1
|
Matsuta K, Kamiyama K, Imamoto T, Takeda I, Masunaga S, Kobayashi M, Takahashi N, Kasuno K, Hara M, Iwano M, Toyama T, Kimura H. PPAR-α Insufficiency Enhances Doxorubicin-Induced Nephropathy in PPAR-α Knockout Mice and a Murine Podocyte Cell Line. Cells 2024; 13:1446. [PMID: 39273018 PMCID: PMC11394432 DOI: 10.3390/cells13171446] [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: 07/31/2024] [Revised: 08/24/2024] [Accepted: 08/25/2024] [Indexed: 09/15/2024] Open
Abstract
Peroxisome proliferator-activated receptor-alpha (PPAR-α) and its exogenous activators (fibrates) promote autophagy. However, whether the deleterious effects of PPAR-α deficiency on doxorubicin (DOX)-induced podocytopathy are associated with reduced autophagy remains to be clarified. We investigated the mechanisms of PPAR-α in DOX-induced podocytopathy and tubular injury in PPAR-α knockout (PAKO) mice and in a murine podocyte cell line. DOX-treated PAKO mice showed higher serum levels of triglycerides and non-esterified fatty acids and more severe podocytopathy than DOX-treated wild-type mice, as evidenced by higher urinary levels of proteins and podocalyxin at 3 days to 2 weeks and higher blood urea nitrogen and serum creatinine levels at 4 weeks. Additionally, there was an increased accumulation of p62, a negative autophagy marker, in the glomerular and tubular regions in DOX-treated PAKO mice at Day 9. Moreover, DOX-treated PAKO mice showed more severe glomerulosclerosis and tubular damage and lower podocalyxin expression in the kidneys than DOX-treated control mice at 4 weeks. Furthermore, DOX treatment increased p-p53, an apoptosis marker, and cleaved the caspase-3 levels and induced apoptosis, which was ameliorated by fenofibrate, a PPAR-α activator. Fenofibrate further enhanced AMPK activation and autophagy under fed and fasting conditions. Conclusively, PPAR-α deficiency enhances DOX-induced podocytopathy, glomerulosclerosis, and tubular injury, possibly by reducing autophagic activity in mouse kidneys.
Collapse
Affiliation(s)
- Kohei Matsuta
- Department of Clinical Laboratory, University of Fukui Hospital, Fukui 910-1193, Japan; (K.M.); (K.K.); (T.I.); (I.T.); (S.M.); (T.T.)
| | - Kazuko Kamiyama
- Department of Clinical Laboratory, University of Fukui Hospital, Fukui 910-1193, Japan; (K.M.); (K.K.); (T.I.); (I.T.); (S.M.); (T.T.)
| | - Toru Imamoto
- Department of Clinical Laboratory, University of Fukui Hospital, Fukui 910-1193, Japan; (K.M.); (K.K.); (T.I.); (I.T.); (S.M.); (T.T.)
| | - Izumi Takeda
- Department of Clinical Laboratory, University of Fukui Hospital, Fukui 910-1193, Japan; (K.M.); (K.K.); (T.I.); (I.T.); (S.M.); (T.T.)
| | - Shinya Masunaga
- Department of Clinical Laboratory, University of Fukui Hospital, Fukui 910-1193, Japan; (K.M.); (K.K.); (T.I.); (I.T.); (S.M.); (T.T.)
| | - Mamiko Kobayashi
- Division of Nephrology, Department of General Medicine, School of Medicine, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan; (M.K.); (N.T.); (K.K.); (M.I.)
| | - Naoki Takahashi
- Division of Nephrology, Department of General Medicine, School of Medicine, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan; (M.K.); (N.T.); (K.K.); (M.I.)
| | - Kenji Kasuno
- Division of Nephrology, Department of General Medicine, School of Medicine, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan; (M.K.); (N.T.); (K.K.); (M.I.)
| | - Masanori Hara
- Iwamuro Health Promotion Center, Niigata 953-0104, Japan;
| | - Masayuki Iwano
- Division of Nephrology, Department of General Medicine, School of Medicine, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan; (M.K.); (N.T.); (K.K.); (M.I.)
| | - Tadashi Toyama
- Department of Clinical Laboratory, University of Fukui Hospital, Fukui 910-1193, Japan; (K.M.); (K.K.); (T.I.); (I.T.); (S.M.); (T.T.)
- Division of Nephrology, Department of General Medicine, School of Medicine, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan; (M.K.); (N.T.); (K.K.); (M.I.)
| | - Hideki Kimura
- Department of Clinical Laboratory, University of Fukui Hospital, Fukui 910-1193, Japan; (K.M.); (K.K.); (T.I.); (I.T.); (S.M.); (T.T.)
- Division of Nephrology, Department of General Medicine, School of Medicine, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan; (M.K.); (N.T.); (K.K.); (M.I.)
| |
Collapse
|
2
|
Lee LE, Doke T, Mukhi D, Susztak K. The key role of altered tubule cell lipid metabolism in kidney disease development. Kidney Int 2024; 106:24-34. [PMID: 38614389 PMCID: PMC11193624 DOI: 10.1016/j.kint.2024.02.025] [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: 02/26/2023] [Revised: 02/16/2024] [Accepted: 02/27/2024] [Indexed: 04/15/2024]
Abstract
Kidney epithelial cells have very high energy requirements, which are largely met by fatty acid oxidation. Complex changes in lipid metabolism are observed in patients with kidney disease. Defects in fatty acid oxidation and increased lipid uptake, especially in the context of hyperlipidemia and proteinuria, contribute to this excess lipid build-up and exacerbate kidney disease development. Recent studies have also highlighted the role of increased de novo lipogenesis in kidney fibrosis. The defect in fatty acid oxidation causes energy starvation. Increased lipid uptake, synthesis, and lower fatty acid oxidation can cause toxic lipid build-up, reactive oxygen species generation, and mitochondrial damage. A better understanding of these metabolic processes may open new treatment avenues for kidney diseases by targeting lipid metabolism.
Collapse
Affiliation(s)
- Lauren E Lee
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Penn-Children's Hospital of Philadelphia Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Tomohito Doke
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Penn-Children's Hospital of Philadelphia Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Dhanunjay Mukhi
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Penn-Children's Hospital of Philadelphia Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Penn-Children's Hospital of Philadelphia Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
3
|
Han Z, Ma K, Tao H, Liu H, Zhang J, Sai X, Li Y, Chi M, Nian Q, Song L, Liu C. A Deep Insight Into Regulatory T Cell Metabolism in Renal Disease: Facts and Perspectives. Front Immunol 2022; 13:826732. [PMID: 35251009 PMCID: PMC8892604 DOI: 10.3389/fimmu.2022.826732] [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: 12/01/2021] [Accepted: 01/24/2022] [Indexed: 11/29/2022] Open
Abstract
Kidney disease encompasses a complex set of diseases that can aggravate or start systemic pathophysiological processes through their complex metabolic mechanisms and effects on body homoeostasis. The prevalence of kidney disease has increased dramatically over the last two decades. CD4+CD25+ regulatory T (Treg) cells that express the transcription factor forkhead box protein 3 (Foxp3) are critical for maintaining immune homeostasis and preventing autoimmune disease and tissue damage caused by excessive or unnecessary immune activation, including autoimmune kidney diseases. Recent studies have highlighted the critical role of metabolic reprogramming in controlling the plasticity, stability, and function of Treg cells. They are also likely to play a vital role in limiting kidney transplant rejection and potentially promoting transplant tolerance. Metabolic pathways, such as mitochondrial function, glycolysis, lipid synthesis, glutaminolysis, and mammalian target of rapamycin (mTOR) activation, are involved in the development of renal diseases by modulating the function and proliferation of Treg cells. Targeting metabolic pathways to alter Treg cells can offer a promising method for renal disease therapy. In this review, we provide a new perspective on the role of Treg cell metabolism in renal diseases by presenting the renal microenvironment、relevant metabolites of Treg cell metabolism, and the role of Treg cell metabolism in various kidney diseases.
Collapse
Affiliation(s)
- Zhongyu Han
- Department of Nephrology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Sichuan Renal Disease Clinical Research Center, University of Electronic Science and Technology of China, Chengdu, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China.,Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kuai Ma
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hongxia Tao
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hongli Liu
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiong Zhang
- Department of Nephrology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Sichuan Renal Disease Clinical Research Center, University of Electronic Science and Technology of China, Chengdu, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Xiyalatu Sai
- Affiliated Hospital of Inner Mongolia University for the Nationalities, Tongliao, China
| | - Yunlong Li
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mingxuan Chi
- Department of Nephrology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Sichuan Renal Disease Clinical Research Center, University of Electronic Science and Technology of China, Chengdu, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Qing Nian
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China.,Department of Blood Transfusion Sicuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Linjiang Song
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chi Liu
- Department of Nephrology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Sichuan Renal Disease Clinical Research Center, University of Electronic Science and Technology of China, Chengdu, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| |
Collapse
|
4
|
Goto H, Shoda S, Nakashima H, Noguchi M, Imakiire T, Ohshima N, Kinoshita M, Tomimatsu S, Kumagai H. Early biomarkers for kidney injury in heat-related illness patients: a prospective observational study at Japanese Self-Defense Force Fuji Hospital. Nephrol Dial Transplant 2022; 38:644-654. [PMID: 35511214 PMCID: PMC9976769 DOI: 10.1093/ndt/gfac166] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Since heatstroke-induced acute kidney injury (AKI) can progress to chronic kidney disease, it would be useful to detect heatstroke-induced AKI and severe heat-related illness in the early phase. We studied the epidemiology of heat-related illness among patients in the Japanese Ground Self-Defense Force and evaluated the relationship between heat-related illness severity and early urinary biomarkers for AKI. METHODS We enrolled patients who were diagnosed with heat-related illness at the Self-Defense Force Fuji Hospital from 1 May to 30 September 2020. We compared the urinary kidney injury molecule-1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL), liver fatty acid-binding protein (L-FABP), N-acetyl-β-D-glucosaminidase (NAG) and β2-microglobulin levels according to the severity of heat-related illness as defined by positive scores for the Japanese Association of Acute Medicine Heatstroke Working Group (JAAM-HS-WG) criteria (0, mild; 1, moderate; ≥2, severe). RESULTS Of the 44 patients, kidney injury, defined as serum creatinine (sCr) ≥1.2 mg/dL, was seen in 9 (20.5%) patients. Urinary NAG, NGAL and L-FABP levels were significantly higher in the ≥2 JAAM-HS-WG criteria group than in the 0 group. Furthermore, urinary L-FABP levels were positively correlated with sCr levels. In contrast, the urinary KIM-1 levels showed the best correlation with serum cystatin C (sCysC) among these biomarkers. CONCLUSIONS We conclude even mild to moderate heatstroke could lead to AKI. Urinary L-FABP is useful for detecting heatstroke-induced AKI and patients with severe heat-related illness requiring immediate treatment. Urinary KIM-1 may detect heatstroke-induced AKI in terms of sCysC, although it was not related to the severity of heat-related illness.
Collapse
Affiliation(s)
| | - Shinichi Shoda
- Self-Defense Force Fuji Hospital, Subashiri, Shizuoka, Japan
| | - Hiroyuki Nakashima
- Department of Immunology and Microbiology, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Midori Noguchi
- Department of Nephrology and Endocrinology, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Toshihiko Imakiire
- Department of Nephrology and Endocrinology, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Naoki Ohshima
- Department of Nephrology and Endocrinology, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Manabu Kinoshita
- Department of Immunology and Microbiology, National Defense Medical College, Tokorozawa, Saitama, Japan
| | | | - Hiroo Kumagai
- Department of Nephrology and Endocrinology, National Defense Medical College, Tokorozawa, Saitama, Japan
| |
Collapse
|
5
|
Sun T, Wang D, Wang B, Liu X, Li N, Shi K. Melatonin attenuates cisplatin-induced acute kidney injury in mice: Involvement of PPARα and fatty acid oxidation. Food Chem Toxicol 2022; 163:112970. [PMID: 35367536 DOI: 10.1016/j.fct.2022.112970] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/21/2022] [Accepted: 03/24/2022] [Indexed: 10/18/2022]
Abstract
The present study focused on the protective effects of melatonin against cisplatin-induced acute kidney injury in mice and its possible mechanism of action in relation to the major regulator of fatty acid oxidation (FAO), peroxidase proliferative receptor α (PPARα). The experiment consisted of the following four groups: vehicle control, cisplatin (15 mg/kg), cisplatin & melatonin (20 mg/kg/day), and melatonin (20 mg/kg/day). Concomitant administration of melatonin significantly ameliorated cisplatin-induced acute kidney injury in mice by decreasing serum levels of triglyceride, blood urea nitrogen and creatinine, reducing the number and size of lipid droplets in tubular epithelial cells, and decreasing the incidence of histopathological changes including tubular cell apoptosis. Moreover, melatonin administration protected kidney tissue by significantly upregulating the levels of PPARα reduced by cisplatin injection, resulting in increased FAO pathway-associated genes (PGC-1a, Acadm, Acat1, Acsm2, Acsm3, Bdh2, Echs and Pecr) as well as reducing protein levels of caspase-3, -9 and Bax. Melatonin not only partially modulated FAO via PPARα signaling, but also decreased cisplatin-induced apoptosis by inhibiting the caspase-3, -9 and Bax pathways. Our findings suggest that melatonin prevents cisplatin-induced acute kidney injury in mice, possibly by upregulating the expression of PPARα, resulting in enhanced FAO and anti-apoptotic properties.
