1
|
Gwon MG, Leem J, An HJ, Gu H, Bae S, Kim JH, Park KK. The decoy oligodeoxynucleotide against HIF-1α and STAT5 ameliorates atopic dermatitis-like mouse model. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 34:102036. [PMID: 37799329 PMCID: PMC10550406 DOI: 10.1016/j.omtn.2023.102036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 09/15/2023] [Indexed: 10/07/2023]
Abstract
Atopic dermatitis (AD) is a common inflammatory skin disease caused by an immune disorder. Mast cells are known to be activated and granulated to maintain an allergic reaction, including rhinitis, asthma, and AD. Although hypoxia-inducible factor-1 alpha (HIF-1α) and signal transducer and activator of transcription 5 (STAT5) play crucial roles in mast cell survival and granulation, their effects need to be clarified in allergic disorders. Thus, we designed decoy oligodeoxynucleotide (ODN) synthetic DNA, without open ends, containing complementary sequences for HIF-1α and STAT5 to suppress the transcriptional activities of HIF-1α and STAT5. In this study, we demonstrated the effects of HIF-1α/STAT5 ODN using AD-like in vivo and in vitro models. The HIF-1α/STAT5 decoy ODN significantly alleviated cutaneous symptoms similar to AD, including morphology changes, immune cell infiltration, skin barrier dysfunction, and inflammatory response. In the AD model, it also inhibited mast cell infiltration and degranulation in skin tissue. These results suggest that the HIF-1α/STAT5 decoy ODN ameliorates the AD-like disorder and immunoglobulin E (IgE)-induced mast cell activation by disrupting HIF-1α/STAT5 signaling pathways. Taken together, these findings suggest the possibility of HIF-1α/STAT5 as therapeutic targets and their decoy ODN as a potential therapeutic tool for AD.
Collapse
Affiliation(s)
- Mi-Gyeong Gwon
- Department of Pathology, School of Medicine, Daegu Catholic University, Daegu 42472, Republic of Korea
| | - Jaechan Leem
- Department of Immunology, School of Medicine, Daegu Catholic University, Daegu 42472, Republic of Korea
| | - Hyun-Jin An
- Department of Pathology, School of Medicine, Daegu Catholic University, Daegu 42472, Republic of Korea
| | - Hyemin Gu
- Department of Pathology, School of Medicine, Daegu Catholic University, Daegu 42472, Republic of Korea
| | - Seongjae Bae
- Department of Pathology, School of Medicine, Daegu Catholic University, Daegu 42472, Republic of Korea
| | - Jong Hyun Kim
- Department of Biochemistry, School of Medicine, Daegu Catholic University, Daegu 42472, Republic of Korea
| | - Kwan-Kyu Park
- Department of Pathology, School of Medicine, Daegu Catholic University, Daegu 42472, Republic of Korea
| |
Collapse
|
2
|
Song L, Wang X, Qu X, Lv C. Transcription Factor Specificity Protein 1 Regulates Inflammation and Fibrin Deposition in Nasal Polyps Via the Regulation of microRNA-125b and the Wnt/β-catenin Signaling Pathway. Inflammation 2022; 45:1118-1132. [PMID: 34988755 DOI: 10.1007/s10753-021-01605-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 11/19/2021] [Accepted: 11/24/2021] [Indexed: 12/20/2022]
Abstract
Nasal polyps (NPs) are multifactorial soft growths inside the nasal passages and are associated with chronic inflammation that originate from the nasal and paranasal sinus mucosae. This study focused on the role of microRNA (miR)-125b and the molecules associated with NP development. Differentially expressed miRNAs between nasal tissues from patients with chronic rhinosinusitis (CRS) with NP (CRSwNP) and CRS without NP (CRSsNP) were screened using microarray analysis. A murine model of CRSwNP was established. The expression of miR-125b in murine tissues was examined using reverse transcription quantitative polymerase chain reaction. Candidate upstream regulators of miR-125b were predicted using bioinformatics tools, and the binding relationship between specificity protein 1 (Sp1) and miR-125b was validated using luciferase and chromatin immunoprecipitation assays. Altered expression of Sp1 and miR-125b was induced to evaluate their relevance to the progression of NPs. miR-125b expression was significantly upregulated in NP tissues from patients with CRSwNP. Sp1 was confirmed as an upstream regulator that promotes miR-125b transcription in NPs. Overexpression of Sp1 reduced levels of d-dimer (an indicator of fibrinogen degradation products) and tissue-type plasminogen activator (t-PA) but increased eosinophil cationic protein and peroxidase levels, as well as the levels of inflammatory factors interleukin-5 (IL-5) and IL-8 in murine NP tissues. However, these trends were reversed after miR-125b downregulation. Sp1 and miR-125b were found to activate the Wnt/β-catenin signaling pathway in NPs. This study demonstrated that Sp1, an upstream transcription factor of miR-125b, accumulates on the miR-125b promoter to activate its transcription, which induces inflammation and fibrin deposition in NP by activating the Wnt/β-catenin axis.
