1
|
Suarez R, Villarreal C, Nahuelpán Y, Jara C, Oyarzún C, Alarcón S, Díaz-Encarnación MM, Guillén-Gómez E, Quezada C, San Martín R. Defective insulin-stimulated equilibrative nucleoside transporter-2 activity and altered subcellular transporter distribution drive the loss of adenosine homeostasis in diabetic kidney disease progression. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166890. [PMID: 37734469 DOI: 10.1016/j.bbadis.2023.166890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 08/23/2023] [Accepted: 09/16/2023] [Indexed: 09/23/2023]
Abstract
AIM Progression of diabetic nephropathy (DN) is linked to the dysregulated increase of adenosine and altered signaling properties. A major contribution to the maintenance of physiological extracellular adenosine levels relies on cellular uptake activity through plasma membrane nucleoside transporters. Because kidney cells are responsive to insulin, this study aims to determine how DN affects insulin regulation of the equilibrative nucleoside transporter-2 (ENT2). METHODS Human Podocytes and rat glomeruli were used to study ENT2 regulation. The effects of diabetes and insulin on ENT2 mediated transport activity were determined measuring the fraction of total adenosine uptake in sodium-free medium which is inhibitable by hypoxanthine. Alterations in ENT2 subcellular distribution were assessed in the kidney of people affected with DN and diabetic rats. The consequences of impaired ENT2 activity on the kidney were evaluated using dipyridamole in an animal model. RESULTS Insulin upregulates ENT2 uptake activity by increasing the Vmax, thus counteracting decreased adenosine uptake due to high d-glucose and achieving extracellular adenosine homeostasis. Insulin promoted ENT2 translocation to the plasma membrane dependent on PI3-kinase/Akt signaling and actin cytoskeleton integrity. However, in diabetic rats, the insulin-mediated induction of ENT2 activity was lost. Additionally, reduced Akt activation in response to insulin correlated with decreased ENT2 distribution at the plasma membrane. Kidney tissues from diabetic rats and human DN biopsies showed ENT2 redistribution to an intracellular pattern, evidencing dysfunctional adenosine uptake. Through ENT inhibition, we evidenced increased proteinuria and induced alpha-smooth muscle actin as a result of profibrotic activation of cells in the kidney. CONCLUSION Deficient insulin regulation of ENT2 activity contributes to chronically high adenosine levels and glomerular alterations that underline diabetic kidney disease progression.
Collapse
Affiliation(s)
- Raibel Suarez
- Institute of Biochemistry and Microbiology, Science Faculty, Universidad Austral de Chile, Valdivia, Chile
| | - Carolina Villarreal
- Institute of Biochemistry and Microbiology, Science Faculty, Universidad Austral de Chile, Valdivia, Chile
| | - Yessica Nahuelpán
- Institute of Biochemistry and Microbiology, Science Faculty, Universidad Austral de Chile, Valdivia, Chile
| | - Claudia Jara
- Institute of Biochemistry and Microbiology, Science Faculty, Universidad Austral de Chile, Valdivia, Chile
| | - Carlos Oyarzún
- Institute of Biochemistry and Microbiology, Science Faculty, Universidad Austral de Chile, Valdivia, Chile
| | - Sebastián Alarcón
- Institute of Biochemistry and Microbiology, Science Faculty, Universidad Austral de Chile, Valdivia, Chile
| | - Montserrat M Díaz-Encarnación
- Nephrology Service Fundació Puigvert, Biomedical Research Institute Sant Pau (IIB Sant Pau), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Elena Guillén-Gómez
- Nephrology Service Fundació Puigvert, Biomedical Research Institute Sant Pau (IIB Sant Pau), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Claudia Quezada
- Institute of Biochemistry and Microbiology, Science Faculty, Universidad Austral de Chile, Valdivia, Chile; Millennium Institute on Immunology and Immunotherapy, Valdivia, Chile
| | - Rody San Martín
- Institute of Biochemistry and Microbiology, Science Faculty, Universidad Austral de Chile, Valdivia, Chile.
| |
Collapse
|
2
|
Schwartz L, de Dios Ruiz-Rosado J, Stonebrook E, Becknell B, Spencer JD. Uropathogen and host responses in pyelonephritis. Nat Rev Nephrol 2023; 19:658-671. [PMID: 37479904 PMCID: PMC10913074 DOI: 10.1038/s41581-023-00737-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2023] [Indexed: 07/23/2023]
Abstract
Urinary tract infections (UTIs) are among the most common bacterial infections seen in clinical practice. The ascent of UTI-causing pathogens to the kidneys results in pyelonephritis, which can trigger kidney injury, scarring and ultimately impair kidney function. Despite sizable efforts to understand how infections develop or are cleared in the bladder, our appreciation of the mechanisms by which infections develop, progress or are eradicated in the kidney is limited. The identification of virulence factors that are produced by uropathogenic Escherichia coli to promote pyelonephritis have begun to fill this knowledge gap, as have insights into the mechanisms by which kidney tubular epithelial cells oppose uropathogenic E. coli infection to prevent or eradicate UTIs. Emerging data also illustrate how specific cellular immune responses eradicate infection whereas other immune cell populations promote kidney injury. Insights into the mechanisms by which uropathogenic E. coli circumvent host immune defences or antibiotic therapy to cause pyelonephritis is paramount to the development of new prevention and treatment strategies to mitigate pyelonephritis and its associated complications.
Collapse
Affiliation(s)
- Laura Schwartz
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA.
- The Ohio State University College of Medicine, Columbus, OH, USA.
| | - Juan de Dios Ruiz-Rosado
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA
- The Ohio State University College of Medicine, Columbus, OH, USA
| | - Emily Stonebrook
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA
- The Ohio State University College of Medicine, Columbus, OH, USA
| | - Brian Becknell
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA
- The Ohio State University College of Medicine, Columbus, OH, USA
| | - John David Spencer
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA.
- The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
3
|
Chelangarimiyandoab F, Mungara P, Batta M, Cordat E. Urinary Tract Infections: Renal Intercalated Cells Protect against Pathogens. J Am Soc Nephrol 2023; 34:1605-1614. [PMID: 37401780 PMCID: PMC10561816 DOI: 10.1681/asn.0000000000000187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 06/22/2023] [Indexed: 07/05/2023] Open
Abstract
Urinary tract infections affect more than 1 in 2 women during their lifetime. Among these, more than 10% of patients carry antibiotic-resistant bacterial strains, highlighting the urgent need to identify alternative treatments. While innate defense mechanisms are well-characterized in the lower urinary tract, it is becoming evident that the collecting duct (CD), the first renal segment encountered by invading uropathogenic bacteria, also contributes to bacterial clearance. However, the role of this segment is beginning to be understood. This review summarizes the current knowledge on CD intercalated cells in urinary tract bacterial clearance. Understanding the innate protective role of the uroepithelium and of the CD offers new opportunities for alternative therapeutic strategies.
Collapse
Affiliation(s)
- Forough Chelangarimiyandoab
- Department of Physiology and Membrane Protein Disease Research Group, Faculty of Medicine & Dentistry, College of Health Sciences, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | |
Collapse
|
4
|
Insulin Downregulated the Infection of Uropathogenic Escherichia coli (UPEC) in Bladder Cells in a High-Glucose Environment through JAK/STAT Signaling Pathway. Microorganisms 2021; 9:microorganisms9122421. [PMID: 34946023 PMCID: PMC8704104 DOI: 10.3390/microorganisms9122421] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/16/2021] [Accepted: 11/19/2021] [Indexed: 12/31/2022] Open
Abstract
Diabetic individuals have a higher incidence of urinary tract infection (UTI) than non-diabetic individuals, and also require longer treatment. We evaluated the effects of insulin pretreatment on the regulation of JAK/STAT transduction pathways in UPEC-infected bladder cells in a high-glucose environment. A bladder cell model with GFP-UPEC and fluorescent-labeled TLR4, STAT1, STAT3, and insulin receptor antibodies, was used to evaluate the relationship between insulin receptor signaling, TLR-4-mediated, and JAK/STAT-dependent pathways. Pretreatment with 20 and 40 µg/mL insulin for 24 h significantly and dose-dependently reduced UPEC infection in SV-HUC-1 cells. Additionally, the expression levels of STAT1 and STAT3 were downregulated in a dose-dependent manner. However, insulin receptor (IR) expression was not affected by insulin pretreatment. Our results showed that insulin-mediated reduction of UPEC infection in a high-glucose environment was not only due to the downregulation of JAK1/2 and phosphorylated STAT-1/3, but also because of the decreased expression of TLR-4 proteins and pro-inflammatory IL-6. Here, we demonstrated that insulin reduced not only UPEC infection in bladder epithelial cells, but also inhibited the JAK/STAT transduction pathway during infection in a high-glucose environment. This study provides evidence to support the use of insulin in the treatment of UPEC infection in patients with type 2 diabetes (T2D).