Collapse
Affiliation(s)
- Tao Sun
- Henan Medical College, Zhengzhou, 451191, China
| | - Di Wang
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Baoying Wang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, 450046, China
| | - Xianghua Liu
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, 450046, China
| | - Ningning Li
- Henan Medical College, Zhengzhou, 451191, China.
| | - Ke Shi
- Henan Medical College, Zhengzhou, 451191, China
| |
Collapse
|
6
|
Gao J, Gu Z. The Role of Peroxisome Proliferator-Activated Receptors in Kidney Diseases. Front Pharmacol 2022; 13:832732. [PMID: 35308207 PMCID: PMC8931476 DOI: 10.3389/fphar.2022.832732] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/14/2022] [Indexed: 12/20/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear hormone receptor superfamily of ligand-activated transcription factors. Accumulating evidence suggests that PPARs may play an important role in the pathogenesis of kidney disease. All three members of the PPAR subfamily, PPARα, PPARβ/δ, and PPARγ, have been implicated in many renal pathophysiological conditions, including acute kidney injury, diabetic nephropathy, and chronic kidney disease, among others. Emerging data suggest that PPARs may be potential therapeutic targets for renal disease. This article reviews the physiological roles of PPARs in the kidney and discusses the therapeutic utility of PPAR agonists in the treatment of kidney disease.
Collapse
Affiliation(s)
- Jianjun Gao
- Department of Nephrology, Chinese PLA Strategic Support Force Characteristic Medical Center, Beijing, China
| | - Zhaoyan Gu
- Department of Endocrinology, Second Medical Center, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
- *Correspondence: Zhaoyan Gu,
| |
Collapse
|
7
|
Xu S, Jia P, Fang Y, Jin J, Sun Z, Zhou W, Li J, Zhang Y, Wang X, Ren T, Zou Z, Ding X. Nuclear farnesoid X receptor attenuates acute kidney injury through fatty acid oxidation. Kidney Int 2022; 101:987-1002. [DOI: 10.1016/j.kint.2022.01.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 12/28/2021] [Accepted: 01/11/2022] [Indexed: 12/14/2022]
|
8
|
Lin PH, Duann P. Dyslipidemia in Kidney Disorders: Perspectives on Mitochondria Homeostasis and Therapeutic Opportunities. Front Physiol 2020; 11:1050. [PMID: 33013450 PMCID: PMC7494972 DOI: 10.3389/fphys.2020.01050] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 07/30/2020] [Indexed: 12/11/2022] Open
Abstract
To excrete body nitrogen waste and regulate electrolyte and fluid balance, the kidney has developed into an energy factory with only second to the heart in mitochondrial content in the body to meet the high-energy demand and regulate homeostasis. Energy supply from the renal mitochondria majorly depends on lipid metabolism, with programed enzyme systems in fatty acid β-oxidation and Krebs cycle. Renal mitochondria integrate several metabolic pathways, including AMPK/PGC-1α, PPARs, and CD36 signaling to maintain energy homeostasis for dynamic and static requirements. The pathobiology of several kidney disorders, including diabetic nephropathy, acute and chronic kidney injuries, has been primarily linked to impaired mitochondrial bioenergetics. Such homeostatic disruption in turn stimulates a pathological adaptation, with mitochondrial enzyme system reprograming possibly leading to dyslipidemia. However, this alteration, while rescuing oncotic pressure deficit secondary to albuminuria and dissipating edematous disorder, also imposes an ominous lipotoxic consequence. Reprograming of lipid metabolism in kidney injury is essential to preserve the integrity of kidney mitochondria, thereby preventing massive collateral damage including excessive autophagy and chronic inflammation. Here, we review dyslipidemia in kidney disorders and the most recent advances on targeting mitochondrial energy metabolism as a therapeutic strategy to restrict renal lipotoxicity, achieve salutary anti-edematous effects, and restore mitochondrial homeostasis.
Collapse
Affiliation(s)
- Pei-Hui Lin
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Pu Duann
- Research and Development, Salem Veteran Affairs Medical Center, Salem, VA, United States
| |
Collapse
|
9
|
Jang HS, Noh MR, Kim J, Padanilam BJ. Defective Mitochondrial Fatty Acid Oxidation and Lipotoxicity in Kidney Diseases. Front Med (Lausanne) 2020; 7:65. [PMID: 32226789 PMCID: PMC7080698 DOI: 10.3389/fmed.2020.00065] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 02/13/2020] [Indexed: 12/31/2022] Open
Abstract
The kidney is a highly metabolic organ and uses high levels of ATP to maintain electrolyte and acid-base homeostasis and reabsorb nutrients. Energy depletion is a critical factor in development and progression of various kidney diseases including acute kidney injury (AKI), chronic kidney disease (CKD), and diabetic and glomerular nephropathy. Mitochondrial fatty acid β-oxidation (FAO) serves as the preferred source of ATP in the kidney and its dysfunction results in ATP depletion and lipotoxicity to elicit tubular injury and inflammation and subsequent fibrosis progression. This review explores the current state of knowledge on the role of mitochondrial FAO dysfunction in the pathophysiology of kidney diseases including AKI and CKD and prospective views on developing therapeutic interventions based on mitochondrial energy metabolism.
Collapse
Affiliation(s)
- Hee-Seong Jang
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Mi Ra Noh
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Jinu Kim
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States.,Department of Anatomy, Jeju National University School of Medicine, Jeju, South Korea.,Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju, South Korea
| | - Babu J Padanilam
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States.,Internal Medicine, Section of Nephrology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
10
|
Naruse H, Ishii J, Takahashi H, Kitagawa F, Nishimura H, Kawai H, Muramatsu T, Harada M, Yamada A, Fujiwara W, Hayashi M, Motoyama S, Sarai M, Watanabe E, Izawa H, Ozaki Y. Urinary Liver-Type Fatty-Acid-Binding Protein Predicts Long-Term Adverse Outcomes in Medical Cardiac Intensive Care Units. J Clin Med 2020; 9:jcm9020482. [PMID: 32050627 PMCID: PMC7073895 DOI: 10.3390/jcm9020482] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/06/2020] [Accepted: 02/07/2020] [Indexed: 12/14/2022] Open
Abstract
We prospectively investigated the prognostic value of urinary liver-type fatty-acid-binding protein (L-FABP) levels on hospital admission, both independently and in combination with serum creatinine-defined acute kidney injury (AKI), to predict long-term adverse outcomes in 1119 heterogeneous patients (mean age; 68 years) treated at medical (non-surgical) cardiac intensive care units (CICUs). Patients with stage 5 chronic kidney disease were excluded from the study. Of these patients, 47% had acute coronary syndrome and 38% had acute decompensated heart failure. The creatinine-defined AKI was diagnosed according to the "Kidney Disease: Improving Global Outcomes" criteria. The primary endpoint was a composite of all-cause death or progression to end-stage kidney disease, indicating the initiation of maintenance dialysis therapy or kidney transplantation. Creatinine-defined AKI occurred in 207 patients, with 44 patients having stage 2 or 3 disease. During a mean follow-up period of 41 months after enrollment, the primary endpoint occurred in 242 patients. Multivariate Cox regression analyses revealed L-FABP levels as independent predictors of the primary endpoint (p < 0.001). Adding L-FABP to a baseline model with established risk factors further enhanced reclassification and discrimination beyond that of the baseline model alone, for primary-endpoint prediction (both; p < 0.01). On Kaplan-Meier analyses, increased L-FABP (≥4th quintile value of 9.0 ng/mL) on admission or presence of creatinine-defined AKI, correlated with an increased risk of the primary endpoint (p < 0.001). Thus, urinary L-FABP levels on admission are potent and independent predictors of long-term adverse outcomes, and they might improve the long-term risk stratification of patients admitted at medical CICUs, when used in combination with creatinine-defined AKI.
Collapse
Affiliation(s)
- Hiroyuki Naruse
- Department of Joint Research Laboratory of Clinical Medicine, Fujita Health University School of Medicine, Toyoake 470-1192, Japan (F.K.)
| | - Junnichi Ishii
- Department of Joint Research Laboratory of Clinical Medicine, Fujita Health University School of Medicine, Toyoake 470-1192, Japan (F.K.)
- Correspondence: ; Tel.: +81-562-93-2312
| | - Hiroshi Takahashi
- Division of Statistics, Fujita Health University School of Medicine, Toyoake 470-1192, Japan;
| | - Fumihiko Kitagawa
- Department of Joint Research Laboratory of Clinical Medicine, Fujita Health University School of Medicine, Toyoake 470-1192, Japan (F.K.)
| | - Hideto Nishimura
- Department of Cardiology, Fujita Health University School of Medicine, Toyoake 470-1192, Japan (H.K.); (T.M.); (M.H.); (A.Y.); (S.M.); (M.S.); (E.W.)
| | - Hideki Kawai
- Department of Cardiology, Fujita Health University School of Medicine, Toyoake 470-1192, Japan (H.K.); (T.M.); (M.H.); (A.Y.); (S.M.); (M.S.); (E.W.)
| | - Takashi Muramatsu
- Department of Cardiology, Fujita Health University School of Medicine, Toyoake 470-1192, Japan (H.K.); (T.M.); (M.H.); (A.Y.); (S.M.); (M.S.); (E.W.)
| | - Masahide Harada
- Department of Cardiology, Fujita Health University School of Medicine, Toyoake 470-1192, Japan (H.K.); (T.M.); (M.H.); (A.Y.); (S.M.); (M.S.); (E.W.)
| | - Akira Yamada
- Department of Cardiology, Fujita Health University School of Medicine, Toyoake 470-1192, Japan (H.K.); (T.M.); (M.H.); (A.Y.); (S.M.); (M.S.); (E.W.)
| | - Wakaya Fujiwara
- Department of Cardiology, Bantane Hospital, Nagoya 454-8509, Japan; (W.F.); (M.H.); (H.I.)
| | - Mutsuharu Hayashi
- Department of Cardiology, Bantane Hospital, Nagoya 454-8509, Japan; (W.F.); (M.H.); (H.I.)
| | - Sadako Motoyama
- Department of Cardiology, Fujita Health University School of Medicine, Toyoake 470-1192, Japan (H.K.); (T.M.); (M.H.); (A.Y.); (S.M.); (M.S.); (E.W.)
| | - Masayoshi Sarai
- Department of Cardiology, Fujita Health University School of Medicine, Toyoake 470-1192, Japan (H.K.); (T.M.); (M.H.); (A.Y.); (S.M.); (M.S.); (E.W.)
| | - Eiichi Watanabe
- Department of Cardiology, Fujita Health University School of Medicine, Toyoake 470-1192, Japan (H.K.); (T.M.); (M.H.); (A.Y.); (S.M.); (M.S.); (E.W.)
| | - Hideo Izawa
- Department of Cardiology, Bantane Hospital, Nagoya 454-8509, Japan; (W.F.); (M.H.); (H.I.)
| | - Yukio Ozaki
- Department of Cardiology, Fujita Health University School of Medicine, Toyoake 470-1192, Japan (H.K.); (T.M.); (M.H.); (A.Y.); (S.M.); (M.S.); (E.W.)
