1
|
Fu Q, Gao Q, Jiao S, Da F, Guo J, Liu Y, Liu J. Adipose-derived stem cells ameliorate radiation-induced lung injury by activating the DDAH1/ADMA/eNOS signaling pathway. Regen Ther 2024; 27:398-407. [PMID: 38694446 PMCID: PMC11061648 DOI: 10.1016/j.reth.2024.04.001] [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: 10/24/2023] [Revised: 03/29/2024] [Accepted: 04/11/2024] [Indexed: 05/04/2024] Open
Abstract
Background Ionizing radiation-induced lung injury is caused by the initial inflammatory reaction and leads to advanced fibrosis of lung tissue. Adipose-derived stem cells (ASCs) are a type of mesenchymal stem cell that can differentiate into various functional cell types with broad application prospects in the treatment of tissue damage. The purpose of this study was to explore the protective effect of ASCs against radiation-induced lung injury and to provide a novel basis for prevention and treatment of radiation-induced lung injury. Materials and methods Fifty mice were randomly divided into a control group (Ctrl), radiation exposure group (IR), radiation exposure plus ASC treatment group (IR + ASC), radiation exposure plus L-257 group (IR + L-257), and radiation exposure plus ASC treatment and L-257 group (IR + ASC + L-257). Mice in IR, IR + ASC, and IR + ASC + L-257 groups were exposed to a single whole-body dose of 5 Gy X-rays (160 kV/25 mA, 1.25 Gy/min). Within 2 h after irradiation, mice in IR + ASC and IR + ASC + L-257 groups were injected with 5 × 106 ASCs via the tail vein. Mice in IR + L-257 and IR + ASC + L-257 groups were intraperitoneally injected with 30 mg/kg L-257 in 0.5 mL saline. Results The mice in the IR group exhibited lung hemorrhage, edema, pulmonary fibrosis, and inflammatory cell infiltration, increased release of proinflammatory cytokines, elevation of oxidative stress and apoptosis, and inhibition of the dimethylarginine dimethylamino hydratase 1 (DDAH1)/ADMA/eNOS signaling pathway. ASC treatment alleviated radiation-induced oxidative stress, apoptosis, and inflammation, and restored the DDAH1/ADMA/eNOS signaling pathway. However, L-257 pretreatment offset the protective effect of ASCs against lung inflammation, oxidative stress, and apoptosis. Conclusions These data suggest that ASCs ameliorate radiation-induced lung injury, and the mechanism may be mediated through the DDAH1/ADMA/eNOS signaling pathway.
Collapse
Affiliation(s)
- Quanwei Fu
- Department of Radiation Medical Protection, School of Military Preventive Medicine, Air Force Medical University, Xi’an 710038, China
| | - Qiaohui Gao
- Department of Radiation Medical Protection, School of Military Preventive Medicine, Air Force Medical University, Xi’an 710038, China
| | - Shengyuan Jiao
- Department of Radiation Medical Protection, School of Military Preventive Medicine, Air Force Medical University, Xi’an 710038, China
| | - Fei Da
- Department of Radiation Medical Protection, School of Military Preventive Medicine, Air Force Medical University, Xi’an 710038, China
| | - Juan Guo
- Department of Radiation Medical Protection, School of Military Preventive Medicine, Air Force Medical University, Xi’an 710038, China
| | - Yunen Liu
- Shenyang Medical College, No. 146, Huanghe North Street, Shenyang 110034, China
- Department of Emergency Medicine, General Hospital of Northern Theater Command, No. 83 Road, Shenhe District, Shenyang l10016, China
| | - Junye Liu
- Department of Radiation Medical Protection, School of Military Preventive Medicine, Air Force Medical University, Xi’an 710038, China
| |
Collapse
|
2
|
Chen X, Xiong Y, Zeng S, Delić D, Gaballa M, Kalk P, Klein T, Krämer BK, Hocher B. Comparison of sGC activator and sGC stimulator in 5/6 nephrectomized rats on high-salt-diet. Front Pharmacol 2024; 15:1480186. [PMID: 39494352 PMCID: PMC11527642 DOI: 10.3389/fphar.2024.1480186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/07/2024] [Indexed: 11/05/2024] Open
Abstract
Introduction Soluble guanylate cyclase (sGC) stimulators and activators are known to enhance kidney function in various models of chronic kidney disease (CKD) by increasing cyclic guanosine monophosphate (cGMP). Their differential effects on CKD progression, particularly under conditions of oxidative stress, remain unexplored by direct comparative studies. Methods We conducted a side-by-side comparison using 5/6 nephrectomized rats on a high salt diet (5/6Nx+HSD) to evaluate the efficacy of the sGC stimulator BAY 41-8543 and the sGC activator BAY 60-2770 in CKD progression. BAY 41-8543 (1 mg/kg; twice daily) and BAY 60-2770 (1 mg/kg; once daily) were administered by gavage for 11 weeks. Results The 5/6Nx+HSD model led to increased plasma creatinine, proteinuria, and blood pressure. Both BAY 41-8543 and BAY 60-2770 significantly reduced systolic and diastolic blood pressure to a similar extent but did not improve renal function parameters. Notably, BAY 60-2770 reduced renal fibrosis, including interstitial fibrosis and glomerulosclerosis, whereas BAY 41-8543 did not. These antifibrotic effects of BAY 60-2770 were independent of blood pressure reduction. Proteomic analysis revealed that BAY 60-2770 corrected the upregulation of 9 proteins associated with apoptosis and fibrosis, including Caspase-3, MKK6 (Mitogen-Activated Protein Kinase Kinase 6), Prdx5 (Peroxiredoxin-5), in the 5/6Nx+HSD group. Discussion In contrast, BAY 41-8543 had no significant impact on these proteins. sGC activators were more effective than sGC stimulators in reducing renal fibrosis in 5/6 nephrectomized rats on a high salt diet, and this effect was due to modulation of apoptosis-associated proteins beyond the control of blood pressure.
Collapse
Affiliation(s)
- Xin Chen
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
- Department of Nephrology, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
| | - Yingquan Xiong
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
- Department of Nephrology, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
| | - Shufei Zeng
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Denis Delić
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Mohamed Gaballa
- Department of Pathology, Faculty of Veterinary Medicine, Benha University, Toukh, Egypt
- Academy of Scientific Research and Technology, Cairo, Egypt
| | - Philipp Kalk
- Department of Nephrology, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
| | - Thomas Klein
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Bernhard K. Krämer
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
- European Center for Angioscience, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Berthold Hocher
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, China
- Department of Endocrinology, IMD Institut für Medizinische Diagnostik Berlin-Potsdam GbR, Berlin, Germany
- Key Laboratory of Reproductive and Stem Cell Engineering, Central South University, Changsha, Hunan, China
| |
Collapse
|
3
|
Gonzalez-Candia A, Figueroa EG, Krause BJ. Pharmacological and molecular mechanisms of miRNA-based therapies for targeting cardiovascular dysfunction. Biochem Pharmacol 2024; 228:116318. [PMID: 38801924 DOI: 10.1016/j.bcp.2024.116318] [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: 04/09/2024] [Revised: 05/13/2024] [Accepted: 05/24/2024] [Indexed: 05/29/2024]
Abstract
Advances in understanding gene expression regulation through epigenetic mechanisms have contributed to elucidating the regulatory mechanisms of noncoding RNAs as pharmacological targets in several diseases. MicroRNAs (miRs) are a class of evolutionarily conserved, short, noncoding RNAs regulating in a concerted manner gene expression at the post-transcriptional level by targeting specific sequences of the 3'-untranslated region of mRNA. Conversely, mechanisms of cardiovascular disease (CVD) remain largely elusive due to their life-course origins, multifactorial pathophysiology, and co-morbidities. In this regard, CVD treatment with conventional medications results in therapeutic failure due to progressive resistance to monotherapy, which overlooks the multiple factors involved, and reduced adherence to poly-pharmacology approaches. Consequently, considering its role in regulating complete gene pathways, miR-based drugs have appreciably progressed into preclinical and clinical testing. This review summarizes the current knowledge about the mechanisms of miRs in cardiovascular disease, focusing specifically on describing how clinical chemistry and physics have improved the stability of the miR molecule. In addition, a comprehensive review of the main miRs involved in cardiovascular disease and the clinical trials in which these molecules are used as active pharmacological molecules is provided.