Collapse
|
5
|
Akhigbe RE, Ajayi LO, Ajayi AF. Codeine exerts cardiorenal injury via upregulation of adenine deaminase/xanthine oxidase and caspase 3 signaling. Life Sci 2021; 273:118717. [PMID: 33159958 DOI: 10.1016/j.lfs.2020.118717] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 10/28/2020] [Accepted: 11/02/2020] [Indexed: 12/12/2022]
Abstract
AIMS Codeine treatment has been shown to be associated with glucolipid deregulation, though data reporting this are inconsistent and the mechanisms are not well understood. Perturbation of glutathione-dependent antioxidant defense and adenosine deaminase (ADA)/xanthine oxidase (XO) signaling has been implicated in the pathogenesis of cardiometabolic disorders. We thus, hypothesized that depletion of glutathione contents and upregulation of ADA/XO are involved in codeine-induced glucolipid deregulation. The present study also investigated whether or not codeine administration would induce genotoxicity and apoptosis in cardiac and renal tissues. MATERIALS AND METHODS Male New Zealand rabbits received per os distilled water or codeine, either in low dose (4 mg/kg) or high dose (10 mg/kg) for 6 weeks. KEY FINDINGS Codeine treatment led to reduced absolute and relative cardiac and renal mass independent of body weight change, increased blood glucose, total cholesterol (TC), triglycerides (TG), and low-density lipoprotein (LDL-C), as well as increased atherogenic indices and triglyceride-glucose index (TyG). Codeine administration significantly increased markers of cardiac and renal injury, as well as impaired cardiorenal functions. Codeine treatment also resulted in increased cardiac and renal malondialdehyde, Advanced Glycation Endproducts (AGE) and 8-hydroxydeoxyguanosine (8-OH-dG), and myeloperoxidase (MPO), ADA, XO, and caspase 3 activities. These observations were accompanied by impaired activities of cardiac and renal proton pumps. SIGNIFICANCE Findings of this study demonstrate that upregulation of ADA/XO and caspase 3 signaling are, at least partly, contributory to the glucolipid deregulation and cardiorenal injury induced by codeine.
Collapse
Affiliation(s)
- R E Akhigbe
- Department of Physiology, College of Medicine, Ladoke Akintola University of Technology, Ogbomoso, Oyo, Nigeria; Reproductive Biology and Toxicology Research Laboratories, Oasis of Grace Hospital, Osogbo, Nigeria
| | - L O Ajayi
- Department of Biochemistry, Adeleke University, Ede, Osun State, Nigeria
| | - A F Ajayi
- Department of Physiology, College of Medicine, Ladoke Akintola University of Technology, Ogbomoso, Oyo, Nigeria.
| |
Collapse
|
6
|
Kirigiti MA, Frazee T, Bennett B, Arik A, Blundell P, Bader L, Bagley J, Frias AE, Sullivan EL, Roberts CT, Kievit P. Effects of pre- and postnatal protein restriction on maternal and offspring metabolism in the nonhuman primate. Am J Physiol Regul Integr Comp Physiol 2020; 318:R929-R939. [PMID: 32130027 DOI: 10.1152/ajpregu.00150.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Women in low- and middle-income countries frequently consume a protein-deficient diet during pregnancy and breastfeeding. The effects of gestational malnutrition on fetal and early postnatal development can have lasting adverse effects on offspring metabolism. Expanding on previous studies in rodent models, we utilized a nonhuman primate model of gestational and early-life protein restriction (PR) to evaluate effects on the organ development and glucose metabolism of juvenile offspring. Offspring were born to dams that had consumed a control diet containing 26% protein or a PR diet containing 13% protein. Offspring were maintained on the PR diet and studied [body and serum measurements, intravenous glucose tolerance tests (ivGTTs), and dual-energy X-ray absorptiometry scans] up to 7 mo of age, at which time tissues were collected for analysis. PR offspring had age-appropriate body weight and were euglycemic but exhibited elevated fasting insulin and reduced initial, but increased total, insulin secretion during an ivGTT at 6 mo of age. No changes were detected in pancreatic islets of PR juveniles; however, PR did induce changes, including reduced kidney size, and changes in liver, adipose tissue, and muscle gene expression in other peripheral organs. Serum osteocalcin was elevated and bone mineral content and density were reduced in PR juveniles, indicating a significant impact of PR on early postnatal bone development.
Collapse
Affiliation(s)
- Melissa A Kirigiti
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, Oregon
| | - Tim Frazee
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, Oregon
| | - Baylin Bennett
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, Oregon
| | - Anam Arik
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, Oregon
| | - Peter Blundell
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, Oregon
| | - Lindsay Bader
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, Oregon
| | - Jennifer Bagley
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon
| | - Antonio E Frias
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon.,Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, Oregon
| | - Elinor L Sullivan
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon.,Department of Human Physiology, University of Oregon, Eugene, Oregon
| | - Charles T Roberts
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, Oregon.,Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon
| | - Paul Kievit
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, Oregon
| |
Collapse
|
7
|
Olaniyi KS, Woru Sabinari I, Olatunji LA. l-glutamine supplementation exerts cardio-renal protection in estrogen-progestin oral contraceptive-treated female rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2020; 74:103305. [PMID: 31790957 DOI: 10.1016/j.etap.2019.103305] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/01/2019] [Accepted: 11/21/2019] [Indexed: 06/10/2023]
Abstract
Glycogen and lipid disruptions represent a spectrum of metabolic disorders that are crucial risk factors for cardiovascular disease in estrogen-progestin oral contraceptive (COC) users. l-glutamine (GLN) has been shown to exert a modulatory effect in metabolic disorders-related syndromes. We therefore hypothesized that GLN supplementation would protect against myocardial and renal glycogen-lipid mishandling in COC-treated animals by modulation of Glucose-6-phosphate dehydrogenase (G6PD) and xanthine oxidase (XO) activities. Adult female Wistar rats were randomly allotted into control, GLN, COC and COC + GLN groups (six rats per group). The groups received vehicle (distilled water, p.o.), GLN (1 g/kg), COC containing 1.0 μg ethinylestradiol plus 5.0 μg levonorgestrel and COC plus GLN respectively, daily for 8 weeks. Data showed that treatment with COC led to metabolically-induced obesity with correspondent increased visceral and epicardial fat mass. It also led to increased plasma, myocardial and renal triglyceride, free fatty acid, malondialdehyde (MDA), XO activity, uric acid content and decreased glutathione content and G6PD activity. In addition, COC increased myocardial but not renal glycogen content, and increased myocardial and renal glycogen synthase activity, increased plasma and renal lactate production and plasma aspartate transaminase/alanine aminotransferase (AST/ALT) ratio. However, these alterations were attenuated when supplemented with GLN except plasma AST/ALT ratio. Collectively, the present results indicate that estrogen-progestin oral contraceptive causes metabolically-induced obesity that is accompanied by differential myocardial and renal metabolic disturbances. The findings also suggest that irrespective of varying metabolic phenotypes, GLN exerts protection against cardio-renal dysmetabolism by modulation of XO and G6PD activities.
Collapse
Affiliation(s)
- Kehinde Samuel Olaniyi
- HOPE Cardiometabolic Research Team & Department of Physiology, College of Health Sciences, University of Ilorin, P.M.B. 1515, Ilorin, Nigeria; Department of Physiology, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Isaiah Woru Sabinari
- HOPE Cardiometabolic Research Team & Department of Physiology, College of Health Sciences, University of Ilorin, P.M.B. 1515, Ilorin, Nigeria
| | - Lawrence Aderemi Olatunji
- HOPE Cardiometabolic Research Team & Department of Physiology, College of Health Sciences, University of Ilorin, P.M.B. 1515, Ilorin, Nigeria.