| |
Collapse
|
11
|
Jang HS, Noh MR, Jung EM, Kim WY, Southekal S, Guda C, Foster KW, Oupicky D, Ferrer FA, Padanilam BJ. Proximal tubule cyclophilin D regulates fatty acid oxidation in cisplatin-induced acute kidney injury. Kidney Int 2020; 97:327-339. [PMID: 31733829 PMCID: PMC6983334 DOI: 10.1016/j.kint.2019.08.019] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 07/25/2019] [Accepted: 08/02/2019] [Indexed: 12/12/2022]
Abstract
Regardless of the etiology, acute kidney injury involves aspects of mitochondrial dysfunction and ATP depletion. Fatty acid oxidation is the preferred energy source of the kidney and is inhibited during acute kidney injury. A pivotal role for the mitochondrial matrix protein, cyclophilin D in regulating overall cell metabolism is being unraveled. We hypothesize that mitochondrial interaction of proximal tubule cyclophilin D and the transcription factor PPARα modulate fatty acid beta-oxidation in cisplatin-induced acute kidney injury. Cisplatin injury resulted in histological and functional damage in the kidney with downregulation of fatty acid oxidation genes and increase of intrarenal lipid accumulation. However, proximal tubule-specific deletion of cyclophilin D protected the kidneys from the aforementioned effects. Mitochondrial translocation of PPARα, its binding to cyclophilin D, and sequestration led to inhibition of its nuclear translocation and transcription of PPARα-regulated fatty acid oxidation genes during cisplatin-induced acute kidney injury. Genetic or pharmacological inhibition of cyclophilin D preserved nuclear expression and transcriptional activity of PPARα and prevented the impairment of fatty acid oxidation and intracellular lipid accumulation. Docking analysis identified potential binding sites between PPARα and cyclophilin D. Thus, our results indicate that proximal tubule cyclophilin D elicits impaired mitochondrial fatty acid oxidation via mitochondrial interaction between cyclophilin D and PPARα. Hence, targeting their interaction may be a potential therapeutic strategy to prevent energy depletion, lipotoxicity and cell death in cisplatin-induced acute kidney injury.
Collapse
Affiliation(s)
- Hee-Seong Jang
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA.
| | - Mi Ra Noh
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Eui-Man Jung
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Woo-Yang Kim
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| | - Siddesh Southekal
- Department of Genetics, Cell Biology, and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Chittibabu Guda
- Department of Genetics, Cell Biology, and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Kirk W Foster
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - David Oupicky
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Fernando A Ferrer
- Department of Surgery, Children's Hospital and Medical Center, Omaha, Nebraska, USA; Department of Pediatrics, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Babu J Padanilam
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA; Department of Internal Medicine, Section of Nephrology, University of Nebraska Medical Center, Omaha, Nebraska, USA.
| |
Collapse
|
12
|
Tajima S, Yamamoto N, Masuda S. Clinical prospects of biomarkers for the early detection and/or prediction of organ injury associated with pharmacotherapy. Biochem Pharmacol 2019; 170:113664. [PMID: 31606409 DOI: 10.1016/j.bcp.2019.113664] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/08/2019] [Indexed: 12/31/2022]
Abstract
Several biomarkers are used to monitor organ damage caused by drug toxicity. Traditional markers of kidney function, such as serum creatinine and blood urea nitrogen are commonly used to estimate glomerular filtration rate. However, these markers have several limitations including poor specificity and sensitivity. A number of serum and urine biomarkers have recently been described to detect kidney damage caused by drugs such as cisplatin, gentamicin, vancomycin, and tacrolimus. Neutrophil gelatinase-associated lipocalin (NGAL), liver-type fatty acid-binding protein (L-FABP), kidney injury molecule-1 (KIM-1), monocyte chemotactic protein-1 (MCP-1), and cystatin C have been identified as biomarkers for early kidney damage. Hy's Law is widely used as to predict a high risk of severe drug-induced liver injury caused by drugs such as acetaminophen. Recent reports have indicated that glutamate dehydrogenase (GLDH), high-mobility group box 1 (HMGB-1), Keratin-18 (k18), MicroRNA-122 and ornithine carbamoyltransferase (OCT) are more sensitive markers of hepatotoxicity compared to the traditional markers including the blood levels of amiotransferases and total bilirubin. Additionally, the rapid development of proteomic technologies in biofluids and tissue provides a new multi-marker panel, leading to the discovery of more sensitive biomarkers. In this review, an update topics of biomarkers for the detection of kidney or liver injury associated with pharmacotherapy.
Collapse
Affiliation(s)
- Soichiro Tajima
- Department of Pharmacy, Kyushu University Hospital, Fukuoka 812-8582, Japan
| | - Nanae Yamamoto
- Department of Pharmacy, Kyushu University Hospital, Fukuoka 812-8582, Japan
| | - Satohiro Masuda
- Department of Pharmacy, Kyushu University Hospital, Fukuoka 812-8582, Japan; Department of Clinical Pharmacology and Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; Department of Pharmacy, International University of Health and Welfare Narita Hospital, Japan; Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, International University of Health and Welfare Narita Hospital, Japan.
| |
Collapse
|
13
|
Iwaki T, Bennion BG, Stenson EK, Lynn JC, Otinga C, Djukovic D, Raftery D, Fei L, Wong HR, Liles WC, Standage SW. PPARα contributes to protection against metabolic and inflammatory derangements associated with acute kidney injury in experimental sepsis. Physiol Rep 2019; 7:e14078. [PMID: 31102342 PMCID: PMC6525329 DOI: 10.14814/phy2.14078] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/02/2019] [Accepted: 04/04/2019] [Indexed: 01/05/2023] Open
Abstract
Sepsis-associated acute kidney injury (AKI) is a significant problem in critically ill children and adults resulting in increased morbidity and mortality. Fundamental mechanisms contributing to sepsis-associated AKI are poorly understood. Previous research has demonstrated that peroxisome proliferator-activated receptor α (PPARα) expression is associated with reduced organ system failure in sepsis. Using an experimental model of polymicrobial sepsis, we demonstrate that mice deficient in PPARα have worse kidney function, which is likely related to reduced fatty acid oxidation and increased inflammation. Ultrastructural evaluation with electron microscopy reveals that the proximal convoluted tubule is specifically injured in septic PPARα deficient mice. In this experimental group, serum metabolomic analysis reveals unanticipated metabolic derangements in tryptophan-kynurenine-NAD+ and pantothenate pathways. We also show that a subgroup of children with sepsis whose genome-wide expression profiles are characterized by repression of the PPARα signaling pathway has increased incidence of severe AKI. These findings point toward interesting associations between sepsis-associated AKI and PPARα-driven fatty acid metabolism that merit further investigation.
Collapse
Affiliation(s)
- Takuma Iwaki
- Department of PediatricsUniversity of Washington School of MedicineSeattleWashington
- Department of PediatricsUniversity HospitalFaculty of MedicineKagawa UniversityKagawaJapan
| | - Brock G. Bennion
- Department of PediatricsUniversity of Washington School of MedicineSeattleWashington
- Department of Pathology and ImmunologyWashington University School of MedicineSt. LouisMissouri
| | - Erin K. Stenson
- Department of PediatricsSection of Critical CareUniversity of Colorado School of MedicineAnschutz Medical CenterChildren's Hospital ColoradoAuroraColorado
- Division of Critical Care MedicineCincinnati Children's Hospital Medical CenterCincinnatiOhio
| | - Jared C. Lynn
- Department of PediatricsUniversity of Washington School of MedicineSeattleWashington
| | - Cynthia Otinga
- Department of PediatricsUniversity of Washington School of MedicineSeattleWashington
| | - Danijel Djukovic
- Department of Chemistry and BiochemistryUniversity of ColoradoBoulderColorado
- Department of Anesthesiology and Pain MedicineUniversity of Washington School of MedicineSeattleWashington
| | - Daniel Raftery
- Department of Anesthesiology and Pain MedicineUniversity of Washington School of MedicineSeattleWashington
| | - Lin Fei
- Division of Biostatistics and EpidemiologyCincinnati Children's Hospital Medical CenterCincinnatiOhio
- Department of PediatricsUniversity of CincinnatiCincinnatiOhio
| | - Hector R. Wong
- Division of Critical Care MedicineCincinnati Children's Hospital Medical CenterCincinnatiOhio
- Department of PediatricsUniversity of CincinnatiCincinnatiOhio
| | - W. Conrad Liles
- Department of MedicineUniversity of Washington School of MedicineSeattleWashington
| | - Stephen W. Standage
- Department of PediatricsUniversity of Washington School of MedicineSeattleWashington
- Division of Critical Care MedicineCincinnati Children's Hospital Medical CenterCincinnatiOhio
- Department of PediatricsUniversity of CincinnatiCincinnatiOhio
| |
Collapse
|
14
|
Nephroprotective Effect of Embryonic Stem Cells Reducing Lipid Peroxidation in Kidney Injury Induced by Cisplatin. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:5420624. [PMID: 31001374 PMCID: PMC6437739 DOI: 10.1155/2019/5420624] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 02/03/2019] [Indexed: 12/25/2022]
Abstract
Introduction The acute kidney injury (AKI) is characterized by a sudden glomerular filtration reduction. Renal or intrinsic causes of AKI include nephrotoxicity induced by exogenous agents like cisplatin, which causes oxidative stress altering the biochemical process and leading to apoptosis. Therefore, this research is aimed at analyzing the embryonic stem cells (ESC) nephroprotective effect in AKI induced by cisplatin, employing genetic, phenotypic, and microspectroscopic techniques. Methods Thirty mice were randomly divided into three groups (n = 10): the healthy, isotonic salt solution (ISS), and mouse embryonic stem cells (mESC) groups. The ISS and mESC groups were subjected to AKI using cisplatin; 24 h post-AKI received an intraperitoneal injection of ISS or 1 × 106 mESC, respectively. At days 4 and 8 post-AKI, five mice of each group were sacrificed to analyze the histopathological, genetic (PDK4 and HO-1), protein (p53), and vibrational microspectroscopic changes. Results Histopathologically, interstitial nephritis and acute tubular necrosis were observed; however, the mESC group showed a more preserved microarchitecture with high cellularity. Additionally, the PDK4 and HO-1 gene expression only increased in the ISS group on day 4 post-AKI. Likewise, p53 was more immunoexpressed at day 8 post-AKI in the ISS group. About biomolecular analysis by microspectroscopy, bands associated with lipids, proteins, and nucleic acids were evidenced. Besides, ratios related to membrane function (protein/lipid), unsaturated lipid content (olefinic/total lipid, olefinic/total CH2, and CH2/CH3), and lipid peroxidation demonstrated oxidative stress induction and lipid peroxidation increase mainly in the ISS group. Finally, the principal component analysis discriminated against each group; nonetheless, some data of the healthy and mESC groups at day 8 were correlated. Conclusions The mESC implant diminishes cisplatin nephrotoxicity, once the protective effect in the reduction of lipid peroxidation was demonstrated, reflecting a functional and histological restoration.
Collapse
|
15
|
AMP-Activated Protein Kinase (AMPK)-Dependent Regulation of Renal Transport. Int J Mol Sci 2018; 19:ijms19113481. [PMID: 30404151 PMCID: PMC6274953 DOI: 10.3390/ijms19113481] [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] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 10/26/2018] [Accepted: 10/30/2018] [Indexed: 02/06/2023] Open
Abstract
AMP-activated kinase (AMPK) is a serine/threonine kinase that is expressed in most cells and activated by a high cellular AMP/ATP ratio (indicating energy deficiency) or by Ca2+. In general, AMPK turns on energy-generating pathways (e.g., glucose uptake, glycolysis, fatty acid oxidation) and stops energy-consuming processes (e.g., lipogenesis, glycogenesis), thereby helping cells survive low energy states. The functional element of the kidney, the nephron, consists of the glomerulus, where the primary urine is filtered, and the proximal tubule, Henle's loop, the distal tubule, and the collecting duct. In the tubular system of the kidney, the composition of primary urine is modified by the reabsorption and secretion of ions and molecules to yield final excreted urine. The underlying membrane transport processes are mainly energy-consuming (active transport) and in some cases passive. Since active transport accounts for a large part of the cell's ATP demands, it is an important target for AMPK. Here, we review the AMPK-dependent regulation of membrane transport along nephron segments and discuss physiological and pathophysiological implications.
