1
|
Kim SK, Bae GS, Bae T, Ku SK, Choi BH, Kwak MK. Renal microRNA-144-3p is associated with transforming growth factor-β1-induced oxidative stress and fibrosis by suppressing the NRF2 pathway in hypertensive diabetic kidney disease. Free Radic Biol Med 2024; 225:546-559. [PMID: 39423929 DOI: 10.1016/j.freeradbiomed.2024.10.286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 08/28/2024] [Accepted: 10/12/2024] [Indexed: 10/21/2024]
Abstract
Chronic kidney disease (CKD) is a global health problem characterized by progressive renal fibrosis and excessive extracellular matrix deposition. Oxidative stress and epigenetic regulation, particularly through microRNAs (miRNAs), play crucial roles in the pathogenesis of CKD. In this study, we investigated the role of urinary miR-144-3p, which is upregulated in rats with CKD induced by diabetes and hypertension, in renal fibrosis progression, particularly its regulation of the nuclear factor erythroid-2-related factor 2 (NRF2) pathway. Our findings revealed elevated miR-144-3p levels and reduced NRF2 and target gene levels in kidney tissues of streptozotocin-treated spontaneously hypertensive rats. In vitro experiments demonstrated that miR-144-3p directly binds to the 3'-untranslated region of nrf2, suppressing the NRF2 pathway in renal tubular epithelial cells. Additionally, the profibrogenic factor transforming growth factor (TGF)-β1 increased miR-144-3p expression. TGF-β1-induced NRF2 suppression and reactive oxygen species elevation were found to be mediated through miR-144-3p upregulation. In vivo, cilostazol, an antiplatelet drug with an NRF2-activating effect, ameliorated renal injury in diabetic hypertensive rats by decreasing TGF-β1 and miR-144-3p levels while increasing NRF2 and its target gene levels in the kidneys. These findings highlight the potential therapeutic value of targeting the miR-144-3p/NRF2 pathway to attenuate CKD progression in hypertensive diabetic conditions.
Collapse
Affiliation(s)
- Seung Ki Kim
- Department of Pharmacy and BK21FOUR Advanced Program for SmartPharma Leaders, Graduate School of The Catholic University of Korea, Gyeonggi-do, 14662, Republic of Korea
| | - Gwang Sun Bae
- Department of Pharmacy and BK21FOUR Advanced Program for SmartPharma Leaders, Graduate School of The Catholic University of Korea, Gyeonggi-do, 14662, Republic of Korea
| | - Taegeun Bae
- Integrated Research Institute for Pharmaceutical Sciences, The Catholic University of Korea, Gyeonggi-do, 14662, Republic of Korea
| | - Sae-Kwang Ku
- College of Korean Medicine, Daegu Haany University, Gyeonsangbuk-do, 712-715, Republic of Korea
| | - Bo-Hyun Choi
- Department of Pharmacology, School of Medicine, Daegu Catholic University, Daegu, 42472, Republic of Korea
| | - Mi-Kyoung Kwak
- Department of Pharmacy and BK21FOUR Advanced Program for SmartPharma Leaders, Graduate School of The Catholic University of Korea, Gyeonggi-do, 14662, Republic of Korea; Integrated Research Institute for Pharmaceutical Sciences, The Catholic University of Korea, Gyeonggi-do, 14662, Republic of Korea; College of Pharmacy, The Catholic University of Korea, Gyeonggi-do, 14662, Republic of Korea.
| |
Collapse
|
2
|
Fu Y, Xiang Y, Wei Q, Ilatovskaya D, Dong Z. Rodent models of AKI and AKI-CKD transition: an update in 2024. Am J Physiol Renal Physiol 2024; 326:F563-F583. [PMID: 38299215 PMCID: PMC11208034 DOI: 10.1152/ajprenal.00402.2023] [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: 12/13/2023] [Revised: 01/29/2024] [Accepted: 01/29/2024] [Indexed: 02/02/2024] Open
Abstract
Despite known drawbacks, rodent models are essential tools in the research of renal development, physiology, and pathogenesis. In the past decade, rodent models have been developed and used to mimic different etiologies of acute kidney injury (AKI), AKI to chronic kidney disease (CKD) transition or progression, and AKI with comorbidities. These models have been applied for both mechanistic research and preclinical drug development. However, current rodent models have their limitations, especially since they often do not fully recapitulate the pathophysiology of AKI in human patients, and thus need further refinement. Here, we discuss the present status of these rodent models, including the pathophysiologic compatibility, clinical translational significance, key factors affecting model consistency, and their main limitations. Future efforts should focus on establishing robust models that simulate the major clinical and molecular phenotypes of human AKI and its progression.
Collapse
Affiliation(s)
- Ying Fu
- Department of Nephrology, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, People's Republic of China
| | - Yu Xiang
- Department of Nephrology, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, People's Republic of China
| | - Qingqing Wei
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, United States
| | - Daria Ilatovskaya
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Zheng Dong
- Department of Nephrology, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, People's Republic of China
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, United States
- Research Department, Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, United States
| |
Collapse
|
3
|
Colombijn JM, Hooft L, Jun M, Webster AC, Bots ML, Verhaar MC, Vernooij RW. Antioxidants for adults with chronic kidney disease. Cochrane Database Syst Rev 2023; 11:CD008176. [PMID: 37916745 PMCID: PMC10621004 DOI: 10.1002/14651858.cd008176.pub3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
BACKGROUND Chronic kidney disease (CKD) is a significant risk factor for cardiovascular disease (CVD) and death. Increased oxidative stress in people with CKD has been implicated as a potential causative factor. Antioxidant therapy decreases oxidative stress and may consequently reduce cardiovascular morbidity and death in people with CKD. This is an update of a Cochrane review first published in 2012. OBJECTIVES To examine the benefits and harms of antioxidant therapy on death and cardiovascular and kidney endpoints in adults with CKD stages 3 to 5, patients undergoing dialysis, and kidney transplant recipients. SEARCH METHODS We searched the Cochrane Kidney and Transplant Register of Studies until 15 November 2022 using search terms relevant to this review. Studies in the Register are identified through searches of CENTRAL, MEDLINE, and EMBASE, conference proceedings, the International Clinical Trials Registry Platform (ICTRP) Search Portal, and ClinicalTrials.gov. SELECTION CRITERIA We included all randomised controlled trials investigating the use of antioxidants, compared with placebo, usual or standard care, no treatment, or other antioxidants, for adults with CKD on cardiovascular and kidney endpoints. DATA COLLECTION AND ANALYSIS Titles and abstracts were screened independently by two authors who also performed data extraction using standardised forms. Results were pooled using random effects models and expressed as risk ratios (RR) or mean difference (MD) with 95% confidence intervals (CI). Confidence in the evidence was assessed using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach. MAIN RESULTS We included 95 studies (10,468 randomised patients) that evaluated antioxidant therapy in adults with non-dialysis-dependent CKD (31 studies, 5342 patients), dialysis-dependent CKD (41 studies, 3444 patients) and kidney transplant recipients (21 studies, 1529 patients). Two studies enrolled dialysis and non-dialysis patients (153 patients). Twenty-one studies assessed the effects of vitamin antioxidants, and 74 assessed the effects of non-vitamin antioxidants. Overall, the quality of included studies was moderate to low or very low due to unclear or high risk of bias for randomisation, allocation concealment, blinding, and loss to follow-up. Compared with placebo, usual care, or no treatment, antioxidant therapy may have little or no effect on cardiovascular death (8 studies, 3813 patients: RR 0.94, 95% CI 0.64 to 1.40; I² = 33%; low certainty of evidence) and probably has little to no effect on death (any cause) (45 studies, 7530 patients: RR 0.95, 95% CI 0.82 to 1.11; I² = 0%; moderate certainty of evidence), CVD (16 studies, 4768 patients: RR 0.79, 95% CI 0.63 to 0.99; I² = 23%; moderate certainty of evidence), or loss of kidney transplant (graft loss) (11 studies, 1053 patients: RR 0.88, 95% CI 0.67 to 1.17; I² = 0%; moderate certainty of evidence). Compared with placebo, usual care, or no treatment, antioxidants had little to no effect on the slope of urinary albumin/creatinine ratio (change in UACR) (7 studies, 1286 patients: MD -0.04 mg/mmol, 95% CI -0.55 to 0.47; I² = 37%; very low certainty of evidence) but the evidence is very uncertain. Antioxidants probably reduced the progression to kidney failure (10 studies, 3201 patients: RR 0.65, 95% CI 0.41 to 1.02; I² = 41%; moderate certainty of evidence), may improve the slope of estimated glomerular filtration rate (change in eGFR) (28 studies, 4128 patients: MD 3.65 mL/min/1.73 m², 95% CI 2.81 to 4.50; I² = 99%; low certainty of evidence), but had uncertain effects on the slope of serum creatinine (change in SCr) (16 studies, 3180 patients: MD -13.35 µmol/L, 95% CI -23.49 to -3.23; I² = 98%; very low certainty of evidence). Possible safety concerns are an observed increase in the risk of infection (14 studies, 3697 patients: RR 1.30, 95% CI 1.14 to 1.50; I² = 3%; moderate certainty of evidence) and heart failure (6 studies, 3733 patients: RR 1.40, 95% CI 1.11 to 1.75; I² = 0; moderate certainty of evidence) among antioxidant users. Results of studies with a low risk of bias or longer follow-ups generally were comparable to the main analyses. AUTHORS' CONCLUSIONS We found no evidence that antioxidants reduced death or improved kidney transplant outcomes or proteinuria in patients with CKD. Antioxidants likely reduce cardiovascular events and progression to kidney failure and may improve kidney function. Possible concerns are an increased risk of infections and heart failure among antioxidant users. However, most studies were of suboptimal quality and had limited follow-up, and few included people undergoing dialysis or kidney transplant recipients. Furthermore, the large heterogeneity in interventions hampers drawing conclusions on the efficacy and safety of individual agents.
Collapse
Affiliation(s)
- Julia Mt Colombijn
- Department of Nephrology and Hypertension, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, Netherlands
| | - Lotty Hooft
- Cochrane Netherlands, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Min Jun
- The George Institute for Global Health, UNSW, Sydney, Australia
| | - Angela C Webster
- Sydney School of Public Health, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Westmead Applied Research Centre, The University of Sydney at Westmead, Westmead, Australia
- NHMRC Clinical Trials Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Department of Transplant and Renal Medicine, Westmead Hospital, Westmead, Australia
| | - Michiel L Bots
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands
| | - Marianne C Verhaar
- Department of Nephrology and Hypertension, University of Utrecht, Utrecht, Netherlands
| | - Robin Wm Vernooij
- Department of Nephrology and Hypertension, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
4
|
Su K, Zhao SL, Yang WX, Lo CS, Chenier I, Liao MC, Pang YC, Peng JZ, Miyata KN, Cailhier JF, Ethier J, Lattouf JB, Filep JG, Ingelfinger JR, Zhang SL, Chan JSD. NRF2 Deficiency Attenuates Diabetic Kidney Disease in Db/Db Mice via Down-Regulation of Angiotensinogen, SGLT2, CD36, and FABP4 Expression and Lipid Accumulation in Renal Proximal Tubular Cells. Antioxidants (Basel) 2023; 12:1715. [PMID: 37760019 PMCID: PMC10525648 DOI: 10.3390/antiox12091715] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/17/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
The role(s) of nuclear factor erythroid 2-related factor 2 (NRF2) in diabetic kidney disease (DKD) is/are controversial. We hypothesized that Nrf2 deficiency in type 2 diabetes (T2D) db/db mice (db/dbNrf2 knockout (KO)) attenuates DKD progression through the down-regulation of angiotensinogen (AGT), sodium-glucose cotransporter-2 (SGLT2), scavenger receptor CD36, and fatty -acid-binding protein 4 (FABP4), and lipid accumulation in renal proximal tubular cells (RPTCs). Db/dbNrf2 KO mice were studied at 16 weeks of age. Human RPTCs (HK2) with NRF2 KO via CRISPR-Cas9 genome editing and kidneys from patients with or without T2D were examined. Compared with db/db mice, db/dbNrf2 KO mice had lower systolic blood pressure, fasting blood glucose, kidney hypertrophy, glomerular filtration rate, urinary albumin/creatinine ratio, tubular lipid droplet accumulation, and decreased expression of AGT, SGLT2, CD36, and FABP4 in RPTCs. Male and female mice had similar results. NRF2 KO attenuated the stimulatory effect of the Nrf2 activator, oltipraz, on AGT, SGLT2, and CD36 expression and high-glucose/free fatty acid (FFA)-stimulated lipid accumulation in HK2. Kidneys from T2D patients exhibited markedly higher levels of CD36 and FABP4 in RPTCs than kidneys from non-diabetic patients. These data suggest that NRF2 exacerbates DKD through the stimulation of AGT, SGLT2, CD36, and FABP4 expression and lipid accumulation in RPTCs of T2D.
Collapse
Affiliation(s)
- Ke Su
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Département de Médecine, Université de Montréal, 900 Saint Denis Street, Montréal, QC H2X 0A9, Canada; (K.S.); (S.-L.Z.); (W.-X.Y.); (C.-S.L.); (I.C.); (M.-C.L.); (Y.-C.P.); (J.-Z.P.); (K.N.M.); (J.-F.C.); (J.E.); (J.-B.L.)
| | - Shui-Ling Zhao
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Département de Médecine, Université de Montréal, 900 Saint Denis Street, Montréal, QC H2X 0A9, Canada; (K.S.); (S.-L.Z.); (W.-X.Y.); (C.-S.L.); (I.C.); (M.-C.L.); (Y.-C.P.); (J.-Z.P.); (K.N.M.); (J.-F.C.); (J.E.); (J.-B.L.)
| | - Wen-Xia Yang
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Département de Médecine, Université de Montréal, 900 Saint Denis Street, Montréal, QC H2X 0A9, Canada; (K.S.); (S.-L.Z.); (W.-X.Y.); (C.-S.L.); (I.C.); (M.-C.L.); (Y.-C.P.); (J.-Z.P.); (K.N.M.); (J.-F.C.); (J.E.); (J.-B.L.)
| | - Chao-Sheng Lo
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Département de Médecine, Université de Montréal, 900 Saint Denis Street, Montréal, QC H2X 0A9, Canada; (K.S.); (S.-L.Z.); (W.-X.Y.); (C.-S.L.); (I.C.); (M.-C.L.); (Y.-C.P.); (J.-Z.P.); (K.N.M.); (J.-F.C.); (J.E.); (J.-B.L.)
| | - Isabelle Chenier
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Département de Médecine, Université de Montréal, 900 Saint Denis Street, Montréal, QC H2X 0A9, Canada; (K.S.); (S.-L.Z.); (W.-X.Y.); (C.-S.L.); (I.C.); (M.-C.L.); (Y.-C.P.); (J.-Z.P.); (K.N.M.); (J.-F.C.); (J.E.); (J.-B.L.)
| | - Min-Chun Liao
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Département de Médecine, Université de Montréal, 900 Saint Denis Street, Montréal, QC H2X 0A9, Canada; (K.S.); (S.-L.Z.); (W.-X.Y.); (C.-S.L.); (I.C.); (M.-C.L.); (Y.-C.P.); (J.-Z.P.); (K.N.M.); (J.-F.C.); (J.E.); (J.-B.L.)