Collapse
Affiliation(s)
- Alejandro Gonzalez-Candia
- Laboratory of Fetal Neuroprogramming (www.neurofetal-lab.cl), Institute of Health Sciences, Universidad de O'Higgins, Rancagua, Chile
| | - Esteban G Figueroa
- Laboratory of Fetal Neuroprogramming (www.neurofetal-lab.cl), Institute of Health Sciences, Universidad de O'Higgins, Rancagua, Chile
| | - Bernardo J Krause
- Institute of Health Sciences, Universidad de O'Higgins, Rancagua, Chile.
| |
Collapse
|
4
|
Zhang Z, Wang Q, Zhang H, Wang S, Ma X, Wang H. Golm1 facilitates the CaO2-DOPC-DSPE200-PEI -CsPbBr3 QDs -induced apoptotic death of hepatocytes through the stimulation of mitochondrial autophagy and mitochondrial reactive oxygen species production through interactions with P53/Beclin-1/Bcl-2. Chem Biol Interact 2024; 398:111076. [PMID: 38815669 DOI: 10.1016/j.cbi.2024.111076] [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: 01/15/2024] [Revised: 05/14/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
Mitophagy is a distinct physiological process that can have beneficial or deleterious effects in particular tissues. Prior research suggests that mitophagic activity can be triggered by CaO2-PM-CsPbBr3 QDs, yet the specific role that mitophagy plays in hepatic injury induced by CaO2-PM-CsPbBr3 QDs has yet to be established. Accordingly, in this study a series of mouse model- and cell-based experiments were performed that revealed the ability of CaO2-PM-CsPbBr3 QDs to activate mitophagic activity. Golm1 was upregulated in response to CaO2-PM-CsPbBr3 QDs treatment, and overexpressing Golm1 induced autophagic flux in the murine liver and hepatocytes, whereas knocking down Golm1 had the opposite effect. CaO2-PM-CsPbBr3 QDs were also able to Golm1 expression, in turn promoting the degradation of P53 and decreasing the half-life of this protein. Overexpressing Golm1 was sufficient to suppress the apoptotic death of hepatocytes in vitro and in vivo, whereas the knockdown of Golm1 had the opposite effect. The ability of Golm1 to promote p53-mediated autophagy was found to be associated with the disruption of Beclin-1 binding to Bcl-2, and the Golm1 N-terminal domain was determined to be required for p53 interactions, inducing autophagic activity in a manner independent of helicase activity or RNA binding. Together, these results indicate that inhibiting Golm1 can promote p53-dependent autophagy via disrupting Beclin-1 binding to Bcl-2, highlighting a novel approach to mitigating liver injury induced by CaO2-PM-CsPbBr3 QDs.
Collapse
Affiliation(s)
- Zhiqiang Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450045, Henan Province, China.
| | - Qinglong Wang
- College of Animal Medicine, Henan University of Animal Husbandry and Economy, Zhengzhou, 450046, Henan Province, China
| | - Haibo Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450045, Henan Province, China
| | - Shengchao Wang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450045, Henan Province, China
| | - Xia Ma
- College of Animal Medicine, Henan University of Animal Husbandry and Economy, Zhengzhou, 450046, Henan Province, China
| | - Hui Wang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450045, Henan Province, China.
| |
Collapse
|
5
|
Yuan J, Yu Z, Zhang P, Luo K, Xu Y, Lan T, Zhang M, Chen Y, Lu Z. DDAH1 recruits peroxiredoxin 1 and sulfiredoxin 1 to preserve its activity and regulate intracellular redox homeostasis. Redox Biol 2024; 70:103080. [PMID: 38354630 PMCID: PMC10876909 DOI: 10.1016/j.redox.2024.103080] [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: 01/18/2024] [Revised: 02/04/2024] [Accepted: 02/07/2024] [Indexed: 02/16/2024] Open
Abstract
Growing evidence suggests that dimethylarginine dimethylaminohydrolase 1 (DDAH1), a crucial enzyme for the degradation of asymmetric dimethylarginine (ADMA), is closely related to oxidative stress during the development of multiple diseases. However, the underlying mechanism by which DDAH1 regulates the intracellular redox state remains unclear. In the present study, DDAH1 was shown to interact with peroxiredoxin 1 (PRDX1) and sulfiredoxin 1 (SRXN1), and these interactions could be enhanced by oxidative stress. In HepG2 cells, H2O2-induced downregulation of DDAH1 and accumulation of ADMA were attenuated by overexpression of PRDX1 or SRXN1 but exacerbated by knockdown of PRDX1 or SRXN1. On the other hand, DDAH1 also maintained the expression of PRDX1 and SRXN1 in H2O2-treated cells. Furthermore, global knockout of Ddah1 (Ddah1-/-) or liver-specific knockout of Ddah1 (Ddah1HKO) exacerbated, while overexpression of DDAH1 alleviated liver dysfunction, hepatic oxidative stress and downregulation of PRDX1 and SRXN1 in CCl4-treated mice. Overexpression of liver PRDX1 improved liver function, attenuated hepatic oxidative stress and DDAH1 downregulation, and diminished the differences between wild type and Ddah1-/- mice after CCl4 treatment. Collectively, our results suggest that the regulatory effect of DDAH1 on cellular redox homeostasis under stress conditions is due, at least in part, to the interaction with PRDX1 and SRXN1.
Collapse
Affiliation(s)
- Juntao Yuan
- College of Life Science, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhuoran Yu
- College of Life Science, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ping Zhang
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, 55455, USA
| | - Kai Luo
- College of Life Science, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ying Xu
- College of Life Science, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ting Lan
- College of Life Science, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Min Zhang
- Department of Nephrology, Affiliated Beijing Chaoyang Hospital of Capital Medical University, Beijing, 100020, China.
| | - Yingjie Chen
- Department of Physiology & Biophysics, University of Mississippi Medical Center, Jackson, MS, 39216, USA.
| | - Zhongbing Lu
- College of Life Science, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
6
|
Feng F, Cui B, Fang L, Lan T, Luo K, Xu X, Lu Z. DDAH1 Protects against Cardiotoxin-Induced Muscle Injury and Regeneration. Antioxidants (Basel) 2023; 12:1754. [PMID: 37760057 PMCID: PMC10525962 DOI: 10.3390/antiox12091754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Nitric oxide (NO) is an important biological signaling molecule affecting muscle regeneration. The activity of NO synthase (NOS) is regulated by dimethylarginine dimethylaminohydrolase 1 (DDAH1) through degradation of the endogenous NOS inhibitor asymmetric dimethylarginine (ADMA). To investigate the role of DDAH1 in muscle injury and regeneration, muscle-specific Ddah1-knockout mice (Ddah1MKO) and their littermates (Ddah1f/f) were used to examine the progress of cardiotoxin (CTX)-induced muscle injury and subsequent muscle regeneration. After CTX injection, Ddah1MKO mice developed more severe muscle injury than Ddah1f/f mice. Muscle regeneration was also delayed in Ddah1MKO mice on Day 5 after CTX injection. These phenomena were associated with higher serum ADMA and LDH levels as well as a great induction of inflammatory response, oxidative stress and cell apoptosis in the gastrocnemius (GA) muscle of Ddah1MKO mice. In the GA muscle of CTX-treated mice, Ddah1 deficiency decreased the protein expression of M-cadherin, myogenin, Bcl-2, peroxiredoxin 3 (PRDX3) and PRDX5, and increased the protein expression of MyoD, TNFα, Il-6, iNOS and Bax. In summary, our data suggest that DDAH1 exerts a protective role in muscle injury and regeneration.
