1
|
Cinakova A, Vavrincova-Yaghi D, Krenek P, Klimas J, Kralova E. Combination of dapagliflozin and pioglitazone lacks superiority against monotherapy in streptozotocin-induced nephropathy. Sci Rep 2025; 15:1464. [PMID: 39789116 PMCID: PMC11718164 DOI: 10.1038/s41598-024-84487-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/24/2024] [Indexed: 01/12/2025] Open
Abstract
Oxidative stress and apoptosis are highly engaged in development of diabetic nephropathy (DN). In monotherapy, dapagliflozin and pioglitazone positively modulate target organ damage even independently of their hypoglycaemic effect. This study evaluated whether a simultaneous PPARγ activation and SGLT cotransporter inhibition offer superior protection against DN-related oxidative and apoptotic processes in a T1DM rat model. Diabetes was induced in Wistar rats using streptozotocin (55 mg/kg, i.p.). The rats received daily chow containing dapagliflozin (10 mg/kg), pioglitazone (12 mg/kg) or their combination. Six weeks after STZ administration, histological and molecular analyses were performed in excised kidneys. STZ-induced DN was demonstrated by the propagation of apoptotic (Bax, p53, Casp3) and oxidative reactions (Gp91phox, MnSOD) and disrupted nitric oxide signalling (eNOS, Hsp90, Cav1). Kidney damage molecule expression (Kim1, Nphs1) revealed a deceleration of kidney damage by pioglitazone and dapagliflozine monotherapies. The monotherapy also reduced apoptosis, oxidative stress, and partially restored NO signalling. The combined therapy ameliorated glomerulosclerosis but in other measured parameters, it reached the effect of the monotherapies except for Hsp90 expression modulation. Both dapagliflozin and pioglitazone exert protective character in kidneys when used in monotherapy. The combined therapy does not exhibit an expected additive effect within modulating oxidative stress, NO signalling or apoptosis.
Collapse
Affiliation(s)
- Aneta Cinakova
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Comenius University Bratislava, SK-83232, Bratislava, Slovakia
| | - Diana Vavrincova-Yaghi
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Comenius University Bratislava, SK-83232, Bratislava, Slovakia
| | - Peter Krenek
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Comenius University Bratislava, SK-83232, Bratislava, Slovakia
| | - Jan Klimas
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Comenius University Bratislava, SK-83232, Bratislava, Slovakia
| | - Eva Kralova
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Comenius University Bratislava, SK-83232, Bratislava, Slovakia.
| |
Collapse
|
2
|
Lee SR, Lee HE, Yoo JY, An EJ, Song SJ, Han KH, Cha DR, Bae YS. Nox4-SH3YL1 complex is involved in diabetic nephropathy. iScience 2024; 27:108868. [PMID: 38318360 PMCID: PMC10839645 DOI: 10.1016/j.isci.2024.108868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 10/25/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
Nox4-derived H2O2 generation plays an important role in the pathogenesis of chronic kidney diseases (CKDs) such as diabetic nephropathy (DN). Here, we showed that SH3 domain-containing Ysc84-like 1 (SH3YL1), a Nox4 cytosolic activator, regulated DN. Streptozotocin (STZ)-induced type Ⅰ diabetic models in SH3YL1 whole-body knockout (KO) mice and podocyte-specific SH3YL1 conditional KO (Nphs2-Cre/SH3YL1fl/fl) mice were established to investigate the function of SH3YL1 in DN. The expression of fibrosis markers and inflammatory cytokines, the generation of oxidative stress, and the loss of podocytes were suppressed in diabetic SH3YL1 KO and Nphs2-Cre/SH3YL1fl/fl mice, compared to diabetic control mice. To extrapolate the observations derived from diabetic mice to clinical implication, we measured the protein level of SH3YL1 in patients DN. In fact, the SH3YL1 level was increased in patients DN. Overall, the SH3YL1-Nox4 complex was identified to play an important role in renal inflammation and fibrosis, resulting in the development of DN.
Collapse
Affiliation(s)
- Sae Rom Lee
- Department of Life Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Hye Eun Lee
- Department of Life Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Jung-Yeon Yoo
- Department of Life Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Eun Jung An
- Department of Life Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Soo-Jin Song
- Department of Anatomy, Ewha Womans University College of Medicine, Seoul 07804, Korea
| | - Ki-Hwan Han
- Department of Anatomy, Ewha Womans University College of Medicine, Seoul 07804, Korea
| | - Dae Ryong Cha
- Department of Internal Medicine, Division of Nephrology, Korea University Ansan Hospital, 516 Kojan-Dong, Ansan City, Kyungki-Do 15355, Korea
| | - Yun Soo Bae
- Department of Life Sciences, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
3
|
Mitrofanova A, Merscher S, Fornoni A. Kidney lipid dysmetabolism and lipid droplet accumulation in chronic kidney disease. Nat Rev Nephrol 2023; 19:629-645. [PMID: 37500941 DOI: 10.1038/s41581-023-00741-w] [Citation(s) in RCA: 87] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2023] [Indexed: 07/29/2023]
Abstract
Chronic kidney disease (CKD) is a global health problem with rising incidence and prevalence. Among several pathogenetic mechanisms responsible for disease progression, lipid accumulation in the kidney parenchyma might drive inflammation and fibrosis, as has been described in fatty liver diseases. Lipids and their metabolites have several important structural and functional roles, as they are constituents of cell and organelle membranes, serve as signalling molecules and are used for energy production. However, although lipids can be stored in lipid droplets to maintain lipid homeostasis, lipid accumulation can become pathogenic. Understanding the mechanisms linking kidney parenchymal lipid accumulation to CKD of metabolic or non-metabolic origin is challenging, owing to the tremendous variety of lipid species and their functional diversity across different parenchymal cells. Nonetheless, multiple research reports have begun to emphasize the effect of dysregulated kidney lipid metabolism in CKD progression. For example, altered cholesterol and fatty acid metabolism contribute to glomerular and tubular cell injury. Newly developed lipid-targeting agents are being tested in clinical trials in CKD, raising expectations for further therapeutic development in this field.
Collapse
Affiliation(s)
- Alla Mitrofanova
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Sandra Merscher
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL, USA.
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
4
|
Leodori G, Pellicano C, Basile V, Colalillo A, Navarini L, Gigante A, Gulli F, Marino M, Basile U, Rosato E. Serum Adiponectin, a Novel Biomarker Correlates with Skin Thickness in Systemic Sclerosis. J Pers Med 2022; 12:jpm12101737. [PMID: 36294874 PMCID: PMC9604668 DOI: 10.3390/jpm12101737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/11/2022] [Accepted: 10/17/2022] [Indexed: 12/03/2022] Open
Abstract
The aim was to evaluate the longitudinal association between basal serum adiponectin and repeated measurements of skin thickness during 12 months of follow-up in systemic sclerosis (SSc) patients. We enrolled SSc patients with disease duration > 2 years in a prospective observational study. Skin thickness was measured at baseline and after 12 months of follow-up with modified Rodnan skin score (mRSS). Baseline serum adiponectin was determined using a commercial ELISA kit. We enrolled 66 female SSc patients (median age 54 years, IQR 42−62 years). The median disease duration was 12 (IQR 8−16) years and median baseline serum adiponectin was 9.8 (IQR 5.6−15.6) mcg/mL. The median mRSS was 10 (IQR 6−18) at baseline and 12 (IQR 7−18) at follow-up. A significant correlation was observed between baseline serum adiponectin and disease duration (r = 0.264, p < 0.05), age (r = 0.515, p < 0.0001), baseline mRSS (r = −0.303, p < 0.05), and mRSS at follow-up (r = −0.322, p < 0.001). In multiple regression analysis, only mRSS at follow-up showed an inverse correlation with baseline serum adiponectin (β = −0.132, p < 0.01). The reduction in serum adiponectin levels is correlated with skin thickness.
Collapse
Affiliation(s)
- Giorgia Leodori
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00189 Rome, Italy
| | - Chiara Pellicano
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00189 Rome, Italy
| | - Valerio Basile
- Clinical Pathology Unit and Cancer Biobank, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Amalia Colalillo
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00189 Rome, Italy
| | - Luca Navarini
- Unit of Allergology, Clinical Immunology and Rheumatology, Campus Bio-Medico University of Rome, 00128 Rome, Italy
| | - Antonietta Gigante
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00189 Rome, Italy
| | - Francesca Gulli
- Clinical Biochemistry Laboratory, IRCCS “Bambino Gesù” Children’s Hospital, 00165 Rome, Italy
| | - Mariapaola Marino
- Department of Translational Medicine and Surgery, Section of General Pathology, “A. Gemelli” IRCCS, Catholic University of the Sacred Heart, 00168 Rome, Italy
- Correspondence: (M.M.); (U.B.)
| | - Umberto Basile
- Department of Laboratory and Infectious Disease Sciences, “A. Gemelli” IRCCS, Catholic University of the Sacred Heart, 00168 Rome, Italy
- Correspondence: (M.M.); (U.B.)
| | - Edoardo Rosato
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00189 Rome, Italy
| |
Collapse
|
5
|
Liu Y, Chen S, Yu L, Deng Y, Li D, Yu X, Chen D, Lu Y, Liu S, Chen R. Pemafibrate attenuates pulmonary fibrosis by inhibiting myofibroblast differentiation. Int Immunopharmacol 2022; 108:108728. [PMID: 35397395 DOI: 10.1016/j.intimp.2022.108728] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/26/2022] [Accepted: 03/18/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND OBJECTIVE Idiopathic pulmonary fibrosis is a chronic progressive disease associated with substantial morbidity and mortality despite advances in medical therapy. Increasing evidence suggests that peroxisome proliferator-activated receptors (PPARs) play important roles in the fibrosis-related diseases and their agonists may become effective therapeutic targets. Pemafibrate is a selective PPARα agonist, but the efficacy against pulmonary fibrosis and mechanisms involved have not been systematically evaluated. Thus, the aims of this study were to explore the role of PPARα in the pulmonary fibrosis and to assess the effect of pemafibrate in vivo and in vitro. METHODS The effects of pemafibrate were evaluated in bleomycin-challenged murine pulmonary fibrosis model and transforming growth factor-beta 1 (TGF-β1) stimulated lung fibroblasts. RESULTS Bleomycin instillation induced body weight loss, declined lung function, pulmonary fibrosis, and extensive collagen deposition in the mice, accompanied with decreased pulmonary expression of PPARα, all of which were partially improved by pemafibrate at a dose of 2 mg/kg. Besides, pemafibrate effectively inhibits TGF-β1-induced myofibroblast differentiation and extracellular matrix (ECM) production in vivo and in vitro. Furthermore, we showed that pemafibrate not only inhibited pulmonary expression of NLRP3 and cleaved caspase-1 in bleomycin-inhaled mice, but also repressed activation of NLRP3/caspase-1 axis in TGF-β1 stimulated lung fibroblasts. CONCLUSION Our data suggest that pemafibrate exerts a marked protection against from the development of pulmonary fibrosis, which could constitute a novel candidate for the treatment for pulmonary fibrosis.