| | - Yu-Chao Pang
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Département de Médecine, Université de Montréal, 900 Saint Denis Street, Montréal, QC H2X 0A9, Canada; (K.S.); (S.-L.Z.); (W.-X.Y.); (C.-S.L.); (I.C.); (M.-C.L.); (Y.-C.P.); (J.-Z.P.); (K.N.M.); (J.-F.C.); (J.E.); (J.-B.L.)
| | - Jun-Zheng Peng
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Département de Médecine, Université de Montréal, 900 Saint Denis Street, Montréal, QC H2X 0A9, Canada; (K.S.); (S.-L.Z.); (W.-X.Y.); (C.-S.L.); (I.C.); (M.-C.L.); (Y.-C.P.); (J.-Z.P.); (K.N.M.); (J.-F.C.); (J.E.); (J.-B.L.)
| | - Kana N. Miyata
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Département de Médecine, Université de Montréal, 900 Saint Denis Street, Montréal, QC H2X 0A9, Canada; (K.S.); (S.-L.Z.); (W.-X.Y.); (C.-S.L.); (I.C.); (M.-C.L.); (Y.-C.P.); (J.-Z.P.); (K.N.M.); (J.-F.C.); (J.E.); (J.-B.L.)
| | - Jean-Francois Cailhier
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Département de Médecine, Université de Montréal, 900 Saint Denis Street, Montréal, QC H2X 0A9, Canada; (K.S.); (S.-L.Z.); (W.-X.Y.); (C.-S.L.); (I.C.); (M.-C.L.); (Y.-C.P.); (J.-Z.P.); (K.N.M.); (J.-F.C.); (J.E.); (J.-B.L.)
| | - Jean Ethier
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Département de Médecine, Université de Montréal, 900 Saint Denis Street, Montréal, QC H2X 0A9, Canada; (K.S.); (S.-L.Z.); (W.-X.Y.); (C.-S.L.); (I.C.); (M.-C.L.); (Y.-C.P.); (J.-Z.P.); (K.N.M.); (J.-F.C.); (J.E.); (J.-B.L.)
| | - Jean-Baptiste Lattouf
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Département de Médecine, Université de Montréal, 900 Saint Denis Street, Montréal, QC H2X 0A9, Canada; (K.S.); (S.-L.Z.); (W.-X.Y.); (C.-S.L.); (I.C.); (M.-C.L.); (Y.-C.P.); (J.-Z.P.); (K.N.M.); (J.-F.C.); (J.E.); (J.-B.L.)
| | - Janos G. Filep
- Centre de Recherche, Hôpital Maisonneuve-Rosemont, Département de Pathologie et Biologie Cellulaire, Université de Montréal, 5415 Boul. de l’Assomption, Montréal, QC H1T 2M4, Canada;
| | - Julie R. Ingelfinger
- Pediatric Nephrology Unit, Massachusetts General Hospital, Harvard Medical School, 15 Parkman Street, WAC 709, Boston, MA 02114, USA;
| | - Shao-Ling Zhang
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Département de Médecine, Université de Montréal, 900 Saint Denis Street, Montréal, QC H2X 0A9, Canada; (K.S.); (S.-L.Z.); (W.-X.Y.); (C.-S.L.); (I.C.); (M.-C.L.); (Y.-C.P.); (J.-Z.P.); (K.N.M.); (J.-F.C.); (J.E.); (J.-B.L.)
| | - John S. D. Chan
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Département de Médecine, Université de Montréal, 900 Saint Denis Street, Montréal, QC H2X 0A9, Canada; (K.S.); (S.-L.Z.); (W.-X.Y.); (C.-S.L.); (I.C.); (M.-C.L.); (Y.-C.P.); (J.-Z.P.); (K.N.M.); (J.-F.C.); (J.E.); (J.-B.L.)
| |
Collapse
|
5
|
Sharma N, Sircar A, Anders HJ, Gaikwad AB. Crosstalk between kidney and liver in non-alcoholic fatty liver disease: mechanisms and therapeutic approaches. Arch Physiol Biochem 2022; 128:1024-1038. [PMID: 32223569 DOI: 10.1080/13813455.2020.1745851] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Liver and kidney are vital organs that maintain homeostasis and injury to either of them triggers pathogenic pathways affecting the other. For example, non-alcoholic fatty liver disease (NAFLD) promotes the progression of chronic kidney disease (CKD), vice versa acute kidney injury (AKI) endorses the induction and progression of liver dysfunction. Progress in clinical and basic research suggest a role of excessive fructose intake, insulin resistance, inflammatory cytokines production, activation of the renin-angiotensin system, redox imbalance, and their impact on epigenetic regulation of gene expression in this context. Recent developments in experimental and clinical research have identified several biochemical and molecular pathways for AKI-liver interaction, including altered liver enzymes profile, metabolic acidosis, oxidative stress, activation of inflammatory and regulated cell death pathways. This review focuses on the current preclinical and clinical findings on kidney-liver crosstalk in NAFLD-CKD and AKI-liver dysfunction settings and highlights potential molecular mechanisms and therapeutic targets.
Collapse
Affiliation(s)
- Nisha Sharma
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, Rajasthan, India
| | - Anannya Sircar
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, Rajasthan, India
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Internal Medicine IV, University Hospital of the Ludwig Maximilians University Munich, Munich, Germany
| | - Anil Bhanudas Gaikwad
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, Rajasthan, India
| |
Collapse
|
6
|
Aranda-Rivera AK, Cruz-Gregorio A, Pedraza-Chaverri J, Scholze A. Nrf2 Activation in Chronic Kidney Disease: Promises and Pitfalls. Antioxidants (Basel) 2022; 11:antiox11061112. [PMID: 35740009 PMCID: PMC9220138 DOI: 10.3390/antiox11061112] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/01/2022] [Accepted: 06/01/2022] [Indexed: 11/16/2022] Open
Abstract
The nuclear factor erythroid 2-related factor 2 (Nrf2) protects the cell against oxidative damage. The Nrf2 system comprises a complex network that functions to ensure adequate responses to redox perturbations, but also metabolic demands and cellular stresses. It must be kept within a physiologic activity range. Oxidative stress and alterations in Nrf2-system activity are central for chronic-kidney-disease (CKD) progression and CKD-related morbidity. Activation of the Nrf2 system in CKD is in multiple ways related to inflammation, kidney fibrosis, and mitochondrial and metabolic effects. In human CKD, both endogenous Nrf2 activation and repression exist. The state of the Nrf2 system varies with the cause of kidney disease, comorbidities, stage of CKD, and severity of uremic toxin accumulation and inflammation. An earlier CKD stage, rapid progression of kidney disease, and inflammatory processes are associated with more robust Nrf2-system activation. Advanced CKD is associated with stronger Nrf2-system repression. Nrf2 activation is related to oxidative stress and moderate uremic toxin and nuclear factor kappa B (NF-κB) elevations. Nrf2 repression relates to high uremic toxin and NF-κB concentrations, and may be related to Kelch-like ECH-associated protein 1 (Keap1)-independent Nrf2 degradation. Furthermore, we review the effects of pharmacological Nrf2 activation by bardoxolone methyl, curcumin, and resveratrol in human CKD and outline strategies for how to adapt future Nrf2-targeted therapies to the requirements of patients with CKD.
Collapse
Affiliation(s)
- Ana Karina Aranda-Rivera
- Laboratory F-315, Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico, Mexico City 04510, Mexico; (A.K.A.-R.); (A.C.-G.); (J.P.-C.)
| | - Alfredo Cruz-Gregorio
- Laboratory F-315, Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico, Mexico City 04510, Mexico; (A.K.A.-R.); (A.C.-G.); (J.P.-C.)
| | - José Pedraza-Chaverri
- Laboratory F-315, Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico, Mexico City 04510, Mexico; (A.K.A.-R.); (A.C.-G.); (J.P.-C.)
| | - Alexandra Scholze
- Department of Nephrology, Odense University Hospital, 5000 Odense C, Denmark
- Institute of Clinical Research, University of Southern Denmark, 5000 Odense C, Denmark
- Correspondence:
| |
Collapse
|
7
|
Wang H, Xu H, Chen W, Cheng M, Zou L, Yang Q, Chan CB, Zhu H, Chen C, Nie JY, Jiao B. Rab13 sustains breast cancer stem cells by supporting tumor-stroma crosstalk. Cancer Res 2022; 82:2124-2140. [PMID: 35395074 DOI: 10.1158/0008-5472.can-21-4097] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/02/2022] [Accepted: 04/05/2022] [Indexed: 11/16/2022]
Abstract
Cancer stem cells (CSC) are supported by the tumor microenvironment, and non-CSCs can regain CSC phenotypes in certain niches, leading to limited clinical benefits of CSC-targeted therapy. A better understanding of the mechanisms governing the orchestration of the CSC niche could help improve the therapeutic targeting of CSCs. Here, we report that Rab13, a small GTPase, is highly expressed in breast CSCs (BCSCs). Rab13 depletion suppressed breast cancer cell stemness, tumorigenesis, and chemoresistance by reducing tumor-stroma crosstalk. Accordingly, Rab13 controlled the membrane translocation of CXCR1/2, allowing tumor cells to interact with tumor-associated macrophages and cancer-associated fibroblasts to establish a supportive BCSC niche. Targeting the Rab13-mediated BCSC niche with bardoxolone-methyl (CDDO-Me) prevented BCSC stemness in vitro and in vivo. These findings highlight the novel regulatory mechanism of Rab13 in BCSC, with important implications for the development of therapeutic strategies for disrupting the BCSC niche.
Collapse
Affiliation(s)
- Hui Wang
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Haibo Xu
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, 518035, China., Shenzhen, Guangdong, China
| | - Wei Chen
- Kunming Institute of Zoology, Chinese Academy of Sciences, China
| | - Mei Cheng
- Kunming Institute of Zoology, Chinese Academy of Sciences, China
| | - Li Zou
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Qin Yang
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | | | - Hao Zhu
- Southern Medical University, Guangzhou, China
| | - Ceshi Chen
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Jian-Yun Nie
- The Third Affiliated Hospital of Kunming Medical University, KUNMING, Yunnan, China
| | - Baowei Jiao
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| |
Collapse
|
8
|
In vitro study on effect of bardoxolone methyl on cisplatin-induced cellular senescence in human proximal tubular cells. Mol Cell Biochem 2022; 477:689-699. [PMID: 34973124 PMCID: PMC8857011 DOI: 10.1007/s11010-021-04295-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 11/04/2021] [Indexed: 11/29/2022]
Abstract
Bardoxolone methyl [methyl-2-cyano-3, 12-dioxooleana-1, 9(11)dien-28-oate (CDDO-Me)], an activator of the nuclear factor erythroid-derived 2-related factor2 pathway, is a potential therapeutic candidate for the treatment of kidney diseases. However, its effect against cellular senescence remains unclear. This study aimed to investigate whether CDDO-Me protects cells against cisplatin-induced cellular senescence using an in vitro model. The human renal proximal tubular epithelial cell line HK-2 was treated with cisplatin for 6 h, followed by treatment with or without CDDO-Me (0.1 or 0.2 μmol/L). Senescence markers were analyzed using western blotting and real-time PCR. Apoptosis was evaluated through TUNEL staining. Cisplatin induced changes in the levels of markers specific for proliferation, cell cycle, and senescence in a time- and dose-dependent manner. Furthermore, IL-6 and IL-8 levels in the culture medium increased markedly. These data suggested that cellular senescence-like alterations occurred in HK-2 cells exposed to cisplatin. CDDO-Me treatment reversed the cisplatin-mediated alterations in the levels of cellular senescence markers. The antioxidant enzymes, HO1, NQO1, GPX1, and CAT were upregulated by CDDO-Me treatment. Furthermore, CDDO-Me treatment induced apoptosis in cisplatin-exposed HK-2 cells. Pretreatment with Ac-DEVD-CHO, the caspase inhibitor, suppressed the reversal effect of CDDO-Me against cisplatin-induced cellular senescence-like alterations. This study showed that CDDO-Me attenuated cisplatin-induced premature senescence of HK-2 cells. This beneficial effect may be related to Nrf2 activation. Our findings also showed that CDDO-Me induced apoptosis in cisplatin-treated HK-2 cells, potentially protecting the kidneys from cellular senescence. CDDO-Me appears to be a candidate treatment for acute kidney injury.
Collapse
|
9
|
Phillips J, Chen JHC, Ooi E, Prunster J, Lim WH. Global Epidemiology, Health Outcomes, and Treatment Options for Patients With Type 2 Diabetes and Kidney Failure. FRONTIERS IN CLINICAL DIABETES AND HEALTHCARE 2021; 2:731574. [PMID: 36994340 PMCID: PMC10012134 DOI: 10.3389/fcdhc.2021.731574] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 07/29/2021] [Indexed: 12/15/2022]
Abstract
The burden of type 2 diabetes and related complications has steadily increased over the last few decades and is one of the foremost global public health threats in the 21st century. Diabetes is one of the leading causes of chronic kidney disease and kidney failure and is an important contributor to the cardiovascular morbidity and mortality in this population. In addition, up to one in three patients who have received kidney transplants develop post-transplant diabetes, but the management of this common complication continues to pose a significant challenge for clinicians. In this review, we will describe the global prevalence and temporal trend of kidney failure attributed to diabetes mellitus in both developing and developed countries. We will examine the survival differences between treated kidney failure patients with and without type 2 diabetes, focusing on the survival differences in those on maintenance dialysis or have received kidney transplants. With the increased availability of novel hypoglycemic agents, we will address the potential impacts of these novel agents in patients with diabetes and kidney failure and in those who have developed post-transplant diabetes.
