented albuminuria, resulted in lower basal GFR (16%), higher renal blood flow (~22%), and lower filtration fraction ( 35%), but similar BP compared to ~ vehicle-treated SFK sheep. Together with greater recruitment of RFR (~14%) in SFK+ACEi animals than SFK, this indicates reduction in glomerular hyperfiltration-mediated kidney dysfunction. During NOS inhibition, the decrease in GFR ( 14%) was greater among SFK+ACEi than among SFK animals. Increased ( 85%) basal urinary total NOx in SFK+ACEi animals compared to SFK indicates elevated NO bioavailability likely contributing to improvements in kidney function and prevention of albuminuria. Conclusions: Brief and early ACEi in SFK is associated with reduced glomerular hyperfiltration-mediated kidney disease up to 8 months of age in a sheep model.
Collapse
Affiliation(s)
- Zoe McArdle
- Z McArdle, Cardiovascular Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Australia
| | - Reetu Singh
- R Singh, Cardiovascular Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Australia
| | - Helle Bielefeldt-Ohmann
- H Bielefeldt-Ohmann, School of Veterinary Science, The University of Queensland, Gatton, Australia
| | - Karen Moritz
- K Moritz, Child Health Research Centre and School of Biomedical Sciences, The University of Queensland, Saint Lucia, Australia
| | - Michiel Schreuder
- M Schreuder, Department of Pediatric Nephrology , Amalia Children's Hospital, Nijmegen, Netherlands
| | - Kate Denton
- K Denton, Cardiovascular Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Australia
| |
Collapse
|
6
|
Srivastava T, Joshi T, Heruth DP, Rezaiekhaligh MH, Garola RE, Zhou J, Boinpelly VC, Ali MF, Alon US, Sharma M, Vanden Heuvel GB, Mahajan P, Priya L, Jiang Y, McCarthy ET, Savin VJ, Sharma R, Sharma M. A mouse model of prenatal exposure to Interleukin-6 to study the developmental origin of health and disease. Sci Rep 2021; 11:13260. [PMID: 34168254 PMCID: PMC8225793 DOI: 10.1038/s41598-021-92751-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022] Open
Abstract
Systemic inflammation in pregnant obese women is associated with 1.5- to 2-fold increase in serum Interleukin-6 (IL-6) and newborns with lower kidney/body weight ratio but the role of IL-6 in increased susceptibility to chronic kidney (CKD) in adult progeny is not known. Since IL-6 crosses the placental barrier, we administered recombinant IL-6 (10 pg/g) to pregnant mice starting at mid-gestation yielded newborns with lower body (p < 0.001) and kidney (p < 0.001) weights. Histomorphometry indicated decreased nephrogenic zone width (p = 0.039) with increased numbers of mature glomeruli (p = 0.002) and pre-tubular aggregates (p = 0.041). Accelerated maturation in IL-6 newborns was suggested by early expression of podocyte-specific protein podocin in glomeruli, increased 5-methyl-cytosine (LC–MS analysis for CpG DNA methylation) and altered expression of certain genes of cell-cycle and apoptosis (RT-qPCR array-analysis). Western blotting showed upregulated pJAK2/pSTAT3. Thus, treating dams with IL-6 as a surrogate provides newborns to study effects of maternal systemic inflammation on future susceptibility to CKD in adulthood.
Collapse
Affiliation(s)
- Tarak Srivastava
- Section of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City, 2401 Gillham Road, Kansas City, MO, 64108, USA. .,Midwest Veterans' Biomedical Research Foundation (MVBRF), Kansas City, MO, USA. .,Department of Oral and Craniofacial Sciences, University of Missouri at Kansas City-School of Dentistry, Kansas City, MO, USA.
| | - Trupti Joshi
- Department of Health Management and Informatics and MU Informatics Institute, University of Missouri, Columbia, MO, USA.,Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO, USA.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA.,MU Data Science and Informatics Institute, University of Missouri, Columbia, MO, USA
| | - Daniel P Heruth
- Children's Mercy Research Institute, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, MO, USA
| | - Mohammad H Rezaiekhaligh
- Section of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City, 2401 Gillham Road, Kansas City, MO, 64108, USA
| | - Robert E Garola
- Department of Pathology and Laboratory Medicine, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, MO, USA
| | - Jianping Zhou
- Midwest Veterans' Biomedical Research Foundation (MVBRF), Kansas City, MO, USA.,Kansas City VA Medical Center, Kansas City, MO, USA
| | - Varun C Boinpelly
- Midwest Veterans' Biomedical Research Foundation (MVBRF), Kansas City, MO, USA.,Kansas City VA Medical Center, Kansas City, MO, USA
| | - Mohammed Farhan Ali
- Section of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City, 2401 Gillham Road, Kansas City, MO, 64108, USA
| | - Uri S Alon
- Section of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City, 2401 Gillham Road, Kansas City, MO, 64108, USA
| | - Madhulika Sharma
- Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Gregory B Vanden Heuvel
- Department of Biomedical Sciences, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, USA
| | - Pramod Mahajan
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Drake University, Des Moines, IA, USA
| | - Lakshmi Priya
- Section of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City, 2401 Gillham Road, Kansas City, MO, 64108, USA
| | - Yuexu Jiang
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO, USA.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Ellen T McCarthy
- Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Virginia J Savin
- Kansas City VA Medical Center, Kansas City, MO, USA.,Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Ram Sharma
- Kansas City VA Medical Center, Kansas City, MO, USA
| | - Mukut Sharma
- Midwest Veterans' Biomedical Research Foundation (MVBRF), Kansas City, MO, USA.,Kansas City VA Medical Center, Kansas City, MO, USA.,Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
7
|
Srivastava T, Heruth DP, Duncan RS, Rezaiekhaligh MH, Garola RE, Priya L, Zhou J, Boinpelly VC, Novak J, Ali MF, Joshi T, Alon US, Jiang Y, McCarthy ET, Savin VJ, Sharma R, Johnson ML, Sharma M. Transcription Factor β-Catenin Plays a Key Role in Fluid Flow Shear Stress-Mediated Glomerular Injury in Solitary Kidney. Cells 2021; 10:cells10051253. [PMID: 34069476 PMCID: PMC8159099 DOI: 10.3390/cells10051253] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/06/2021] [Accepted: 05/14/2021] [Indexed: 01/21/2023] Open
Abstract
Increased fluid flow shear stress (FFSS) in solitary kidney alters podocyte function in vivo. FFSS-treated cultured podocytes show upregulated AKT-GSK3β-β-catenin signaling. The present study was undertaken to confirm (i) the activation of β-catenin signaling in podocytes in vivo using unilaterally nephrectomized (UNX) TOPGAL mice with the β-galactosidase reporter gene for β-catenin activation, (ii) β-catenin translocation in FFSS-treated mouse podocytes, and (iii) β-catenin signaling using publicly available data from UNX mice. The UNX of TOPGAL mice resulted in glomerular hypertrophy and increased the mesangial matrix consistent with hemodynamic adaptation. Uninephrectomized TOPGAL mice showed an increased β-galactosidase expression at 4 weeks but not at 12 weeks, as assessed using immunofluorescence microscopy (p < 0.001 at 4 weeks; p = 0.16 at 12 weeks) and X-gal staining (p = 0.008 at 4 weeks; p = 0.65 at 12 weeks). Immunofluorescence microscopy showed a significant increase in phospho-β-catenin (Ser552, p = 0.005) at 4 weeks but not at 12 weeks (p = 0.935) following UNX, and the levels of phospho-β-catenin (Ser675) did not change. In vitro FFSS caused a sustained increase in the nuclear translocation of phospho-β-catenin (Ser552) but not phospho-β-catenin (Ser675) in podocytes. The bioinformatic analysis of the GEO dataset, #GSE53996, also identified β-catenin as a key upstream regulator. We conclude that transcription factor β-catenin mediates FFSS-induced podocyte (glomerular) injury in solitary kidney.