Collapse
Affiliation(s)
- Li Song
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Affiliated Hospital of Binzhou Medical College, Yantai, 264100, Shandong, People's Republic of China
| | - Xi Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Affiliated Hospital of Binzhou Medical College, Yantai, 264100, Shandong, People's Republic of China
| | - Xiangyang Qu
- Department of Orthopedic Trauma, Yantai Affiliated Hospital of Binzhou Medical College, Yantai, 264100, Shandong, People's Republic of China
| | - Chao Lv
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Mountain Hospital, Laishan District, No. 10087, Keji Avenue, Yantai, 264001, Shandong, People's Republic of China.
| |
Collapse
|
3
|
Kong D, Zhang Z, Chen L, Huang W, Zhang F, Wang L, Wang Y, Cao P, Zheng S. Curcumin blunts epithelial-mesenchymal transition of hepatocytes to alleviate hepatic fibrosis through regulating oxidative stress and autophagy. Redox Biol 2020; 36:101600. [PMID: 32526690 PMCID: PMC7287144 DOI: 10.1016/j.redox.2020.101600] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/08/2020] [Accepted: 05/28/2020] [Indexed: 02/06/2023] Open
Abstract
The massive production and activation of myofibroblasts (MFB) is key to the development of liver fibrosis. In many studies, it has been proven that hepatocytes are an important part of MFB, and can be transformed into MFB through epithelial-mesenchymal transition (EMT) during hepatic fibrogenesis. In our previous study, we confirmed that curcumin inhibited EMT procession and differentiation of hepatocytes into MFB. In addition, in previous studies, it has been shown that autophagy plays an important role in the regulation of cellular EMT procession. In the current study, we showed that curcumin inhibited TGF-β/Smad signaling transmission by activating autophagy, thereby inhibiting EMT. The mechanism of degradative polyubiquitylation of Smad2 and Smad3 is likely through inhibiting tetratricopeptide repeat domain 3 (TTC3) and by inducing ubiquitylation and proteasomal degradation of Smad ubiquitination regulatory factor 2 (SMURF2), which on account of the increase of autophagy in hepatocytes. Curcumin inhibits levels of reactive oxygen species (ROS) and oxidative stress in hepatocytes by activating PPAR-α, and regulates upstream signaling pathways of autophagy AMPK and PI3K/AKT/mTOR, leading to an increase of the autophagic flow in hepatocytes. In this study, we confirm that curcumin effectively reduced the occurrence of EMT in hepatocytes and inhibited production of the extracellular matrix (ECM) by activating autophagy, which provides a potential novel therapeutic strategy for hepatic fibrosis.
Collapse
Affiliation(s)
- Desong Kong
- Chinese Medicine Modernization and Big Data Research Center, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210022, China; Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zili Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Liping Chen
- Chinese Medicine Modernization and Big Data Research Center, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210022, China
| | - Weifang Huang
- Department of Pharmacology, School of Integral Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Feng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ling Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yu Wang
- Department of Oncology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210022, China
| | - Peng Cao
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China.
| | - Shizhong Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
4
|
Beneficial Effects of SREBP Decoy Oligodeoxynucleotide in an Animal Model of Hyperlipidemia. Int J Mol Sci 2020; 21:ijms21020552. [PMID: 31952262 PMCID: PMC7014099 DOI: 10.3390/ijms21020552] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/08/2020] [Accepted: 01/13/2020] [Indexed: 12/15/2022] Open
Abstract
Hyperlipidemia is a chronic disorder that plays an important role in the development of cardiovascular diseases, type II diabetes, atherosclerosis, hypertension, and non-alcoholic fatty liver disease. Hyperlipidemias have created a worldwide health crisis and impose a substantial burden not only on personal health but also on societies and economies. Transcription factors in the sterol regulatory element binding protein (SREBP) family are key regulators of the lipogenic genes in the liver. SREBPs regulate lipid homeostasis by controlling the expression of a range of enzymes required for the synthesis of endogenous cholesterol, fatty acids, triacylglycerol, and phospholipids. Thereby, SREBPs have been considered as targets for the treatment of metabolic diseases. The aim of this study was to investigate the beneficial functions and the possible underlying molecular mechanisms of SREBP decoy ODN, which is a novel inhibitor of SREBPs, in high-fat diet (HFD)-fed hyperlipidemic mice. Our studies using HFD-induced hyperlipidemia animal model revealed that SREBB decoy ODN inhibited the increased expression of fatty acid synthetic pathway, such as SREBP-1c, FAS, SCD-1, ACC1, and HMGCR. In addition, SREBP decoy ODN decreased pro-inflammatory cytokines, including TNF-α, IL-1β, IL-8, and IL-6 expression. These results suggest that SREBP decoy ODN exerts its anti-hyperlipidemia effects in HFD-induced hyperlipidemia mice by regulating their lipid metabolism and inhibiting lipogenesis through inactivation of the SREPB pathway.
