1
|
Liu X, Gao X, Yang Y, Yang D, Guo Q, Li L, Liu S, Cong W, Lu S, Hou L, Wang B, Li N. EVA1A reverses lenvatinib resistance in hepatocellular carcinoma through regulating PI3K/AKT/p53 signaling axis. Apoptosis 2024; 29:1161-1184. [PMID: 38743191 DOI: 10.1007/s10495-024-01967-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2024] [Indexed: 05/16/2024]
Abstract
Lenvatinib is a commonly used first-line drug for the treatment of advanced hepatocellular carcinoma (HCC). However, its clinical efficacy is limited due to the drug resistance. EVA1A was a newly identified tumor suppressor, nevertheless, the impact of EVA1A on resistance to lenvatinib treatment in HCC and the potential molecular mechanisms remain unknown. In this study, the expression of EVA1A in HCC lenvatinib-resistant cells is decreased and its low expression was associated with a poor prognosis of HCC. Overexpression of EVA1A reversed lenvatinib resistance in vitro and in vivo, as demonstrated by its ability to promote cell apoptosis and inhibit cell proliferation, invasion, migration, EMT, and tumor growth. Silencing EVA1A in lenvatinib-sensitive parental HCC cells exerted the opposite effect and induced resistance to lenvatinib. Mechanistically, upregulated EVA1A inhibited the PI3K/AKT/MDM2 signaling pathway, resulting in a reduced interaction between MDM2 and p53, thereby stabilizing p53 and enhancing its antitumor activity. In addition, upregulated EVA1A suppressed the PI3K/AKT/mTOR signaling pathway and promoted autophagy, leading to the degradation of mutant p53 and attenuating its oncogenic impact. On the contrary, loss of EVA1A activated the PI3K/AKT/MDM2 signaling pathway and inhibited autophagy, promoting p53 proteasomal degradation and mutant p53 accumulation respectively. These findings establish a crucial role of EVA1A loss in driving lenvatinib resistance involving a mechanism of modulating PI3K/AKT/p53 signaling axis and suggest that upregulating EVA1A is a promising therapeutic strategy for alleviating resistance to lenvatinib, thereby improving the efficacy of HCC treatment.
Collapse
MESH Headings
- Humans
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Quinolines/pharmacology
- Quinolines/therapeutic use
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Phenylurea Compounds/pharmacology
- Phenylurea Compounds/therapeutic use
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/drug effects
- Tumor Suppressor Protein p53/metabolism
- Tumor Suppressor Protein p53/genetics
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphatidylinositol 3-Kinases/genetics
- Signal Transduction/drug effects
- Proto-Oncogene Proteins c-akt/metabolism
- Proto-Oncogene Proteins c-akt/genetics
- Animals
- Cell Line, Tumor
- Mice
- Apoptosis/drug effects
- Cell Proliferation/drug effects
- Mice, Nude
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Gene Expression Regulation, Neoplastic/drug effects
- Male
- Xenograft Model Antitumor Assays
- Mice, Inbred BALB C
- Proto-Oncogene Proteins c-mdm2/metabolism
- Proto-Oncogene Proteins c-mdm2/genetics
- Female
Collapse
Affiliation(s)
- Xiaokun Liu
- School of Basic Medicine, College of Electronic Information, Micro-Nano Technology College, Qingdao University, Qingdao, China
| | - Xiao Gao
- School of Basic Medicine, College of Electronic Information, Micro-Nano Technology College, Qingdao University, Qingdao, China
| | - Yuling Yang
- Department of Infectious Diseases, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Di Yang
- School of Basic Medicine, College of Electronic Information, Micro-Nano Technology College, Qingdao University, Qingdao, China
| | - Qingming Guo
- Clinical Laboratory, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Lianhui Li
- School of Basic Medicine, College of Electronic Information, Micro-Nano Technology College, Qingdao University, Qingdao, China
| | - Shunlong Liu
- Department of Clinical Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Wanxin Cong
- Department of Clinical Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Sen Lu
- Department of Medical Laboratory, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Lin Hou
- School of Basic Medicine, College of Electronic Information, Micro-Nano Technology College, Qingdao University, Qingdao, China
| | - Bin Wang
- School of Basic Medicine, College of Electronic Information, Micro-Nano Technology College, Qingdao University, Qingdao, China
| | - Ning Li
- School of Basic Medicine, College of Electronic Information, Micro-Nano Technology College, Qingdao University, Qingdao, China.