Collapse
Affiliation(s)
- Tarak Srivastava
- Section of Nephrology, Children’s Mercy Hospital and University of Missouri at Kansas City, Kansas City, MO 64108, USA; (M.H.R.); (L.P.); (M.F.A.); (U.S.A.)
- Midwest Veterans’ Biomedical Research Foundation (MVBRF), Kansas City, MO 64128, USA; (J.Z.); (V.C.B.); (M.S.)
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri at Kansas City, Kansas City, MO 64108, USA;
- Correspondence: ; Tel.: +1-816-234-3010; Fax: +1-816-302-9919
| | - Daniel P. Heruth
- Children’s Mercy Research Institute, Children’s Mercy Hospital and University of Missouri at Kansas City, Kansas City, MO 64108, USA;
| | - R. Scott Duncan
- School of Biological Sciences, University of Missouri at Kansas City, Kansas City, MO 64108, USA;
| | - Mohammad H. Rezaiekhaligh
- Section of Nephrology, Children’s Mercy Hospital and University of Missouri at Kansas City, Kansas City, MO 64108, USA; (M.H.R.); (L.P.); (M.F.A.); (U.S.A.)
| | - Robert E. Garola
- Department of Pathology and Laboratory Medicine, Children’s Mercy Hospital and University of Missouri at Kansas City, Kansas City, MO 64108, USA;
| | - Lakshmi Priya
- Section of Nephrology, Children’s Mercy Hospital and University of Missouri at Kansas City, Kansas City, MO 64108, USA; (M.H.R.); (L.P.); (M.F.A.); (U.S.A.)
| | - Jianping Zhou
- Midwest Veterans’ Biomedical Research Foundation (MVBRF), Kansas City, MO 64128, USA; (J.Z.); (V.C.B.); (M.S.)
- Kansas City VA Medical Center, Kansas City, MO 64128, USA; (V.J.S.); (R.S.)
| | - Varun C. Boinpelly
- Midwest Veterans’ Biomedical Research Foundation (MVBRF), Kansas City, MO 64128, USA; (J.Z.); (V.C.B.); (M.S.)
- Kansas City VA Medical Center, Kansas City, MO 64128, USA; (V.J.S.); (R.S.)
| | - Jan Novak
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35487, USA;
| | - Mohammed Farhan Ali
- Section of Nephrology, Children’s Mercy Hospital and University of Missouri at Kansas City, Kansas City, MO 64108, USA; (M.H.R.); (L.P.); (M.F.A.); (U.S.A.)
| | - Trupti Joshi
- Department of Health Management and Informatics, University of Missouri, Columbia, MO 65211, USA;
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO 65211, USA;
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
- MU Data Science and Informatics Institute, University of Missouri, Columbia, MO 65211, USA
| | - Uri S. Alon
- Section of Nephrology, Children’s Mercy Hospital and University of Missouri at Kansas City, Kansas City, MO 64108, USA; (M.H.R.); (L.P.); (M.F.A.); (U.S.A.)
| | - Yuexu Jiang
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO 65211, USA;
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Ellen T. McCarthy
- Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - Virginia J. Savin
- Kansas City VA Medical Center, Kansas City, MO 64128, USA; (V.J.S.); (R.S.)
| | - Ram Sharma
- Kansas City VA Medical Center, Kansas City, MO 64128, USA; (V.J.S.); (R.S.)
| | - Mark L. Johnson
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri at Kansas City, Kansas City, MO 64108, USA;
| | - Mukut Sharma
- Midwest Veterans’ Biomedical Research Foundation (MVBRF), Kansas City, MO 64128, USA; (J.Z.); (V.C.B.); (M.S.)
- Kansas City VA Medical Center, Kansas City, MO 64128, USA; (V.J.S.); (R.S.)
- Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| |
Collapse
|
8
|
Sholokh A, Klussmann E. Local cyclic adenosine monophosphate signalling cascades-Roles and targets in chronic kidney disease. Acta Physiol (Oxf) 2021; 232:e13641. [PMID: 33660401 DOI: 10.1111/apha.13641] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 12/20/2022]
Abstract
The molecular mechanisms underlying chronic kidney disease (CKD) are poorly understood and treatment options are limited, a situation underpinning the need for elucidating the causative molecular mechanisms and for identifying innovative treatment options. It is emerging that cyclic 3',5'-adenosine monophosphate (cAMP) signalling occurs in defined cellular compartments within nanometre dimensions in processes whose dysregulation is associated with CKD. cAMP compartmentalization is tightly controlled by a specific set of proteins, including A-kinase anchoring proteins (AKAPs) and phosphodiesterases (PDEs). AKAPs such as AKAP18, AKAP220, AKAP-Lbc and STUB1, and PDE4 coordinate arginine-vasopressin (AVP)-induced water reabsorption by collecting duct principal cells. However, hyperactivation of the AVP system is associated with kidney damage and CKD. Podocyte injury involves aberrant AKAP signalling. cAMP signalling in immune cells can be local and slow the progression of inflammatory processes typical for CKD. A major risk factor of CKD is hypertension. cAMP directs the release of the blood pressure regulator, renin, from juxtaglomerular cells, and plays a role in Na+ reabsorption through ENaC, NKCC2 and NCC in the kidney. Mutations in the cAMP hydrolysing PDE3A that cause lowering of cAMP lead to hypertension. Another major risk factor of CKD is diabetes mellitus. AKAP18 and AKAP150 and several PDEs are involved in insulin release. Despite the increasing amount of data, an understanding of functions of compartmentalized cAMP signalling with relevance for CKD is fragmentary. Uncovering functions will improve the understanding of physiological processes and identification of disease-relevant aberrations may guide towards new therapeutic concepts for the treatment of CKD.