Collapse
Affiliation(s)
- Jessica Phillips
- Department of Renal Medicine, Sir Charles Gairdner Hospital, Perth, WA, Australia
- *Correspondence: Jessica Phillips,
| | - Jenny H. C. Chen
- School of Medicine, University of Wollongong, Wollongong, NSW, Australia
- Depatment of Nephrology, Wollongong Hospital, Wollongong, NSW, Australia
| | - Esther Ooi
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Janelle Prunster
- Department of Renal Medicine, Cairns Hospital, Cairns, QLD, Australia
| | - Wai H. Lim
- Department of Renal Medicine, Sir Charles Gairdner Hospital, Perth, WA, Australia
- Medical School, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
10
|
Conley MM, McFarlane CM, Johnson DW, Kelly JT, Campbell KL, MacLaughlin HL. Interventions for weight loss in people with chronic kidney disease who are overweight or obese. Cochrane Database Syst Rev 2021; 3:CD013119. [PMID: 33782940 PMCID: PMC8094234 DOI: 10.1002/14651858.cd013119.pub2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Obesity and chronic kidney disease (CKD) are highly prevalent worldwide and result in substantial health care costs. Obesity is a predictor of incident CKD and progression to kidney failure. Whether weight loss interventions are safe and effective to impact on disease progression and clinical outcomes, such as death remains unclear. OBJECTIVES This review aimed to evaluate the safety and efficacy of intentional weight loss interventions in overweight and obese adults with CKD; including those with end-stage kidney disease (ESKD) being treated with dialysis, kidney transplantation, or supportive care. SEARCH METHODS We searched the Cochrane Kidney and Transplant Register of Studies up to 14 December 2020 through contact with the Information Specialist using search terms relevant to this review. Studies in the Register are identified through searches of CENTRAL, MEDLINE, EMBASE, conference proceedings, the International Clinical Trials Register (ICTRP) Search Portal and ClinicalTrials.gov. SELECTION CRITERIA Randomised controlled trials (RCTs) and quasi-RCTs of more than four weeks duration, reporting on intentional weight loss interventions, in individuals with any stage of CKD, designed to promote weight loss as one of their primary stated goals, in any health care setting. DATA COLLECTION AND ANALYSIS Two authors independently assessed study eligibility and extracted data. We applied the Cochrane 'Risk of Bias' tool and used the GRADE process to assess the certainty of evidence. We estimated treatment effects using random-effects meta-analysis. Results were expressed as risk ratios (RR) for dichotomous outcomes together with 95% confidence intervals (CI) or mean differences (MD) or standardised mean difference (SMD) for continuous outcomes or in descriptive format when meta-analysis was not possible. MAIN RESULTS We included 17 RCTs enrolling 988 overweight or obese adults with CKD. The weight loss interventions and comparators across studies varied. We categorised comparisons into three groups: any weight loss intervention versus usual care or control; any weight loss intervention versus dietary intervention; and surgical intervention versus non-surgical intervention. Methodological quality was varied, with many studies providing insufficient information to accurately judge the risk of bias. Death (any cause), cardiovascular events, successful kidney transplantation, nutritional status, cost effectiveness and economic analysis were not measured in any of the included studies. Across all 17 studies many clinical parameters, patient-centred outcomes, and adverse events were not measured limiting comparisons for these outcomes. In studies comparing any weight loss intervention to usual care or control, weight loss interventions may lead to weight loss or reduction in body weight post intervention (6 studies, 180 participants: MD -3.69 kg, 95% CI -5.82 to -1.57; follow-up: 5 weeks to 12 months, very low-certainty evidence). In very low certainty evidence any weight loss intervention had uncertain effects on body mass index (BMI) (4 studies, 100 participants: MD -2.18 kg/m², 95% CI -4.90 to 0.54), waist circumference (2 studies, 53 participants: MD 0.68 cm, 95% CI -7.6 to 6.24), proteinuria (4 studies, 84 participants: 0.29 g/day, 95% CI -0.76 to 0.18), systolic (4 studies, 139 participants: -3.45 mmHg, 95% CI -9.99 to 3.09) and diastolic blood pressure (4 studies, 139 participants: -2.02 mmHg, 95% CI -3.79 to 0.24). Any weight loss intervention made little or no difference to total cholesterol, high density lipoprotein cholesterol, and inflammation, but may lower low density lipoprotein cholesterol. There was little or no difference between any weight loss interventions (lifestyle or pharmacological) compared to dietary-only weight loss interventions for weight loss, BMI, waist circumference, proteinuria, and systolic blood pressure, however diastolic blood pressure was probably reduced. Furthermore, studies comparing the efficacy of different types of dietary interventions failed to find a specific dietary intervention to be superior for weight loss or a reduction in BMI. Surgical interventions probably reduced body weight (1 study, 11 participants: MD -29.50 kg, 95% CI -36.4 to -23.35), BMI (2 studies, 17 participants: MD -10.43 kg/m², 95% CI -13.58 to -7.29), and waist circumference (MD -30.00 cm, 95% CI -39.93 to -20.07) when compared to non-surgical weight loss interventions after 12 months of follow-up. Proteinuria and blood pressure were not reported. All results across all comparators should be interpreted with caution due to the small number of studies, very low quality of evidence and heterogeneity across interventions and comparators. AUTHORS' CONCLUSIONS All types of weight loss interventions had uncertain effects on death and cardiovascular events among overweight and obese adults with CKD as no studies reported these outcome measures. Non-surgical weight loss interventions (predominately lifestyle) appear to be an effective treatment to reduce body weight, and LDL cholesterol. Surgical interventions probably reduce body weight, waist circumference, and fat mass. The current evidence is limited by the small number of included studies, as well as the significant heterogeneity and a high risk of bias in most studies.
Collapse
Affiliation(s)
- Marguerite M Conley
- Department of Nutrition and Dietetics, Princess Alexandra Hospital, Woolloongabba, Australia
- School of Exercise and Nutrition Sciences, Queensland University of Technology, Brisbane, Australia
| | | | - David W Johnson
- Department of Nephrology, Princess Alexandra Hospital, Woolloongabba, Australia
| | - Jaimon T Kelly
- Centre for Applied Health Economics, Menzies Health Institute Queensland, Griffith University, Nathan, Australia
| | - Katrina L Campbell
- Centre for Applied Health Economics, Menzies Health Institute Queensland, Griffith University, Nathan, Australia
| | - Helen L MacLaughlin
- School of Exercise and Nutrition Sciences, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
11
|
Gu X, Liu Y, Wang N, Zhen J, Zhang B, Hou S, Cui Z, Wan Q, Feng H. Transcription of MRPL12 regulated by Nrf2 contributes to the mitochondrial dysfunction in diabetic kidney disease. Free Radic Biol Med 2021; 164:329-340. [PMID: 33444714 DOI: 10.1016/j.freeradbiomed.2021.01.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 12/30/2020] [Accepted: 01/03/2021] [Indexed: 02/06/2023]
Abstract
Diabetic kidney disease (DKD) is the leading cause of chronic kidney disease (CKD) and end-stage renal disease (ESRD). Increasing evidences suggested that DKD correlates more closely to mitochondrial dysfunction than to hyperglycemia. Our previous study has reported that mitochondrial ribosomal protein L7/L12 (MRPL12) could positively control the mitochondrial oxidative phosphorylation (OXPHOS) and mtDNA copy number. The present study further investigated the role of MRPL12 in mitochondrial dysfunction of DKD. Using a mass spectrometry-based proteomics and immunohistochemistry, we found that MRPL12 underwent significant decreases in diabetic kidneys. Moreover, decreased expression of MRPL12 was associated with reduced mitochondrial OXPHOS in proximal tubular epithelial cells (PTECs) and overexpression of MRPL12 could alleviated the impairment of OXPHOS induced by long term high glucose. We further explored the upstream mechanism and identified nuclear factor erythroid 2-related factor 2 (Nrf2) as a potential transcription factor for MRPL12. Nrf2 changes consistently with MRPL12 in DKD and correlates with alterations of mitochondrial function, fibrosis and apoptosis of PTECs treated with high glucose challenge. Thus, the role of MRPL12 in the maintenance of mitochondrial function in DKD may be regulated by Nrf2, and provides new potential therapeutic targets for DKD.
Collapse
Affiliation(s)
- Xia Gu
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yi Liu
- Department of Pulmonary and Critical Care Medicine, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Na Wang
- Medical Department, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Junhui Zhen
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Bo Zhang
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Shaoshuai Hou
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Zhengguo Cui
- Department of Public Health, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Qiang Wan
- Department of Endocrinology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| | - Hong Feng
- Cancer Center, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
12
|
Gutierrez B, Gallardo I, Ruiz L, Alvarez Y, Cachofeiro V, Margolles A, Hernandez M, Nieto ML. Oleanolic acid ameliorates intestinal alterations associated with EAE. J Neuroinflammation 2020; 17:363. [PMID: 33246492 PMCID: PMC7697371 DOI: 10.1186/s12974-020-02042-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 11/19/2020] [Indexed: 12/18/2022] Open
Abstract
Background Multiple sclerosis (MS) is a chronic demyelinating autoimmune disease affecting the CNS. Recent studies have indicated that intestinal alterations play key pathogenic roles in the development of autoimmune diseases, including MS. The triterpene oleanolic acid (OA), due to its anti-inflammatory properties, has shown to beneficially influence the severity of the experimental autoimmune encephalomyelitis (EAE), a preclinical model of MS. We herein investigate EAE-associated gut intestinal dysfunction and the effect of OA treatment. Methods Mice with MOG35–55-induced EAE were treated with OA or vehicle from immunization day and were daily analyzed for clinical deficit. We performed molecular and histological analysis in serum and intestinal tissues to measure oxidative and inflammatory responses. We used Caco-2 and HT29-MTX-E12 cells to elucidate OA in vitro effects. Results We found that OA protected from EAE-induced changes in intestinal permeability and preserved the mucin-containing goblet cells along the intestinal tract. Serum levels of the markers for intestinal barrier damage iFABP and monocyte activation sCD14 were consistently and significantly reduced in OA-treated EAE mice. Beneficial OA effects also included a decrease of pro-inflammatory mediators both in serum and colonic tissue of treated-EAE mice. Moreover, the levels of some immunoregulatory cytokines, the neurotrophic factor GDNF, and the gastrointestinal hormone motilin were preserved in OA-treated EAE mice. Regarding oxidative stress, OA treatment prevented lipid peroxidation and superoxide anion accumulation in intestinal tissue, while inducing the expression of the ROS scavenger Sestrin-3. Furthermore, short-chain fatty acids (SCFA) quantification in the cecal content showed that OA reduced the high iso-valeric acid concentrations detected in EAE-mice. Lastly, using in vitro cell models which mimic the intestinal epithelium, we verified that OA protected against intestinal barrier dysfunction induced by injurious agents produced in both EAE and MS. Conclusion These findings reveal that OA ameliorates the gut dysfunction found in EAE mice. OA normalizes the levels of gut mucosal dysfunction markers, as well as the pro- and anti-inflammatory immune bias during EAE, thus reinforcing the idea that OA is a beneficial compound for treating EAE and suggesting that OA may be an interesting candidate to be explored for the treatment of human MS. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-020-02042-6.
Collapse
Affiliation(s)
- Beatriz Gutierrez
- Instituto de Biología y Genética Molecular (IBGM-CSIC/UVa), Valladolid, Spain
| | - Isabel Gallardo
- Instituto de Biología y Genética Molecular (IBGM-CSIC/UVa), Valladolid, Spain
| | - Lorena Ruiz
- Dairy Research Institute of Asturias, Spanish National Research Council (IPLA-CSIC), Paseo Río Linares s/n, Villaviciosa, Asturias, Spain.,MicroHealth Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Asturias, Spain
| | - Yolanda Alvarez
- Instituto de Biología y Genética Molecular (IBGM-CSIC/UVa), Valladolid, Spain
| | - Victoria Cachofeiro
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid and Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Abelardo Margolles
- Dairy Research Institute of Asturias, Spanish National Research Council (IPLA-CSIC), Paseo Río Linares s/n, Villaviciosa, Asturias, Spain.,MicroHealth Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Asturias, Spain
| | - Marita Hernandez
- Instituto de Biología y Genética Molecular (IBGM-CSIC/UVa), Valladolid, Spain
| | - Maria Luisa Nieto
- Instituto de Biología y Genética Molecular (IBGM-CSIC/UVa), Valladolid, Spain.
| |
Collapse
|
13
|
O’Brien J, Wendell SG. Electrophile Modulation of Inflammation: A Two-Hit Approach. Metabolites 2020; 10:metabo10110453. [PMID: 33182676 PMCID: PMC7696920 DOI: 10.3390/metabo10110453] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/11/2022] Open
Abstract
Electrophilic small molecules have gained significant attention over the last decade in the field of covalent drug discovery. Long recognized as mediators of the inflammatory process, recent evidence suggests that electrophiles may modulate the immune response through the regulation of metabolic networks. These molecules function as pleiotropic signaling mediators capable of reversibly reacting with nucleophilic biomolecules, most notably at reactive cysteines. More specifically, electrophiles target critical cysteines in redox regulatory proteins to activate protective pathways such as the nuclear factor erythroid 2-related factor 2-Kelch-like ECH-associated protein 1 (Nrf2-Keap1) antioxidant signaling pathway while also inhibiting Nuclear Factor κB (NF-κB). During inflammatory states, reactive species broadly alter cell signaling through the oxidation of lipids, amino acids, and nucleic acids, effectively propagating the inflammatory sequence. Subsequent changes in metabolic signaling inform immune cell maturation and effector function. Therapeutic strategies targeting inflammatory pathologies leverage electrophilic drug compounds, in part, because of their documented effect on the redox balance of the cell. With mounting evidence demonstrating the link between redox signaling and metabolism, electrophiles represent ideal therapeutic candidates for the treatment of inflammatory conditions. Through their pleiotropic signaling activity, electrophiles may be used strategically to both directly and indirectly target immune cell metabolism.
Collapse
|
14
|
Anti-Inflammatory Activity of Kurarinone Involves Induction of HO-1 via the KEAP1/Nrf2 Pathway. Antioxidants (Basel) 2020; 9:antiox9090842. [PMID: 32916869 PMCID: PMC7554885 DOI: 10.3390/antiox9090842] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/31/2020] [Accepted: 09/07/2020] [Indexed: 02/07/2023] Open
Abstract
Kurarinone, a flavonoid isolated from the roots of Sophora flavescens, was suggested to exert potent antioxidant and immunosuppressive effects. However, the underlying mechanisms remain unclear. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a key transcription factor that regulates the antioxidant defense system with anti-inflammatory activity. In the present study, we demonstrated that kurarinone activated Nrf2 and increased the expression of antioxidant enzymes, including heme oxygenase-1 (HO-1). Mechanistically, kurarinone downregulated the expression of kelch-like ECH-associated protein 1 (KEAP1), subsequently leading to the activation of Nrf2. Kurarinone also inhibited the expression of the inflammatory cytokine, interleukin (IL)-1β, and inducible nitric oxide synthase (iNos) in lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages. The overexpression of HO-1 suppressed the LPS-induced production of inflammatory mediators in RAW264.7 cells, and the immunosuppressive effects of kurarinone were partially inhibited by a treatment with Tin Protomorphyrin IX (TinPPIX), an inhibitor of HO-1. These results indicate that kurarinone activates the KEAP1/Nrf2 pathway to induce HO-1 expression, thereby exerting immunosuppressive effects.
Collapse
|
15
|
Lewis JH, Jadoul M, Block GA, Chin MP, Ferguson DA, Goldsberry A, Meyer CJ, O'Grady M, Pergola PE, Reisman SA, Wigley WC, Chertow GM. Effects of Bardoxolone Methyl on Hepatic Enzymes in Patients with Type 2 Diabetes Mellitus and Stage 4 CKD. Clin Transl Sci 2020; 14:299-309. [PMID: 32860734 PMCID: PMC7877861 DOI: 10.1111/cts.12868] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 07/16/2020] [Indexed: 12/29/2022] Open
Abstract
In a multinational placebo‐controlled phase III clinical trial in 2,185 patients with type 2 diabetes mellitus and stage 4 chronic kidney disease, treatment with the Nrf2 activator bardoxolone methyl increased estimated glomerular filtration rate, a measure of kidney function, but also resulted in increases in serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), and gamma glutamyl transferase. These increases in liver enzyme level(s) were maximal after 4 weeks of treatment and reversible, trending back toward baseline through week 48. Total bilirubin concentrations did not increase, and no cases met Hy’s Law criteria, although two subjects had ALT concentrations that exceeded 10 × the upper limit of the population reference range leading to discontinuation of treatment. Animal and cell culture experiments suggested that the increases in ALT and AST induced by bardoxolone methyl may be related to its pharmacological activity. Bardoxolone methyl significantly induced the mRNA expression of ALT and AST isoforms in cultured cells. Expression of ALT and AST isoforms in liver and kidney also positively correlated with Nrf2 status in mice. Overall, these data suggest that the increases in ALT and AST observed clinically were, at least in part, related to the pharmacological induction of aminotransferases via Nrf2 activation, rather than to any intrinsic form of hepatotoxicity.