Collapse
Affiliation(s)
- Yuanyuan Liu
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China; Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Shuyu Chen
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China; Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Li Yu
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
| | - Yao Deng
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
| | - Difei Li
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
| | - Xiu Yu
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
| | - Dandan Chen
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
| | - Ye Lu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shengming Liu
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Jinan University, Guangzhou, China.
| | - Rongchang Chen
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China.
| |
Collapse
|
6
|
Wang S, Liang Y, Dai C. Metabolic Regulation of Fibroblast Activation and Proliferation during Organ Fibrosis. KIDNEY DISEASES (BASEL, SWITZERLAND) 2022; 8:115-125. [PMID: 35527985 DOI: 10.1159/000522417] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/29/2022] [Indexed: 12/13/2022]
Abstract
Background Activated fibroblasts are present in the injury response, tumorigenesis, fibrosis, and inflammation in a variety of tissues and myriad disease types. Summary During normal tissue repair, quiescent fibroblasts transform into a proliferative and contractile phenotype termed myofibroblasts and are then lost as repair resolves to form a scar. When excessive levels are reached, activated fibroblasts proliferate and produce large amounts of extracellular matrix, which accumulates in the interstitial space of different organs. This accumulation leads to fibrotic dysfunction and multiple-organ dysfunction syndrome. To date, there are limited effective treatments for these conditions. Cellular metabolism is the cornerstone of all biological activities. Emerging evidence shows that metabolic alterations in fibroblasts are important for the activation process and illness progression. These discoveries, along with current clinical advances showing decreased lung fibrosis after targeting specific metabolic pathways, thus offer new possibilities for therapeutic interventions. The purpose of this review was to summarize the most recent knowledge of the major metabolic changes that occur during fibroblast transition from quiescent to activated states and the evidence linking alterations in fibroblast metabolism to the pathobiology of several common fibrotic diseases and tumor-related diseases. Key Messages Metabolic disorders are associated with the progression of chronic kidney diseases. Interfering with fibroblast metabolism may be a promising therapeutic strategy for renal fibrosis and other fibrosis-related diseases.
Collapse
Affiliation(s)
- Sudan Wang
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Yan Liang
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Chunsun Dai
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, China.,Department of Clinical Genetics, 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
7
|
Ung CY, Onoufriadis A, Parsons M, McGrath JA, Shaw TJ. Metabolic perturbations in fibrosis disease. Int J Biochem Cell Biol 2021; 139:106073. [PMID: 34461262 DOI: 10.1016/j.biocel.2021.106073] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/09/2021] [Accepted: 08/25/2021] [Indexed: 12/19/2022]
Abstract
Metabolic changes occur in all forms of disease but their impact on fibrosis is a relatively recent area of interest. This review provides an overview of the major metabolic pathways, glycolysis, amino acid metabolism and lipid metabolism, and highlights how they influence fibrosis at a cellular and tissue level, drawing on key discoveries in dermal, renal, pulmonary and hepatic fibrosis. The emerging influence of adipose tissue-derived cytokines is discussed and brings a link between fibrosis and systemic metabolism. To close, the concept of targeting metabolism for fibrotic therapy is reviewed, drawing on lessons from the more established field of cancer metabolism, with an emphasis on important considerations for clinical translation.
Collapse
Affiliation(s)
- Chuin Ying Ung
- St John's Institute of Dermatology, King's College London, London, SE19RT, UK.
| | | | - Maddy Parsons
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, SE11UL, UK.
| | - John A McGrath
- St John's Institute of Dermatology, King's College London, London, SE19RT, UK.
| | - Tanya J Shaw
- Centre for Inflammation Biology & Cancer Immunology, King's College London, London, SE1 1UL, UK.
| |
Collapse
|
8
|
Nuwormegbe S, Park NY, Kim SW. Lobeglitazone attenuates fibrosis in corneal fibroblasts by interrupting TGF-beta-mediated Smad signaling. Graefes Arch Clin Exp Ophthalmol 2021; 260:149-162. [PMID: 34468828 DOI: 10.1007/s00417-021-05370-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 07/30/2021] [Accepted: 08/06/2021] [Indexed: 12/13/2022] Open
Abstract
PURPOSE Transforming growth factor beta 1 (TGF-β1) is an important cytokine released after ocular surface injury to promote wound healing. However, its persistence at the injury site triggers a fibrotic response that leads to corneal scarring and opacity. Thiazolidinediones (TZDs) are synthetic peroxisome proliferator-activated receptor gamma (PPAR-γ) ligands used to regulate glucose and lipid metabolism in the management of type 2 diabetes. Studies have also showed TZDs have antifibrotic effect. In this study, we investigated the antifibrotic effect of the TZD lobeglitazone on TGF-β1-induced fibrosis in corneal fibroblasts. METHODS Human primary corneal fibroblasts were cultivated and treated with TGF-β1 (5 ng/mL) to induce fibrosis, with or without pre-treatments with different concentrations of lobeglitazone. Myofibroblast differentiation and extracellular matrix (ECM) protein expression was evaluated by western blotting, immunofluorescence, real-time PCR, and collagen gel contraction assay. The effect of lobeglitazone on TGF-β1-induced reactive oxygen species (ROS) generation was evaluated by DCFDA-cellular ROS detection assay kit. Signaling proteins were evaluated by western blotting to determine the mechanism underlying the antifibrotic effect. RESULTS Our results showed lobeglitazone attenuated TGF-β1-induced ECM synthesis and myofibroblast differentiation of corneal fibroblasts. This antifibrotic effect appeared to be independent of PPAR signaling and rather due to the inhibition of the TGF-β1-induced Smad signaling. Lobeglitazone also blocked TGF-β1-induced ROS generation and nicotinamide adenine dinucleotide phosphate oxidase (Nox) 4 transcription. CONCLUSION These findings indicate that lobeglitazone may be a promising therapeutic agent for corneal scarring. KEY MESSAGES.
Collapse
Affiliation(s)
- Selikem Nuwormegbe
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju, Ilsan-ro, Gangwon-do, 26426, Republic of Korea
| | - Na-Young Park
- Department of Ophthalmology, Wonju College of Medicine, Yonsei University, Wonju, Ilsan-ro, Gangwon-do, 26426, Republic of Korea
| | - Sun Woong Kim
- Department of Ophthalmology, Wonju College of Medicine, Yonsei University, Wonju, Ilsan-ro, Gangwon-do, 26426, Republic of Korea.
| |
Collapse
|
9
|
Use of Lipid-Modifying Agents for the Treatment of Glomerular Diseases. J Pers Med 2021; 11:jpm11080820. [PMID: 34442464 PMCID: PMC8401447 DOI: 10.3390/jpm11080820] [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: 07/16/2021] [Accepted: 08/17/2021] [Indexed: 01/14/2023] Open
Abstract
Although dyslipidemia is associated with chronic kidney disease (CKD), it is more common in nephrotic syndrome (NS), and guidelines for the management of hyperlipidemia in NS are largely opinion-based. In addition to the role of circulating lipids, an increasing number of studies suggest that intrarenal lipids contribute to the progression of glomerular diseases, indicating that proteinuric kidney diseases may be a form of "fatty kidney disease" and that reducing intracellular lipids could represent a new therapeutic approach to slow the progression of CKD. In this review, we summarize recent progress made in the utilization of lipid-modifying agents to lower renal parenchymal lipid accumulation and to prevent or reduce kidney injury. The agents mentioned in this review are categorized according to their specific targets, but they may also regulate other lipid-relevant pathways.
Collapse
|
10
|
Libby AE, Jones B, Lopez-Santiago I, Rowland E, Levi M. Nuclear receptors in the kidney during health and disease. Mol Aspects Med 2020; 78:100935. [PMID: 33272705 DOI: 10.1016/j.mam.2020.100935] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/24/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023]
Abstract
Over the last 30 years, nuclear receptors (NRs) have been increasingly recognized as key modulators of systemic homeostasis and as contributing factors in many diseases. In the kidney, NRs play numerous important roles in maintaining homeostasis-many of which continue to be unraveled. As "master regulators", these important transcription factors integrate and coordinate many renal processes such as circadian responses, lipid metabolism, fatty acid oxidation, glucose handling, and inflammatory responses. The use of recently-developed genetic tools and small molecule modulators have allowed for detailed studies of how renal NRs contribute to kidney homeostasis. Importantly, while NRs are intimately involved in proper kidney function, they are also implicated in a variety of renal diseases such as diabetes, acute kidney injury, and other conditions such as aging. In the last 10 years, our understanding of renal disease etiology and progression has been greatly shaped by knowledge regarding how NRs are dysregulated in these conditions. Importantly, NRs have also become attractive therapeutic targets for attenuation of renal diseases, and their modulation for this purpose has been the subject of intense investigation. Here, we review the role in health and disease of six key renal NRs including the peroxisome proliferator-activated receptors (PPAR), estrogen-related receptors (ERR), the farnesoid X receptors (FXR), estrogen receptors (ER), liver X receptors (LXR), and vitamin D receptors (VDR) with an emphasis on recent findings over the last decade. These NRs have generated a wealth of data over the last 10 years that demonstrate their crucial role in maintaining normal renal homeostasis as well as their capacity to modulate disease progression.
Collapse
Affiliation(s)
- Andrew E Libby
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, 3900 Reservoir Rd, Washington, DC, 20007, USA.
| | - Bryce Jones
- Department of Pharmacology and Physiology, Georgetown University, 3900 Reservoir Rd, Washington, DC, 20007, USA.
| | - Isabel Lopez-Santiago
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, 3900 Reservoir Rd, Washington, DC, 20007, USA.
| | - Emma Rowland
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, 3900 Reservoir Rd, Washington, DC, 20007, USA.
| | - Moshe Levi
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, 3900 Reservoir Rd, Washington, DC, 20007, USA.
| |
Collapse
|
11
|
Involvement of FATP2-mediated tubular lipid metabolic reprogramming in renal fibrogenesis. Cell Death Dis 2020; 11:994. [PMID: 33219209 PMCID: PMC7679409 DOI: 10.1038/s41419-020-03199-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 10/04/2020] [Accepted: 10/06/2020] [Indexed: 12/13/2022]
Abstract
Following a chronic insult, renal tubular epithelial cells (TECs) contribute to the development of kidney fibrosis through dysregulated lipid metabolism that lead to lipid accumulation and lipotoxicity. Intracellular lipid metabolism is tightly controlled by fatty acids (FAs) uptake, oxidation, lipogenesis, and lipolysis. Although it is widely accepted that impaired fatty acids oxidation (FAO) play a crucial role in renal fibrosis progression, other lipid metabolic pathways, especially FAs uptake, has not been investigated in fibrotic kidney. In this study, we aim to explore the potential mechanically role of FAs transporter in the pathogenesis of renal fibrosis. In the present study, the unbiased gene expression studies showed that fatty acid transporter 2 (FATP2) was one of the predominant expressed FAs transport in TECs and its expression was tightly associated with the decline of renal function. Treatment of unilateral ureteral obstruction (UUO) kidneys and TGF-β induced TECs with FATP2 inhibitor (FATP2i) lipofermata restored the FAO activities and alleviated fibrotic responses both in vivo and in vitro. Moreover, the expression of profibrotic cytokines including TGF-β, connective tissue growth factor (CTGF), fibroblast growth factor (FGF), and platelet-derived growth factor subunit B (PDGFB) were all decreased in FATP2i-treated UUO kidneys. Mechanically, FATP2i can effectively attenuate cell apoptosis and endoplasmic reticulum (ER) stress induced by TGF-β treatment in cultured TECs. Taking together, these findings reveal that FATP2 elicits a profibrotic response to renal interstitial fibrosis by inducing lipid metabolic reprogramming including abnormal FAs uptake and defective FAO in TECs.
