1
|
Yao Z, Li J, Yu J, Cheng Y, Fang C, Chen X, Chen X, Wang Y, Gao D, Lin F. Glycometabolic Regulation of Angiogenesis: Mechanisms and Therapeutic Strategies. Int J Mol Sci 2025; 26:2386. [PMID: 40141029 PMCID: PMC11942008 DOI: 10.3390/ijms26062386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/02/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
Angiogenesis, the process by which new blood vessels emerge from pre-existing vasculature, forms the fundamental biological basis for therapeutic angiogenesis. In recent years, this field has garnered significant attention, particularly in the context of understanding the mechanisms of angiogenesis through the lens of glycometabolism. The potential clinical applications of this research have been widely acknowledged within the medical community. In this article, the role of angiogenesis and the principal molecular mechanisms that govern it are first delineated. The influence of glycometabolism on angiogenesis is then explored, with a focus on glycolysis. Finally, research on therapeutic angiogenesis based on the regulation of glycometabolism is presented, offering novel perspectives for ongoing research and clinical applications.
Collapse
Affiliation(s)
- Zhifeng Yao
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (Z.Y.); (J.L.); (J.Y.)
| | - Junting Li
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (Z.Y.); (J.L.); (J.Y.)
| | - Jiaming Yu
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (Z.Y.); (J.L.); (J.Y.)
| | - Ye Cheng
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (Z.Y.); (J.L.); (J.Y.)
| | - Chang Fang
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (Z.Y.); (J.L.); (J.Y.)
| | - Xinlei Chen
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (Z.Y.); (J.L.); (J.Y.)
| | - Xiaoqi Chen
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (Z.Y.); (J.L.); (J.Y.)
| | - Yizheng Wang
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (Z.Y.); (J.L.); (J.Y.)
- Key Laboratory of Integrative Medicine on Chronic Diseases, Fujian Province University, Fuzhou 350122, China
| | - Dong Gao
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (Z.Y.); (J.L.); (J.Y.)
- Key Laboratory of Integrative Medicine on Chronic Diseases, Fujian Province University, Fuzhou 350122, China
| | - Fan Lin
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (Z.Y.); (J.L.); (J.Y.)
- Key Laboratory of Integrative Medicine on Chronic Diseases, Fujian Province University, Fuzhou 350122, China
| |
Collapse
|
2
|
Luo G, Hu W, Yang J, Ding H, Xu C, Tong X, Ding C, Zhao J. Identification of G protein subunit alpha i3 as a promising oncotarget of LUAD. Cell Signal 2025; 127:111582. [PMID: 39733926 DOI: 10.1016/j.cellsig.2024.111582] [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: 10/29/2024] [Revised: 12/10/2024] [Accepted: 12/24/2024] [Indexed: 12/31/2024]
Abstract
Exploring new oncotargets essential for lung adenocarcinoma (LUAD) cell growth is important. Here the bioinformatical studies revealed that Gαi3 expression is elevated in LUAD tissues and its overexpression correlates with poor survival of the patients. Moreover, overexpression of Gαi3 mRNA and protein was detected in LUAD tissues of patients as well as in primary/immortalized LUAD cells. In both primary and immortalized LUAD cells, genetic silencing (by viral shRNA) or knockout ("KO", through CRISPR/Cas9 method) of Gαi3 potently inhibited LUAD cell proliferation and mobility. The results of caspase-3 activity assay, caspase-9 activity assay, histone DNA ELISA, TUNEL nuclear staining and Annexin V staining showed that inhibition of Gαi3 expression promoted apoptosis. In addition, a significant decrease in mitochondrial membrane potential was found in Gαi3-deficient LUAD cells by JC-1 staining. Overexpression of Gαi3 strengthened the proliferation and migration of LUAD cell. Gene set enrichment analysis revealed that Gαi3 was closely related to PI3k/Akt/mTOR, which we validated experimentally. Akt-S6K phosphorylation was downregulated following Gαi3 silencing or KO, but augmented after Gαi3 overexpression in primary LUAD cells. Restoring Akt-S6K phosphorylation by a S473D constitutively-active mutant Akt1 ameliorated Gαi3 KO-induced LUAD cell proliferation inhibition, migration suppression and apoptosis. In vivo, the growth of subcutaneous LUAD xenografts was largely inhibited after intratumoral injection of Gαi3 shRNA-expressing adeno-associated virus (AAV). Gαi3 downregulation, Akt-mTOR inhibition, proliferation inactivation and apoptosis were detected in the Gαi3 shRNA-treated LUAD xenografts. Together, targeting Gαi3 potently inhibited LUAD cell growth in vitro and in vivo.
