1
|
Huang Z, Tan Y. The Potential of Cylindromatosis (CYLD) as a Therapeutic Target in Oxidative Stress-Associated Pathologies: A Comprehensive Evaluation. Int J Mol Sci 2023; 24:8368. [PMID: 37176077 PMCID: PMC10179184 DOI: 10.3390/ijms24098368] [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: 03/29/2023] [Revised: 04/25/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
Oxidative stress (OS) arises as a consequence of an imbalance between the formation of reactive oxygen species (ROS) and the capacity of antioxidant defense mechanisms to neutralize them. Excessive ROS production can lead to the damage of critical biomolecules, such as lipids, proteins, and DNA, ultimately contributing to the onset and progression of a multitude of diseases, including atherosclerosis, chronic obstructive pulmonary disease, Alzheimer's disease, and cancer. Cylindromatosis (CYLD), initially identified as a gene linked to familial cylindromatosis, has a well-established and increasingly well-characterized function in tumor inhibition and anti-inflammatory processes. Nevertheless, burgeoning evidence suggests that CYLD, as a conserved deubiquitination enzyme, also plays a pivotal role in various key signaling pathways and is implicated in the pathogenesis of numerous diseases driven by oxidative stress. In this review, we systematically examine the current research on the function and pathogenesis of CYLD in diseases instigated by oxidative stress. Therapeutic interventions targeting CYLD may hold significant promise for the treatment and management of oxidative stress-induced human diseases.
Collapse
Affiliation(s)
| | - Yanjie Tan
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250358, China;
| |
Collapse
|
2
|
Zhou JJ, Li H, Li L, Li Y, Wang PH, Meng XM, He JG. CYLD mediates human pulmonary artery smooth muscle cell dysfunction in congenital heart disease-associated pulmonary arterial hypertension. J Cell Physiol 2021; 236:6297-6311. [PMID: 33507567 DOI: 10.1002/jcp.30298] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/31/2020] [Accepted: 01/11/2021] [Indexed: 11/11/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a common complication of congenital heart disease (CHD). Deubiquitinase cylindromatosis (CYLD) has been reported to significantly aggravate vascular smooth muscle cell (VSMC) phenotypic transformation, proliferation, and migration. Here, we aimed to further investigate its roles and underlying mechanisms in the CHD-PAH development. The expression of CYLD in the lung tissues from CHD-PAH patients and monocrotaline (MCT) plus aortocaval (AV)-induced PAH rats, pulmonary artery smooth muscle cells (PASMCs) from MCT-AV-induced PAH rats, and human PASMCs (HPASMCs) was evaluated. After infection with CYLD siRNA or pcNDA3.1-CYLD, the proliferation, migration, and apoptosis of HPASMCs were measured using an EdU assay, transwell and scratch wound healing assays, and flow cytometric assay, respectively. An adeno-associated virus (AAV) vector encoding CYLD was used to suppress CYLD expression by being intratracheally instilled in rats 7 days before MCT-AV treatment. The results showed that CYLD was increased in the lung tissues from CHD-PAH patients and MCT-AV-induced PAH rats, and in PASMCs from MCT-AV-induced PAH rats. The contractile-type HPASMCs expressed low levels of CYLD, while the proliferative synthetic-type HPASMCs expressed high levels of CYLD. In addition, CYLD could mediate HPASMC dysfunction, which regulated HPASMC phenotypic transformation and proliferation via the modulation of p38 and ERK activation, while CYLD regulated HPASMC migration via the modulation of p38 activation. In vivo results demonstrated that the local suppression of CYLD expression could attenuate the increased levels of PAH and its associated pulmonary vascular remodeling in MCT-AV-induced PAH rats. Collectively, these results indicated that CYLD might be a potential novel therapeutic target for the prevention of PAH and pulmonary vascular remodeling in CHD-PAH through the modulation of HPASMC dysfunction.