Collapse
|
12
|
Improved Renoprotection in Diabetes with Combination Therapy of Coccinia indica Leaf Extract and Low-Dose Pioglitazone. SEPARATIONS 2020. [DOI: 10.3390/separations7040058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: The metabolic changes associated with diabetes can lead to nephropathy eventually resulting in end-stage renal disease. Current antidiabetic therapies do not effectively prevent the onset of diabetic kidney diseases as well as progression. Aim: To evaluate the effect of Coccinia indica leaf extract alone and in combination with pioglitazone, an antihyperglycemic agent was used to modulate the progressive kidney damage induced by type 2 diabetes in rats. Hypotheses: Pioglitazone causes severe adverse effects when administered for long-term therapy. The hypotheses in this study is to examine the renoprotective effect of Coccinia indica leaf extract (200 mg/kg p.o.) when co-administered with low-dose pioglitazone (7 mg/kg) in type-2-diabetes-induced nephropathy in rats and simultaneously evaluate the hypoglycemic response as well. Methods: Rats (Males, Sprague Dawley) were kept on a high-fat diet and were given a single dose of streptozotocin (35 mg/kg, i.p.) to induce diabetic nephropathy. Treatment groups received either Coccinia indica leaf extract or pioglitazone or pioglitazone with Coccinia indica extract, fenofibrate, or lisinopril for 7 weeks. Blood glucose, antioxidant status, triglycerides, total cholesterol, creatinine, blood urea nitrogen, and proteinuria levels were estimated and compared with the normal control and disease control (untreated) groups. Results: The untreated diabetic rats showed increased blood glucose levels, lipid profiles, and renal oxidative stress, along with an increase in nephropathy markers such as blood urea nitrogen, creatinine, and proteinuria. Histopathological examination revealed glomerular damage. Combination treatment with Coccinia indica leaf extract and a low dose of pioglitazone normalized the nephropathic markers as well as histopathological changes. Conclusion: Coccinia indica leaf extract when co-administered with a low dose of pioglitazone as antidiabetic therapy showed good glycemic control and a beneficial renoprotective effect. Combination therapy would lower the dose of pioglitazone and also protect kidneys from drug-induced toxicity as observed from normalized nephropathic markers in a diabetic rat model.
Collapse
|
13
|
Capsaicin up-regulates pro-apoptotic activity of thiazolidinediones in glioblastoma cell line. Biomed Pharmacother 2020; 132:110741. [PMID: 33038582 DOI: 10.1016/j.biopha.2020.110741] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/02/2020] [Accepted: 09/07/2020] [Indexed: 12/13/2022] Open
Abstract
Capsaicin (N-vanillyl-8-methyl-alpha-nonenamide), a spicy, neurotoxic component of hot pepper is a ligand of vanilloid type-I (TRPV1) receptor of anti-cancer potential. However, molecular mechanism of its action is not fully understood. We found that capsaicin stimulated intrinsic and extrinsic pathway of apoptosis in human glioblastoma LN-18 cell line and this phenomenon was not dependent on TRPV1. Activation of peroxisome proliferator-activated receptor gamma (PPARγ), a ligand-dependent transcription factor, also induced apoptosis in glioblastoma cells. Although PPARγ ligands (thiazolidinediones - rosiglitazone, pioglitazone) promoted apoptosis in LN-18 cells, capsaicin augmented this effect. We found that capsaicin in a dose dependent manner induced expression of PPARγ in glioblastoma LN-18 cells. These findings suggest that capsaicin-dependent up-regulation of PPARγ represent the mechanism for augmentation of cell death by thiazolidinediones.
Collapse
|
14
|
Mulder S, Perco P, Oxlund C, Mehdi UF, Hankemeier T, Jacobsen IA, Toto R, Heerspink HJL, Pena MJ. Baseline urinary metabolites predict albuminuria response to spironolactone in type 2 diabetes. Transl Res 2020; 222:17-27. [PMID: 32438071 DOI: 10.1016/j.trsl.2020.04.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 04/09/2020] [Accepted: 04/11/2020] [Indexed: 12/14/2022]
Abstract
The mineralocorticoid receptor antagonist spironolactone significantly reduces albuminuria in subjects with diabetic kidney disease, albeit with a large variability between individuals. Identifying novel biomarkers that predict response to therapy may help to tailor spironolactone therapy. We aimed to identify a set of metabolites for prediction of albuminuria response to spironolactone in subjects with type 2 diabetes. Systems biology molecular process analysis was performed a priori to identify metabolites linked to molecular disease processes and drug mechanism of action. Individual subject data and urine samples were used from 2 randomized placebo controlled double blind clinical trials (NCT01062763, NCT00381134). A urinary metabolite score was developed to predict albuminuria response to spironolactone therapy using penalized ridge regression with leave-one-out cross validation. Bioinformatic analysis identified a set of 18 metabolites linked to a diabetic kidney disease molecular model and potentially affected by spironolactone mechanism of action. Spironolactone reduced UACR relative to placebo by median -42% (25th to 75% percentile -65 to 6) and -29% (25th to 75% percentile -37 to -1) in the test and replication cohorts, respectively. In the test cohort, UACR reduction was higher in the lowest tertile of the baseline urinary metabolite score compared with middle and upper tertiles -58% (25th to 75% percentile -78 to 33), -28% (25th to 75% percentile -46 to 8), -40% (25th to 75% percentile -52% to 31), respectively, P = 0.001 for trend). In the replication cohort, UACR reduction was -54% (25th to 75% percentile -65 to -50), -41 (25th to 75% percentile -46% to 30), and -17% (25th to 75% percentile -36 to 5), respectively, P = 0.010 for trend). We identified a set of 18 urinary metabolites through systems biology to predict albuminuria response to spironolactone in type 2 diabetes. These data suggest that urinary metabolites may be used as a tool to tailor optimal therapy and move in the direction of personalized medicine.
Collapse
Affiliation(s)
- Skander Mulder
- University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Paul Perco
- Medical University of Innsbruck, Innsbruck, Austria
| | | | - Uzma F Mehdi
- University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | | | - Robert Toto
- University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Hiddo J L Heerspink
- University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Michelle J Pena
- University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
15
|
Ge M, Fontanesi F, Merscher S, Fornoni A. The Vicious Cycle of Renal Lipotoxicity and Mitochondrial Dysfunction. Front Physiol 2020; 11:732. [PMID: 32733268 PMCID: PMC7358947 DOI: 10.3389/fphys.2020.00732] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 06/08/2020] [Indexed: 12/15/2022] Open
Abstract
The kidney is one of the most energy-demanding organs that require abundant and healthy mitochondria to maintain proper function. Increasing evidence suggests a strong association between mitochondrial dysfunction and chronic kidney diseases (CKDs). Lipids are not only important sources of energy but also essential components of mitochondrial membrane structures. Dysregulation of mitochondrial oxidative metabolism and increased reactive oxygen species (ROS) production lead to compromised mitochondrial lipid utilization, resulting in lipid accumulation and renal lipotoxicity. However, lipotoxicity can be either the cause or the consequence of mitochondrial dysfunction. Imbalanced lipid metabolism, in turn, can hamper mitochondrial dynamics, contributing to the alteration of mitochondrial lipids and reduction in mitochondrial function. In this review, we summarize the interplay between renal lipotoxicity and mitochondrial dysfunction, with a focus on glomerular diseases.
Collapse
Affiliation(s)
- Mengyuan Ge
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States.,Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Flavia Fontanesi
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Sandra Merscher
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States.,Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States.,Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
16
|
PPAR γ and Its Agonists in Chronic Kidney Disease. Int J Nephrol 2020; 2020:2917474. [PMID: 32158560 PMCID: PMC7060840 DOI: 10.1155/2020/2917474] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/11/2020] [Accepted: 02/03/2020] [Indexed: 02/06/2023] Open
Abstract
Chronic kidney disease (CKD) has become a global healthcare issue. CKD can progress to irreversible end-stage renal diseases (ESRD) or renal failure. The major risk factors for CKD include obesity, diabetes, and cardiovascular diseases. Understanding the key process involved in the disease development may lead to novel interventive strategies, which is currently lagging behind. Peroxisome proliferator-activated receptor γ (PPARγ) is one of the ligand-activated transcription factor superfamily members and is globally expressed in human tissues. Its agonists such as thiazolidinediones (TZDs) have been applied as effective antidiabetic drugs as they control insulin sensitivity in multiple metabolic tissues. Besides, TZDs exert protective effects in multiple other CKD risk disease contexts. As PPARγ is abundantly expressed in major kidney cells, its physiological roles in those cells have been studied in both cell and animal models. The function of PPARγ in the kidney ranges from energy metabolism, cell proliferation to inflammatory suppression, although major renal side effects of existing agonists (including TZDs) have been reported, which limited their application in treating CKD. In the current review, we systemically assess the function of PPARγ in CKDs and the benefits and current limitations of its agonists in the clinical applications.
Collapse
|
17
|
Vallée A, Lecarpentier Y. TGF-β in fibrosis by acting as a conductor for contractile properties of myofibroblasts. Cell Biosci 2019; 9:98. [PMID: 31827764 PMCID: PMC6902440 DOI: 10.1186/s13578-019-0362-3] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 11/30/2019] [Indexed: 12/21/2022] Open
Abstract
Myofibroblasts are non-muscle contractile cells that play a key physiologically role in organs such as the stem villi of the human placenta during physiological pregnancy. They are able to contract and relax in response to changes in the volume of the intervillous chamber. Myofibroblasts have also been observed in several diseases and are involved in wound healing and the fibrotic processes affecting several organs, such as the liver, lungs, kidneys and heart. During the fibrotic process, tissue retraction rather than contraction is correlated with collagen synthesis in the extracellular matrix, leading to irreversible fibrosis and, finally, apoptosis of myofibroblasts. The molecular motor of myofibroblasts is the non-muscle type IIA and B myosin (NMMIIA and NMMIIB). Fibroblast differentiation into myofibroblasts is largely governed by the transforming growth factor-β1 (TGF-β1). This system controls the canonical WNT/β-catenin pathway in a positive manner, and PPARγ in a negative manner. The WNT/β-catenin pathway promotes fibrosis, while PPARγ prevents it. This review focuses on the contractile properties of myofibroblasts and the conductor, TGF-β1, which together control the opposing interplay between PPARγ and the canonical WNT/β-catenin pathway.
Collapse
Affiliation(s)
- Alexandre Vallée
- 1Délégation à la Recherche Clinique (DRCI), Hôpital Foch, Suresnes, France.,DACTIM-MIS, Laboratoire de Mathématiques et Applications (LMA), CNRS, UMR 7348, Université de Poitiers, CHU de Poitiers, Poitiers, France
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien (GHEF), Meaux, France
| |
Collapse
|
18
|
Abstract
Fibrosis is the abnormal deposition of extracellular matrix, which can lead to organ dysfunction, morbidity, and death. The disease burden caused by fibrosis is substantial, and there are currently no therapies that can prevent or reverse fibrosis. Metabolic alterations are increasingly recognized as an important pathogenic process that underlies fibrosis across many organ types. As a result, metabolically targeted therapies could become important strategies for fibrosis reduction. Indeed, some of the pathways targeted by antifibrotic drugs in development - such as the activation of transforming growth factor-β and the deposition of extracellular matrix - have metabolic implications. This Review summarizes the evidence to date and describes novel opportunities for the discovery and development of drugs for metabolic reprogramming, their associated challenges, and their utility in reducing fibrosis. Fibrotic therapies are potentially relevant to numerous common diseases such as cirrhosis, non-alcoholic steatohepatitis, chronic renal disease, heart failure, diabetes, idiopathic pulmonary fibrosis, and scleroderma.