Collapse
|
5
|
Kim JY, Park JH, Jeon EJ, Leem J, Park KK. Melatonin Prevents Transforming Growth Factor-β1-Stimulated Transdifferentiation of Renal Interstitial Fibroblasts to Myofibroblasts by Suppressing Reactive Oxygen Species-Dependent Mechanisms. Antioxidants (Basel) 2020; 9:antiox9010039. [PMID: 31906396 PMCID: PMC7022732 DOI: 10.3390/antiox9010039] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 12/20/2019] [Accepted: 12/30/2019] [Indexed: 12/18/2022] Open
Abstract
Accumulating evidence suggests that the pineal hormone melatonin displays protective effects against renal fibrosis, but the mechanisms remain poorly understood. Here, we investigate the effect of the pineal hormone on transdifferentiation of renal fibroblasts to myofibroblasts invoked by transforming growth factor-β1 (TGF-β1). Increased proliferation and activation of renal interstitial fibroblasts after TGF-β1 treatment were attenuated by melatonin pretreatment. Mechanistically, melatonin suppressed Smad2/3 phosphorylation and nuclear co-localization of their phosphorylated forms and Smad4 after TGF-β1 stimulation. In addition, increased phosphorylations of Akt, extracellular signal-regulated kinase 1/2, and p38 after TGF-β1 treatment were also suppressed by the hormone. These effects of melatonin were not affected by pharmacological and genetic inhibition of its membrane receptors. Furthermore, melatonin significantly reversed an increase of intracellular reactive oxygen species (ROS) and malondialdehyde levels, and a decrease of the reduced glutathione/oxidized glutathione ratio after TGF-β1 treatment. Finally, TGF-β1-induced proliferation and activation were also suppressed by N-acetylcysteine. Altogether, these findings suggest that the pineal hormone melatonin prevents TGF-β1-induced transdifferentiation of renal interstitial fibroblasts to myofibroblasts via inhibition of Smad and non-Smad signaling cadcades by inhibiting ROS-mediated mechanisms in its receptor-independent manner.
Collapse
Affiliation(s)
- Jung-Yeon Kim
- Department of Immunology, School of Medicine, Catholic University of Daegu, Daegu 42472, Korea;
| | - Jae-Hyung Park
- Department of Physiology, School of Medicine, Keimyung University, Daegu 42601, Korea;
| | - Eon Ju Jeon
- Department of Internal Medicine, School of Medicine, Catholic University of Daegu, Daegu 42472, Korea
- Correspondence: (E.J.J.); (J.L.)
| | - Jaechan Leem
- Department of Immunology, School of Medicine, Catholic University of Daegu, Daegu 42472, Korea;
- Correspondence: (E.J.J.); (J.L.)
| | - Kwan-Kyu Park
- Department of Pathology, School of Medicine, Catholic University of Daegu, Daegu 42472, Korea;
| |
Collapse
|
6
|
Gwon MG, An HJ, Kim JY, Kim WH, Gu H, Kim HJ, Leem J, Jung HJ, Park KK. Anti-fibrotic effects of synthetic TGF-β1 and Smad oligodeoxynucleotide on kidney fibrosis in vivo and in vitro through inhibition of both epithelial dedifferentiation and endothelial-mesenchymal transitions. FASEB J 2019; 34:333-349. [PMID: 31914629 DOI: 10.1096/fj.201901307rr] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 10/16/2019] [Accepted: 10/21/2019] [Indexed: 01/10/2023]
Abstract
Kidney fibrosis is a common process of various kidney diseases leading to end-stage renal failure irrespective of etiology. Myofibroblasts are crucial mediators in kidney fibrosis through production of extracellular matrix (ECM), but their origin has not been clearly identified. Many study proposed that epithelial and endothelial cells become myofibroblasts by epithelial dedifferentiation and endothelial-mesenchymal transition (EndoMT). TGF-β1/Smad signaling plays a crucial role in partly epithelial-mensencymal transition (EMT) and EndoMT. Thus, we designed the TGF-β1/Smad oligodeoxynucleotide (ODN), a synthetic short DNA containing complementary sequence for Smad transcription factor and TGF-β1 mRNA. Therefore, this study investigated the anti-fibrotic effect of synthetic TGF-β1/Smad ODN on UUO-induced kidney fibrosis in vivo model and TGF-β1-induced in vitro model. To examine the effect of TGF-β1/Smad ODN, we performed various experiments to evaluate kidney fibrosis. The results showed that UUO induced inflammation, ECM accumulation, epithelial dedifferentiation and EndoMT processes, and tubular atrophy. However, synthetic TGF-β1/Smad ODN significantly suppressed UUO-induced fibrosis. Furthermore, synthetic ODN attenuated TGF-β1-induced epithelial dedifferentiation and EndoMT program via blocking TGF-β1/Smad signaling. In conclusion, this study demonstrated that administration of synthetic TGF-β1/Smad ODN attenuates kidney fibrosis, epithelial dedifferentiation, and EndoMT processes. The findings propose the possibility of synthetic ODN as a new effective therapeutic tool for kidney fibrosis.