Collapse
Affiliation(s)
- Jing-Jing Zhou
- Center of Pulmonary Vascular Disease, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huang Li
- Department of Cardiology, Guangdong Cardiovascular Institute Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Li Li
- Department of Pathology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yue Li
- The Animal Experimental Centre, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pei-He Wang
- The Animal Experimental Centre, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xian-Min Meng
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jian-Guo He
- Center of Pulmonary Vascular Disease, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
3
|
Fu YW, Li L, Wang XQ, Zhou Y, Zhu LF, Mei YM, Xu Y. The inhibitory effect of the deubiquitinase cylindromatosis (CYLD) on inflammatory responses in human gingival fibroblasts. Oral Dis 2020; 27:1487-1497. [PMID: 33031609 DOI: 10.1111/odi.13672] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 09/22/2020] [Accepted: 10/01/2020] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Experiments were performed to evaluate CYLD expression in human gingival tissue samples and to examine the effects of CYLD on inflammatory responses in lipopolysaccharide (LPS)- or TNF-α-stimulated human gingival fibroblasts (HGFs). METHODS Immunohistochemistry for CYLD and p65 expression was performed with healthy and inflamed gingival tissue samples. siRNA was used to knock down the expression of CYLD in HGFs. Upon LPS or TNF-α stimulation, NF-κB activation was detected in control and CYLD-knockdown HGFs. RT-PCR was applied to determine gene expression. Western blot analyses were employed to assess protein expression. Immunofluorescence staining was carried out to evaluate the nuclear translocation of p65. RESULTS Immunohistochemical staining showed the expression of CYLD in human gingival tissues. In addition, CYLD protein expression was reduced in inflamed gingival tissue samples compared with healthy tissue samples. CYLD knockdown greatly enhanced the mRNA expression of proinflammatory cytokines in LPS- or TNF-α-stimulated HGFs. Furthermore, knocking down CYLD expression increased LPS-stimulated NF-κB activation in HGFs. Unexpectedly, CYLD knockdown did not affect TNF-α-induced NF-κB activation. CONCLUSIONS Our results suggest that CYLD participates in periodontal inflammatory responses by negatively regulating LPS-induced NF-κB signalling.
Collapse
Affiliation(s)
- Yong-Wei Fu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China.,Department of Stomatology, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Lu Li
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Xiao-Qian Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Yi Zhou
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Li-Fang Zhu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - You-Min Mei
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Yan Xu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
4
|
Imaizumi Y, Takami Y, Yamamoto K, Nagasawa M, Nozato Y, Nozato S, Takeshita H, Wang C, Yokoyama S, Hayashi H, Hongyo K, Akasaka H, Takeya Y, Sugimoto K, Nakagami H, Rakugi H. Pathophysiological significance of cylindromatosis in the vascular endothelium and macrophages for the initiation of age-related atherogenesis. Biochem Biophys Res Commun 2019; 508:1168-1174. [DOI: 10.1016/j.bbrc.2018.12.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 12/04/2018] [Indexed: 10/27/2022]
|
5
|
Salvianolic Acid A Inhibits OX-LDL Effects on Exacerbating Choroidal Neovascularization via Downregulating CYLD. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:6210694. [PMID: 29081889 PMCID: PMC5610829 DOI: 10.1155/2017/6210694] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 02/28/2017] [Accepted: 03/08/2017] [Indexed: 11/17/2022]
Abstract
Backgrounds Age-related macular degeneration is closely related to lipid oxidation, while relationship between OX-LDL and choroidal neovascularization is unclear. Recently, cylindromatosis is proved to regulate angiogenesis. However, its role in CNV progression remained unclear. Salvianolic acid A is widely used in vascular diseases. We investigated the relationship between OX-LDL and CNV and explore antineovascularization mechanism of Sal A. Methods C57BL6/J mice were randomized into four groups and injected with PBS or OX-LDL, together with Sal A for one week. CNV was induced by laser; CNV severity was analyzed by fundus fluorescein angiography, H&E staining, and choroid flat mount after 1 week. In in vitro experiments, ARPE-19 and HUVECs were cultured with OX-LDL (with or without Sal A) for 48 hours. Angiogenic proteins, cell junction integrity, and tube formation were measured. CYLD siRNA and specific inhibitors were used to explore mechanisms of CYLD in promoting OX-LDL-induced CNV progression. Results OX-LDL promoted laser-induced CNV volume by increasing VEGF, PDGF, and CYLD levels. Sal A antagonized OX-LDL effects and restrained CNV progression by decreasing VEGF/PDGF/CYLD, increasing antiangiostatin levels, and promoting P62-CYLD-TRAF6 interaction. Conclusions We demonstrated oxidation damage exacerbates CNV progression, and Sal A could be a clinical therapeutic reagent to exudative AMD.