Collapse
|
19
|
Hoerst K, van den Broek L, Sachse C, Klein O, von Fritschen U, Gibbs S, Hedtrich S. Regenerative potential of adipocytes in hypertrophic scars is mediated by myofibroblast reprogramming. J Mol Med (Berl) 2019; 97:761-775. [DOI: 10.1007/s00109-019-01772-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 03/06/2019] [Accepted: 03/08/2019] [Indexed: 12/19/2022]
|
20
|
Lecarpentier Y, Gourrier E, Gobert V, Vallée A. Bronchopulmonary Dysplasia: Crosstalk Between PPARγ, WNT/β-Catenin and TGF-β Pathways; The Potential Therapeutic Role of PPARγ Agonists. Front Pediatr 2019; 7:176. [PMID: 31131268 PMCID: PMC6509750 DOI: 10.3389/fped.2019.00176] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/16/2019] [Indexed: 12/21/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a serious pulmonary disease which occurs in preterm infants. Mortality remains high due to a lack of effective treatment, despite significant progress in neonatal resuscitation. In BPD, a persistently high level of canonical WNT/β-catenin pathway activity at the canalicular stage disturbs the pulmonary maturation at the saccular and alveolar stages. The excessive thickness of the alveolar wall impairs the normal diffusion of oxygen and carbon dioxide, leading to hypoxia. Transforming growth factor (TGF-β) up-regulates canonical WNT signaling and inhibits the peroxysome proliferator activated receptor gamma (PPARγ). This profile is observed in BPD, especially in animal models. Following a premature birth, hypoxia activates the canonical WNT/TGF-β axis at the expense of PPARγ. This gives rise to the differentiation of fibroblasts into myofibroblasts, which can lead to pulmonary fibrosis that impairs the respiratory function after birth, during childhood and even adulthood. Potential therapeutic treatment could target the inhibition of the canonical WNT/TGF-β pathway and the stimulation of PPARγ activity, in particular by the administration of nebulized PPARγ agonists.
Collapse
Affiliation(s)
- Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien, Meaux, France
| | - Elizabeth Gourrier
- Service de néonatologie, Grand Hôpital de l'Est Francilien, Meaux, France
| | - Vincent Gobert
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien, Meaux, France
| | - Alexandre Vallée
- Diagnosis and Therapeutic Center, Hypertension and Cardiovascular Prevention Unit, Hôtel-Dieu Hospital, AP-HP Paris, Paris-Descartes University, Paris, France
| |
Collapse
|
21
|
Platt C, Coward RJ. Peroxisome proliferator activating receptor-γ and the podocyte. Nephrol Dial Transplant 2017; 32:423-433. [PMID: 27697843 DOI: 10.1093/ndt/gfw320] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 07/24/2016] [Indexed: 12/13/2022] Open
Abstract
Over the past two decades it has become clear that the glomerular podocyte is a key cell in preventing albuminuria, kidney failure and cardiovascular morbidity. Understanding the key pathways that protect the podocyte in times of glomerular stress, which can also be therapeutically manipulated, are highly attractive. In the following review we assess the evidence that the peroxisome proliferator activating receptor (PPAR) agonists are beneficial for podocyte and kidney function with a focus on PPAR-γ. We explain our current understanding of the mechanisms of action of these agonists and the evidence they are beneficial in diabetic and non-diabetic kidney disease. We also outline why these drugs have not been widely used for kidney disease in the past but they may be in the future.
Collapse
Affiliation(s)
| | - Richard J Coward
- Department of Paediatric Nephrology, Bristol Royal Hospital for Children, Bristol, United Kingdom
| |
Collapse
|
22
|
Lecarpentier Y, Schussler O, Claes V, Vallée A. The Myofibroblast: TGFβ-1, A Conductor which Plays a Key Role in Fibrosis by Regulating the Balance between PPARγ and the Canonical WNT Pathway. NUCLEAR RECEPTOR RESEARCH 2017. [DOI: 10.11131/2017/101299] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l’Est Francilien (GHEP), Meaux, France
| | - Olivier Schussler
- Department of Cardiovascular Surgery, Cardiovascular Research Laboratory, HUG/CMU, Geneva, Switzerland
| | - Victor Claes
- Department of Pharmaceutical Sciences, University of Antwerp, Wilrijk, Belgium
| | - Alexandre Vallée
- Experimental and Clinical Neurosciences Laboratory, INSERM U1084, University of Poitiers, Poitiers, France
| |
Collapse
|
23
|
Vallée A, Lecarpentier Y, Guillevin R, Vallée JN. Interactions between TGF-β1, canonical WNT/β-catenin pathway and PPAR γ in radiation-induced fibrosis. Oncotarget 2017; 8:90579-90604. [PMID: 29163854 PMCID: PMC5685775 DOI: 10.18632/oncotarget.21234] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 08/17/2017] [Indexed: 12/16/2022] Open
Abstract
Radiation therapy induces DNA damage and inflammation leading to fibrosis. Fibrosis can occur 4 to 12 months after radiation therapy. This process worsens with time and years. Radiation-induced fibrosis is characterized by fibroblasts proliferation, myofibroblast differentiation, and synthesis of collagen, proteoglycans and extracellular matrix. Myofibroblasts are non-muscle cells that can contract and relax. Myofibroblasts evolve towards irreversible retraction during fibrosis process. In this review, we discussed the interplays between transforming growth factor-β1 (TGF-β1), canonical WNT/β-catenin pathway and peroxisome proliferator-activated receptor gamma (PPAR γ) in regulating the molecular mechanisms underlying the radiation-induced fibrosis, and the potential role of PPAR γ agonists. Overexpression of TGF-β and canonical WNT/β-catenin pathway stimulate fibroblasts accumulation and myofibroblast differentiation whereas PPAR γ expression decreases due to the opposite interplay of canonical WNT/β-catenin pathway. Both TGF-β1 and canonical WNT/β-catenin pathway stimulate each other through the Smad pathway and non-Smad pathways such as phosphatidylinositol 3-kinase/serine/threonine kinase (PI3K/Akt) signaling. WNT/β-catenin pathway and PPAR γ interact in an opposite manner. PPAR γ agonists decrease β-catenin levels through activation of inhibitors of the WNT pathway such as Smad7, glycogen synthase kinase-3 (GSK-3 β) and dickkopf-related protein 1 (DKK1). PPAR γ agonists also stimulate phosphatase and tensin homolog (PTEN) expression, which decreases both TGF-β1 and PI3K/Akt pathways. PPAR γ agonists by activating Smad7 decrease Smads pathway and then TGF-β signaling leading to decrease radiation-induced fibrosis. TGF-β1 and canonical WNT/β-catenin pathway promote radiation-induced fibrosis whereas PPAR γ agonists can prevent radiation-induced fibrosis.
Collapse
Affiliation(s)
- Alexandre Vallée
- Experimental and Clinical Neurosciences Laboratory, INSERM U1084, University of Poitiers, Poitiers, France.,Laboratory of Mathematics and Applications (LMA), UMR CNRS 7348, University of Poitiers, Poitiers, France
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien (GHEF), Meaux, France
| | - Rémy Guillevin
- DACTIM, UMR CNRS 7348, University of Poitiers et CHU de Poitiers, Poitiers, France
| | - Jean-Noël Vallée
- Laboratory of Mathematics and Applications (LMA), UMR CNRS 7348, University of Poitiers, Poitiers, France.,CHU Amiens Picardie, University of Picardie Jules Verne (UPJV), Amiens, France
| |
Collapse
|
24
|
Marangoni RG, Masui Y, Fang F, Korman B, Lord G, Lee J, Lakota K, Wei J, Scherer PE, Otvos L, Yamauchi T, Kubota N, Kadowaki T, Asano Y, Sato S, Tourtellotte WG, Varga J. Adiponectin is an endogenous anti-fibrotic mediator and therapeutic target. Sci Rep 2017; 7:4397. [PMID: 28667272 PMCID: PMC5493638 DOI: 10.1038/s41598-017-04162-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 05/22/2017] [Indexed: 12/21/2022] Open
Abstract
Skin fibrosis in systemic sclerosis (SSc) is accompanied by attrition of dermal white adipose tissue (dWAT) and reduced levels of circulating adiponectin. Since adiponectin has potent regulatory effects on fibroblasts, we sought to assess adiponectin signaling in SSc skin biopsies, and evaluate fibrosis in mice with adiponectin gain- and loss-of-function mutations. Furthermore, we investigated the effects and mechanism of action of agonist peptides targeting adiponectin receptors in vitro and in vivo. We found that adiponectin pathway activity was significantly reduced in a subset of SSc skin biopsies. Mice lacking adiponectin mounted an exaggerated dermal fibrotic response, while transgenic mice with constitutively elevated adiponectin showed selective dWAT expansion and protection from skin and peritoneal fibrosis. Adiponectin receptor agonists abrogated ex vivo fibrotic responses in explanted normal and SSc fibroblasts and in 3D human skin equivalents, in part by attenuating focal adhesion complex assembly, and prevented and reversed experimentally-induced organ fibrosis in mice. These results implicate aberrant adiponectin pathway activity in skin fibrosis, identifying a novel function for this pleiotropic adipokine in regulation of tissue remodeling. Restoring adiponectin signaling in SSc patients therefore might represent an innovative pharmacological strategy for intractable organ fibrosis.
Collapse
Affiliation(s)
- Roberta G Marangoni
- Northwestern Scleroderma Program, Feinberg School of Medicine, Chicago, IL, 60611, USA.
| | - Yuri Masui
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Feng Fang
- Northwestern Scleroderma Program, Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Benjamin Korman
- Northwestern Scleroderma Program, Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Gabriel Lord
- Northwestern Scleroderma Program, Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Junghwa Lee
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Katja Lakota
- Department of Rheumatology, University Medical Centre Ljubljana, 1000, Ljubljana, Slovenia
| | - Jun Wei
- Northwestern Scleroderma Program, Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Laszlo Otvos
- Department of Biology, Temple University, Philadelphia, PA, 19140, USA
| | - Toshimasa Yamauchi
- Department of Diabetes and Metabolic Diseases, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Naoto Kubota
- Department of Diabetes and Metabolic Diseases, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Takashi Kadowaki
- Department of Diabetes and Metabolic Diseases, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yoshihide Asano
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Shinichi Sato
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Warren G Tourtellotte
- Department of Pathology and Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - John Varga
- Northwestern Scleroderma Program, Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
25
|
Li J, Wang W, He Y, Li Y, Yan EZ, Zhang K, Irvine DJ, Hammond PT. Structurally Programmed Assembly of Translation Initiation Nanoplex for Superior mRNA Delivery. ACS NANO 2017; 11:2531-2544. [PMID: 28157292 PMCID: PMC5629916 DOI: 10.1021/acsnano.6b08447] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Messenger RNA (mRNA) represents a promising class of nucleic-acid-based therapeutics. While numerous nanocarriers have been developed for mRNA delivery, the inherent labile nature of mRNA results in a very low transfection efficiency and poor expression of desired protein. Here we preassemble the mRNA translation initiation structure through an inherent molecular recognition between 7-methylguanosine (m7G)-capped mRNA and eukaryotic initiation factor 4E (eIF4E) protein to form ribonucleoproteins (RNPs), thereby mimicking the first step of protein synthesis inside cells. Subsequent electrostatic stabilization of RNPs with structurally tunable cationic carriers leads to nanosized complexes (nanoplexes), which elicit high levels of mRNA transfection in different cell types by enhancing intracellular mRNA stability and protein synthesis. By investigating a family of synthetic polypeptides bearing different side group arrangements of cationic charge, we find that the molecular structure modulates the nanoscale distance between the mRNA strand and the eIF4E protein inside the nanoplex, which directly impacts the enhancement of mRNA transfection. To demonstrate the biomedical potential of this approach, we use this approach to introduce mRNA/eIF4E nanoplexes to murine dendritic cells, resulting in increased activation of cytotoxic CD8 T cells ex vivo. More importantly, eIF4E enhances gene expression in lungs following a systemic delivery of luciferase mRNA/eIF4E in mice. Collectively, this bioinspired molecular assembly method could lead to a new paradigm of gene delivery.