Collapse
Affiliation(s)
- Anastasiia Sholokh
- Max‐Delbrück‐Center for Molecular Medicine (MDC) Helmholtz Association Berlin Germany
| | - Enno Klussmann
- Max‐Delbrück‐Center for Molecular Medicine (MDC) Helmholtz Association Berlin Germany
- DZHK (German Centre for Cardiovascular Research) Berlin Germany
| |
Collapse
|
9
|
Oyagbemi AA, Bolaji-Alabi FB, Ajibade TO, Adejumobi OA, Ajani OS, Jarikre TA, Omobowale TO, Ola-Davies OE, Soetan KO, Aro AO, Emikpe BO, Saba AB, Adedapo AA, Oyeyemi MO, Nkadimeng SM, Kayoka-Kabongo PN, McGaw LJ, Oguntibeju OO, Yakubu MA. Novel antihypertensive action of rutin is mediated via inhibition of angiotensin converting enzyme/mineralocorticoid receptor/angiotensin 2 type 1 receptor (ATR1) signaling pathways in uninephrectomized hypertensive rats. J Food Biochem 2020; 44:e13534. [PMID: 33089540 DOI: 10.1111/jfbc.13534] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/18/2020] [Accepted: 10/02/2020] [Indexed: 12/01/2022]
Abstract
Hypertension is the most common cardiovascular disease that affects approximately 26% of adult population, worldwide. Rutin is one of the important flavonoids that is consumed in the daily diet, and found in many food items, vegetables, and beverages. Uninephrectomy (UNX) of the left kidney was performed, followed by induction of hypertension. The rats were randomly divided into four groups of 10 rats: group 1-Sham-operated rats; group 2-UNX rats, group 3-UNX-L-NAME (40 mg/kg) plus rutin (100 mg/kg bwt), and groups 4-UNX-L-NAME plus lisinopril (10 mg/kg bwt), orally for 3 weeks. Results revealed significant heightening of arterial pressure and oxidative stress indices, while hypertensive rats treated with rutin had lower expressions of angiotensin converting enzyme (ACE) and mineralocorticoid receptor in uninephrectomized rats. Together, rutin as a novel antihypertensive flavonoid could provide an unimaginable benefits for the management of hypertension through inhibition of angiotensin converting enzyme and mineralocorticoid receptor. PRACTICAL APPLICATIONS: Hypertension has been reported to be the most common cardiovascular disease, affecting approximately 26% of the adult population worldwide with predicted prevalence to increase by 60% by 2025. Recent advances in phytomedicine have shown flavonoids to be very helpful in the treatment of many diseases. Flavonoids have been used in the treatment and management of cardiovascular diseases, obesity and hypertension. The study revealed that rutin, a known flavonoid inhibited angiotensin converting enzyme (ACE), angiotensin 2 type 1 receptor (ATR1), and mineralocorticoid receptor (MCR), comparable to the classic ACE inhibitor, Lisinopril, indicating the novel antihypertensive property of rutin. Therefore, flavonoids such as rutin found in fruits and vegetables could, therefore, serve as an antihypertensive drug regimen. Combining all, functional foods rich in flavonoids could be used as potential therapeutic candidates for managing uninephrectomized hypertensive patients.
Collapse
Affiliation(s)
- Ademola Adetokunbo Oyagbemi
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Foluso Bolawaye Bolaji-Alabi
- Department of Veterinary Surgery and Radiology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Temitayo Olabisi Ajibade
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olumuyiwa Abiola Adejumobi
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olumide Samuel Ajani
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Theophilus Aghogho Jarikre
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Temidayo Olutayo Omobowale
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olufunke Eunice Ola-Davies
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Kehinde Olugboyega Soetan
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Abimbola Obemisola Aro
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Benjamin Obukowho Emikpe
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adebowale Benard Saba
- Department of Agriculture and Animal Health, College of Agriculture and Environmental Sciences, University of South Africa, Gauteng, South Africa
| | - Adeolu Alex Adedapo
- Department of Agriculture and Animal Health, College of Agriculture and Environmental Sciences, University of South Africa, Gauteng, South Africa
| | | | - Sanah Malomile Nkadimeng
- Phytomedicine Programme, Department of Paraclinical Science, University of Pretoria Faculty of Veterinary Science, Pretoria, South Africa
| | | | - Lyndy Joy McGaw
- Phytomedicine Programme, Department of Paraclinical Science, University of Pretoria Faculty of Veterinary Science, Pretoria, South Africa
| | - Oluwafemi Omoniyi Oguntibeju
- Phytomedicine and Phytochemistry Group, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Cape Town, South Africa
| | - Momoh Audu Yakubu
- Department of Environmental & Interdisciplinary Sciences, College of Science, Engineering & Technology, Vascular Biology Unit, Center for Cardiovascular Diseases, Texas Southern University, Houston, TX, USA
| |
Collapse
|
10
|
Srivastava T, Joshi T, Jiang Y, Heruth DP, Rezaiekhaligh MH, Novak J, Staggs VS, Alon US, Garola RE, El-Meanawy A, McCarthy ET, Zhou J, Boinpelly VC, Sharma R, Savin VJ, Sharma M. Upregulated proteoglycan-related signaling pathways in fluid flow shear stress-treated podocytes. Am J Physiol Renal Physiol 2020; 319:F312-F322. [PMID: 32628542 DOI: 10.1152/ajprenal.00183.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The ultrafiltrate flow over the major processes and cell body generates fluid flow shear stress (FFSS) on podocytes. Hyperfiltration-associated increase in FFSS can lead to podocyte injury and detachment. Previously, we showed that FFSS-induced upregulation of the cyclooxygenase 2 (COX2)-PGE2-prostaglandin E receptor 2 (EP2) axis in podocytes activates Akt-glycogen synthase kinase-3β-β-catenin and MAPK/ERK signaling in response to FFSS. Integrative MultiOmics Pathway Resolution (IMPRes) is a new bioinformatic tool that enables simultaneous time-series analysis of more than two groups to identify pathways and molecular connections. In the present study, we used previously characterized COX2 [prostaglandin-endoperoxide synthase 2 (Ptgs2)], EP2 (Ptger2), and β1-catenin (Ctnnb1) as "seed genes" from an array data set of four groups analyzed over a time course. The 3 seed genes shared 7 pathways and 50 genes of 14 pathways and 89 genes identified by IMPRes. A composite of signaling pathways highlighted the temporal molecular connections during mechanotransduction signaling in FFSS-treated podocytes. We investigated the "proteoglycans in cancer" and "galactose metabolism" pathways predicted by IMPRes. A custom-designed PCR array validated 60.7% of the genes predicted by IMPRes analysis, including genes for the above-named pathways. Further validation using Western blot analysis showed increased expression of phosho-Erbb2, phospho-mammalian target of rapamycin (mTOR), CD44, and hexokinase II (Hk2); decreased total Erbb2, galactose mutarotase (Galm), and β-1,4-galactosyltransferase 1 (B4galt1); and unchanged total mTOR and AKT3. These findings corroborate our previously reported results. This study demonstrates the potential of the IMPRes method to identify novel pathways. Identifying the "proteoglycans in cancer" and "galactose metabolism" pathways has generated a lead to study the significance of FFSS-induced glycocalyx remodeling and possible detachment of podocytes from the glomerular matrix.