Collapse
Affiliation(s)
- Mi-Gyeong Gwon
- Department of Pathology, School of Medicine, Catholic University of Daegu, Daegu, Republic of Korea
| | - Hyun-Jin An
- Department of Pathology, School of Medicine, Catholic University of Daegu, Daegu, Republic of Korea
| | - Jung-Yeon Kim
- Department of Immunology, School of Medicine, Catholic University of Daegu, Daegu, Republic of Korea
| | - Woon-Hae Kim
- Department of New Biology, DGIST, Daegu, Republic of Korea
| | - Hyemin Gu
- Department of Pathology, School of Medicine, Catholic University of Daegu, Daegu, Republic of Korea
| | - Hyun-Ju Kim
- Department of Pathology, School of Medicine, Catholic University of Daegu, Daegu, Republic of Korea
| | - Jaechan Leem
- Department of Immunology, School of Medicine, Catholic University of Daegu, Daegu, Republic of Korea
| | - Hyun Jin Jung
- Department of Urology, School of Medicine, Catholic University of Daegu, Daegu, Republic of Korea
| | - Kwan-Kyu Park
- Department of Pathology, School of Medicine, Catholic University of Daegu, Daegu, Republic of Korea
| |
Collapse
|
7
|
Miyake T, Miyake T, Kurashiki T, Morishita R. Molecular Pharmacological Approaches for Treating Abdominal Aortic Aneurysm. Ann Vasc Dis 2019; 12:137-146. [PMID: 31275464 PMCID: PMC6600097 DOI: 10.3400/avd.ra.18-00076] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 02/07/2019] [Indexed: 12/12/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is considered to be a potent life-threatening disorder in elderly individuals. Although many patients with a small AAA are detected during routine abdominal screening, there is no effective therapeutic option to prevent the progression or regression of AAA in the clinical setting. Recent advances in molecular biology have led to the identification of several important molecules, including microRNA and transcription factor, in the process of AAA formation. Regulation of these factors using nucleic acid drugs is expected to be a novel therapeutic option for AAA. Nucleic acid drugs can bind to target factors, mRNA, microRNA, and transcription factors in a sequence-specific fashion, resulting in a loss of function of the target molecule at the transcriptional or posttranscriptional level. Of note, inhibition of a transcription factor using a decoy strategy effectively suppresses experimental AAA formation, by regulating the expression of several genes associated with the disease progression. This review focuses on recent advances in molecular therapy of using nucleic acid drugs to treat AAA.
Collapse
Affiliation(s)
- Takashi Miyake
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Tetsuo Miyake
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Tomohiro Kurashiki
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
8
|
Antifibrotic Effect of Smad Decoy Oligodeoxynucleotide in a CCl₄-Induced Hepatic Fibrosis Animal Model. Molecules 2018; 23:molecules23081991. [PMID: 30103395 PMCID: PMC6222866 DOI: 10.3390/molecules23081991] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/08/2018] [Accepted: 08/08/2018] [Indexed: 01/18/2023] Open
Abstract
Hepatic fibrosis is the wound-healing process of chronic hepatic disease that leads to the end-stage of hepatocellular carcinoma and demolition of hepatic structures. Epithelial–mesenchymal transition (EMT) has been identified to phenotypic conversion of the epithelium to mesenchymal phenotype that occurred during fibrosis. Smad decoy oligodeoxynucleotide (ODN) is a synthetic DNA fragment containing a complementary sequence of Smad transcription factor. Thus, this study evaluated the antifibrotic effects of Smad decoy ODN on carbon tetrachloride (CCl4)-induced hepatic fibrosis in mice. As shown in histological results, CCl4 treatment triggered hepatic fibrosis and increased Smad expression. On the contrary, Smad decoy ODN administration suppressed fibrogenesis and EMT process. The expression of Smad signaling and EMT-associated protein was markedly decreased in Smad decoy ODN-treated mice compared with CCl4-injured mice. In conclusion, these data indicate the practicability of Smad decoy ODN administration for preventing hepatic fibrosis and EMT processes.