| |
Collapse
|
2
|
Chen Y, Wang D, Yin J, Krafft PR, Luo X, Hao D, Li C, Liu Y, Li L, Zhang Y, Zhu C. Increased TMEM166 Level in Patients with Postoperative Stroke after Carotid Endarterectomy. Neuroscience 2024; 549:138-144. [PMID: 38734302 DOI: 10.1016/j.neuroscience.2024.05.007] [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: 10/24/2023] [Revised: 04/21/2024] [Accepted: 05/05/2024] [Indexed: 05/13/2024]
Abstract
Postoperative stroke is a challenging and potentially devastating complication after elective carotid endarterectomy (CEA). We previously demonstrated that transmembrane protein 166 (TMEM166) levels were directly related to neuronal damage after cerebral ischemia-reperfusion injury in rats. In this subsequent clinical study, we aimed to evaluate the prognostic value of TMEM166 in patients suffering from post-CEA strokes. Thirty-five patients undergoing uncomplicated elective CEA and 8 patients who suffered ischemic strokes after CEA were recruited. We evaluated the protein level and expression of TMEM166 in patients diagnosed with postoperative strokes and compared it to those in patients who underwent uncomplicated elective CEA. Blood samples and carotid artery plaques were collected and analyzed. High expressions of TMEM166 were detected by immunofluorescence staining and Western Blot in carotid artery plaques of all patients who underwent CEA. Furthermore, circulating TMEM166 concentrations were statistically higher in post-CEA stroke patients than in patients allocated to the control group. Mean plasma concentrations of inflammatory markers, including interleukin 6 (IL-6) and C-reactive protein (CRP), were also elevated in patients with postoperative strokes. Therefore, based on these findings, we hypothesize that elevated TMEM166 levels, accompanied by a strong inflammatory response, serve as a useful biomarker for risk assessment of postoperative stroke following CEA.
Collapse
Affiliation(s)
- Yifei Chen
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Danyang Wang
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jie Yin
- Department of Anesthesiology, the First Affiliated Hospital of Xinxiang Medical University, Henan, China
| | - Paul R Krafft
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Xin Luo
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Dandan Hao
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Chengwen Li
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Ye Liu
- Department of Anaesthesiology, Beijing Obstetrics and Gynecology Hospital,Capital Medical University, Beijing, China.
| | - Li Li
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Yongbo Zhang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Chen Zhu
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
3
|
Xiang QM, Jiang N, Liu YF, Wang YB, Mu DA, Liu R, Sun LY, Zhang W, Guo Q, Li K. Overexpression of SH2D1A promotes cancer progression and is associated with immune cell infiltration in hepatocellular carcinoma via bioinformatics and in vitro study. BMC Cancer 2023; 23:1005. [PMID: 37858067 PMCID: PMC10585762 DOI: 10.1186/s12885-023-11315-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 08/18/2023] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND SH2 domain containing 1A (SH2D1A) expression has been linked to cancer progression. However, the functions of SH2D1A in hepatocellular carcinoma (HCC) have not been reported. METHODS The effects of SH2D1A on the proliferation, migration, and invasion of HCC cells and the related pathways were re-explored in cell models with SH2D1A overexpression using the CCK-8, migration and invasion assays and western blotting. The functions and mechanisms of genes co-expressed with SH2D1A were analyzed using gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. The relationship between SH2D1A expression and immune microenvironment features in HCC was explored. RESULTS Elevated SH2D1A expression promoted cell proliferation, migration, and invasion, which was related to the overexpression of p-Nf-κB and BCL2A1 protein levels in HCC. SH2D1A expression was related to the immune, stromal, and ESTIMATE scores, and the abundance of immune cells, such as B cells, CD8+ T cells, and T cells. SH2D1A expression was significantly related to the expression of immune cell markers, such as PDCD1, CD8A, and CTLA4 in HCC. CONCLUSION SH2D1A overexpression was found to promote cell growth and metastasis via the Nf-κB signaling pathway and may be related to the immune microenvironment in HCC. The findings indicate that SH2D1A can function as a biomarker in HCC.