Collapse
|
6
|
Yu B, Liu Z, Fu Y, Wang Y, Zhang L, Cai Z, Yu F, Wang X, Zhou J, Kong W. CYLD Deubiquitinates Nicotinamide Adenine Dinucleotide Phosphate Oxidase 4 Contributing to Adventitial Remodeling. Arterioscler Thromb Vasc Biol 2017; 37:1698-1709. [PMID: 28751569 DOI: 10.1161/atvbaha.117.309859] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 07/13/2017] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Transdifferentiation of adventitial fibroblasts (AFs) into myofibroblasts plays a critical role during the vascular remodeling that occurs during atherosclerosis, restenosis, and aortic aneurysm. The ubiquitination/deubiquitination regulatory system is essential for the quality control of proteins. The involvement of ubiquitination/deubiquitination during AF transdifferentiation remains largely unknown. In this study, we determined the role of cylindromatosis (CYLD), a deubiquitinase, in the process of AF differentiation and activation in vitro and in vivo. APPROACH AND RESULTS Transforming growth factor-β1 and homocysteine, 2 known inducers of AF transdifferentiation, greatly upregulated CYLD expression in a time- and dose-dependent manner. The silencing of CYLD significantly inhibited AF transdifferentiation and activation as evidenced by the expression of contractile proteins, the production of the proinflammatory cytokines MCP-1 (monocyte chemotactic protein 1) and IL-6 (interleukin-6), the deposition of extracellular matrix, and cell migration. We further asked whether CYLD mediates AF activation via the regulation of nicotinamide adenine dinucleotide phosphate oxidase 4 (Nox4) as it is an essential factor during AF transdifferentiation. Indeed, the silencing of CYLD repressed transforming growth factor-β1-induced and homocysteine-induced Nox4 upregulation and reactive oxygen species production, whereas Nox4 overexpression greatly rescued the inhibitory effect on AF activation by CYLD silencing. Most interestingly, transforming growth factor-β1 and homocysteine repressed Nox4 ubiquitination and prolonged the half-life of Nox4. Moreover, Nox4 was deubiquitinated via a direct interaction with the ubiquitin-specific protease domain of CYLD. In accordance, hyperhomocysteinemia significantly increased adventitial CYLD and Nox4 expression, promoted AF transdifferentiation, and aggravated CaPO4-induced abdominal aortic aneurysm in mice. These effects were abolished in CYLD-/- mice. CONCLUSIONS CYLD contributes to the transdifferentiation of AFs via deubiquitinating Nox4 and may play a role in vascular remodeling.
Collapse
Affiliation(s)
- Bing Yu
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Ziyi Liu
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Yi Fu
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Yingbao Wang
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Lu Zhang
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Zeyu Cai
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Fang Yu
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Xian Wang
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Jun Zhou
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.).
| | - Wei Kong
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.).
| |
Collapse
|
7
|
Zhang Y, Zhang XJ, Wang PX, Zhang P, Li H. Reprogramming Innate Immune Signaling in Cardiometabolic Disease. Hypertension 2017; 69:747-760. [PMID: 28320852 DOI: 10.1161/hypertensionaha.116.08192] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Yaxing Zhang
- From the Department of Cardiology, Renmin Hospital (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), School of Basic Medical Sciences (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), Institute of Model Animal (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), and Medical Research Institute, School of Medicine (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), Wuhan University, P.R. China
| | - Xiao-Jing Zhang
- From the Department of Cardiology, Renmin Hospital (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), School of Basic Medical Sciences (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), Institute of Model Animal (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), and Medical Research Institute, School of Medicine (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), Wuhan University, P.R. China
| | - Pi-Xiao Wang
- From the Department of Cardiology, Renmin Hospital (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), School of Basic Medical Sciences (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), Institute of Model Animal (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), and Medical Research Institute, School of Medicine (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), Wuhan University, P.R. China
| | - Peng Zhang
- From the Department of Cardiology, Renmin Hospital (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), School of Basic Medical Sciences (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), Institute of Model Animal (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), and Medical Research Institute, School of Medicine (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), Wuhan University, P.R. China
| | - Hongliang Li
- From the Department of Cardiology, Renmin Hospital (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), School of Basic Medical Sciences (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), Institute of Model Animal (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), and Medical Research Institute, School of Medicine (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), Wuhan University, P.R. China.
| |
Collapse
|
8
|
Yin L, Liu S, Li C, Ding S, Bi D, Niu Z, Han L, Li W, Gao D, Liu Z, Lu J. CYLD downregulates Livin and synergistically improves gemcitabine chemosensitivity and decreases migratory/invasive potential in bladder cancer: the effect is autophagy-associated. Tumour Biol 2016; 37:12731-12742. [PMID: 27448305 DOI: 10.1007/s13277-016-5157-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 07/12/2016] [Indexed: 12/11/2022] Open
Abstract
Although GC (gemcitabine and cisplatin) chemotherapy remains an effective method for treating bladder cancer (BCa), chemoresistance is a major obstacle in chemotherapy. In this study, we determined whether gemcitabine resistance correlates with migratory/invasive potential in BCa and whether this relationship is regulated by the cylindromatosis (CYLD)-Livin module. First, we independently investigated the correlation of CYLD/Livin and gemcitabine resistance with the potential for tumor migration and invasiveness. Second, we found that co-transfected CYLD and Livin dramatically improved sensitivity to gemcitabine chemotherapy and decreased migration/invasion potential. Next, we determined that CYLD may regulate Livin by the NF-κB-dependent pathway. We also found that CYLD overexpression and Livin knockdown might improve gemcitabine chemosensitivity by decreasing autophagy and increasing apoptosis in BCa cells. Finally, the effects of CYLD-Livin on tumor growth in vivo were evaluated. Our study demonstrates that CYLD-Livin might represent a potential therapeutic for chemoresistant BCa.
