1
|
Bangar NS, Dixit A, Apte MM, Tupe RS. Syzygium cumini (L.) skeels mitigate diabetic nephropathy by regulating Nrf2 pathway and mitocyhondrial dysfunction: In vitro and in vivo studies. JOURNAL OF ETHNOPHARMACOLOGY 2025; 336:118684. [PMID: 39127117 DOI: 10.1016/j.jep.2024.118684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/12/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
ETHNOPHARMACOLOGICAL PREVALENCE Hyperglycemia in diabetes increases the generation of advanced glycation end products (AGEs) through non-enzymatic reactions. The interaction between AGEs and their receptors (RAGE) leads to oxidative and inflammatory stress, which plays a pivotal role in developing diabetic nephropathy. Syzygium cumini (SC) L. (DC.) homeopathic preparations viz. 200C, 30C, and mother tincture [MT] are used to treat diabetes. This study aimed to elucidate the regulatory effects of SC preparations (200C, 30C, and MT) on the nuclear factor erythroid 2-related factor 2 (Nrf2) - nuclear factor-κB (NF-κB) pathways and mitochondrial dysfunction in mitigating diabetic nephropathy (DN). MATERIALS AND METHODS Streptozotocin-induced diabetic rats were treated with SC preparations (200C, 30C, MT; 1:20 dilution in distilled water; 600 μL/kg body weight) and metformin (45 mg/kg body weight) twice daily for 40 days. DN was evaluated through biochemical parameters and histological examination. Renal tissue lysates were analyzed for glycation markers. Protein and gene levels of Nrf2, NF-κB, and mitochondrial dysfunctional signaling were determined via western blotting and RT-qPCR. An immunohistochemical analysis of the kidneys was performed. In vitro, human serum albumin (HSA - 10 mg/ml) was glycated with methylglyoxal (MGO - 55 mM) in the presence of SC preparations (200C, 30C, MT) for eight days. Glycated samples (400 μg/mL) were incubated with renal cells (HEK-293) for 24 h. Further reactive oxygen species production, Nrf2 nuclear translocation, and protein or gene expression of Nrf2 and apoptosis markers were analyzed by western blotting, RT-qPCR, and flow cytometry. Molecular docking of gallic and ellagic acid with the HSA-MGO complex was performed. RESULT In vivo experiments using streptozotocin-induced diabetic rats treated with SC preparations exhibited improved biochemical parameters, preserved kidney function, and reduced glycation adduct formation in a dose-dependent manner. Furthermore, SC preparations downregulated inflammatory mediators such as RAGE, NF-κB, vascular endothelial growth factor (VEGF), and Tumor necrosis factor α (TNF-α) while upregulating the Nrf2-dependent antioxidant and detoxification pathways. They downregulated B-cell lymphoma 2 (Bcl-2) associated X-protein (BAX), C/EBP homologous protein (CHOP), Dynamin-related protein 1 (DRP1), and upregulated BCL 2 gene expression. Notably, SC preparations facilitated nuclear translocation of Nrf2, leading to the upregulation of antioxidant enzymes and the downregulation of oxidative stress markers. Molecular docking studies revealed favorable interactions between gallic (-5.26 kcal/mol) and ellagic acid (-4.71 kcal/mol) with the HSA-MGO complex. CONCLUSION SC preparations mitigate renal cell apoptosis and mitochondrial dysfunction through Nrf2-dependent mechanisms.
Collapse
Affiliation(s)
- Nilima S Bangar
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, Maharashtra State, India.
| | - Aditi Dixit
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, Maharashtra State, India.
| | - Mayura M Apte
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, Maharashtra State, India.
| | - Rashmi S Tupe
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, Maharashtra State, India.
| |
Collapse
|
2
|
Chen X, Hu K, Zhang Y, He SM, Wang DD. Targeting CXCR2 ameliorated tacrolimus-induced nephrotoxicity by alleviating overactivation of PI3K/AKT/mTOR pathway and calcium overload. Biomed Pharmacother 2024; 180:117526. [PMID: 39378682 DOI: 10.1016/j.biopha.2024.117526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 10/10/2024] Open
Abstract
OBJECTIVES The purposes of this study were to (i) verify the role of CXCR2 in tacrolimus-induced nephrotoxicity, (ii) explore the specific mechanism of CXCR2-mediated tacrolimus nephrotoxicity, and (iii) target the antagonism of CXCR2 and provide a potential target for the treatment of tacrolimus-induced nephrotoxicity in children. METHODS CXCR2 knockout (CXCR2-KO) mice were used to evaluate the role of CXCR2 in tacrolimus-induced nephrotoxicity. Wistar rats were used to explore the underlying mechanism. RESULTS In the knockout mice, compared with N-WT group, the renal function index was deteriorative (P < 0.01), the degree of renal fibrosis was aggravated (P < 0.01), the pathological expression of E-cadherin (P < 0.01) and α-SMA (P < 0.01) were occurred in T-WT group. Inversely, compared with T-WT group, the above indicators were improved in T-KO group (P < 0.01). In wistar rats, compared with N group, the renal function index was deteriorative (P < 0.05 or P < 0.01), fibrosis and calcium overload occurred (P < 0.01), CXCL2-CXCR2 was activated (P < 0.05), and meanwhile PI3K/AKT/mTOR pathway was activated (P < 0.05 or P < 0.01) in T group. Inversely, compared with T group, the above indicators were reversed in C group (P < 0.05 or P < 0.01). CONCLUSION The present study was firstly to report that CXCL2-CXCR2 activated PI3K/AKT/mTOR pathway and calcium overload in tacrolimus-induced nephrotoxicity, and targeting CXCR2 could inhibit the progression of tacrolimus-induced nephrotoxicity.
Collapse
Affiliation(s)
- Xiao Chen
- School of Nursing, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Ke Hu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy & School of Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yue Zhang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy & School of Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Su-Mei He
- Department of Pharmacy, Suzhou Research Center of Medical School, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, Jiangsu 215153, China.
| | - Dong-Dong Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy & School of Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
3
|
Yang H, Sun J, Sun A, Wei Y, Xie W, Xie P, Zhang L, Zhao L, Huang Y. Podocyte programmed cell death in diabetic kidney disease: Molecular mechanisms and therapeutic prospects. Biomed Pharmacother 2024; 177:117140. [PMID: 39018872 DOI: 10.1016/j.biopha.2024.117140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 06/28/2024] [Accepted: 07/10/2024] [Indexed: 07/19/2024] Open
Abstract
Diabetic kidney disease (DKD) is the primary cause of chronic kidney and end-stage renal disease. Glomerular podocyte loss and death are pathological hallmarks of DKD, and programmed cell death (PCD) in podocytes is crucial in DKD progression. PCD involves apoptosis, autophagy, ferroptosis, pyroptosis, and necroptosis. During DKD, PCD in podocytes is severely impacted and primarily characterized by accelerated podocyte apoptosis and suppressed autophagy. These changes lead to a gradual decrease in podocyte numbers, impairing the glomerular filtration barrier function and accelerating DKD progression. However, research on the interactions between the different types of PCD in podocytes is lacking. This review focuses on the novel roles and mechanisms of PCD in the podocytes of patients with DKD. Additionally, we summarize clinical drugs capable of regulating podocyte PCD, present challenges and prospects faced in developing drugs related to podocyte PCD and suggest that future research should further explore the detailed mechanisms of podocyte PCD and interactions among different types of PCD.
Collapse
Affiliation(s)
- Haoyu Yang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Jun Sun
- Changchun University of Chinese Medicine, Changchun 130117, China
| | - Aru Sun
- Changchun University of Chinese Medicine, Changchun 130117, China
| | - Yu Wei
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Weinan Xie
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Pengfei Xie
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Lili Zhang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Linhua Zhao
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Yishan Huang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; China-Japan Friendship Hospital, Beijing 100029, China.
| |
Collapse
|
4
|
Zhou C, Zhao D, Wu C, Wu Z, Zhang W, Chen S, Zhao X, Wu S. Role of histone deacetylase inhibitors in non-neoplastic diseases. Heliyon 2024; 10:e33997. [PMID: 39071622 PMCID: PMC11283006 DOI: 10.1016/j.heliyon.2024.e33997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 07/30/2024] Open
Abstract
Background Epigenetic dysregulation has been implicated in the development and progression of a variety of human diseases, but epigenetic changes are reversible, and epigenetic enzymes and regulatory proteins can be targeted using small molecules. Histone deacetylase inhibitors (HDACis), as a class of epigenetic drugs, are widely used to treat various cancers and other diseases involving abnormal gene expression. Results Specially, HDACis have emerged as a promising strategy to enhance the therapeutic effect of non-neoplastic conditions, including neurological disorders, cardiovascular diseases, renal diseases, autoimmune diseases, inflammatory diseases, infectious diseases and rare diseases, along with their related mechanisms. However, their clinical efficacy has been limited by drug resistance and toxicity. Conclusions To date, most clinical trials of HDAC inhibitors have been related to the treatment of cancer rather than the treatment of non-cancer diseases, for which experimental studies are gradually underway. Discussions regarding non-neoplastic diseases often concentrate on specific disease types. Therefore, this review highlights the development of HDACis and their potential therapeutic applications in non-neoplastic diseases, either as monotherapy or in combination with other drugs or therapies.
Collapse
Affiliation(s)
- Chunxiao Zhou
- College of Medicine, Qingdao University, Qingdao, 266000, China
| | - Dengke Zhao
- Harbin Medical University, Harbin, 150000, China
| | - Chunyan Wu
- College of Medicine, Qingdao University, Qingdao, 266000, China
| | - Zhimin Wu
- College of Medicine, Qingdao University, Qingdao, 266000, China
| | - Wen Zhang
- College of Medicine, Qingdao University, Qingdao, 266000, China
| | - Shilv Chen
- College of Medicine, Qingdao University, Qingdao, 266000, China
| | - Xindong Zhao
- College of Medicine, Qingdao University, Qingdao, 266000, China
| | - Shaoling Wu
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| |
Collapse
|
5
|
Bi Y, Wei H, Yu T, Li X, Xu S. New insights into resveratrol attenuates hepatotoxicity in emamectin benzoate-exposed grass carp (Ctenopharyngodon idella) via NO system/NF-κB signaling pathway. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 202:105941. [PMID: 38879332 DOI: 10.1016/j.pestbp.2024.105941] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/22/2024] [Accepted: 05/01/2024] [Indexed: 07/02/2024]
Abstract
Emamectin benzoate (EMB) is extensively used as a crop protection agent. Overuse of EMB poses a serious threat to the quality of water and non-target organisms in the environment. Resveratrol (RES) is a natural phytoalexin with the function of anti-oxidation and anti-inflammation. Nonetheless, it is unclear whether EMB affects the expression of cytokines and induces autophagy, apoptosis, and necroptosis of hepatocytes (L8824 cell) in grass carp (Ctenopharyngodon idella), and whether RES has an attenuate function in this process. Therefore, we established the L8824 cells model of EMB exposure and treated it with RES. The results showed that compared with the control (CON) group, EMB exposure significantly increased the nitric oxide (NO) content, inducible nitric oxide synthase (iNOS) activity, and the expression of iNOS and phosphorylated nuclear factor kappa B (p-NF-κB) (P < 0.05). In addition, compared with the CON group, the results of flow cytometry and dansylcadaverine (MDC) staining showed a significant increase in apoptosis and autophagy in the EMB-exposed group (P < 0.05) with the activation of the B-cell lymphoma-2 (Bcl-2)/Bcl-2 associated X (Bax)/cysteine-aspartic acid protease 3 (Caspase-3)/cysteine-aspartic acid protease 9 (Caspase-9) pathway and microtubule-associated protein light chain 3 (LC3)/sequestosome 1 (p62)/Beclin1 pathway. EMB exposure significantly increased the mRNA and protein expression of receptor-interacting protein 1 (RIPK1)/receptor-interacting protein 3 (RIPK3)/mixed the lineage kinase domain-like (MLKL) pathway (P < 0.05). Moreover, EMB exposure significantly increased the expression of genes related to immunity (immunoglobulin G (IgG), immunoglobulin M (IgM), and immunoglobulin D (IgD), and antimicrobial peptide-related genes expression including β-defensin and hepcidin) (P < 0.05). The addition of RES significantly diminished autophagy, apoptosis, necroptosis, and immunity-related gene expression by inhibiting iNOS activity, NO content, and the protein expression of iNOS and p-NF-κB. In conclusion, RES attenuated autophagy, apoptosis, and necroptosis in EMB-exposed L8824 cells via suppression of the NO system/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yanju Bi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Haidong Wei
- College of Life Science, Northeast Agricultural University, Harbin 150030, PR China
| | - Tingting Yu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Xiaojing Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
6
|
Wang N, Zhang C. Oxidative Stress: A Culprit in the Progression of Diabetic Kidney Disease. Antioxidants (Basel) 2024; 13:455. [PMID: 38671903 PMCID: PMC11047699 DOI: 10.3390/antiox13040455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/01/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Diabetic kidney disease (DKD) is the principal culprit behind chronic kidney disease (CKD), ultimately developing end-stage renal disease (ESRD) and necessitating costly dialysis or kidney transplantation. The limited therapeutic efficiency among individuals with DKD is a result of our finite understanding of its pathogenesis. DKD is the result of complex interactions between various factors. Oxidative stress is a fundamental factor that can establish a link between hyperglycemia and the vascular complications frequently encountered in diabetes, particularly DKD. It is crucial to recognize the essential and integral role of oxidative stress in the development of diabetic vascular complications, particularly DKD. Hyperglycemia is the primary culprit that can trigger an upsurge in the production of reactive oxygen species (ROS), ultimately sparking oxidative stress. The main endogenous sources of ROS include mitochondrial ROS production, NADPH oxidases (Nox), uncoupled endothelial nitric oxide synthase (eNOS), xanthine oxidase (XO), cytochrome P450 (CYP450), and lipoxygenase. Under persistent high glucose levels, immune cells, the complement system, advanced glycation end products (AGEs), protein kinase C (PKC), polyol pathway, and the hexosamine pathway are activated. Consequently, the oxidant-antioxidant balance within the body is disrupted, which triggers a series of reactions in various downstream pathways, including phosphoinositide 3-kinase/protein kinase B (PI3K/Akt), transforming growth factor beta/p38-mitogen-activated protein kinase (TGF-β/p38-MAPK), nuclear factor kappa B (NF-κB), adenosine monophosphate-activated protein kinase (AMPK), and the Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling. The disease might persist even if strict glucose control is achieved, which can be attributed to epigenetic modifications. The treatment of DKD remains an unresolved issue. Therefore, reducing ROS is an intriguing therapeutic target. The clinical trials have shown that bardoxolone methyl, a nuclear factor erythroid 2-related factor 2 (Nrf2) activator, blood glucose-lowering drugs, such as sodium-glucose cotransporter 2 inhibitors, and glucagon-like peptide-1 receptor agonists can effectively slow down the progression of DKD by reducing oxidative stress. Other antioxidants, including vitamins, lipoic acid, Nox inhibitors, epigenetic regulators, and complement inhibitors, present a promising therapeutic option for the treatment of DKD. In this review, we conduct a thorough assessment of both preclinical studies and current findings from clinical studies that focus on targeted interventions aimed at manipulating these pathways. We aim to provide a comprehensive overview of the current state of research in this area and identify key areas for future exploration.