Collapse
Affiliation(s)
- Qian-Ming Xiang
- Department of Cardiothoracic surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, China
- Department of General Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ni Jiang
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yue-Feng Liu
- Department of Ophthalmology surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Yuan-Biao Wang
- Department of Yunnan Tumor Research Institute, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - De-An Mu
- Department of Hepatobiliary and Pancreatic Surgery, The People's Hospital of Jianyang city, Jianyang, China
| | - Rong Liu
- Department of General Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lu-Yun Sun
- Department of General Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Wei Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The People's Hospital of Jianyang city, Jianyang, China.
| | - Qiang Guo
- Department of Cardiothoracic surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, China.
| | - Kai Li
- Department of Hepatobiliary and Pancreatic Surgery, The People's Hospital of Jianyang city, Jianyang, China.
| |
Collapse
|
4
|
Hao Y, Song T, Wang M, Li T, Zhao C, Li T, Hou Y, He H. Dual targets of lethal apoptosis and protective autophagy in liver cancer with periplocymarin elicit a limited therapeutic effect. Int J Oncol 2023; 62:44. [PMID: 36825592 PMCID: PMC9946806 DOI: 10.3892/ijo.2023.5492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 01/27/2023] [Indexed: 02/25/2023] Open
Abstract
Cardiac glycosides (CGs) are candidate anticancer agents that function by increasing [Ca2+]i to induce apoptotic cell death in several types of cancer cells. However, new findings have shown that the anti‑cancer effects of CGs involve complex cell‑signal transduction mechanisms. Hence, exploring the potential mechanisms of action of CGs may provide insight into their anti‑cancer effects and thus aid in the selection of the appropriate CG. Periplocymarin (PPM), which is a cardiac glycoside, is an active ingredient extracted from Cortex periplocae. The role of PPM was evaluated in HepG2 cells and xenografted nude mice. Cell proliferation, real‑time ATP rate assays, western blotting, cell apoptosis assays, short interfering RNA transfection, the patch clamp technique, electron microscopy, JC‑1 staining, immunofluorescence staining and autophagic flux assays were performed to evaluate the function and regulatory mechanisms of PPM in vitro. The in vivo activity of the PPM was assessed using a mouse xenograft model. The present study demonstrated that PPM synchronously activated lethal apoptosis and protective autophagy in liver cancer, and the initiation of autophagy counteracted the inherent pro‑apoptotic capacity and impaired the anti‑cancer effects. Specifically, PPM exerted a pro‑-apoptotic effect in HepG2 cells and activated macroautophagy by initiation of the AMPK/ULK1 and mTOR signaling pathways. Activation of macroautophagy counteracted the pro‑apoptotic effects of PPM, but when it was combined with an autophagy inhibitor, the anti‑cancer effects of PPM in mice bearing HepG2 xenografts were observed. Collectively, these results indicated that a self‑limiting effect impaired the pro‑apoptotic effects of PPM in liver cancer, but when combined with an autophagy inhibitor, it may serve as a novel therapeutic option for the management of liver cancer.
Collapse
Affiliation(s)
- Yuanyuan Hao
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China,Hebei Yiling Chinese Medicine Research Institute, Shijiazhuang, Hebei 050035, P.R. China,New Drug Evaluation Center, Shijiazhuang Yiling Pharmaceutical Co., Ltd, Shijiazhuang, Hebei 050035, P.R. China
| | - Tao Song
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China,Hebei Yiling Chinese Medicine Research Institute, Shijiazhuang, Hebei 050035, P.R. China,New Drug Evaluation Center, Shijiazhuang Yiling Pharmaceutical Co., Ltd, Shijiazhuang, Hebei 050035, P.R. China
| | - Mingye Wang
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Tongtong Li
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Chi Zhao
- Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Ting Li
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Yunlong Hou