Collapse
Affiliation(s)
- Lei Yin
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jingwu Road, No 324, Jinan, 250021, Shandong, China
| | - Shuai Liu
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jingwu Road, No 324, Jinan, 250021, Shandong, China
| | - Chensheng Li
- Department of Digestive Diseases, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, China
| | - Sentai Ding
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jingwu Road, No 324, Jinan, 250021, Shandong, China
| | - Dongbin Bi
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jingwu Road, No 324, Jinan, 250021, Shandong, China
| | - Zhihong Niu
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jingwu Road, No 324, Jinan, 250021, Shandong, China
| | - Liping Han
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250014, China
| | - Wenjia Li
- Shandong University, Jinan, 250000, China
| | - Dexuan Gao
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jingwu Road, No 324, Jinan, 250021, Shandong, China
| | - Zheng Liu
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jingwu Road, No 324, Jinan, 250021, Shandong, China
| | - Jiaju Lu
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jingwu Road, No 324, Jinan, 250021, Shandong, China.
| |
Collapse
|
9
|
Li SW, Wang CY, Jou YJ, Huang SH, Hsiao LH, Wan L, Lin YJ, Kung SH, Lin CW. SARS Coronavirus Papain-Like Protease Inhibits the TLR7 Signaling Pathway through Removing Lys63-Linked Polyubiquitination of TRAF3 and TRAF6. Int J Mol Sci 2016; 17:ijms17050678. [PMID: 27164085 PMCID: PMC4881504 DOI: 10.3390/ijms17050678] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 04/26/2016] [Accepted: 04/26/2016] [Indexed: 12/12/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV) papain-like protease (PLPro) reportedly inhibits the production of type I interferons (IFNs) and pro-inflammatory cytokines in Toll-like receptor 3 (TLR3) and retinoic acid-inducible gene 1 (RIG-I) pathways. The study investigated the inhibitory effect and its antagonistic mechanism of SARS-CoV PLPro on TLR7-mediated cytokine production. TLR7 agonist (imiquimod (IMQ)) concentration-dependently induced activation of ISRE-, NF-κB- and AP-1-luciferase reporters, as well as the production of IFN-α, IFN-β, TNF-α, IL-6 and IL-8 in human promonocyte cells. However, SARS-CoV PLPro significantly inhibited IMQ-induced cytokine production through suppressing the activation of transcription factors IRF-3, NF-κB and AP-1. Western blot analysis with anti-Lys48 and anti-Lys63 ubiquitin antibodies indicated the SARS-CoV PLPro removed Lys63-linked ubiquitin chains of TRAF3 and TRAF6, but not Lys48-linked ubiquitin chains in un-treated and treated cells. The decrease in the activated state of TRAF3 and TRAF6 correlated with the inactivation of TBK1 in response to IMQ by PLPro. The results revealed that the antagonism of SARS-CoV PLPro on TLR7-mediated innate immunity was associated with the negative regulation of TRAF3/6-TBK1-IRF3/NF-κB/AP1 signals.
Collapse
Affiliation(s)
- Shih-Wen Li
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404, Taiwan.
| | - Ching-Ying Wang
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404, Taiwan.
| | - Yu-Jen Jou
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404, Taiwan.
| | - Su-Hua Huang
- Department of Biotechnology, College of Health Science, Asia University, Wufeng, Taichung 413, Taiwan.
| | - Li-Hsin Hsiao
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404, Taiwan.
| | - Lei Wan
- Department of Medical Genetics and Medical Research, China Medical University Hospital, Taichung 404, Taiwan.
| | - Ying-Ju Lin
- Department of Medical Genetics and Medical Research, China Medical University Hospital, Taichung 404, Taiwan.
| | - Szu-Hao Kung
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming University, Taipei 112, Taiwan.
| | - Cheng-Wen Lin
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404, Taiwan.