Collapse
|
9
|
Lv W, Booz GW, Wang Y, Fan F, Roman RJ. Inflammation and renal fibrosis: Recent developments on key signaling molecules as potential therapeutic targets. Eur J Pharmacol 2017; 820:65-76. [PMID: 29229532 DOI: 10.1016/j.ejphar.2017.12.016] [Citation(s) in RCA: 228] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/07/2017] [Accepted: 12/07/2017] [Indexed: 12/21/2022]
Abstract
Chronic kidney disease (CKD) is a major public health issue. At the histological level, renal fibrosis is the final common pathway of progressive kidney disease irrespective of the initial injury. Considerable evidence now indicates that renal inflammation plays a central role in the initiation and progression of CKD. Some of the inflammatory signaling molecules involved in CKD include: monocyte chemoattractant protein-1 (MCP-1), bradykinin B1 receptor (B1R), nuclear factor κB (NF-κB), tumor necrosis factor-α (TNFα), transforming growth factor β (TGF-β), and platelet-derived growth factor (PDGF). Multiple antifibrotic factors, such as interleukin-10 (IL-10), interferon-γ (IFN-γ), bone morphogenetic protein-7 (BMP-7), hepatocyte growth factor (HGF) are also downregulated in CKD. Therefore, restoration of the proper balance between pro- and antifibrotic signaling pathways could serve as a guiding principle for the design of new antifibrotic strategies that simultaneously target many pathways. The purpose of this review is to summarize the existing body of knowledge regarding activation of cytokine pathways and infiltration of inflammatory cells as a starting point for developing novel antifibrotic therapies to prevent progression of CKD.
Collapse
Affiliation(s)
- Wenshan Lv
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; Department of Endocrinology and Metabolism, the Affiliated Hospital of Qingdao University, Qingdao 26003, China
| | - George W Booz
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Yangang Wang
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Qingdao University, Qingdao 26003, China
| | - Fan Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Richard J Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA.
| |
Collapse
|
10
|
Wang J, Kang M, Wen Q, Qin YT, Wei ZX, Xiao JJ, Wang RS. Berberine sensitizes nasopharyngeal carcinoma cells to radiation through inhibition of Sp1 and EMT. Oncol Rep 2017; 37:2425-2432. [PMID: 28350122 DOI: 10.3892/or.2017.5499] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 06/07/2016] [Indexed: 11/05/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a tumor of epithelial origin with radiotherapy as its standard treatment. However, radioresistance remains a critical issue in the treatment of NPC. This study aimed to investigate the effect of berberine on the proliferation, cell cycle regulation, apoptosis, radioresistance of NPC cells and whether specificity protein 1 (Sp1) is a functional target of berberine. Our results showed that treatment with berberine reduced the proliferation and viability of CNE-2 cells in a dose- and time‑dependent manner. Berberine induced cell cycle arrest in the G0/G1 phase and apoptosis. In CNE-2 cells exposed to gamma‑ray irradiation, berberine reduced cell viability at various concentrations (25, 50, 75 and 100 µmol/l). Berberine significantly decreased mRNA and protein expression of Sp1 in the CNE-2 cells. Mithramycin A, a selective Sp1 inhibitor, enhanced the radiosensitivity and the rate of apoptosis in the CNE-2 cells. Berberine inhibited transforming growth factor-β (TGF-β)-induced tumor invasion and suppressed epithelial-to-mesenchymal transition (EMT) process, as evidenced by increased E-cadherin and decreased vimentin proteins. Sp1 may be required for the TGF-β1-induced invasion and EMT by berberine. In conclusion, berberine demonstrated the ability to suppress proliferation, induce cell cycle arrest and apoptosis, and enhance radiosensitivity of the CNE-2 NPC cells. Sp1 may be a target of berberine which is decreased during the radiosensitization of berberine.