Collapse
Affiliation(s)
- Tarak Srivastava
- Section of Nephrology, Children's Mercy Hospital and University of Missouri, Kansas City, Missouri.,Midwest Veterans' Biomedical Research Foundation, Kansas City, Missouri.,Department of Oral and Craniofacial Sciences, University of Missouri School of Dentistry, Kansas City, Missouri
| | - Trupti Joshi
- Department of Health Management and Informatics and University of Missouri Informatics Institute, University of Missouri, Columbia, Missouri.,Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, Missouri.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri.,MU Data Science and Informatics Institute, University of Missouri, Columbia, Missouri
| | - Yuexu Jiang
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, Missouri.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri
| | - Daniel P Heruth
- Children's Mercy Research Institute, Children's Mercy Hospital and University of Missouri, Kansas City, Missouri
| | - Mohamed H Rezaiekhaligh
- Section of Nephrology, Children's Mercy Hospital and University of Missouri, Kansas City, Missouri
| | - Jan Novak
- Department of Microbiology, University of Alabama, Birmingham, Alabama
| | - Vincent S Staggs
- Biostatistics and Epidemiology Core, Children's Mercy Kansas City, Department of Pediatrics, University of Missouri, Kansas City, Missouri
| | - Uri S Alon
- Section of Nephrology, Children's Mercy Hospital and University of Missouri, Kansas City, Missouri
| | - Robert E Garola
- Department of Pathology and Laboratory Medicine, Children's Mercy Hospital and University of Missouri, Kansas City
| | - Ashraf El-Meanawy
- Division of Nephrology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Ellen T McCarthy
- Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Jianping Zhou
- Midwest Veterans' Biomedical Research Foundation, Kansas City, Missouri.,Kansas City Veterans Affairs Medical Center, Kansas City, Missouri
| | - Varun C Boinpelly
- Midwest Veterans' Biomedical Research Foundation, Kansas City, Missouri.,Kansas City Veterans Affairs Medical Center, Kansas City, Missouri
| | - Ram Sharma
- Midwest Veterans' Biomedical Research Foundation, Kansas City, Missouri.,Kansas City Veterans Affairs Medical Center, Kansas City, Missouri
| | - Virginia J Savin
- Midwest Veterans' Biomedical Research Foundation, Kansas City, Missouri.,Kansas City Veterans Affairs Medical Center, Kansas City, Missouri
| | - Mukut Sharma
- Midwest Veterans' Biomedical Research Foundation, Kansas City, Missouri.,Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas.,Kansas City Veterans Affairs Medical Center, Kansas City, Missouri
| |
Collapse
|
11
|
McArdle Z, Schreuder MF, Moritz KM, Denton KM, Singh RR. Physiology and Pathophysiology of Compensatory Adaptations of a Solitary Functioning Kidney. Front Physiol 2020; 11:725. [PMID: 32670095 PMCID: PMC7332829 DOI: 10.3389/fphys.2020.00725] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/03/2020] [Indexed: 12/15/2022] Open
Abstract
Children born with a solitary functioning kidney (SFK) have an increased risk of hypertension and kidney disease from early in adulthood. In response to a reduction in kidney mass, the remaining kidney undergoes compensatory kidney growth. This is associated with both an increase in size of the kidney tubules and the glomeruli and an increase in single nephron glomerular filtration rate (SNGFR). The compensatory hypertrophy and increase in filtration at the level of the individual nephron results in normalization of total glomerular filtration rate (GFR). However, over time these same compensatory mechanisms may contribute to kidney injury and hypertension. Indeed, approximately 50% of children born with a SFK develop hypertension by the age of 18 and 20–40% require dialysis by the age of 30. The mechanisms that result in kidney injury are only partly understood, and early biomarkers that distinguish those at an elevated risk of kidney injury are needed. This review will outline the compensatory adaptations to a SFK, and outline how these adaptations may contribute to kidney injury and hypertension later in life. These will be based largely on the mechanisms we have identified from our studies in an ovine model of SFK, that implicate the renal nitric oxide system, the renin angiotensin system and the renal nerves to kidney disease and hypertension associated with SFK. This discussion will also evaluate current, and speculate on next generation, prognostic factors that may predict those children at a higher risk of future kidney disease and hypertension.
Collapse
Affiliation(s)
- Zoe McArdle
- Cardiovascular Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, VIC, Australia
| | - Michiel F Schreuder
- Department of Pediatric Nephrology, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, Netherlands
| | - Karen M Moritz
- Child Health Research Centre and School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
| | - Kate M Denton
- Cardiovascular Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, VIC, Australia
| | - Reetu R Singh
- Cardiovascular Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
12
|
Concerted EP2 and EP4 Receptor Signaling Stimulates Autocrine Prostaglandin E 2 Activation in Human Podocytes. Cells 2020; 9:cells9051256. [PMID: 32438662 PMCID: PMC7290667 DOI: 10.3390/cells9051256] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/29/2020] [Accepted: 05/14/2020] [Indexed: 12/14/2022] Open
Abstract
Glomerular hyperfiltration is an important mechanism in the development of albuminuria. During hyperfiltration, podocytes are exposed to increased fluid flow shear stress (FFSS) in Bowman’s space. Elevated Prostaglandin E2 (PGE2) synthesis and upregulated cyclooxygenase 2 (Cox2) are associated with podocyte injury by FFSS. We aimed to elucidate a PGE2 autocrine/paracrine pathway in human podocytes (hPC). We developed a modified liquid chromatography tandem mass spectrometry (LC/ESI-MS/MS) protocol to quantify cellular PGE2, 15-keto-PGE2, and 13,14-dihydro-15-keto-PGE2 levels. hPC were treated with PGE2 with or without separate or combined blockade of prostaglandin E receptors (EP), EP2, and EP4. Furthermore, the effect of FFSS on COX2, PTGER2, and PTGER4 expression in hPC was quantified. In hPC, stimulation with PGE2 led to an EP2- and EP4-dependent increase in cyclic adenosine monophosphate (cAMP) and COX2, and induced cellular PGE2. PTGER4 was downregulated after PGE2 stimulation in hPC. In the corresponding LC/ESI-MS/MS in vivo analysis at the tissue level, increased PGE2 and 15-keto-PGE2 levels were observed in isolated glomeruli obtained from a well-established rat model with glomerular hyperfiltration, the Munich Wistar Frömter rat. COX2 and PTGER2 were upregulated by FFSS. Our data thus support an autocrine/paracrine COX2/PGE2 pathway in hPC linked to concerted EP2 and EP4 signaling.