Collapse
Affiliation(s)
- Jiahe Li
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Wade Wang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Yanpu He
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Yingzhong Li
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Emily Z. Yan
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Ketian Zhang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Darrell J. Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
- Howard Hughes Medical Institute, Chevy Chase, MD 20815
| | - Paula T. Hammond
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
- Correspondence: David H. Koch Professor in Engineering, Bayer Chair Professor of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, United States.
| |
Collapse
|
26
|
Wu L, Wang Q, Guo F, Ma X, Ji H, Liu F, Zhao Y, Qin G. MicroRNA-27a Induces Mesangial Cell Injury by Targeting of PPARγ, and its In Vivo Knockdown Prevents Progression of Diabetic Nephropathy. Sci Rep 2016; 6:26072. [PMID: 27184517 PMCID: PMC4869109 DOI: 10.1038/srep26072] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 04/26/2016] [Indexed: 11/09/2022] Open
Abstract
MicroRNAs play important roles in the pathogenesis of diabetic nephropathy (DN). In this study, we found that high glucose upregulated miR-27a expression in cultured glomerular mesangial cells and in the kidney glomeruli of streptozotocin (STZ)-induced diabetic rats. miR-27a knockdown prevented high glucose-induced mesangial cell proliferation and also blocked the upregulation of extracellular matrix (ECM)-associated profibrotic genes. Reduction of cell proliferation and profibrotic gene expression by a miR-27a inhibitor depended upon the expression of peroxisome proliferator-activated receptor γ (PPARγ). Further studies showed that miR-27a negatively regulated PPARγ expression by binding to the 3'-untranslated region of rat PPARγ. An antisense oligonucleotide specific to miR-27a (antagomir-27a) significantly reduced renal miR-27a expression in STZ-induced diabetic rats and significantly increased PPARγ levels. Antagomir-27a also reduced kidney ECM accumulation and proteinuria in STZ-induced diabetic rats. These findings suggest that specific reduction of renal miR-27a decreases renal fibrosis, which may be explained in part by its regulation of PPARγ, and that targeting miR-27a may represent a novel therapeutic approach for DN.
Collapse
Affiliation(s)
- Lina Wu
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.,Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Qingzhu Wang
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Feng Guo
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xiaojun Ma
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Hongfei Ji
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.,Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Fei Liu
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yanyan Zhao
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Guijun Qin
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| |
Collapse
|
27
|
Ramot Y, Mastrofrancesco A, Camera E, Desreumaux P, Paus R, Picardo M. The role of PPARγ-mediated signalling in skin biology and pathology: new targets and opportunities for clinical dermatology. Exp Dermatol 2016; 24:245-51. [PMID: 25644500 DOI: 10.1111/exd.12647] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2015] [Indexed: 12/19/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors that modulate the expression of multiple different genes involved in the regulation of lipid, glucose and amino acid metabolism. PPARs and cognate ligands also regulate important cellular functions, including cell proliferation and differentiation, as well as inflammatory responses. This includes a role in mediating skin and pilosebaceous unit homoeostasis: PPARs appear to be essential for maintaining skin barrier permeability, inhibit keratinocyte cell growth, promote keratinocyte terminal differentiation and regulate skin inflammation. They also may have protective effects on human hair follicle (HFs) epithelial stem cells, while defects in PPARγ-mediated signalling may promote the death of these stem cells and thus facilitate the development of cicatricial alopecia (lichen planopilaris). Overall, however, selected PPARγ modulators appear to act as hair growth inhibitors that reduce the proliferation and promote apoptosis of hair matrix keratinocytes. The fact that commonly prescribed PPARγ-modulatory drugs of the thiazolidine-2,4-dione class can exhibit a battery of adverse cutaneous effects underscores the importance of distinguishing beneficial from clinically undesired cutaneous activities of PPARγ ligands and to better understand on the molecular level how PPARγ-regulated cutaneous lipid metabolism and PPARγ-mediated signalling impact on human skin physiology and pathology. Surely, the therapeutic potential that endogenous and exogenous PPARγ modulators may possess in selected skin diseases, ranging from chronic inflammatory hyperproliferative dermatoses like psoriasis and atopic dermatitis, via scarring alopecia and acne can only be harnessed if the complexities of PPARγ signalling in human skin and its appendages are systematically dissected.
Collapse
Affiliation(s)
- Yuval Ramot
- Department of Dermatology, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | | | | | | | | | | |
Collapse
|
28
|
Abstract
In the last few decades, rapid changes in lifestyle have led to an alarming increase in the prevalence of obesity and obesity-associated complications. Obese patients are at increased risk of developing hypertension, heart disease, insulin resistance, dyslipidemia, type 2 diabetes and kidney disease. The surplus of calories is normally stored as triglycerides in adipose tissue. However, excess lipids can also accumulate ectopically in other organs, including the kidney, contributing to their damage through toxic processes named lipotoxicity. The kidney is negatively affected by dyslipidemia, lipid accumulation and changes in circulating adipokines that bring about alterations in renal lipid metabolism and promote insulin resistance, generation of reactive oxygen species and endoplasmic reticulum stress, ultimately leading to alterations in the glomerular filtration barrier and renal failure. This review focuses on the pathogenic molecular mechanisms associated with renal lipotoxicity, and presents new insights about potential new therapeutic targets and biomarkers such as microRNAs and long non-coding RNAs, of relevance for the early detection of lipid-associated kidney disease.
Collapse
|
29
|
Mohey V, Singh M, Puri N, Kaur T, Pathak D, Singh AP. Sildenafil obviates ischemia-reperfusion injury-induced acute kidney injury through peroxisome proliferator-activated receptor γ agonism in rats. J Surg Res 2015; 201:69-75. [PMID: 26850186 DOI: 10.1016/j.jss.2015.09.035] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 09/22/2015] [Accepted: 09/25/2015] [Indexed: 11/25/2022]
Abstract
BACKGROUND Sildenafil is a phosphodiesterase inhibitor used clinically for treating erectile dysfunction. Few studies suggest sildenafil to be a renoprotective agent. The present study investigated the involvement of peroxisome proliferator-activated receptor γ (PPAR-γ) in sildenafil-mediated protection against ischemia-reperfusion-induced acute kidney injury (AKI) in rats. MATERIALS AND METHODS The rats were subjected to ischemia-reperfusion injury (IRI) with 40 min of bilateral renal ischemia followed by reperfusion for 24 h. The renal damage was assessed by measuring creatinine clearance, blood urea nitrogen, plasma uric acid, electrolytes, and microproteinuria in rats. The thiobarbituric acid reactive substances, superoxide anion generation, and reduced glutathione levels were measured to assess oxidative stress in renal tissues. The hematoxylin-eosin staining was carried out to demonstrate histopathologic changes in renal tissues. Sildenafil (0.5 and 1.0 mg/kg, intraperitoneal) was administered 1 h before subjecting the rats to renal IRI. In a separate group, bisphenol A diglycidyl ether (30 mg/kg, intraperitoneal), a PPAR-γ receptor antagonist, was given before sildenafil administration followed by IRI. RESULTS The ischemia-reperfusion demonstrated marked AKI with significant changes in serum and urinary parameters, enhanced oxidative stress, and histopathologic changes in renal tissues. The administration of sildenafil demonstrated significant protection against ischemia-reperfusion-induced AKI. The prior treatment with bisphenol A diglycidyl ether abolished sildenafil-mediated renal protection, thereby confirming involvement of PPAR-γ agonism in the sildenafil-mediated renoprotective effect. CONCLUSIONS It is concluded that sildenafil protects against ischemia-reperfusion-induced AKI through PPAR-γ agonism in rats.
Collapse
Affiliation(s)
- Vinita Mohey
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| | - Manjinder Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| | - Nikkita Puri
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| | - Tajpreet Kaur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India; Department of Pharmacology, Khalsa College of Pharmacy, Amritsar, India
| | - Devendra Pathak
- Department of Veterinary Anatomy, Guru Angad Dev Veterinary and Animal Science University, Ludhiana, India
| | - Amrit Pal Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India.
| |
Collapse
|
30
|
Muñoz-Félix JM, González-Núñez M, Martínez-Salgado C, López-Novoa JM. TGF-β/BMP proteins as therapeutic targets in renal fibrosis. Where have we arrived after 25 years of trials and tribulations? Pharmacol Ther 2015; 156:44-58. [PMID: 26493350 DOI: 10.1016/j.pharmthera.2015.10.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The understanding of renal fibrosis in chronic kidney disease (CKD) remains as a challenge. More than 10% of the population of developed countries suffer from CKD. Proliferation and activation of myofibroblasts and accumulation of extracellular matrix proteins are the main features of kidney fibrosis, a process in which a large number of cytokines are involved. Targeting cytokines responsible for kidney fibrosis development might be an important strategy to face the problem of CKD. The increasing knowledge of the signaling pathway network of the transforming growth factor beta (TGF-β) superfamily members, such as the profibrotic cytokine TGF-β1 or the bone morphogenetic proteins (BMPs), and their involvement in the regulation of kidney fibrosis, has stimulated numerous research teams to look for potential strategies to inhibit profibrotic cytokines or to enhance the anti-fibrotic actions of other cytokines. The consequence of all these studies is a better understanding of all these canonical (Smad-mediated) and non-canonical signaling pathways. In addition, the different receptors involved for signaling of each cytokine, the different combinations of type I-type II receptors, and the presence and function of co-receptors that can influence the biological response have been also described. However, are these studies leading to suitable strategies to block the appearance and progression of kidney fibrosis? In this review, we offer a critical perspective analyzing the achievements using the most important strategies developed up till now: TGF-β antibodies, chemical inhibitors of TGF-β receptors, miRNAs and signaling pathways and BMP agonists with a potential role as therapeutic molecules against kidney fibrosis.