| |
Collapse
|
8
|
Patel BM, Goyal RK. Liver and insulin resistance: New wine in old bottle!!! Eur J Pharmacol 2019; 862:172657. [DOI: 10.1016/j.ejphar.2019.172657] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 09/02/2019] [Accepted: 09/05/2019] [Indexed: 12/20/2022]
|
9
|
Polianskyte-Prause Z, Tolvanen TA, Lindfors S, Dumont V, Van M, Wang H, Dash SN, Berg M, Naams JB, Hautala LC, Nisen H, Mirtti T, Groop PH, Wähälä K, Tienari J, Lehtonen S. Metformin increases glucose uptake and acts renoprotectively by reducing SHIP2 activity. FASEB J 2019; 33:2858-2869. [PMID: 30321069 PMCID: PMC6338644 DOI: 10.1096/fj.201800529rr] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 09/24/2018] [Indexed: 01/05/2023]
Abstract
Metformin, the first-line drug to treat type 2 diabetes (T2D), inhibits mitochondrial glycerolphosphate dehydrogenase in the liver to suppress gluconeogenesis. However, the direct target and the underlying mechanisms by which metformin increases glucose uptake in peripheral tissues remain uncharacterized. Lipid phosphatase Src homology 2 domain-containing inositol-5-phosphatase 2 (SHIP2) is upregulated in diabetic rodent models and suppresses insulin signaling by reducing Akt activation, leading to insulin resistance and diminished glucose uptake. Here, we demonstrate that metformin directly binds to and reduces the catalytic activity of the recombinant SHIP2 phosphatase domain in vitro. Metformin inhibits SHIP2 in cultured cells and in skeletal muscle and kidney of db/db mice. In SHIP2-overexpressing myotubes, metformin ameliorates reduced glucose uptake by slowing down glucose transporter 4 endocytosis. SHIP2 overexpression reduces Akt activity and enhances podocyte apoptosis, and both are restored to normal levels by metformin. SHIP2 activity is elevated in glomeruli of patients with T2D receiving nonmetformin medication, but not in patients receiving metformin, compared with people without diabetes. Furthermore, podocyte loss in kidneys of metformin-treated T2D patients is reduced compared with patients receiving nonmetformin medication. Our data unravel a novel molecular mechanism by which metformin enhances glucose uptake and acts renoprotectively by reducing SHIP2 activity.-Polianskyte-Prause, Z., Tolvanen, T. A., Lindfors, S., Dumont, V., Van, M., Wang, H., Dash, S. N., Berg, M., Naams, J.-B., Hautala, L. C., Nisen, H., Mirtti, T., Groop, P.-H., Wähälä, K., Tienari, J., Lehtonen, S. Metformin increases glucose uptake and acts renoprotectively by reducing SHIP2 activity.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Gene Expression Regulation, Enzymologic/drug effects
- Humans
- Hypoglycemic Agents/pharmacology
- Kidney Diseases/prevention & control
- Male
- Metformin/pharmacology
- Mice
- Mice, Inbred C57BL
- Muscle Fibers, Skeletal/cytology
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/metabolism
- Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/antagonists & inhibitors
- Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism
- Podocytes/cytology
- Podocytes/drug effects
- Podocytes/metabolism
- Rats
Collapse
Affiliation(s)
| | | | - Sonja Lindfors
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Vincent Dumont
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Mervi Van
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Hong Wang
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Surjya N. Dash
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Mika Berg
- Department of Chemistry, University of Helsinki, Helsinki, Finland
| | | | - Laura C. Hautala
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Harry Nisen
- Department of Urology, Helsinki University Hospital, Helsinki, Finland
| | - Tuomas Mirtti
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Per-Henrik Groop
- Folkhälsan Research Center, Folkhälsan Institute of Genetics, University of Helsinki, Helsinki, Finland
- Abdominal Center Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
- Central Clinical School, Monash University, Melbourne, Victoria, Australia; and
| | - Kristiina Wähälä
- Department of Chemistry, University of Helsinki, Helsinki, Finland
| | - Jukka Tienari
- Department of Pathology, University of Helsinki, Helsinki, Finland
- Helsinki University Hospital, Hyvinkää, Finland
| | - Sanna Lehtonen
- Department of Pathology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
10
|
Singh S, Sharma R, Kumari M, Tiwari S. Insulin receptors in the kidneys in health and disease. World J Nephrol 2019; 8:11-22. [PMID: 30705868 PMCID: PMC6354081 DOI: 10.5527/wjn.v8.i1.11] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 11/15/2018] [Accepted: 12/10/2018] [Indexed: 02/06/2023] Open
Abstract
Insulin is an important hormone that affects various metabolic processes, including kidney function. Impairment in insulin’s action leads to insulin resistance in the target tissue. Besides defects in post-receptor insulin signaling, impairment at the receptor level could significantly affect insulin sensitivity of the target tissue. The kidney is a known target of insulin; however, whether the kidney develops “insulin resistance” is debatable. Regulation of the insulin receptor (IR) expression and its function is very well studied in major metabolic tissues like liver, skeletal muscles, and adipose tissue. The physiological relevance of IRs in the kidney has recently begun to be clarified. The credit goes to studies that showed a wide distribution of IR throughout the nephron segments and their reduced expression in the insulin resistance state. Moreover, altered renal and systemic metabolism observed in mice with targeted deletion of the IR from various epithelial cells of the kidney has strengthened this proposition. In this review, we recapitulate the crucial findings from literature that have expanded our knowledge regarding the significance of the renal IR in normal- and insulin-resistance states.
Collapse
Affiliation(s)
- Sarojini Singh
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Rajni Sharma
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Manju Kumari
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Swasti Tiwari
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| |
Collapse
|
11
|
Murtha MJ, Eichler T, Bender K, Metheny J, Li B, Schwaderer AL, Mosquera C, James C, Schwartz L, Becknell B, Spencer JD. Insulin receptor signaling regulates renal collecting duct and intercalated cell antibacterial defenses. J Clin Invest 2018; 128:5634-5646. [PMID: 30418175 DOI: 10.1172/jci98595] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 09/20/2018] [Indexed: 12/14/2022] Open
Abstract
People with diabetes mellitus have increased infection risk. With diabetes, urinary tract infection (UTI) is more common and has worse outcomes. Here, we investigate how diabetes and insulin resistance impact the kidney's innate defenses and urine sterility. We report that type 2 diabetic mice have increased UTI risk. Moreover, insulin-resistant prediabetic mice have increased UTI susceptibility, independent of hyperglycemia or glucosuria. To identify how insulin resistance affects renal antimicrobial defenses, we genetically deleted the insulin receptor in the kidney's collecting tubules and intercalated cells. Intercalated cells, located within collecting tubules, contribute to epithelial defenses by acidifying the urine and secreting antimicrobial peptides (AMPs) into the urinary stream. Collecting duct and intercalated cell-specific insulin receptor deletion did not impact urine acidification, suppressed downstream insulin-mediated targets and AMP expression, and increased UTI susceptibility. Specifically, insulin receptor-mediated signaling regulates AMPs, including lipocalin 2 and ribonuclease 4, via phosphatidylinositol-3-kinase signaling. These data suggest that insulin signaling plays a critical role in renal antibacterial defenses.
Collapse
Affiliation(s)
- Matthew J Murtha
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Tad Eichler
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Kristin Bender
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Jackie Metheny
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Birong Li
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Andrew L Schwaderer
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,The Ohio State University College of Medicine, Columbus, Ohio, USA.,Division of Nephrology, Department of Pediatrics, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Claudia Mosquera
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Cindy James
- Mass Spectrometry and Proteomics Facility, The Ohio State University, Columbus, Ohio, USA
| | - Laura Schwartz
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Brian Becknell
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,The Ohio State University College of Medicine, Columbus, Ohio, USA.,Division of Nephrology, Department of Pediatrics, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - John David Spencer
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,The Ohio State University College of Medicine, Columbus, Ohio, USA.,Division of Nephrology, Department of Pediatrics, Nationwide Children's Hospital, Columbus, Ohio, USA
| |
Collapse
|
12
|
Scheen AJ. Effects of glucose-lowering agents on surrogate endpoints and hard clinical renal outcomes in patients with type 2 diabetes. DIABETES & METABOLISM 2018; 45:110-121. [PMID: 30477733 DOI: 10.1016/j.diabet.2018.10.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 09/17/2018] [Accepted: 10/08/2018] [Indexed: 12/12/2022]
Abstract
Diabetic kidney disease (DKD) represents an enormous burden in patients with type 2 diabetes mellitus (T2DM). Preclinical studies using most glucose-lowering agents have suggested renal-protective effects, but the proposed mechanisms of renoprotection have yet to be defined, and the promising results from experimental studies remain to be translated into human clinical findings to improve the prognosis of patients at risk of DKD. Also, it is important to distinguish effects on surrogate endpoints, such as decreases in albuminuria and estimated glomerular filtration rate (eGFR), and hard clinical endpoints, such as progression to end-stage renal disease (ESRD) and death from renal causes. Data regarding insulin therapy are surprisingly scarce, and it is nearly impossible to separate the effects of better glucose control from those of insulin per se, whereas favourable preclinical data with metformin, thiazolidinediones and dipeptidyl peptidase (DPP)-4 inhibitors are plentiful, and positive effects have been observed in clinical studies, at least for surrogate endpoints. The most favourable renal results have been reported with glucagon-like peptide-1 receptor agonists (GLP-1RAs) and sodium-glucose cotransporter type-2 inhibitors (SGLT2is). Significant reductions in both albuminuria and eGFR decline have been reported with these classes of glucose-lowering medications compared with placebo and other glucose-lowering agents. Moreover, in large prospective cardiovascular outcome trials using composite renal outcomes as secondary endpoints, both GLP-1RAs and SGLT2is added to standard care reduced renal outcomes combining persistent macro-albuminuria, doubling of serum creatinine, progression to ESRD and kidney-related death; however, to date, only SGLT2is have been clearly shown to reduce such hard clinical outcomes. Yet, as the renoprotective effects of SGLT2is and GLP-1RAs appear to be independent of glucose-lowering activity, the underlying mechanisms are still a matter of debate. For this reason, further studies with renal outcomes as primary endpoints are now awaited in T2DM patients at high risk of DKD, including trials evaluating the potential add-on benefits of combined GLP-1RA-SGLT2i therapies.