Collapse
Affiliation(s)
- James H Lewis
- Division of Gastroenterology & Hepatology, Georgetown University Hospital, Washington, District of Columbia, USA
| | - Michel Jadoul
- Department of Nephrology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | - Glenn M Chertow
- Division of Nephrology, Stanford University School of Medicine, Palo Alto, California, USA
| |
Collapse
|
16
|
Infante B, Franzin R, Madio D, Calvaruso M, Maiorano A, Sangregorio F, Netti GS, Ranieri E, Gesualdo L, Castellano G, Stallone G. Molecular Mechanisms of AKI in the Elderly: From Animal Models to Therapeutic Intervention. J Clin Med 2020; 9:jcm9082574. [PMID: 32784471 PMCID: PMC7464895 DOI: 10.3390/jcm9082574] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 07/21/2020] [Accepted: 08/05/2020] [Indexed: 12/17/2022] Open
Abstract
Acute kidney injury (AKI), a critical syndrome characterized by a sudden reduction of renal function, is a common disorder among elderly patients particularly in Intensive Care Unit (ICU). AKI is closely associated with both short- and long-term mortality and length of hospital stay and is considered a predictor of chronic kidney disease (CKD). Specific hemodynamic, metabolic, and molecular changes lead to increased susceptibility to injury in the aged kidney; therefore, certain causes of AKI such as the prerenal reduction in renal perfusion or vascular obstructive conditions are more common in the elderly; moreover, AKI is often multifactorial and iatrogenic. Older patients present several comorbidities (diabetes, hypertension, heart failure) and are exposed to multiple medical interventions such as the use of nephrotoxic contrasts media and medications, which can also trigger AKI. Considering the emerging relevance of this condition, prevention and treatment of AKI in the elderly should be crucial in the internist and emergency setting. This review article summarizes the incidence, the risk factors, the pathophysiology, the molecular mechanisms and the strategies of prevention and treatment of AKI in elderly patients.
Collapse
Affiliation(s)
- Barbara Infante
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto Luigi 251, 71122 Foggia, Italy; (B.I.); (D.M.); (A.M.); (F.S.); (G.S.)
| | - Rossana Franzin
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, 70124 Bari, Italy; (R.F.); (L.G.)
| | - Desirèe Madio
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto Luigi 251, 71122 Foggia, Italy; (B.I.); (D.M.); (A.M.); (F.S.); (G.S.)
| | - Martina Calvaruso
- Nephrology, Dialysis and Transplantation Unit, Department of Biomedical Sciences, University of Foggia, 71122 Foggia, Italy;
| | - Annamaria Maiorano
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto Luigi 251, 71122 Foggia, Italy; (B.I.); (D.M.); (A.M.); (F.S.); (G.S.)
| | - Fabio Sangregorio
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto Luigi 251, 71122 Foggia, Italy; (B.I.); (D.M.); (A.M.); (F.S.); (G.S.)
| | - Giuseppe Stefano Netti
- Clinical Pathology, Department of Surgical and Medical Sciences, University of Foggia, Viale Pinto Luigi 251, 71122 Foggia, Italy; (G.S.N.); (E.R.)
| | - Elena Ranieri
- Clinical Pathology, Department of Surgical and Medical Sciences, University of Foggia, Viale Pinto Luigi 251, 71122 Foggia, Italy; (G.S.N.); (E.R.)
| | - Loreto Gesualdo
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, 70124 Bari, Italy; (R.F.); (L.G.)
| | - Giuseppe Castellano
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto Luigi 251, 71122 Foggia, Italy; (B.I.); (D.M.); (A.M.); (F.S.); (G.S.)
- Correspondence: ; Tel.: +39-088-173-2610; Fax: +39-088-173-6001
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto Luigi 251, 71122 Foggia, Italy; (B.I.); (D.M.); (A.M.); (F.S.); (G.S.)
| |
Collapse
|
17
|
Common Drug Pipelines for the Treatment of Diabetic Nephropathy and Hepatopathy: Can We Kill Two Birds with One Stone? Int J Mol Sci 2020; 21:ijms21144939. [PMID: 32668632 PMCID: PMC7404115 DOI: 10.3390/ijms21144939] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/30/2020] [Accepted: 06/30/2020] [Indexed: 12/17/2022] Open
Abstract
Type 2 diabetes (T2D) is associated with diabetic nephropathy as well as nonalcoholic steatohepatitis (NASH), which can be called "diabetic hepatopathy or diabetic liver disease". NASH, a severe form of nonalcoholic fatty disease (NAFLD), can sometimes progress to cirrhosis, hepatocellular carcinoma and hepatic failure. T2D patients are at higher risk for liver-related mortality compared with the nondiabetic population. NAFLD is closely associated with chronic kidney disease (CKD) or diabetic nephropathy according to cross-sectional and longitudinal studies. Simultaneous kidney liver transplantation (SKLT) is dramatically increasing in the United States, because NASH-related cirrhosis often complicates end-stage renal disease. Growing evidence suggests that NAFLD and CKD share common pathogenetic mechanisms and potential therapeutic targets. Glucagon-like peptide 1 (GLP-1) receptor agonists and sodium-glucose cotransporter 2 (SGLT2) inhibitors are expected to ameliorate NASH and diabetic nephropathy/CKD. There are no approved therapies for NASH, but a variety of drug pipelines are now under development. Several agents of them can also ameliorate diabetic nephropathy/CKD, including peroxisome proliferator-activated receptors agonists, apoptosis signaling kinase 1 inhibitor, nuclear factor-erythroid-2-related factor 2 activator, C-C chemokine receptor types 2/5 antagonist and nonsteroidal mineral corticoid receptor antagonist. This review focuses on common drug pipelines in the treatment of diabetic nephropathy and hepatopathy.
Collapse
|
18
|
Wu J, Sun X, Jiang Z, Jiang J, Xu L, Tian A, Sun X, Meng H, Li Y, Huang W, Jia Y, Wu H. Protective role of NRF2 in macrovascular complications of diabetes. J Cell Mol Med 2020; 24:8903-8917. [PMID: 32628815 PMCID: PMC7417734 DOI: 10.1111/jcmm.15583] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/05/2020] [Accepted: 05/07/2020] [Indexed: 02/07/2023] Open
Abstract
Macrovascular complications develop in over a half of the diabetic individuals, resulting in high morbidity and mortality. This poses a severe threat to public health and a heavy burden to social economy. It is therefore important to develop effective approaches to prevent or slow down the pathogenesis and progression of macrovascular complications of diabetes (MCD). Oxidative stress is a major contributor to MCD. Nuclear factor (erythroid‐derived 2)‐like 2 (NRF2) governs cellular antioxidant defence system by activating the transcription of various antioxidant genes, combating diabetes‐induced oxidative stress. Accumulating experimental evidence has demonstrated that NRF2 activation protects against MCD. Structural inhibition of Kelch‐like ECH‐associated protein 1 (KEAP1) is a canonical way to activate NRF2. More recently, novel approaches, such as activation of the Nfe2l2 gene transcription, decreasing KEAP1 protein level by microRNA‐induced degradation of Keap1 mRNA, prevention of proteasomal degradation of NRF2 protein and modulation of other upstream regulators of NRF2, have emerged in prevention of MCD. This review provides a brief introduction of the pathophysiology of MCD and the role of oxidative stress in the pathogenesis of MCD. By reviewing previous work on the activation of NRF2 in MCD, we summarize strategies to activate NRF2, providing clues for future intervention of MCD. Controversies over NRF2 activation and future perspectives are also provided in this review.
Collapse
Affiliation(s)
- Junduo Wu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, China
| | - Xiaodan Sun
- Intensive Care Unit, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ziping Jiang
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, China
| | - Jun Jiang
- Department of Neurosurgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Linlin Xu
- Department of Neurology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ao Tian
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xuechun Sun
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Huali Meng
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ying Li
- Department of Dermatology, Affiliated Hospital of Beihua University, Jilin, China
| | - Wenlin Huang
- School of Science and Technology, Georgia Gwinnett College, Lawrenceville, GA, USA
| | - Ye Jia
- Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Hao Wu
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
19
|
Balhorn R, Hartmann C, Schupp N. Aldosterone Induces DNA Damage and Activation of Nrf2 Mainly in Tubuli of Mouse Kidneys. Int J Mol Sci 2020; 21:ijms21134679. [PMID: 32630085 PMCID: PMC7370281 DOI: 10.3390/ijms21134679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 06/28/2020] [Indexed: 12/11/2022] Open
Abstract
Hypertensive patients have an increased risk of developing chronic kidney disease (CKD). Many of these patients have increased levels of the blood pressure regulating mineralocorticoid aldosterone. As a protection against aldosterone-induced damage, kidney cells can upregulate key regulators of the antioxidant defense, such as nuclear factor-erythroid-2-related factor 2 (Nrf2). In the present study aldosterone-induced kidney damage and Nrf2 activation in kidney cells of mice treated with three different concentrations of aldosterone for 4 weeks was localized. Increased albumin and neutrophil gelatinase-associated lipocalin (NGAL) in urine revealed an impaired kidney function of the aldosterone-infused mice. Localization of aldosterone-induced oxidative damage (in the form of DNA lesions) in specific kidney cells showed an increase in proximal tubuli and to an even greater extend in distal tubuli. Phosphorylated Nrf2 was increased in distal tubule cells after aldosterone-infusion. Nrf2 activation in proximal tubuli or in glomeruli after aldosterone-treatment could not be observed. Nrf2 target genes and proteins analyzed, paradoxically, showed a downregulation in the whole kidney. Aldosterone-treated mice exhibited an increased kidney injury and DNA damage in distal and proximal tubuli. Nrf2 seemed only to be specifically activated in distal tubule cells, where we also detected the highest amount of oxidative damage.
Collapse
|
20
|
Nezu M, Suzuki N. Roles of Nrf2 in Protecting the Kidney from Oxidative Damage. Int J Mol Sci 2020; 21:ijms21082951. [PMID: 32331329 PMCID: PMC7215459 DOI: 10.3390/ijms21082951] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/16/2020] [Accepted: 04/21/2020] [Indexed: 12/12/2022] Open
Abstract
Over 10% of the global population suffers from kidney disease. However, only kidney replacement therapies, which burden medical expenses, are currently effective in treating kidney disease. Therefore, elucidating the complicated molecular pathology of kidney disease is an urgent priority for developing innovative therapeutics for kidney disease. Recent studies demonstrated that intertwined renal vasculature often causes ischemia-reperfusion injury (IRI), which generates oxidative stress, and that the accumulation of oxidative stress is a common pathway underlying various types of kidney disease. We reported that activating the antioxidative transcription factor Nrf2 in renal tubules in mice with renal IRI effectively mitigates tubular damage and interstitial fibrosis by inducing the expression of genes related to cytoprotection against oxidative stress. Additionally, since the kidney performs multiple functions beyond blood purification, renoprotection by Nrf2 activation is anticipated to lead to various benefits. Indeed, our experiments indicated the possibility that Nrf2 activation mitigates anemia, which is caused by impaired production of the erythroid growth factor erythropoietin from injured kidneys, and moderates organ damage worsened by anemic hypoxia. Clinical trials investigating Nrf2-activating compounds in kidney disease patients are ongoing, and beneficial effects are being obtained. Thus, Nrf2 activators are expected to emerge as first-in-class innovative medicine for kidney disease treatment.
Collapse
Affiliation(s)
- Masahiro Nezu
- Department of Endocrinology and Diabetes, Yamanashi Prefectural Central Hospital, Fujimi 1-1-1, Kofu, Japan;
- Division of Oxygen Biology, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Aoba-ku, Sendai, Japan
| | - Norio Suzuki
- Division of Oxygen Biology, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Aoba-ku, Sendai, Japan
- Correspondence: ; Tel.: +81-22-717-8206
| |
Collapse
|
21
|
Borella R, Forti L, Gibellini L, De Gaetano A, De Biasi S, Nasi M, Cossarizza A, Pinti M. Synthesis and Anticancer Activity of CDDO and CDDO-Me, Two Derivatives of Natural Triterpenoids. Molecules 2019; 24:molecules24224097. [PMID: 31766211 PMCID: PMC6891335 DOI: 10.3390/molecules24224097] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/25/2019] [Accepted: 11/10/2019] [Indexed: 01/05/2023] Open
Abstract
Triterpenoids are natural compounds synthesized by plants through cyclization of squalene, known for their weak anti-inflammatory activity. 2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oic acid (CDDO), and its C28 modified derivative, methyl-ester (CDDO-Me, also known as bardoxolone methyl), are two synthetic derivatives of oleanolic acid, synthesized more than 20 years ago, in an attempt to enhance the anti-inflammatory behavior of the natural compound. These molecules have been extensively investigated for their strong ability to exert antiproliferative, antiangiogenic, and antimetastatic activities, and to induce apoptosis and differentiation in cancer cells. Here, we discuss the chemical properties of natural triterpenoids, the pathways of synthesis and the biological effects of CDDO and its derivative CDDO-Me. At nanomolar doses, CDDO and CDDO-Me have been shown to protect cells and tissues from oxidative stress by increasing the transcriptional activity of the nuclear factor (erythroid-derived 2)-like 2 (Nrf2). At doses higher than 100 nM, CDDO and CDDO-Me are able to modulate the differentiation of a variety of cell types, both tumor cell lines or primary culture cell, while at micromolar doses these compounds exert an anticancer effect in multiple manners; by inducing extrinsic or intrinsic apoptotic pathways, or autophagic cell death, by inhibiting telomerase activity, by disrupting mitochondrial functions through Lon protease inhibition, and by blocking the deubiquitylating enzyme USP7. CDDO-Me demonstrated its efficacy as anticancer drugs in different mouse models, and versus several types of cancer. Several clinical trials have been started in humans for evaluating CDDO-Me efficacy as anticancer and anti-inflammatory drug; despite promising results, significant increase in heart failure events represented an obstacle for the clinical use of CDDO-Me.
Collapse
Affiliation(s)
- Rebecca Borella
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (R.B.); (L.F.); (A.D.G.)
| | - Luca Forti
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (R.B.); (L.F.); (A.D.G.)
| | - Lara Gibellini
- Department of Medical and Surgical Sciences of Children and Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (L.G.); (S.D.B.)
| | - Anna De Gaetano
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (R.B.); (L.F.); (A.D.G.)
| | - Sara De Biasi
- Department of Medical and Surgical Sciences of Children and Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (L.G.); (S.D.B.)
| | - Milena Nasi
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (M.N.); (A.C.)
| | - Andrea Cossarizza
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (M.N.); (A.C.)
| | - Marcello Pinti
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (R.B.); (L.F.); (A.D.G.)