Collapse
Affiliation(s)
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
7
|
Zuo Z, Li Q, Zhou S, Yu R, Wu C, Chen J, Xiao Y, Chen H, Song J, Pan Y, Wang W. Berberine ameliorates contrast-induced acute kidney injury by regulating HDAC4-FoxO3a axis-induced autophagy: In vivo and in vitro. Phytother Res 2024; 38:1761-1780. [PMID: 37922559 DOI: 10.1002/ptr.8059] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 09/30/2023] [Accepted: 10/15/2023] [Indexed: 11/07/2023]
Abstract
In hospitals, contrast-induced acute kidney injury (CI-AKI) is a major cause of renal failure. This study evaluates berberine's (BBR) renal protection and its potential HDAC4 mechanism. CI-AKI in rats was induced with 10 mL kg-1 ioversol. Rats were divided into five groups: Ctrl, BBR, CI-AKI, CI-AKI + BBR, and CI-AKI + Tasq. The renal function of CI-AKI rats was determined by measuring serum creatinine and blood urea nitrogen. Histopathological changes and apoptosis of renal tubular epithelial cells were observed by HE and terminal deoxynucleotidyl transferase (TdTase)-mediated dUTP-biotin nick end labeling (TUNEL) staining. Transmission electron microscopy was used to observe autophagic structures. In vitro, a CI-AKI cell model was created with ioversol-treated HK-2 cells. Treatments included BBR, Rapa, HCQ, and Tasq. Analyses focused on proteins and genes associated with kidney injury, apoptosis, autophagy, and the HDAC4-FoxO3a axis. BBR showed significant protective effects against CI-AKI both in vivo and in vitro. It inhibited apoptosis by increasing Bcl-2 protein levels and decreasing Bax levels. BBR also activated autophagy, as indicated by changes in autophagy-related proteins and autophagic flux. The study further revealed that the contrast agent ioversol increased the expression of HDAC4, which led to elevated levels of phosphorylated FoxO3a (p-FoxO3a) and acetylated FoxO3a (Ac-FoxO3a). However, BBR inhibited HDAC4 expression, resulting in decreased levels of p-FoxO3a and Ac-FoxO3a. This activation of autophagy-related genes, regulated by the transcription factor FoxO3a, played a role in BBR's protective effects. BBR, a traditional Chinese medicine, shows promise against CI-AKI. It may counteract CI-AKI by modulating HDAC4 and FoxO3a, enhancing autophagy, and limiting apoptosis.
Collapse
Affiliation(s)
- Zhi Zuo
- Department of Cardiology, The First Affiliated Hospital with Nanjing Medical University/Jiangsu Province Hospital, Nanjing, China
| | - Qingju Li
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
- School of Clinical Medicine, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou, China
- Jiangsu College of Nursing, Huai'an, China
| | - Suqin Zhou
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
| | - Ran Yu
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
- School of Clinical Medicine, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou, China
- Jiangsu College of Nursing, Huai'an, China
| | - Caixia Wu
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
| | - Jiajia Chen
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
| | - Yao Xiao
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
- Jiangsu College of Nursing, Huai'an, China
| | - Haoyu Chen
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
| | - Jian Song
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
| | - Yan Pan
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
| | - Wanpeng Wang
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
- School of Clinical Medicine, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou, China
- Jiangsu College of Nursing, Huai'an, China
| |
Collapse
|
8
|
He M, Wei W, Zhang Y, Xiang Z, Peng D, Kasimumali A, Rong S. Gut microbial metabolites SCFAs and chronic kidney disease. J Transl Med 2024; 22:172. [PMID: 38369469 PMCID: PMC10874542 DOI: 10.1186/s12967-024-04974-6] [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: 11/18/2023] [Accepted: 02/11/2024] [Indexed: 02/20/2024] Open
Abstract
The global incidence of Chronic Kidney Disease (CKD) is steadily escalating, with discernible linkage to the intricate terrain of intestinal microecology. The intestinal microbiota orchestrates a dynamic equilibrium in the organism, metabolizing dietary-derived compounds, a process which profoundly impacts human health. Among these compounds, short-chain fatty acids (SCFAs), which result from microbial metabolic processes, play a versatile role in influencing host energy homeostasis, immune function, and intermicrobial signaling, etc. SCFAs emerge as pivotal risk factors influencing CKD's development and prognosis. This paper review elucidates the impact of gut microbial metabolites, specifically SCFAs, on CKD, highlighting their role in modulating host inflammatory responses, oxidative stress, cellular autophagy, the immune milieu, and signaling cascades. An in-depth comprehension of the interplay between SCFAs and kidney disease pathogenesis may pave the way for their utilization as biomarkers for CKD progression and prognosis or as novel adjunctive therapeutic strategies.
Collapse
Affiliation(s)
- Meng He
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Wenqian Wei
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yichen Zhang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Zhouxia Xiang
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Dan Peng
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Ayijiaken Kasimumali
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Shu Rong
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
9
|
Tseng CH, Shah KM, Chiu IJ, Hsiao LL. The Role of Autophagy in Type 2 Diabetic Kidney Disease Management. Cells 2023; 12:2691. [PMID: 38067119 PMCID: PMC10705810 DOI: 10.3390/cells12232691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/17/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
Diabetic kidney disease (DKD), or diabetic nephropathy (DN), is one of the most prevalent complications of type 2 diabetes mellitus (T2DM) and causes severe burden on the general welfare of T2DM patients around the world. While several new agents have shown promise in treating this condition and potentially halting the progression of the disease, more work is needed to understand the complex regulatory network involved in the disorder. Recent studies have provided new insights into the connection between autophagy, a physiological metabolic process known to maintain cellular homeostasis, and the pathophysiological pathways of DKD. Typically, autophagic activity plays a role in DKD progression mainly by promoting an inflammatory response to tissue damage, while both overactivated and downregulated autophagy worsen disease outcomes in different stages of DKD. This correlation demonstrates the potential of autophagy as a novel therapeutic target for the disease, and also highlights new possibilities for utilizing already available DN-related medications. In this review, we summarize findings on the relationship between autophagy and DKD, and the impact of these results on clinical management strategies.
Collapse
Affiliation(s)
- Che-Hao Tseng
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (C.-H.T.); (K.M.S.)
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Kavya M. Shah
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (C.-H.T.); (K.M.S.)
| | - I-Jen Chiu
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (C.-H.T.); (K.M.S.)
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- TMU-Research Center of Urology and Kidney (TMU-RCUK), Taipei Medical University, Taipei 11031, Taiwan
| | - Li-Li Hsiao
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (C.-H.T.); (K.M.S.)
| |
Collapse
|
10
|
Kumar P, Brooks HL. Sex-specific epigenetic programming in renal fibrosis and inflammation. Am J Physiol Renal Physiol 2023; 325:F578-F594. [PMID: 37560775 DOI: 10.1152/ajprenal.00091.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/18/2023] [Accepted: 07/31/2023] [Indexed: 08/11/2023] Open
Abstract
The growing prevalence of hypertension, heart disease, diabetes, and obesity along with an aging population is leading to a higher incidence of renal diseases in society. Chronic kidney disease (CKD) is characterized mainly by persistent inflammation, fibrosis, and gradual loss of renal function leading to renal failure. Sex is a known contributor to the differences in incidence and progression of CKD. Epigenetic programming is an essential regulator of renal physiology and is critically involved in the pathophysiology of renal injury and fibrosis. Epigenetic signaling integrates intrinsic and extrinsic signals onto the genome, and various environmental and hormonal stimuli, including sex hormones, which regulate gene expression and downstream cellular responses. The most extensively studied epigenetic alterations that play a critical role in renal damage include histone modifications and DNA methylation. Notably, these epigenetic alterations are reversible, making them candidates for potential therapeutic targets for the treatment of renal diseases. Here, we will summarize the current knowledge on sex differences in epigenetic modulation of renal fibrosis and inflammation and highlight some possible epigenetic therapeutic strategies for CKD treatment.
Collapse
Affiliation(s)
- Prerna Kumar
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana, United States
| | - Heddwen L Brooks
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana, United States
| |
Collapse
|
11
|
Takebayashi G, Chiba Y, Wakamatsu K, Murakami R, Miyai Y, Matsumoto K, Uemura N, Yanase K, Shirakami G, Ogino Y, Ueno M. E-Cadherin Is Expressed in Epithelial Cells of the Choroid Plexus in Human and Mouse Brains. Curr Issues Mol Biol 2023; 45:7813-7826. [PMID: 37886936 PMCID: PMC10605538 DOI: 10.3390/cimb45100492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/18/2023] [Accepted: 09/23/2023] [Indexed: 10/28/2023] Open
Abstract
Evidence showing the functional significance of the choroid plexus is accumulating. Epithelial cells with tight and adherens junctions of the choroid plexus play important roles in cerebrospinal fluid production and circadian rhythm formation. Although specific types of cadherin expressed in adherens junctions of choroid plexus epithelium (CPE) have been examined, they remained uncertain. Recent mass spectrometry and immunolocalization analysis revealed that non-epithelial cadherins, P- and N-cadherins, are expressed in the lateral membrane of CPE, whereas E-cadherin expression has not been confirmed in CPE of humans or mice. In this study, we examined E-cadherin expression in CPE of mice and humans by RT-PCR, immunohistochemical-, and Western blotting analyses. We confirmed, by using RT-PCR analysis, the mRNA expression of E-cadherin in the choroid plexus of mice. The immunohistochemical expression of E-cadherin was noted in the lateral membrane of CPE of mice and humans. We further confirmed, in Western blotting, the specific immunoreactivity for E-cadherin. Immunohistochemically, the expression of E- and N-cadherins or vimentin was unevenly distributed in some CPE, whereas that of E- and P-cadherins or β-catenin frequently co-existed in other CPE. These findings indicate that E-cadherin is expressed in the lateral membrane of CPE, possibly correlated with the expression of other cadherins and cytoplasmic proteins.
Collapse
Affiliation(s)
- Genta Takebayashi
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Takamatsu 761-0793, Kagawa, Japan; (G.T.); (Y.C.); (K.W.); (R.M.); (Y.M.); (K.M.)