Collapse
Affiliation(s)
- A J Scheen
- Division of Clinical Pharmacology, Centre for Interdisciplinary Research on Medicines (CIRM), University of Liège, Liège, Belgium; Division of Diabetes, Nutrition and Metabolic Disorders, Department of Medicine, CHU de Liège, Liège, Belgium.
| |
Collapse
|
13
|
Tokarz VL, MacDonald PE, Klip A. The cell biology of systemic insulin function. J Cell Biol 2018; 217:2273-2289. [PMID: 29622564 PMCID: PMC6028526 DOI: 10.1083/jcb.201802095] [Citation(s) in RCA: 276] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 03/21/2018] [Accepted: 03/23/2018] [Indexed: 12/12/2022] Open
Abstract
Insulin is the paramount anabolic hormone, promoting carbon energy deposition in the body. Its synthesis, quality control, delivery, and action are exquisitely regulated by highly orchestrated intracellular mechanisms in different organs or "stations" of its bodily journey. In this Beyond the Cell review, we focus on these five stages of the journey of insulin through the body and the captivating cell biology that underlies the interaction of insulin with each organ. We first analyze insulin's biosynthesis in and export from the β-cells of the pancreas. Next, we focus on its first pass and partial clearance in the liver with its temporality and periodicity linked to secretion. Continuing the journey, we briefly describe insulin's action on the blood vasculature and its still-debated mechanisms of exit from the capillary beds. Once in the parenchymal interstitium of muscle and adipose tissue, insulin promotes glucose uptake into myofibers and adipocytes, and we elaborate on the intricate signaling and vesicle traffic mechanisms that underlie this fundamental function. Finally, we touch upon the renal degradation of insulin to end its action. Cellular discernment of insulin's availability and action should prove critical to understanding its pivotal physiological functions and how their failure leads to diabetes.
Collapse
Affiliation(s)
- Victoria L Tokarz
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Patrick E MacDonald
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Amira Klip
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
14
|
Abstract
BACKGROUND Systems biology is an important field for understanding whole biological mechanisms composed of interactions between biological components. One approach for understanding complex and diverse mechanisms is to analyze biological pathways. However, because these pathways consist of important interactions and information on these interactions is disseminated in a large number of biomedical reports, text-mining techniques are essential for extracting these relationships automatically. RESULTS In this study, we applied node2vec, an algorithmic framework for feature learning in networks, for relationship extraction. To this end, we extracted genes from paper abstracts using pkde4j, a text-mining tool for detecting entities and relationships. Using the extracted genes, a co-occurrence network was constructed and node2vec was used with the network to generate a latent representation. To demonstrate the efficacy of node2vec in extracting relationships between genes, performance was evaluated for gene-gene interactions involved in a type 2 diabetes pathway. Moreover, we compared the results of node2vec to those of baseline methods such as co-occurrence and DeepWalk. CONCLUSIONS Node2vec outperformed existing methods in detecting relationships in the type 2 diabetes pathway, demonstrating that this method is appropriate for capturing the relatedness between pairs of biological entities involved in biological pathways. The results demonstrated that node2vec is useful for automatic pathway construction.
Collapse
Affiliation(s)
- Munui Kim
- Department of Library and Information Science, Yonsei University, 50, Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea
| | - Seung Han Baek
- Department of Library and Information Science, Yonsei University, 50, Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea
| | - Min Song
- Department of Library and Information Science, Yonsei University, 50, Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea.
| |
Collapse
|
15
|
Rossi C, Marzano V, Consalvo A, Zucchelli M, Levi Mortera S, Casagrande V, Mavilio M, Sacchetta P, Federici M, Menghini R, Urbani A, Ciavardelli D. Proteomic and metabolomic characterization of streptozotocin-induced diabetic nephropathy in TIMP3-deficient mice. Acta Diabetol 2018; 55:121-129. [PMID: 29134286 DOI: 10.1007/s00592-017-1074-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 11/03/2017] [Indexed: 10/18/2022]
Abstract
AIMS The tissue inhibitor of metalloproteinase TIMP3 is a stromal protein that restrains the activity of both protease and receptor in the extracellular matrix and has been found to be down-regulated in diabetic nephropathy (DN), the leading cause of end-stage renal disease in developed countries. METHODS In order to gain deeper insights on the association of loss of TIMP3 and DN, we performed differential proteomic analysis of kidney and blood metabolic profiling of wild-type and Timp3-knockout mice before and after streptozotocin (STZ) treatment, widely used to induce insulin deficiency and hyperglycemia. RESULTS Kidney proteomic data and blood metabolic profiles suggest significant alterations of peroxisomal and mitochondrial fatty acids β-oxidation in Timp3-knockout mice compared to wild-type mice under basal condition. These alterations were exacerbated in response to STZ treatment. CONCLUSIONS Proteomic and metabolomic approaches showed that loss of TIMP3 alone or in combination with STZ treatment results in significant alterations of kidney lipid metabolism and peripheral acylcarnitine levels, supporting the idea that loss of TIMP3 may generate a phenotype more prone to DN.
Collapse
Affiliation(s)
- Claudia Rossi
- Laboratorio di Biochimica Analitica e Proteomica, Centro Scienze dell'Invecchiamento e Medicina Traslazionale - CeSI-MeT, Via Luigi Polacchi 11, 66100, Chieti, Italy.
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Via dei Vestini 29, 66013, Chieti, Italy.
| | - Valeria Marzano
- Proteomic and Metabonomic Laboratory, Santa Lucia Foundation IRCCS, Via del Fosso di Fiorano 65, 00143, Rome, Italy
- Human Microbiome Unit, Genetic and Rare Diseases Area, Bambino Gesù Children's Hospital IRCCS, Piazza S. Onofrio 4, 00165, Rome, Italy
| | - Ada Consalvo
- Laboratorio di Biochimica Analitica e Proteomica, Centro Scienze dell'Invecchiamento e Medicina Traslazionale - CeSI-MeT, Via Luigi Polacchi 11, 66100, Chieti, Italy
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Via dei Vestini 29, 66013, Chieti, Italy
| | - Mirco Zucchelli
- Laboratorio di Biochimica Analitica e Proteomica, Centro Scienze dell'Invecchiamento e Medicina Traslazionale - CeSI-MeT, Via Luigi Polacchi 11, 66100, Chieti, Italy
| | - Stefano Levi Mortera
- Proteomic and Metabonomic Laboratory, Santa Lucia Foundation IRCCS, Via del Fosso di Fiorano 65, 00143, Rome, Italy
- Human Microbiome Unit, Genetic and Rare Diseases Area, Bambino Gesù Children's Hospital IRCCS, Piazza S. Onofrio 4, 00165, Rome, Italy
| | - Viviana Casagrande
- Department of Systems Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy
| | - Maria Mavilio
- Department of Systems Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy
| | - Paolo Sacchetta
- Laboratorio di Biochimica Analitica e Proteomica, Centro Scienze dell'Invecchiamento e Medicina Traslazionale - CeSI-MeT, Via Luigi Polacchi 11, 66100, Chieti, Italy
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Via dei Vestini 29, 66013, Chieti, Italy
| | - Massimo Federici
- Department of Systems Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy
| | - Rossella Menghini
- Department of Systems Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy
| | - Andrea Urbani
- Proteomic and Metabonomic Laboratory, Santa Lucia Foundation IRCCS, Via del Fosso di Fiorano 65, 00143, Rome, Italy
- Institute of Biochemistry and Biochemical Clinic, Faculty of Medicine and Surgery - Policlinico A. Gemelli, Catholic University of Sacred Heart, Largo F. Vito 1, 00168, Rome, Italy
| | - Domenico Ciavardelli
- Laboratorio di Biochimica Analitica e Proteomica, Centro Scienze dell'Invecchiamento e Medicina Traslazionale - CeSI-MeT, Via Luigi Polacchi 11, 66100, Chieti, Italy
- School of Human and Social Science, 'Kore' University of Enna, Via della Cooperazione, 94100, Enna, Italy
| |
Collapse
|
16
|
Bertinat R, Westermeier F, Silva P, Gatica R, Oliveira JM, Nualart F, Gomis R, Yáñez AJ. The Antidiabetic Agent Sodium Tungstate Induces Abnormal Glycogen Accumulation in Renal Proximal Tubules from Diabetic IRS2-Knockout Mice. J Diabetes Res 2018; 2018:5697970. [PMID: 30003110 PMCID: PMC5996472 DOI: 10.1155/2018/5697970] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/19/2017] [Accepted: 01/28/2018] [Indexed: 11/22/2022] Open
Abstract
The kidney is an insulin-sensitive organ involved in glucose homeostasis. One major effect of insulin is to induce glycogen storage in the liver and muscle. However, no significant glycogen stores are detected in normal kidneys, but diabetic subjects present a characteristic renal histopathological feature resulting from extensive glycogen deposition mostly in nonproximal tubules. The mechanism of renal glycogen accumulation is yet poorly understood. Here, we studied in situ glycogen accumulation in the kidney from diabetic IRS2-knockout mice and the effect of the insulin-mimetic agent sodium tungstate (NaW). IRS2-knockout mice displayed hyperglycemia and hyperinsulinemia. NaW only normalized glycemia. There was no evident morphological difference between kidneys from untreated wild-type (WT), NaW-treated WT, and untreated IRS2-knockout mice. However, NaW-treated IRS2-knockout mice showed tubular alterations resembling clear cells in the cortex, but not in the outer medulla, that were correlated with glycogen accumulation. Immunohistochemical detection of the gluconeogenic enzyme phosphoenolpyruvate carboxykinase, mostly expressed by renal proximal tubules, showed that altered tubules were of proximal origin. Our preliminary study suggests that IRS2 differentially regulates glycogen accumulation in renal tubules and that NaW treatment in the context of IRS2 ablation induces abnormal glycogen accumulation in cortical proximal tubules.