Collapse
Affiliation(s)
- Jun Wang
- Department of Radiotherapy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Min Kang
- Department of Radiotherapy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Qin Wen
- Department of Radiotherapy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yu-Tao Qin
- Department of Radiotherapy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Zhu-Xin Wei
- Department of Radiotherapy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jing-Jian Xiao
- Department of Radiotherapy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Ren-Sheng Wang
- Department of Radiotherapy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
11
|
Jia D, Ni YR, Zhang YQ, Rao C, Hou J, Tang HQ, Liu CB, Wu JF. SP1 and UTE1 Decoy ODNs inhibit activation and proliferation of hepatic stellate cells by targeting tissue inhibitors of metalloproteinase 1. Cell Biosci 2016; 6:31. [PMID: 27175276 PMCID: PMC4863342 DOI: 10.1186/s13578-016-0094-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 04/13/2016] [Indexed: 01/10/2023] Open
Abstract
Background The excessive accumulation of extracellular matrix of hepatic fibrosis is positively correlated with tissue inhibitors of metalloproteinase 1 (TIMP1). Here we aimed to investigate whether TIMP1 may be down-regulated by Decoy ODNs strategy to capture transcriptional factor upstream TIMP1 element 1 (UTE1) and specificity protein 1(SP1). Results By luciferase reporter assays, we confirmed that these Decoy ODNs could influence the promoter activation of TIMP-1, α-SMA and Collagen Iα2 (COLΙα2) genes as well as the enhancer activation of TRE in HSC-T6 cells, and the combination tended to be more effective than SP1 or UTE1 Decoy ODN alone. Western blot analysis also demonstrated down-regulation of the expression of those target genes except for TGF-β. Furthermore, we observed that the viability of HSC-T6 cells at 72 h was significantly in decline in combination group. Conclusion The combination of SP1 and UTE1 Decoy ODNs treatments inhibit the activation and proliferation of HSCs more effectively than one of the Decoy ODNs through co-regulation of TIMP1 and TGF-β signal pathway but not the expression of TGF-β itself.
Collapse
Affiliation(s)
- Dong Jia
- Medical College, China Three Gorges University, 8 Daxue Road, Xiling District, Yichang, 443002 Hubei Province China.,Institute of Liver Diseases, China Three Gorges University, Yichang, Hubei Province China.,Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, Hubei Province China
| | - Yi-Ran Ni
- Medical College, China Three Gorges University, 8 Daxue Road, Xiling District, Yichang, 443002 Hubei Province China
| | - Yan-Qiong Zhang
- Medical College, China Three Gorges University, 8 Daxue Road, Xiling District, Yichang, 443002 Hubei Province China
| | - Chun Rao
- Medical College, China Three Gorges University, 8 Daxue Road, Xiling District, Yichang, 443002 Hubei Province China.,Institute of Liver Diseases, China Three Gorges University, Yichang, Hubei Province China.,Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, Hubei Province China
| | - Jun Hou
- First Clinical Medical College, China Three Gorges University, Yichang, Hubei Province China
| | - He-Qing Tang
- First Clinical Medical College, China Three Gorges University, Yichang, Hubei Province China
| | - Chang-Bai Liu
- Medical College, China Three Gorges University, 8 Daxue Road, Xiling District, Yichang, 443002 Hubei Province China.,Institute of Liver Diseases, China Three Gorges University, Yichang, Hubei Province China.,Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, Hubei Province China
| | - Jiang-Feng Wu
- Medical College, China Three Gorges University, 8 Daxue Road, Xiling District, Yichang, 443002 Hubei Province China.,Institute of Liver Diseases, China Three Gorges University, Yichang, Hubei Province China.,Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, Hubei Province China
| |
Collapse
|
12
|
Zhang R, Yu Y, Deng J, Zhang C, Zhang J, Cheng Y, Luo X, Han B, Yang H. Sesamin Ameliorates High-Fat Diet-Induced Dyslipidemia and Kidney Injury by Reducing Oxidative Stress. Nutrients 2016; 8:nu8050276. [PMID: 27171111 PMCID: PMC4882689 DOI: 10.3390/nu8050276] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 04/26/2016] [Accepted: 05/04/2016] [Indexed: 01/26/2023] Open
Abstract
The study explored the protective effect of sesamin against lipid-induced renal injury and hyperlipidemia in a rat model. An animal model of hyperlipidemia was established in Sprague-Dawley rats. Fifty-five adult Sprague-Dawley rats were divided into five groups. The control group was fed a standard diet, while the other four groups were fed a high-fat diet for 5 weeks to induce hyperlipidemia. Three groups received oral sesamin in doses of 40, 80, or 160 mg/(kg·day). Seven weeks later, the blood lipids, renal function, antioxidant enzyme activities, and hyperoxide levels in kidney tissues were measured. The renal pathological changes and expression levels of collagen type IV (Col-IV) and α-smooth muscle actin (α-SMA) were analyzed. The administration of sesamin improved the serum total cholesterol, triglyceride, low-density lipoprotein cholesterol, apolipoprotein-B, oxidized-low-density lipoprotein, and serum creatinine levels in hyperlipidemic rats, while it increased the high-density lipoprotein cholesterol and apolipoprotein-A levels. Sesamin reduced the excretion of 24-h urinary protein and urinary albumin and downregulated α-SMA and Col-IV expression. Moreover, sesamin ameliorated the superoxide dismutase activity and reduced malondialdehyde levels in kidney tissue. Sesamin could mediate lipid metabolism and ameliorate renal injury caused by lipid metabolism disorders in a rat model of hyperlipidemia.