Collapse
|
13
|
Srivastava T, Ju W, Milne GL, Rezaiekhaligh MH, Staggs VS, Alon US, Sharma R, Zhou J, El-Meanawy A, McCarthy ET, Savin VJ, Sharma M. Urinary prostaglandin E 2 is a biomarker of early adaptive hyperfiltration in solitary functioning kidney. Prostaglandins Other Lipid Mediat 2019; 146:106403. [PMID: 31838197 DOI: 10.1016/j.prostaglandins.2019.106403] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 11/20/2019] [Accepted: 12/10/2019] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Hyperfiltration is a major contributor to progression of chronic kidney disease (CKD) in diabetes, obesity and in individuals with solitary functioning kidney (SFK). We have proposed hyperfiltration-induced injury as a continuum of overlapping glomerular changes caused by increased biomechanical forces namely, fluid flow shear stress (FFSS) and tensile stress. We have shown that FFSS is elevated in animals with SFK and, it upregulates prostaglandin E2 (PGE2), cyclooxygenase-2 and PGE2 receptor EP2 in cultured podocytes and in uninephrectomized mice. We conceptualized urinary PGE2 as a biomarker of early effects of hyperfiltration-induced injury preceding microalbuminuria in individuals with SFK. We studied children with SFK to validate our hypothesis. METHODS Urine samples from children with SFK and controls were analyzed for PGE2, albumin (glomerular injury biomarker) and epidermal growth factor (EGF, tubular injury biomarker). Age, gender, and Z-scores for height, weight, BMI, and blood pressure were obtained. RESULTS Children with SFK were comparable to controls except for lower BMI Z-scores. The median values were elevated in SFK compared to control for urine PGE2 [9.1 (n = 57) vs. 5.7 (n = 72), p = 0.009] ng/mgCr and albumin [7.6 (n = 40) vs. 7.0 (n = 41), p = 0.085] μg/mgCr, but not for EGF [20098 (n = 44) vs. 18637 (n = 44), p = 0.746] pg/mgCr. Significant increase in urinary PGE2 (p = 0.024) and albumin (p = 0.019) but not EGF (p = 0.412) was observed using additional regression modeling. These three urinary analytes were independent of each other. CONCLUSION Increased urinary PGE2 from elevated SNGFR and consequently increased FFSS during early stage of CKD precedes overt microalbuminuria and is a biomarker for early hyperfiltration-induced injury in individuals with SFK.
Collapse
Affiliation(s)
- Tarak Srivastava
- Division of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, MO, United States; Research and Development, Nephrology, Kansas City VA Medical Center, Kansas City, MO, United States.
| | - Wenjun Ju
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Ginger L Milne
- Division of Clinical Pharmacology, Vanderbilt University, Nashville, TN, United States
| | - Mohamed H Rezaiekhaligh
- Division of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, MO, United States
| | - Vincent S Staggs
- Biostatistics & Epidemiology Core, Children's Mercy Kansas City, Department of Pediatrics, University of Missouri, Kansas City, United States
| | - Uri S Alon
- Division of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, MO, United States
| | - Ram Sharma
- Research and Development, Nephrology, Kansas City VA Medical Center, Kansas City, MO, United States
| | - Jianping Zhou
- Research and Development, Nephrology, Kansas City VA Medical Center, Kansas City, MO, United States; Midwest Biomedical Research Foundation (MBRF), KCVA Medical Center, Kansas City, MO, United States
| | - Ashraf El-Meanawy
- Division of Nephrology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Ellen T McCarthy
- Kidney Institute, Kansas University Medical Center, Kansas City, KS, United States
| | - Virginia J Savin
- Research and Development, Nephrology, Kansas City VA Medical Center, Kansas City, MO, United States; Kidney Institute, Kansas University Medical Center, Kansas City, KS, United States
| | - Mukut Sharma
- Research and Development, Nephrology, Kansas City VA Medical Center, Kansas City, MO, United States; Midwest Biomedical Research Foundation (MBRF), KCVA Medical Center, Kansas City, MO, United States; Kidney Institute, Kansas University Medical Center, Kansas City, KS, United States
| |
Collapse
|
14
|
Arachidonic Acid Metabolism and Kidney Inflammation. Int J Mol Sci 2019; 20:ijms20153683. [PMID: 31357612 PMCID: PMC6695795 DOI: 10.3390/ijms20153683] [Citation(s) in RCA: 184] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 07/19/2019] [Accepted: 07/20/2019] [Indexed: 12/17/2022] Open
Abstract
As a major component of cell membrane lipids, Arachidonic acid (AA), being a major component of the cell membrane lipid content, is mainly metabolized by three kinds of enzymes: cyclooxygenase (COX), lipoxygenase (LOX), and cytochrome P450 (CYP450) enzymes. Based on these three metabolic pathways, AA could be converted into various metabolites that trigger different inflammatory responses. In the kidney, prostaglandins (PG), thromboxane (Tx), leukotrienes (LTs) and hydroxyeicosatetraenoic acids (HETEs) are the major metabolites generated from AA. An increased level of prostaglandins (PGs), TxA2 and leukotriene B4 (LTB4) results in inflammatory damage to the kidney. Moreover, the LTB4-leukotriene B4 receptor 1 (BLT1) axis participates in the acute kidney injury via mediating the recruitment of renal neutrophils. In addition, AA can regulate renal ion transport through 19-hydroxystilbenetetraenoic acid (19-HETE) and 20-HETE, both of which are produced by cytochrome P450 monooxygenase. Epoxyeicosatrienoic acids (EETs) generated by the CYP450 enzyme also plays a paramount role in the kidney damage during the inflammation process. For example, 14 and 15-EET mitigated ischemia/reperfusion-caused renal tubular epithelial cell damage. Many drug candidates that target the AA metabolism pathways are being developed to treat kidney inflammation. These observations support an extraordinary interest in a wide range of studies on drug interventions aiming to control AA metabolism and kidney inflammation.
Collapse
|
15
|
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are among the most widely used therapeutic class in clinical medicine. These are sub-divided based on their selectivity for inhibition of cyclooxygenase (COX) isoforms (COX-1 and COX-2) into: (1) non-selective (ns-NSAIDs), and (2) selective NSAIDs (s-NSAIDs) with preferential inhibition of COX-2 isozyme. The safety and pathophysiology of NSAIDs on the renal and cardiovascular systems have continued to evolve over the years following short- and long-term treatment in both preclinical models and humans. This review summarizes major learnings on cardiac and renal complications associated with pharmaceutical inhibition of COX-1 and COX-2 with focus on preclinical to clinical translatability of cardio-renal data.