Collapse
Affiliation(s)
- Fei Feng
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China;
| | - Bingqing Cui
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China; (B.C.); (K.L.)
| | - Li Fang
- Department of Endocrinology, Dongtai Renmin Hospital, Dongtai 224233, China;
| | - Ting Lan
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China; (B.C.); (K.L.)
| | - Kai Luo
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China; (B.C.); (K.L.)
| | - Xin Xu
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China;
| | - Zhongbing Lu
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China; (B.C.); (K.L.)
| |
Collapse
|
7
|
Zhang X, Ma L, Wang J. Cross-Regulation Between Redox and Epigenetic Systems in Tumorigenesis: Molecular Mechanisms and Clinical Applications. Antioxid Redox Signal 2023; 39:445-471. [PMID: 37265163 DOI: 10.1089/ars.2023.0253] [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: 06/03/2023]
Abstract
Significance: Redox and epigenetics are two important regulatory processes of cell physiological functions. The cross-regulation between these processes has critical effects on the occurrence and development of various types of tumors. Recent Advances: The core factor that influences redox balance is reactive oxygen species (ROS) generation. The ROS functions as a double-edged sword in tumors: Low levels of ROS promote tumors, whereas excessive ROS induces various forms of tumor cell death, including apoptosis and ferroptosis as well as necroptosis and pyroptosis. Many studies have shown that the redox balance is influenced by epigenetic mechanisms such as DNA methylation, histone modification, chromatin remodeling, non-coding RNAs (microRNA, long non-coding RNA, and circular RNA), and RNA N6-methyladenosine modification. Several oxidizing or reducing substances also affect the epigenetic state. Critical Issues: In this review, we summarize research on the cross-regulation between redox and epigenetics in cancer and discuss the relevant molecular mechanisms. We also discuss the current research on the clinical applications. Future Directions: Future research can use high-throughput methods to analyze the molecular mechanisms of the cross-regulation between redox and epigenetics using both in vitro and in vivo models in more detail, elucidate regulatory mechanisms, and provide guidance for clinical treatment. Antioxid. Redox Signal. 39, 445-471.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Lifang Ma
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Jiayi Wang
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| |
Collapse
|
8
|
Asha Parveen SM, Kami Reddy KR, Ummanni R. Dimethylarginine Dimethylaminohydrolase - 1 expression is increased under tBHP-induced oxidative stress regulates nitric oxide production in PCa cells attenuates mitochondrial ROS-mediated apoptosis. Nitric Oxide 2023; 138-139:70-84. [PMID: 37423418 DOI: 10.1016/j.niox.2023.07.002] [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: 04/21/2023] [Revised: 07/03/2023] [Accepted: 07/07/2023] [Indexed: 07/11/2023]
Abstract
Dimethylarginine dimethylaminohydrolase-1 (DDAH1) expression is frequently elevated in different cancers including prostate cancer (PCa) and enhances nitric oxide (NO) production in tumor cells by metabolising endogenous nitric oxide synthase (NOS) inhibitors. DDAH1 protects the PCa cells from cell death and promotes survival. In this study, we have investigated the cytoprotective role of DDAH1 and determined the mechanism of DDAH1 in protecting the cells in tumor microenvironment. Proteomic analysis of PCa cells with stable overexpression of DDAH1 has identified that oxidative stress-related activity is altered. Oxidative stress promotes cancer cell proliferation, survival and causes chemoresistance. A known inducer of oxidative stress, tert-Butyl Hydroperoxide (tBHP) treatment to PCa cells led to elevated DDAH1 level that is actively involved in protecting the PCa cells from oxidative stress induced cell damage. In PC3-DDAH1- cells, tBHP treatment led to higher mROS levels indicating that the loss of DDAH1 increases the oxidative stress and eventually leads to cell death. Under oxidative stress, nuclear Nrf2 controlled by SIRT1 positively regulates DDAH1 expression in PC3 cells. In PC3-DDAH1+ cells, tBHP induced DNA damage is well tolerated compared to wild-type cells while PC3-DDAH1- became sensitive to tBHP. In PC3 cells, tBHPexposure has increased the production of NO and GSH which may be acting as an antioxidant defence to overcome oxidative stress. Furthermore, in tBHP treated PCa cells, DDAH1 is controlling the expression of Bcl2, active PARP and caspase 3. Taken together, these results confirm that DDAH1 is involved in the antioxidant defence system and promotes cell survival.
Collapse
Affiliation(s)
- Sakkarai Mohamed Asha Parveen
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Karthik Reddy Kami Reddy
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India
| | - Ramesh Ummanni
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
9
|
Shen X, Luo K, Yuan J, Gao J, Cui B, Yu Z, Lu Z. Hepatic DDAH1 mitigates hepatic steatosis and insulin resistance in obese mice: Involvement of reduced S100A11 expression. Acta Pharm Sin B 2023; 13:3352-3364. [PMID: 37655336 PMCID: PMC10465955 DOI: 10.1016/j.apsb.2023.05.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 03/16/2023] [Accepted: 04/03/2023] [Indexed: 09/02/2023] Open
Abstract
Dimethylarginine dimethylaminohydrolase 1 (DDAH1) is an important regulator of plasma asymmetric dimethylarginine (ADMA) levels, which are associated with insulin resistance in patients with nonalcoholic fatty liver disease (NAFLD). To elucidate the role of hepatic DDAH1 in the pathogenesis of NAFLD, we used hepatocyte-specific Ddah1-knockout mice (Ddah1HKO) to examine the progress of high-fat diet (HFD)-induced NAFLD. Compared to diet-matched flox/flox littermates (Ddah1f/f), Ddah1HKO mice exhibited higher serum ADMA levels. After HFD feeding for 16 weeks, Ddah1HKO mice developed more severe liver steatosis and worse insulin resistance than Ddah1f/f mice. On the contrary, overexpression of DDAH1 attenuated the NAFLD-like phenotype in HFD-fed mice and ob/ob mice. RNA-seq analysis showed that DDAH1 affects NF-κB signaling, lipid metabolic processes, and immune system processes in fatty livers. Furthermore, DDAH1 reduces S100 calcium-binding protein A11 (S100A11) possibly via NF-κB, JNK and oxidative stress-dependent manner in fatty livers. Knockdown of hepatic S100a11 by an AAV8-shS100a11 vector alleviated hepatic steatosis and insulin resistance in HFD-fed Ddah1HKO mice. In summary, our results suggested that the liver DDAH1/S100A11 axis has a marked effect on liver lipid metabolism in obese mice. Strategies to increase liver DDAH1 activity or decrease S100A11 expression could be a valuable approach for NAFLD therapy.
Collapse
Affiliation(s)
- Xiyue Shen
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
- Institute of Respiratory Medicine, Tongji University School of Medicine, Shanghai 200433, China
| | - Kai Luo
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Juntao Yuan
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Junling Gao
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bingqing Cui
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhuoran Yu
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhongbing Lu
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
10
|
Li T, Zhang T, Wang H, Zhang Q, Gao H, Liu R, Yin C. The ADMA-DDAH1 axis in ovarian apoptosis of polycystic ovary syndrome. J Steroid Biochem Mol Biol 2023; 225:106180. [PMID: 36243205 DOI: 10.1016/j.jsbmb.2022.106180] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/16/2022] [Accepted: 09/17/2022] [Indexed: 11/07/2022]
Abstract
Dimethylarginine dimethylaminohydrolase 1 (DDAH1) mainly degrades asymmetric dimethylarginine (ADMA), an endogenous nitric oxide synthase inhibitor. Emerging evidence suggested that plasma ADMA is accumulated in patients with polycystic ovary syndrome (PCOS). However, ADMA-DDAH1 involvement in PCOS pathogenesis is unclear. Here, we used dehydroepiandrosterone (DHEA)-induced PCOS rats and the ovarian granulosa cell line KGN to investigate the effect of the ADMA-DDAH1 pathway on ovarian apoptosis. Moreover, we also quantified the ADMA levels and redox status in human serum specimens, Sprague Dawley rats and KGN cells to investigate the effect of ADMA-DDAH1 on redox status and ovarian apoptosis in PCOS. We enrolled 19 women with PCOS and 17 healthy women (controls) in this study. The women with PCOS had increased serum ADMA levels and decreased glutathione peroxidase (GSH-PX) compared with the controls. In Sprague Dawley rats, 21-day DHEA treatment established PCOS and the rat contained higher ADMA levels in serum and lower DDAH1 expression in ovaries. Moreover, the PCOS rat serum and ovaries exhibited increased levels of the oxidative stress marker malondialdehyde (MDA). ADMA treatment of the KGN cells induced reactive oxygen species accumulation and led to apoptosis. Contrastingly, overexpressing DDAH1 in the KGN cells significantly decreased ADMA levels, enhanced cell viability, and inhibited oxidative stress, while the effect was inverse in DDAH1 knockdown cells. Overall, our results demonstrated that PCOS involves elevated ADMA levels and redox imbalance. The ADMA-DDAH1 pathway exerted a marked effect on oxidative stress and ovarian apoptosis in PCOS. Our findings suggested that strategies for increasing DDAH1 activity in ovarian cells may provide a novel approach for ameliorating PCOS.