Collapse
|
16
|
Balampanis K, Chasapi A, Kourea E, Tanoglidi A, Hatziagelaki E, Lambadiari V, Dimitriadis G, Lambrou GI, Kalfarentzos F, Melachrinou M, Sotiropoulou-Bonikou G. Inter-tissue expression patterns of the key metabolic biomarker PGC-1α in severely obese individuals: Implication in obesity-induced disease. Hellenic J Cardiol 2018; 60:282-293. [PMID: 30138744 DOI: 10.1016/j.hjc.2018.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 07/29/2018] [Accepted: 08/03/2018] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE PGC-1α is already known as a significant regulator of mitochondrial biogenesis, oxidative phosphorylation and fatty acid metabolism. Our study focuses on the role of PGC1α in morbid obesity, in five different tissues, collected from 50 severely obese patients during planned bariatric surgery. METHODS The investigated tissues included subcutaneous adipose tissue (SAT), visceral adipose tissue (VAT), skeletal muscle (SM), extramyocellular adipose tissue (EMAT) and liver. PGC1α expression was investigated with immunohistochemistry and evaluated with microscopy. RESULTS Our findings highlighted significant positive inter-tissue correlations regarding PGC-1α expression between several tissue pairs (VAT-SAT, VAT-SM, VAT-EMAT, SAT-SM, SAT-EMAT, SM-EMAT). Moreover, we found significant negative correlations between PGC1α expression in VAT with CD68 expression in skeletal muscle and EMAT, implying a possible protective role of PGC1α against obesity-induced inflammation. CONCLUSION Unmasking the inter-tissue communication networks regarding PGC-1α expression in morbid obesity, will give more insight into its significant role in obesity-induced diseases. PGC1α could potentially represent a future preventive and therapeutic target against obesity-induced disease, probably through enhancing mitochondrial biogenesis and metabolism.
Collapse
Affiliation(s)
- Konstantinos Balampanis
- Department of Pathology, Medical School, University of Patras, 26500 Patras, Greece; Second Department of Internal Medicine, Research Unit and Diabetes Center, Attikon University Hospital, National and Kapodistrian University of Athens, Medical School, Rimini 1, Haidari, 12462 Athens, Greece.
| | - Athina Chasapi
- Department of Pathology, Medical School, University of Patras, 26500 Patras, Greece.
| | - Eleni Kourea
- Department of Pathology, Medical School, University of Patras, 26500 Patras, Greece.
| | - Anna Tanoglidi
- Department of Clinical Pathology, Akademiska University, Uppsala, Sweden.
| | - Erifili Hatziagelaki
- Second Department of Internal Medicine, Research Unit and Diabetes Center, Attikon University Hospital, National and Kapodistrian University of Athens, Medical School, Rimini 1, Haidari, 12462 Athens, Greece.
| | - Vaia Lambadiari
- Second Department of Internal Medicine, Research Unit and Diabetes Center, Attikon University Hospital, National and Kapodistrian University of Athens, Medical School, Rimini 1, Haidari, 12462 Athens, Greece.
| | - George Dimitriadis
- Second Department of Internal Medicine, Research Unit and Diabetes Center, Attikon University Hospital, National and Kapodistrian University of Athens, Medical School, Rimini 1, Haidari, 12462 Athens, Greece.
| | - George I Lambrou
- First Department of Pediatrics, Choremeio Research Laboratory, National and Kapodistrian University of Athens, Medical School, Thivon & Levadeias 8, Goudi, 11527 Athens, Greece.
| | - Fotios Kalfarentzos
- Department of Surgery, Medical School, University of Patras, 26500 Patras, Greece.
| | - Maria Melachrinou
- Department of Pathology, Medical School, University of Patras, 26500 Patras, Greece.
| | | |
Collapse
|
17
|
Bontha SV, Maluf DG, Archer KJ, Dumur CI, Dozmorov M, King A, Akalin E, Mueller TF, Gallon L, Mas VR. Effects of DNA Methylation on Progression to Interstitial Fibrosis and Tubular Atrophy in Renal Allograft Biopsies: A Multi-Omics Approach. Am J Transplant 2017; 17:3060-3075. [PMID: 28556588 PMCID: PMC5734859 DOI: 10.1111/ajt.14372] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 05/01/2017] [Accepted: 05/20/2017] [Indexed: 01/25/2023]
Abstract
Progressive fibrosis of the interstitium is the dominant final pathway in renal destruction in native and transplanted kidneys. Over time, the continuum of molecular events following immunological and nonimmunological insults lead to interstitial fibrosis and tubular atrophy and culminate in kidney failure. We hypothesize that these insults trigger changes in DNA methylation (DNAm) patterns, which in turn could exacerbate injury and slow down the regeneration processes, leading to fibrosis development and graft dysfunction. Herein, we analyzed biopsy samples from kidney allografts collected 24 months posttransplantation and used an integrative multi-omics approach to understand the underlying molecular mechanisms. The role of DNAm and microRNAs on the graft gene expression was evaluated. Enrichment analyses of differentially methylated CpG sites were performed using GenomeRunner. CpGs were strongly enriched in regions that were variably methylated among tissues, implying high tissue specificity in their regulatory impact. Corresponding to this methylation pattern, gene expression data were related to immune response (activated state) and nephrogenesis (inhibited state). Preimplantation biopsies showed similar DNAm patterns to normal allograft biopsies at 2 years posttransplantation. Our findings demonstrate for the first time a relationship among epigenetic modifications and development of interstitial fibrosis, graft function, and inter-individual variation on long-term outcomes.
Collapse
Affiliation(s)
- Sai Vineela Bontha
- Translational Genomics Transplant Laboratory, Transplant Division, University of Virginia, Department of Surgery, PO Box 800625. 409 Lane Rd, Charlottesville, VA, 22908- 0625, USA
| | - Daniel G. Maluf
- Translational Genomics Transplant Laboratory, Transplant Division, University of Virginia, Department of Surgery, PO Box 800625. 409 Lane Rd, Charlottesville, VA, 22908- 0625, USA
| | - Kellie J. Archer
- Division of Biostatistics, The Ohio State University, 1841 Neil Avenue, 240 Cunz Hall, Columbus, OH 43210
| | - Catherine I. Dumur
- Department of Pathology, Virginia Commonwealth University, PO Box 980662, 1101 E. Marshall Street, Richmond, VA 23298-0662
| | - Mikhail Dozmorov
- Department of Biostatistics, Virginia Commonwealth University, One Capitol Square, room 730, 830 East Main Street, Richmond, Virginia 23298
| | - Anne King
- Division of Nephrology, Internal Medicine. Virginia commonwealth University, VA, 1101 E. Marshall Street, Richmond, VA 23298-0662
| | - Enver Akalin
- Departments of Clinical Medicine and Surgery, Albert Einstein College of Medicine Montefiore Medical Center, 11 E 210th St, Bronx, NY 10467
| | - Thomas F. Mueller
- Division of Nephorology, Internal Medicine, University Hospital Zurich, Ramistrasse 100, Zurich-8091
| | - Lorenzo Gallon
- Department of Medicine-Nephrology, Northwestern University676 N St Clair St # 100, Chicago, IL 60611
| | - Valeria R. Mas
- Translational Genomics Transplant Laboratory, Transplant Division, University of Virginia, Department of Surgery, PO Box 800625. 409 Lane Rd, Charlottesville, VA, 22908- 0625, USA
| |
Collapse
|
18
|
Ezaki T, Nishiumi S, Azuma T, Yoshida M. Metabolomics for the early detection of cisplatin-induced nephrotoxicity. Toxicol Res (Camb) 2017; 6:843-853. [PMID: 30090547 PMCID: PMC6062266 DOI: 10.1039/c7tx00171a] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 08/23/2017] [Indexed: 01/21/2023] Open
Abstract
Cisplatin, which is an inorganic molecule containing a platinum ion, is an antineoplastic agent that has been used to treat various solid tumors. However, its side effects include nephrotoxicity, neurotoxicity, bone marrow toxicity, gastrointestinal toxicity, and ototoxicity, which can limit its use. In this study, nephrotoxicity was caused by the intraperitoneal injection of cisplatin into rats, and then metabolome analysis was performed using gas chromatography/mass spectrometry (GC/MS) and liquid chromatography/mass spectrometry (LC/MS) to find plasma metabolite biomarker candidates that would facilitate the early detection of cisplatin-induced nephrotoxicity. As a result, chronological changes were detected in the plasma levels of cysteine-cystine and 3-hydroxy-butyrate in the GC/MS-based metabolomics study. In the LC/MS-based metabolomics study, 3 acylcarnitines and a phosphatidylethanolamine with C18:2-C18:2 were identified as potential plasma biomarkers of cisplatin-induced nephrotoxicity. The plasma levels of these 6 metabolites altered significantly after the administration of cisplatin, and these alterations occurred quicker than the equivalent changes in the plasma levels of creatinine and blood urea nitrogen, which are usually used as indicators of renal dysfunction. These results indicate that the abovementioned metabolites might be reliable biomarkers that would allow the earlier detection of cisplatin-induced nephrotoxicity and that metabolomics is a useful tool for discovering biomarkers that could be used to predict the side effects of cancer therapy.
Collapse
Affiliation(s)
- Takeshi Ezaki
- Division of Gastroenterology , Department of Internal Medicine , Kobe University Graduate School of Medicine , Japan . ; ; Tel: +81-78-382-6305
| | - Shin Nishiumi
- Division of Gastroenterology , Department of Internal Medicine , Kobe University Graduate School of Medicine , Japan . ; ; Tel: +81-78-382-6305
| | - Takeshi Azuma
- Division of Gastroenterology , Department of Internal Medicine , Kobe University Graduate School of Medicine , Japan . ; ; Tel: +81-78-382-6305
| | - Masaru Yoshida
- Division of Gastroenterology , Department of Internal Medicine , Kobe University Graduate School of Medicine , Japan . ; ; Tel: +81-78-382-6305
- Metabolomics Research , Department of Internal Related , Kobe University Graduate School of Medicine , Japan
- AMED-CREST , AMED , Japan
| |
Collapse
|
19
|
Su W, Cao R, He YC, Guan YF, Ruan XZ. Crosstalk of Hyperglycemia and Dyslipidemia in Diabetic Kidney Disease. KIDNEY DISEASES 2017; 3:171-180. [PMID: 29344511 DOI: 10.1159/000479874] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 07/28/2017] [Indexed: 01/02/2023]
Abstract
Background Diabetic kidney disease (DKD) is defined by the functional, structural, and clinical abnormalities of the kidney that are caused by diabetes. Summary One-third of both type 1 diabetes and type 2 diabetes patients suffer from DKD, which is the leading cause of end-stage renal disease, and is also associated with cardiovascular disease and high public health care costs. Serum glucose level and lipid level are key factors in the pathogenesis of DKD and are modifiable. The goal of this review is to provide an update on the roles of glucose and lipid metabolism in DKD and their crosstalk at the molecular level. We will further discuss the recent advances regarding metabolic nuclear receptors in glucose-lipid crosstalk, which may provide new potential therapeutic targets for DKD. Key Message AMPK, SREBP-1, and some metabolic hormone receptors including liver X receptors, farnesoid X receptors, and peroxisome proliferator-activated receptors mediate the crosstalk of hyperglycemia and dyslipidemia in diabetic kidney disease and might be potential treatment candidates.
Collapse
Affiliation(s)
- Wen Su
- AstraZeneca - Shenzhen University Joint Institute of Nephrology, Center for Nephrology and Urology, Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, Shenzhen, China
| | - Rong Cao
- Department of Nephrology, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Yong Cheng He
- Department of Nephrology, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - You Fei Guan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Xiong Zhong Ruan
- John Moorhead Research Laboratory, Centre for Nephrology, University College London Medical School, Royal Free Campus, London, UK
| |
Collapse
|
20
|
Mizokami F, Mizuno T. Acute kidney injury induced by antimicrobial agents in the elderly: awareness and mitigation strategies. Drugs Aging 2016; 32:1-12. [PMID: 25491560 DOI: 10.1007/s40266-014-0232-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The use of antimicrobial agents has increased in recent years as treatments have diversified and resistant bacteria have appeared. With increased use of antimicrobial agents, elderly patients are prone to adverse drug reactions (ADRs) as a result of factors such as drug-drug interactions, polypharmacy, long-term use, and over- or under-dosage. In particular, elderly patients using antimicrobials are at increased risk to develop drug-induced acute kidney injury (AKI), which is the most common severe ADR in such patients. AKI is a serious problem that is associated with mortality amongst hospitalized patients. Antimicrobial-induced AKI can be classified into three different types: acute tubular necrosis (ATN), acute interstitial nephritis (AIN), and renal tubule lumen obstruction. AKI can generally be prevented by proper maintenance of fluid balance. To design dosage regimens that ensure efficient drug excretion via the kidney, it is necessary to accurately estimate renal function; however, the kidney undergoes age-dependent structural and functional alterations over time. Therefore, proper management of antimicrobial agents by an antimicrobial stewardship team may lead to decreased incidence of AKI. This article reviews antimicrobial-induced AKI and discusses potential strategies for increasing awareness of AKI and mitigating its clinical effects.