Collapse
Affiliation(s)
- José M Muñoz-Félix
- Unidad de Fisiopatología Renal y Cardiovascular, Instituto Reina Sofía de Investigación Nefrológica, Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - María González-Núñez
- Unidad de Fisiopatología Renal y Cardiovascular, Instituto Reina Sofía de Investigación Nefrológica, Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Carlos Martínez-Salgado
- Unidad de Fisiopatología Renal y Cardiovascular, Instituto Reina Sofía de Investigación Nefrológica, Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain; Instituto de Estudios de Ciencias de la Salud de Castilla y León (IECSCYL), Hospital Universitario de Salamanca, Salamanca, Spain
| | - José M López-Novoa
- Unidad de Fisiopatología Renal y Cardiovascular, Instituto Reina Sofía de Investigación Nefrológica, Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain.
| |
Collapse
|
31
|
Chen YH, Chiang MH, Liu JS, Chang YK, Kuo KL, Hung SC, Tai HL, Hsu CC, Tarng DC. Thiazolidinediones and Risk of Long-Term Dialysis in Diabetic Patients with Advanced Chronic Kidney Disease: A Nationwide Cohort Study. PLoS One 2015; 10:e0129922. [PMID: 26083376 PMCID: PMC4470911 DOI: 10.1371/journal.pone.0129922] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Accepted: 05/14/2015] [Indexed: 11/23/2022] Open
Abstract
Thiazolidinediones (TZDs) reduce urinary albumin excretion and proteinuria in diabetic nephropathy. The effect of TZDs on hard renal outcome in diabetic patients with chronic kidney disease (CKD) is unknown. We investigate the association of TZDs and risk of long-term dialysis or death in diabetic patients with advanced CKD. The nationwide population-based cohort study was conducted using Taiwan’s National Health Insurance Research Database. From January 2000 to June 2009, 12350 diabetic patients with advanced CKD (serum creatinine levels greater than 6 mg/dL but not yet receiving renal replacement therapy) were selected for the study. We used multivariable Cox regression models and a propensity score-based matching technique to estimate hazard ratios (HRs) for development of long-term dialysis and the composite outcome of long-term dialysis or death for TZD users (n=1224) as compared to nonusers (n=11126). During a median follow-up of 6 months, 8270 (67.0%) patients required long-term dialysis and 2593 (21.0%) patients died before starting long-term dialysis. Using propensity score matched analysis, we found TZD users were associated with a lower risk for long-term dialysis (HR, 0.80; 95% confidence interval [CI], 0.74-0.86) and the composite outcome of long-term dialysis or death (HR, 0.85; 95% CI, 0.80-0.91). The results were consistent across most patient subgroups. Use of TZDs among diabetic patients with advanced CKD was associated with lower risk for progression to end-stage renal disease necessitating long-term dialysis or death. Further randomized controlled studies are required to validate this association.
Collapse
Affiliation(s)
- Yu-Hsin Chen
- Division of Nephrology, Department of Internal Medicine, Taipei City Hospital Yang-Ming Branch, Taipei, Taiwan
- Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Ming-Han Chiang
- Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - Jia-Sin Liu
- Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - Yu-Kang Chang
- Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - Ko-Lin Kuo
- Division of Nephrology, Taipei Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, Taipei, Taiwan
| | - Szu-Chun Hung
- Division of Nephrology, Taipei Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, Taipei, Taiwan
| | - Hsin-Ling Tai
- Nursing Department, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chih-Cheng Hsu
- Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan
- Department of Health Services Administration, China Medical University, Taichung, Taiwan
- * E-mail: (CCH); (DCT)
| | - Der-Cherng Tarng
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Department and Institute of Physiology and Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
- * E-mail: (CCH); (DCT)
| |
Collapse
|
32
|
Li Y, Shen Y, Li M, Su D, Xu W, Liang X, Li R. Inhibitory effects of peroxisome proliferator-activated receptor γ agonists on collagen IV production in podocytes. Mol Cell Biochem 2015; 405:233-41. [PMID: 25920446 DOI: 10.1007/s11010-015-2414-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 04/18/2015] [Indexed: 12/14/2022]
Abstract
Peroxisome proliferator-activated receptor-γ (PPAR-γ) agonists have beneficial effects on the kidney diseases through preventing microalbuminuria and glomerulosclerosis. However, the mechanisms underlying these effects remain to be fully understood. In this study, we investigate the effects of PPAR-γ agonist, rosiglitazone (Rosi) and pioglitazone (Pio), on collagen IV production in mouse podocytes. The endogenous expression of PPAR-γ was found in the primary podocytes and can be upregulated by Rosi and Pio, respectively, detected by RT-PCR and Western blot. PPAR-γ agonist markedly blunted the increasing of collagen IV expression and extraction in podocytes induced by TGF-β. In contrast, adding PPAR-γ antagonist, GW9662, to podocytes largely prevented the inhibition of collagen IV expression from Pio treatment. Our data also showed that phosphorylation of Smad2/3 enhanced by TGF-β in a time-dependent manner was significantly attenuated by adding Pio. The promoter region of collagen IV gene contains one putative consensus sequence of Smad-binding element (SBE) by promoter analysis, Rosi and Pio significantly ameliorated TGF-β-induced SBE4-luciferase activity. In conclusion, PPAR-γ activation by its agonist, Rosi or Pio, in vitro directly inhibits collagen IV expression and synthesis in primary mouse podocytes. The suppression of collagen IV production was related to the inhibition of TGF-β-driven phosphorylation of Smad2/3 and decreased response activity of SBEs of collagen IV in PPAR-γ agonist-treated mouse podocytes. This represents a novel mechanistic support regarding PPAR-γ agonists as podocyte protective agents.
Collapse
Affiliation(s)
- Yanjiao Li
- Department of Nephrology, Shanxi Provincial People's Hospital, The Affiliated People's Hospital of Shanxi Medical University, Taiyuan, 030012, Shanxi, China
| | | | | | | | | | | | | |
Collapse
|
33
|
Gounder VK, Arumugam S, Arozal W, Thandavarayan RA, Pitchaimani V, Harima M, Suzuki K, Nomoto M, Watanabe K. Olmesartan protects against oxidative stress possibly through the Nrf2 signaling pathway and inhibits inflammation in daunorubicin-induced nephrotoxicity in rats. Int Immunopharmacol 2014; 18:282-9. [DOI: 10.1016/j.intimp.2013.11.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 11/14/2013] [Accepted: 11/18/2013] [Indexed: 01/10/2023]
|
34
|
Wei J, Zhu H, Komura K, Lord G, Tomcik M, Wang W, Doniparthi S, Tamaki Z, Hinchcliff M, Distler JHW, Varga J. A synthetic PPAR-γ agonist triterpenoid ameliorates experimental fibrosis: PPAR-γ-independent suppression of fibrotic responses. Ann Rheum Dis 2014; 73:446-54. [PMID: 23515440 PMCID: PMC4028127 DOI: 10.1136/annrheumdis-2012-202716] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Persistent fibroblast activation initiated by transforming growth factor β (TGF-β) is a fundamental event in the pathogenesis of systemic sclerosis, and its pharmacological inhibition represents a potential therapeutic strategy. The nuclear receptor, peroxisome proliferator-activated receptor γ (PPAR-γ), exerts potent fibrotic activity. The synthetic oleanane triterpenoid, 2-cyano-3,12-dioxo-olean-1,9-dien-28-oic acid (CDDO), is a PPAR-γ agonist with potential effects on TGF-β signalling and dermal fibrosis. OBJECTIVE To examine the modulation of fibrogenesis by CDDO in explanted fibroblasts, skin organ cultures and murine models of scleroderma. MATERIAL AND METHODS The effects of CDDO on experimental fibrosis induced by bleomycin injection or by overexpression of constitutively active type I TGF-β receptor (TgfbR1ca) were evaluated. Modulation of fibrotic gene expression was examined in human skin organ cultures. To delineate the mechanisms underlying the antifibrotic effects of CDDO, explanted skin fibroblasts cultured in two-dimensional monolayers or in three-dimensional full-thickness human skin equivalents were studied. RESULTS CDDO significantly ameliorated dermal fibrosis in two complementary mouse models of scleroderma, as well as in human skin organ cultures and in three-dimensional human skin equivalents. In two-dimensional monolayer cultures of explanted normal fibroblasts, CDDO abrogated fibrogenic responses induced by TGF-β. These CDDO effects occurred via disruption of Smad-dependent transcription and were associated with inhibition of Akt activation. In scleroderma fibroblasts, CDDO attenuated the elevated synthesis of collagen. Remarkably, the in vitro antifibrotic effects of CDDO were independent of PPAR-γ. CONCLUSIONS The PPAR-γ agonist triterpenoid CDDO attenuates fibrogenesis by antagonistically targeting canonical TGF-β/Smad and Akt signalling in a PPAR-γ-independent manner. These findings identify this synthetic triterpenoid as a potential new therapy for the control of fibrosis.
Collapse
Affiliation(s)
- Jun Wei
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Hongyan Zhu
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Kazuhiro Komura
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Gabriel Lord
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Michal Tomcik
- Institute of Rheumatology, Department of Clinical and Experimental Rheumatology, Charles University, Prague, Czech Republic
| | - Wenxia Wang
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Sruthi Doniparthi
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Zenshiro Tamaki
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Monique Hinchcliff
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Joerg H. W. Distler
- Department of Internal Medicine, University of Erlangen-Nuremberg, Erlangen, Germany
| | - John Varga
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, USA
| |
Collapse
|
35
|
Ghosh AK, Quaggin SE, Vaughan DE. Molecular basis of organ fibrosis: potential therapeutic approaches. Exp Biol Med (Maywood) 2013; 238:461-81. [PMID: 23856899 DOI: 10.1177/1535370213489441] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Fibrosis, a non-physiological wound healing in multiple organs, is associated with end-stage pathological symptoms of a wide variety of vascular injury and inflammation related diseases. In response to chemical, immunological and physical insults, the body's defense system and matrix synthetic machinery respond to healing the wound and maintain tissue homeostasis. However, uncontrolled wound healing leads to scarring or fibrosis, a pathological condition characterized by excessive synthesis and accumulation of extracellular matrix proteins, loss of tissue homeostasis and organ failure. Understanding the actual cause of pathological wound healing and identification of igniter(s) of fibrogenesis would be helpful to design novel therapeutic approaches to control pathological wound healing and to prevent fibrosis related morbidity and mortality. In this article, we review the significance of a few key cytokines (TGF-β, IFN-γ, IL-10) transcriptional activators (Sp1, Egr-1, Smad3), repressors (Smad7, Fli-1, PPAR-γ, p53, Klotho) and epigenetic modulators (acetyltransferase, methyltransferases, deacetylases, microRNAs) involved in major matrix protein collagen synthesis under pathological stage of wound healing, and the potentiality of these regulators as therapeutic targets for fibrosis treatment. The significance of endothelial to mesenchymal transition (EndMT) and senescence, two newly emerged fields in fibrosis research, has also been discussed.
Collapse
Affiliation(s)
- Asish K Ghosh
- Feinberg Cardiovascular Research Institute & Division of Nephrology, Northwestern University, Chicago, IL, USA.
| | | | | |
Collapse
|
36
|
Liang YJ, Jian JH, Chen CY, Hsu CY, Shih CY, Leu JG. L-165,041, troglitazone and their combination treatment to attenuate high glucose-induced receptor for advanced glycation end products (RAGE) expression. Eur J Pharmacol 2013; 715:33-8. [PMID: 23831394 DOI: 10.1016/j.ejphar.2013.06.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 06/06/2013] [Accepted: 06/21/2013] [Indexed: 01/14/2023]
Abstract
Diabetic nephropathy is the leading cause of end-stage renal disease in the most developed countries of the world. Hyperglycemia-induced advanced glycation end products (AGEs) and receptor for AGEs (RAGE) production, pro-inflammatory cytokine secretion, and oxidative stress activation play major roles in kidney cell injury and apoptosis. Peroxisome proliferator-activated receptor-gamma (PPARγ) agonists are used clinically as insulin sensitizers. This study evaluated the renoprotective effect of PPARγ (troglitazone) and PPARδ (L-165,041) agonists on human embryonic kidney 293 (HEK) and mesangial cells. Troglitazone (10 μM) and L-165,041 (1 μM) significantly inhibited high glucose (25mM)-induced interleukin-6 and TNF-α production, RAGE expression and NF-κB translocation in HEK cells. Furthermore, Troglitazone (10 μM) and L-165,041(1 μM) significantly increased SOD expression and attenuated apoptosis in HEK and mesangial cells. The inhibitory effect between 1 μM L-165,041 and 10 μM troglitazone showed no difference. Furthermore L-165,041 and troglitazone together did not increase the effects. These results provide important information for future application of PPAR agonists in diabetic nephropathy treatment.