- Correspondence: ; Tel.: +39 059 205 5386; Fax: +39 059 205 5426
| |
Collapse
|
22
|
Song MK, Lee JH, Ryoo IG, Lee SH, Ku SK, Kwak MK. Bardoxolone ameliorates TGF-β1-associated renal fibrosis through Nrf2/Smad7 elevation. Free Radic Biol Med 2019; 138:33-42. [PMID: 31059771 DOI: 10.1016/j.freeradbiomed.2019.04.033] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 04/24/2019] [Accepted: 04/25/2019] [Indexed: 01/05/2023]
Abstract
Transforming growth factor-β (TGF-β) is a potent pathogenic factor of renal injury through the upregulation of extracellular matrix (ECM) expression and facilitation of renal fibrosis. Nuclear factor erythroid 2-like 2 (Nfe2l2; Nrf2), a master regulator of antioxidant and detoxifying systems, is mainly controlled by the binding with cytosolic protein Kelch-like ECH-associated protein 1 (Keap1) and subsequent proteasomal degradation. The protective effect of Nrf2 on renal injury has been attributed to its antioxidant role, where it aids in coping with oxidative stress-associated progression of renal disease. In this study, we investigated the effect of Nrf2 activation on ECM production and TGF-β/Smad signaling using Keap1-silenced MES-13 cells (a genetic glomerular mesangial cell model with Nrf2 overexpression). The TGF-β1-inducible expression of fibronectin and α-smooth muscle actin (α-Sma) was suppressed and Smad2/3 phosphorylation was blocked in Nrf2-high mesangial cells as compared with that in control cells. Notably, in these Nrf2-high mesangial cells, levels of TGF-β1 receptor 1 (TβR1) were substantially diminished, and the protein levels of Smad7, an inhibitor TGF-β1/Smad signaling, were increased. Nrf2-mediated Smad7 elevation and its anti-fibrotic role in Keap1-silenced cells were confirmed by studies with Nrf2-or Smad7-silencing. As a molecular link for Smad7 elevation in Nrf2-high cells, the reduction of Smad-ubiquitination-regulatory factor 1 (Smurf1), an E3 ubiquitin ligase for Smad7, was notable. Silencing of Smurf1 increased Smad7 in the control mesangial cells; however, forced expression of Smurf1 repressed Smad7 levels in Keap1-silenced cells. Additionally, we demonstrate that bardoxolone (BARD; CDDO-methyl), a pharmacological activator of Nrf2, increased Smad7 levels and attenuated TGF-β/Smad/ECM expression in MES-13. Moreover, in an aristolochic acid (AA)-mediated nephropathy mouse model, the renal expression of Nrf2 and Smad7 was elevated by BARD treatment, and AA-induced tubular necrosis and interstitial fibrosis were substantially ameliorated by BARD. Collectively, these results indicate that the Nrf2-Smad7 axis plays a key role in the protection of TGF-β-induced renal fibrosis, and further suggest a novel molecular mechanism of beneficial effect of BARD on renal disease.
Collapse
Affiliation(s)
- Min-Kyun Song
- Department of Pharmacy and BK21PLUS Team for Creative Leader Program for Pharmacomics-based Future Pharmacy, Graduate School of The Catholic University of Korea, 43 Jibong-ro, Bucheon, Gyeonggi-do, 14662, Republic of Korea
| | - Jin-Hee Lee
- Integrated Research Institute for Pharmaceutical Sciences, The Catholic University of Korea, Republic of Korea
| | - In-Geun Ryoo
- Integrated Research Institute for Pharmaceutical Sciences, The Catholic University of Korea, Republic of Korea
| | - Sang-Hwan Lee
- Department of Pharmacy and BK21PLUS Team for Creative Leader Program for Pharmacomics-based Future Pharmacy, Graduate School of The Catholic University of Korea, 43 Jibong-ro, Bucheon, Gyeonggi-do, 14662, Republic of Korea
| | - Sae-Kwang Ku
- College of Korean Medicine, Daegu Haany University, Gyeongsan, Gyeonsangbuk-do, 712-715, Republic of Korea
| | - Mi-Kyoung Kwak
- Department of Pharmacy and BK21PLUS Team for Creative Leader Program for Pharmacomics-based Future Pharmacy, Graduate School of The Catholic University of Korea, 43 Jibong-ro, Bucheon, Gyeonggi-do, 14662, Republic of Korea; Integrated Research Institute for Pharmaceutical Sciences, The Catholic University of Korea, Republic of Korea; College of Pharmacy, The Catholic University of Korea, Republic of Korea.
| |
Collapse
|
23
|
Rizk DV, Silva AL, Pergola PE, Toto R, Warnock DG, Chin MP, Goldsberry A, O'Grady M, Meyer CJ, McCullough PA. Effects of Bardoxolone Methyl on Magnesium in Patients with Type 2 Diabetes Mellitus and Chronic Kidney Disease. Cardiorenal Med 2019; 9:316-325. [PMID: 31170712 DOI: 10.1159/000500612] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 04/18/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Treatment with bardoxolone methyl (Bard) in a multinational phase 3 trial, Bardoxolone Methyl Evaluation in Patients with Chronic Kidney Disease and Type 2 Diabetes (BEACON), resulted in increases in estimated glomerular filtration rate with concurrent reductions in serum magnesium. We analyzed data from several trials to characterize reductions in magnesium with Bard. METHODS BEACON randomized patients (n = 2,185) with type 2 diabetes (T2DM) and stage 4 chronic kidney disease (CKD) 1:1 to receive Bard (20 mg) or placebo once daily. In a separate open-label study, magnesium levels from 24-hour urine and sublingual epithelial cell samples were analyzed in patients with stage 3b-4 CKD and T2DM administered 20 mg Bard for 56 consecutive days. RESULTS BEACON patients randomized to Bard experienced significant reductions in serum magnesium from baseline relative to patients randomized to placebo (-0.17 mEq/L, 95% CI -0.18 to -0.60 mEq/L; p < 0.001). A separate study showed intracellular and urinary magnesium levels were unchanged with Bard treatment. CONCLUSIONS Bard treatment results in significant decreases in serum magnesium that are not associated with changes in intracellular and urinary magnesium levels, indicating that magnesium decreases are not due to renal magnesium wasting or total body magnesium depletion. Importantly, the decreases in serum magnesium with Bard are not associated with adverse effects on QT interval.
Collapse
Affiliation(s)
- Dana V Rizk
- Department of Medicine, Nephrology Division, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Arnold L Silva
- Boise Kidney and Hypertension Institute, Meridian, Idaho, USA
| | | | - Robert Toto
- Internal Medicine and Clinical Science, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - David G Warnock
- Department of Medicine, Nephrology Division, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Melanie P Chin
- Product Development, Reata Pharmaceuticals, Irving, Texas, USA
| | | | - Megan O'Grady
- Product Development, Reata Pharmaceuticals, Irving, Texas, USA
| | - Colin J Meyer
- Product Development, Reata Pharmaceuticals, Irving, Texas, USA
| | - Peter A McCullough
- Baylor Heart and Vascular Institute, Baylor Jack and Jane Hamilton Heart and Vascular Hospital, The Heart Hospital, Baylor University Medical Center, Dallas, Texas, USA,
| |
Collapse
|
24
|
Fernandez-Fernandez B, Fernandez-Prado R, Górriz JL, Martinez-Castelao A, Navarro-González JF, Porrini E, Soler MJ, Ortiz A. Canagliflozin and Renal Events in Diabetes with Established Nephropathy Clinical Evaluation and Study of Diabetic Nephropathy with Atrasentan: what was learned about the treatment of diabetic kidney disease with canagliflozin and atrasentan? Clin Kidney J 2019; 12:313-321. [PMID: 31198532 PMCID: PMC6543971 DOI: 10.1093/ckj/sfz070] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Indexed: 12/11/2022] Open
Abstract
In April 2019, two major Phase 3 randomized clinical trials were published that assessed primary renal outcomes in diabetic kidney disease (DKD) in type 2 diabetes mellitus (T2DM). The Canagliflozin and Renal Events in Diabetes with Established Nephropathy Clinical Evaluation (CREDENCE) tested an already available antidiabetic drug, canagliflozin, and the Study of Diabetic Nephropathy with Atrasentan (SONAR) tested a novel molecule, the endothelin-1 receptor blocker atrasentan, both on top of renin-angiotensin system blockade. Both trials demonstrated significant nephroprotection in patients with overt DKD (albuminuria >300 mg/g urinary creatinine) for combined primary endpoints of end-stage kidney disease (ESKD), doubling of serum creatinine or death from renal or cardiovascular causes in CREDENCE {hazard ratio [HR] 0.70 [95% confidence interval (CI) 0.59-0.82]} and ESKD and doubling of serum creatinine in SONAR [HR 0.65 (95% CI 0.49-0.88)]. Canagliflozin also decreased the secondary renal endpoint ESKD, doubling of serum creatinine or renal death [HR 0.66 (95% CI 0.53-0.81)], which was similar in nature and impact to the primary endpoint in SONAR. In addition, canagliflozin decreased a secondary endpoint of cardiovascular death or hospitalization for heart failure [HR 0.69 (95% CI 0.57-0.83)], whereas atrasentan had no significant impact on a secondary cardiovascular composite endpoint or on hospital admissions for heart failure and, despite restrictive exclusion criteria, there was a non-significant trend towards more frequent episodes of heart failure. Based on these results, canagliflozin will likely be approved for the indication of treating DKD in T2DM and the estimated glomerular filtration rate threshold for prescribing it will be lifted, whereas the future and place of atrasentan in the treatment of DKD remain unclear.
Collapse
Affiliation(s)
- Beatriz Fernandez-Fernandez
- IIS-Fundacion Jimenez Diaz UAM and School of Medicine, UAM, Madrid, Spain
- REDINREN, Madrid, Spain
- GEENDIAB, Barcelona, Spain
| | - Raul Fernandez-Prado
- IIS-Fundacion Jimenez Diaz UAM and School of Medicine, UAM, Madrid, Spain
- REDINREN, Madrid, Spain
- GEENDIAB, Barcelona, Spain
| | - Jose Luis Górriz
- GEENDIAB, Barcelona, Spain
- Hospital Clínico Universitario, Universitat de Valencia-INCLIVA, Valencia, Spain
| | | | - Juan F Navarro-González
- GEENDIAB, Barcelona, Spain
- Unidad de Investigación y Servicio de Nefrología, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Esteban Porrini
- GEENDIAB, Barcelona, Spain
- Instituto de Tecnologías Biomédicas, Hospital Universitario de Canarias, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| | - María José Soler
- GEENDIAB, Barcelona, Spain
- Nephrology Department, Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
- Nephrology Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
| | - Alberto Ortiz
- IIS-Fundacion Jimenez Diaz UAM and School of Medicine, UAM, Madrid, Spain
- REDINREN, Madrid, Spain
- GEENDIAB, Barcelona, Spain
| |
Collapse
|
25
|
Daenen K, Andries A, Mekahli D, Van Schepdael A, Jouret F, Bammens B. Oxidative stress in chronic kidney disease. Pediatr Nephrol 2019; 34:975-991. [PMID: 30105414 DOI: 10.1007/s00467-018-4005-4] [Citation(s) in RCA: 460] [Impact Index Per Article: 76.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 06/03/2018] [Accepted: 06/14/2018] [Indexed: 12/13/2022]
Abstract
Oxidative stress (OS), defined as disturbances in the pro-/antioxidant balance, is harmful to cells due to the excessive generation of highly reactive oxygen (ROS) and nitrogen (RNS) species. When the balance is not disturbed, OS has a role in physiological adaptations and signal transduction. However, an excessive amount of ROS and RNS results in the oxidation of biological molecules such as lipids, proteins, and DNA. Oxidative stress has been reported in kidney disease, due to both antioxidant depletions as well as increased ROS production. The kidney is a highly metabolic organ, rich in oxidation reactions in mitochondria, which makes it vulnerable to damage caused by OS, and several studies have shown that OS can accelerate kidney disease progression. Also, in patients at advanced stages of chronic kidney disease (CKD), increased OS is associated with complications such as hypertension, atherosclerosis, inflammation, and anemia. In this review, we aim to describe OS and its influence on CKD progression and its complications. We also discuss the potential role of various antioxidants and pharmacological agents, which may represent potential therapeutic targets to reduce OS in both pediatric and adult CKD patients.
Collapse
Affiliation(s)
- Kristien Daenen
- Department of Microbiology and Immunology, Laboratory of Nephrology, KU Leuven - University of Leuven, 3000, Leuven, Belgium.
- Department of Nephrology, Dialysis and Renal Transplantation, University Hospitals Leuven, 3000, Leuven, Belgium.
- Department of Nephrology, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Asmin Andries
- Department of Pharmaceutical and Pharmacological Sciences, Pharmaceutical Analysis, KU Leuven - University of Leuven, 3000, Leuven, Belgium
| | - Djalila Mekahli
- Department of Development and Regeneration, Laboratory of Pediatrics, PKD Group, KU Leuven - University of Leuven, 3000, Leuven, Belgium
- Department of Pediatric Nephrology, University Hospitals Leuven, 3000, Leuven, Belgium
| | - Ann Van Schepdael
- Department of Pharmaceutical and Pharmacological Sciences, Pharmaceutical Analysis, KU Leuven - University of Leuven, 3000, Leuven, Belgium
| | - François Jouret
- Division of Nephrology, Department of Internal Medicine, University of Liège Hospital (ULg CHU), Liège, Belgium
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), Cardiovascular Science, University of Liège, Liège, Belgium
| | - Bert Bammens
- Department of Microbiology and Immunology, Laboratory of Nephrology, KU Leuven - University of Leuven, 3000, Leuven, Belgium
- Department of Nephrology, Dialysis and Renal Transplantation, University Hospitals Leuven, 3000, Leuven, Belgium
| |
Collapse
|
26
|
Effect of bardoxolone methyl on the urine albumin-to-creatinine ratio in patients with type 2 diabetes and stage 4 chronic kidney disease. Kidney Int 2019; 96:1030-1036. [PMID: 31377056 DOI: 10.1016/j.kint.2019.04.027] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 04/01/2019] [Accepted: 04/19/2019] [Indexed: 12/29/2022]
Abstract
Bardoxolone methyl attenuates inflammation by inducing nuclear factor erythroid-derived 2-related factor 2 and suppressing nuclear factor κB. The Bardoxolone Methyl Evaluation in Patients With Chronic Kidney Disease and Type 2 Diabetes (BEACON) trial was a phase 3 placebo-controlled, randomized, double-blind, parallel-group, international, multicenter trial in 2185 patients with type 2 diabetes mellitus and stage 4 chronic kidney disease. BEACON was terminated because of safety concerns, largely related to a significant increase in early heart failure events in patients randomized to bardoxolone methyl. Bardoxolone methyl resulted in increased estimated glomerular filtration rate (eGFR) and urine albumin-to-creatinine ratio. Herein, we present post hoc analyses characterizing the relation between the urine albumin-to-creatinine ratio and eGFR. The urine albumin-to-creatinine ratio and eGFR were assessed every four weeks through Week 12, followed by assessments every eight weeks thereafter, and 4 weeks after the last dose of bardoxolone methyl was administered. The initial increases in urine albumin-to-creatinine ratio observed in patients randomized to bardoxolone methyl were attenuated after six months. Multivariable regression analysis identified baseline eGFR and eGFR over time as the dominant factors associated with change in the urine albumin-to-creatinine ratio. Relative to placebo, bardoxolone methyl resulted in a significant decrease in albuminuria when indexed to eGFR (least-squared means: -0.035 [95% confidence interval -0.031 to -0.039]). Thus, among patients with type 2 diabetes mellitus and stage 4 chronic kidney disease treated with bardoxolone methyl, changes in albuminuria are directly related to changes in eGFR, challenging the conventional construct that increases in albuminuria universally reflect kidney injury and denote harm.