- Department of Anesthesiology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Takamatsu 761-0793, Kagawa, Japan; (N.U.); (K.Y.); (G.S.); (Y.O.)
| | - Yoichi Chiba
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Takamatsu 761-0793, Kagawa, Japan; (G.T.); (Y.C.); (K.W.); (R.M.); (Y.M.); (K.M.)
| | - Keiji Wakamatsu
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Takamatsu 761-0793, Kagawa, Japan; (G.T.); (Y.C.); (K.W.); (R.M.); (Y.M.); (K.M.)
| | - Ryuta Murakami
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Takamatsu 761-0793, Kagawa, Japan; (G.T.); (Y.C.); (K.W.); (R.M.); (Y.M.); (K.M.)
| | - Yumi Miyai
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Takamatsu 761-0793, Kagawa, Japan; (G.T.); (Y.C.); (K.W.); (R.M.); (Y.M.); (K.M.)
| | - Koichi Matsumoto
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Takamatsu 761-0793, Kagawa, Japan; (G.T.); (Y.C.); (K.W.); (R.M.); (Y.M.); (K.M.)
| | - Naoya Uemura
- Department of Anesthesiology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Takamatsu 761-0793, Kagawa, Japan; (N.U.); (K.Y.); (G.S.); (Y.O.)
| | - Ken Yanase
- Department of Anesthesiology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Takamatsu 761-0793, Kagawa, Japan; (N.U.); (K.Y.); (G.S.); (Y.O.)
| | - Gotaro Shirakami
- Department of Anesthesiology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Takamatsu 761-0793, Kagawa, Japan; (N.U.); (K.Y.); (G.S.); (Y.O.)
| | - Yuichi Ogino
- Department of Anesthesiology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Takamatsu 761-0793, Kagawa, Japan; (N.U.); (K.Y.); (G.S.); (Y.O.)
| | - Masaki Ueno
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Takamatsu 761-0793, Kagawa, Japan; (G.T.); (Y.C.); (K.W.); (R.M.); (Y.M.); (K.M.)
| |
Collapse
|
12
|
Liu Z, Liu J, Wang W, An X, Luo L, Yu D, Sun W. Epigenetic modification in diabetic kidney disease. Front Endocrinol (Lausanne) 2023; 14:1133970. [PMID: 37455912 PMCID: PMC10348754 DOI: 10.3389/fendo.2023.1133970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 05/30/2023] [Indexed: 07/18/2023] Open
Abstract
Diabetic kidney disease (DKD) is a common microangiopathy in diabetic patients and the main cause of death in diabetic patients. The main manifestations of DKD are proteinuria and decreased renal filtration capacity. The glomerular filtration rate and urinary albumin level are two of the most important hallmarks of the progression of DKD. The classical treatment of DKD is controlling blood glucose and blood pressure. However, the commonly used clinical therapeutic strategies and the existing biomarkers only partially slow the progression of DKD and roughly predict disease progression. Therefore, novel therapeutic methods, targets and biomarkers are urgently needed to meet clinical requirements. In recent years, increasing attention has been given to the role of epigenetic modification in the pathogenesis of DKD. Epigenetic variation mainly includes DNA methylation, histone modification and changes in the noncoding RNA expression profile, which are deeply involved in DKD-related inflammation, oxidative stress, hemodynamics, and the activation of abnormal signaling pathways. Since DKD is reversible at certain disease stages, it is valuable to identify abnormal epigenetic modifications as early diagnosis and treatment targets to prevent the progression of end-stage renal disease (ESRD). Because the current understanding of the epigenetic mechanism of DKD is not comprehensive, the purpose of this review is to summarize the role of epigenetic modification in the occurrence and development of DKD and evaluate the value of epigenetic therapies in DKD.
Collapse
Affiliation(s)
- Zhe Liu
- Public Research Platform, First Hospital of Jilin University, Changchun, Jilin, China
- College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Jiahui Liu
- Public Research Platform, First Hospital of Jilin University, Changchun, Jilin, China
| | - Wanning Wang
- Department of Nephrology, First Hospital of Jilin University, Changchun, Jilin, China
| | - Xingna An
- Public Research Platform, First Hospital of Jilin University, Changchun, Jilin, China
| | - Ling Luo
- Public Research Platform, First Hospital of Jilin University, Changchun, Jilin, China
| | - Dehai Yu
- Public Research Platform, First Hospital of Jilin University, Changchun, Jilin, China
| | - Weixia Sun
- Department of Nephrology, First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
13
|
Li Y, Li K, Zhao W, Wang H, Xue X, Chen X, Li W, Xu P, Wang K, Liu P, Tian X, Fu R. VPA improves ferroptosis in tubular epithelial cells after cisplatin-induced acute kidney injury. Front Pharmacol 2023; 14:1147772. [PMID: 37153759 PMCID: PMC10155836 DOI: 10.3389/fphar.2023.1147772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/31/2023] [Indexed: 05/10/2023] Open
Abstract
Background: As a novel non-apoptotic cell death, ferroptosis has been reported to play a crucial role in acute kidney injury (AKI), especially cisplatin-induced AKI. Valproic acid (VPA), an inhibitor of histone deacetylase (HDAC) 1 and 2, is used as an antiepileptic drug. Consistent with our data, a few studies have demonstrated that VPA protects against kidney injury in several models, but the detailed mechanism remains unclear. Results: In this study, we found that VPA prevents against cisplatin-induced renal injury via regulating glutathione peroxidase 4 (GPX4) and inhibiting ferroptosis. Our results mainly indicated that ferroptosis presented in tubular epithelial cells of AKI humans and cisplatin-induced AKI mice. VPA or ferrostatin-1 (ferroptosis inhibitor, Fer-1) reduced cisplatin-induced AKI functionally and pathologically, which was characterized by reduced serum creatinine, blood urea nitrogen, and tissue damage in mice. Meanwhile, VPA or Fer-1 treatment in both in vivo and in vitro models, decreased cell death, lipid peroxidation, and expression of acyl-CoA synthetase long-chain family member 4 (ACSL4), reversing downregulation of GPX4. In addition, our study in vitro indicated that GPX4 inhibition by siRNA significantly weakened the protective effect of VPA after cisplatin treatment. Conclusion: Ferroptosis plays an essential role in cisplatin-induced AKI and inhibiting ferroptosis through VPA to protect against renal injury is a viable treatment in cisplatin-induced AKI.
Collapse
Affiliation(s)
- Yan Li
- Department of Nephrology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Ke Li
- Department of Nephrology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Weihao Zhao
- Department of Nephrology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Haodong Wang
- Department of Nephrology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xiaodong Xue
- School of Computer Science, National University of Singapore, Singapore, Singapore
| | - Xianghui Chen
- Department of Nephrology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Wantao Li
- Department of Nephrology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Peihao Xu
- School of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Kexin Wang
- Department of Nephrology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Pengfei Liu
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xuefei Tian
- Section of Nephrology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
- *Correspondence: Rongguo Fu, ; Xuefei Tian,
| | - Rongguo Fu
- Department of Nephrology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- *Correspondence: Rongguo Fu, ; Xuefei Tian,
| |
Collapse
|
14
|
Gao M, Yang N, Lei Y, Zhang W, Liu H, Lin H. Tannic acid antagonizes atrazine exposure-induced autophagy and DNA damage crosstalk in grass carp hepatocytes via NO/iNOS/NF-κB signaling pathway to maintain stable immune function. FISH & SHELLFISH IMMUNOLOGY 2022; 131:1075-1084. [PMID: 36396070 DOI: 10.1016/j.fsi.2022.11.024] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/10/2022] [Accepted: 11/12/2022] [Indexed: 06/16/2023]
Abstract
Atrazine (ATR) is a herbicide widely used in grass crops. The pollution of the soil and water environment is extremely harmful to aquatic animals and their offspring. iNOS/NO upregulation, DNA damage and cellular autophagy affect the immune function of fish liver cells. The effects of ATR at exposure doses on grass carp hepatocytes in terms of autophagy and DNA damage effects in genotoxicity, as well as the antagonistic effects of TAN on the above phenotypes and the internal mechanisms are not known. Therefore, we constructed control (Con group), ATR exposure (ATR group), TAN exposure (TAN group) and mixed group (ATR + TAN group) models on grass carp hepatocytes. Validation was performed by comet assay, MDC staining, qRT-PCR and protein blotting assay as well as iNOS/NO indicator levels and expression of immune factors as these experimental methods. Our data indicate that iNOS/NO assay kit measured that ATR treatment resulted in a significant increase in iNOS/NO activity and levels in grass carp hepatocytes (p < 0.05). We also found that NO/iNOS/NF-κB pathway genes were significantly activated (p < 0.05) at the exposure dose of ATR (3 μg mL-1). In addition, the proportion of cells that died due to DNA damage, autophagy, and immunotoxic effects was significantly increased at the exposure dose of ATR. Comet assay protein blotting detected increased DNA damage in cells at the ATR exposure dose (p < 0.05). MDC staining and qRT-PCR and protein blotting to detect the proportion of autophagic cells and autophagy-related genes also appeared upregulated at the exposed dose of ATR (p < 0.05). In brief, this study showed that ATR exposure caused cellular DNA damage and autophagy via the NO/iNOS/NF-κB axis, which led to immunotoxic effects and eventual death of grass carp hepatocytes. The present study facilitates the demonstration of the molecular mechanism of TAN alleviation of ATR cytotoxicity from the perspective of NO-mediated iNOS/NF-κB axis. It provides insights into the protection of farmed fish from agricultural contaminants and opens up new horizons in the use of natural plant-derived monomers for the clinical treatment of antagonistic triazine pesticide poisoning.
Collapse
Affiliation(s)
- Meichen Gao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Naixi Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Yutian Lei
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Wenyue Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Huanyi Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Hongjin Lin
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China.
| |
Collapse
|
15
|
Valproic acid attenuates cellular senescence in diabetic kidney disease through the inhibition of complement C5a receptors. Sci Rep 2022; 12:20278. [PMID: 36434087 PMCID: PMC9700697 DOI: 10.1038/s41598-022-24851-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022] Open
Abstract
Despite increasing knowledge about the factors involved in the progression of diabetic complications, diabetic kidney disease (DKD) continues to be a major health burden. Current therapies only slow but do not prevent the progression of DKD. Thus, there is an urgent need to develop novel therapy to halt the progression of DKD and improve disease prognosis. In our preclinical study where we administered a histone deacetylase (HDAC) inhibitor, valproic acid, to streptozotocin-induced diabetic mice, albuminuria and glomerulosclerosis were attenuated. Furthermore, we discovered that valproic acid attenuated diabetes-induced upregulation of complement C5a receptors, with a concomitant reduction in markers of cellular senescence and senescence-associated secretory phenotype. Interestingly, further examination of mice lacking the C5a receptor 1 (C5aR1) gene revealed that cellular senescence was attenuated in diabetes. Similar results were observed in diabetic mice treated with a C5aR1 inhibitor, PMX53. RNA-sequencing analyses showed that PMX53 significantly regulated genes associated with cell cycle pathways leading to cellular senescence. Collectively, these results for the first time demonstrated that complement C5a mediates cellular senescence in diabetic kidney disease. Cellular senescence has been implicated in the pathogenesis of diabetic kidney disease, thus therapies to inhibit cellular senescence such as complement inhibitors present as a novel therapeutic option to treat diabetic kidney disease.
Collapse
|
16
|
Siddhi J, Sherkhane B, Kalavala AK, Arruri V, Velayutham R, Kumar A. Melatonin prevents diabetes‐induced nephropathy by modulating the AMPK/SIRT1 axis: Focus on autophagy and mitochondrial dysfunction. Cell Biol Int 2022; 46:2142-2157. [DOI: 10.1002/cbin.11899] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/26/2022] [Accepted: 08/22/2022] [Indexed: 02/06/2023]
Affiliation(s)
- Jain Siddhi
- Department of Pharmacology and Toxicology National Institute of Pharmaceutical Education and Research (NIPER)‐Hyderabad Balanagar India
| | - Bhoomika Sherkhane
- Department of Pharmacology and Toxicology National Institute of Pharmaceutical Education and Research (NIPER)‐Hyderabad Balanagar India
| | - Anil Kumar Kalavala
- Department of Pharmaceutics, College of Pharmacy and Pharmaceutical Science Florida A&M University Tallahassee Florida USA
| | - Vijay Arruri
- Department of Neurological Surgery University of Wisconsin‐Madison Madison Wisconsin USA
| | - Ravichandiran Velayutham
- Department of Natural Products National Institute of Pharmaceutical Education and Research (NIPER)‐Kolkata Kolkata India
| | - Ashutosh Kumar
- Department of Pharmacology and Toxicology National Institute of Pharmaceutical Education and Research (NIPER)‐Hyderabad Balanagar India
- Department of Pharmacology and Toxicology National Institute of Pharmaceutical Education and Research (NIPER)‐Kolkata Kolkata India
| |
Collapse
|
17
|
Wang S, Zhang X, Wang Q, Wang R. Histone modification in podocyte injury of diabetic nephropathy. J Mol Med (Berl) 2022; 100:1373-1386. [PMID: 36040515 DOI: 10.1007/s00109-022-02247-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 07/31/2022] [Accepted: 08/17/2022] [Indexed: 11/24/2022]
Abstract
Diabetic nephropathy (DN), an important complication of diabetic microvascular disease, is one of the leading causes of end-stage renal disease (ESRD), which brings heavy burdens to the whole society. Podocytes are terminally differentiated glomerular cells, which act as a pivotal component of glomerular filtration barrier. When podocytes are injured, glomerular filtration barrier is damaged, and proteinuria would occur. Dysfunction of podocytes contributes to DN. And degrees of podocyte injury influence prognosis of DN. Growing evidences have shown that epigenetics does a lot in the evolvement of podocyte injury. Epigenetics includes DNA methylation, histone modification, and non-coding RNA. Among them, histone modification plays an indelible role. Histone modification includes histone methylation, histone acetylation, and other modifications such as histone phosphorylation, histone ubiquitination, histone ADP-ribosylation, histone crotonylation, and histone β-hydroxybutyrylation. It can affect chromatin structure and regulate gene transcription to exert its function. This review is to summarize documents about pathogenesis of podocyte injury, most importantly, histone modification of podocyte injury in DN recently to provide new ideas for further molecular research, diagnosis, and treatment.