Collapse
Affiliation(s)
- Yao-Jen Liang
- Department and Institute of Life Science, Fu-Jen Catholic University, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
37
|
Tavafi M. Complexity of diabetic nephropathy pathogenesis and design of investigations. J Renal Inj Prev 2013; 2:59-62. [PMID: 25340129 PMCID: PMC4206008 DOI: 10.12861/jrip.2013.20] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Accepted: 03/12/2013] [Indexed: 01/07/2023] Open
Abstract
Diabetic nephropathy (DN) pathogenesis is very complex and multifactorial. There are several mechanisms or pathways that hyperglycemia leads to renal injuries. Each pathway makes renal injuries via several mediators. Some mediators are common between the pathways such as reactive oxygen species (ROS) and TGF-β and there are many overlaps and interference between the pathways. This review summarized complexity of DN pathogenesis and overlaps or interfering of mediators between the pathogenesis pathways. Besides, in the review suggested new designs of researches based on this complexity pathogenesis. The pathogenesis of DN is certainly very complex and multifactorial. From the overview of molecular mechanisms of DN pathogenesis, there are many pathways and many mediators with many interferences and overlaps between them. The focal point of this pathogenesis still unknown but it seems that RAAS system, oxidative stress and TGF-β relatively are common between these complex tangle webs of pathogenesis.
Collapse
Affiliation(s)
- Majid Tavafi
- Department of Anatomy, Faculty of Medicine, Lorestan University of Medical sciences, Khoram Abad, Iran
| |
Collapse
|
38
|
Arora MK, Singh UK. Molecular mechanisms in the pathogenesis of diabetic nephropathy: an update. Vascul Pharmacol 2013; 58:259-71. [PMID: 23313806 DOI: 10.1016/j.vph.2013.01.001] [Citation(s) in RCA: 152] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 01/04/2013] [Accepted: 01/04/2013] [Indexed: 12/13/2022]
Abstract
Diabetes mellitus is known to trigger retinopathy, neuropathy and nephropathy. Diabetic nephropathy, a long-term major microvascular complication of uncontrolled hyperglycemia, affects a large population worldwide. Recent findings suggest that numerous pathways are activated during the course of diabetes mellitus and that these pathways individually or collectively play a role in the induction and progression of diabetic nephropathy. However, clinical strategies targeting these pathways to manage diabetic nephropathy remain unsatisfactory, as the number of diabetic patients with nephropathy is increasing yearly. To develop ground-breaking therapeutic options to prevent the development and progression of diabetic nephropathy, a comprehensive understanding of the molecular mechanisms involved in the pathogenesis of the disease is mandatory. Therefore, the purpose of this paper is to discuss the underlying mechanisms and downstream pathways involved in the pathogenesis of diabetic nephropathy.
Collapse
Affiliation(s)
- Mandeep Kumar Arora
- Faculty of Pharmacy, Swami Vivekanand Subharti University, Meerut 250005, Uttar Pradesh, India.
| | | |
Collapse
|
39
|
Abstract
PURPOSE Diabetes is the leading cause of end-stage renal failure. The present study was undertaken to characterize the effects of Corni Fructus on diabetic nephropathy in streptozotocin-induced diabetic rats and their mechanisms. MATERIALS AND METHODS Streptozotocin-diabetic rats were orally administrated with Corni Fructus at a dose of 100, 200 or 400 mg/kg body mass for 40 days. RESULTS Corni Fructus-treated diabetic rats showed significant decreases of blood glucose, urinary protein levels and water consumption. Corni Fructus also reduced serum total cholesterol, total triglyceride and low-density lipoprotein cholesterol levels, and showed a tendency of enhancing high-density lipoprotein cholesterol level. Levels of serum albumin and creatinine in diabetic rats were also significantly reduced by Corni Fructus administration at a dose of 200 and 400 mg/kg body mass compared with non-treated diabetic rats. Corni Fructus increased catalase (CAT), superoxide dismutase (SOD) and glutathione peroxidose (GSH-px) activities in the kidneys of diabetic rats. Furthermore, Corni Fructus treatment enhanced renal peroxisome proliferator-activated receptor-γ (PPARγ) expression in diabetic rats. CONCLUSION These results demonstrated that Corni Fructus may have the potential to protect the animals from diabetic nephropathy by amelioration of oxidative stress and stimulation of PPARγ expression.
Collapse
Affiliation(s)
- Dawei Gao
- Department of Biological Engineering, College of Environmental and Chemical Engineering, Yanshan University, No.438 Hebei Street, Qinhuangdao 066004, China.
| | | | | | | |
Collapse
|
40
|
Wei J, Bhattacharyya S, Jain M, Varga J. Regulation of Matrix Remodeling by Peroxisome Proliferator-Activated Receptor-γ: A Novel Link Between Metabolism and Fibrogenesis. Open Rheumatol J 2012; 6:103-15. [PMID: 22802908 PMCID: PMC3396343 DOI: 10.2174/1874312901206010103] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2012] [Revised: 03/27/2012] [Accepted: 04/04/2012] [Indexed: 02/07/2023] Open
Abstract
The intractable process of fibrosis underlies the pathogenesis of systemic sclerosis (SSc) and other diseases, and in aggregate contributes to 45% of deaths worldwide. Because currently there is no effective anti-fibrotic therapy, a better understanding of the pathways and cellular differentiation programs underlying fibrosis are needed. Emerging evidence points to a fundamental role of the nuclear hormone receptor peroxisome proliferator activated receptor-γ (PPAR-γ) in modulating fibrogenesis. While PPAR-γ has long been known to be important in lipid metabolism and in glucose homeostasis, its role in regulating mesenchymal cell biology and its association with pathological fibrosis had not been appreciated until recently. This article highlights recent studies revealing a consistent association of fibrosis with aberrant PPAR-γ expression and activity in various forms of human fibrosis and in rodent models, and reviews studies linking genetic manipulation of the PPAR-γ pathway in rodents and fibrosis. We survey the broad range of anti-fibrotic activities associated with PPAR-γ and the underlying mechanisms. We also summarize the emerging data linking PPAR-γ dysfunction and pulmonary arterial hypertension (PAH), which together with fibrosis is responsible for the mortality in patients in SSc. Finally, we consider current and potential future strategies for targeting PPAR-γ activity or expression as a therapy for controlling fibrosis.
Collapse
Affiliation(s)
- Jun Wei
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Swati Bhattacharyya
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Manu Jain
- Respiratory and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - John Varga
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, USA
| |
Collapse
|
41
|
Tovar-Palacio C, Torres N, Diaz-Villaseñor A, Tovar AR. The role of nuclear receptors in the kidney in obesity and metabolic syndrome. GENES AND NUTRITION 2012; 7:483-98. [PMID: 22532116 DOI: 10.1007/s12263-012-0295-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 04/02/2012] [Indexed: 02/06/2023]
Abstract
Nuclear receptors are ligand-activated transcriptional regulators of several key aspects of renal physiology and pathophysiology. As such, nuclear receptors control a large variety of metabolic processes, including kidney lipid metabolism, drug clearance, inflammation, fibrosis, cell differentiation, and oxidative stress. Derangement of nuclear receptor regulation, that is, mainly due to obesity may induce metabolic syndrome, may contribute to the pathogenesis and progression of chronic renal disease and may result in end-stage renal disease. This places nuclear receptors at the forefront of novel therapeutic approaches for a broad range of kidney disorders and diseases, including glomerulosclerosis, tubulointerstitial disease, renal lipotoxicity, kidney fibrosis, and hypertension. This review focuses on the importance of the transcription factors peroxisome proliferator-activated receptor alpha, peroxisome proliferator-activated receptor beta, peroxisome proliferator-activated receptor gamma, liver X receptors, farnesoid X receptor, and the pregnane X receptor/steroid and xenobiotic receptor (PXR) on the physiology and pathophysiology of renal diseases associated with obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Claudia Tovar-Palacio
- Department of Nephrology and Mineral Metabolism, National Medical Science and Nutrition Institute, Salvador Zubirán, Vasco de Quiroga No. 15, Tlalpan, 14000, Mexico, D.F., Mexico,
| | | | | | | |
Collapse
|
42
|
Bolignano D, Zoccali C. Glitazones in chronic kidney disease: potential and concerns. Nutr Metab Cardiovasc Dis 2012; 22:167-175. [PMID: 22364889 DOI: 10.1016/j.numecd.2011.11.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Revised: 11/07/2011] [Accepted: 11/13/2011] [Indexed: 11/17/2022]
Abstract
AIMS Glitazones rank now among the most used hypoglycemic agents in patients with type-2 diabetes. This systematic review focuses on the cardiovascular and renal outcomes in chronic kidney disease (CKD) patients treated with these drugs. DATA SYNTHESIS Data from randomized clinical trials and a meta-analysis indicate that glitazones (particularly rosiglitazone) may increase the risk of myocardial infarction, heart failure and cardiovascular death in type-2 diabetics. Observational studies looking at survival and cardiovascular outcomes in diabetic patients with kidney failure show controversial results. Studies in experimental models and clinical studies suggest that glitazones may have favorable effects on renal disease progression, because these drugs coherently reduce urinary albumin excretion and proteinuria in diabetic and non-diabetic nephropathies. No clinical trial based on clinical end-points like kidney failure has until now tested the effect of glitazones on the evolution of chronic renal failure in these patients. CONCLUSIONS Whether the use of glitazones has a positive or a negative impact upon major cardiovascular and renal outcomes in diabetic patients remains an open, unanswered question. Specific studies are needed to assess the efficacy and safety of glitazones in a high risk population like type-2 diabetics with chronic kidney disease.
Collapse
Affiliation(s)
- D Bolignano
- CNR-IBIM, Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension of Reggio Calabria, Reggio Calabria, Italy
| | | |
Collapse
|
43
|
The renoprotective actions of peroxisome proliferator-activated receptors agonists in diabetes. PPAR Res 2012; 2012:456529. [PMID: 22448165 PMCID: PMC3289856 DOI: 10.1155/2012/456529] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Revised: 11/21/2011] [Accepted: 11/21/2011] [Indexed: 12/17/2022] Open
Abstract
Pharmaceutical agonists of peroxisome proliferator-activated receptors (PPARs) are widely used in the management of type 2 diabetes, chiefly as lipid-lowering agents and oral hypoglycaemic agents. Although most of the focus has been placed on their cardiovascular effects, both positive and negative, these agents also have significant renoprotective actions in the diabetic kidney. Over and above action on metabolic control and effects on blood pressure, PPAR agonists also appear to have independent effects on a number of critical pathways that are implicated in the development and progression of diabetic kidney disease, including oxidative stress, inflammation, hypertrophy, and podocyte function. This review will examine these direct and indirect actions of PPAR agonists in the diabetic kidney and explore recent findings of clinical trials of PPAR agonists in patients with diabetes.