Collapse
|
27
|
Chertow GM, Appel GB, Block GA, Chin MP, Coyne DW, Goldsberry A, Kalantar-Zadeh K, Meyer CJ, Molitch ME, Pergola PE, Raskin P, Silva AL, Spinowitz B, Sprague SM, Rossing P. Effects of bardoxolone methyl on body weight, waist circumference and glycemic control in obese patients with type 2 diabetes mellitus and stage 4 chronic kidney disease. J Diabetes Complications 2018; 32:1113-1117. [PMID: 30318163 DOI: 10.1016/j.jdiacomp.2018.09.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/04/2018] [Accepted: 09/09/2018] [Indexed: 02/05/2023]
Abstract
AIMS Obesity is associated with progression of chronic kidney disease (CKD). Treatment with bardoxolone methyl in a multinational phase 3 trial, Bardoxolone Methyl Evaluation in Patients with Chronic Kidney Disease and Type 2 Diabetes (BEACON), resulted in increases in estimated glomerular filtration rate (eGFR) with concurrent reductions in body weight. We performed post-hoc analyses to further characterize reductions in body weight with bardoxolone methyl. METHODS Eligible patients with type 2 diabetes (T2DM) and CKD stage 4 (eGFR 15 to <30 mL/min/1.73 m2) were randomized 1:1 to receive once-daily oral dose of bardoxolone methyl (20 mg) or placebo. RESULTS BEACON enrolled 2185 patients. Patients randomized to bardoxolone methyl experienced significant reductions in body weight from baseline relative to patients randomized to placebo (-5.7 kg; 95% CI: -6.0 to -5.3 kg; p < 0.001). In patients randomized to bardoxolone methyl, rate and magnitude of body weight loss were proportional to baseline BMI. Bardoxolone methyl resulted in significant reductions in waist circumference and improved glycemic control. CONCLUSIONS Bardoxolone methyl resulted in significant weight loss in a generally obese patient population with T2DM and stage 4 CKD, with the magnitude and rate dependent on baseline BMI.
Collapse
Affiliation(s)
- Glenn M Chertow
- Stanford University School of Medicine, Palo Alto, CA 94304, United States.
| | - Gerald B Appel
- Columbia University Medical Center, Glomerular Kidney Disease Center, New York, NY 10032, United States.
| | | | - Melanie P Chin
- Reata Pharmaceuticals, Product Development, Irving, TX 75063, United States.
| | - Daniel W Coyne
- Washington University, Division of Nephrology, St. Louis, MO 63110, United States.
| | - Angie Goldsberry
- Reata Pharmaceuticals, Product Development, Irving, TX 75063, United States.
| | | | - Colin J Meyer
- Reata Pharmaceuticals, Product Development, Irving, TX 75063, United States.
| | - Mark E Molitch
- Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, United States.
| | | | - Philip Raskin
- University of Texas, Southwestern Medical Center, Division of Endocrinology, Dallas, TX 75390, United States.
| | - Arnold L Silva
- Boise Kidney and Hypertension Institute, Meridian, ID 83642, United States.
| | - Bruce Spinowitz
- New York Presbyterian Queens, Flushing, NY 11355, United States.
| | - Stuart M Sprague
- Northshore Medical University Health System - University of Chicago, Evanston, IL 60201, United States
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, University of Copenhagen, Copenhagen, DK-2820, Gentofte, Denmark.
| |
Collapse
|
28
|
Matzinger M, Fischhuber K, Heiss EH. Activation of Nrf2 signaling by natural products-can it alleviate diabetes? Biotechnol Adv 2018; 36:1738-1767. [PMID: 29289692 PMCID: PMC5967606 DOI: 10.1016/j.biotechadv.2017.12.015] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 12/19/2017] [Accepted: 12/26/2017] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes mellitus (DM) has reached pandemic proportions and effective prevention strategies are wanted. Its onset is accompanied by cellular distress, the nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor boosting cytoprotective responses, and many phytochemicals activate Nrf2 signaling. Thus, Nrf2 activation by natural products could presumably alleviate DM. We summarize function, regulation and exogenous activation of Nrf2, as well as diabetes-linked and Nrf2-susceptible forms of cellular stress. The reported amelioration of insulin resistance, β-cell dysfunction and diabetic complications by activated Nrf2 as well as the status quo of Nrf2 in precision medicine for DM are reviewed.
Collapse
Affiliation(s)
- Manuel Matzinger
- University of Vienna, Department of Pharmacognosy, Althanstrasse 14, 1090 Vienna, Austria
| | - Katrin Fischhuber
- University of Vienna, Department of Pharmacognosy, Althanstrasse 14, 1090 Vienna, Austria
| | - Elke H Heiss
- University of Vienna, Department of Pharmacognosy, Althanstrasse 14, 1090 Vienna, Austria.
| |
Collapse
|
29
|
Hishikawa A, Hayashi K, Itoh H. Transcription Factors as Therapeutic Targets in Chronic Kidney Disease. Molecules 2018; 23:molecules23051123. [PMID: 29747407 PMCID: PMC6100497 DOI: 10.3390/molecules23051123] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/05/2018] [Accepted: 05/07/2018] [Indexed: 12/23/2022] Open
Abstract
The growing number of patients with chronic kidney disease (CKD) is recognized as an emerging problem worldwide. Recent studies have indicated that deregulation of transcription factors is associated with the onset or progression of kidney disease. Several clinical trials indicated that regression of CKD may be feasible via activation of the transcription factor nuclear factor erythroid-2 related factor 2 (Nrf2), which suggests that transcription factors may be potential drug targets for CKD. Agents stabilizing hypoxia-inducible factor (HIF), which may be beneficial for renal anemia and renal protection, are also now under clinical trial. Recently, we have reported that the transcription factor Kruppel-like factor 4 (KLF4) regulates the glomerular podocyte epigenome, and that the antiproteinuric effect of the renin–angiotensin system blockade may be partially mediated by KLF4. KLF4 is one of the Yamanaka factors that induces iPS cells and is reported to be involved in epigenetic remodeling. In this article, we summarize the transcription factors associated with CKD and particularly focus on the possibility of transcription factors being novel drug targets for CKD through epigenetic modulation.
Collapse
Affiliation(s)
- Akihito Hishikawa
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan.
| | - Kaori Hayashi
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan.
| | - Hiroshi Itoh
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan.
| |
Collapse
|
30
|
MDM2 controls NRF2 antioxidant activity in prevention of diabetic kidney disease. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:1034-1045. [PMID: 29704532 DOI: 10.1016/j.bbamcr.2018.04.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 04/15/2018] [Accepted: 04/23/2018] [Indexed: 01/03/2023]
Abstract
Oxidative stress and P53 contribute to the pathogenesis of diabetic kidney disease (DKD). Nuclear factor erythroid 2-related factor 2 (NRF2) is a master regulator of cellular antioxidant defense system, is negatively regulated by P53 and prevents DKD. Recent findings revealed an important role of mouse double minute 2 (MDM2) in protection against DKD. However, the mechanism remained unclear. We hypothesized that MDM2 enhances NRF2 antioxidant signaling in DKD given that MDM2 is a key negative regulator of P53. The MDM2 inhibitor nutlin3a elevated renal P53, inhibited NRF2 signaling and induced oxidative stress, inflammation, fibrosis, DKD-like renal pathology and albuminuria in the wild-type (WT) non-diabetic mice. These effects exhibited more prominently in nutlin3a-treated WT diabetic mice. Interestingly, nutlin3a failed to induce greater renal injuries in the Nrf2 knockout (KO) mice under both the diabetic and non-diabetic conditions, indicating that NRF2 predominantly mediates MDM2's action. On the contrary, P53 inhibition by pifithrin-α activated renal NRF2 signaling and the expression of Mdm2, and attenuated DKD in the WT diabetic mice, but not in the Nrf2 KO diabetic mice. In high glucose-treated mouse mesangial cells, P53 gene silencing completely abolished nutlin3a's inhibitory effect on NRF2 signaling. The present study demonstrates for the first time that MDM2 controls renal NRF2 antioxidant activity in DKD via inhibition of P53, providing MDM2 activation and P53 inhibition as novel strategies in the management of DKD.
Collapse
|
31
|
Lv W, Booz GW, Fan F, Wang Y, Roman RJ. Oxidative Stress and Renal Fibrosis: Recent Insights for the Development of Novel Therapeutic Strategies. Front Physiol 2018; 9:105. [PMID: 29503620 PMCID: PMC5820314 DOI: 10.3389/fphys.2018.00105] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 01/31/2018] [Indexed: 12/12/2022] Open
Abstract
Chronic kidney disease (CKD) is a significant worldwide healthcare problem. Regardless of the initial injury, renal fibrosis is the common final pathway leading to end stage renal disease. Although the underlying mechanisms are not fully defined, evidence indicates that besides inflammation, oxidative stress plays a crucial role in the etiology of renal fibrosis. Oxidative stress results from an imbalance between the production of free radicals that are often increased by inflammation and mitochondrial dysfunction, and reduced anti-oxidant defenses. Several studies have demonstrated that oxidative stress may occur secondary to activation of transforming growth factor β1 (TGF-β1) activity, consistent with its role to increase nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (Nox) activity. A number of other oxidative stress-related signal pathways have also been identified, such as nuclear factor erythroid-2 related factor 2 (Nrf2), the nitric oxide (NO)-cyclic guanosine monophosphate (cGMP)-cGMP-dependent protein kinase 1-phosphodiesterase (cGMP-cGK1-PDE) signaling pathway, and the peroxisome proliferator-activated receptor gamma (PPARγ) pathway. Several antioxidant and renoprotective agents, including cysteamine bitartrate, epoxyeicosatrienoic acids (EETs), and cytoglobin (Cygb) have demonstrated ameliorative effects on renal fibrosis in preclinical or clinical studies. The mechanism of action of many traditional Chinese medicines used to treat renal disorders is based on their antioxidant properties, which could form the basis for new therapeutic approaches. This review focuses on the signaling pathways triggered by oxidative stress that lead to renal fibrosis and provides an update on the development of novel anti-oxidant therapies for CKD.
Collapse
Affiliation(s)
- Wenshan Lv
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States.,Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - George W Booz
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Fan Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Yangang Wang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Richard J Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
32
|
Chin MP, Bakris GL, Block GA, Chertow GM, Goldsberry A, Inker LA, Heerspink HJL, O'Grady M, Pergola PE, Wanner C, Warnock DG, Meyer CJ. Bardoxolone Methyl Improves Kidney Function in Patients with Chronic Kidney Disease Stage 4 and Type 2 Diabetes: Post-Hoc Analyses from Bardoxolone Methyl Evaluation in Patients with Chronic Kidney Disease and Type 2 Diabetes Study. Am J Nephrol 2018; 47:40-47. [PMID: 29402767 DOI: 10.1159/000486398] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 12/19/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND Increases in measured inulin clearance, measured creatinine clearance, and estimated glomerular filtration rate (eGFR) have been observed with bardoxolone methyl in 7 studies enrolling approximately 2,600 patients with type 2 diabetes (T2D) and chronic kidney disease (CKD). The largest of these studies was Bardoxolone Methyl Evaluation in Patients with Chronic Kidney Disease and Type 2 Diabetes (BEACON), a multinational, randomized, double-blind, placebo-controlled phase 3 trial which enrolled patients with T2D and CKD stage 4. The BEACON trial was terminated after preliminary analyses showed that patients randomized to bardoxolone methyl experienced significantly higher rates of heart failure events. We performed post-hoc analyses to characterize changes in kidney function induced by bardoxolone methyl. METHODS Patients in -BEACON (n = 2,185) were randomized 1: 1 to receive once-daily bardoxolone methyl (20 mg) or placebo. We compared the effects of bardoxolone methyl and placebo on a post-hoc composite renal endpoint consisting of ≥30% decline from baseline in eGFR, eGFR <15 mL/min/1.73 m2, and end-stage renal disease (ESRD) events (provision of dialysis or kidney transplantation). RESULTS Consistent with prior studies, patients randomized to bardoxolone methyl experienced mean increases in eGFR that were sustained through study week 48. Moreover, increases in eGFR from baseline were sustained 4 weeks after cessation of treatment. Patients randomized to bardoxolone methyl were significantly less likely to experience the composite renal endpoint (hazards ratio 0.48 [95% CI 0.36-0.64]; p < 0.0001). CONCLUSIONS Bardoxolone methyl preserves kidney function and may delay the onset of ESRD in patients with T2D and stage 4 CKD.
Collapse
Affiliation(s)
| | | | | | - Glenn M Chertow
- Stanford University School of Medicine, Stanford, California, USA
| | | | | | - Hiddo J L Heerspink
- Department of Clinical Pharmacy and Pharmacology University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | | | | | | | - David G Warnock
- University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | |
Collapse
|
33
|
Barrett EJ, Liu Z, Khamaisi M, King GL, Klein R, Klein BEK, Hughes TM, Craft S, Freedman BI, Bowden DW, Vinik AI, Casellini CM. Diabetic Microvascular Disease: An Endocrine Society Scientific Statement. J Clin Endocrinol Metab 2017; 102:4343-4410. [PMID: 29126250 PMCID: PMC5718697 DOI: 10.1210/jc.2017-01922] [Citation(s) in RCA: 305] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 08/29/2017] [Indexed: 01/18/2023]
Abstract
Both type 1 and type 2 diabetes adversely affect the microvasculature in multiple organs. Our understanding of the genesis of this injury and of potential interventions to prevent, limit, or reverse injury/dysfunction is continuously evolving. This statement reviews biochemical/cellular pathways involved in facilitating and abrogating microvascular injury. The statement summarizes the types of injury/dysfunction that occur in the three classical diabetes microvascular target tissues, the eye, the kidney, and the peripheral nervous system; the statement also reviews information on the effects of diabetes and insulin resistance on the microvasculature of skin, brain, adipose tissue, and cardiac and skeletal muscle. Despite extensive and intensive research, it is disappointing that microvascular complications of diabetes continue to compromise the quantity and quality of life for patients with diabetes. Hopefully, by understanding and building on current research findings, we will discover new approaches for prevention and treatment that will be effective for future generations.