Collapse
Affiliation(s)
- Tianhe Li
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Tingting Zhang
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Huanhuan Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qiaoli Zhang
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Huimin Gao
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Ruixia Liu
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Chenghong Yin
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China.
| |
Collapse
|
11
|
Gao J, Lei T, Wang H, Luo K, Wang Y, Cui B, Yu Z, Hu X, Zhang F, Chen Y, Ding W, Lu Z. Dimethylarginine dimethylaminohydrolase 1 protects PM 2.5 exposure-induced lung injury in mice by repressing inflammation and oxidative stress. Part Fibre Toxicol 2022; 19:64. [PMID: 36242005 PMCID: PMC9569114 DOI: 10.1186/s12989-022-00505-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 10/06/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Airborne fine particulate matter with aerodynamic diameter ≤ 2.5 μm (PM2.5) pollution is associated with the prevalence of respiratory diseases, including asthma, bronchitis and chronic obstructive pulmonary disease. In patients with those diseases, circulating asymmetric dimethylarginine (ADMA) levels are increased, which contributes to airway nitric oxide deficiency, oxidative stress and inflammation. Overexpression of dimethylarginine dimethylaminohydrolase 1 (DDAH1), an enzyme degrading ADMA, exerts protective effects in animal models. However, the impact of DDAH1/ADMA on PM2.5-induced lung injury has not been investigated. METHODS Ddah1-/- and DDAH1-transgenic mice, as well as their respective wild-type (WT) littermates, were exposed to either filtered air or airborne PM2.5 (mean daily concentration ~ 50 µg/m3) for 6 months through a whole-body exposure system. Mice were also acutely exposed to 10 mg/kg PM2.5 and/or exogenous ADMA (2 mg/kg) via intratracheal instillation every other day for 2 weeks. Inflammatory response, oxidative stress and related gene expressions in the lungs were examined. In addition, RAW264.7 cells were exposed to PM2.5 and/or ADMA and the changes in intracellular oxidative stress and inflammatory response were determined. RESULTS Ddah1-/- mice developed more severe lung injury than WT mice after long-term PM2.5 exposure, which was associated with greater induction of pulmonary oxidative stress and inflammation. In the lungs of PM2.5-exposed mice, Ddah1 deficiency increased protein expression of p-p65, iNOS and Bax, and decreased protein expression of Bcl-2, SOD1 and peroxiredoxin 4. Conversely, DDAH1 overexpression significantly alleviated lung injury, attenuated pulmonary oxidative stress and inflammation, and exerted opposite effects on those proteins in PM2.5-exposed mice. In addition, exogenous ADMA administration could mimic the effect of Ddah1 deficiency on PM2.5-induced lung injury, oxidative stress and inflammation. In PM2.5-exposed macrophages, ADMA aggravated the inflammatory response and oxidative stress in an iNOS-dependent manner. CONCLUSION Our data revealed that DDAH1 has a marked protective effect on long-term PM2.5 exposure-induced lung injury.
Collapse
Affiliation(s)
- Junling Gao
- College of Life Sciences, University of Chinese Academy of Sciences, 19A Yuquanlu, Beijing, 100049, China
| | - Tong Lei
- College of Life Sciences, University of Chinese Academy of Sciences, 19A Yuquanlu, Beijing, 100049, China
| | - Hongyun Wang
- College of Life Sciences, University of Chinese Academy of Sciences, 19A Yuquanlu, Beijing, 100049, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Kai Luo
- College of Life Sciences, University of Chinese Academy of Sciences, 19A Yuquanlu, Beijing, 100049, China
| | - Yuanli Wang
- College of Life Sciences, University of Chinese Academy of Sciences, 19A Yuquanlu, Beijing, 100049, China
| | - Bingqing Cui
- College of Life Sciences, University of Chinese Academy of Sciences, 19A Yuquanlu, Beijing, 100049, China
| | - Zhuoran Yu
- College of Life Sciences, University of Chinese Academy of Sciences, 19A Yuquanlu, Beijing, 100049, China
| | - Xiaoqi Hu
- College of Life Sciences, University of Chinese Academy of Sciences, 19A Yuquanlu, Beijing, 100049, China
| | - Fang Zhang
- College of Life Sciences, University of Chinese Academy of Sciences, 19A Yuquanlu, Beijing, 100049, China
| | - Yingjie Chen
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Wenjun Ding
- College of Life Sciences, University of Chinese Academy of Sciences, 19A Yuquanlu, Beijing, 100049, China.
| | - Zhongbing Lu
- College of Life Sciences, University of Chinese Academy of Sciences, 19A Yuquanlu, Beijing, 100049, China.
| |
Collapse
|
12
|
DDAH1 Protects against Acetaminophen-Induced Liver Hepatoxicity in Mice. Antioxidants (Basel) 2022; 11:antiox11050880. [PMID: 35624743 PMCID: PMC9137993 DOI: 10.3390/antiox11050880] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/23/2022] [Accepted: 04/28/2022] [Indexed: 02/04/2023] Open
Abstract
In many developed countries, acetaminophen (APAP) overdose-induced acute liver injury is a significant therapeutic problem. Dimethylarginine dimethylaminohydrolase 1 (DDAH1) is a critical enzyme for asymmetric dimethylarginine (ADMA) metabolism. Growing evidence suggests that liver dysfunction is associated with increased plasma ADMA levels and reduced hepatic DDAH1 activity/expression. The purpose of this study was to investigate the involvement of DDAH1 in APAP-mediated hepatotoxicity using Ddah1-/- and DDAH1 transgenic mice. After APAP challenge, Ddah1-/- mice developed more severe liver injury than wild type (WT) mice, which was associated with a greater induction of fibrosis, oxidative stress, inflammation, cell apoptosis and phosphorylation of JNK. In contrast, overexpression of DDAH1 attenuated APAP-induced liver injury. RNA-seq analysis showed that DDAH1 affects xenobiotic metabolism and glutathione metabolism pathways in APAP-treated livers. Furthermore, we found that DDAH1 knockdown aggravated APAP-induced cell death, oxidative stress, phosphorylation of JNK and p65, upregulation of CYP2E1 and downregulation of GSTA1 in HepG2 cells. Collectively, our data suggested that DDAH1 has a marked protective effect against APAP-induced liver oxidative stress, inflammation and injury. Strategies to increase hepatic DDAH1 expression/activity may be novel approaches for drug-induced acute liver injury therapy.
Collapse
|
13
|
BACH1, the master regulator of oxidative stress, has a dual effect on CFTR expression. Biochem J 2021; 478:3741-3756. [PMID: 34605540 PMCID: PMC8589331 DOI: 10.1042/bcj20210252] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 02/07/2023]
Abstract
The cystic fibrosis transmembrane conductance regulator (CFTR) gene lies within a topologically associated domain (TAD) in which multiple cis-regulatory elements (CREs) and transcription factors (TFs) regulate its cell-specific expression. The CREs are recruited to the gene promoter by a looping mechanism that depends upon both architectural proteins and specific TFs. An siRNA screen to identify TFs coordinating CFTR expression in airway epithelial cells suggested an activating role for BTB domain and CNC homolog 1 (BACH1). BACH1 is a ubiquitous master regulator of the cellular response to oxidative stress. Here, we show that BACH1 may have a dual effect on CFTR expression by direct occupancy of CREs at physiological oxygen (∼8%), while indirectly modulating expression under conditions of oxidative stress. Hence BACH1, can activate or repress the same gene, to fine tune expression in response to environmental cues such as cell stress. Furthermore, our 4C-seq data suggest that BACH1 can also directly regulate CFTR gene expression by modulating locus architecture through occupancy at known enhancers and structural elements, and depletion of BACH1 alters the higher order chromatin structure.