Collapse
|
44
|
Saglam F, Cavdar Z, Sarioglu S, Kolatan E, Oktay G, Yilmaz O, Camsari T. Pioglitazone reduces peritoneal fibrosis via inhibition of TGF-β, MMP-2, and MMP-9 in a model of encapsulating peritoneal sclerosis. Ren Fail 2011; 34:95-102. [PMID: 22136281 DOI: 10.3109/0886022x.2011.623498] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE Matrix metalloproteinases (MMPs) and transforming growth factor beta (TGF-β) were increased in peritoneal dialysis patients with encapsulating peritoneal sclerosis (EPS) and in chlorhexidine gluconate (CG)-induced peritoneal sclerosing animal models. Peroxisome proliferator-activated receptors (PPARs) are the major regulators of key metabolic pathways of various inflammatory responses in fibrosing processes in most tissues. The objective of this study was to investigate the effect of pioglitazone (Pio), a synthetic PPAR-γ ligand, on the development of peritoneal fibrosis in CG-induced EPS rats. METHODS Thirty-two Wistar albino rats were intraperitoneally injected with saline (C group n = 8) or with CG (1.5 mL/100 g; CG group, n = 8). Pio (30 mg/kg/day) was administered orally to another group of CG injected rats (the CG + Pio group, n = 8) and to another control group (Pio group, n = 8) from initiation to the end of this study. After 14 days of Pio administration, the rats were killed and the parietal and visceral peritoneum were harvested. TGF-β, MMP-2, MMP-9, tissue inhibitor of metalloproteinases (TIMP)-1, and TIMP-2 activity assays and a morphological examination of the peritoneal tissues were performed. RESULTS Pio significantly inhibited thickening of the submesothelial layer, fibrosis, and inflammation in the peritoneum. It also prevented increases in pro-MMP-2, pro-MMP-9, TIMP-1, and TGF-β activities. CONCLUSION Pio, via MMP and TGF-β inhibition, may lessen accumulation of peritoneal extracellular matrix and fibrosis to some extent in an EPS model and might be a new approach to the amelioration of EPS.
Collapse
Affiliation(s)
- Funda Saglam
- Department of Nephrology, School of Medicine, Dokuz Eylul University, Izmir, Balcova, Turkey.
| | | | | | | | | | | | | |
Collapse
|
45
|
Advanced-glycation-end-product-cholesterol-aggregated-protein accelerates the proliferation of mesangial cells mediated by transforming-growth-factor-beta 1 receptors and the ERK-MAPK pathway. Eur J Pharmacol 2011; 672:159-68. [DOI: 10.1016/j.ejphar.2011.09.185] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Revised: 09/21/2011] [Accepted: 09/24/2011] [Indexed: 01/30/2023]
|
46
|
Bhattacharyya S, Wei J, Varga J. Understanding fibrosis in systemic sclerosis: shifting paradigms, emerging opportunities. Nat Rev Rheumatol 2011; 8:42-54. [PMID: 22025123 PMCID: PMC3954787 DOI: 10.1038/nrrheum.2011.149] [Citation(s) in RCA: 288] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Fibrosis in multiple organs is a prominent pathological finding and distinguishing hallmark of systemic sclerosis (SSc). Findings during the past 5 years have contributed to a more complete understanding of the complex cellular and molecular underpinning of fibrosis in SSc. Fibroblasts, the principal effector cells, are activated in the profibrotic cellular milieu by cytokines and growth factors, developmental pathways, endothelin 1 and thrombin. Innate immune signaling via Toll-like receptors, matrix-generated biomechanical stress signaling via integrins, hypoxia and oxidative stress seem to be implicated in perpetuating the process. Beyond chronic fibroblast activation, fibrosis represents a failure to terminate tissue repair, coupled with an expanded population of mesenchymal cells originating from bone marrow and transdifferentiation of epithelial cells, endothelial cells and pericytes. In addition, studies have identified intrinsic alterations in SSc fibroblasts resulting from epigenetic changes, as well as altered microRNA expression that might underlie the cell-autonomous, persistent activation phenotype of these cells. Precise characterization of the deregulated extracellular and intracellular signaling pathways, mediators and cellular differentiation programs that contribute to fibrosis in SSc will facilitate the development of selective, targeted therapeutic strategies. Effective antifibrotic therapy will ultimately involve novel compounds and repurposing of drugs that are already approved for other indications.
Collapse
Affiliation(s)
- Swati Bhattacharyya
- Division of Rheumatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, McGaw M300, 240 East Huron Street, Chicago, IL 60611, USA
| | | | | |
Collapse
|
47
|
Wang WM, Chen H, Zhong F, Lu Y, Han L, Chen N. Inhibitory effects of rosiglitazone on lipopolysaccharide-induced inflammation in a murine model and HK-2 cells. Am J Nephrol 2011; 34:152-62. [PMID: 21734368 DOI: 10.1159/000329120] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2010] [Accepted: 05/04/2011] [Indexed: 11/19/2022]
Abstract
BACKGROUND Inflammation may play an important role in the pathogenesis of kidney disease. Agonists of the peroxisome proliferator-activated receptor-γ (PPAR-γ), such as rosiglitazone, have been recently demonstrated to regulate inflammation by modulating the production of inflammatory mediators. The purpose of this study was to examine the effects of rosiglitazone on lipopolysaccharide (LPS)-induced kidney inflammation and to explore the mechanism of its renoprotection. METHODS Mice were treated with LPS with or without pretreatment with rosiglitazone. Blood urea nitrogen (BUN), creatinine levels, the urinary albumin-to-creatinine ratio, macrophage infiltration, monocyte chemoattractant protein-1 (MCP-1) expression, PPAR-γ expression, and NF-κB and PPAR-γ activity were investigated. HK-2 cells were maintained under defined in vitro conditions, treated with either rosiglitazone and/or the PPAR-γ antagonist GW9662, and then stimulated with LPS. MCP-1, IL-8, IL-6, NF-κB activity and PPAR-γ expression were investigated. RESULTS Compared to the LPS only group, pretreatment with rosiglitazone in vivo significantly attenuated the BUN levels macrophage infiltration, MCP-1 overexpression and NF-κB activity (p < 0.05). Rosiglitazone also restored PPAR-γ expression and protein activity, which were reduced significantly in the LPS only group (p < 0.05). Furthermore, in the LPS-stimulated HK-2 cells, rosiglitazone downregulated MCP-1, IL-8 and IL-6 expression as well as NF-κB activation and increased PPAR-γ expression (p < 0.05). These effects were diminished by GW9662. CONCLUSION These results showed that pretreatment with rosiglitazone could attenuate kidney inflammation through the activation of PPAR-γ, suppression of MCP-1 overproduction and NF-κB activation. Rosiglitazone had a protective effect via a PPAR-γ-dependent pathway in LPS-treated HK-2 cells.
Collapse
Affiliation(s)
- W M Wang
- Department of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University, PR China
| | | | | | | | | | | |
Collapse
|
48
|
Peroxisome proliferator-activated receptor γ: innate protection from excessive fibrogenesis and potential therapeutic target in systemic sclerosis. Curr Opin Rheumatol 2011; 22:671-6. [PMID: 20693905 DOI: 10.1097/bor.0b013e32833de1a7] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW Progressive organ fibrosis and pulmonary arterial hypertension (PAH) are the leading causes of death in patients with systemic sclerosis (SSc). However, the pathogenesis and the link between these two processes remain obscure. A better understanding of these events is needed in order to facilitate the discovery and development of effective therapies for SSc. RECENT FINDINGS Recent reports provide evidence that the orphan receptor peroxisome proliferator-activated receptor γ (PPARγ), better known for its pivotal role in metabolism, has potent effects on inflammation, fibrogenesis and vascular remodeling and is important in the pathogenesis of fibrosis and PAH, and as a potential therapeutic target in SSc. The studies discussed in this review indicate that ligands of PPARγ potently modulate connective tissue turnover and suggest that aberrant expression or function of PPARγ is associated with, and very likely contributes to, the progression of pathological fibrosis and vascular remodeling. These observations are of particularly relevance because FDA-approved drugs of the thiazolidinedione class currently used for the treatment of obesity-associated type 2 diabetes activate PPARγ signaling. Moreover, novel PPARγ ligands with selective activity are under development or in clinical trials for inflammatory diseases, asthma, Alzheimer disease and cancer. SUMMARY Drugs targeting the PPARγ pathway might be effective for the control of fibrosis as well as pathological vascular remodeling underlying PAH and, therefore, might have a therapeutic potential in SSc. A greater understanding of the mechanisms underlying the antifibrogenic and vascular remodeling activities of PPARγ ligands will be necessary in order to advance these drugs into clinical use.
Collapse
|
49
|
Qi W, Mu J, Luo ZF, Zeng W, Guo YH, Pang Q, Ye ZL, Liu L, Yuan FH, Feng B. Attenuation of diabetic nephropathy in diabetes rats induced by streptozotocin by regulating the endoplasmic reticulum stress inflammatory response. Metabolism 2011; 60:594-603. [PMID: 20817186 DOI: 10.1016/j.metabol.2010.07.021] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 06/15/2010] [Accepted: 07/14/2010] [Indexed: 01/22/2023]
Abstract
The endoplasmic reticulum (ER) is capable of sensing metabolic and stress parameters and integrating intra- and extracellular signals to support a coordinated cell response. In the present study, we verified the hypothesis that 4-phenylbutyric acid (4-PBA), a chemical chaperone, prevented the progression of diabetic nephropathy (DN). Male Sprague-Dawley rats were randomly divided into 3 groups: a normal control group, a DN group, and a DN model plus 4-PBA treatment group (PBA). The DN model was induced by injection of streptozotocin with uninephrectomy. The dosage of 4-PBA treatment was gavaged at a dose of 1 g/kg body weight each day for 12 weeks. The expression of the ER stress indicators significantly increased in the kidney of DN rats within the indicated period. Moreover, the expression of phosphorylated c-JUN NH(2)-terminal kinase, the monocyte chemoattractant protein-1, and the final fibrotic effector all elevated markedly in the kidney of DN rats. Urinary protein excretion rate and the concentration of urinary monocyte chemoattractant protein-1 were higher than those in the normal control group. Treatment with 4-PBA can suppress the expression of the glucose-regulated protein 78 and the phosphorylation of the PKR-like ER kinase, both of which are ER stress indicators; renoinflammatory signal; and the expression of inflammatory cytokines and fibrosis factors. It also can inhibit the increase in urinary protein excretion rate and urinary monocyte chemoattractant protein-1. In conclusion, 4-PBA exerts a marked renoprotective effect possibly due to modulating ER stress and related inflammatory cascade.
Collapse
Affiliation(s)
- Wei Qi
- Department of Nephrology, Xinqiao Hospital, The Third Military Medical University, Chongqing, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Peroxisome proliferator-activated receptor-α is renoprotective in doxorubicin-induced glomerular injury. Kidney Int 2011; 79:1302-11. [PMID: 21368746 DOI: 10.1038/ki.2011.17] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Doxorubicin (DOX) is an anthracycline antibiotic utilized in antitumor therapy; however, its clinical use is frequently impeded by renal toxic effects. As peroxisome proliferator-activated receptor-α (PPAR-α) has renoprotective effects in drug-related kidney injuries, we tested its ability to inhibit DOX-induced renal injury. Although both male PPAR-α knockout mice and their wild-type littermates (pure 129/SvJ background) had significant proteinuria 4 weeks after DOX treatment, those with deletion of PPAR-α had more severe proteinuria. This was associated with more serious podocyte foot process effacement compared with wild-type mice. In contrast, the PPAR-α agonist fenofibrate effectively reduced proteinuria and attenuated DOX-induced podocyte foot process effacement. Consistently, glomerular nephrin expression was significantly lower in the knockout compared with wild-type mice following DOX treatment. Fenofibrate therapy significantly blunted the reduction in glomerular nephrin levels in DOX-treated wild-type mice. In cultured podocytes, DOX induced apoptosis, increased cleaved caspase-3 levels, and decreased Bcl-2 expression, all attenuated by pretreatment with fenofibrate. Thus, PPAR-α deficiency exacerbates DOX-related renal injury, in part, due to increased podocyte apoptosis.
Collapse
|