Collapse
Affiliation(s)
- Simeng Wang
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, 250012, Shandong, China
| | - Xinyu Zhang
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, 250012, Shandong, China
| | - Qinglian Wang
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, 250012, Shandong, China. .,Department of Nephrology, Shandong Provincial Hospital, Shandong First Medical University, No. 324 Jingwu Street, Jinan, 250021, Shandong, China.
| | - Rong Wang
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, 250012, Shandong, China. .,Department of Nephrology, Shandong Provincial Hospital, Shandong First Medical University, No. 324 Jingwu Street, Jinan, 250021, Shandong, China.
| |
Collapse
|
18
|
Zhang Z, Sun Y, Xue J, Jin D, Li X, Zhao D, Lian F, Qi W, Tong X. The critical role of dysregulated autophagy in the progression of diabetic kidney disease. Front Pharmacol 2022; 13:977410. [PMID: 36091814 PMCID: PMC9453227 DOI: 10.3389/fphar.2022.977410] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/04/2022] [Indexed: 11/30/2022] Open
Abstract
Diabetic kidney disease (DKD) is one of the major public health problems in society today. It is a renal complication caused by diabetes mellitus with predominantly microangiopathy and is a major cause of end-stage renal disease (ESRD). Autophagy is a metabolic pathway for the intracellular degradation of cytoplasmic products and damaged organelles and plays a vital role in maintaining homeostasis and function of the renal cells. The dysregulation of autophagy in the hyperglycaemic state of diabetes mellitus can lead to the progression of DKD, and the activation or restoration of autophagy through drugs is beneficial to the recovery of renal function. This review summarizes the physiological process of autophagy, illustrates the close link between DKD and autophagy, and discusses the effects of drugs on autophagy and the signaling pathways involved from the perspective of podocytes, renal tubular epithelial cells, and mesangial cells, in the hope that this will be useful for clinical treatment.
Collapse
Affiliation(s)
- Ziwei Zhang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yuting Sun
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiaojiao Xue
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - De Jin
- Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, China
| | - Xiangyan Li
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Daqing Zhao
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Fengmei Lian
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Fengmei Lian, ; Wenxiu Qi, ; Xiaolin Tong,
| | - Wenxiu Qi
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
- *Correspondence: Fengmei Lian, ; Wenxiu Qi, ; Xiaolin Tong,
| | - Xiaolin Tong
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Fengmei Lian, ; Wenxiu Qi, ; Xiaolin Tong,
| |
Collapse
|
19
|
Kushwaha K, Garg SS, Gupta J. Targeting epigenetic regulators for treating diabetic nephropathy. Biochimie 2022; 202:146-158. [PMID: 35985560 DOI: 10.1016/j.biochi.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 07/01/2022] [Accepted: 08/02/2022] [Indexed: 11/25/2022]
Abstract
Diabetes is accompanied by the worsening of kidney functions. The reasons for kidney dysfunction mainly include high blood pressure (BP), high blood sugar levels, and genetic makeup. Vascular complications are the leading cause of the end-stage renal disorder (ESRD) and death of diabetic patients. Epigenetics has emerged as a new area to explain the inheritance of non-mendelian conditions like diabetic kidney diseases. Aberrant post-translational histone modifications (PTHMs), DNA methylation (DNAme), and miRNA constitute major epigenetic mechanisms that progress diabetic nephropathy (DN). Increased blood sugar levels alter PTHMs, DNAme, and miRNA in kidney cells results in aberrant gene expression that causes fibrosis, accumulation of extracellular matrix (ECM), increase in reactive oxygen species (ROS), and renal injuries. Histone acetylation (HAc) and histone deacetylation (HDAC) are the most studied epigenetic modifications with implications in the occurrence of kidney disorders. miRNAs induced by hyperglycemia in renal cells are also responsible for ECM accumulation and dysfunction of the glomerulus. In this review, we highlight the role of epigenetic modifications in DN progression and current strategies employed to ameliorate DN.
Collapse
Affiliation(s)
- Kriti Kushwaha
- Department of Biotechnology, School of Bioengineering and Bioscience, Lovely Professional University, Phagwara, Punjab, India
| | - Sourbh Suren Garg
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Jeena Gupta
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India.
| |
Collapse
|
20
|
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: 2.5] [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
|
21
|
Naito S, Kawashima N, Ishii D, Fujita T, Iwamura M, Takeuchi Y. Decreased GM3 correlates with proteinuria in minimal change nephrotic syndrome and focal segmental glomerulosclerosis. Clin Exp Nephrol 2022; 26:1078-1085. [PMID: 35804208 DOI: 10.1007/s10157-022-02249-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/18/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Glycolipids on cell membrane rafts play various roles by interacting with glycoproteins. Recently, it was reported that the glycolipid GM3 is expressed in podocytes and may play a role in podocyte protection. In this report, we describe the correlation between changes in GM3 expression in glomeruli and proteinuria in minimal change nephrotic syndrome (MCNS) and focal segmental glomerulosclerosis (FSGS) patients. METHODS We performed a case-control study of the correlation between nephrin/GM3 expression levels and proteinuria in MCNS and FSGS patients who underwent renal biopsy at our institution between 2009 and 2014. Normal renal tissue sites were used from patients who had undergone nephrectomy at our institution and gave informed consent. RESULTS Both MCNS and FSGS had decreased GM3 and Nephrin expression compared with the normal (normal vs. MCNS, FSGS; all p < 0.01). Furthermore, in both MCNS and FSGS, GM3 expression was negatively correlated with proteinuria (MCNS: r = - 0.61, p < 0.01, FSGS: r = - 0.56, p < 0.05). However, nephrin expression had a trend to correlate with proteinuria in FSGS (MCNS: r = 0.19, p = 0.58, FSGS: r = - 0.48, p = 0.06). Furthermore, in a simple linear regression analysis, GM3 expression also correlated with proteinuric change after 12 months of treatment (MCNS: r = 0.40, p = 0.38, FSGS: r = 0. 68, p < 0.05). CONCLUSION We showed for the first time that decreased GM3 expression correlates with proteinuria in MCNS and FSGS patients. Further studies are needed on the podocyte-protective effects of GM3.
Collapse
Affiliation(s)
- Shokichi Naito
- Department of Nephrology, School of Medicine, Kitasato University, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa, 252-0374, Japan.
| | - Nagako Kawashima
- Department of Nephrology, School of Medicine, Kitasato University, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa, 252-0374, Japan
| | - Daisuke Ishii
- Department of Urology, School of Medicine, Kitasato University, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa, 252-0374, Japan
| | - Tetsuo Fujita
- Department of Urology, School of Medicine, Kitasato University, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa, 252-0374, Japan
| | - Masatsugu Iwamura
- Department of Urology, School of Medicine, Kitasato University, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa, 252-0374, Japan
| | - Yasuo Takeuchi
- Department of Nephrology, School of Medicine, Kitasato University, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa, 252-0374, Japan
| |
Collapse
|
22
|
Shen F, Zhuang S. Histone Acetylation and Modifiers in Renal Fibrosis. Front Pharmacol 2022; 13:760308. [PMID: 35559244 PMCID: PMC9086452 DOI: 10.3389/fphar.2022.760308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 04/04/2022] [Indexed: 12/23/2022] Open
Abstract
Histones are the most abundant proteins bound to DNA in eukaryotic cells and frequently subjected to post-modifications such as acetylation, methylation, phosphorylation and ubiquitination. Many studies have shown that histone modifications, especially histone acetylation, play an important role in the development and progression of renal fibrosis. Histone acetylation is regulated by three families of proteins, including histone acetyltransferases (HATs), histone deacetylases (HDACs) and bromodomain and extraterminal (BET) proteins. These acetylation modifiers are involved in a variety of pathophysiological processes leading to the development of renal fibrosis, including partial epithelial-mesenchymal transition, renal fibroblast activation, inflammatory response, and the expression of pro-fibrosis factors. In this review, we summarize the role and regulatory mechanisms of HATs, HDACs and BET proteins in renal fibrosis and provide evidence for targeting these modifiers to treat various chronic fibrotic kidney diseases in animal models.
Collapse
Affiliation(s)
- Fengchen Shen
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shougang Zhuang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, RI, United States
| |
Collapse
|
23
|
Dhawan P, Vasishta S, Balakrishnan A, Joshi MB. Mechanistic insights into glucose induced vascular epigenetic reprogramming in type 2 diabetes. Life Sci 2022; 298:120490. [DOI: 10.1016/j.lfs.2022.120490] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/22/2022] [Accepted: 03/16/2022] [Indexed: 12/13/2022]
|
24
|
The HDAC2/SP1/miR-205 feedback loop contributes to tubular epithelial cell extracellular matrix production in diabetic kidney disease. Clin Sci (Lond) 2022; 136:223-238. [PMID: 35084460 DOI: 10.1042/cs20210470] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 01/23/2022] [Accepted: 01/27/2022] [Indexed: 11/17/2022]
Abstract
Extracellular matrix (ECM) accumulation is considered an important pathological feature of diabetic kidney disease (DKD). Histone deacetylase (HDAC) inhibitors protect against kidney injury. However, the potential mechanisms of HDACs in DKD are still largely unknown. Here, we describe a novel feedback loop composed of HDAC2 and miR-205 that regulates ECM production in tubular epithelial cells in individuals with DKD. We found that HDAC2 mRNA expression in peripheral blood was markedly higher in patients with DKD than in patients with diabetes. Nuclear HDAC2 protein expression was increased in TGFβ1-stimulated tubular epithelial cells and db/db mice. We also found that miR-205 was regulated by HDAC2 and downregulated in TGFβ1-treated HK2 cells and db/db mice. In addition, HDAC2 reduced histone H3K9 acetylation in the miR-205 promoter region to inhibit its promoter activity and subsequently suppressed miR-205 expression through an SP1-mediated pathway. Furthermore, miR-205 directly targeted HDAC2 and inhibited HDAC2 expression. Intriguingly, miR-205 also regulated its own transcription by inhibiting HDAC2 and increasing histone H3K9 acetylation in its promoter, forming a feedback regulatory loop. Additionally, the miR-205 agonist attenuated ECM production in HK2 cells and renal interstitial fibrosis in db/db mice. In conclusion, the HDAC2/SP1/miR-205 feedback loop may be crucial for the pathogenesis of DKD.