Collapse
Affiliation(s)
- Zaher A Radi
- Drug Safety Research & Development, Pfizer Research, Development & Medical, Cambridge, USA
| | - K Nasir Khan
- Drug Safety Research & Development, Pfizer Research, Development & Medical, Cambridge, USA
| |
Collapse
|
16
|
Abstract
Kidney donors face a small but definite risk of end-stage renal disease 15 to 30 years postdonation. The development of proteinuria, hypertension with gradual decrease in kidney function in the donor after surgical resection of 1 kidney, has been attributed to hyperfiltration. Genetic variations, physiological adaptations, and comorbidities exacerbate the hyperfiltration-induced loss of kidney function in the years after donation. A focus on glomerular hemodynamics and capillary pressure has led to the development of drugs that target the renin-angiotensin-aldosterone system (RAAS), but these agents yield mixed results in transplant recipients and donors. Recent work on glomerular biomechanical forces highlights the differential effects of tensile stress and fluid flow shear stress (FFSS) from hyperfiltration. Capillary wall stretch due to glomerular capillary pressure increases tensile stress on podocyte foot processes that cover the capillary. In parallel, increased flow of the ultrafiltrate due to single-nephron glomerular filtration rate elevates FFSS on the podocyte cell body. Although tensile stress invokes the RAAS, FFSS predominantly activates the cyclooxygenase 2-prostaglandin E2-EP2 receptor axis. Distinguishing these 2 mechanisms is critical, as current therapeutic approaches focus on the RAAS system. A better understanding of the biomechanical forces can lead to novel therapeutic agents to target FFSS through the cyclooxygenase 2-prostaglandin E2-EP2 receptor axis in hyperfiltration-mediated injury. We present an overview of several aspects of the risk to transplant donors and discuss the relevance of FFSS in podocyte injury, loss of glomerular barrier function leading to albuminuria and gradual loss of renal function, and potential therapeutic strategies to mitigate hyperfiltration-mediated injury to the remaining kidney.
Collapse
|
17
|
Li Y, Xia W, Zhao F, Wen Z, Zhang A, Huang S, Jia Z, Zhang Y. Prostaglandins in the pathogenesis of kidney diseases. Oncotarget 2018; 9:26586-26602. [PMID: 29899878 PMCID: PMC5995175 DOI: 10.18632/oncotarget.25005] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 03/14/2018] [Indexed: 12/11/2022] Open
Abstract
Prostaglandins (PGs) are important lipid mediators produced from arachidonic acid via the sequential catalyzation of cyclooxygenases (COXs) and specific prostaglandin synthases. There are five subtypes of PGs, namely PGE2, PGI2, PGD2, PGF2α, and thromboxane A2 (TXA2). PGs exert distinct roles by combining to a diverse family of membrane-spanning G protein-coupled prostanoid receptors. The distribution of these PGs, their specific synthases and receptors vary a lot in the kidney. This review summarized the recent findings of PGs together with the COXs and their specific synthases and receptors in regulating renal function and highlighted the insights into their roles in the pathogenesis of various kidney diseases.
Collapse
Affiliation(s)
- Yuanyuan Li
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Weiwei Xia
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Fei Zhao
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Zhaoying Wen
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Aihua Zhang
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Songming Huang
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Zhanjun Jia
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Yue Zhang
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| |
Collapse
|
18
|
Srivastava T, Thiagarajan G, Alon US, Sharma R, El-Meanawy A, McCarthy ET, Savin VJ, Sharma M. Role of biomechanical forces in hyperfiltration-mediated glomerular injury in congenital anomalies of the kidney and urinary tract. Nephrol Dial Transplant 2018; 32:759-765. [PMID: 28339567 DOI: 10.1093/ndt/gfw430] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 11/15/2016] [Indexed: 11/13/2022] Open
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) including solitary kidney constitute the main cause of progressive chronic kidney disease (CKD) in children. Children born with CAKUT develop signs of CKD only during adolescence and do not respond to renin-angiotensin-aldosterone system blockers. Early cellular changes underlying CKD progression to end-stage renal disease by early adulthood are not well understood. The mechanism of maladaptive hyperfiltration that occurs from loss of functional nephrons, including solitary kidney, is not clear. We re-examine the phenomenon of hyperfiltration in the context of biomechanical forces with special reference to glomerular podocytes. Capillary stretch exerts tensile stress on podocytes through the glomerular basement membrane. The flow of ultrafiltrate over the cell surface directly causes fluid flow shear stress (FFSS) on podocytes. FFSS on the podocyte surface increases 1.5- to 2-fold in animal models of solitary kidney and its effect on podocytes is a subject of ongoing research. Podocytes (i) are mechanosensitive to tensile and shear forces, (ii) use prostaglandin E2, angiotensin-II or nitric oxide for mechanoperception and (iii) use specific signaling pathways for mechanotransduction. We discuss (i) the nature of and differences in cellular responses to biomechanical forces, (ii) methods to study biomechanical forces and (iii) effects of biomechanical forces on podocytes and glomeruli. Future studies on FFSS will likely identify novel targets for strategies for early intervention to complement and strengthen the current regimen for treating children with CAKUT.
Collapse
Affiliation(s)
- Tarak Srivastava
- Section of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, MO, USA.,Renal Research Laboratory, Research and Development, Kansas City VA Medical Center, Kansas City, MO, USA
| | - Ganesh Thiagarajan
- School of Computing and Engineering, University of Missouri at Kansas City, MO, USA
| | - Uri S Alon
- Section of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, MO, USA
| | - Ram Sharma
- Renal Research Laboratory, Research and Development, Kansas City VA Medical Center, Kansas City, MO, USA
| | - Ashraf El-Meanawy
- Division of Nephrology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ellen T McCarthy
- Kidney Institute, Kansas University Medical Center, Kansas City, KS, USA
| | - Virginia J Savin
- Renal Research Laboratory, Research and Development, Kansas City VA Medical Center, Kansas City, MO, USA
| | - Mukut Sharma
- Renal Research Laboratory, Research and Development, Kansas City VA Medical Center, Kansas City, MO, USA
| |
Collapse
|
19
|
Abstract
Life with a solitary functioning kidney (SFK) may be different from that when born with two kidneys. Based on the hyperfiltration hypothesis, a SFK may lead to glomerular damage with hypertension, albuminuria and progression towards end-stage renal disease. As the prognosis of kidney donors was considered to be very good, having a SFK has been considered to be a benign condition. In contrast, our research group has demonstrated that being born with or acquiring a SFK in childhood results in renal injury before adulthood in over 50% of those affected. Most congenital cases will be detected during antenatal ultrasound screening, but up to 38% of cases of unilateral renal agenesis are missed. In about 25-50% of cases of antenatally detected SFK there will be signs of hypertrophy, which could indicate additional nephron formation and is associated with a somewhat reduced risk of renal injury. Additional renal and extrarenal anomalies are frequently detected and may denote a genetic cause for the SFK, even though for the majority of cases no explanation can (yet) be found. The ongoing glomerular hyperfiltration results in renal injury, for which early markers are lacking. Individuals with SFK should avoid obesity and excessive salt intake to limit additional hyperfiltration. As conditions like hypertension, albuminuria and a mildly reduced glomerular filtration rate generally do not result in specific complaints but may pose a threat to long-term health, screening for renal injury in any individual with a SFK would appear to be imperative, starting from infancy. With early treatment, secondary consequences may be diminished, thereby providing the optimal life for anyone born with a SFK.