Collapse
|
14
|
tert-Butyl Hydroperoxide (tBHP)-Induced Lipid Peroxidation and Embryonic Defects Resemble Glucose-6-Phosphate Dehydrogenase (G6PD) Deficiency in C. elegans. Int J Mol Sci 2020; 21:ijms21228688. [PMID: 33217954 PMCID: PMC7698637 DOI: 10.3390/ijms21228688] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/16/2020] [Accepted: 11/16/2020] [Indexed: 11/17/2022] Open
Abstract
G6PD is required for embryonic development in animals, as severe G6PD deficiency is lethal to mice, zebrafish and nematode. Lipid peroxidation is linked to membrane-associated embryonic defects in Caenorhabditis elegans (C. elegans). However, the direct link between lipid peroxidation and embryonic lethality has not been established. The aim of this study was to delineate the role of lipid peroxidation in gspd-1-knockdown (ortholog of g6pd) C. elegans during reproduction. tert-butyl hydroperoxide (tBHP) was used as an exogenous inducer. Short-term tBHP administration reduced brood size and enhanced germ cell death in C. elegans. The altered phenotypes caused by tBHP resembled GSPD-1 deficiency in C. elegans. Mechanistically, tBHP-induced malondialdehyde (MDA) production and stimulated calcium-independent phospholipase A2 (iPLA) activity, leading to disturbed oogenesis and embryogenesis. The current study provides strong evidence to support the notion that enhanced lipid peroxidation in G6PD deficiency promotes death of germ cells and impairs embryogenesis in C. elegans.
Collapse
|
15
|
Maitiabola G, Tian F, Sun H, Zhang L, Gao X, Xue B, Wang X. Proteome characteristics of liver tissue from patients with parenteral nutrition-associated liver disease. Nutr Metab (Lond) 2020; 17:43. [PMID: 32518576 PMCID: PMC7268697 DOI: 10.1186/s12986-020-00453-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 04/13/2020] [Indexed: 12/15/2022] Open
Abstract
Background Parenteral nutrition (PN)-associated liver disease (PNALD) is a common and life-threatening complication in patients receiving PN. However, its definitive etiology is not yet clear. Therefore, performed proteomic analyses of human liver tissue to explore the same. Methods Liver tissue was derived and compared across selected patients with (n = 3) /without (n = 4) PNALD via isobaric Tag for Relative and Absolute Quantitation (iTRAQ)-based quantitative proteomics. Bioinformatics analysis was performed using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases to explore the mechanisms of PNALD based on differentially expressed proteins (DEPs). The essential proteins that were differentially expressed between the two groups were explored and verified by western blotting. Results A total of 112 proteins were found to be differentially expressed, of which 73 were downregulated, and 39 were upregulated in the PNALD group. Bioinformatics analysis showed DEPs to be associated with mitochondrial oxidative phosphorylation (mainly involved in mitochondrial respiratory chain complex I assembly), hepatic glycolipid metabolism (involved primarily in glycogen formation and gluconeogenesis), and oxidative stress (mainly involved in antioxidant change). Conclusion Overall, our results indicated that mitochondrial energy metabolism impairment, hepatic glycolipid metabolism disorder, and excessive oxidative stress injury might explain the comprehensive mechanism underlying PNALD. Moreover, we have provided multiple potential targets for further exploring the PNALD mechanism.
Collapse
Affiliation(s)
- Gulisudumu Maitiabola
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, East Zhongshan Road 305, Nanjing, 210002 P.R. China
| | - Feng Tian
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, East Zhongshan Road 305, Nanjing, 210002 P.R. China
| | - Haifeng Sun
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, East Zhongshan Road 305, Nanjing, 210002 P.R. China
| | - Li Zhang
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, East Zhongshan Road 305, Nanjing, 210002 P.R. China
| | - Xuejin Gao
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, East Zhongshan Road 305, Nanjing, 210002 P.R. China
| | - Bin Xue
- Core Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166 China
| | - Xinying Wang
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, East Zhongshan Road 305, Nanjing, 210002 P.R. China
| |
Collapse
|
16
|
Han X, Liu H, Zhang Z, Yang W, Wu C, Liu X, Zhang F, Sun B, Zhao Y, Jiang G, Yang YG, Ding W. Epitranscriptomic 5-Methylcytosine Profile in PM 2.5-induced Mouse Pulmonary Fibrosis. GENOMICS, PROTEOMICS & BIOINFORMATICS 2020; 18:41-51. [PMID: 32135311 PMCID: PMC7393542 DOI: 10.1016/j.gpb.2019.11.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 10/26/2019] [Accepted: 11/27/2019] [Indexed: 11/25/2022]
Abstract
Exposure of airborne particulate matter (PM) with an aerodynamic diameter less than 2.5 μm (PM2.5) is epidemiologically associated with lung dysfunction and respiratory symptoms, including pulmonary fibrosis. However, whether epigenetic mechanisms are involved in PM2.5-induced pulmonary fibrosis is currently poorly understood. Herein, using a PM2.5-induced pulmonary fibrosis mouse model, we found that PM2.5 exposure leads to aberrant mRNA 5-methylcytosine (m5C) gain and loss in fibrotic lung tissues. Moreover, we showed the m5C-mediated regulatory map of gene functions in pulmonary fibrosis after PM2.5 exposure. Several genes act as m5C gain-upregulated factors, probably critical for the development of PM2.5-induced fibrosis in mouse lungs. These genes, including Lcn2, Mmp9, Chi3l1, Adipoq, Atp5j2, Atp5l, Atpif1, Ndufb6, Fgr, Slc11a1, and Tyrobp, are highly related to oxidative stress response, inflammatory responses, and immune system processes. Our study illustrates the first epitranscriptomic RNA m5C profile in PM2.5-induced pulmonary fibrosis and will be valuable in identifying biomarkers for PM2.5 exposure-related lung pathogenesis with translational potential.
Collapse
Affiliation(s)
- Xiao Han
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, College of Future Technology, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; Sino-Danish College, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Hanchen Liu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zezhong Zhang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; Sino-Danish College, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Wenlan Yang
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, College of Future Technology, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; Sino-Danish College, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Chunyan Wu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; Sino-Danish College, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Xueying Liu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fang Zhang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Baofa Sun
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, College of Future Technology, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yongliang Zhao
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, College of Future Technology, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Guibin Jiang
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Yun-Gui Yang
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, College of Future Technology, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; Sino-Danish College, University of Chinese Academy of Sciences, Beijing 101408, China; Institute of Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China.
| | - Wenjun Ding
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; Sino-Danish College, University of Chinese Academy of Sciences, Beijing 101408, China.
| |
Collapse
|
17
|
Al-Modawi RN, Brinchmann JE, Karlsen TA. Multi-pathway Protective Effects of MicroRNAs on Human Chondrocytes in an In Vitro Model of Osteoarthritis. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 17:776-790. [PMID: 31446120 PMCID: PMC6716067 DOI: 10.1016/j.omtn.2019.07.011] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 07/12/2019] [Accepted: 07/16/2019] [Indexed: 01/15/2023]
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease. One of the main pathogenic factors of OA is thought to be inflammation. Other factors associated with OA are dysregulation of microRNAs, reduced autophagic activity, oxidative stress, and altered metabolism. microRNAs are small non-coding RNAs that are powerful regulators of gene expression. miR-140-5p is considered a cartilage-specific microRNA, is necessary for in vitro chondrogenesis, has anti-inflammatory properties, and is downregulated in osteoarthritic cartilage. Its passenger strand, miR-140-3p, is the most highly expressed microRNA in healthy cartilage and increases during in vitro chondrogenesis. miR-146a is a well-known anti-inflammatory microRNA. Several studies have illustrated its role in OA and autoimmune diseases. We show that, when human chondrocytes were transfected individually with miR-140-5p, miR-140-3p, or miR-146a prior to stimulation with interleukin-1 beta and tumor factor necrosis-alpha as an inflammatory model of OA, each of these microRNAs exhibited similar protective effects. Mass spectrometry analysis provided an insight to the altered proteome. All three microRNAs downregulated important inflammatory mediators. In addition, they affected different proteins belonging to the same biological processes, suggesting an overall inhibition of inflammation and oxidative stress, enhancement of autophagy, and restoration of other homeostatic cellular mechanisms, including metabolism.