- Department of Biotechnology, College of Health Science, Asia University, Wufeng, Taichung 413, Taiwan.
| |
Collapse
|
10
|
Mathis BJ, Lai Y, Qu C, Janicki JS, Cui T. CYLD-mediated signaling and diseases. Curr Drug Targets 2016; 16:284-94. [PMID: 25342597 DOI: 10.2174/1389450115666141024152421] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 09/26/2014] [Accepted: 10/03/2014] [Indexed: 02/07/2023]
Abstract
The conserved cylindromatosis (CYLD) codes for a deubiquitinating enzyme and is a crucial regulator of diverse cellular processes such as immune responses, inflammation, death, and proliferation. It directly regulates multiple key signaling cascades, such as the Nuclear Factor kappa B [NFkB] and the Mitogen-Activated Protein Kinase (MAPK) pathways, by its catalytic activity on polyubiquitinated key intermediates. Several lines of emerging evidence have linked CYLD to the pathogenesis of various maladies, including cancer, poor infection control, lung fibrosis, neural development, and now cardiovascular dysfunction. While CYLD-mediated signaling is cell type and stimuli specific, the activity of CYLD is tightly controlled by phosphorylation and other regulators such as Snail. This review explores a broad selection of current and past literature regarding CYLD's expression, function and regulation with emerging reports on its role in cardiovascular disease.
Collapse
Affiliation(s)
| | | | | | | | - Taixing Cui
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209, USA.
| |
Collapse
|
11
|
Kraus WE, Muoio DM, Stevens R, Craig D, Bain JR, Grass E, Haynes C, Kwee L, Qin X, Slentz DH, Krupp D, Muehlbauer M, Hauser ER, Gregory SG, Newgard CB, Shah SH. Metabolomic Quantitative Trait Loci (mQTL) Mapping Implicates the Ubiquitin Proteasome System in Cardiovascular Disease Pathogenesis. PLoS Genet 2015; 11:e1005553. [PMID: 26540294 PMCID: PMC4634848 DOI: 10.1371/journal.pgen.1005553] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 09/04/2015] [Indexed: 12/15/2022] Open
Abstract
Levels of certain circulating short-chain dicarboxylacylcarnitine (SCDA), long-chain dicarboxylacylcarnitine (LCDA) and medium chain acylcarnitine (MCA) metabolites are heritable and predict cardiovascular disease (CVD) events. Little is known about the biological pathways that influence levels of most of these metabolites. Here, we analyzed genetics, epigenetics, and transcriptomics with metabolomics in samples from a large CVD cohort to identify novel genetic markers for CVD and to better understand the role of metabolites in CVD pathogenesis. Using genomewide association in the CATHGEN cohort (N = 1490), we observed associations of several metabolites with genetic loci. Our strongest findings were for SCDA metabolite levels with variants in genes that regulate components of endoplasmic reticulum (ER) stress (USP3, HERC1, STIM1, SEL1L, FBXO25, SUGT1) These findings were validated in a second cohort of CATHGEN subjects (N = 2022, combined p = 8.4x10-6–2.3x10-10). Importantly, variants in these genes independently predicted CVD events. Association of genomewide methylation profiles with SCDA metabolites identified two ER stress genes as differentially methylated (BRSK2 and HOOK2). Expression quantitative trait loci (eQTL) pathway analyses driven by gene variants and SCDA metabolites corroborated perturbations in ER stress and highlighted the ubiquitin proteasome system (UPS) arm. Moreover, culture of human kidney cells in the presence of levels of fatty acids found in individuals with cardiometabolic disease, induced accumulation of SCDA metabolites in parallel with increases in the ER stress marker BiP. Thus, our integrative strategy implicates the UPS arm of the ER stress pathway in CVD pathogenesis, and identifies novel genetic loci associated with CVD event risk. Cardiovascular disease is a strongly heritable trait. Despite application of the latest genomic technologies, the genetic architecture of disease risk remains poorly defined, and mechanisms underlying this susceptibility are incompletely understood. In this study, we performed genome-wide mapping of heart disease-related metabolites measured in the blood as the genetic traits of interest (instead of the disease itself), in a large cohort of 3512 patients at risk of heart disease from the CATHGEN study. Our goal was to discover new cardiovascular disease genes and thereby mechanisms of disease pathogenesis by understanding the genes that regulate levels of these metabolites. These analyses identified novel genetic variants associated with metabolite levels and with cardiovascular disease itself. Importantly, by utilizing an unbiased systems-based approach integrating genetics, gene expression, epigenetics and metabolomics, we uncovered a novel pathway of heart disease pathogenesis, that of endoplasmic reticulum (ER) stress, represented by elevated levels of circulating short-chain dicarboxylacylcarnitine (SCDA) metabolites.