Collapse
Affiliation(s)
- Gaomeng Luo
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China; Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wenxuan Hu
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China; Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jian Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China; Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hao Ding
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China; Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chun Xu
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China; Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xin Tong
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China; Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Cheng Ding
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China; Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Jun Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China; Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
3
|
Zhu XR, Zhu JQ, Gu QH, Liu N, Lu JJ, Li XH, Liu YY, Zheng X, Chen MB, Ji Y. A novel identified epithelial ligand-receptor-associated gene signature highlights POPDC3 as a potential therapy target for non-small cell lung cancer. Cell Death Dis 2025; 16:114. [PMID: 39971925 PMCID: PMC11840029 DOI: 10.1038/s41419-025-07410-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 01/13/2025] [Accepted: 01/30/2025] [Indexed: 02/21/2025]
Abstract
The tumor microenvironment (TME) is pivotal in non-small cell lung cancer (NSCLC) progression, influencing drug resistance and immune cell behavior through complex ligand-receptor (LR) interactions. This study developed an epithelial LR-related prognostic risk score (LRrisk) to identify biomarkers and targets in NSCLC. We identified twenty epithelial LRs with significant prognostic implications and delineated three molecular NSCLC subtypes with distinct outcomes, pathological characteristics, biological pathways, and immune profiles. The LRrisk model was constructed using twelve differentially expressed ligand-receptor interaction-related genes (LRGs), with a focus on POPDC3 (popeye domain-containing protein 3), which was overexpressed in NSCLC cells. Functional assays revealed that POPDC3 knockdown reduced cell proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT), while its overexpression promoted cancerous activities. In vivo, POPDC3 silencing hindered, and its overexpression accelerated the growth of NSCLC xenografts in nude mice. Additionally, high expression levels of POPDC3 in NSCLC tissues were associated with enhanced CD4+ T cell infiltration and increased PD-1 expression within the TME. Moreover, ectopic POPDC3 overexpression in C57BL/6 J mouse Lewis lung carcinoma (LLC) xenografts enhanced CD4+ T cell infiltration and PD-1 expression in the TME. This research establishes a robust epithelial LR-related signature, highlighting POPDC3 as a critical facilitator of NSCLC progression and a potential therapeutic target.
Collapse
Affiliation(s)
- Xiao-Ren Zhu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
- Clinical Research and Lab Center, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
- Medical School of Jiangsu University, Zhenjiang, China
| | - Jia-Qi Zhu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Qian-Hui Gu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Na Liu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
- Medical School of Jiangsu University, Zhenjiang, China
| | - Jing-Jing Lu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
- Medical School of Jiangsu University, Zhenjiang, China
| | - Xiao-Hong Li
- Department of Clinical Laboratory, The First People's Hospital of Taicang, Taicang, China
| | - Yuan-Yuan Liu
- Clinical Research and Lab Center, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
- Medical School of Jiangsu University, Zhenjiang, China
| | - Xian Zheng
- Medical School of Jiangsu University, Zhenjiang, China.
- Department of Pharmacy, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China.
| | - Min-Bin Chen
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China.