Collapse
Affiliation(s)
- Ruijuan Zhang
- Department of Nutrition and Food Safety, School of Public Health of Xi'an Jiao Tong University, Xi'an 710061, China.
| | - Yan Yu
- Department of Nutrition and Food Safety, School of Public Health of Xi'an Jiao Tong University, Xi'an 710061, China.
| | - Jianjun Deng
- Shaanxi Key laboratory of Degradable Biomedical Materials, Department of Food Science and Engineering, College of Chemical Engineering, Northwest University, Xi'an 710069, China.
| | - Chao Zhang
- Department of Nutrition and Food Safety, School of Public Health of Xi'an Jiao Tong University, Xi'an 710061, China.
| | - Jinghua Zhang
- Department of Nutrition and Food Safety, School of Public Health of Xi'an Jiao Tong University, Xi'an 710061, China.
| | - Yue Cheng
- Department of Nutrition and Food Safety, School of Public Health of Xi'an Jiao Tong University, Xi'an 710061, China.
| | - Xiaoqin Luo
- Department of Nutrition and Food Safety, School of Public Health of Xi'an Jiao Tong University, Xi'an 710061, China.
| | - Bei Han
- Department of Nutrition and Food Safety, School of Public Health of Xi'an Jiao Tong University, Xi'an 710061, China.
| | - Haixia Yang
- Department of Nutrition and Food Safety, School of Public Health of Xi'an Jiao Tong University, Xi'an 710061, China.
| |
Collapse
|
13
|
Jia D, Zhang YQ, Wu JF. Decoy oligonucleotide technology in fibrosis: Application and delivery strategy. Shijie Huaren Xiaohua Zazhi 2015; 23:4931-4938. [DOI: 10.11569/wcjd.v23.i31.4931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Fibrosis is a pathological condition caused by a variety of etiologies, which is characterized by an increase in the fibrous connective tissue and a reduction in the parenchymal cells of several organs and can result in structural damage and functional impairment of organs. With the development of molecular biology and cellular biology technology in recent years, gene therapy methods for fibrosis are drawing attention, including antisense oligonucleotides, RNA interference, Decoy oligonucleotide (ODN) technology and so on. Among them, Decoy ODN technology can block the target gene expression by capturing specific transcription factors, having the potential to interfere with the expression of the fibrosis related genes. This paper will review the application of Decoy ODN technology in fibrosis as well as the delivery strategy in vivo.
Collapse
|
14
|
Youn SW, Park KK. Small-nucleic-acid-based therapeutic strategy targeting the transcription factors regulating the vascular inflammation, remodeling and fibrosis in atherosclerosis. Int J Mol Sci 2015; 16:11804-33. [PMID: 26006249 PMCID: PMC4463731 DOI: 10.3390/ijms160511804] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 05/15/2015] [Accepted: 05/18/2015] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis arises when injury to the arterial wall induces an inflammatory cascade that is sustained by a complex network of cytokines, together with accumulation of lipids and fibrous material. Inflammatory cascades involve leukocyte adherence and chemotaxis, which are coordinated by the local secretion of adhesion molecules, chemotactic factors, and cytokines. Transcription factors are critical to the integration of the various steps of the cascade response to mediators of vascular injury, and are induced in a stimulus-dependent and cell-type-specific manner. Several small-nucleic-acid-based therapeutic strategies have recently been developed to target transcription factors: antisense oligodeoxynucleotides, RNA interference, microRNA, and decoy oligodeoxynucleotides. The aim of this review was to provide an overview of these particular targeted therapeutic strategies, toward regulation of the vascular inflammation, remodeling and fibrosis associated with atherosclerosis.
Collapse
Affiliation(s)
- Sung Won Youn
- Department of Radiology, Catholic University of Daegu Medical Center, School of Medicine, Catholic University of Daegu, Daegu 705-718, Korea.
| | - Kwan-Kyu Park
- Department of Pathology, Catholic University of Daegu Medical Center, School of Medicine, Catholic University of Daegu, Daegu 705-718, Korea.