Collapse
Affiliation(s)
- Romina Bertinat
- Centro de Microscopía Avanzada (CMA BIO-BIO), Universidad de Concepción, Concepción, Chile
| | - Francisco Westermeier
- Institute of Biomedical Science, FH Joanneum Gesellschaft mbH University of Applied Sciences, Eggenberger Allee 13, 8020 Graz, Austria
- Facultad de Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Pamela Silva
- Facultad de Salud, Universidad Santo Tomás, Osorno, Chile
| | - Rodrigo Gatica
- Escuela de Veterinaria, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Joana Moitinho Oliveira
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
- Diabetes and Obesity Research Laboratory, IDIBAPS, Barcelona, Spain
| | - Francisco Nualart
- Centro de Microscopía Avanzada (CMA BIO-BIO), Universidad de Concepción, Concepción, Chile
| | - Ramón Gomis
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
- Diabetes and Obesity Research Laboratory, IDIBAPS, Barcelona, Spain
- Department of Endocrinology and Nutrition, Hospital Clinic, Barcelona, Spain
- Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | | |
Collapse
|
17
|
Klawitter J, Pennington A, Klawitter J, Thurman JM, Christians U. Mitochondrial cyclophilin D ablation is associated with the activation of Akt/p70S6K pathway in the mouse kidney. Sci Rep 2017; 7:10540. [PMID: 28874678 PMCID: PMC5585384 DOI: 10.1038/s41598-017-10076-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 08/04/2017] [Indexed: 01/03/2023] Open
Abstract
The mitochondrial matrix protein cyclophilin D (CypD) is an essential component of the mitochondrial permeability transition pore (MPTP). Here we characterized the effects of CypD ablation on bioenergetics in the kidney. CypD loss triggers a metabolic shift in Ppif-/- male and female mouse kidneys towards glycolysis and Krebs cycle activity. The shift is accompanied by increased glucose consumption and a transcriptional upregulation of effectors of glucose metabolism in the kidney. These included activation of Akt, AMPK (only in males) and p70S6K kinases. Gender specific differences between the Ppif-/- male and female mouse kidneys were observed including activation of pro-surviving ERK1/2 kinase and inhibited expression of pro-apoptotic and pro-fibrotic JNK and TGFβ1 proteins in Ppif-/- females. They also showed the highest expression of phosphorylated-ERK1/2 and Akt S473 proteins of all four investigated animal groups. Furthermore, Ppif-/- females showed higher lactate concentrations and ATP/ADP-ratios in the kidney than males. These metabolic and transcriptional modifications could provide an additional level of protection to Ppif-/- females. In summary, loss of mitochondrial CypD results in a shift in bioenergetics and in activation of glucose-metabolism regulating Akt/AMPK/p70S6 kinase pathways that is expected to affect the capability of Ppif-/- mice kidneys to react to stimuli and injury.
Collapse
Affiliation(s)
- Jelena Klawitter
- Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, USA.
- Division of Renal Disease and Hypertension, University of Colorado Denver, Aurora, Colorado, USA.
| | - Alexander Pennington
- Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, USA
| | - Jost Klawitter
- Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, USA
| | - Joshua M Thurman
- Division of Renal Disease and Hypertension, University of Colorado Denver, Aurora, Colorado, USA
| | - Uwe Christians
- Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, USA
| |
Collapse
|
18
|
Sibiya N, Ngubane P, Mabandla M. The Ameliorative Effect of Pectin-Insulin Patch On Renal Injury in Streptozotocin-Induced Diabetic Rats. Kidney Blood Press Res 2017; 42:530-540. [PMID: 28854437 DOI: 10.1159/000480395] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 05/09/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND/AIMS Renal damage and dysfunction is attributed to sustained hyperglycaemia in overt diabetes. Subcutaneous insulin injections are beneficial in delaying the progression of renal dysfunction and damage in diabetics. However, the current mode of administration is associated with severe undesirable effects. In this study, we evaluated the ameliorative effects of pectin-insulin dermal patches on renal dysfunction in diabetes. METHODS Pectin-insulin patches (20.0, 40.8 and 82.9 µg/kg) were applied on the skin of streptozotocin-induced diabetic rats, thrice daily for 5 weeks. Blood glucose concentration, blood pressure and urine output volume were recorded on week 5 after which the animals were sacrificed after which the kidneys and plasma were collected. Kidney nephrin expression and urinary nephrin concentration, albumin excretion rate (AER), creatinine clearance (CC) and albumin creatinine ratio (ACR) were assessed. RESULTS Patch application resulted in reduced blood glucose concentration and blood pressure. Furthermore, pectin-insulin patch treatment resulted in increased kidney nephrin expression and reduced urinary nephrin concentration. AER, CC ACR were also reduced post patch application. CONCLUSIONS The application of pectin-insulin patch limited diabetes associated kidney damaged and improved kidney function. These observations suggest that pectin-insulin patches may ameliorate kidney dysfunction that is associated with chronic subcutaneous insulin administration.
Collapse
|
19
|
Escudero CA, Herlitz K, Troncoso F, Guevara K, Acurio J, Aguayo C, Godoy AS, González M. Pro-angiogenic Role of Insulin: From Physiology to Pathology. Front Physiol 2017; 8:204. [PMID: 28424632 PMCID: PMC5380736 DOI: 10.3389/fphys.2017.00204] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 03/20/2017] [Indexed: 12/13/2022] Open
Abstract
The underlying molecular mechanisms involve in the regulation of the angiogenic process by insulin are not well understood. In this review article, we aim to describe the role of insulin and insulin receptor activation on the control of angiogenesis and how these mechanisms can be deregulated in human diseases. Functional expression of insulin receptors and their signaling pathways has been described on endothelial cells and pericytes, both of the main cells involved in vessel formation and maturation. Consequently, insulin has been shown to regulate endothelial cell migration, proliferation, and in vitro tubular structure formation through binding to its receptors and activation of intracellular phosphorylation cascades. Furthermore, insulin-mediated pro-angiogenic state is potentiated by generation of vascular growth factors, such as the vascular endothelial growth factor, produced by endothelial cells. Additionally, diseases such as insulin resistance, obesity, diabetes, and cancer may be associated with the deregulation of insulin-mediated angiogenesis. Despite this knowledge, the underlying molecular mechanisms need to be elucidated in order to provide new insights into the role of insulin on angiogenesis.
Collapse
Affiliation(s)
- Carlos A Escudero
- Group of Investigation in Tumor Angiogenesis, Vascular Physiology Laboratory, Basic Sciences Department, Universidad del Bío BíoChillán, Chile.,Group of Research and Innovation in Vascular Health, Department of Basic Sciences, Universidad del Bío-BíoChillán, Chile
| | - Kurt Herlitz
- Group of Investigation in Tumor Angiogenesis, Vascular Physiology Laboratory, Basic Sciences Department, Universidad del Bío BíoChillán, Chile
| | - Felipe Troncoso
- Group of Investigation in Tumor Angiogenesis, Vascular Physiology Laboratory, Basic Sciences Department, Universidad del Bío BíoChillán, Chile
| | - Katherine Guevara
- Group of Investigation in Tumor Angiogenesis, Vascular Physiology Laboratory, Basic Sciences Department, Universidad del Bío BíoChillán, Chile
| | - Jesenia Acurio
- Group of Investigation in Tumor Angiogenesis, Vascular Physiology Laboratory, Basic Sciences Department, Universidad del Bío BíoChillán, Chile
| | - Claudio Aguayo
- Group of Research and Innovation in Vascular Health, Department of Basic Sciences, Universidad del Bío-BíoChillán, Chile.,Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of ConcepciónConcepción, Chile
| | - Alejandro S Godoy
- Department of Physiology, Pontificia Universidad Católica de ChileSantiago, Chile.,Department of Urology, Roswell Park Cancer InstituteBuffalo, NY, USA
| | - Marcelo González
- Group of Research and Innovation in Vascular Health, Department of Basic Sciences, Universidad del Bío-BíoChillán, Chile.,Vascular Physiology Laboratory, Department of Physiology, Faculty of Biological Sciences, Universidad of ConcepciónConcepción, Chile
| |
Collapse
|
20
|
Shanmugasundaram K, Block K. Renal Carcinogenesis, Tumor Heterogeneity, and Reactive Oxygen Species: Tactics Evolved. Antioxid Redox Signal 2016; 25:685-701. [PMID: 27287984 PMCID: PMC5069729 DOI: 10.1089/ars.2015.6569] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 06/07/2016] [Accepted: 06/10/2016] [Indexed: 12/13/2022]
Abstract
SIGNIFICANCE The number of kidney cancers is growing 3-5% each year due to unknown etiologies. Intra- and inter-tumor mediators increase oxidative stress and drive tumor heterogeneity. Recent Advances: Technology advancement in state-of-the-art instrumentation and methodologies allows researchers to detect and characterize global landscaping modifications in genes, proteins, and pathophysiology patterns at the single-cell level. CRITICAL ISSUES We postulate that the sources of reactive oxygen species (ROS) and their activation within subcellular compartments will change over a timeline of tumor evolvement and contribute to tumor heterogeneity. Therefore, the complexity of intracellular changes within a tumor and ROS-induced tumor heterogeneity coupled to the advancement of detecting these events globally are limited at the level of data collection, organization, and interpretation using software algorithms and bioinformatics. FUTURE DIRECTIONS Integrative and collaborative research, combining the power of numbers with careful experimental design, protocol development, and data interpretation, will translate cancer biology and therapeutics to a heightened level or leave the abundant raw data as stagnant and underutilized. Antioxid. Redox Signal. 25, 685-701.