Collapse
Affiliation(s)
- Eugene J. Barrett
- Division of Endocrinology, Department of Medicine, University of Virginia, Charlottesville, Virginia 22908
| | - Zhenqi Liu
- Division of Endocrinology, Department of Medicine, University of Virginia, Charlottesville, Virginia 22908
| | - Mogher Khamaisi
- Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts 02215
| | - George L. King
- Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts 02215
| | - Ronald Klein
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53705
| | - Barbara E. K. Klein
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53705
| | - Timothy M. Hughes
- Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Suzanne Craft
- Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Barry I. Freedman
- Divisions of Nephrology and Endocrinology, Department of Internal Medicine, Centers for Diabetes Research, and Center for Human Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Donald W. Bowden
- Divisions of Nephrology and Endocrinology, Department of Internal Medicine, Centers for Diabetes Research, and Center for Human Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Aaron I. Vinik
- EVMS Strelitz Diabetes Center, Eastern Virginia Medical Center, Norfolk, Virginia 23510
| | - Carolina M. Casellini
- EVMS Strelitz Diabetes Center, Eastern Virginia Medical Center, Norfolk, Virginia 23510
| |
Collapse
|
34
|
Gleixner KV, Schneeweiss M, Eisenwort G, Berger D, Herrmann H, Blatt K, Greiner G, Byrgazov K, Hoermann G, Konopleva M, Waliul I, Cumaraswamy AA, Gunning PT, Maeda H, Moriggl R, Deininger M, Lion T, Andreeff M, Valent P. Combined targeting of STAT3 and STAT5: a novel approach to overcome drug resistance in chronic myeloid leukemia. Haematologica 2017; 102:1519-1529. [PMID: 28596283 PMCID: PMC5685220 DOI: 10.3324/haematol.2016.163436] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 06/07/2017] [Indexed: 12/14/2022] Open
Abstract
In chronic myeloid leukemia, resistance against BCR-ABL1 tyrosine kinase inhibitors can develop because of BCR-ABL1 mutations, activation of additional pro-oncogenic pathways, and stem cell resistance. Drug combinations covering a broad range of targets may overcome resistance. CDDO-Me (bardoxolone methyl) is a drug that inhibits the survival of leukemic cells by targeting different pro-survival molecules, including STAT3. We found that CDDO-Me inhibits proliferation and survival of tyrosine kinase inhibitor-resistant BCR-ABL1+ cell lines and primary leukemic cells, including cells harboring BCR-ABL1T315I or T315I+ compound mutations. Furthermore, CDDO-Me was found to block growth and survival of CD34+/CD38− leukemic stem cells (LSC). Moreover, CDDO-Me was found to produce synergistic growth-inhibitory effects when combined with BCR-ABL1 tyrosine kinase inhibitors. These drug-combinations were found to block multiple signaling cascades and molecules, including STAT3 and STAT5. Furthermore, combined targeting of STAT3 and STAT5 by shRNA and STAT5-targeting drugs also resulted in synergistic growth-inhibition, pointing to a new efficient concept of combinatorial STAT3 and STAT5 inhibition. However, CDDO-Me was also found to increase the expression of heme-oxygenase-1, a heat-shock-protein that triggers drug resistance and cell survival. We therefore combined CDDO-Me with the heme-oxygenase-1 inhibitor SMA-ZnPP, which also resulted in synergistic growth-inhibitory effects. Moreover, SMA-ZnPP was found to sensitize BCR-ABL1+ cells against the combination ‘CDDO-Me+ tyrosine kinase inhibitor’. Together, combined targeting of STAT3, STAT5, and heme-oxygenase-1 overcomes resistance in BCR-ABL1+ cells, including stem cells and highly resistant sub-clones expressing BCR-ABL1T315I or T315I-compound mutations. Whether such drug-combinations are effective in tyrosine kinase inhibitor-resistant patients with chronic myeloid leukemia remains to be elucidated.
Collapse
Affiliation(s)
- Karoline V Gleixner
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Austria .,Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Austria
| | | | - Gregor Eisenwort
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Austria
| | - Daniela Berger
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Austria
| | - Harald Herrmann
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Austria.,Department of Radiation Therapy, Medical University of Vienna, Austria
| | - Katharina Blatt
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Austria
| | - Georg Greiner
- Department of Laboratory Medicine, Medical University of Vienna, Austria
| | | | - Gregor Hoermann
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Austria.,Department of Laboratory Medicine, Medical University of Vienna, Austria
| | - Marina Konopleva
- Department of Leukemia, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Islam Waliul
- Institute of Drug Delivery Sciences, Sojo University, Kumamoto and BioDynamics Research Laboratory, Kumamoto, Japan
| | | | | | - Hiroshi Maeda
- Institute of Drug Delivery Sciences, Sojo University, Kumamoto and BioDynamics Research Laboratory, Kumamoto, Japan
| | - Richard Moriggl
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria.,Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
| | - Michael Deininger
- Division of Hematology and Hematologic Malignancies, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Thomas Lion
- Children's Cancer Research Institute (CCRI), Vienna, Austria.,Department of Pediatrics, Medical University of Vienna, Austria
| | - Michael Andreeff
- Department of Leukemia, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Austria.,Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Austria
| |
Collapse
|
35
|
Refaat A, Pararasa C, Arif M, Brown JEP, Carmichael A, Ali SS, Sakurai H, Griffiths HR. Bardoxolone-methyl inhibits migration and metabolism in MCF7 cells. Free Radic Res 2017; 51:211-221. [PMID: 28277986 DOI: 10.1080/10715762.2017.1295452] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Bardoxolone-methyl (BAR) is reported to have anti-inflammatory, anti-proliferative and anti-fibrotic effects. BAR activates Nrf2 and may ameliorate oxidative stress through induction of antioxidant genes. However, off-target effects, probably concentration and NFkB-dependent, have limited the clinical use of BAR. Nrf2 regulates expression of antioxidant and mitochondrial genes and has been proposed as a target for both obesity and breast cancer. Therefore, we explored whether BAR can alter migration and proliferation in the MCF7 cell line and whether metabolic function is affected by BAR. Incubation with BAR caused a time-dependent migratory inhibition and an associated decrease in mitochondrial respiration. Both migratory and mitochondrial inhibition by BAR were further enhanced in the presence of fatty acids. In addition to the activation of Nrf2, BAR altered the expression of target mRNA GCLC and UCP1. After 24 h, BAR inhibited both glycolytic capacity, reserve (p < 0.05) and oxidative phosphorylation (p < 0.001) with an associated increase in mitochondrial ROS and loss of intracellular glutathione in MCF7 cells; however, impairment of mitochondrial activity was prevented by N-acetyl cysteine. The fatty acid, palmitate, increased mitochondrial ROS, impaired migration and oxidative phosphorylation but palmitate toxicity towards MCF7 could not be inhibited by N-acetyl cysteine suggesting that they exert effects through different pathways. BAR-activated AKT, induced DNA damage and inhibited cell proliferation. When the proteasome was inhibited, there was loss of BAR-mediated changes in p65 phosphorylation and SOD2 expression suggesting non-canonical NFkB signaling effects. These data suggest that BAR-induced ROS are important in inhibiting MCF7 migration and metabolism by negatively affecting glycolytic capacity and mitochondrial function.
Collapse
Affiliation(s)
- Alaa Refaat
- a Life & Health Sciences , Aston University , Birmingham , UK.,b Helmy Institute of Medical Sciences, Zewail City of Science and Technology , Giza , Egypt.,c Department of Cancer Cell Biology, Graduate School of Medicine and Pharmaceutical Sciences , University of Toyama , Toyama , Japan
| | | | - Muhammed Arif
- a Life & Health Sciences , Aston University , Birmingham , UK
| | - James E P Brown
- a Life & Health Sciences , Aston University , Birmingham , UK
| | | | - Sameh S Ali
- b Helmy Institute of Medical Sciences, Zewail City of Science and Technology , Giza , Egypt
| | - Hiroaki Sakurai
- c Department of Cancer Cell Biology, Graduate School of Medicine and Pharmaceutical Sciences , University of Toyama , Toyama , Japan
| | - Helen R Griffiths
- a Life & Health Sciences , Aston University , Birmingham , UK.,d Faculty of Health and Medical Sciences , University of Surrey , Guildford , UK
| |
Collapse
|
36
|
The nuclear factor (erythroid-derived 2)-like 2 (Nrf2) activator dh404 protects against diabetes-induced endothelial dysfunction. Cardiovasc Diabetol 2017; 16:33. [PMID: 28253885 PMCID: PMC5335831 DOI: 10.1186/s12933-017-0513-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 02/22/2017] [Indexed: 12/13/2022] Open
Abstract
Background Vascular dysfunction is a pivotal event in the development of diabetes-associated vascular disease. Increased inflammation and oxidative stress are major contributors to vascular dysfunction. Nrf2, a master regulator of several anti-oxidant genes and a suppressor of inflammatory NF-κB, has potential as a target to combat oxidative stress and inflammation. The aim of this study was to investigate the effects of a novel Nrf2 activator, the bardoxolone methyl derivative dh404, on endothelial function in vitro and in vivo. Methods dh404 at 3 mg/kg was administered to male Akita mice, an established diabetic mouse model of insulin insufficiency and hyperglycemia, from 6 weeks of age. At 26 weeks of age, vascular reactivity was assessed by wire myography, pro-inflammatory expression was assessed in the aortas by qRT-PCR and immunohistochemistry, and systemic and vascular oxidative stress measurements were determined. Additionally, studies in human aortic endothelial cells (HAECs) derived from normal and diabetic patients in the presence or absence of dh404 included assessment of pro-inflammatory genes by qRT-PCR and western blotting. Oxidative stress was assessed by three methods; L-012, DCFDA and amplex red. Static adhesion assays were performed to determine the leukocyte–endothelial interaction in the presence or absence of dh404. Results Dh404 significantly attenuated endothelial dysfunction in diabetic Akita mice characterized by reduced contraction in response to phenylephrine and the downregulation of inflammatory genes (VCAM-1, ICAM-1, p65, IL-1β) and pro-oxidant genes (Nox1 and Nox2). Furthermore, reduced systemic and vascular oxidative stress levels were observed in diabetic Akita mice. dh404 exhibited cytoprotective effects in diabetic HAECs in vitro, reflected by significant upregulation of Nrf2-responsive genes, NAD(P)H quinone oxidoreductase 1 (NQO1) and heme oxygenase-1 (HO-1), reduction of oxidative stress markers (O2·− and H2O2), inhibition of inflammatory genes (VCAM-1 and the p65 subunit of NF-κB) and attenuation of leukocyte–endothelial interactions (P < 0.05 for all in vitro and in vivo parameters; one or two-way ANOVA as appropriate with post hoc testing). Conclusion These studies demonstrate that upregulation of Nrf2 by dh404 represents a novel therapeutic strategy to limit diabetes-associated vascular injury. Electronic supplementary material The online version of this article (doi:10.1186/s12933-017-0513-y) contains supplementary material, which is available to authorized users.
Collapse
|
37
|
Lacava V, Pellicanò V, Ferrajolo C, Cernaro V, Visconti L, Conti G, Buemi M, Santoro D. Novel avenues for treating diabetic nephropathy: new investigational drugs. Expert Opin Investig Drugs 2017; 26:445-462. [PMID: 28277032 DOI: 10.1080/13543784.2017.1293039] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Viviana Lacava
- Unit of Nephrology and Dialysis, University of Messina, Messina, Italy
| | | | - Carmen Ferrajolo
- Department of Experimental Medicine, Second University of Naples, Napoli, Italy
| | - Valeria Cernaro
- Unit of Nephrology and Dialysis, University of Messina, Messina, Italy
| | - Luca Visconti
- Unit of Nephrology and Dialysis, University of Messina, Messina, Italy
| | - Giovanni Conti
- Unit of Pediatric Nephrology and Rheumatology, University of Messina, Messina, Italy
| | - Michele Buemi
- Unit of Nephrology and Dialysis, University of Messina, Messina, Italy
| | - Domenico Santoro
- Unit of Nephrology and Dialysis, University of Messina, Messina, Italy
| |
Collapse
|
38
|
Musso G, Cassader M, Cohney S, De Michieli F, Pinach S, Saba F, Gambino R. Fatty Liver and Chronic Kidney Disease: Novel Mechanistic Insights and Therapeutic Opportunities. Diabetes Care 2016; 39:1830-45. [PMID: 27660122 DOI: 10.2337/dc15-1182] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 07/10/2016] [Indexed: 02/03/2023]
Abstract
Chronic kidney disease (CKD) is a risk factor for end-stage renal disease (ESRD) and cardiovascular disease (CVD). ESRD or CVD develop in a substantial proportion of patients with CKD receiving standard-of-care therapy, and mortality in CKD remains unchanged. These data suggest that key pathogenetic mechanisms underlying CKD progression go unaffected by current treatments. Growing evidence suggests that nonalcoholic fatty liver disease (NAFLD) and CKD share common pathogenetic mechanisms and potential therapeutic targets. Common nutritional conditions predisposing to both NAFLD and CKD include excessive fructose intake and vitamin D deficiency. Modulation of nuclear transcription factors regulating key pathways of lipid metabolism, inflammation, and fibrosis, including peroxisome proliferator-activated receptors and farnesoid X receptor, is advancing to stage III clinical development. The relevance of epigenetic regulation in the pathogenesis of NAFLD and CKD is also emerging, and modulation of microRNA21 is a promising therapeutic target. Although single antioxidant supplementation has yielded variable results, modulation of key effectors of redox regulation and molecular sensors of intracellular energy, nutrient, or oxygen status show promising preclinical results. Other emerging therapeutic approaches target key mediators of inflammation, such as chemokines; fibrogenesis, such as galectin-3; or gut dysfunction through gut microbiota manipulation and incretin-based therapies. Furthermore, NAFLD per se affects CKD through lipoprotein metabolism and hepatokine secretion, and conversely, targeting the renal tubule by sodium-glucose cotransporter 2 inhibitors can improve both CKD and NAFLD. Implications for the treatment of NAFLD and CKD are discussed in light of this new therapeutic armamentarium.
Collapse
Affiliation(s)
- Giovanni Musso
- Humanitas Gradenigo Hospital, University of Turin, Turin, Italy
| | - Maurizio Cassader
- Department of Medical Sciences, San Giovanni Battista Hospital, University of Turin, Turin, Italy
| | - Solomon Cohney
- Department of Nephrology, Western & Royal Melbourne Hospitals, Melbourne, VIC, Australia
| | - Franco De Michieli
- Department of Medical Sciences, San Giovanni Battista Hospital, University of Turin, Turin, Italy
| | - Silvia Pinach
- Department of Medical Sciences, San Giovanni Battista Hospital, University of Turin, Turin, Italy
| | - Francesca Saba
- Department of Medical Sciences, San Giovanni Battista Hospital, University of Turin, Turin, Italy
| | - Roberto Gambino
- Department of Medical Sciences, San Giovanni Battista Hospital, University of Turin, Turin, Italy
| |
Collapse
|
39
|
Musso G, Cassader M, Cohney S, Pinach S, Saba F, Gambino R. Emerging Liver-Kidney Interactions in Nonalcoholic Fatty Liver Disease. Trends Mol Med 2016; 21:645-662. [PMID: 26432021 DOI: 10.1016/j.molmed.2015.08.005] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Revised: 08/07/2015] [Accepted: 08/16/2015] [Indexed: 12/12/2022]
Abstract
Mounting evidence connects non-alcoholic fatty liver disease (NAFLD) to chronic kidney disease (CKD). We review emerging mechanistic links between NAFLD and CKD, including altered activation of angiotensin converting enzyme (ACE)-2, nutrient/energy sensors sirtuin-1 and AMP-activated kinase, as well as impaired antioxidant defense mediated by nuclear factor erythroid 2-related factor-2 (Nrf2). Dietary fructose excess may also contribute to NAFLD and CKD. NAFLD affects renal injury through lipoprotein dysmetabolism and altered secretion of the hepatokines fibroblast growth factor-21, fetuin-A, insulin-like growth factor-1, and syndecan-1. CKD may mutually aggravate NAFLD and associated metabolic disturbances through altered intestinal barrier function and microbiota composition, the accumulation of uremic toxic metabolites, and alterations in pre-receptor glucocorticoid metabolism. We conclude by discussing the implications of these findings for the treatment of NAFLD and CKD.