Collapse
Affiliation(s)
- Fumihiro Mizokami
- Department of Pharmacy, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, Aichi, 474-8511, Japan,
| | | |
Collapse
|
21
|
Kuncewitch M, Yang WL, Jacob A, Khader A, Giangola M, Nicastro J, Coppa GF, Wang P. Inhibition of fatty acid synthase with C75 decreases organ injury after hemorrhagic shock. Surgery 2015; 159:570-9. [PMID: 26372196 DOI: 10.1016/j.surg.2015.07.036] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 06/30/2015] [Accepted: 07/01/2015] [Indexed: 01/28/2023]
Abstract
BACKGROUND Hemorrhagic shock is the primary cause of morbidity and mortality in the intensive care units in patients under the age of 35. Several organs, including the lungs, are seriously affected by hemorrhagic shock and inadequate resuscitation. Excess free fatty acids have shown to trigger inflammation in various disease conditions. C75 is a small compound that inhibits fatty acid synthase, a key enzyme in the control of fatty acid metabolism that also stimulates fatty acid oxidation. We hypothesized that C75 treatment would be protective against hemorrhagic shock. METHODS Adult male Sprague-Dawley rats were cannulated with a femoral artery catheter and subjected to controlled bleeding. Blood was shed to maintain a mean arterial pressure of 30 mm Hg for 90 minutes, then resuscitated over 30 minutes with a crystalloid volume equal to twice the volume of shed blood. Fifteen minutes into the 30-minute resuscitation, the rats received either intravenous infusion of C75 (1 mg/kg body weight) or vehicle (20% dimethyl sulfoxide). Blood and tissue samples were collected 6 hours after resuscitation (ie, 7.5 hours after hemorrhage) for analysis. RESULTS After hemorrhage and resuscitation, C75 treatment decreased the increase in serum free fatty acids by 48%, restored adenosine triphosphate levels, and stimulated carnitine palmitoyl transferase-1 activity. Administration of C75 decreased serum levels of markers of injury (aspartate aminotransferase, lactate, and lactate dehydrogenase) by 38%, 32%, and 78%, respectively. Serum creatinine and blood urea nitrogen were also decreased significantly by 38% and 40%, respectively. These changes correlated with decreases in neutrophil infiltration in the lung, evidenced by decreases in Gr-1-stained cells and myeloperoxidase activity and improved lung histology. Finally, administration of C75 decreased pulmonary mRNA levels of cyclooxygenase-2 and interleukin-6 by 87% and 65%, respectively. CONCLUSION Administration of C75 after hemorrhage and resuscitation decreased the increase in serum free fatty acids, decreased markers of tissue injury, downregulated the expression of inflammatory mediators, and decreased neutrophil infiltration and lung injury. Thus, the dual action of inhibiting fatty acid synthesis and stimulating fatty acid oxidation by C75 could be developed as a promising adjuvant therapy strategy to protect against hemorrhagic shock.
Collapse
Affiliation(s)
- Michael Kuncewitch
- Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, NY
| | - Weng Lang Yang
- Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, NY; Center for Translational Research, The Feinstein Institute for Medical Research, Manhasset, NY
| | - Asha Jacob
- Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, NY; Center for Translational Research, The Feinstein Institute for Medical Research, Manhasset, NY
| | - Adam Khader
- Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, NY
| | - Matthew Giangola
- Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, NY
| | - Jeff Nicastro
- Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, NY
| | - Gene F Coppa
- Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, NY
| | - Ping Wang
- Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, NY; Center for Translational Research, The Feinstein Institute for Medical Research, Manhasset, NY.
| |
Collapse
|
22
|
Sari EK, Bakir B, Aydin BD, Sozmen M. The effects of kefir, koumiss, yogurt and commercial probiotic formulations on PPARα and PPAR-β/δ expressions in mouse kidney. Biotech Histochem 2013; 89:287-95. [DOI: 10.3109/10520295.2013.844274] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
23
|
Arany I, Clark JS, Reed DK, Juncos LA, Dixit M. Role of p66shc in renal toxicity of oleic acid. Am J Nephrol 2013; 38:226-32. [PMID: 23988748 DOI: 10.1159/000354357] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 07/11/2013] [Indexed: 01/09/2023]
Abstract
BACKGROUND/AIMS Adult and childhood obesity is an independent risk factor in development of chronic kidney disease (CKD) and its progression to end-stage kidney disease. Pathologic consequences of obesity include non-esterified fatty acid-induced oxidative stress and consequent injury. Since the serine36-phosphorylated p66shc is a newly recognized mediator of oxidative stress and kidney injury, we studied its role in oleic acid (OA)-induced production of reactive oxygen species (ROS), mitochondrial depolarization and injury in cultured renal proximal tubule cells. METHODS Renal proximal tubule cells were used and treated with OA: ROS production, mitochondrial depolarization as well as injury were determined. Transcriptional effects of OA on the p66shc gene were determined in a reporter luciferase assay. The role of p66shc in adverse effects of OA was determined using knockdown, p66shc serine36 phosphorylation and cytochrome c binding-deficient cells. RESULTS We found that OA increased ROS production via the mitochondria - and to a less extent via the NADPH oxidase - resulting in ROS-dependent mitochondrial depolarization and consequent injury. Interestingly, OA also stimulated the promoter of p66shc. Hence, knockdown of p66shc, impairment its Ser36 phosphorylation (mutation of Ser36 residue to alanine) or cytochrome c binding (W134F mutation) significantly attenuated OA-dependent lipotoxicity. CONCLUSION These results offer a novel mechanism by which obesity may lead to renal tubular injury and consequently development of CKD. Manipulation of this pathway may offer therapeutic means to ameliorate obesity-dependent renal lipotoxicity.
Collapse
Affiliation(s)
- Istvan Arany
- Division of Pediatric Nephrology, Department of Pediatrics, University of Mississippi Medical Center, Jackson, MO 39110, USA.
| | | | | | | | | |
Collapse
|
24
|
Li S, Mariappan N, Megyesi J, Shank B, Kannan K, Theus S, Price PM, Duffield JS, Portilla D. Proximal tubule PPARα attenuates renal fibrosis and inflammation caused by unilateral ureteral obstruction. Am J Physiol Renal Physiol 2013; 305:F618-27. [PMID: 23804447 DOI: 10.1152/ajprenal.00309.2013] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
We examined the effects of increased expression of proximal tubule peroxisome proliferator-activated receptor (PPAR)α in a mouse model of renal fibrosis. After 5 days of unilateral ureteral obstruction (UUO), PPARα expression was significantly reduced in kidney tissue of wild-type mice but this downregulation was attenuated in proximal tubules of PPARα transgenic (Tg) mice. When compared with wild-type mice subjected to UUO, PPARα Tg mice had reduced mRNA and protein expression of proximal tubule transforming growth factor (TGF)-β1, with reduced production of extracellular matrix proteins including collagen 1, fibronectin, α-smooth muscle actin, and reduced tubulointerstitial fibrosis. UUO-mediated increased expression of microRNA 21 in kidney tissue was also reduced in PPARα Tg mice. Overexpression of PPARα in cultured proximal tubular cells by adenoviral transduction reduced aristolochic acid-mediated increased production of TGF-β, demonstrating PPARα signaling reduces epithelial TGF-β production. Flow cytometry studies of dissociated whole kidneys demonstrated reduced macrophage infiltration to kidney tissue in PPARα Tg mice after UUO. Increased expression of proinflammatory cytokines including IL-1β, IL-6, and TNF-α in wild-type mice was also significantly reduced in kidney tissue of PPARα Tg mice. In contrast, the expression of anti-inflammatory cytokines IL-10 and arginase-1 was significantly increased in kidney tissue of PPARα Tg mice when compared with wild-type mice subjected to UUO. Our studies demonstrate several mechanisms by which preserved expression of proximal tubule PPARα reduces tubulointerstitial fibrosis and inflammation associated with obstructive uropathy.
Collapse
Affiliation(s)
- Shenyang Li
- Division of Nephrology, Univ. of Arkansas for Medical Sciences, 4301 West Markham St., Slot 501, Little Rock, AR 72205, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Chau BN, Xin C, Hartner J, Ren S, Castano AP, Linn G, Li J, Tran PT, Kaimal V, Huang X, Chang AN, Li S, Kalra A, Grafals M, Portilla D, MacKenna DA, Orkin SH, Duffield JS. MicroRNA-21 promotes fibrosis of the kidney by silencing metabolic pathways. Sci Transl Med 2012; 4:121ra18. [PMID: 22344686 DOI: 10.1126/scitranslmed.3003205] [Citation(s) in RCA: 417] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Scarring of the kidney is a major public health concern, directly promoting loss of kidney function. To understand the role of microRNA (miRNA) in the progression of kidney scarring in response to injury, we investigated changes in miRNA expression in two kidney fibrosis models and identified 24 commonly up-regulated miRNAs. Among them, miR-21 was highly elevated in both animal models and in human transplanted kidneys with nephropathy. Deletion of miR-21 in mice resulted in no overt abnormality. However, miR-21(-/-) mice suffered far less interstitial fibrosis in response to kidney injury, a phenotype duplicated in wild-type mice treated with anti-miR-21 oligonucleotides. Global derepression of miR-21 target mRNAs was readily detectable in miR-21(-/-) kidneys after injury. Analysis of gene expression profiles up-regulated in the absence of miR-21 identified groups of genes involved in metabolic pathways, including the lipid metabolism pathway regulated by peroxisome proliferator-activated receptor-α (Pparα), a direct miR-21 target. Overexpression of Pparα prevented ureteral obstruction-induced injury and fibrosis. Pparα deficiency abrogated the antifibrotic effect of anti-miR-21 oligonucleotides. miR-21 also regulated the redox metabolic pathway. The mitochondrial inhibitor of reactive oxygen species generation Mpv17l was repressed by miR-21, correlating closely with enhanced oxidative kidney damage. These studies demonstrate that miR-21 contributes to fibrogenesis and epithelial injury in the kidney in two mouse models and is a candidate target for antifibrotic therapies.
Collapse
|
26
|
Li S, Nagothu K, Ranganathan G, Ali SM, Shank B, Gokden N, Ayyadevara S, Megyesi J, Olivecrona G, Chugh SS, Kersten S, Portilla D. Reduced kidney lipoprotein lipase and renal tubule triglyceride accumulation in cisplatin-mediated acute kidney injury. Am J Physiol Renal Physiol 2012; 303:F437-48. [PMID: 22622461 DOI: 10.1152/ajprenal.00111.2012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Peroxisome proliferator-activated receptor-α (PPARα) activation attenuates cisplatin (CP)-mediated acute kidney injury by increasing fatty acid oxidation, but mechanisms leading to reduced renal triglyceride (TG) accumulation could also contribute. Here, we investigated the effects of PPARα and CP on expression and enzyme activity of kidney lipoprotein lipase (LPL) as well as on expression of angiopoietin protein-like 4 (Angptl4), glycosylphosphatidylinositol-anchored-HDL-binding protein (GPIHBP1), and lipase maturation factor 1 (Lmf1), which are recognized as important proteins that modulate LPL activity. CP caused a 40% reduction in epididymal white adipose tissue (WAT) mass, with a reduction of LPL expression and activity. CP also reduced kidney LPL expression and activity. Angptl4 mRNA levels were increased by ninefold in liver and kidney tissue and by twofold in adipose tissue of CP-treated mice. Western blots of two-dimensional gel electrophoresis identified increased expression of a neutral pI Angptl4 protein in kidney tissue of CP-treated mice. Immunolocalization studies showed reduced staining of LPL and increased staining of Angptl4 primarily in proximal tubules of CP-treated mice. CP also increased TG accumulation in kidney tissue, which was ameliorated by PPARα ligand. In summary, a PPARα ligand ameliorates CP-mediated nephrotoxicity by increasing LPL activity via increased expression of GPHBP1 and Lmf1 and by reducing expression of Angptl4 protein in the proximal tubule.