Collapse
Affiliation(s)
| | - Karen Block
- Department of Medicine, University of Texas Health Science Center, San Antonio, Texas
- South Texas Veterans Health Care System, Audie L. Murphy Memorial Hospital Division, San Antonio, Texas
| |
Collapse
|
21
|
Eichler TE, Becknell B, Easterling RS, Ingraham SE, Cohen DM, Schwaderer AL, Hains DS, Li B, Cohen A, Metheny J, Tridandapani S, Spencer JD. Insulin and the phosphatidylinositol 3-kinase signaling pathway regulate Ribonuclease 7 expression in the human urinary tract. Kidney Int 2016; 90:568-79. [PMID: 27401534 DOI: 10.1016/j.kint.2016.04.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 04/12/2016] [Accepted: 04/28/2016] [Indexed: 10/21/2022]
Abstract
Diabetes mellitus is a systemic disease associated with a deficiency of insulin production or action. Diabetic patients have an increased susceptibility to infection with the urinary tract being the most common site. Recent studies suggest that Ribonuclease 7 (RNase 7) is a potent antimicrobial peptide that plays an important role in protecting the urinary tract from bacterial insult. Because the impact of diabetes on RNase 7 expression and function are unknown, we investigated the effects of insulin on RNase 7 using human urine specimens. The urinary RNase 7 concentrations were measured in healthy control patients and insulin-deficient type 1 diabetics before and after starting insulin therapy. Compared with controls, diabetic patients had suppressed urinary RNase 7 concentrations, which increased with insulin. Using primary human urothelial cells, the mechanisms by which insulin stimulates RNase 7 synthesis were next explored. Insulin induced RNase 7 production via the phosphatidylinositide 3-kinase signaling pathway (PI3K/AKT) to shield urothelial cells from uropathogenic E. coli. In contrast, uropathogenic E. coli suppressed PI3K/AKT activity and RNase 7 production. Thus, insulin and PI3K/AKT signaling are essential for RNase 7 expression and increased infection risks in diabetic patients may be secondary to suppressed RNase 7 production. Our data may provide unique insight into novel urinary tract infection therapeutic strategies in at-risk populations.
Collapse
Affiliation(s)
- Tad E Eichler
- Center for Clinical and Translational Research, Department of Pediatrics, The Research Institute at Nationwide Children's, Columbus, Ohio, USA
| | - Brian Becknell
- Center for Clinical and Translational Research, Department of Pediatrics, The Research Institute at Nationwide Children's, Columbus, Ohio, USA; Division of Nephrology, Department of Pediatrics, Nationwide Children's, Columbus, Ohio, USA
| | - Robert S Easterling
- Center for Clinical and Translational Research, Department of Pediatrics, The Research Institute at Nationwide Children's, Columbus, Ohio, USA; University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Susan E Ingraham
- Center for Clinical and Translational Research, Department of Pediatrics, The Research Institute at Nationwide Children's, Columbus, Ohio, USA; Division of Nephrology, Department of Pediatrics, Nationwide Children's, Columbus, Ohio, USA
| | - Daniel M Cohen
- Division of Emergency Medicine, Department of Pediatrics, Nationwide Children's, Columbus, Ohio, USA
| | - Andrew L Schwaderer
- Center for Clinical and Translational Research, Department of Pediatrics, The Research Institute at Nationwide Children's, Columbus, Ohio, USA; Division of Nephrology, Department of Pediatrics, Nationwide Children's, Columbus, Ohio, USA
| | - David S Hains
- Innate Immunity Translational Research Center, Department of Pediatrics, Children's Foundation Research Institute at Le Bonheur Children's Hospital, Memphis, Tennessee, USA
| | - Birong Li
- Center for Clinical and Translational Research, Department of Pediatrics, The Research Institute at Nationwide Children's, Columbus, Ohio, USA
| | - Ariel Cohen
- Center for Clinical and Translational Research, Department of Pediatrics, The Research Institute at Nationwide Children's, Columbus, Ohio, USA
| | - Jackie Metheny
- Center for Clinical and Translational Research, Department of Pediatrics, The Research Institute at Nationwide Children's, Columbus, Ohio, USA
| | - Susheela Tridandapani
- Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University James Cancer Hospital and Richard J. Solove Research Institute, Columbus, Ohio, USA; Department of Molecular Virology, Immunology, and Medical Genetics, Comprehensive Cancer Center, The Ohio State University James Cancer Hospital and Richard J. Solove Research Institute, Columbus, Ohio, USA
| | - John David Spencer
- Center for Clinical and Translational Research, Department of Pediatrics, The Research Institute at Nationwide Children's, Columbus, Ohio, USA; Division of Nephrology, Department of Pediatrics, Nationwide Children's, Columbus, Ohio, USA.
| |
Collapse
|
22
|
Obesity-related glomerulopathy: clinical and pathologic characteristics and pathogenesis. Nat Rev Nephrol 2016; 12:453-71. [PMID: 27263398 DOI: 10.1038/nrneph.2016.75] [Citation(s) in RCA: 481] [Impact Index Per Article: 53.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The prevalence of obesity-related glomerulopathy is increasing in parallel with the worldwide obesity epidemic. Glomerular hypertrophy and adaptive focal segmental glomerulosclerosis define the condition pathologically. The glomerulus enlarges in response to obesity-induced increases in glomerular filtration rate, renal plasma flow, filtration fraction and tubular sodium reabsorption. Normal insulin/phosphatidylinositol 3-kinase/Akt and mTOR signalling are critical for podocyte hypertrophy and adaptation. Adipokines and ectopic lipid accumulation in the kidney promote insulin resistance of podocytes and maladaptive responses to cope with the mechanical forces of renal hyperfiltration. Although most patients have stable or slowly progressive proteinuria, up to one-third develop progressive renal failure and end-stage renal disease. Renin-angiotensin-aldosterone blockade is effective in the short-term but weight loss by hypocaloric diet or bariatric surgery has induced more consistent and dramatic antiproteinuric effects and reversal of hyperfiltration. Altered fatty acid and cholesterol metabolism are increasingly recognized as key mediators of renal lipid accumulation, inflammation, oxidative stress and fibrosis. Newer therapies directed to lipid metabolism, including SREBP antagonists, PPARα agonists, FXR and TGR5 agonists, and LXR agonists, hold therapeutic promise.
Collapse
|
23
|
Zaika O, Tomilin V, Mamenko M, Bhalla V, Pochynyuk O. New perspective of ClC-Kb/2 Cl- channel physiology in the distal renal tubule. Am J Physiol Renal Physiol 2016; 310:F923-30. [PMID: 26792067 PMCID: PMC5002062 DOI: 10.1152/ajprenal.00577.2015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 01/14/2016] [Indexed: 12/17/2022] Open
Abstract
Since its identification as the underlying molecular cause of Bartter's syndrome type 3, ClC-Kb (ClC-K2 in rodents, henceforth it will be referred as ClC-Kb/2) is proposed to play an important role in systemic electrolyte balance and blood pressure regulation by controlling basolateral Cl(-) exit in the distal renal tubular segments from the cortical thick ascending limb to the outer medullary collecting duct. Considerable experimental and clinical effort has been devoted to the identification and characterization of disease-causing mutations as well as control of the channel by its cofactor, barttin. However, we have only begun to unravel the role of ClC-Kb/2 in different tubular segments and to reveal the regulators of its expression and function, e.g., insulin and IGF-1. In this review we discuss recent experimental evidence in this regard and highlight unexplored questions critical to understanding ClC-Kb/2 physiology in the kidney.