Collapse
Affiliation(s)
- William E. Kraus
- Division of Cardiology, Department of Medicine, Duke University, Durham, North Carolina, United States of America
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, United States of America
| | - Deborah M. Muoio
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, United States of America
- Division of Endocrinology, Department of Medicine, Duke University, Durham, North Carolina, United States of America
| | - Robert Stevens
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, United States of America
| | - Damian Craig
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, United States of America
| | - James R. Bain
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, United States of America
| | - Elizabeth Grass
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, United States of America
| | - Carol Haynes
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, United States of America
| | - Lydia Kwee
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, United States of America
| | - Xuejun Qin
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, United States of America
| | - Dorothy H. Slentz
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, United States of America
| | - Deidre Krupp
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, United States of America
| | - Michael Muehlbauer
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, United States of America
| | - Elizabeth R. Hauser
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, United States of America
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, United States of America
| | - Simon G. Gregory
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, United States of America
| | - Christopher B. Newgard
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, United States of America
| | - Svati H. Shah
- Division of Cardiology, Department of Medicine, Duke University, Durham, North Carolina, United States of America
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
12
|
Xu B, Yang H, Sun M, Chen H, Jiang L, Zheng X, Ding G, Liu Y, Sheng Y, Cui D, Duan Y. 2,3',4,4',5-Pentachlorobiphenyl Induces Inflammatory Responses in the Thyroid Through JNK and Aryl Hydrocarbon Receptor-Mediated Pathway. Toxicol Sci 2015; 149:300-11. [PMID: 26519956 DOI: 10.1093/toxsci/kfv235] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Polychlorinated biphenyls (PCBs) are durable and widely distributed environmental contaminants that can compromise the normal functions of multiple organs and systems; one important mechanism is the induction of inflammatory disorders. In this study, we explored the influences of 2,3',4,4',5-pentachlorobiphenyl (PCB118) on inflammatory responses and its underlying mechanisms in the thyroid. Wistar rats were administered PCB118 intraperitoneally at 0, 10, 100, and 1000 μg/kg/d, 5 days a week for 13 weeks; rat thyroid FRTL-5 cells were treated with PCB118 (0, 0.25, 2.5, and 25 nM) for indicated time. Results revealed that PCB118 promoted the generation of interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), and intercellular adhesion molecule-1 (ICAM-1) in a time- and dose-related manner and decreased sodium/iodide symporter (NIS) protein expression. Moreover, stimulation with PCB118 resulted in the upregulation of the aryl hydrocarbon receptor (AhR)-responsive gene cytochrome P450 1A1 in FRTL-5 cells; whereas pretreatment with the AhR inhibitor α-naphthoflavone or AhR small interfering RNA (siRNA) suppressed AhR, CYP1A1, IL-6, and ICAM-1 and restored NIS expression. In vivo and in vitro studies also suggested that the c-Jun N-terminal kinase (JNK) pathway was activated on PCB118 exposure, and the experiments using siRNA for JNK partially blocked PCB118-induced upregulation of IL-6 and ICAM-1 and downregulation of NIS. Altogether, PCB118 stimulates production of IL-6, TNF-α, and ICAM-1 in the thyroid through AhR and JNK activations and subsequently interferes with NIS expression, resulting in the disruption of thyroid structure and function.
Collapse
Affiliation(s)
- Bojin Xu
- *Department of Endocrinology and
| | - Hui Yang
- *Department of Endocrinology and
| | | | | | | | | | | | - Yun Liu
- Department of Gerontology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yunlu Sheng
- Department of Gerontology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Dai Cui
- *Department of Endocrinology and
| | - Yu Duan
- *Department of Endocrinology and
| |
Collapse
|
13
|
AGUIRIANO-MOSER VICTOR, SVEJDA BERNHARD, LI ZENGXIA, STURM SONJA, STUPPNER HERMANN, INGOLIC ELISABETH, HÖGER HARALD, SIEGL VERONIKA, MEIER-ALLARD NATHALIE, SADJAK ANTON, PFRAGNER ROSWITHA. Ursolic acid from Trailliaedoxa gracilis induces apoptosis in medullary thyroid carcinoma cells. Mol Med Rep 2015; 12:5003-11. [PMID: 26151624 PMCID: PMC4581794 DOI: 10.3892/mmr.2015.4053] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 02/10/2015] [Indexed: 01/08/2023] Open
Abstract
Medullary thyroid carcinoma (MTC) originates from the C‑cells of the thyroid and is not sensitive to radiation or chemotherapy. Therefore, surgical removal of the tumor tissue in its entirety is the only curative treatment for MTC. The present study aimed to examine the potential mechanisms of action of extracts of Trailliaedoxa gracilis (TG; WW Smith & Forrest), a plant from the province of Sichuan, China, and of ursolic acid (UA), a pentacyclic triterpen present in TG, on the MTC‑SK MTC cell line. A total of 13 TG fractions and UA were examined in vitro for their effects on cell morphology, cell number, proliferation and rates of apoptosis. Reverse transcription‑quantitative polymerase chain reaction of nuclear factor‑κB essential modifier (NEMO) was performed to delineate the role of the apoptotic pathway following treatment with UA. TG and UA were examined in vivo in xenotransplanted MTC‑bearing severe combined immunodeficient mice. The TG fractions exhibited antiproliferative effects, with inhibition of mitochondrial activity in the tumor cells at concentrations, which caused no impairment of the normal control cells. The apoptotic rates of the MTC‑SK cells treated with the TG fractions and UA were determined, in which no marked tumor inhibition was observed in the treated MTC‑mice, and no change in the expression of NEMO was detected in the treated MTC‑SK cells. The observation of early‑onset activation of caspase 8 suggested that the responsible factor was linked to NEMO, an anti‑apoptotic protein. However, no differences in the mRNA transcription levels of NEMO were detected in MTC‑SK cells treated with UA, suggesting that this protein was not associated with the signal transducer and activator of transcription 3 pathway.