Collapse
|
25
|
Charan HV, Dwivedi DK, Khan S, Jena G. Mechanisms of NLRP3 inflammasome-mediated hepatic stellate cell activation: therapeutic potential for liver fibrosis. Genes Dis 2022; 10:480-494. [DOI: 10.1016/j.gendis.2021.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 11/09/2021] [Accepted: 12/01/2021] [Indexed: 01/18/2023] Open
|
26
|
Varghese R, Majumdar A. A New Prospect for the Treatment of Nephrotic Syndrome Based on Network Pharmacology Analysis. Curr Res Physiol 2022; 5:36-47. [PMID: 35098155 PMCID: PMC8783131 DOI: 10.1016/j.crphys.2021.12.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 11/10/2021] [Accepted: 12/29/2021] [Indexed: 12/11/2022] Open
Abstract
Network pharmacology is an emerging field which is currently capturing interest in drug discovery and development. Chronic kidney conditions have become a threat globally due to its associated lifelong therapies. Nephrotic syndrome (NS) is a common glomerular disease that is seen in paediatric and adult population with characteristic manifestation of proteinuria, oedema, hypoalbuminemia, and hyperlipidemia. It involves podocyte damage with tubulointerstitial fibrosis and glomerulosclerosis. Till date there has been no specific treatment available for this condition that provides complete remission. Repurposing of drugs can thus be a potential strategy for the treatment of NS. Recently, epigenetic mechanisms were identified that promote progression of many renal diseases. Therefore, in the present study, we investigated two epigenetic drugs valproic acid (VPA) and all-trans retinoic acid (ATRA). Epigenetic drugs act by binging about changes in gene expression without altering the DNA sequence. The changes include DNA methylation or histone modifications. The targets for the two drugs ATRA and VPA were collated from ChEMBL and Binding DB. All the genes associated with NS were collected from DisGeNET and KEGG database. Interacting proteins for the target genes were acquired from STRING database. The genes were then subjected to gene ontology and pathway enrichment analysis using a functional enrichment software tool. A drug-target and drug-potential target-protein interaction network was constructed using the Cytoscape software. Our results revealed that the two drugs VPA and ATRA had 65 common targets that contributed to kidney diseases. Out of which, 25 targets were specifically NS associated. Further, our work exhibited that ATRA and VPA were synergistically involved in pathways of inflammation, renal fibrosis, glomerulosclerosis and possibly mitochondrial biogenesis and endoplasmic reticulum stress. We thus propose a synergistic potential of the two drugs for treating chronic kidney diseases, specifically NS. The outcomes will undoubtedly invigorate further preclinical and clinical explorative studies. We identify network pharmacology as an initial inherent approach in identifying drug candidates for repurposing and synergism.
Collapse
Affiliation(s)
- Rini Varghese
- Bombay College of Pharmacy, Kalina, Santacruz (E), Mumbai, Maharashtra, 400098, India
| | - Anuradha Majumdar
- Bombay College of Pharmacy, Kalina, Santacruz (E), Mumbai, Maharashtra, 400098, India
| |
Collapse
|
27
|
Teh YM, Mualif SA, Lim SK. A comprehensive insight into autophagy and its potential signaling pathways as a therapeutic target in podocyte injury. Int J Biochem Cell Biol 2021; 143:106153. [PMID: 34974186 DOI: 10.1016/j.biocel.2021.106153] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 12/23/2021] [Accepted: 12/29/2021] [Indexed: 02/06/2023]
Abstract
As part of the glomerular filtration membrane, podocyte is terminally differentiated, structurally unique, and highly specialized in maintaining kidney function. Proteinuria caused by podocyte injury (foot process effacement) is the clinical symptom of various kidney diseases (CKD), including nephrotic syndrome. Podocyte autophagy has become a powerful therapeutic strategy target in ameliorating podocyte injury. Autophagy is known to be associated significantly with sirtuin-1, proteinuria, and podocyte injury. Various key findings in podocyte autophagy were reported in the past ten years, such as the role of endoplasmic reticulum (ER) stress in podocyte autophagy impairment, podocyte autophagy-related gene, essential roles of the signaling pathways: Mammalian Target of Rapamycin (mTOR)/ Phosphoinositide 3-kinase (PI3k)/ serine/threonine kinase 1 (Akt) in podocyte autophagy. These significant factors caused podocyte injury associated with autophagy impairment. Sirtuin-1 was reported to have a vital key role in mTOR signaling, 5'AMP-activated protein kinase (AMPK) regulation, autophagy activation, and various critical pathways associated with podocyte's function and health; it has potential value to podocyte injury pathogenesis investigation. From these findings, podocyte autophagy has become an attractive therapeutic strategy to ameliorate podocyte injury, and this review will provide an in-depth review on therapeutic targets he podocyte autophagy.
Collapse
Affiliation(s)
- Yoong Mond Teh
- School of Biomedical Engineering and Health Sciences, Faculty of Engineering, Universiti Teknologi Malaysia (UTM), Johor Bahru, Malaysia
| | - Siti Aisyah Mualif
- School of Biomedical Engineering and Health Sciences, Faculty of Engineering, Universiti Teknologi Malaysia (UTM), Johor Bahru, Malaysia; Medical Device and Technology Centre (MEDiTEC), Universiti Teknologi Malaysia, Malaysia
| | - Soo Kun Lim
- Renal Division, Department of Medicine, Faculty of Medicine, University of Malaya (UM), Kuala Lumpur, Malaysia.
| |
Collapse
|
28
|
Abstract
Diabetic nephropathy (DN), which is a common microvascular complication with a high incidence in diabetic patients, greatly increases the mortality of patients. With further study on DN, it is found that epigenetics plays a crucial role in the pathophysiological process of DN. Epigenetics has an important impact on the development of DN through a variety of mechanisms, and promotes the generation and maintenance of metabolic memory, thus ultimately leading to a poor prognosis. In this review we discuss the methylation of DNA, modification of histone, and regulation of non-coding RNA involved in the progress of cell dysfunction, inflammation and fibrosis in the kidney, which ultimately lead to the deterioration of DN.
Collapse
|
29
|
Singh D, Gupta S, Verma I, Morsy MA, Nair AB, Ahmed ASF. Hidden pharmacological activities of valproic acid: A new insight. Biomed Pharmacother 2021; 142:112021. [PMID: 34463268 DOI: 10.1016/j.biopha.2021.112021] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 07/28/2021] [Accepted: 08/07/2021] [Indexed: 12/24/2022] Open
Abstract
Valproic acid (VPA) is an approved drug for managing epileptic seizures, bipolar disorders, and migraine. VPA has been shown to elevate the level of gamma-aminobutyric acid (GABA) in the brain through competitive inhibition of GABA transaminase, thus promoting the availability of synaptic GABA and facilitating GABA-mediated responses. VPA, which is a small chain of fatty acids, prevents histone deacetylases (HDACs). HDACs play a crucial role in chromatin remodeling and gene expression through posttranslational changes of chromatin-associated histones. Recent studies reported a possible effect of VPA against particular types of cancers. This effect was partially attributed to its role in regulating epigenetic modifications through the inhibition of HDACs, which affect the expression of genes associated with cell cycle control, cellular differentiation, and apoptosis. In this review, we summarize the current information on the actions of VPA in diseases such as diabetes mellitus, kidney disorders, neurodegenerative diseases, muscular dystrophy, and cardiovascular disorders.
Collapse
Affiliation(s)
- Dhirendra Singh
- Department of Pharmacology, M.M. College of Pharmacy, M.M. (Deemed to be University), Mullana, Ambala, Haryana, India
| | - Sumeet Gupta
- Department of Pharmacology, M.M. College of Pharmacy, M.M. (Deemed to be University), Mullana, Ambala, Haryana, India.
| | - Inderjeet Verma
- Department of Pharmacology, M.M. College of Pharmacy, M.M. (Deemed to be University), Mullana, Ambala, Haryana, India
| | - Mohamed A Morsy
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia; Department of Pharmacology, Faculty of Medicine, Minia University, El-Minia, Egypt
| | - Anroop B Nair
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Al-Shaimaa F Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, El-Minia, Egypt
| |
Collapse
|
30
|
Abstract
Epigenetics examines heritable changes in DNA and its associated proteins except mutations in gene sequence. Epigenetic regulation plays fundamental roles in kidney cell biology through the action of DNA methylation, chromatin modification via epigenetic regulators and non-coding RNA species. Kidney diseases, including acute kidney injury, chronic kidney disease, diabetic kidney disease and renal fibrosis are multistep processes associated with numerous molecular alterations even in individual kidney cells. Epigenetic alterations, including anomalous DNA methylation, aberrant histone alterations and changes of microRNA expression all contribute to kidney pathogenesis. These changes alter the genome-wide epigenetic signatures and disrupt essential pathways that protect renal cells from uncontrolled growth, apoptosis and development of other renal associated syndromes. Molecular changes impact cellular function within kidney cells and its microenvironment to drive and maintain disease phenotype. In this chapter, we briefly summarize epigenetic mechanisms in four kidney diseases including acute kidney injury, chronic kidney disease, diabetic kidney disease and renal fibrosis. We primarily focus on current knowledge about the genome-wide profiling of DNA methylation and histone modification, and epigenetic regulation on specific gene(s) in the pathophysiology of these diseases and the translational potential of identifying new biomarkers and treatment for prevention and therapy. Incorporating epigenomic testing into clinical research is essential to elucidate novel epigenetic biomarkers and develop precision medicine using emerging therapies.
Collapse
|
31
|
Abdolahi M, Karimi E, Sarraf P, Tafakhori A, Siri G, Salehinia F, Sedighiyan M, Asanjarani B, Badeli M, Abdollahi H, Yoosefi N, Yousefi A, Rad AS, Djalali M. The omega-3 and Nano-curcumin effects on vascular cell adhesion molecule (VCAM) in episodic migraine patients: a randomized clinical trial. BMC Res Notes 2021; 14:283. [PMID: 34301320 PMCID: PMC8305494 DOI: 10.1186/s13104-021-05700-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 07/14/2021] [Indexed: 02/07/2023] Open
Abstract
Objective The purpose of this clinical trial was to examine the effect of omega-3 fatty acids (W-3 FAs), nanocurcumin and their combination on serum levels and gene expression of VCAM in patients with episodic migraine. Results In this study, 80 patients were randomly divided in to 4 groups to receive for 2 months. Both serum levels and gene expression of VCAM showed remarkable decreases after single W-3 and after combined W-3 and nanocurcumin interventions. However, a borderline significant change and no remarkable change were observed after single nanocurcumin supplementation and in control group, respectively. While a significant difference between study groups in VCAM concentrations existed, there was no meaningful difference in VCAM gene expression among groups. It appears that the W-3 and combined W-3 and nanocurcumin can relieve VCAM serum level and its gene expression in patients with episodic migraine. Moreover, the combination of W-3 with nanocurcumin might cause more significant declines in VCAM level in the serum of migraine patients than when W-3 is administered alone. Trial Registration: This study was registered in Iranian Registry of Clinical Trials (IRCT) with ID number: NCT02532023. Supplementary Information The online version contains supplementary material available at 10.1186/s13104-021-05700-x.
Collapse
Affiliation(s)
- Mina Abdolahi
- Amir Alam Hospital Complexes, Tehran University of Medical Sciences, Sa'adi Street, Tehran, Iran
| | - Elmira Karimi
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Poursina Street, Tehran, Iran
| | - Payam Sarraf
- Iranian Centre of Neurological Research, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.,Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Tafakhori
- Iranian Centre of Neurological Research, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.,Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Goli Siri
- Department of Internal Medicine, Amiralam Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Farahnaz Salehinia
- Department of Internal Medicine, Amiralam Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Sedighiyan
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Poursina Street, PO Box: 14155-6446, Tehran, Iran
| | - Behzad Asanjarani
- Department of Internal Medicine, Amiralam Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mostafa Badeli
- Department of Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Poursina Street, Tehran, Iran
| | - Hamed Abdollahi
- Department of Anesthesiology, Amir Alam Hospital Complexes, Tehran University of Medical Sciences, Sa'adi Street, Tehran, Iran
| | - Niyoosha Yoosefi
- Honours Cellular Anatomical Physiology, University of British Columbia, Vancouver, BC, Canada
| | - Abolghasem Yousefi
- Department of Anesthesiology, Amir Alam Hospital Complexes, Tehran University of Medical Sciences, Sa'adi Street, Tehran, Iran
| | - Amir Shayegan Rad
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Poursina Street, PO Box: 14155-6446, Tehran, Iran
| | - Mahmoud Djalali
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Poursina Street, PO Box: 14155-6446, Tehran, Iran.
| |
Collapse
|
32
|
Shao BY, Zhang SF, Li HD, Meng XM, Chen HY. Epigenetics and Inflammation in Diabetic Nephropathy. Front Physiol 2021; 12:649587. [PMID: 34025445 PMCID: PMC8131683 DOI: 10.3389/fphys.2021.649587] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 04/12/2021] [Indexed: 12/11/2022] Open
Abstract
Diabetic nephropathy (DN) leads to high morbidity and disability. Inflammation plays a critical role in the pathogenesis of DN, which involves renal cells and immune cells, the microenvironment, as well as extrinsic factors, such as hyperglycemia, chemokines, cytokines, and growth factors. Epigenetic modifications usually regulate gene expression via DNA methylation, histone modification, and non-coding RNAs without altering the DNA sequence. During the past years, numerous studies have been published to reveal the mechanisms of epigenetic modifications that regulate inflammation in DN. This review aimed to summarize the latest evidence on the interplay of epigenetics and inflammation in DN, and highlight the potential targets for treatment and diagnosis of DN.