Collapse
Affiliation(s)
- Shenyang Li
- Division of Nephrology, Department of Internal Medicine, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
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: 1.5] [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
|
28
|
Post-treatment with the combination of 5-aminoimidazole-4-carboxyamide ribonucleoside and carnitine improves renal function after ischemia/reperfusion injury. Shock 2012; 37:39-46. [PMID: 21841537 DOI: 10.1097/shk.0b013e31823185d7] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Renal ischemia/reperfusion (I/R) injury is a major clinical problem where main metabolic pathways are compromised and cellular homeostasis crashes after ATP depletion. Fatty acids are major energy source in the kidneys. Carnitine palmitoyltransferase I (CPT1), a mitochondrial membrane enzyme, utilizes carnitine to transport fatty acids to mitochondria for the process of β-oxidation and ATP generation. In addition, CPT1 activity is indirectly regulated by adenosine monophosphate-activated protein kinase, which can be activated by 5-aminoimidazole-4-carboxyamide ribonucleoside (AICAR). We hypothesized that administration of carnitine and AICAR could reestablish the energetic balance after reperfusion and ameliorate renal I/R injury. Male adult rats were subjected to renal I/R by bilateral renal pedicle clamping for 60 min, followed by administration of saline (vehicle), carnitine (250 mg/kg BW), AICAR (30 mg/kg BW), or combination of both drugs. Blood and renal tissues were collected 24 h after reperfusion for various measurements. Renal carnitine levels decreased 53% after I/R. The combined treatment significantly increased CPT1 activity and ATP levels and lowered renal malondialdehyde and serum TNF-α levels against the vehicle group. It led to improvement in renal morphology and histological damage score associated with diminution in serum creatinine, blood urea nitrogen, and aspartate aminotransferase levels. Moreover, the combined treatment significantly improved the survival rate in comparison to the vehicle group. In contrast, administration of either drug alone did not show a significant improvement in most of the measurements. In conclusion, enhancing energy metabolism by combination of carnitine and AICAR provides a novel modality to treat renal I/R injury.
Collapse
|
29
|
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: 3.6] [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
|
30
|
Role of PPARα and Its Agonist in Renal Diseases. PPAR Res 2010; 2010:345098. [PMID: 21076544 PMCID: PMC2976496 DOI: 10.1155/2010/345098] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Accepted: 10/17/2010] [Indexed: 01/08/2023] Open
Abstract
Peroxisome proliferator-activated receptor (PPAR)-α, a member of a large nuclear receptor superfamily, plays a major role in the regulation of lipid metabolism. Recently, PPARα activation has been shown to confer additional benefits on endothelial function, kidney function, and anti-inflammation, suggesting that PPARα agonists may be good candidates for treating acute renal failure. In clinical application, PPAR-α activators, such as hypolipidemic drugs in fibric acid class, were proven to have therapeutic effects on metabolic syndrome and cardiovascular disease. This paper focuses on signaling pathways, ligand selectivity, and physio-pathological roles of PPARα in kidney diseases and the therapeutic utility of PPARα modulators in the treatment of diabetes and inflammation-induced nephropathy. Implication of new and more potent PPAR-α activators could provide important insights into the overall benefits of activating PPAR-α clinically for the treatment of dyslipidemia and the prevention of diabetic or inflammation-induced nephropathy in the future.
Collapse
|
31
|
Hiukka A, Maranghi M, Matikainen N, Taskinen MR. PPARalpha: an emerging therapeutic target in diabetic microvascular damage. Nat Rev Endocrinol 2010; 6:454-63. [PMID: 20567246 DOI: 10.1038/nrendo.2010.89] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The global pandemic of diabetes mellitus portends an alarming rise in the prevalence of microvascular complications, despite advanced therapies for hyperglycemia, hypertension and dyslipidemia. Peroxisome proliferator-activated receptor alpha (PPARalpha) is expressed in organs affected by diabetic microvascular disease (retina, kidney and nerves), and its expression is regulated specifically in these tissues. Experimental evidence suggests that PPARalpha activation attenuates or inhibits several mediators of vascular damage, including lipotoxicity, inflammation, reactive oxygen species generation, endothelial dysfunction, angiogenesis and thrombosis, and thus might influence intracellular signaling pathways that lead to microvascular complications. PPARalpha has emerged as a novel target to prevent microvascular disease, via both its lipid-related and lipid-unrelated actions. Despite strong experimental evidence of the potential benefits of PPARalpha agonists in the prevention of vascular damage, the evidence from clinical studies in patients with diabetes mellitus remains limited. Promising findings from the Fenofibrate Intervention and Event Lowering in Diabetes (FIELD) study on microvascular outcomes are countered by elevations in participants' homocysteine and creatinine levels that might potentially attenuate the benefits of PPARalpha activation. This Review focuses on the role of PPARalpha activation in diabetic microvascular disease and highlights the available experimental and clinical evidence from studies of PPARalpha agonists.
Collapse
Affiliation(s)
- Anne Hiukka
- Division of Cardiology, Department of Medicine, Helsinki University Central Hospital and Biomedicum, Haartmaninkatu 8, 00029 Helsinki, Finland
| | | | | | | |
Collapse
|
32
|
Lin S, Chan W, Li J, Cai Z. Liquid chromatography/mass spectrometry for investigating the biochemical effects induced by aristolochic acid in rats: the plasma metabolome. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2010; 24:1312-1318. [PMID: 20391603 DOI: 10.1002/rcm.4516] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
In this study, high-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry in conjunction with chemometric methods including principal components analysis was used to investigate metabolic profiling in plasma samples from rats dosed with aristolochic acid (AA). Differentiating metabolites were identified by high-resolution mass spectrometry and tandem mass spectrometry analyses, database searching and comparison with the analytical results of authentic standards. Several metabolites in plasma including a glucuronide conjugate, bile acids, lysophosphatidylcholines and fatty acids were detected in rats exposed to AA. To improve visualization, a z-score plot and a cluster heat map were generated for the concentration fluctuations of the metabolites in different dosage groups. The results obtained from this study indicated that class-specific metabolomic patterns were obviously differentiated for each metabolite in the different dosage groups of AA.
Collapse
Affiliation(s)
- Shuhai Lin
- Department of Chemistry, Hong Kong Baptist University, Hong Kong SAR, China
| | | | | | | |
Collapse
|
33
|
Li S, Nagothu KK, Desai V, Lee T, Branham W, Moland C, Megyesi JK, Crew MD, Portilla D. Transgenic expression of proximal tubule peroxisome proliferator-activated receptor-alpha in mice confers protection during acute kidney injury. Kidney Int 2009; 76:1049-62. [PMID: 19710628 DOI: 10.1038/ki.2009.330] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Our previous studies suggest that peroxisome proliferator-activated receptor-alpha (PPARalpha) plays a critical role in regulating fatty acid beta-oxidation in kidney tissue and this directly correlated with preservation of kidney morphology and function during acute kidney injury. To further study this, we generated transgenic mice expressing PPARalpha in the proximal tubule under the control of the promoter of KAP2 (kidney androgen-regulated protein 2). Segment-specific upregulation of PPARalpha expression by testosterone treatment of female transgenic mice improved kidney function during cisplatin or ischemia-reperfusion-induced acute kidney injury. Ischemia-reperfusion injury or treatment with cisplatin in wild-type mice caused inhibition of fatty-acid oxidation, reduction of mitochondrial genes of oxidative phosphorylation, mitochondrial DNA, fatty-acid metabolism, and the tricarboxylic acid cycle. Similar injury in testosterone-treated transgenic mice resulted in amelioration of these effects. Similarly, there were increases in the levels of 4-hydroxy-2-hexenal-derived lipid peroxidation products in wild-type mice, which were also reduced in the transgenic mice. Similarly, necrosis of the S3 segment was reduced in the two injury models in transgenic mice compared to wild type. Our results suggest proximal tubule PPARalpha activity serves as a metabolic sensor. Its increased expression without the use of an exogenous PPARalpha ligand in the transgenic mice is sufficient to protect kidney function and morphology, and to prevent abnormalities in lipid metabolism associated with acute kidney injury.
Collapse
Affiliation(s)
- Shenyang Li
- Division of Nephrology, Departments of Internal Medicine and Immunology, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, Arkansas 72205, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Peroxisome proliferator-activated receptors in diabetic nephropathy. PPAR Res 2009; 2008:879523. [PMID: 19277201 PMCID: PMC2652581 DOI: 10.1155/2008/879523] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2008] [Accepted: 12/08/2008] [Indexed: 02/08/2023] Open
Abstract
Diabetic nephropathy is a leading cause of end-stage renal disease, which is increasing in incidence worldwide, despite intensive treatment approaches such as glycemic and blood pressure control in patients with diabetes mellitus. New therapeutic strategies are needed to prevent the onset of diabetic nephropathy. Peroxisome proliferator-activated receptors (PPARs) are ligand-activated nuclear transcription factors that play important roles in lipid and glucose homeostases. These agents might prevent the progression of diabetic nephropathy, since PPAR agonists improve dyslipidemia and insulin resistance. Furthermore, data from murine models suggest that PPAR agonists also have independent renoprotective effects by suppressing inflammation, oxidative stress, lipotoxicity, and activation of the renin-angiotensin system. This review summarizes data from clinical and experimental studies regarding the relationship between PPARs and diabetic nephropathy. The therapeutic potential of PPAR agonists in the treatment of diabetic nephropathy is also discussed.
Collapse
|
35
|
Arafa HMM. Carnitine deficiency aggravates carboplatin nephropathy through deterioration of energy status, oxidant/anti-oxidant balance, and inflammatory endocoids. Toxicology 2008; 254:51-60. [PMID: 18852009 DOI: 10.1016/j.tox.2008.09.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2008] [Revised: 09/04/2008] [Accepted: 09/05/2008] [Indexed: 12/30/2022]
Abstract
We have recently shown that carnitine deficiency could represent a risk factor in paracetamol hepatotoxicity. By the same token, d-carnitine-induced carnitine deficiency aggravated carboplatin nephropathy following challenge with a single dose (35mg/kg, IP) of the platinum drug in male Swiss albino rats. The combination modality induced marked degenerative changes and severe inflammation in kidney tissues that surpassed either carboplatin or d-carnitine given alone. The combined regimen synergistically increased the serum levels of creatinine, blood urea nitrogen (BUN), tumor necrosis factor alpha (TNF-alpha), palmitate, and kidney malondialdehyde (MDA), adenosine triphosphate (ATP), nitric oxide (NO) contents as well as kidney myeloperoxidase (MPO) activity. The only parameter that has been notably decreased was the kidney reduced glutathione (GSH) level. Exaggeration by carnitine deficit of the deleterious effects of carboplatin is most probably ascribed to energy starvation. The reduction in kidney content of ATP parcels was associated with elevation of serum palmitate level that reflected debilitated fatty acid oxidation, and this further deteriorated energy resources in kidney tissues. Compromising the oxidant/anti-oxidant balance and modulating the release of some inflammatory endocoids namely, TNF-alpha and NO could also possibly account for such combinatorial detrimental toxicity. The current study was further extended to elucidate any possible nephroprotective effects of l-carnitine. Interestingly, carnitine supplementation ahead of carboplatin challenge ameliorated and almost normalized all the biochemical parameters and also mitigated the injurious effects of the cytotoxic drug. Thus, one could conclude that carnitine deficiency, whether being a causative clue or a sequela, might represent a risk factor in carboplatin nephropathy.