Collapse
Affiliation(s)
- VICTOR AGUIRIANO-MOSER
- Department of Pathophysiology and Immunology, Center of Molecular Medicine, Medical University of Graz, Graz A-8010, Austria
| | - BERNHARD SVEJDA
- Department of Pathophysiology and Immunology, Center of Molecular Medicine, Medical University of Graz, Graz A-8010, Austria
| | - ZENG-XIA LI
- Department of Pathophysiology and Immunology, Center of Molecular Medicine, Medical University of Graz, Graz A-8010, Austria
- Department of Biochemistry & Molecular Biology, Shanghai Medical School, Fudan University, Shanghai 200433, P.R. China
| | - SONJA STURM
- Department of Pharmacognosy, Institute of Pharmacy, Center of Molecular Biosciences, Leopold Franzens University of Innsbruck, Innsbruck A-6010, Austria
| | - HERMANN STUPPNER
- Department of Pharmacognosy, Institute of Pharmacy, Center of Molecular Biosciences, Leopold Franzens University of Innsbruck, Innsbruck A-6010, Austria
| | - ELISABETH INGOLIC
- Core Unit of Biomedical Research, ivision of Laboratory Animal Science and Genetics, Medical University of Vienna, Himberg A-2325, Austria
| | - HARALD HÖGER
- Research Institute for Electron Microscopy and Fine Structure Research, University of Technology Graz, Graz A-8010, Austria
| | - VERONIKA SIEGL
- Department of Pathophysiology and Immunology, Center of Molecular Medicine, Medical University of Graz, Graz A-8010, Austria
| | - NATHALIE MEIER-ALLARD
- Department of Pathophysiology and Immunology, Center of Molecular Medicine, Medical University of Graz, Graz A-8010, Austria
| | - ANTON SADJAK
- Department of Pathophysiology and Immunology, Center of Molecular Medicine, Medical University of Graz, Graz A-8010, Austria
| | - ROSWITHA PFRAGNER
- Department of Pathophysiology and Immunology, Center of Molecular Medicine, Medical University of Graz, Graz A-8010, Austria
| |
Collapse
|
14
|
Deubiquitinating enzyme CYLD mediates pressure overload-induced cardiac maladaptive remodeling and dysfunction via downregulating Nrf2. J Mol Cell Cardiol 2015; 84:143-53. [PMID: 25935309 DOI: 10.1016/j.yjmcc.2015.04.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 03/26/2015] [Accepted: 04/10/2015] [Indexed: 01/04/2023]
Abstract
Ubiquitin proteasome system (UPS) consists of ubiquitin, ubiquitin-activating enzymes (E1s), ubiquitin-conjugating enzymes (E2s), ubiquitin ligases (E3s), proteasomes, and deubiquitinating enzymes (DUBs). Ubiquitin, E1s, several E2s, E3s, and proteasomes play an important role in the regulation of cardiac homeostasis and dysfunction; however, less is known about the role of DUBs in the heart. Here, we uncovered a crucial role of cyclindromatosis (CYLD), a DUB, in mediating cardiac maladaptive remodeling and dysfunction. CYLD expression was dramatically upregulated in the cardiomyocytes of hypertrophic and failing human and murine hearts. Knockout of CYLD improved survival rate and alleviated cardiac hypertrophy, fibrosis, apoptosis, oxidative stress, and dysfunction in mice that were subjected to sustained pressure overload induced by transverse aortic constriction. Deep sequencing and gene array analyses revealed that the most dramatically changed genes are those involving in the free radical scavenging pathway and cardiovascular disease, including fos, jun, myc, and nuclear factor erythroid-2 related factor 2 (Nrf2) in the heart. Moreover, knockdown of CYLD enhanced mitogen-activated protein kinase (MAPK) ERK- and p38-mediated expression of c-jun, c-fos, and c-myc, which govern Nrf2 expression in cardiomyocytes. The CYLD deficiency-induced suppression of reactive oxygen species (ROS) formation, death and hypertrophy in cardiomyocytes was blocked by additional knockdown of Nrf2. Taken together, our findings demonstrate for the first time that CYLD mediates cardiac maladaptive remodeling and dysfunction, most likely via enhancing myocardial oxidative stress in response to pressure overload. At the molecular level, CYLD interrupts the ERK- and p38-/AP-1 and c-Myc pathways to suppress Nrf2-operated antioxidative capacity, thereby enhancing oxidative stress in the heart.