Collapse
Affiliation(s)
- Bao-Yi Shao
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Shao-Fei Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Hai-Di Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Xiao-Ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Hai-Yong Chen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China
- Department of Chinese Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
33
|
Zheng W, Guo J, Liu ZS. Effects of metabolic memory on inflammation and fibrosis associated with diabetic kidney disease: an epigenetic perspective. Clin Epigenetics 2021; 13:87. [PMID: 33883002 PMCID: PMC8061201 DOI: 10.1186/s13148-021-01079-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/13/2021] [Indexed: 01/19/2023] Open
Abstract
Diabetic kidney disease (DKD) is one of the most common microvascular complication of both type 1 (T1DM) and type 2 diabetes mellitus (T2DM), and the leading cause of end-stage renal disease (ESRD) worldwide. Persistent inflammation and subsequent chronic fibrosis are major causes of loss of renal function, which is associated with the progression of DKD to ESRD. In fact, DKD progression is affected by a combination of genetic and environmental factors. Approximately, one-third of diabetic patients progress to develop DKD despite intensive glycemic control, which propose an essential concept "metabolic memory." Epigenetic modifications, an extensively studied mechanism of metabolic memory, have been shown to contribute to the susceptibility to develop DKD. Epigenetic modifications also play a regulatory role in the interactions between the genes and the environmental factors. The epigenetic contributions to the processes of inflammation and fibrogenesis involved in DKD occur at different regulatory levels, including DNA methylation, histone modification and non-coding RNA modulation. Compared with genetic factors, epigenetics represents a new therapeutic frontier in understanding the development DKD and may lead to therapeutic breakthroughs due to the possibility to reverse these modifications therapeutically. Early recognition of epigenetic events and biomarkers is crucial for timely diagnosis and intervention of DKD, and for the prevention of the progression of DKD to ESRD. Herein, we will review the latest epigenetic mechanisms involved in the renal pathology of both type 1 (T1DN) and type 2 diabetic nephropathy (T2DN) and highlight the emerging role and possible therapeutic strategies based on the understanding of the role of epigenetics in DKD-associated inflammation and fibrogenesis.
Collapse
Affiliation(s)
- Wen Zheng
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, People's Republic of China
- Core Unit of National Clinical Medical Research Center of Kidney Disease, No. 1, Jianshe East Road, Zhengzhou, 450052, Henan Province, People's Republic of China
| | - Jia Guo
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, People's Republic of China
- Core Unit of National Clinical Medical Research Center of Kidney Disease, No. 1, Jianshe East Road, Zhengzhou, 450052, Henan Province, People's Republic of China
| | - Zhang-Suo Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China.
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, People's Republic of China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, People's Republic of China.
- Core Unit of National Clinical Medical Research Center of Kidney Disease, No. 1, Jianshe East Road, Zhengzhou, 450052, Henan Province, People's Republic of China.
| |
Collapse
|
34
|
Lin Q, Banu K, Ni Z, Leventhal JS, Menon MC. Podocyte Autophagy in Homeostasis and Disease. J Clin Med 2021; 10:jcm10061184. [PMID: 33809036 PMCID: PMC7998595 DOI: 10.3390/jcm10061184] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 02/16/2021] [Accepted: 02/18/2021] [Indexed: 12/19/2022] Open
Abstract
Autophagy is a protective mechanism that removes dysfunctional components and provides nutrition for cells. Podocytes are terminally differentiated specialized epithelial cells that wrap around the capillaries of the glomerular filtration barrier and show high autophagy level at the baseline. Here, we provide an overview of cellular autophagy and its regulation in homeostasis with specific reference to podocytes. We discuss recent data that have focused on the functional role and regulation of autophagy during podocyte injury in experimental and clinical glomerular diseases. A thorough understanding of podocyte autophagy could shed novel insights into podocyte survival mechanisms with injury and offer potential targets for novel therapeutics for glomerular disease.
Collapse
Affiliation(s)
- Qisheng Lin
- Division of Nephrology, Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (Q.L.); (K.B.); (J.S.L.)
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China;
| | - Khadija Banu
- Division of Nephrology, Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (Q.L.); (K.B.); (J.S.L.)
- Division of Nephrology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Zhaohui Ni
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China;
| | - Jeremy S. Leventhal
- Division of Nephrology, Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (Q.L.); (K.B.); (J.S.L.)
| | - Madhav C. Menon
- Division of Nephrology, Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (Q.L.); (K.B.); (J.S.L.)
- Division of Nephrology, Yale School of Medicine, New Haven, CT 06510, USA
- Correspondence:
| |
Collapse
|
35
|
Valproic acid influences the expression of genes implicated with hyperglycaemia-induced complement and coagulation pathways. Sci Rep 2021; 11:2163. [PMID: 33495488 PMCID: PMC7835211 DOI: 10.1038/s41598-021-81794-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 01/11/2021] [Indexed: 01/04/2023] Open
Abstract
Because the liver plays a major role in metabolic homeostasis and secretion of clotting factors and inflammatory innate immune proteins, there is interest in understanding the mechanisms of hepatic cell activation under hyperglycaemia and whether this can be attenuated pharmacologically. We have previously shown that hyperglycaemia stimulates major changes in chromatin organization and metabolism in hepatocytes, and that the histone deacetylase inhibitor valproic acid (VPA) is able to reverse some of these metabolic changes. In this study, we have used RNA-sequencing (RNA-seq) to investigate how VPA influences gene expression in hepatocytes. Interesting, we observed that VPA attenuates hyperglycaemia-induced activation of complement and coagulation cascade genes. We also observe that many of the gene activation events coincide with changes to histone acetylation at the promoter of these genes indicating that epigenetic regulation is involved in VPA action.
Collapse
|
36
|
Current Therapies in Nephrotic Syndrome: HDAC inhibitors, an Emerging Therapy for Kidney Diseases. CURRENT RESEARCH IN BIOTECHNOLOGY 2021. [DOI: 10.1016/j.crbiot.2021.05.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
|
37
|
Liu M, Qiao Z, Zhang Y, Zhan P, Yi F. Histone Deacetylases Take Center Stage on Regulation of Podocyte Function. KIDNEY DISEASES (BASEL, SWITZERLAND) 2020; 6:236-246. [PMID: 32903938 PMCID: PMC7445693 DOI: 10.1159/000507117] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 03/10/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Podocytes (highly specialized and terminally differentiated epithelial cells) are integral components of the glomerular filtration barrier that are vulnerable to a variety of injuries and, as a result, they undergo a series of changes ranging from hypertrophy to detachment and apoptosis. Podocyte injury is a major determinant in proteinuric kidney disease and identification of potential therapeutic targets for preventing podocyte injury has clinical importance. Although numerous studies have achieved dramatic advances in the understanding of podocyte biology and its relevance to renal injury, few effective and specific therapies are available. SUMMARY Epigenetic modifications have been proven to play important roles in the pathogenesis of kidney diseases. Among them, histone deacetylase (HDAC)-mediated epigenetic acetylation in the kidney has attracted much attention, which may play multiple roles in both kidney development and the pathogenesis of kidney disease. Recent studies have demonstrated that HDAC protect against podocyte injury by regulation of inflammation, apoptosis, autophagy, mitochondrial function, and insulin resistance. In this review, we summarize recent advances in the understanding of the functions and regulatory mechanisms of HDAC in podocytes and associated proteinuric kidney diseases. In addition, we provide evidence of the potential therapeutic effects of HDAC inhibitors for proteinuric kidney disease. KEY MESSAGES Pharmacological targeting of HDAC-mediated epigenetic processes may open new therapeutic avenues for chronic kidney disease.
Collapse
Affiliation(s)
| | | | | | | | - Fan Yi
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| |
Collapse
|
38
|
Pereira BP, do Valle GT, Salles BCC, Costa KCM, Ângelo ML, Torres LHL, Novaes RD, Ruginsk SG, Tirapelli CR, de Araújo Paula FB, Ceron CS. Pyrrolidine dithiocarbamate reduces alloxan-induced kidney damage by decreasing nox4, inducible nitric oxide synthase, and metalloproteinase-2. Naunyn Schmiedebergs Arch Pharmacol 2020; 393:1899-1910. [PMID: 32440769 DOI: 10.1007/s00210-020-01906-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 05/10/2020] [Indexed: 12/18/2022]
Abstract
We examined the effect of the NFκB inhibitor pyrrolidine-1-carbodithioic acid (PDTC) on inducible nitric oxide synthase (iNOS), matrix metalloproteinase-2 (MMP-2) activity, and oxidative and inflammatory kidney damage in alloxan-induced diabetes. Two weeks after diabetes induction (alloxan-130 mg/kg), control and diabetic rats received PDTC (100 mg/kg) or vehicle for 8 weeks. Body weight, glycemia, urea, and creatinine were measured. Kidney changes were measured in hematoxylin/eosin sections and ED1 by immunohistochemistry. Kidney thiobarbituric acid reactive substances (TBARS), superoxide anion (O2-), and nitrate/nitrite (NOx) levels, and catalase and superoxide dismutase (SOD) activities were analyzed. Also, kidney nox4 and iNOS expression, and NFkB nuclear translocation were measured by western blot, and MMP-2 by zymography. Glycemia and urea increased in alloxan rats, which were not modified by PDTC treatment. However, PDTC attenuated kidney structural alterations and macrophage infiltration in diabetic rats. While diabetes increased both TBARS and O2- levels, PDTC treatment reduced TBARS in diabetic and O2- in control kidneys. A decrease in NOx levels was found in diabetic kidneys, which was prevented by PDTC. Diabetes reduced catalase activity, and PDTC increased catalase and SOD activities in both control and diabetic kidneys. PDTC treatment reduced MMP-2 activity and iNOS and p65 NFκB nuclear expression found increased in diabetic kidneys. Our results show that the NFκB inhibitor PDTC reduces renal damage through reduction of Nox4, iNOS, macrophages, and MMP-2 in the alloxan-induced diabetic model. These findings suggest that PDTC inhibits alloxan kidney damage via antioxidative and anti-inflammatory mechanisms.
Collapse
Affiliation(s)
- Bruna Pinheiro Pereira
- Departamento de Alimentos e Medicamentos, Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, Minas Gerais,, Brazil
| | - Gabriel Tavares do Valle
- Escola de Enfermagem de Ribeirão Preto (EERP), Universidade de São Paulo - USP, Sao Paulo, Brazil
| | - Bruno César Côrrea Salles
- Departamento de Análises Clínicas, Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, Minas Gerais, Brazil
| | - Karla Cristinne Mancini Costa
- Departamento de Alimentos e Medicamentos, Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, Minas Gerais,, Brazil
| | - Marilene Lopes Ângelo
- Departamento de Alimentos e Medicamentos, Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, Minas Gerais,, Brazil
| | - Larissa Helena Lobo Torres
- Departamento de Alimentos e Medicamentos, Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, Minas Gerais,, Brazil
| | - Rômulo Dias Novaes
- Departamento de Biologia Estrutural, Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, Minas Gerais, Brazil
| | - Sílvia Graciela Ruginsk
- Departamento de Ciências Fisiológicas, Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, Minas Gerais, Brazil
| | - Carlos Renato Tirapelli
- Escola de Enfermagem de Ribeirão Preto (EERP), Universidade de São Paulo - USP, Sao Paulo, Brazil
| | | | - Carla Speroni Ceron
- Departamento de Alimentos e Medicamentos, Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, Minas Gerais,, Brazil.
| |
Collapse
|
39
|
Sharma D, Kumar Tekade R, Kalia K. Kaempferol in ameliorating diabetes-induced fibrosis and renal damage: An in vitro and in vivo study in diabetic nephropathy mice model. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 76:153235. [PMID: 32563017 DOI: 10.1016/j.phymed.2020.153235] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 03/03/2020] [Accepted: 04/24/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Kaempferol is a natural polyflavonol that has gained considerable attention as antidiabetic therapeutics. Recent reports emphasize the role of hyperglycemia and RhoA/Rho Kinase activity in the pathogenesis of diabetic nephropathy (DN). This study aims to evaluate the GLP-1 and insulin release along with RhoA/Rho Kinase inhibition pertaining to the anti-fibrotic and reno-protective effects of Kaempferol in DN. METHODS The effect of Kaempferol on GLP-1 and insulin release along with underlying mechanisms (Ca2+ and cAMP levels) in GLUTag and MIN6 cells as well as in their co-culture has been evaluated. Further, the effect of Kaempferol on GLP-1 and insulin release was evaluated under in-vivo circumstances in the DN C57BL/6 mouse model. Histology and fibrosis specific staining was performed to study the renal injuries and fibrosis, while the expression of mRNA and protein of interest was evaluated by RT-PCR and western blot analysis. RESULTS Kaempferol treatment promoted the GLP-1 and insulin release, which was accompanied by increased intracellular levels of cAMP and Ca2+ in GLUTag and MIN6 cells. In agreement with in vitro studies, Kaempferol also increased the release of GLP-1 and insulin in the DN mouse model. Notably, Kaempferol showed the potential to ameliorate the histological changes as well as renal fibrosis while decreasing the expression levels of DN markers including TGF-β1, CTGF, fibronectin, collagen IV, IL-1β, RhoA, ROCK2, and p-MYPT1 in DN kidney tissues. A rise in the expression of E-cadherin and nephrin was also noted in the same study. CONCLUSION This study establishes that Kaempferol ameliorates renal injury and fibrosis by enhancing the release of GLP-1, insulin, and inhibition of RhoA/Rho Kinase. This study recommends Kaempferol for further clinical trials to be developed as novel therapeutics for improving the renal function in DN patients.