| |
Collapse
|
15
|
Zhang Y, Chen HX, Zhou SY, Wang SX, Zheng K, Xu DD, Liu YT, Wang XY, Wang X, Yan HZ, Zhang L, Liu QY, Chen WQ, Wang YF. Sp1 and c-Myc modulate drug resistance of leukemia stem cells by regulating survivin expression through the ERK-MSK MAPK signaling pathway. Mol Cancer 2015; 14:56. [PMID: 25890196 PMCID: PMC4357193 DOI: 10.1186/s12943-015-0326-0] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 02/23/2015] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is initiated and maintained by a subset of self-renewing leukemia stem cells (LSCs), which contribute to the progression, recurrence and therapeutic resistance of leukemia. However, the mechanisms underlying the maintenance of LSCs drug resistance have not been fully defined. In this study, we attempted to elucidate the mechanisms of LSCs drug resistance. METHODS We performed reverse phase protein arrays to analyze the expression of anti-apoptotic proteins in the LSC-enriched leukemia cell line KG-1a. Immuno-blotting, cell viability and clinical AML samples were evaluated to verify the micro-assay results. The characteristics and transcriptional regulation of survivin were analyzed with the relative luciferase reporter assay, mutant constructs, chromatin immuno-precipitation (ChIP), quantitative real-time reverse transcription polymerase chain reaction (RT-qPCR), and western blotting. The levels of Sp1, c-Myc, phospho-extracellular signal-regulated kinase (p-ERK), phospho-mitogen and stress-activated protein kinase (p-MSK) were investigated in paired CD34+ and CD34- AML patient samples. RESULTS Survivin was highly over-expressed in CD34 + CD38- KG-1a cells and paired CD34+ AML patients compared with their differentiated counterparts. Functionally, survivin contributes to the drug resistance of LSCs, and Sp1 and c-Myc concurrently regulate levels of survivin transcription. Clinically, Sp1 and c-Myc were significantly up-regulated and positively correlated with survivin in CD34+ AML patients. Moreover, Sp1 and c-Myc were further activated by the ERK/MSK mitogen-activated protein kinase (MAPK) signaling pathway, modulating survivin levels. CONCLUSION Our findings demonstrated that ERK/MSK/Sp1/c-Myc axis functioned as a critical regulator of survivin expression in LSCs, offering a potential new therapeutic strategy for LSCs therapy.
Collapse
Affiliation(s)
- Yi Zhang
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R, China.
- Institute of Biomedicine, Jinan University, 510632, Guangzhou, P.R China.
| | - Hai-xuan Chen
- College of Medicine, Jinan University, 510632, Guangzhou, P.R China.
| | - Shu-yan Zhou
- Department of Pathological Physiology, Wan-nan Medical College, 241000, Wuhu, P.R China.
| | - Shao-xiang Wang
- College of Medicine, Shenzhen University, 518020, Shenzhen, P.R China.
| | - Kai Zheng
- College of Medicine, Shenzhen University, 518020, Shenzhen, P.R China.
| | - Dan-dan Xu
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R, China.
| | - Yu-ting Liu
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R, China.
| | - Xiao-yan Wang
- Institute of Biomedicine, Jinan University, 510632, Guangzhou, P.R China.
| | - Xiao Wang
- Institute of Biomedicine, Jinan University, 510632, Guangzhou, P.R China.
| | - Hai-Zhao Yan
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R, China.
| | - Li Zhang
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R, China.
| | - Qiu-ying Liu
- Institute of Biomedicine, Jinan University, 510632, Guangzhou, P.R China.
| | - Wan-qun Chen
- College of Medicine, Jinan University, 510632, Guangzhou, P.R China.
| | - Yi-fei Wang
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R, China.
- Institute of Biomedicine, Jinan University, 510632, Guangzhou, P.R China.
| |
Collapse
|
16
|
Kim KH, Park KK. Small RNA- and DNA-based gene therapy for the treatment of liver cirrhosis, where we are? World J Gastroenterol 2014; 20:14696-14705. [PMID: 25356032 PMCID: PMC4209535 DOI: 10.3748/wjg.v20.i40.14696] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 04/03/2014] [Accepted: 06/05/2014] [Indexed: 02/06/2023] Open
Abstract
Chronic liver diseases with different aetiologies rely on the chronic activation of liver injuries which result in a fibrogenesis progression to the end stage of cirrhosis and liver failure. Based on the underlying cellular and molecular mechanisms of a liver fibrosis, there has been proposed several kinds of approaches for the treatment of liver fibrosis. Recently, liver gene therapy has been developed as an alternative way to liver transplantation, which is the only effective therapy for chronic liver diseases. The activation of hepatic stellate cells, a subsequent release of inflammatory cytokines and an accumulation of extracellular matrix during the liver fibrogenesis are the major obstacles to the treatment of liver fibrosis. Several targeted strategies have been developed, such as antisense oligodeoxynucleotides, RNA interference and decoy oligodeoxynucleotides to overcome this barriers. With this report an overview will be provided of targeted strategies for the treatment of liver cirrhosis, and particularly, of the targeted gene therapy using short RNA and DNA segments.
Collapse
|