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China,Hebei Yiling Chinese Medicine Research Institute, Shijiazhuang, Hebei 050035, P.R. China,New Drug Evaluation Center, Shijiazhuang Yiling Pharmaceutical Co., Ltd, Shijiazhuang, Hebei 050035, P.R. China,Correspondence to: Professor Yunlong Hou, College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, 3 Xingyuan Road, Shijiazhuang, Hebei 050200, P.R. China, E-mail:
| | - Hongjiang He
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China,Professor Hongjiang He, Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang 150081, P.R. China, E-mail:
| |
Collapse
|
5
|
The Degradation of TMEM166 by Autophagy Promotes AMPK Activation to Protect SH-SY5Y Cells Exposed to MPP+. Cells 2022; 11:cells11172706. [PMID: 36078115 PMCID: PMC9454683 DOI: 10.3390/cells11172706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 08/22/2022] [Accepted: 08/25/2022] [Indexed: 11/19/2022] Open
Abstract
Neuronal oxidative stress caused by mitochondrial dysfunction plays a crucial role in the development of Parkinson’s disease (PD). Growing evidence shows that autophagy confers neuroprotection in oxidative-stress-associated PD. This work aims to investigate the involvement of TMEM166, an endoplasmic-reticulum-localized autophagy-regulating protein, in the process of PD-associated oxidative stress through the classic cellular PD model of neuroblastoma SH-SY5Y cells exposed to 1-methyl-4-phenylpyridinium (MPP+). Reactive oxygen species (ROS) production and mitochondrial membrane potential were checked to assess the oxidative stress induced by MPP+ and the cellular ATP generated was determined to evaluate mitochondrial function. The effect on autophagy induction was evaluated by analyzing p62 and LC3-II/I expression and by observing the LC3 puncta and the colocalization of LC3 with LAMP1/ LAMP2. The colocalization of mitochondria with LC3, the colocalization of Tom20 with LAMP1 and Tom20 expression were analyzed to evaluate mitophagy. We found that TMEM166 is up-regulated in transcript levels, but up-regulated first and then down-regulated by autophagic degradation in protein levels upon MPP+-treatment. Overexpression of TMEM166 induces mitochondria fragmentation and dysfunction and exacerbates MPP+-induced oxidative stress and cell viability reduction. Overexpression of TMEM166 is sufficient to induce autophagy and mitophagy and promotes autophagy and mitophagy under MPP+ treatment, while knockdown of TMEM166 inhibits basal autophagic degradation. In addition, overexpressed TMEM166 suppresses AMPK activation, while TMEM166 knockdown enhances AMPK activation. Pharmacological activation of AMPK alleviates the exacerbation of oxidative stress induced by TMEM166 overexpression and increases cell viability, while pharmacological inhibition mitophagy aggravates the oxidative stress induced by MPP+ treatment combined with TMEM166 overexpression. Finally, we find that overexpressed TMEM166 partially localizes to mitochondria and, simultaneously, the active AMPK in mitochondria is decreased. Collectively, these findings suggest that TMEM166 can translocate from ER to mitochondria and inhibit AMPK activation and, in response to mitochondrial oxidative stress, neuronal cells choose to up-regulate TMEM166 to promote autophagy/mitophagy; then, the enhancing autophagy/mitophagy degrades the TMEM166 to activate AMPK, by the two means to maintain cell survival. The continuous synthesis and degradation of TMEM166 in autophagy/mitochondria flux suggest that TMEM166 may act as an autophagy/mitochondria adaptor.
Collapse
|
6
|
The Emerging Role of EVA1A in Different Types of Cancers. Int J Mol Sci 2022; 23:ijms23126665. [PMID: 35743108 PMCID: PMC9224241 DOI: 10.3390/ijms23126665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 06/04/2022] [Accepted: 06/14/2022] [Indexed: 12/06/2022] Open
Abstract
Eva-1 homolog A (EVA1A), also known as transmembrane protein 166 (TMEM166) and regulator of programmed cell death, is an endoplasmic reticulum associated protein, which can play an important role in many diseases, including a variety of cancers, by regulating autophagy/apoptosis. However, the related mechanism, especially the role of EVA1A in cancers, has not been fully understood. In this review, we summarize the recent studies on the role of EVA1A in different types of cancers, including breast cancer, papillary thyroid cancer, non-small cell lung cancer, hepatocellular carcinoma, glioblastoma and pancreatic cancer, and analyze the relevant mechanisms to provide a theoretical basis for future related research.