Collapse
Affiliation(s)
- Dilip Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, 382355, Gujarat, India
| | - Rakesh Kumar Tekade
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, 382355, Gujarat, India
| | - Kiran Kalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, 382355, Gujarat, India; Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, 382355, Gujarat, India.
| |
Collapse
|
40
|
Exploring the inhibitory activity of valproic acid against the HDAC family using an MMGBSA approach. J Comput Aided Mol Des 2020; 34:857-878. [PMID: 32180123 DOI: 10.1007/s10822-020-00304-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 03/07/2020] [Indexed: 12/30/2022]
Abstract
Valproic acid (VPA) is a compound currently used in clinical practice for the treatment of epilepsy as well as bipolar and mood disorders. VPA targets histone deacetylases (HDACs), which participate in the removal of acetyl groups from lysine in several proteins, regulating a wide variety of functions within the organism. An imbalance or malfunction of these enzymes is associated with the development and progression of several diseases, such as cancer and neurodegenerative diseases. HDACs are divided into four classes, but VPA only targets Class I (HDAC1-3 and 8) and Class IIa (HDAC4-5, 7 and 9) HDACs; however, structural and energetic information regarding the manner by which VPA inhibits these HDACs is lacking. Here, the structural and energetic features that determine this recognition were studied using molecular docking and molecular dynamics (MD) simulation. It was found that VPA reaches the catalytic site in HDAC1-3 and 7, whereas in HDAC6, VPA only reaches the catalytic tunnel. In HDAC4, VPA was bound adjacent to L1 and L2, a zone that participates in corepressor binding, and in HDAC8, VPA was bound to the hydrophobic active site channel (HASC), in line with previous reports.
Collapse
|
41
|
Gao Q, Wang Y, Ma N, Dai H, Roy AC, Chang G, Shi X, Shen X. Sodium valproate attenuates the iE-DAP induced inflammatory response by inhibiting the NOD1-NF-κB pathway and histone modifications in bovine mammary epithelial cells. Int Immunopharmacol 2020; 83:106392. [PMID: 32182568 DOI: 10.1016/j.intimp.2020.106392] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/14/2020] [Accepted: 03/08/2020] [Indexed: 12/30/2022]
Abstract
The anti-inflammatory effects of sodium valproate (VPA) in vivo and in vitro have been demonstrated in recent studies. The aim of this study was to evaluate whether VPA can suppress inflammation in bovine mammary epithelial cells (BMECs) stimulated by γ-D-glutamyl-meso-diaminopimelic acid (iE-DAP). First, the concentration and treatment points of iE-DAP and VPA were optimized. Then, BMECs were cultured in complete media and separated into four groups: untreated control cells (CON group), cells stimulated by 10 μg/mL iE-DAP for 6 h (DAP group), cells stimulated by 0.5 mmol/L VPA for 6 h (VPA group), and cells pretreated with VPA (0.5 mmol/L) for 6 h followed by 10 μg/mL of iE-DAP for 6 h (VD group). The results showed that the level of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) in the culture medium increased in the iE-DAP-treated cells and that pretreatment with VPA reversed this increase. iE-DAP increased both mRNA and protein expression levels of nucleotide-binding oligomerization domain-containing protein 1 (NOD1) and receptor-interacting protein kinas (RIPK2) and activated inhibitor of NF-κB (IκB) and nuclear factor-kappa B p65 (NF-κB p65) through phosphorylation. Upon activation of the NF-κB pathway, the expression of the pro-inflammatory cytokines IL-6, interleukin-8 (IL-8) and interleukin-1β (IL-1β), the acute phase protein serum amyloid A 3 (SAA3) and the lingual antimicrobial peptide (LAP) but not haptoglobi (HP) or bovine neutrophil beta defensing 5 (BNBD5) were increased in the DAP group. The VPA pretreatment induced the acetylation of signal transducers and activators of transcription(STAT1) and histone 3 (H3) by inhibiting histone deacetylase (HDAC) and then suppressed the NF-κB pathway. Moreover, VPA induced autophagy and reduced apoptosis in BMECs in the VD group. These results suggested that VPA treatment can attenuate the inflammatory response induced by iE-DAP.
Collapse
Affiliation(s)
- Qianyun Gao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yan Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Nana Ma
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Hongyu Dai
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Animesh Chandra Roy
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Guangjun Chang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Xiaoli Shi
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Xiangzhen Shen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
42
|
Fontecha-Barriuso M, Martin-Sanchez D, Ruiz-Andres O, Poveda J, Sanchez-Niño MD, Valiño-Rivas L, Ruiz-Ortega M, Ortiz A, Sanz AB. Targeting epigenetic DNA and histone modifications to treat kidney disease. Nephrol Dial Transplant 2019. [PMID: 29534238 DOI: 10.1093/ndt/gfy009] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Epigenetics refers to heritable changes in gene expression patterns not caused by an altered nucleotide sequence, and includes non-coding RNAs and covalent modifications of DNA and histones. This review focuses on functional evidence for the involvement of DNA and histone epigenetic modifications in the pathogenesis of kidney disease and the potential therapeutic implications. There is evidence of activation of epigenetic regulatory mechanisms in acute kidney injury (AKI), chronic kidney disease (CKD) and the AKI-to-CKD transition of diverse aetiologies, including ischaemia-reperfusion injury, nephrotoxicity, ureteral obstruction, diabetes, glomerulonephritis and polycystic kidney disease. A beneficial in vivo effect over preclinical kidney injury has been reported for drugs that decrease DNA methylation by either inhibiting DNA methylation (e.g. 5-azacytidine and decitabine) or activating DNA demethylation (e.g. hydralazine), decrease histone methylation by inhibiting histone methyltransferases, increase histone acetylation by inhibiting histone deacetylases (HDACs, e.g. valproic acid, vorinostat, entinostat), increase histone crotonylation (crotonate) or interfere with histone modification readers [e.g. inhibits of bromodomain and extra-terminal proteins (BET)]. Most preclinical studies addressed CKD or the AKI-to-CKD transition. Crotonate administration protected from nephrotoxic AKI, but evidence is conflicting on DNA methylation inhibitors for preclinical AKI. Several drugs targeting epigenetic regulators are in clinical development or use, most of them for malignancy. The BET inhibitor apabetalone is in Phase 3 trials for atherosclerosis, kidney function being a secondary endpoint, but nephrotoxicity was reported for DNA and HDAC inhibitors. While research into epigenetic modulators may provide novel therapies for kidney disease, caution should be exercised based on the clinical nephrotoxicity of some drugs.
Collapse
Affiliation(s)
- Miguel Fontecha-Barriuso
- Research Institute IIS-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain.,IRSIN, Madrid, Spain.,REDINREN, Madrid, Spain
| | - Diego Martin-Sanchez
- Research Institute IIS-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain.,IRSIN, Madrid, Spain.,REDINREN, Madrid, Spain
| | - Olga Ruiz-Andres
- Research Institute IIS-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain.,IRSIN, Madrid, Spain.,REDINREN, Madrid, Spain
| | - Jonay Poveda
- Research Institute IIS-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain.,IRSIN, Madrid, Spain.,REDINREN, Madrid, Spain
| | - Maria Dolores Sanchez-Niño
- Research Institute IIS-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain.,IRSIN, Madrid, Spain.,REDINREN, Madrid, Spain
| | - Lara Valiño-Rivas
- Research Institute IIS-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain.,IRSIN, Madrid, Spain.,REDINREN, Madrid, Spain
| | - Marta Ruiz-Ortega
- Research Institute IIS-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain.,IRSIN, Madrid, Spain.,REDINREN, Madrid, Spain
| | - Alberto Ortiz
- Research Institute IIS-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain.,IRSIN, Madrid, Spain.,REDINREN, Madrid, Spain
| | - Ana Belén Sanz
- Research Institute IIS-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain.,IRSIN, Madrid, Spain.,REDINREN, Madrid, Spain
| |
Collapse
|
43
|
Hou F, Wei W, Qin X, Liang J, Han S, Han A, Kong Q. The posttranslational modification of HDAC4 in cell biology: Mechanisms and potential targets. J Cell Biochem 2019; 121:930-937. [PMID: 31588631 DOI: 10.1002/jcb.29365] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 08/20/2019] [Indexed: 12/15/2022]
Abstract
Histone deacetylase 4 (HDAC4) is a member of the HDACs family, its expression is closely related to the cell development. The cell is an independent living entity that undergoes proliferation, differentiation, senescence, apoptosis, and pathology, and each process has a strict and complex regulatory system. With deepening of its research, the expression of HDAC4 is critical in the life process. This review focuses on the posttranslational modification of HDAC4 in cell biology, providing an important target for future disease treatment.
Collapse
Affiliation(s)
- Fei Hou
- Lupus Research Institute, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China.,Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong, Jining, China
| | - Wei Wei
- Lupus Research Institute, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China.,Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong, Jining, China
| | - Xiao Qin
- Lupus Research Institute, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China.,Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong, Jining, China
| | - Jing Liang
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong, Jining, China.,College of Life Sciences, Qufu Normal University, Qufu, China
| | - Sha Han
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong, Jining, China
| | - Aizhong Han
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong, Jining, China
| | - Qingsheng Kong
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong, Jining, China
| |
Collapse
|
44
|
Jia Y, Reddy MA, Das S, Oh HJ, Abdollahi M, Yuan H, Zhang E, Lanting L, Wang M, Natarajan R. Dysregulation of histone H3 lysine 27 trimethylation in transforming growth factor-β1-induced gene expression in mesangial cells and diabetic kidney. J Biol Chem 2019; 294:12695-12707. [PMID: 31266808 DOI: 10.1074/jbc.ra119.007575] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 06/13/2019] [Indexed: 12/20/2022] Open
Abstract
Transforming growth factor-β1 (TGF-β)-induced fibrotic and inflammatory genes in renal mesangial cells (MCs) play important roles in glomerular dysfunction associated with diabetic nephropathy (DN). TGF-β regulates gene expression in MCs by altering key chromatin histone modifications at target gene promoters. However, the role of the repressive histone H3 lysine 27 trimethylation (H3K27me3) modification is unclear. Here we show that TGF-β reduces H3K27me3 at the Ctgf, Serpine1, and Ccl2 gene promoters in rat MCs (RMCs) and reciprocally up-regulates the expression of these pro-fibrotic and inflammatory genes. In parallel, TGF-β down-regulates Enhancer of Zeste homolog 2 (Ezh2), an H3K27me3 methyltransferase, and decreases its recruitment at Ctgf and Ccl2 but not Serpine1 promoters. Ezh2 knockdown with siRNAs enhances TGF-β-induced expression of these genes, supporting its repressive function. Mechanistically, Ezh2 down-regulation is mediated by TGF-β-induced microRNA, miR-101b, which targets Ezh2 3'-UTR. TGF-β also up-regulates Jmjd3 and Utx in RMCs, suggesting a key role for these H3K27me3 demethylases in H3K27me3 inhibition. In RMCs, Utx knockdown inhibits hypertrophy, a key event in glomerular dysfunction. The H3K27me3 regulators are similarly altered in human and mouse MCs. High glucose inhibits Ezh2 and increases miR-101b in a TGF-β-dependent manner. Furthermore, in kidneys from rodent models of DN, fibrotic genes, miR-101b, and H3K27me3 demethylases are up-regulated, whereas Ezh2 protein levels as well as enrichment of Ezh2 and H3K27me3 at target genes are decreased, demonstrating in vivo relevance. These results suggest that H3K27me3 inhibition by TGF-β via dysregulation of related histone-modifying enzymes and miRNAs augments pathological genes mediating glomerular mesangial dysfunction and DN.