Collapse
Affiliation(s)
- Rua Nader Al-Modawi
- Norwegian Center for Stem Cell Research, Department of Immunology and Transfusion Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.
| | - Jan E Brinchmann
- Norwegian Center for Stem Cell Research, Department of Immunology and Transfusion Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Department of Molecular Medicine, University of Oslo, Oslo, Norway.
| | - Tommy A Karlsen
- Norwegian Center for Stem Cell Research, Department of Immunology and Transfusion Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| |
Collapse
|
18
|
Wang T, Jiang L, Wei X, Liu B, Zhao J, Xie P, Yang B, Wang L. MiR-21-3p aggravates injury in rats with acute hemorrhagic necrotizing pancreatitis by activating TRP signaling pathway. Biomed Pharmacother 2018; 107:1744-1753. [PMID: 30257393 DOI: 10.1016/j.biopha.2018.08.164] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 08/27/2018] [Accepted: 08/31/2018] [Indexed: 02/03/2023] Open
Abstract
To evaluate the expression and effect of miR-21-3p in pancreas and lung injury of acute hemorrhagic necrotizing pancreatitis (AHNP) rats. AHNP rat model was constructed via retrograde injection of 5% sodium taurocholate into biliary pancreatic duct. Then rats were divided into normal, sham, AHNP, mimics negative control (NC), miR-21-3p mimics, inhibitors NC, miR-21-3p inhibitors, miR-21-3p mimics + phosphate buffer saline and miR-21-3p mimics + Gd3+ groups (N = 10 in each group). The expression of miR-21-3p, TRP signaling pathway factor, apoptosis related protein and histology were studied in pancreatic and lung tissues. Apoptosis of pancreatic acinar cells was detected. Oxidative stress indexes were detected in lung tissues. The level of PaO2 and PaCO2 and the expression of amylase, lipase and inflammatory factors were detected in blood. Compared with normal and sham groups, the miR-21-3p expression was increased in pancreatic and lung tissues of AHNP rats. MiR-21-3p expression was successfully regulated. Down-regulated miR-21-3p promoted apoptosis of pancreatic acinar cells and restored its function in AHNP rats. Up-regulated miR-21-3p reduced the lung oxygenation function, promoted pathological damage, and aggravated oxidative stress injury in AHNP rats. Meanwhile, up-regulated miR-21-3p also promoted the expression of serum enzymes and inflammatory factors, and activated TRP signaling pathway in AHNP rats. And miR-21-3p aggravated pancreatitis and lung injury by activating transient receptor potential (TRP) signaling pathway in AHNP rats. miR-21-3p promoted the pancreatic injury, inhibited apoptosis of necrotic acinar cells and aggravated lung oxidative stress injury by activating TRP signaling pathway in AHNP rats.
Collapse
Affiliation(s)
- Tao Wang
- Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University, No. 7, Zhengzhou Kangfufront Street, Zhengzhou City, 450052, Henan Province, China
| | - Lihua Jiang
- Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University, No. 7, Zhengzhou Kangfufront Street, Zhengzhou City, 450052, Henan Province, China.
| | - Xiaoyong Wei
- Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University, No. 7, Zhengzhou Kangfufront Street, Zhengzhou City, 450052, Henan Province, China
| | - Bo Liu
- Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University, No. 7, Zhengzhou Kangfufront Street, Zhengzhou City, 450052, Henan Province, China
| | - Junbo Zhao
- Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University, No. 7, Zhengzhou Kangfufront Street, Zhengzhou City, 450052, Henan Province, China
| | - Peilin Xie
- Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University, No. 7, Zhengzhou Kangfufront Street, Zhengzhou City, 450052, Henan Province, China
| | - Bo Yang
- Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University, No. 7, Zhengzhou Kangfufront Street, Zhengzhou City, 450052, Henan Province, China
| | - Lijuan Wang
- Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University, No. 7, Zhengzhou Kangfufront Street, Zhengzhou City, 450052, Henan Province, China
| |
Collapse
|
19
|
Liu S, Yuan J, Yue W, Bi Y, Shen X, Gao J, Xu X, Lu Z. GCN2 deficiency protects against high fat diet induced hepatic steatosis and insulin resistance in mice. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3257-3267. [PMID: 30006154 DOI: 10.1016/j.bbadis.2018.07.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 06/27/2018] [Accepted: 07/09/2018] [Indexed: 02/05/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterized by hepatic lipid deposition and oxidative stress. It has been demonstrated that general control nonderepressible 2 (GCN2) is required to maintain hepatic fatty acid homeostasis under conditions of amino acid deprivation. However, the impact of GCN2 on the development of NAFLD has not been investigated. In this study, we used Gcn2-/- mice to investigate the effect of GCN2 on high fat diet (HFD)-induced hepatic steatosis. After HFD feeding for 12 weeks, Gcn2-/- mice were less obese than wild-type (WT) mice, and Gcn2-/- significantly attenuated HFD-induced liver dysfunction, hepatic steatosis and insulin resistance. In the livers of the HFD-fed mice, GCN2 deficiency resulted in higher levels of lipolysis genes, lower expression of genes related to FA synthesis, transport and lipogenesis, and less induction of oxidative stress. Furthermore, we found that knockdown of GCN2 attenuated, whereas overexpression of GCN2 exacerbated, palmitic acid-induced steatosis, oxidative & ER stress, and changes of peroxisome proliferator-activated receptor gamma (PPARγ), fatty acid synthase (FAS) and metallothionein (MT) expression in HepG2 cells. Collectively, our data provide evidences that GCN2 deficiency protects against HFD-induced hepatic steatosis by inhibiting lipogenesis and reducing oxidative stress. Our findings suggest that strategies to inhibit GCN2 activity in the liver may provide a novel approach to attenuate NAFLD development.
Collapse
Affiliation(s)
- Shasha Liu
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Juntao Yuan
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenhui Yue
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China; Department of Cardiology, Pan-Vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Yanwei Bi
- Shantou University Medical College, Shantou 515041, China
| | - Xiyue Shen
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Junling Gao
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xin Xu
- Department of Exercise Rehabilitation, Shanghai University of Sport, Shanghai 200438, China.
| | - Zhongbing Lu
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
20
|
Abstract
SIGNIFICANCE RNA is a heterogeneous class of molecules with the minority being protein coding. Noncoding RNAs (ncRNAs) are involved in translation and epigenetic control mechanisms of gene expression. Recent Advances: In recent years, the number of identified ncRNAs has dramatically increased and it is now clear that ncRNAs provide a complex layer of differential gene expression control. CRITICAL ISSUES NcRNAs exhibit interplay with redox regulation. Redox regulation alters the expression of ncRNAs; conversely, ncRNAs alter the expression of generator and effector systems of redox regulation in a complex manner, which will be the focus of this review article. FUTURE DIRECTIONS Understanding the role of ncRNA in redox control will lead to the development of new strategies to alter redox programs. Given that many ncRNAs (particularly microRNAs [miRNAs]) change large gene sets, these molecules are attractive drug candidates; already, now miRNAs can be targeted in patients. Therefore, the development of ncRNA therapies focusing on these molecules is an attractive future strategy. Antioxid. Redox Signal. 29, 793-812.