Collapse
|
7
|
Flubendazole induces mitochondrial dysfunction and DRP1-mediated mitophagy by targeting EVA1A in breast cancer. Cell Death Dis 2022; 13:375. [PMID: 35440104 PMCID: PMC9019038 DOI: 10.1038/s41419-022-04823-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 03/23/2022] [Accepted: 04/04/2022] [Indexed: 02/06/2023]
Abstract
Breast cancer is still one of the most common malignancies worldwide and remains a major clinical challenge. We previously reported that the anthelmintic drug flubendazole induced autophagy and apoptosis via upregulation of eva-1 homolog A (EVA1A) in triple-negative breast cancer (TNBC) and was repurposed as a novel anti-tumor agent. However, the detailed underlying mechanisms remain unclear and need further investigation. Here, we found that flubendazole impairs the permeability of the mitochondrial outer membrane and mitochondrial function in breast cancer. Meanwhile, flubendazole increased dynamin-related protein (DRP1) expression, leading to the accumulation of PTEN induced putative kinase 1 (PINK1) and subsequent mitochondrial translocation of Parkin, thereby promoting excessive mitophagy. The resultant excessive mitophagy contributed to mitochondrial damage and dysfunction induced by flubendazole, thus inhibiting breast cancer cells proliferation and migration. Moreover, we demonstrated that excessive DRP1-mediated mitophagy played a critical role in response to the anti-tumor effects of EVA1A in breast cancer. Taken together, our results provide new insights into the molecular mechanisms in relation to the anti-tumor activities of flubendazole, and may be conducive to its rational use in potential clinical applications.
Collapse
|
8
|
Li Y, Miller CA, Shea LK, Jiang X, Guzman MA, Chandler RJ, Ramakrishnan SM, Smith SN, Venditti CP, Vogler CA, Ory DS, Ley TJ, Sands MS. Enhanced Efficacy and Increased Long-Term Toxicity of CNS-Directed, AAV-Based Combination Therapy for Krabbe Disease. Mol Ther 2021; 29:691-701. [PMID: 33388420 PMCID: PMC7854295 DOI: 10.1016/j.ymthe.2020.12.031] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/25/2020] [Accepted: 12/22/2020] [Indexed: 12/13/2022] Open
Abstract
Infantile globoid cell leukodystrophy (GLD, Krabbe disease) is a demyelinating disease caused by the deficiency of the lysosomal enzyme galactosylceramidase (GALC) and the progressive accumulation of the toxic metabolite psychosine. We showed previously that central nervous system (CNS)-directed, adeno-associated virus (AAV)2/5-mediated gene therapy synergized with bone marrow transplantation and substrate reduction therapy (SRT) to greatly increase therapeutic efficacy in the murine model of Krabbe disease (Twitcher). However, motor deficits remained largely refractory to treatment. In the current study, we replaced AAV2/5 with an AAV2/9 vector. This single change significantly improved several endpoints primarily associated with motor function. However, nearly all (14/16) of the combination-treated Twitcher mice and all (19/19) of the combination-treated wild-type mice developed hepatocellular carcinoma (HCC). 10 out of 10 tumors analyzed had AAV integrations within the Rian locus. Several animals had additional integrations within or near genes that regulate cell growth or death, are known or potential tumor suppressors, or are associated with poor prognosis in human HCC. Finally, the substrate reduction drug L-cycloserine significantly decreased the level of the pro-apoptotic ceramide 18:0. These data demonstrate the value of AAV-based combination therapy for Krabbe disease. However, they also suggest that other therapies or co-morbidities must be taken into account before AAV-mediated gene therapy is considered for human therapeutic trials.