- Medical School of Jiangsu University, Zhenjiang, China.
| | - Yong Ji
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China.
| |
Collapse
|
4
|
Xie H, Ji J, Liu Z, Lu N, Wei Y, Zhou A, Liu J, Jiao Q. Gαi1/3 signaling mediates IL-5-induced eosinophil activation and type 2 inflammation in eosinophilic chronic rhinosinusitis. Front Immunol 2025; 15:1460104. [PMID: 39840047 PMCID: PMC11746084 DOI: 10.3389/fimmu.2024.1460104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 12/12/2024] [Indexed: 01/30/2025] Open
Abstract
Background Uncontrolled severe eosinophilic chronic rhinosinusitis (eCRS) is associated with elevated levels of Th2 cells and raised immunoglobulin concentrations in nasal polyp tissue. eCRS is characterized by high eosinophilic infiltration and type 2 inflammation. Gαi1/3 proteins participate in allergic inflammation by regulating immune cells. Whether Gαi1/3 proteins have a role in the development of eCRS remains unknown. Objectives To investigate the association between Gαi1/3 expression levels and CRS and the underlying mechanisms. Methods Western blotting and immunohistology were used to detect Gαi1/3 expression. Correlations between Gαi1/3 and immune cells and clinical parameters were analyzed. Signaling pathway activation in IL-5-induced Gαi1/3-knockout or knockdown mouse embryonic fibroblasts (MEFs) and eosinophils (EoL-1 cells) was detected by western blotting. EdU/DAPI was used to evaluate the proliferation of EoL-1 cells. A CRS model was established using Gαi1/3-knockout mice, and histological analysis and inflammatory cytokine measurements were performed. Results Compared with the non-eCRS subset, the eCRS subset showed significantly increased Gαi1/3 expression levels. High nasal tissue Gαi1/3 levels were linked to high tissue eosinophil infiltration, and high disease severity and allergic conditions in CRS patients. Gαi1/3 were required for IL-5-induced Akt-mTOR and Erk activation in MEFs. In EoL-1 cells, Gαi1/3 was associated with IL-5-activated IL-5Rα, promoting IL-5Rα endocytosis and transducing downstream signaling. IL-5-induced EoL-1 cell proliferation and degranulation were suppressed after Gαi1/3 silencing. In a CRS murine model, immune cell infiltration and type 2 inflammation were largely impaired in Gαi1/3-double-knockout mice. Conclusion Increased Gαi1/3 expression levels in nasal tissue are linked to eosinophil infiltration and increased disease severity in CRS patients. Gαi1/3 contributes to eosinophil activation and participates in regulating allergic inflammation in CRS patients.
Collapse
Affiliation(s)
- Huanxia Xie
- Department of Ear, Nose, and Throat, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiang Ji
- Department of Dermatology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhichen Liu
- Department of Ear, Nose, and Throat, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ning Lu
- Department of Ear, Nose, and Throat, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuqian Wei
- Department of Dermatology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Aina Zhou
- Department of Ear, Nose, and Throat, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jisheng Liu
- Department of Ear, Nose, and Throat, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qingqing Jiao
- Central Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Dermatology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
5
|
Xu W, Cao L, Liu H. CAMK2D and Complement Factor I-Involved Calcium/Calmodulin Signaling Modulates Sodium Iodate-Induced Mouse Retinal Degeneration. Invest Ophthalmol Vis Sci 2025; 66:63. [PMID: 39873650 PMCID: PMC11781327 DOI: 10.1167/iovs.66.1.63] [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: 07/27/2024] [Accepted: 12/06/2024] [Indexed: 01/30/2025] Open
Abstract
Purpose To investigate the effect of Ca2+/calmodulin-dependent protein kinase II (CAMKII) δ subtypes (CAMK2D) on sodium iodate (NaIO3)-induced retinal degeneration in mice. Methods Bioinformatics analysis and Western blot experiments were used to screen the significantly differentially expressed genes in age-related macular degeneration (AMD) disease. CAMK2D knockdown and overexpression models were constructed by lentivirus (LV) infection of adult retinal pigment epithelial cell line-19 (ARPE-19) cells in vitro. Flow cytometry was used to detect ARPE-19 cell apoptosis induced by NaIO3. In vivo, CAMK2D knockdown and overexpression mouse models were generated by infecting mouse retinal pigment epithelium (RPE) with adeno-associated virus (AAV). Retinography, optical coherence tomography (OCT), and histological analysis (hematoxylin and eosin staining) were used to detect NaIO3-induced retinal structural changes in mice. Electroretinography (ERG) was used to detect NaIO3-induced retinal function changes in mice. TdT-mediated dUTP nick-end labeling (TUNEL) staining was used to detect the apoptosis of retinal cells induced by NaIO3. RNA sequencing (RNA-Seq) and bioinformatics analysis were used to screen for target genes affected by CAMK2D in CAMK2D-overexpressing ARPE-19 cells. And flow cytometry, OCT, and ERG were used to evaluate the regulatory effect of CAMK2D on target genes. Results Bioinformatics analysis found the expression of genes related to Ca2+ signal was significantly reduced in AMD patients. Western blot showed that in a mouse model of dry AMD induced by NaIO3, CAMK2D expression in RPE-Choroid tissue significantly lower than normal mice. In vitro, our results showed that overexpression of CAMK2D in ARPE-19 cells decreased apoptosis induced by NaIO3 and knockdown increased apoptosis. In vivo, CAMK2D overexpression in RPE cells can attenuate the retina degeneration induced by NaIO3 and CAMK2D knockdown aggravated degeneration. The bioinformatics analysis indicated that CAMK2D might affect AMD pathology through complement factor I (CFI). In vitro, knockdown of CFI in ARPE-19 cells increased apoptosis induced by NaIO3. In knockdown CFI ARPE-19 cells, overexpression of CAMK2D reduced the above apoptosis. In mice retina, CFI knockdown can aggravate the retina degeneration induced by NaIO3. In knockdown CFI mice, overexpression of CAMK2D in RPE can attenuate the above retina degeneration. Western blot confirmed that CAMK2D regulated the expression of CFI in mice. Conclusions CAMK2D can attenuate the retinal degeneration induced by NaIO3, which was achieved by regulating the CFI.
Collapse
Affiliation(s)
- Weixing Xu
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou City, China
- School of Graduate, Dalian Medical University, Dalian City, China
- Key Laboratory of Age-related Macular Degeneration of Liaoning Province, Jinzhou Medical University, Jinzhou City, China
| | - Liu Cao
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang City, China
| | - Hua Liu
- School of Graduate, Dalian Medical University, Dalian City, China
- Key Laboratory of Age-related Macular Degeneration of Liaoning Province, Jinzhou Medical University, Jinzhou City, China
| |
Collapse
|
6
|
Liu N, Zhu XR, Wu CY, Liu YY, Chen MB, Gu JH. PCK1 as a target for cancer therapy: from metabolic reprogramming to immune microenvironment remodeling. Cell Death Discov 2024; 10:478. [PMID: 39578429 PMCID: PMC11584723 DOI: 10.1038/s41420-024-02240-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/24/2024] Open
Abstract
Recently, changes in metabolites and metabolism-related enzymes related to tumor cell proliferation, metastasis, drug resistance, and immunosuppression have become a research hotspot, and researchers have attempted to determine the clinical correlation between specific molecular lesions and metabolic phenotypes. Convincing evidence shows that metabolic reprogramming is closely related to the proliferation, invasion, metastasis, and poor prognosis of malignant tumors. Therefore, targeting metabolic reprogramming is a new direction for cancer treatment. However, how molecular alterations in tumors contribute to metabolic diversity and unique targeting dependencies remains unclear. A full understanding of the underlying mechanisms of metabolic reprogramming in cancer may lead to better identification of therapeutic targets and the development of therapeutic strategies. Evidence for the importance of PCK1, a phosphoenolpyruvate carboxykinase 1, in tumorigenesis and development is accumulating. PCK1 can regulate cell proliferation and metastasis by remodeling cell metabolism. Additionally, PCK1 has "nonclassical" nonmetabolic functions, involving the regulation of gene expression, angiogenesis, epigenetic modification, and other processes, and has an impact on cell survival, apoptosis, and other biological activities, as well as the remodeling of the tumor immune microenvironment. Herein, we provide a comprehensive overview of the functions of PCK1 under physiological and pathological conditions and suggest that PCK1 is a potential target for cancer therapy. We also propose a future exploration direction for targeting PCK1 for cancer therapy from a clinical perspective. Finally, in view of the collective data, the results of our discussion suggest the potential clinical application of targeted PCK1 therapy in combination with chemotherapy and immunotherapy for cancer treatment.