Collapse
Affiliation(s)
- Matthias S Leisegang
- 1 Institute for Cardiovascular Physiology, Goethe-University , Frankfurt, Germany .,2 German Center of Cardiovascular Research (DZHK) , Partner Site RheinMain, Frankfurt, Germany
| | - Katrin Schröder
- 1 Institute for Cardiovascular Physiology, Goethe-University , Frankfurt, Germany .,2 German Center of Cardiovascular Research (DZHK) , Partner Site RheinMain, Frankfurt, Germany
| | - Ralf P Brandes
- 1 Institute for Cardiovascular Physiology, Goethe-University , Frankfurt, Germany .,2 German Center of Cardiovascular Research (DZHK) , Partner Site RheinMain, Frankfurt, Germany
| |
Collapse
|
21
|
Ducsay CA, Goyal R, Pearce WJ, Wilson S, Hu XQ, Zhang L. Gestational Hypoxia and Developmental Plasticity. Physiol Rev 2018; 98:1241-1334. [PMID: 29717932 PMCID: PMC6088145 DOI: 10.1152/physrev.00043.2017] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Hypoxia is one of the most common and severe challenges to the maintenance of homeostasis. Oxygen sensing is a property of all tissues, and the response to hypoxia is multidimensional involving complicated intracellular networks concerned with the transduction of hypoxia-induced responses. Of all the stresses to which the fetus and newborn infant are subjected, perhaps the most important and clinically relevant is that of hypoxia. Hypoxia during gestation impacts both the mother and fetal development through interactions with an individual's genetic traits acquired over multiple generations by natural selection and changes in gene expression patterns by altering the epigenetic code. Changes in the epigenome determine "genomic plasticity," i.e., the ability of genes to be differentially expressed according to environmental cues. The genomic plasticity defined by epigenomic mechanisms including DNA methylation, histone modifications, and noncoding RNAs during development is the mechanistic substrate for phenotypic programming that determines physiological response and risk for healthy or deleterious outcomes. This review explores the impact of gestational hypoxia on maternal health and fetal development, and epigenetic mechanisms of developmental plasticity with emphasis on the uteroplacental circulation, heart development, cerebral circulation, pulmonary development, and the hypothalamic-pituitary-adrenal axis and adipose tissue. The complex molecular and epigenetic interactions that may impact an individual's physiology and developmental programming of health and disease later in life are discussed.
Collapse
Affiliation(s)
- Charles A. Ducsay
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Ravi Goyal
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - William J. Pearce
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Sean Wilson
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Xiang-Qun Hu
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Lubo Zhang
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| |
Collapse
|
22
|
Cardiomyocyte dimethylarginine dimethylaminohydrolase1 attenuates left-ventricular remodeling after acute myocardial infarction: involvement in oxidative stress and apoptosis. Basic Res Cardiol 2018; 113:28. [DOI: 10.1007/s00395-018-0685-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 05/29/2018] [Indexed: 12/27/2022]
|
23
|
Wang H, Shen X, Tian G, Shi X, Huang W, Wu Y, Sun L, Peng C, Liu S, Huang Y, Chen X, Zhang F, Chen Y, Ding W, Lu Z. AMPKα2 deficiency exacerbates long-term PM 2.5 exposure-induced lung injury and cardiac dysfunction. Free Radic Biol Med 2018; 121:202-214. [PMID: 29753072 DOI: 10.1016/j.freeradbiomed.2018.05.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 05/07/2018] [Accepted: 05/08/2018] [Indexed: 12/22/2022]
Abstract
Previous studies have demonstrated that long-term exposure to fine particulate matter (PM2.5) increases the risk of respiratory and cardiovascular diseases. As a metabolic sensor, AMP-activated protein kinase (AMPK) is a promising target for cardiovascular disease. However, the impact of AMPK on the adverse health effects of PM2.5 has not been investigated. In this study, we exposed wild-type (WT) and AMPKα2-/- mice to either airborne PM2.5 (mean daily concentration ~64 µg/m3) or filtered air for 6 months through a whole-body exposure system. After exposure, AMPKα2-/- mice developed severe lung injury and left ventricular dysfunction. In the PM2.5-exposed lungs and hearts, loss of AMPKα2 resulted in higher levels of fibrotic genes, more collagen deposition, lower levels of peroxiredoxin 5 (Prdx5), and greater induction of oxidative stress and inflammation than observed in the lungs and hearts of WT mice. In PM2.5-exposed BEAS-2B and H9C2 cells, inhibition of AMPK activity significantly decreased cell viability and Prdx5 expression, and increased the intracellular ROS and p-NF-κB levels. Collectively, our results provide the first direct evidence that AMPK has a marked protective effect on the adverse health effects induced by long-term PM2.5 exposure. Our findings suggest that strategies to increase AMPK activity may provide a novel approach to attenuate air pollution associated disease.
Collapse
Affiliation(s)
- Hongyun Wang
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiyue Shen
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guoxiong Tian
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xili Shi
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Huang
- Institute for Environmental Reference Materials of Ministry of Environmental Protection, Beijing 100029, China
| | - Yongguang Wu
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lei Sun
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Can Peng
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Shasha Liu
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ying Huang
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoyu Chen
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fang Zhang
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yingjie Chen
- Cardiovascular Division and Lillehei Heart Institute; University of Minnesota, Minneapolis, MN 55455, USA
| | - Wenjun Ding
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Zhongbing Lu
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
24
|
Kinoshita C, Aoyama K, Nakaki T. Neuroprotection afforded by circadian regulation of intracellular glutathione levels: A key role for miRNAs. Free Radic Biol Med 2018; 119:17-33. [PMID: 29198727 DOI: 10.1016/j.freeradbiomed.2017.11.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 11/21/2017] [Accepted: 11/27/2017] [Indexed: 01/17/2023]
Abstract
Circadian rhythms are approximately 24-h oscillations of physiological and behavioral processes that allow us to adapt to daily environmental cycles. Like many other biological functions, cellular redox status and antioxidative defense systems display circadian rhythmicity. In the central nervous system (CNS), glutathione (GSH) is a critical antioxidant because the CNS is extremely vulnerable to oxidative stress; oxidative stress, in turn, causes several fatal diseases, including neurodegenerative diseases. It has long been known that GSH level shows circadian rhythm, although the mechanism underlying GSH rhythm production has not been well-studied. Several lines of recent evidence indicate that the expression of antioxidant genes involved in GSH homeostasis as well as circadian clock genes are regulated by post-transcriptional regulator microRNA (miRNA), indicating that miRNA plays a key role in generating GSH rhythm. Interestingly, several reports have shown that alterations of miRNA expression as well as circadian rhythm have been known to link with various diseases related to oxidative stress. A growing body of evidence implicates a strong correlation between antioxidative defense, circadian rhythm and miRNA function, therefore, their dysfunctions could cause numerous diseases. It is hoped that continued elucidation of the antioxidative defense systems controlled by novel miRNA regulation under circadian control will advance the development of therapeutics for the diseases caused by oxidative stress.
Collapse
Affiliation(s)
- Chisato Kinoshita
- Department of Pharmacology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| | - Koji Aoyama
- Department of Pharmacology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| | - Toshio Nakaki
- Department of Pharmacology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan.
| |
Collapse
|
25
|
Shi L, Zhao C, Wang H, Lei T, Liu S, Cao J, Lu Z. Dimethylarginine Dimethylaminohydrolase 1 Deficiency Induces the Epithelial to Mesenchymal Transition in Renal Proximal Tubular Epithelial Cells and Exacerbates Kidney Damage in Aged and Diabetic Mice. Antioxid Redox Signal 2017; 27:1347-1360. [PMID: 28594240 DOI: 10.1089/ars.2017.7022] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
AIMS Asymmetric dimethylarginine (ADMA), an endogenous inhibitor of nitric oxide synthase, is mainly degraded by dimethylarginine dimethylaminohydrolase (DDAH). Emerging evidence suggests that plasma ADMA accumulation and DDAH1 activity/expression reduction are linked to chronic kidney disease (CKD) pathology, but the mechanisms remain largely unknown. Here, we examined the role of ADMA/DDAH1 in the epithelial-mesenchymal transition (EMT) of tubular epithelial cells (TECs), an important mechanism for the pathogenesis of renal fibrosis. RESULTS In HK-2 cells, DDAH1 expression was reduced by aldosterone treatment, and overexpression of DDAH1 significantly attenuated aldosterone-induced EMT. More interestingly, DDAH1 deficiency resulted in EMT-related changes in primary TECs via increasing oxidative stress, impairing adenosine monophosphate-activated kinase (AMPK) signaling, and downregulating of peroxiredoxin 5 (Prdx5). However, those effects could not be mimicked by increasing the ADMA concentration. After regular feeding for 24 months or inducing type 2 diabetes, Ddah1-/- mice had higher serum creatinine levels than wild-type (WT) mice. In the kidneys of the aged or diabetic mice, loss of DDAH1 resulted in more interstitial fibrosis, more collagen deposition, and greater induction of EMT-related changes and oxidative stress than in the WT kidneys. Innovation and Conclusion: Our results provide the first direct evidence that the DDAH1 has a marked effect on kidney fibrosis and oxidative stress induced by aging or diabetes. Our findings suggest that strategies to increase DDAH1 activity in TECs may provide a novel approach to attenuate CKD development. Antioxid. Redox Signal. 27, 1347-1360.