Collapse
Affiliation(s)
- Ye Jia
- Department of Diabetes Complications and Metabolism, Beckman Research Institute of City of Hope, Duarte, California 91010.,Division of Nephrology, First Hospital of Jilin University, Changchun 130021, China
| | - Marpadga A Reddy
- Department of Diabetes Complications and Metabolism, Beckman Research Institute of City of Hope, Duarte, California 91010
| | - Sadhan Das
- Department of Diabetes Complications and Metabolism, Beckman Research Institute of City of Hope, Duarte, California 91010
| | - Hyung Jung Oh
- Department of Diabetes Complications and Metabolism, Beckman Research Institute of City of Hope, Duarte, California 91010.,Ewha Institute of Convergence Medicine, Ewha Womans University Mokdong Hospital, Seoul 07985, South Korea
| | - Maryam Abdollahi
- Department of Diabetes Complications and Metabolism, Beckman Research Institute of City of Hope, Duarte, California 91010
| | - Hang Yuan
- Department of Diabetes Complications and Metabolism, Beckman Research Institute of City of Hope, Duarte, California 91010.,Division of Nephrology, First Hospital of Jilin University, Changchun 130021, China
| | - Erli Zhang
- Department of Diabetes Complications and Metabolism, Beckman Research Institute of City of Hope, Duarte, California 91010.,Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Linda Lanting
- Department of Diabetes Complications and Metabolism, Beckman Research Institute of City of Hope, Duarte, California 91010
| | - Mei Wang
- Department of Diabetes Complications and Metabolism, Beckman Research Institute of City of Hope, Duarte, California 91010
| | - Rama Natarajan
- Department of Diabetes Complications and Metabolism, Beckman Research Institute of City of Hope, Duarte, California 91010
| |
Collapse
|
45
|
Role of Calbindin-D28k in Diabetes-Associated Advanced Glycation End-Products-Induced Renal Proximal Tubule Cell Injury. Cells 2019; 8:cells8070660. [PMID: 31262060 PMCID: PMC6678974 DOI: 10.3390/cells8070660] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/27/2019] [Accepted: 06/29/2019] [Indexed: 12/16/2022] Open
Abstract
Diabetes-associated advanced glycation end-products (AGEs) can increase extracellular matrix (ECM) expression and induce renal fibrosis. Calbindin-D28k, which plays a role in calcium reabsorption in renal distal convoluted tubules, is increased in a diabetic kidney. The role of calbindin-D28k in diabetic nephropathy still remains unclear. Here, calbindin-D28k protein expression was unexpectedly induced in the renal tubules of db/db diabetic mice. AGEs induced the calbindin-D28k expression in human renal proximal tubule cells (HK2), but not in mesangial cells. AGEs induced the expression of fibrotic molecules, ECM proteins, epithelial-mesenchymal transition (EMT) markers, and endoplasmic reticulum (ER) stress-related molecules in HK2 cells, which could be inhibited by a receptor for AGE (RAGE) neutralizing antibody. Calbindin-D28k knockdown by siRNA transfection reduced the cell viability and obviously enhanced the protein expressions of fibrotic factors, EMT markers, and ER stress-related molecules in AGEs-treated HK2 cells. Chemical chaperone 4-Phenylbutyric acid counteracted the AGEs-induced ER stress and ECM and EMT markers expressions. Calbindin-D28k siRNA in vivo delivery could enhance renal fibrosis in db/db diabetic mice. These findings suggest that inducible calbindin-D28k protects against AGEs/RAGE axis-induced ER stress-activated ECM induction and cell injury in renal proximal tubule cells.
Collapse
|
46
|
Dong L, Li J, Lian Y, Tang ZX, Zen Z, Yu P, Li Y. Long-Term Intensive Lifestyle Intervention Promotes Improvement of Stage III Diabetic Nephropathy. Med Sci Monit 2019; 25:3061-3068. [PMID: 31022160 PMCID: PMC6498885 DOI: 10.12659/msm.913512] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Diabetic nephropathy (DN) is a potentially fatal complication of diabetes mellitus. While lifestyle changes can reduce diabetes risk, it is unclear whether improved lifestyle can slow or reverse DN progression. This study evaluated whether an intensive lifestyle intervention (IL-I) targeting weight loss and inflammation through increased physical activity and reduced caloric intake can delay DN progression. MATERIAL AND METHODS Patients were randomly divided into 2 groups. Both groups received diet and exercise guidelines, but one (IL-I) received more frequent external support than the other (control). We compared markers of metabolic and cardiovascular health, redox status, inflammation, and renal function between groups at 3 and 6 months. Metabolic and cardiovascular metrics included BMI, blood pressure, blood glycosylated hemoglobin (HbA1c), and serum HDL-cholesterol. Redox status was evaluated by serum superoxide dismutase (SOD) and the lipid oxidation product malondialdehyde (MDA), while inflammation was assessed by serum concentrations of IL-6 and TNF-alpha. Renal function was assessed by urine/serum 8-OHdG, albumin: creatinine ratio (ACR), and the renal fibrosis marker TGF-ß1. RESULTS Both groups demonstrated initial BMI reduction, lower HbA1c, and higher HDL-cholesterol, but changes were significantly larger in the IL-I group at 6 months. Blood pressure at 6 months was reduced only in the IL-I group. The IL-I group also achieved a greater sustained SOD increase and MDA reduction. Finally, only the IL-I group demonstrated significant reductions in urine ACR, serum/urine 8-OHdG, and plasma TGF-ß1. These indicators deteriorated after IL-I was stopped. CONCLUSIONS Lifestyle changes including exercise and diet can delay renal damage and promote improvement from DN.
Collapse
Affiliation(s)
- Li Dong
- Department of Nephrology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China (mainland)
| | - Jie Li
- Department of Nephrology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China (mainland)
| | - Yu Lian
- Department of Endocrinology, Southwest Hospital affiliated to The Army Military Medical University, Chongqing, China (mainland)
| | - Zu-Xia Tang
- Department of Nephrology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China (mainland)
| | - Zheng Zen
- Department of Nephrology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China (mainland)
| | - Pan Yu
- Department of Burns and Plastic Surgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China (mainland)
| | - Yang Li
- Department of Nephrology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China (mainland)
| |
Collapse
|
47
|
Exploring the Drug Repurposing Versatility of Valproic Acid as a Multifunctional Regulator of Innate and Adaptive Immune Cells. J Immunol Res 2019; 2019:9678098. [PMID: 31001564 PMCID: PMC6437734 DOI: 10.1155/2019/9678098] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 11/30/2018] [Accepted: 01/02/2019] [Indexed: 12/11/2022] Open
Abstract
Valproic acid (VPA) is widely recognized for its use in the control of epilepsy and other neurological disorders in the past 50 years. Recent evidence has shown the potential of VPA in the control of certain cancers, owed in part to its role in modulating epigenetic changes through the inhibition of histone deacetylases, affecting the expression of genes involved in the cell cycle, differentiation, and apoptosis. The direct impact of VPA in cells of the immune system has only been explored recently. In this review, we discuss the effects of VPA in the suppression of some activation mechanisms in several immune cells that lead to an anti-inflammatory response. As expected, immune cells are not exempt from the effect of VPA, as it also affects the expression of genes of the cell cycle and apoptosis through epigenetic modifications. In addition to inhibiting histone deacetylases, VPA promotes RNA interference, activates histone methyltransferases, or represses the activation of transcription factors. However, during the infectious process, the effectiveness of VPA is subject to the biological nature of the pathogen and the associated immune response; this is because VPA can promote the control or the progression of the infection. Due to its various effects, VPA is a promising alternative for the control of autoimmune diseases and hypersensitivity and needs to be further explored.
Collapse
|
48
|
Histone Deacetylase Inhibitors and Diabetic Kidney Disease. Int J Mol Sci 2018; 19:ijms19092630. [PMID: 30189630 PMCID: PMC6165182 DOI: 10.3390/ijms19092630] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 08/29/2018] [Accepted: 08/31/2018] [Indexed: 12/16/2022] Open
Abstract
Despite recent clinical trial advances and improvements in clinical care, kidney disease due to diabetes remains the most common cause of chronic kidney failure worldwide. In the search for new treatments, recent attentions have turned to drug repurposing opportunities, including study of the histone deacetylase (HDAC) inhibitor class of agents. HDACs are a group of enzymes that remove functional acetyl groups from histone and non-histone proteins and they can affect cellular function through both epigenetic and non-epigenetic means. Over the past decade, several HDAC inhibitors have been adopted into clinical practice, primarily for the treatment of hematological malignancy, whereas other existing therapies (for instance valproate) have been found to have HDAC inhibitory effects. Here we review the current HDAC inhibitors in the clinic and under development; the literature evidence supporting the renoprotective effects of HDAC inhibitors in experimental diabetic kidney disease; and the adverse effect profiles that may prevent existing therapies from entering the clinic for this indication. Whereas recent research efforts have shed light on the fundamental actions of HDACs in the diabetic kidney, whether these efforts will translate into novel therapies for patients will require more specific and better-tolerated therapies.
Collapse
|
49
|
Pawlowska E, Szczepanska J, Wisniewski K, Tokarz P, Jaskólski DJ, Blasiak J. NF-κB-Mediated Inflammation in the Pathogenesis of Intracranial Aneurysm and Subarachnoid Hemorrhage. Does Autophagy Play a Role? Int J Mol Sci 2018; 19:E1245. [PMID: 29671828 PMCID: PMC5979412 DOI: 10.3390/ijms19041245] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 04/13/2018] [Accepted: 04/17/2018] [Indexed: 12/12/2022] Open
Abstract
The rupture of saccular intracranial aneurysms (IA) is the commonest cause of non-traumatic subarachnoid hemorrhage (SAH)—the most serious form of stroke with a high mortality rate. Aneurysm walls are usually characterized by an active inflammatory response, and NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) has been identified as the main transcription factor regulating the induction of inflammation-related genes in IA lesions. This transcription factor has also been related to IA rupture and resulting SAH. We and others have shown that autophagy interacts with inflammation in many diseases, but there is no information of such interplay in IA. Moreover, NF-κB, which is a pivotal factor controlling inflammation, is regulated by autophagy-related proteins, and autophagy is regulated by NF-κB signaling. It was also shown that autophagy mediates the normal functioning of vessels, so its disturbance can be associated with vessel-related disorders. Early brain injury, delayed brain injury, and associated cerebral vasospasm are among the most serious consequences of IA rupture and are associated with impaired function of the autophagy⁻lysosomal system. Further studies on the role of the interplay between autophagy and NF-κB-mediated inflammation in IA can help to better understand IA pathogenesis and to identify IA patients with an increased SAH risk.
Collapse
Affiliation(s)
- Elzbieta Pawlowska
- Department of Orthodontics, Medical University of Lodz, 92-216 Lodz, Poland.
| | - Joanna Szczepanska
- Department of Pediatric Dentistry, Medical University of Lodz, 92-216 Lodz, Poland.
| | - Karol Wisniewski
- Department of Neurosurgery and Neurooncology, Medical University of Lodz, Barlicki University Hospital, Kopcinskiego 22, 90-153 Lodz, Poland.
| | - Paulina Tokarz
- Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland.
| | - Dariusz J Jaskólski
- Department of Neurosurgery and Neurooncology, Medical University of Lodz, Barlicki University Hospital, Kopcinskiego 22, 90-153 Lodz, Poland.
| | - Janusz Blasiak
- Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland.
| |
Collapse
|
50
|
Blockade of histone deacetylase 6 protects against cisplatin-induced acute kidney injury. Clin Sci (Lond) 2018; 132:339-359. [PMID: 29358506 DOI: 10.1042/cs20171417] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 01/04/2018] [Accepted: 01/22/2018] [Indexed: 12/21/2022]
Abstract
Histone deacetylase 6 (HDAC6) has been shown to be involved in various pathological conditions, including cancer, neurodegenerative disorders and inflammatory diseases. Nonetheless, its specific role in drug-induced nephrotoxicity is poorly understood. Cisplatin (dichlorodiamino platinum) belongs to an inorganic platinum - fundamental chemotherapeutic drug utilized in the therapy of various solid malignant tumors. However, the use of cisplatin is extremely limited by obvious side effects, for instance bone marrow suppression and nephrotoxicity. In the present study, we utilized a murine model of cisplatin-induced acute kidney injury (AKI) and a highly selective inhibitor of HDAC6, tubastatin A (TA), to assess the role of HDAC6 in nephrotoxicity and its associated mechanisms. Cisplatin-induced AKI was accompanied by increased expression and activation of HDAC6; blocking HDAC6 with TA lessened renal dysfunction, attenuated renal pathological changes, reduced expression of neutrophil gelatinase-associated lipocalin and kidney injury molecule 1, and decreased tubular cell apoptosis. In cultured human epithelial cells, TA or HDAC6 siRNA treatment also inhibited cisplatin-induced apoptosis. Mechanistic studies demonstrated that cisplatin treatment induced phosphorylation of AKT and loss of E-cadherin in the nephrotoxic kidney, and administration of TA enhanced AKT phosphorylation and preserved E-cadherin expression. HDAC6 inhibition also potentiated autophagy as evidenced by increased expression of autophagy-related gene (Atg) 7 (Atg7), Beclin-1, and decreased renal oxidative stress as demonstrated by up-regulation of superoxide dismutase (SOD) activity and down-regulation of malondialdehyde levels. Moreover, TA was effective in inhibiting nuclear factor-κ B (NF-κB) phosphorylation and suppressing the expression of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). Collectively, these data provide strong evidence that HDAC6 inhibition is protective against cisplatin-induced AKI and suggest that HDAC6 may be a potential therapeutic target for AKI treatment.
Collapse
|