Collapse
|
20
|
Srivastava T, Dai H, Heruth DP, Alon US, Garola RE, Zhou J, Duncan RS, El-Meanawy A, McCarthy ET, Sharma R, Johnson ML, Savin VJ, Sharma M. Mechanotransduction signaling in podocytes from fluid flow shear stress. Am J Physiol Renal Physiol 2018; 314:F22-F34. [PMID: 28877882 PMCID: PMC5866353 DOI: 10.1152/ajprenal.00325.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 08/21/2017] [Accepted: 08/28/2017] [Indexed: 12/28/2022] Open
Abstract
Recently, we and others have found that hyperfiltration-associated increase in biomechanical forces, namely, tensile stress and fluid flow shear stress (FFSS), can directly and distinctly alter podocyte structure and function. The ultrafiltrate flow over the major processes and cell body generates FFSS to podocytes. Our previous work suggests that the cyclooxygenase-2 (COX-2)-PGE2-PGE2 receptor 2 (EP2) axis plays an important role in mechanoperception of FFSS in podocytes. To address mechanotransduction of the perceived stimulus through EP2, cultured podocytes were exposed to FFSS (2 dyn/cm2) for 2 h. Total RNA from cells at the end of FFSS treatment, 2-h post-FFSS, and 24-h post-FFSS was used for whole exon array analysis. Differentially regulated genes ( P < 0.01) were analyzed using bioinformatics tools Enrichr and Ingenuity Pathway Analysis to predict pathways/molecules. Candidate pathways were validated using Western blot analysis and then further confirmed to be resulting from a direct effect of PGE2 on podocytes. Results show that FFSS-induced mechanotransduction as well as exogenous PGE2 activate the Akt-GSK3β-β-catenin (Ser552) and MAPK/ERK but not the cAMP-PKA signal transduction cascades. These pathways are reportedly associated with FFSS-induced and EP2-mediated signaling in other epithelial cells as well. The current regimen for treating hyperfiltration-mediated injury largely depends on targeting the renin-angiotensin-aldosterone system. The present study identifies specific transduction mechanisms and provides novel information on the direct effect of FFSS on podocytes. These results suggest that targeting EP2-mediated signaling pathways holds therapeutic significance for delaying progression of chronic kidney disease secondary to hyperfiltration.
Collapse
Affiliation(s)
- Tarak Srivastava
- Section of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City , Kansas City, Missouri
- Renal Research Laboratory, Research and Development, Kansas City Veterans Affairs Medical Center , Kansas City, Missouri
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri at Kansas City , Kansas City, Missouri
| | - Hongying Dai
- Section of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City , Kansas City, Missouri
| | - Daniel P Heruth
- Department of Experimental and Translational Genetics Research, Children's Mercy Hospital and University of Missouri at Kansas City , Kansas City, Missouri
| | - Uri S Alon
- Section of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City , Kansas City, Missouri
| | - Robert E Garola
- Department of Pathology and Laboratory Medicine, Children's Mercy Hospital and University of Missouri at Kansas City , Kansas City, Missouri
| | - Jianping Zhou
- Renal Research Laboratory, Research and Development, Kansas City Veterans Affairs Medical Center , Kansas City, Missouri
| | - R Scott Duncan
- Department of Ophthalmology, University of Missouri at Kansas City , Kansas City, Missouri
| | - Ashraf El-Meanawy
- Division of Nephrology, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Ellen T McCarthy
- Kidney Institute, University of Kansas Medical Center , Kansas City, Kansas
| | - Ram Sharma
- Renal Research Laboratory, Research and Development, Kansas City Veterans Affairs Medical Center , Kansas City, Missouri
| | - Mark L Johnson
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri at Kansas City , Kansas City, Missouri
| | - Virginia J Savin
- Renal Research Laboratory, Research and Development, Kansas City Veterans Affairs Medical Center , Kansas City, Missouri
- Kidney Institute, University of Kansas Medical Center , Kansas City, Kansas
| | - Mukut Sharma
- Renal Research Laboratory, Research and Development, Kansas City Veterans Affairs Medical Center , Kansas City, Missouri
- Kidney Institute, University of Kansas Medical Center , Kansas City, Kansas
| |
Collapse
|
21
|
Endlich K, Kliewe F, Endlich N. Stressed podocytes-mechanical forces, sensors, signaling and response. Pflugers Arch 2017; 469:937-949. [PMID: 28687864 DOI: 10.1007/s00424-017-2025-8] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 06/22/2017] [Indexed: 02/07/2023]
Abstract
Increased glomerular capillary pressure (glomerular hypertension) and increased glomerular filtration rate (glomerular hyperfiltration) have been proven to cause glomerulosclerosis in animal models and are likely to be operative in patients. Since podocytes cover the glomerular basement membrane, they are exposed to tensile stress due to circumferential wall tension and to fluid shear stress arising from filtrate flow through the narrow filtration slits and through Bowman's space. In vitro evidence documents that podocytes respond to tensile stress as well as to fluid shear stress. Several proteins are discussed in this review that are expressed in podocytes and could act as mechanosensors converting mechanical force via a conformational change into a biochemical signal. The cation channels P2X4 and TRPC6 were shown to be involved in mechanosignaling in podocytes. P2X4 is activated by stretch-induced ATP release, while TRPC6 might be inherently mechanosensitive. Membrane, slit diaphragm and cell-matrix contact proteins are connected to the sublemmal actin network in podocytes via various linker proteins. Therefore, actin-associated proteins, like the proven mechanosensor filamin, are ideal candidates to sense forces in the podocyte cytoskeleton. Furthermore, podocytes express talin, p130Cas, and fibronectin that are known to undergo a conformational change in response to mechanical force exposing cryptic binding sites. Downstream of mechanosensors, experimental evidence suggests the involvement of MAP kinases, Ca2+ and COX2 in mechanosignaling and an emerging role of YAP/TAZ. In summary, our understanding of mechanotransduction in podocytes is still sketchy, but future progress holds promise to identify targets to alleviate conditions of increased mechanical load.
Collapse
Affiliation(s)
- Karlhans Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, 17489, Greifswald, Germany.