Collapse
Affiliation(s)
- Linlin Shi
- College of Life Science, University of Chinese Academy of Sciences , Beijing, China
| | - Chenyang Zhao
- College of Life Science, University of Chinese Academy of Sciences , Beijing, China
| | - Hongyun Wang
- College of Life Science, University of Chinese Academy of Sciences , Beijing, China
| | - Tong Lei
- College of Life Science, University of Chinese Academy of Sciences , Beijing, China
| | - Shasha Liu
- College of Life Science, University of Chinese Academy of Sciences , Beijing, China
| | - Jianwei Cao
- College of Life Science, University of Chinese Academy of Sciences , Beijing, China
| | - Zhongbing Lu
- College of Life Science, University of Chinese Academy of Sciences , Beijing, China
| |
Collapse
|
26
|
Li T, Feng R, Zhao C, Wang Y, Wang J, Liu S, Cao J, Wang H, Wang T, Guo Y, Lu Z. Dimethylarginine Dimethylaminohydrolase 1 Protects Against High-Fat Diet-Induced Hepatic Steatosis and Insulin Resistance in Mice. Antioxid Redox Signal 2017; 26:598-609. [PMID: 27565538 DOI: 10.1089/ars.2016.6742] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
AIMS High plasma concentrations of asymmetric dimethylarginine (ADMA), an endogenous nitric oxide synthase inhibitor, are associated with hepatic dysfunction in patients with nonalcoholic fatty liver disease (NAFLD). However, it is unknown whether ADMA is involved in the pathogenesis of NAFLD. Dimethylarginine dimethylaminohydrolase 1 (DDAH1) is an enzyme that degrades ADMA. In this study, we used Ddah1-/- mice to investigate the effects of the ADMA/DDAH1 pathway on high-fat diet (HFD)-induced hepatic steatosis. RESULTS After HFD feeding for 20 weeks, Ddah1-/- mice were more obese and had developed more severe hepatic steatosis and worse insulin resistance compared with wild-type (WT) mice. In the livers of HFD-fed mice, loss of DDAH1 resulted in higher levels of lipogenic genes, lower expression of β-oxidation genes, and greater induction of oxidative stress, endoplasmic reticulum stress, and inflammation than in the WT livers. Furthermore, ADMA treatment in HepG2 cells led to oxidative stress and steatosis, whereas overexpression of DDAH1 attenuated palmitic acid-induced steatosis, oxidative stress, and inflammation. Innovation and Conclusion: Our results provide the first direct evidence that the ADMA/DDAH1 pathway has a marked effect on hepatic lipogenesis and steatosis induced by HFD feeding. Our findings suggest that strategies to increase DDAH1 activity in hepatocytes may provide a novel approach to attenuate NAFLD development. Antioxid. Redox Signal. 26, 598-609.
Collapse
Affiliation(s)
- Tianhe Li
- College of Life Science, University of Chinese Academy of Sciences , Beijing, China
| | - Run Feng
- College of Life Science, University of Chinese Academy of Sciences , Beijing, China
| | - Chenyang Zhao
- College of Life Science, University of Chinese Academy of Sciences , Beijing, China
| | - Yue Wang
- College of Life Science, University of Chinese Academy of Sciences , Beijing, China
| | - Jian Wang
- College of Life Science, University of Chinese Academy of Sciences , Beijing, China
| | - Shasha Liu
- College of Life Science, University of Chinese Academy of Sciences , Beijing, China
| | - Jianwei Cao
- College of Life Science, University of Chinese Academy of Sciences , Beijing, China
| | - Hongyun Wang
- College of Life Science, University of Chinese Academy of Sciences , Beijing, China
| | - Ting Wang
- College of Life Science, University of Chinese Academy of Sciences , Beijing, China
| | - Yuting Guo
- College of Life Science, University of Chinese Academy of Sciences , Beijing, China
| | - Zhongbing Lu
- College of Life Science, University of Chinese Academy of Sciences , Beijing, China
| |
Collapse
|
27
|
Cui Y, She K, Tian D, Zhang P, Xin X. miR-146a Inhibits Proliferation and Enhances Chemosensitivity in Epithelial Ovarian Cancer via Reduction of SOD2. Oncol Res 2017; 23:275-82. [PMID: 27131313 PMCID: PMC7838621 DOI: 10.3727/096504016x14562725373798] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecological malignancy, accounting for 90% of all ovarian cancer. Dysregulation of miRNAs is associated with several types of EOC. In the current research, we aimed to study the role of abnormal expression of miR-146a in the development of EOC and to elucidate the possible molecular mechanisms. Compared with control samples, mRNA expression of miR-146a was significantly decreased in EOC tissues and cell lines. Overexpression of miR-146a prohibited cell proliferation, enhanced apoptosis, and increased sensitivity to chemotherapy drugs in EOC cells. In contrast, downregulation of miR-146a promoted cell proliferation, suppressed apoptosis, and decreased sensitivity to chemotherapy drugs in EOC cells. Overexpression of miR-146a increased the reactive oxygen species (ROS) level and decreased SOD2 mRNA and protein expression. Downregulation of miR-146a increased SOD2 mRNA and protein expression. Overexpression of SOD2 significantly inhibited miR-146a mimics-induced suppression of cell proliferation and the increase of apoptosis and chemosensitivity. In conclusion, we identify miR-146a as a potential tumor suppressor in patients with EOC. miR-146a downregulates the expression of SOD2 and enhances ROS generation, leading to increased apoptosis, inhibition of proliferation, and enhanced sensitivity to chemotherapy. The data demonstrate that the miR-146a/SOD2/ROS pathway may serve as a novel therapeutic target and prognostic marker in patients with EOC.
Collapse
Affiliation(s)
- YaJie Cui
- Department of Obstetrics and Gynecology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | | | | | | | | |
Collapse
|
28
|
DDAH1 plays dual roles in PM2.5 induced cell death in A549 cells. Biochim Biophys Acta Gen Subj 2016; 1860:2793-801. [DOI: 10.1016/j.bbagen.2016.03.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 03/04/2016] [Accepted: 03/15/2016] [Indexed: 11/16/2022]
|
29
|
Kuang DB, Zhou JP, Yu LY, Zeng WJ, Xiao J, Zhu GZ, Zhang ZL, Chen XP. DDAH1-V3 transcript might act as miR-21 sponge to maintain balance of DDAH1-V1 in cultured HUVECs. Nitric Oxide 2016; 60:59-68. [DOI: 10.1016/j.niox.2016.09.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 09/11/2016] [Accepted: 09/20/2016] [Indexed: 01/01/2023]
|
30
|
TGF-β1/Smads and miR-21 in Renal Fibrosis and Inflammation. Mediators Inflamm 2016; 2016:8319283. [PMID: 27610006 PMCID: PMC5005604 DOI: 10.1155/2016/8319283] [Citation(s) in RCA: 228] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 07/15/2016] [Accepted: 07/26/2016] [Indexed: 12/14/2022] Open
Abstract
Renal fibrosis, irrespective of its etiology, is a final common stage of almost all chronic kidney diseases. Increased apoptosis, epithelial-to-mesenchymal transition, and inflammatory cell infiltration characterize the injured kidney. On the molecular level, transforming growth factor-β1 (TGF-β1)-Smad3 signaling pathway plays a central role in fibrotic kidney disease. Recent findings indicate the prominent role of microRNAs, small noncoding RNA molecules that inhibit gene expression through the posttranscriptional repression of their target mRNAs, in different pathologic conditions, including renal pathophysiology. miR-21 was also shown to play a dynamic role in inflammatory responses and in accelerating injury responses to promote organ failure and fibrosis. Understanding the cellular and molecular bases of miR-21 involvement in the pathogenesis of kidney diseases, including inflammatory reaction, could be crucial for their early diagnosis. Moreover, the possibility of influencing miR-21 level by specific antagomirs may be considered as an approach for treatment of renal diseases.
Collapse
|