Collapse
Affiliation(s)
- Oleg Zaika
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas; and
| | - Viktor Tomilin
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas; and
| | - Mykola Mamenko
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas; and
| | - Vivek Bhalla
- Division of Nephrology, Department of Medicine, Stanford University, Stanford, California
| | - Oleh Pochynyuk
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas; and
| |
Collapse
|
24
|
Selective Insulin Resistance in the Kidney. BIOMED RESEARCH INTERNATIONAL 2016; 2016:5825170. [PMID: 27247938 PMCID: PMC4876201 DOI: 10.1155/2016/5825170] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/04/2016] [Accepted: 04/07/2016] [Indexed: 12/12/2022]
Abstract
Insulin resistance has been characterized as attenuation of insulin sensitivity at target organs and tissues, such as muscle and fat tissues and the liver. The insulin signaling cascade is divided into major pathways such as the PI3K/Akt pathway and the MAPK/MEK pathway. In insulin resistance, however, these pathways are not equally impaired. For example, in the liver, inhibition of gluconeogenesis by the insulin receptor substrate (IRS) 2 pathway is impaired, while lipogenesis by the IRS1 pathway is preserved, thus causing hyperglycemia and hyperlipidemia. It has been recently suggested that selective impairment of insulin signaling cascades in insulin resistance also occurs in the kidney. In the renal proximal tubule, insulin signaling via IRS1 is inhibited, while insulin signaling via IRS2 is preserved. Insulin signaling via IRS2 continues to stimulate sodium reabsorption in the proximal tubule and causes sodium retention, edema, and hypertension. IRS1 signaling deficiency in the proximal tubule may impair IRS1-mediated inhibition of gluconeogenesis, which could induce hyperglycemia by preserving glucose production. In the glomerulus, the impairment of IRS1 signaling deteriorates the structure and function of podocyte and endothelial cells, possibly causing diabetic nephropathy. This paper mainly describes selective insulin resistance in the kidney, focusing on the proximal tubule.
Collapse
|
25
|
Zaika O, Palygin O, Tomilin V, Mamenko M, Staruschenko A, Pochynyuk O. Insulin and IGF-1 activate Kir4.1/5.1 channels in cortical collecting duct principal cells to control basolateral membrane voltage. Am J Physiol Renal Physiol 2016; 310:F311-F321. [PMID: 26632606 PMCID: PMC4839479 DOI: 10.1152/ajprenal.00436.2015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 11/30/2015] [Indexed: 01/12/2023] Open
Abstract
Potassium Kir4.1/5.1 channels are abundantly expressed at the basolateral membrane of principal cells in the cortical collecting duct (CCD), where they are thought to modulate transport rates by controlling transepithelial voltage. Insulin and insulin-like growth factor-1 (IGF-1) stimulate apically localized epithelial sodium channels (ENaC) to augment sodium reabsorption in the CCD. However, little is known about their actions on potassium channels localized at the basolateral membrane. In this study, we implemented patch-clamp analysis in freshly isolated murine CCD to assess the effect of these hormones on Kir4.1/5.1 at both single channel and cellular levels. We demonstrated that K(+)-selective conductance via Kir4.1/5.1 is the major contributor to the macroscopic current recorded from the basolateral side in principal cells. Acute treatment with 10 μM amiloride (ENaC blocker), 100 nM tertiapin-Q (TPNQ; ROMK inhibitor), and 100 μM ouabain (Na(+)-K(+)-ATPase blocker) failed to produce a measurable effect on the macroscopic current. In contrast, Kir4.1 inhibitor nortriptyline (100 μM), but not fluoxetine (100 μM), virtually abolished whole cell K(+)-selective conductance. Insulin (100 nM) markedly increased the open probability of Kir4.1/5.1 and nortriptyline-sensitive whole cell current, leading to significant hyperpolarization of the basolateral membrane. Inhibition of the phosphatidylinositol 3-kinase cascade with LY294002 (20 μM) abolished action of insulin on Kir4.1/5.1. IGF-1 had similar stimulatory actions on Kir4.1/5.1-mediated conductance only when applied at a higher (500 nM) concentration and was ineffective at 100 nM. We concluded that both insulin and, to a lesser extent, IGF-1 activate Kir4.1/5.1 channel activity and open probability to hyperpolarize the basolateral membrane, thereby facilitating Na(+) reabsorption in the CCD.
Collapse
Affiliation(s)
- Oleg Zaika
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas
| | - Oleg Palygin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin; and
| | - Viktor Tomilin
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas; Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russian Federation
| | - Mykola Mamenko
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas
| | | | - Oleh Pochynyuk
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas;
| |
Collapse
|
26
|
Marlais M, Coward RJ. Paediatrics, insulin resistance and the kidney. Pediatr Nephrol 2015; 30:1217-24. [PMID: 25060762 DOI: 10.1007/s00467-014-2890-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Revised: 04/05/2014] [Accepted: 04/15/2014] [Indexed: 12/12/2022]
Abstract
Systemic insulin resistance is becoming more prevalent in the young due to modern lifestyles predisposing to the metabolic syndrome and obesity. There is also evidence that there are critical insulin-resistant phases for the developing child, including puberty, and that renal disease per se causes systemic insulin resistance. This review considers the factors that render children insulin resistant, as well as the accumulating evidence that the kidney is an insulin-responsive organ and could be affected by insulin resistance.
Collapse
Affiliation(s)
- Matko Marlais
- Institute of Child Health, University College London, London, United Kingdom
| | | |
Collapse
|
27
|
Ilatovskaya DV, Levchenko V, Brands MW, Pavlov TS, Staruschenko A. Cross-talk between insulin and IGF-1 receptors in the cortical collecting duct principal cells: implication for ENaC-mediated Na+ reabsorption. Am J Physiol Renal Physiol 2015; 308:F713-9. [PMID: 25651558 DOI: 10.1152/ajprenal.00081.2014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 01/20/2015] [Indexed: 12/17/2022] Open
Abstract
Insulin and IGF-1 are recognized as powerful regulators of the epithelial Na+ channel (ENaC) in the aldosterone-sensitive distal nephron. As previously described, these hormones both acutely increase ENaC activity in freshly isolated split open tubules and cultured principal cortical collecting duct cells. The present study was aimed at differentiating the effects of insulin and IGF-1 on Na+ transport in immortalized mpkCCDcl4 cells and defining their interrelations. We have shown that both insulin and IGF-1 applied basolaterally, but not apically, enhanced transepithelial Na+ transport in the mpkCCDcl4 cell line with EC50 values of 8.8 and 14.5 nM, respectively. Insulin treatment evoked phosphorylation of both insulin and IGF-1 receptors, whereas the effects of IGF-1 were more profound on its own receptor rather than the insulin receptor. AG-1024 and PPP, inhibitors of IGF-1 and insulin receptor tyrosine kinase activity, diminished insulin- and IGF-1-stimulated Na+ transport in mpkCCDcl4 cells. The effects of insulin and IGF-1 on ENaC-mediated currents were found to be additive, with insulin likely stimulating both IGF-1 and insulin receptors. We hypothesize that insulin activates IGF-1 receptors in addition to its own receptors, making the effects of these hormones interconnected.
Collapse
Affiliation(s)
- Daria V Ilatovskaya
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin; and
| | - Vladislav Levchenko
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin; and
| | - Michael W Brands
- Department of Physiology, Georgia Regents University, Augusta, Georgia
| | - Tengis S Pavlov
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin; and
| | | |
Collapse
|
28
|
Zaika O, Mamenko M, Boukelmoune N, Pochynyuk O. IGF-1 and insulin exert opposite actions on ClC-K2 activity in the cortical collecting ducts. Am J Physiol Renal Physiol 2015; 308:F39-F48. [PMID: 25339702 PMCID: PMC4281695 DOI: 10.1152/ajprenal.00545.2014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 10/16/2014] [Indexed: 01/07/2023] Open
Abstract
Despite similar stimulatory actions on the epithelial sodium channel (ENaC)-mediated sodium reabsorption in the distal tubule, insulin promotes kaliuresis, whereas insulin-like growth factor-1 (IGF-1) causes a reduction in urinary potassium levels. The factors contributing to this phenomenon remain elusive. Electrogenic distal nephron ENaC-mediated Na(+) transport establishes driving force for Cl(-) reabsorption and K(+) secretion. Using patch-clamp electrophysiology, we document that a Cl(-) channel is highly abundant on the basolateral plasma membrane of intercalated cells in freshly isolated mouse cortical collecting duct (CCD) cells. The channel has characteristics attributable to the ClC-K2: slow gating kinetics, conductance ∼10 pS, voltage independence, Cl(-)>NO3 (-) anion selectivity, and inhibition/activation by low/high pH, respectively. IGF-1 (100 and 500 nM) acutely stimulates ClC-K2 activity in a reversible manner. Inhibition of PI3-kinase (PI3-K) with LY294002 (20 μM) abrogates activation of ClC-K2 by IGF-1. Interestingly, insulin (100 nM) reversibly decreases ClC-K2 activity in CCD cells. This inhibitory action is independent of PI3-K and is mediated by stimulation of a mitogen-activated protein kinase-dependent cascade. We propose that IGF-1, by stimulating ClC-K2 channels, promotes net Na(+) and Cl(-) reabsorption, thus reducing driving force for potassium secretion by the CCD. In contrast, inhibition of ClC-K2 by insulin favors coupling of Na(+) reabsorption with K(+) secretion at the apical membrane contributing to kaliuresis.