Collapse
Affiliation(s)
- Hossam M M Arafa
- Department of Pharmacol & Toxicol, Faculty of Pharmacy, Al-Azhar University, Nasr City, Cairo, Egypt.
| |
Collapse
|
36
|
Obih P, Oyekan AO. Regulation of blood pressure, natriuresis and renal thiazide/amiloride sensitivity in PPARalpha null mice. Blood Press 2008; 17:55-63. [PMID: 18568693 DOI: 10.1080/08037050701789278] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
This study evaluated the role of PPARalpha in renal function and whether PPARalpha knockout (KO) mice are hypertensive or salt-sensitive. We hypothesize that PPARalpha modulation of ion transport defines the capacity for sodium excretion (U(Na)V). PPARalpha KO and wild-type (WT) mice were placed on a normal salt (NS, 0.5% NaCl) or high salt (8% NaCl, HS) diet for 28 days and mean arterial blood pressure (MABP) and heart rate (HR) determined. In a group of anesthetized animals on NS diet, pressure natriuresis (P/N) was determined and in another group, acute sodium load (0.9% NaCl) was administered and U(Na)V compared in mice pretreated with amiloride (200 microg/kg) or hydrochlorothiazide (3 mg/kg), in vivo measurements of sodium hydrogen exchanger or Na-Cl-cotransporter activity, respectively. MABP and HR were similar in PPARalpha KO and WT mice placed on a NS diet (116+/-6 mmHg, 587+/-40 beats/min, KO; 116+/-4 mmHg, 551+/-20 beats/min, WT). HS diet increased MABP to a greater extent in KO mice (Delta = 29+/-3 vs 14+/-3 mmHg, p<0.05) as did proteinuria (8- vs 2.5-fold, p<0.05). P/N was blunted in untreated KO mice. In response to an acute NaCl-load, U(Na)V was faster in PPARalpha KO mice (4.31+/-1.11 vs 0.77+/-0.31 micromol, p<0.05). However, U(Na)V was unchanged in hydrochlorothiazide-treated KO mice but increased 6.9-fold in WT mice. Similarly, U(Na)V was less in amiloride-treated KO mice (3.4- vs 15.5-fold). These data suggest that PPARalpha participates in pressure natriuresis and affects Na transport via amiloride- and thiazide-sensitive mechanisms. Thus, despite defective fatty acid oxidation, PPARalpha null mice are not hypertensive but develop salt-sensitive hypertension.
Collapse
Affiliation(s)
- Patience Obih
- College of Pharmacy, Xavier University of Louisiana, New Orleans, Louisiana, USA
| | | |
Collapse
|
37
|
Renal L-type fatty acid-binding protein mediates the bezafibrate reduction of cisplatin-induced acute kidney injury. Kidney Int 2008; 73:1374-84. [PMID: 18368030 DOI: 10.1038/ki.2008.106] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Fibrates, the PPAR alpha ligand-like compounds increase the expression of proximal tubule liver fatty acid binding protein (L-FABP) and significantly decrease cisplatin-induced acute kidney injury. To study whether the bezafibrate-mediated upregulation of renal L-FABP was involved in this cytoprotective effect we treated transgenic mice of PPAR agonists inducible human L-FABP expression with cisplatin in the presence or absence of bezafibrate. Blood urea nitrogen was unchanged in the first day but increased 3 days after cisplatin. While urinary L-FABP increased over 100-fold 1 day after cisplatin treatment in the transgenic mice it was significantly reduced when these transgenic mice were pretreated with bezafibrate. Cisplatin-induced renal necrosis and apoptosis were significantly reduced in bezafibrate pretreated transgenic mice and this correlated with decreased accumulation of lipid and lipid peroxidation products. Immunohistochemical analysis of kidney tissue of bezafibrate-cisplatin-treated transgenic mice showed preservation of cytoplasmic L-FABP in the proximal tubule, but this was reduced in transgenic mice treated only with cisplatin. L-FABP mRNA and protein levels were significantly increased in bezafibrate-cisplatin-treated transgenic mice when compared to mice not fibrate treated. Our study shows that the bezafibrate-mediated upregulation of proximal tubule L-FABP plays a pivotal role in the reduction of cisplatin-induced acute kidney injury.
Collapse
|
38
|
Hsu YH, Chen CH, Hou CC, Sue YM, Cheng CY, Cheng TH, Lin H, Tsai WL, Chan P, Chen TH. Prostacyclin protects renal tubular cells from gentamicin-induced apoptosis via a PPARα-dependent pathway. Kidney Int 2008; 73:578-87. [DOI: 10.1038/sj.ki.5002704] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
39
|
Ruan X, Zheng F, Guan Y. PPARs and the kidney in metabolic syndrome. Am J Physiol Renal Physiol 2008; 294:F1032-47. [PMID: 18234957 DOI: 10.1152/ajprenal.00152.2007] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The metabolic syndrome (MetS) is defined by a set of metabolic risk factors, including insulin resistance, central obesity, dyslipidemia, hyperglycemia, and hypertension for type 2 diabetes and cardiovascular disease. Although both retrospective and prospective clinical studies have revealed that MetS is associated with chronic renal disease, even with a nondiabetic cause, the cellular and molecular mechanisms in this association remain largely uncharacterized. Recently, increasing evidence suggests that peroxisome proliferator-activated receptors (PPARs), a subgroup of the nuclear hormone receptor superfamily of ligand-activated transcription factors, may play an important role in the pathogenesis of MetS. All three members of the PPAR nuclear receptor subfamily, PPARalpha, -beta/delta, and -gamma, are critical in regulating insulin sensitivity, adipogenesis, lipid metabolism, inflammation, and blood pressure. PPARs have also been implicated in many renal pathophysiological conditions, including diabetic nephropathy and glomerulosclerosis. Ligands for PPARs such as hypolipidemic PPARalpha activators, and antidiabetic thiazolidinedione PPARgamma agonists affect not only diverse aspects of MetS but also renal disease progression. Emerging data suggest that PPARs may be potential therapeutic targets for MetS and its related renal complications. This review focuses on current knowledge of the role of PPARs in MetS and discusses the potential therapeutic utility of PPAR modulators in the treatment of kidney diseases associated with MetS.
Collapse
Affiliation(s)
- Xiongzhong Ruan
- Center for Nephrology, University College of London, London, United Kingdom
| | | | | |
Collapse
|
40
|
Yamamoto T, Noiri E, Ono Y, Doi K, Negishi K, Kamijo A, Kimura K, Fujita T, Kinukawa T, Taniguchi H, Nakamura K, Goto M, Shinozaki N, Ohshima S, Sugaya T. Renal L-type fatty acid--binding protein in acute ischemic injury. J Am Soc Nephrol 2007; 18:2894-902. [PMID: 17942962 DOI: 10.1681/asn.2007010097] [Citation(s) in RCA: 260] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Fatty acid-binding proteins (FABPs) bind unsaturated fatty acids and lipid peroxidation products during tissue injury from hypoxia. We evaluated the potential role of L-type FABP (L-FABP) as a biomarker of renal ischemia in both human kidney transplant patients and animal models. Urinary L-FABP levels were measured in the first urine produced from 12 living-related kidney transplant patients immediately after reperfusion of their transplanted organs, and intravital video analysis of peritubular capillary blood flow was performed simultaneously. A significant direct correlation was found between urinary L-FABP level and both peritubular capillary blood flow and the ischemic time of the transplanted kidney (both P < 0.0001), as well as hospital stay (P < 0.05). In human-L-FABP transgenic mice subjected to ischemia-reperfusion injury, immunohistological analyses demonstrated the transition of L-FABP from the cytoplasm of proximal tubular cells to the tubular lumen. In addition, after injury, these transgenic mice demonstrated lower blood urea nitrogen levels and less histological injury than injured wild-type mice, likely due to a reduction of tissue hypoxia. In vitro experiments using a stable cell line of mouse proximal tubule cells transfected with h-L-FABP cDNA showed reduction of oxidative stress during hypoxia compared to untransfected cells. Taken together, these data show that increased urinary L-FABP after ischemic-reperfusion injury may find future use as a biomarker of acute ischemic injury.
Collapse
Affiliation(s)
- Tokunori Yamamoto
- Department of Urology, University Hospital, Nagoya University, Nagoya, Japan 113-8655
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Negishi K, Noiri E, Sugaya T, Li S, Megyesi J, Nagothu K, Portilla D. A role of liver fatty acid-binding protein in cisplatin-induced acute renal failure. Kidney Int 2007; 72:348-58. [PMID: 17495861 DOI: 10.1038/sj.ki.5002304] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Previous studies from our laboratory showed that increased fatty acid oxidation by the kidney is cytoprotective during cisplatin (CP)-mediated nephrotoxicity. In this study, we determined the effects of CP and fibrates on peroxisome proliferation and the expression of liver fatty acid-binding protein (L-FABP) in normal mice, and in mice transgenically overexpressing human L-FABP (h-L-FABP). Labeling of peroxisomes demonstrated reduced peroxisomal staining in the proximal tubule of CP-treated mice compared with control mice. There was increased peroxisomal labeling in the proximal tubules of both control and CP-treated mice when either was treated with fibrate; a known peroxisome proliferator-activated receptor-alpha ligand. L-FABP protein expression, not detected in control or CP-treated mice, was significantly increased in the proximal tubules of fibrate-treated mice of either group. In the transgenic mice, CP increased the shedding of h-L-FABP in the urine, which was decreased by fibrate as was the acute renal failure. A cytosolic pattern of h-L-FABP expression was found in the proximal tubules of untreated transgenic mice with a nuclear presence in CP-treated mice. Fibrate pretreatment restored the cytosolic expression pattern in CP-treated mice. Our study shows that fibrate may improve CP-induced acute renal failure due to both peroxisome proliferation and increased L-FABP in the cytosol of the proximal tubule.
Collapse
Affiliation(s)
- K Negishi
- Department of Nephrology and Endocrinology, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
42
|
Lu H, Lei X, Klaassen C. Gender differences in renal nuclear receptors and aryl hydrocarbon receptor in 5/6 nephrectomized rats. Kidney Int 2006; 70:1920-8. [PMID: 16985511 DOI: 10.1038/sj.ki.5001880] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
This study was aimed at delineating molecular pathways essential in gender-different pathogenesis of chronic kidney diseases (CKD). Renal transcripts of nuclear receptors and metabolic enzymes in male and female kidneys from 5/6 nephrectomized (Nx) rats 7 weeks post-Nx were examined using branched DNA signal amplification assay. Nx-males had marked kidney injury coupled with anemia and malnutrition. Nx-females had moderate renal injury, and were free of albuminuria, anemia, and malnutrition. Nx-males had systemic and renal inflammation, which were largely absent in Nx-females. Blood 17beta-estradiol, testosterone, and corticosterone did not change, whereas urinary testosterone decreased in both genders. Compared to males, female kidneys had higher androgen receptor (AR) and aryl hydrocarbon receptor (AhR) but lower estrogen receptor alpha (ERalpha). Compared to Nx-males, female remnant kidneys had less decreases in ERalpha and peroxisome proliferator-activated receptor alpha (PPARalpha), had no induction of AR and decrease of acyl-CoA oxidase, whereas had induction of cytochrome P450 4a1 (Cyp4a1) but decrease of AhR. Renal protein expression of a 52-kDa isoform of Wilm's tumor 1 (WT1), transcription factor critical in nephrogenesis, decreased dramatically in Nx-males but largely preserved in Nx-females. In conclusion, gender divergences in basal expression and alteration of ERalpha, AR, AhR, WT1, and PPARalpha/Cyp4a1 during CKD may explain gender differences in CKD progression and outcome of renal transplantation.
Collapse
Affiliation(s)
- H Lu
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160-7417, USA
| | | | | |
Collapse
|
43
|
Portilla D, Li S, Nagothu KK, Megyesi J, Kaissling B, Schnackenberg L, Safirstein RL, Beger RD. Metabolomic study of cisplatin-induced nephrotoxicity. Kidney Int 2006; 69:2194-204. [PMID: 16672910 DOI: 10.1038/sj.ki.5000433] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We have shown that cisplatin inhibits fatty acid oxidation, and that fibrate treatment ameliorates renal function by preventing the inhibition of fatty acid oxidation and proximal tubule cell death. Urine samples of mice treated with single injection of cisplatin (20 mg/kg body weight) were collected for 3 days and analyzed by 1H-nuclear magnetic resonance (NMR) spectroscopy. In a separate group, urine samples of mice treated with peroxisome proliferator-activated receptor-alpha (PPARalpha) ligand WY were also analyzed by NMR after 2 days of cisplatin exposure. Biochemical analysis of endogenous metabolites was performed in serum, urine, and kidney tissue. Electron microscopic studies were carried out to examine the effects of PPARalpha ligand and cisplatin. Principal component analysis demonstrated the presence of glucose, amino acids, and trichloacetic acid cycle metabolites in the urine after 48 h of cisplatin administration. These metabolic alterations precede changes in serum creatinine. Biochemical studies confirmed the presence of glucosuria, but also demonstrated the accumulation of nonesterified fatty acids, and triglycerides in serum, urine, and kidney tissue, in spite of increased levels of plasma insulin. These metabolic alterations were ameliorated by the use of PPARalpha ligand. Electron microscopic analysis confirmed the protective effect of the fibrate on preventing cisplatin-mediated necrosis of the S3 segment of the proximal tubule. Our study shows that cisplatin-induces a unique NMR metabolic profile in urine of mice that developed acute renal failure, and confirms the protective effect of a fibrate class of PPARalpha ligands. We propose that the injury-induced metabolic profile may be used as a biomarker of cisplatin-induced nephrotoxicity.