- Institut für Anatomie and Zellbiologie, Universitätsmedizin Greifswald, Friedrich-Loeffler-Str. 23c, 17489, Greifswald, Germany.
| | - Felix Kliewe
- Department of Anatomy and Cell Biology, University Medicine Greifswald, 17489, Greifswald, Germany
| | - Nicole Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, 17489, Greifswald, Germany
| |
Collapse
|
22
|
Mohammed SG, Arjona FJ, Latta F, Bindels RJM, Roepman R, Hoenderop JGJ. Fluid shear stress increases transepithelial transport of Ca
2+
in ciliated distal convoluted and connecting tubule cells. FASEB J 2017; 31:1796-1806. [DOI: 10.1096/fj.201600687rrr] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 01/03/2017] [Indexed: 11/11/2022]
Affiliation(s)
- Sami G. Mohammed
- Department of PhysiologyRadboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Francisco J. Arjona
- Department of PhysiologyRadboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Femke Latta
- Department of PhysiologyRadboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - René J. M. Bindels
- Department of PhysiologyRadboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Ronald Roepman
- Department of Human GeneticsRadboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Joost G. J. Hoenderop
- Department of PhysiologyRadboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
| |
Collapse
|
23
|
Hyperfiltration-associated biomechanical forces in glomerular injury and response: Potential role for eicosanoids. Prostaglandins Other Lipid Mediat 2017; 132:59-68. [PMID: 28108282 DOI: 10.1016/j.prostaglandins.2017.01.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 12/22/2016] [Accepted: 01/10/2017] [Indexed: 12/29/2022]
Abstract
Hyperfiltration is a well-known risk factor in progressive loss of renal function in chronic kidney disease (CKD) secondary to various diseases. A reduced number of functional nephrons due to congenital or acquired cause(s) results in hyperfiltration in the remnant kidney. Hyperfiltration-associated increase in biomechanical forces, namely pressure-induced tensile stress and fluid flow-induced shear stress (FFSS) determine cellular injury and response. We believe the current treatment of CKD yields limited success because it largely attenuates pressure-induced tensile stress changes but not the effect of FFSS on podocytes. Studies on glomerular podocytes, tubular epithelial cells and bone osteocytes provide evidence for a significant role of COX-2 generated PGE2 and its receptors in response to tensile stress and FFSS. Preliminary observations show increased urinary PGE2 in children born with a solitary kidney. FFSS-induced COX2-PGE2-EP2 signaling provides an opportunity to identify targets and, for developing novel agents to complement currently available treatment.
Collapse
|
24
|
Zhong F, Mallipattu SK, Estrada C, Menon M, Salem F, Jain MK, Chen H, Wang Y, Lee K, He JC. Reduced Krüppel-Like Factor 2 Aggravates Glomerular Endothelial Cell Injury and Kidney Disease in Mice with Unilateral Nephrectomy. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2021-2031. [PMID: 27317905 DOI: 10.1016/j.ajpath.2016.03.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 02/18/2016] [Accepted: 03/29/2016] [Indexed: 12/13/2022]
Abstract
Loss of functional nephrons induces compensatory glomerular hyperfiltration and hypertrophy, leading to the progression of chronic kidney disease. Krüppel-like factor 2 (KLF2), a shear-stress-inducible transcription factor, confers protection against endothelial injury. Because glomerular hyperfiltration is associated with shear stress, we hypothesized that KLF2 may be an important factor in the compensatory response to unilateral nephrectomy (UNX). To test this hypothesis, endothelial cell-specific Klf2 heterozygous knockout mice (KO) and their wild-type littermate control (WT) underwent either UNX or sham-operation. WT-UNX mice developed compensatory renal hypertrophy as expected, whereas KO-UNX mice did not. KO-UNX mice exhibited higher blood pressure, reduced glomerular filtration rate, and significant increase in proteinuria and glomerulosclerosis compared to WT-UNX. Expression of endothelial nitric oxide synthase (official name Nos3), a known transcriptional target gene of KLF2, was significantly reduced and dysregulation of other endothelial genes was also observed in the glomeruli of KO-UNX when compared to WT-UNX and sham-operated mice. Furthermore, both podocyte number and expression of podocyte markers were also significantly reduced in KO-UNX glomeruli, indicating a potential cross talk between glomerular endothelial cells and podocytes. Finally, decreased renal expression of KLF2 in nephrectomy patients was associated with the progression of kidney disease. Taken together, our data demonstrate a protective role of KLF2 against glomerular endothelial cell injury and progression of chronic kidney disease in the model of compensatory renal hypertrophy.
Collapse
Affiliation(s)
- Fang Zhong
- Department of Medicine/Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Nephrology, Hang Zhou Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Sandeep K Mallipattu
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York
| | - Chelsea Estrada
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York
| | - Madhav Menon
- Department of Medicine/Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Fadi Salem
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Mukesh K Jain
- Department of Medicine, Case Cardiovascular Institute Research Institute, Case Western Reserve University, Cleveland, Ohio
| | - Hongyu Chen
- Department of Nephrology, Hang Zhou Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yongjun Wang
- Department of Nephrology, Hang Zhou Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Kyung Lee
- Department of Medicine/Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - John C He
- Department of Medicine/Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York; Renal Section, James J. Peters Veterans Affairs Medical Center, Bronx, New York.
| |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW The kidney mediates the excretion or conservation of water and electrolytes in the face of changing fluid and salt intake and losses. To ultrafilter and reabsorb the exact quantities of free water and salts to maintain euvolemia a range of endocrine, paracrine, and hormonal signaling systems have evolved linking the tubules, capillaries, glomeruli, arterioles, and other intrinsic cells of the kidney. Our understanding of these systems remains incomplete. RECENT FINDINGS Recent work has provided new insights into the workings of the communication pathways between tubular segments and the glomeruli and vasculature, with novel therapeutic agents in development. Particular progress has also been made in the visualization of tubuloglomerular feedback. SUMMARY The review summarizes our current understanding of pathway functions in health and disease, as well as future therapeutic options to protect the healthy and injured kidney.
Collapse
Affiliation(s)
- David A. Ferenbach
- Department of Medicine, Renal Division and Biomedical Engineering Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Joseph V. Bonventre
- Department of Medicine, Renal Division and Biomedical Engineering Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Harvard-Massachusetts Institute of Technology, Division of Health Sciences and Technology, Cambridge, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
26
|
Role of COX-2/mPGES-1/prostaglandin E2 cascade in kidney injury. Mediators Inflamm 2015; 2015:147894. [PMID: 25729216 PMCID: PMC4333324 DOI: 10.1155/2015/147894] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 01/19/2015] [Indexed: 12/26/2022] Open
Abstract
COX-2/mPGES-1/PGE2 cascade plays critical roles in modulating many physiological and pathological actions in different organs. In the kidney, this cascade is of high importance in regulating fluid metabolism, blood pressure, and renal hemodynamics. Under some disease conditions, this cascade displays various actions in response to the different pathological insults. In the present review, the roles of this cascade in the pathogenesis of kidney injuries including diabetic and nondiabetic kidney diseases and acute kidney injuries were introduced and discussed. The new insights from this review not only increase the understanding of the pathological role of the COX-2/mPGES-1/PGE2 pathway in kidney injuries, but also shed new light on the innovation of the strategies for the treatment of kidney diseases.
Collapse
|