Collapse
Affiliation(s)
- Oleg Zaika
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas
| | - Mykola Mamenko
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas
| | - Nabila Boukelmoune
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas
| | - Oleh Pochynyuk
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas
| |
Collapse
|
29
|
Sharma N, Li L, Ecelbarger CM. Sex differences in renal and metabolic responses to a high-fructose diet in mice. Am J Physiol Renal Physiol 2014; 308:F400-10. [PMID: 25537743 DOI: 10.1152/ajprenal.00403.2014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
High fructose intake has been associated with increased incidences of renal disease and hypertension, among other pathologies. Most fructose is cleared by the portal system and metabolized in the liver; however, systemic levels of fructose can rise with increased consumption. We tested whether there were sex differences in the renal responses to a high-fructose diet in mice. Two-month-old male and female C57BL6/129/SV mice (n = 6 mice per sex per treatment) were randomized to receive control or high-fructose (65% by weight) diets as pelleted chow ad libitum for 3 mo. Fructose feeding did not significantly affect body weight but led to a 19% and 10% increase in kidney weight in male and female mice, respectively. In male mice, fructose increased the expression (∼50%) of renal cortical proteins involved in metabolism, including glucose transporter 5 (facilitative fructose transporter), ketohexokinase, and the insulin receptor (β-subunit). Female mice had lower basal levels of glucose transporter 5, which were unresponsive to fructose. However, female mice had increased urine volume and plasma K(+) and decreased plasma Na(+) with fructose, whereas male mice were less affected. Likewise, female mice showed a two- to threefold reduction in the expression Na(+)-K(+)-2Cl(-) cotransporter 2 in the thick ascending limb and aquaporin-2 in the collecting duct with fructose relative to female control mice, whereas male mice had no change. Overall, our results support greater proximal metabolism of fructose in male animals and greater distal tubule/collecting duct (electrolyte homeostasis) alterations in female animals. These sex differences may be important determinants of the specific nature of pathologies that develop in association with high fructose consumption.
Collapse
Affiliation(s)
- Nikhil Sharma
- Division of Endocrinology and Metabolism, Georgetown University, Washington, District of Columbia; and
| | - Lijun Li
- Division of Endocrinology and Metabolism, Georgetown University, Washington, District of Columbia; and Center for the Study of Sex Differences in Health, Aging, and Disease, Department of Medicine, Georgetown University, Washington, District of Columbia
| | - C M Ecelbarger
- Division of Endocrinology and Metabolism, Georgetown University, Washington, District of Columbia; and Center for the Study of Sex Differences in Health, Aging, and Disease, Department of Medicine, Georgetown University, Washington, District of Columbia
| |
Collapse
|
30
|
Zelenchuk LV, Hedge AM, Rowe PSN. PHEX mimetic (SPR4-peptide) corrects and improves HYP and wild type mice energy-metabolism. PLoS One 2014; 9:e97326. [PMID: 24839967 PMCID: PMC4026222 DOI: 10.1371/journal.pone.0097326] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 04/17/2014] [Indexed: 12/19/2022] Open
Abstract
CONTEXT PHEX or DMP1 mutations cause hypophosphatemic-rickets and altered energy metabolism. PHEX binds to DMP1-ASARM-motif to form a complex with α5β3 integrin that suppresses FGF23 expression. ASARM-peptides increase FGF23 by disrupting the PHEX-DMP1-Integrin complex. We used a 4.2 kDa peptide (SPR4) that binds to ASARM-peptide/motif to study the DMP1-PHEX interaction and to assess SPR4 for the treatment of energy metabolism defects in HYP and potentially other bone-mineral disorders. DESIGN Subcutaneously transplanted osmotic pumps were used to infuse SPR4-peptide or vehicle (VE) into wild-type mice (WT) and HYP-mice (PHEX mutation) for 4 weeks. RESULTS SPR4 partially corrected HYP mice hypophosphatemia and increased serum 1.25(OH)2D3. Serum FGF23 remained high and PTH was unaffected. WT-SPR4 mice developed hypophosphatemia and hypercalcemia with increased PTH, FGF23 and 1.25(OH)2D3. SPR4 increased GAPDH HYP-bone expression 60× and corrected HYP-mice hyperglycemia and hypoinsulinemia. HYP-VE serum uric-acid (UA) levels were reduced and SPR4 infusion suppressed UA levels in WT-mice but not HYP-mice. SPR4 altered leptin, adiponectin, and sympathetic-tone and increased the fat mass/weight ratio for HYP and WT mice. Expression of perlipin-2 a gene involved in obesity was reduced in HYP-VE and WT-SPR4 mice but increased in HYP-SPR4 mice. Also, increased expression of two genes that inhibit insulin-signaling, ENPP1 and ESP, occurred with HYP-VE mice. In contrast, SPR4 reduced expression of both ENPP1 and ESP in WT mice and suppressed ENPP1 in HYP mice. Increased expression of FAM20C and sclerostin occurred with HYP-VE mice. SPR4 suppressed expression of FAM20C and sclerostin in HYP and WT mice. CONCLUSIONS ASARM peptides and motifs are physiological substrates for PHEX and modulate osteocyte PHEX-DMP1-α5β3-integrin interactions and thereby FGF23 expression. These interactions also provide a nexus that regulates bone and energy metabolism. SPR4 suppression of sclerostin and/or sequestration of ASARM-peptides improves energy metabolism and may have utility for treating familial rickets, osteoporosis, obesity and diabetes.
Collapse
Affiliation(s)
- Lesya V. Zelenchuk
- Internal Medicine, The Kidney Institute, Kansas University Medical Center (KUMC), Kansas City, Kansas, United States of America
| | - Anne-Marie Hedge
- Internal Medicine, The Kidney Institute, Kansas University Medical Center (KUMC), Kansas City, Kansas, United States of America
| | - Peter S. N. Rowe
- Internal Medicine, The Kidney Institute, Kansas University Medical Center (KUMC), Kansas City, Kansas, United States of America
| |
Collapse
|
31
|
Lay A, Coward RJ. Recent advances in our understanding of insulin signalling to the podocyte. Nephrol Dial Transplant 2013; 29:1127-33. [PMID: 24286976 DOI: 10.1093/ndt/gft471] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
It is becoming increasingly clear that the insulin responses of a number of different cell types within the kidney are important in the maintenance of normal renal function. This review summarizes our current understanding of renal insulin signalling, with specific focus on the podocyte, presenting recent evidence that suggests these responses are altered in systemic insulin-resistant states and chronic kidney disease via a number of different mechanisms.
Collapse
Affiliation(s)
- Abigail Lay
- Academic Renal Unit, Learning and Research Building, Southmead Hospital, School of Clinical Sciences, University of Bristol, Bristol BS10 5NB, UK
| | - Richard J Coward
- Academic Renal Unit, Learning and Research Building, Southmead Hospital, School of Clinical Sciences, University of Bristol, Bristol BS10 5NB, UK
| |
Collapse
|
32
|
Cunard R. Mammalian tribbles homologs at the crossroads of endoplasmic reticulum stress and Mammalian target of rapamycin pathways. SCIENTIFICA 2013; 2013:750871. [PMID: 24490110 PMCID: PMC3892554 DOI: 10.1155/2013/750871] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 11/20/2013] [Indexed: 05/03/2023]
Abstract
In 2000, investigators discovered Tribbles, a Drosophila protein that coordinates morphogenesis by inhibiting mitosis. Further work has delineated Xenopus (Xtrb2), Nematode (Nipi-3), and mammalian homologs of Drosophila tribbles, which include TRB1, TRB2, and TRB3. The sequences of tribbles homologs are highly conserved, and despite their protein kinase structure, to date they have not been shown to have kinase activity. TRB family members play a role in the differentiation of macrophages, lymphocytes, muscle cells, adipocytes, and osteoblasts. TRB isoforms also coordinate a number of critical cellular processes including glucose and lipid metabolism, inflammation, cellular stress, survival, apoptosis, and tumorigenesis. TRB family members modulate multiple complex signaling networks including mitogen activated protein kinase cascades, protein kinase B/AKT signaling, mammalian target of rapamycin, and inflammatory pathways. The following review will discuss metazoan homologs of Drosophila tribbles, their structure, expression patterns, and functions. In particular, we will focus on TRB3 function in the kidney in podocytes. This review will also discuss the key signaling pathways with which tribbles proteins interact and provide a rationale for developing novel therapeutics that exploit these interactions to provide better treatment options for both acute and chronic kidney disease.
Collapse
Affiliation(s)
- Robyn Cunard
- Research Service and Division of Nephrology-Hypertension, Veterans Affairs San Diego Healthcare System, Veterans Medical Research Foundation, Mail Code 151, 3350 La Jolla Village Drive, San Diego, CA 92161, USA
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- *Robyn Cunard:
| |
Collapse
|