Collapse
Affiliation(s)
- D Portilla
- Department of Internal Medicine, Division of Nephrology, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, Arkansas 72205, USA.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
BACKGROUND In previous studies we have shown that cisplatin inhibits peroxisome proliferator-activated receptor-alpha (PPAR-alpha) activity and consequently fatty acid oxidation, and these events precede proximal tubule cell death. In addition the use of fibrate class of PPAR-alpha ligands ameliorate renal function by preventing both inhibition of fatty acid oxidation and proximal tubule cell death. METHODS LLC-PK1 cells were treated with cisplatin and apoptosis was established by the presence of nuclear fragmentation and by cell cycle analysis. Proximal tubular cells treated with cisplatin and bezafibrate were subjected to sub cellular fractionation and the presence of Bax, Bcl-2, cytochrome c, and active caspase-3 in the cytosolic and mitochondrial membrane fractions was determined by Western blot analysis. PPAR-alpha activity was measured by determining luciferase activity after transfection of LLC-PK1 cells with TK-Luc 3x PPAR response elements (PPRE), and the accumulation of nonesterified free fatty acids was measured in lysates obtained from cells treated with cisplatin and bezafibrate. RESULTS Incubation of LLC-PK1 cells with 25 micromol/L cisplatin for 18 hours induced 41.5% apoptosis measured by cell cycle analysis. Cisplatin-induced apoptosis was significantly suppressed by bezafibrate, a fibrate class of PPAR-alpha ligand. Bezafibrate treatment of LLC-PK1 cells prevented cisplatin-induced translocation of proapoptotic Bax from the cytosol to the mitochondrial fraction, and increased the expression of antiapoptotic molecule Bcl-2. Cisplatin-induced inhibition of PPAR-alpha activity was accompanied by increased accumulation of nonesterified free fatty acids. Pretreatment with bezafibrate prevented both the inhibition of PPAR-alpha activity and the accumulation of nonesterified free fatty acids induced by cisplatin. Finally, bezafibrate prevented cisplatin-induced release of cytochrome c from the mitochondria to the cytosol, and the cleavage of procaspase-3 to active caspase-3. CONCLUSION Bezafibrate treatment inhibits cisplatin-mediated tubular injury by preventing the activation of various cellular mechanisms that lead to proximal tubule cell death. These findings support our previous observations where the use of fibrates represents a novel strategy to ameliorate proximal tubule cell death in cisplatin-induced acute renal failure.
Collapse
Affiliation(s)
- Kiran K Nagothu
- Division of Nephrology, Department of Internal Medicine, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA
| | | | | | | |
Collapse
|
45
|
|
46
|
Feldkamp T, Kribben A, Roeser NF, Senter RA, Weinberg JM. Accumulation of nonesterified fatty acids causes the sustained energetic deficit in kidney proximal tubules after hypoxia-reoxygenation. Am J Physiol Renal Physiol 2005; 290:F465-77. [PMID: 16159894 DOI: 10.1152/ajprenal.00305.2005] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Kidney proximal tubules exhibit decreased ATP and reduced, but not absent, mitochondrial membrane potential (Deltapsi(m)) during reoxygenation after severe hypoxia. This energetic deficit, which plays a pivotal role in overall cellular recovery, cannot be explained by loss of mitochondrial membrane integrity, decreased electron transport, or compromised F1F0-ATPase and adenine nucleotide translocase activities. Addition of oleate to permeabilized tubules produced concentration-dependent decreases of Deltapsi(m) measured by safranin O uptake (threshold for oleate = 0.25 microM, 1.6 nmol/mg protein; maximal effect = 4 microM, 26 nmol/mg) that were reversed by delipidated BSA (dBSA). Cell nonesterified fatty acid (NEFA) levels increased from <1 to 17.4 nmol/mg protein during 60- min hypoxia and remained elevated at 7.6 nmol/mg after 60 min reoxygenation, at which time ATP had recovered to only 10% of control values. Safranin O uptake in reoxygenated tubules, which was decreased 85% after 60-min hypoxia, was normalized by dBSA, which improved ATP synthesis as well. dBSA also almost completely normalized Deltapsi(m) when the duration of hypoxia was increased to 120 min. In intact tubules, the protective substrate combination of alpha-ketoglutarate + malate (alpha-KG/MAL) increased ATP three- to fourfold, limited NEFA accumulation during hypoxia by 50%, and lowered NEFA during reoxygenation. Notably, dBSA also improved ATP recovery when added to intact tubules during reoxygenation and was additive to the effect of alpha-KG/MAL. We conclude that NEFA overload is the primary cause of energetic failure of reoxygenated proximal tubules and lowering NEFA substantially contributes to the benefit from supplementation with alpha-KG/MAL.
Collapse
Affiliation(s)
- Thorsten Feldkamp
- Nephrology Division, Department of Internal Medicine, Rm. 1560, MSRB II, University of Michigan Medical Center, Ann Arbor, MI 48109-0676, USA
| | | | | | | | | |
Collapse
|
47
|
Li S, Gokden N, Okusa MD, Bhatt R, Portilla D. Anti-inflammatory effect of fibrate protects from cisplatin-induced ARF. Am J Physiol Renal Physiol 2005; 289:F469-80. [PMID: 15814532 DOI: 10.1152/ajprenal.00038.2005] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Recently, we demonstrated that peroxisome proliferator-activated receptor-α (PPARα) ligand ameliorates cisplatin-induced acute renal failure (ARF) by preventing inhibition of substrate oxidation, and also by preventing apoptosis and necrosis of the proximal tubule (Li S, Bhatt R, Megyesi J, Gokden N, Shah SV, and Portilla D. Am J Physiol Renal Physiol 287: F990–F998, 2004). In the following studies, we examined the protective effect of PPARα ligand on cisplatin-induced inflammatory responses during ARF. Mice subjected to a single intraperitoneal injection of cisplatin developed ARF at day 3. Cisplatin increased mRNA and protein expression of TNF-α, RANTES, and also upregulated endothelial adhesion molecules ICAM-1/VCAM-1 and chemokine receptors CCR1/CCR5. Cisplatin also led to neutrophil infiltration in the corticomedullary region. Pretreatment of wild-type mice with WY-14,643, a fibrate class of PPARα ligands, before cisplatin significantly suppressed cisplatin-induced upregulation of cytokine/chemokine expression, prevented neutrophil accumulation, and ameliorated renal dysfunction. In contrast, treatment with PPARα ligand before cisplatin did not prevent cytokine/chemokine production, neutrophil accumulation, and did not protect kidney function in PPARα null mice. In addition, we observed that cisplatin-induced NF-κB binding activity in nuclear extracts from wild-type mice was markedly reduced by treatment with PPARα ligand. These results demonstrate that PPARα exerts an anti-inflammatory effect in kidney tissue by a mechanism that includes inhibition of NF-κB DNA binding activity, and this effect results in inhibition of neutrophil infiltration, cytokine/chemokine release, and amelioration of cisplatin-induced ARF.
Collapse
Affiliation(s)
- Shenyang Li
- Division of Nephrology, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, 72205, USA
| | | | | | | | | |
Collapse
|
48
|
Mount PF, Hill RE, Fraser SA, Levidiotis V, Katsis F, Kemp BE, Power DA. Acute renal ischemia rapidly activates the energy sensor AMPK but does not increase phosphorylation of eNOS-Ser1177. Am J Physiol Renal Physiol 2005; 289:F1103-15. [PMID: 15914772 DOI: 10.1152/ajprenal.00458.2004] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
A fundamental aspect of acute renal ischemia is energy depletion, manifest as a falling level of ATP that is associated with a simultaneous rise in AMP. The energy sensor AMP-activated protein kinase (AMPK) is activated by a rising AMP-to-ATP ratio, but its role in acute renal ischemia is unknown. AMPK is activated in the ischemic heart and is reported to phosphorylate both endothelial nitric oxide synthase (eNOS) and acetyl-CoA carboxylase. To study activation of AMPK in acute renal ischemia, the renal pedicle of anesthetized Sprague-Dawley rats was cross-clamped for increasing time intervals. AMPK was strongly activated within 1 min and remained so after 30 min. However, despite the robust activation of AMPK, acute renal ischemia did not increase phosphorylation of the AMPK phosphorylation sites eNOS-Ser(1177) or acetyl-CoA carboxylase-Ser(79). Activation of AMPK in bovine aortic endothelial cells by the ATP-depleting agent antimycin A and the antidiabetic drug phenformin also did not increase phosphorylation of eNOS-Ser(1177), confirming that AMPK activation and phosphorylation of eNOS are dissociated in some situations. Immunoprecipitation studies demonstrated that the dissociation between AMPK activation and phosphorylation of eNOS-Ser(1177) was not due to changes in the physical associations between AMPK, eNOS, or heat shock protein 90. In conclusion, acute renal ischemia rapidly activates the energy sensor AMPK, which is known to maintain ATP reserves during energy stress. The substrates it phosphorylates, however, are different from those in other organs such as the heart.
Collapse
Affiliation(s)
- Peter F Mount
- Austin Research Institute, Austin Health, University of Melbourne, Heidelberg 3084, Victoria, Australia.
| | | | | | | | | | | | | |
Collapse
|
49
|
Li S, Basnakian A, Bhatt R, Megyesi J, Gokden N, Shah SV, Portilla D. PPAR-alpha ligand ameliorates acute renal failure by reducing cisplatin-induced increased expression of renal endonuclease G. Am J Physiol Renal Physiol 2004; 287:F990-8. [PMID: 15280156 DOI: 10.1152/ajprenal.00206.2004] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Cisplatin injury to the kidney is characterized, in part, by inhibition of substrate oxidation, inflammation, and tubular cell death in the form of apoptosis and necrosis. Recently, we demonstrated that cisplatin-induced inhibition of substrate oxidation can be reversed by the administration of peroxisome proliferator-activated receptor-alpha (PPAR-alpha) ligands, resulting in amelioration of renal function. We therefore hypothesize that by improving fatty acid oxidation in vivo might protect renal function by reducing both apoptosis and necrosis in cisplatin-treated mice. Mice subjected to a single intraperitoneal injection of cisplatin developed acute renal failure (ARF) at days 3 and 4. At day 4 after cisplatin injection mRNA, protein levels and enzyme activity of proapoptotic renal endonuclease G (Endo G) were increased compared with saline-treated mice. In situ hybridization and immunohistochemical studies localized the increased expression of Endo G mRNA to the cytosolic compartment and Endo G protein to the nuclear compartment of proximal tubules in cisplatin-treated mice. Pretreatment of PPAR-alpha wild-type mice with PPAR-alpha ligand WY-14643 reduced significantly cisplatin-induced increased protein expression and enzyme activity of Endo G and prevented the nuclear translocation of mitochondrial Endo G. Morphological examination of tubular injury in the PPAR-alpha wild-type mice that received PPAR-alpha ligand and cisplatin did show significant amelioration of acute tubular necrosis, as well as a significant reduction in the number of apoptotic cells in the proximal tubule when compared with the cisplatin-treated group. In contrast, in PPAR-alpha-null mice treated with the ligand and cisplatin, Endo G protein expression was not reduced and this was accompanied by lack of protection of kidney function. We conclude that PPAR-alpha ligand protects against cisplatin-induced renal injury via a PPAR-alpha-dependent mechanism by reducing the expression and enzyme activity of proximal tubule Endo G, which results in amelioration of both proximal tubule cell apoptosis and necrosis.
Collapse
Affiliation(s)
- Shenyang Li
- Dept. of Medicine, University of Arkansas for Medical Sciences, Slot 501, 4301 W. Markham Street, Little Rock, AR 72205, USA
| | | | | | | | | | | | | |
Collapse
|