Collapse
|
15
|
Nanduri B, Suvarnapunya AE, Venkatesan M, Edelmann MJ. Deubiquitinating enzymes as promising drug targets for infectious diseases. Curr Pharm Des 2013; 19:3234-47. [PMID: 23151130 DOI: 10.2174/1381612811319180008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 11/01/2012] [Indexed: 01/25/2023]
Abstract
Deubiquitinating enzymes (DUBs) remove ubiquitin and ubiquitin-like modifications from proteins and they have been known to contribute to processes relevant in microbial infection, such as immune responses pathways. Numerous viral and bacterial DUBs have been identified, and activities of several host DUBs are known to be modulated during the infection process, either by a pathogen or by a host. Recently there have been attempts to take advantage of this feature and design therapeutic inhibitors of DUBs that can be used to limit the spread of infection. This review is focused on exploring the potential of DUBs in the treatment of infectious diseases.
Collapse
Affiliation(s)
- Bindu Nanduri
- Institute for Genomics, Biocomputing and Biotechnology, Mississippi State University, Mississippi State, MS 39762, USA
| | | | | | | |
Collapse
|
16
|
Wang TR, Yang G, Liu GN. DNA Enzyme ED5 Depletes Egr-1 and Inhibits Neointimal Hyperplasia in Rats. Cardiology 2013; 125:192-200. [DOI: 10.1159/000350364] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 02/10/2013] [Indexed: 11/19/2022]
|
17
|
Li SW, Yang TC, Wan L, Lin YJ, Tsai FJ, Lai CC, Lin CW. Correlation between TGF-β1 expression and proteomic profiling induced by severe acute respiratory syndrome coronavirus papain-like protease. Proteomics 2012; 12:3193-205. [PMID: 22936401 PMCID: PMC7168038 DOI: 10.1002/pmic.201200225] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Revised: 07/20/2012] [Accepted: 08/09/2012] [Indexed: 01/13/2023]
Abstract
Severe acute respiratory syndrome (SARS) coronavirus (SARS‐CoV) papain‐like protease (PLpro), a deubiquitinating enzyme, demonstrates inactivation of interferon (IFN) regulatory factor 3 and NF‐κB, reduction of IFN induction, and suppression of type I IFN signaling pathway. This study investigates cytokine expression and proteomic change induced by SARS‐CoV PLpro in human promonocyte cells. PLpro significantly increased TGF‐β1 mRNA expression (greater than fourfold) and protein production (greater than threefold). Proteomic analysis, Western blot, and quantitative real‐time PCR assays indicated PLpro upregulating TGF‐β1‐associated genes: HSP27, protein disulfide isomerase A3 precursor, glial fibrillary acidic protein, vimentin, retinal dehydrogenase 2, and glutathione transferase omega‐1. PLpro‐activated ubiquitin proteasome pathway via upregulation of ubiquitin‐conjugating enzyme E2–25k and proteasome subunit alpha type 5. Proteasome inhibitor MG‐132 significantly reduced expression of TGF‐β1 and vimentin. PLpro upregulated HSP27, linking with activation of p38 MAPK and ERK1/2 signaling. Treatment with SB203580 and U0126 reduced PLpro‐induced expression of TGF‐β1, vimentin, and type I collagen. Results point to SARS‐CoV PLpro triggering TGF‐β1 production via ubiquitin proteasome, p38 MAPK, and ERK1/2‐mediated signaling.
Collapse
Affiliation(s)
- Shih-Wen Li
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan; Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan
| | | | | | | | | | | | | |
Collapse
|