Collapse
Affiliation(s)
| | - Maurizio Cassader
- Department of Medical Sciences, San Giovanni Battista Hospital, University of Turin, Turin, Italy
| | - Solomon Cohney
- Department of Nephrology, Royal Melbourne and Western Hospital, Victoria, University of Melbourne, Melbourne, Australia
| | - Silvia Pinach
- Department of Medical Sciences, San Giovanni Battista Hospital, University of Turin, Turin, Italy
| | - Francesca Saba
- Department of Medical Sciences, San Giovanni Battista Hospital, University of Turin, Turin, Italy
| | - Roberto Gambino
- Department of Medical Sciences, San Giovanni Battista Hospital, University of Turin, Turin, Italy
| |
Collapse
|
40
|
Marcuccilli M, Chonchol M. NAFLD and Chronic Kidney Disease. Int J Mol Sci 2016; 17:562. [PMID: 27089331 PMCID: PMC4849018 DOI: 10.3390/ijms17040562] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 03/25/2016] [Accepted: 03/28/2016] [Indexed: 12/13/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease in developed countries and it is now considered a risk factor for cardiovascular disease. Evidence linking NAFLD to the development and progression of chronic kidney disease (CKD) is emerging as a popular area of scientific interest. The rise in simultaneous liver-kidney transplantation as well as the significant cost associated with the presence of chronic kidney disease in the NAFLD population make this entity a worthwhile target for screening and therapeutic intervention. While several cross-sectional and case control studies have been published to substantiate these theories, very little data exists on the underlying cause of NAFLD and CKD. In this review, we will discuss the most recent publications on the diagnosis of NAFLD as well new evidence regarding the pathophysiology of NAFLD and CKD as an inflammatory disorder. These mechanisms include the role of obesity, the renin-angiotensin system, and dysregulation of fructose metabolism and lipogenesis in the development of both disorders. Further investigation of these pathways may lead to novel therapies that aim to target the NAFLD and CKD. However, more prospective studies that include information on both renal and liver histology will be necessary in order to understand the relationship between these diseases.
Collapse
Affiliation(s)
- Morgan Marcuccilli
- Division of Renal Diseases and Hypertension, University of Colorado Hospital, Aurora, CO 80045, USA.
| | - Michel Chonchol
- Division of Renal Diseases and Hypertension, University of Colorado Denver, 13199 East Montview Boulevard, Suite 495, Aurora, CO 80045, USA.
| |
Collapse
|
41
|
Prasad S, Yadav VR, Sung B, Gupta SC, Tyagi AK, Aggarwal BB. Ursolic acid inhibits the growth of human pancreatic cancer and enhances the antitumor potential of gemcitabine in an orthotopic mouse model through suppression of the inflammatory microenvironment. Oncotarget 2016; 7:13182-96. [PMID: 26909608 PMCID: PMC4914350 DOI: 10.18632/oncotarget.7537] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 01/06/2016] [Indexed: 12/29/2022] Open
Abstract
The development of chemoresistance in human pancreatic cancer is one reason for the poor survival rate for patients with this cancer. Because multiple gene products are linked with chemoresistance, we investigated the ability of ursolic acid (UA) to sensitize pancreatic cancer cells to gemcitabine, a standard drug used for the treatment of pancreatic cancer. These investigations were done in AsPC-1, MIA PaCa-2, and Panc-28 cells and in nude mice orthotopically implanted with Panc-28 cells. In vitro, UA inhibited proliferation, induced apoptosis, suppressed NF-κB activation and its regulated proliferative, metastatic, and angiogenic proteins. UA (20 μM) also enhanced gemcitabine (200 nM)-induced apoptosis and suppressed the expression of NF-κB-regulated proteins. In the nude mouse model, oral administration of UA (250 mg/kg) suppressed tumor growth and enhanced the effect of gemcitabine (25 mg/kg). Furthermore, the combination of UA and gemcitabine suppressed the metastasis of cancer cells to distant organs such as liver and spleen. Immunohistochemical analysis showed that biomarkers of proliferation (Ki-67) and microvessel density (CD31) were suppressed by the combination of UA and gemcitabine. UA inhibited the activation of NF-κB and STAT3 and the expression of tumorigenic proteins regulated by these inflammatory transcription factors in tumor tissue. Furthermore, the combination of two agents decreased the expression of miR-29a, closely linked with tumorigenesis, in the tumor tissue. UA was found to be bioavailable in animal serum and tumor tissue. These results suggest that UA can inhibit the growth of human pancreatic tumors and sensitize them to gemcitabine by suppressing inflammatory biomarkers linked to proliferation, invasion, angiogenesis, and metastasis.
Collapse
Affiliation(s)
- Sahdeo Prasad
- Department of Experimental Therapeutics, Cytokine Research Laboratory, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vivek R. Yadav
- Department of Experimental Therapeutics, Cytokine Research Laboratory, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bokyung Sung
- Department of Experimental Therapeutics, Cytokine Research Laboratory, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Subash C. Gupta
- Department of Experimental Therapeutics, Cytokine Research Laboratory, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amit K. Tyagi
- Department of Experimental Therapeutics, Cytokine Research Laboratory, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bharat B. Aggarwal
- Department of Experimental Therapeutics, Cytokine Research Laboratory, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Anti-inflammatory Research Institute, San Deigo, CA, USA
| |
Collapse
|
42
|
Elango B, Dornadula S, Paulmurugan R, Ramkumar KM. Pterostilbene Ameliorates Streptozotocin-Induced Diabetes through Enhancing Antioxidant Signaling Pathways Mediated by Nrf2. Chem Res Toxicol 2016; 29:47-57. [DOI: 10.1021/acs.chemrestox.5b00378] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
| | | | - Ramasamy Paulmurugan
- Department
of Radiology, Stanford University School of Medicine, Palo Alto, California 94305, United States
| | | |
Collapse
|
43
|
Houghton CA, Fassett RG, Coombes JS. Sulforaphane and Other Nutrigenomic Nrf2 Activators: Can the Clinician's Expectation Be Matched by the Reality? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:7857186. [PMID: 26881038 PMCID: PMC4736808 DOI: 10.1155/2016/7857186] [Citation(s) in RCA: 175] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/06/2015] [Indexed: 12/14/2022]
Abstract
The recognition that food-derived nonnutrient molecules can modulate gene expression to influence intracellular molecular mechanisms has seen the emergence of the fields of nutrigenomics and nutrigenetics. The aim of this review is to describe the properties of nutrigenomic activators of transcription factor Nrf2 (nuclear factor erythroid 2-related factor 2), comparing the potential for sulforaphane and other phytochemicals to demonstrate clinical efficacy as complementary medicines. Broccoli-derived sulforaphane emerges as a phytochemical with this capability, with oral doses capable of favourably modifying genes associated with chemoprevention. Compared with widely used phytochemical-based supplements like curcumin, silymarin, and resveratrol, sulforaphane more potently activates Nrf2 to induce the expression of a battery of cytoprotective genes. By virtue of its lipophilic nature and low molecular weight, sulforaphane displays significantly higher bioavailability than the polyphenol-based dietary supplements that also activate Nrf2. Nrf2 activation induces cytoprotective genes such as those playing key roles in cellular defense mechanisms including redox status and detoxification. Both its high bioavailability and significant Nrf2 inducer capacity contribute to the therapeutic potential of sulforaphane-yielding supplements.
Collapse
Affiliation(s)
- Christine A. Houghton
- School of Human Movement and Nutrition Science, The University of Queensland, Brisbane, Australia
| | - Robert G. Fassett
- School of Human Movement and Nutrition Science, The University of Queensland, Brisbane, Australia
| | - Jeff S. Coombes
- School of Human Movement and Nutrition Science, The University of Queensland, Brisbane, Australia
- *Jeff S. Coombes:
| |
Collapse
|
44
|
Camer D, Yu Y, Szabo A, Wang H, Dinh CH, Huang XF. Bardoxolone methyl prevents the development and progression of cardiac and renal pathophysiologies in mice fed a high-fat diet. Chem Biol Interact 2016; 243:10-8. [DOI: 10.1016/j.cbi.2015.11.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 10/21/2015] [Accepted: 11/13/2015] [Indexed: 02/07/2023]
|
45
|
Abstract
The transcription factor Nrf2 (nuclear factor erythroid 2-related factor 2) is a major regulator of oxidative stress defence in the human body. As Nrf2 regulates the expression of a large battery of cytoprotective genes, it plays a crucial role in the prevention of degenerative disease in multiple organs. Thus it has been the focus of research as a pharmacological target that could be used for prevention and treatment of chronic diseases such as multiple sclerosis, chronic kidney disease or cardiovascular diseases. The present review summarizes promising findings from basic research and shows which Nrf2-targeting therapies are currently being investigated in clinical trials and which agents have already entered clinical practice.
Collapse
|
46
|
Singh VK, Mishra A, Gupta KK, Misra R, Patel ML, Shilpa. Reduction of microalbuminuria in type-2 diabetes mellitus with angiotensin-converting enzyme inhibitor alone and with cilnidipine. Indian J Nephrol 2015; 25:334-9. [PMID: 26664207 PMCID: PMC4663769 DOI: 10.4103/0971-4065.151764] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The aim of our study was to find out the antiproteinuric effect of enalapril angiotensin-converting enzyme (ACE inhibitor) alone or in combination with cilnidipine in patients with type-2 diabetes mellitus. The study was conducted on 71 patients with type-2 diabetes mellitus patients with hypertension and microalbuminuria. They were divided into two groups randomly as follows: Group I (enalaprilalone, n = 36) and Group II (enalapril with cilnidipine, n = 35). In both the groups, baseline 24 h urinary albumin was estimated and was repeated every 3 months upto 1-year. After 1-year follow-up, reduction in microalbuminuria was found to be greater in Group II. In Group I microalbuminuria came down by 25.68 ± 21.40 while in Group II it reduced by 54.88 ± 13.84 (P < 0.001). We conclude that in diabetic population, cilnidipine has an additive effect in microalbuminuria reduction over and above the well-proven effect of ACE inhibitors.
Collapse
Affiliation(s)
- V K Singh
- Department of Medicine, King George Medical University, Lucknow, Uttar Pradesh, India
| | - A Mishra
- Department of Medicine, King George Medical University, Lucknow, Uttar Pradesh, India
| | - K K Gupta
- Department of Medicine, King George Medical University, Lucknow, Uttar Pradesh, India
| | - R Misra
- Department of Medicine, King George Medical University, Lucknow, Uttar Pradesh, India
| | - M L Patel
- Department of Medicine, King George Medical University, Lucknow, Uttar Pradesh, India
| | - Shilpa
- Department of Medicine, King George Medical University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
47
|
Montezano AC, Dulak-Lis M, Tsiropoulou S, Harvey A, Briones AM, Touyz RM. Oxidative Stress and Human Hypertension: Vascular Mechanisms, Biomarkers, and Novel Therapies. Can J Cardiol 2015; 31:631-41. [DOI: 10.1016/j.cjca.2015.02.008] [Citation(s) in RCA: 205] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 02/06/2015] [Accepted: 02/06/2015] [Indexed: 02/07/2023] Open
|
48
|
Abstract
Despite the wealth of pre-clinical support for a role for reactive oxygen and nitrogen species (ROS/RNS) in the aetiology of diabetic complications, enthusiasm for antioxidant therapeutic approaches has been dampened by less favourable outcomes in large clinical trials. This has necessitated a re-evaluation of pre-clinical evidence and a more rational approach to antioxidant therapy. The present review considers current evidence, from both pre-clinical and clinical studies, to address the benefits of antioxidant therapy. The main focus of the present review is on the effects of direct targeting of ROS-producing enzymes, the bolstering of antioxidant defences and mechanisms to improve nitric oxide availability. Current evidence suggests that a more nuanced approach to antioxidant therapy is more likely to yield positive reductions in end-organ injury, with considerations required for the types of ROS/RNS involved, the timing and dosage of antioxidant therapy, and the selective targeting of cell populations. This is likely to influence future strategies to lessen the burden of diabetic complications such as diabetes-associated atherosclerosis, diabetic nephropathy and diabetic retinopathy.
Collapse
|
49
|
Moon EJ, Giaccia A. Dual roles of NRF2 in tumor prevention and progression: possible implications in cancer treatment. Free Radic Biol Med 2015; 79:292-9. [PMID: 25458917 PMCID: PMC4339613 DOI: 10.1016/j.freeradbiomed.2014.11.009] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 11/13/2014] [Accepted: 11/13/2014] [Indexed: 12/16/2022]
Abstract
The cap'n'collar (CNC) family serves as cellular sensors of oxidative and electrophilic stresses and shares structural similarities including basic leucine zipper (bZIP) and CNC domains. They form heterodimers with small MAF proteins to regulate antioxidant and phase II enzymes through antioxidant response element (ARE)-mediated transactivation. Among the CNC family members, NRF2 is required for systemic protection against redox-mediated injury and carcinogenesis. On the other hand, NRF2 is activated by oncogenic pathways, metabolism, and hypoxia. Constitutive NRF2 activation is observed in a variety of human cancers and it is highly correlated with tumor progression and aggressiveness. In this review, we will discuss how NRF2 plays dual roles in cancer prevention and progression depending on the cellular context and environment. Therefore, a better understanding of NRF2 will be necessary to exploit this complex network of balancing antioxidant pathways to inhibit tumor progression.
Collapse
Affiliation(s)
- Eui Jung Moon
- Division of Radiation Biology & Oncology, Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Amato Giaccia
- Division of Radiation Biology & Oncology, Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
50
|
Owusu-Ansah A, Choi SH, Petrosiute A, Letterio JJ, Huang AYC. Triterpenoid inducers of Nrf2 signaling as potential therapeutic agents in sickle cell disease: a review. Front Med 2014; 9:46-56. [PMID: 25511620 DOI: 10.1007/s11684-015-0375-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 09/12/2014] [Indexed: 01/06/2023]
Abstract
Sickle cell disease (SCD) is an inherited disorder of hemoglobin in which the abnormal hemoglobin S polymerizes when deoxygenated. This polymerization of hemoglobin S not only results in hemolysis and vasoocclusion but also precipitates inflammation, oxidative stress and chronic organ dysfunction. Oxidative stress is increasingly recognized as an important intermediate in these pathophysiological processes and is therefore an important target for therapeutic intervention. The transcription factor nuclear erythroid derived-2 related factor 2 (Nrf2) controls the expression of anti-oxidant enzymes and is emerging as a protein whose function can be exploited with therapeutic intent. This review article is focused on triterpenoids that activate Nrf2, and their potential for reducing oxidative stress in SCD as an approach to prevent organ dysfunction associated with this disease. A brief overview of oxidative stress in the clinical context of SCD is accompanied by a discussion of several pathophysiological mechanisms contributing to oxidative stress. Finally, these mechanisms are then related to current management strategies in SCD that are either utilized currently or under evaluation. The article concludes with a perspective on the potential of the various therapeutic interventions to reduce oxidative stress and morbidity associated with SCD.
Collapse
Affiliation(s)
- Amma Owusu-Ansah
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | | | | | | | | |
Collapse
|