Collapse
Affiliation(s)
- Yedda Li
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Christopher A Miller
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Lauren K Shea
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Xuntian Jiang
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Miguel A Guzman
- Department of Pathology, St. Louis University School of Medicine, St. Louis, MO, USA
| | - Randy J Chandler
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Sai M Ramakrishnan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Stephanie N Smith
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Charles P Venditti
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Carole A Vogler
- Department of Pathology, St. Louis University School of Medicine, St. Louis, MO, USA
| | - Daniel S Ory
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Timothy J Ley
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA; Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Mark S Sands
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA; Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
9
|
Ma X, Liu L. Knockdown of FAM225B inhibits the progression of the hypertrophic scar following glaucoma surgery by inhibiting autophagy. Mol Med Rep 2021; 23:204. [PMID: 33495826 PMCID: PMC7821338 DOI: 10.3892/mmr.2021.11843] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 12/18/2020] [Indexed: 12/16/2022] Open
Abstract
The formation of a hypertrophic scar (HS) may lead to failure of glaucoma surgery. Long non-coding RNAs (lncRNAs) are involved in the formation of HSs. Moreover, family with sequence similarity 225 member B (FAM225B) is upregulated in HS. However, the role of the lncRNA FAM225B in HS remains unknown. Thus, the present study aimed to investigate the function of FAM225B in HS. Scar fibroblasts were isolated from patients who had undergone glaucoma surgery. Western blotting was used to detect the expressions of Bax, Bcl-2, cleaved caspase 3, p62, ATG7 and Beclin 1, and reverse transcription-quantitative PCR (RT-qPCR) were conducted to determine the level of FAM225B in scar fibroblasts. Microtubule associated protein 1 light chain 3 α staining was performed to examine autophagosomes in scar fibroblasts. Furthermore, cell proliferation was evaluated via 5-ethynyl-2′-deoxyuridine staining. Flow cytometry was conducted to determine cell apoptosis and the levels of reactive oxygen species (ROS) in scar fibroblasts. The cell migratory ability was assessed using a Transwell assay. The results demonstrated that FAM225B knockdown significantly attenuated scar fibroblast proliferation and induced apoptosis. Additionally, transfection of scar fibroblasts with FAM225B small interfering RNA (siRNA) significantly increased the ROS levels and significantly decreased the migration of scar fibroblasts. The FAM225B overexpression-induced increase of scar fibroblast proliferation and migration was significantly reversed by 3-methyladenine administration. The results suggested that knockdown of FAM225B significantly inhibited the proliferation of scar fibroblasts by inhibiting autophagy. Therefore, knockdown of FAM225B could inhibit scar fibroblast proliferation after glaucoma surgery by inhibiting autophagy. These findings may provide a novel perspective of developing treatment strategy for the patients with HSs after glaucoma surgery.
Collapse
Affiliation(s)
- Xianpeng Ma
- Department of Dermatology, Affiliated Hospital of Beihua University, Jilin, Jilin 132001, P.R. China
| | - Lili Liu
- Department of Dermatology, Affiliated Hospital of Beihua University, Jilin, Jilin 132001, P.R. China
| |
Collapse
|
10
|
Yuan H, Xu Y, Luo Y, Wang NX, Xiao JH. Role of Nrf2 in cell senescence regulation. Mol Cell Biochem 2021; 476:247-259. [PMID: 32918185 DOI: 10.1007/s11010-020-03901-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/02/2020] [Indexed: 02/06/2023]
Abstract
Nuclear factor-E2-related factor 2 (Nrf2) is a key transcription factor known to be involved in maintaining cell redox balance and signal transduction and plays central role in reducing intracellular oxidative stress damage, delaying cell senescence and preventing age-related diseases. However, it has been shown that the level of Nrf2 decreases with age and that the silencing of the Nrf2 gene is associated with the induction of premature senescence. Therefore, a plethora of researchers have focused on elucidating the regulatory mechanism of Nrf2 in the prevention of cell senescence. This complex regulatory mechanism of Nrf2 in the cell senescence process involves coordinated regulation of multiple signaling molecules. After summarizing the function of Nrf2 and its relationship with cell senescence pathway, this review focuses on the recent advances and progress made in elucidating the regulatory mechanism of Nrf2 in the cell senescence process. Additionally, the information collected here may provide insights for further research on Nrf2, in particular, on its regulatory mechanism in the cell senescence process.
Collapse
Affiliation(s)
- Huan Yuan
- Zunyi Municiptal Key Laboratory of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, People's Republic of China
| | - Yan Xu
- Zunyi Municiptal Key Laboratory of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, People's Republic of China
- Guizhou Provincial Research Center for Translational Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, People's Republic of China
| | - Yi Luo
- Zunyi Municiptal Key Laboratory of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, People's Republic of China
- Guizhou Provincial Research Center for Translational Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, People's Republic of China
| | - Nuo-Xin Wang
- Zunyi Municiptal Key Laboratory of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, People's Republic of China
- Guizhou Provincial Research Center for Translational Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, People's Republic of China
| | - Jian-Hui Xiao
- Zunyi Municiptal Key Laboratory of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, People's Republic of China.
- Guizhou Provincial Research Center for Translational Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, People's Republic of China.
- Zunyi Municiptal Key Laboratory of Medicinal Biotechnology, Center for Translational Medicine, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Zunyi, 563003, People's Republic of China.
| |
Collapse
|