Collapse
Affiliation(s)
- Na Liu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China.
| | - Xiao-Ren Zhu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Chang-Ying Wu
- Department of Intensive Care Medicine, Chongqing People's Hospital, Chongqing, China
| | - Yuan-Yuan Liu
- Clinical Research and Lab Center, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Min-Bin Chen
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China.
| | - Jin-Hua Gu
- Department of Clinical Laboratory, Kunshan First People's Hospital, Affiliated to Jiangsu University Kunshan, Kunshan, China.
| |
Collapse
|
7
|
Jiang JZ, Qiao YB, Zhu XR, Gu QH, Lu JJ, Ye ZY, Xu L, Liu YY. Identification of Gαi3 as a promising molecular oncotarget of pancreatic cancer. Cell Death Dis 2024; 15:699. [PMID: 39349432 PMCID: PMC11442978 DOI: 10.1038/s41419-024-07079-6] [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: 06/06/2024] [Revised: 09/08/2024] [Accepted: 09/13/2024] [Indexed: 10/02/2024]
Abstract
The increasing mortality rate of pancreatic cancer globally necessitates the urgent identification for novel therapeutic targets. This study investigated the expression, functions, and mechanistic insight of G protein inhibitory subunit 3 (Gαi3) in pancreatic cancer. Bioinformatics analyses reveal that Gαi3 is overexpressed in human pancreatic cancer, correlating with poor prognosis, higher tumor grade, and advanced classification. Elevated Gαi3 levels are also confirmed in human pancreatic cancer tissues and primary/immortalized cancer cells. Gαi3 shRNA or knockout (KO) significantly reduced cell viability, proliferation, cell cycle progression, and mobility in primary/immortalized pancreatic cancer cells. Conversely, Gαi3 overexpression enhanced pancreatic cancer cell growth. RNA-sequencing and bioinformatics analyses of Gαi3-depleted cells indicated Gαi3's role in modulating the Akt-mTOR and PKA-Hippo-YAP pathways. Akt-S6 phosphorylation was decreased in Gαi3-depleted cells, but was increased with Gαi3 overexpression. Additionally, Gαi3 depletion elevated PKA activity and activated the Hippo pathway kinase LATS1/2, leading to YAP/TAZ inactivation, while Gαi3 overexpression exerted the opposite effects. There is an increased binding between Gαi3 promoter and the transcription factor TCF7L2 in pancreatic cancer tissues and cells. Gαi3 expression was significantly decreased following TCF7L2 silencing, but increased with TCF7L2 overexpression. In vivo, intratumoral injection of Gαi3 shRNA-expressing adeno-associated virus significantly inhibited subcutaneous pancreatic cancer xenografts growth in nude mice. A significant growth reduction was also observed in xenografts from Gαi3 knockout pancreatic cancer cells. Akt-mTOR inactivation and increased PKA activity coupled with YAP/TAZ inactivation were also detected in xenograft tumors upon Gαi3 depletion. Furthermore, bioinformatic analysis and multiplex immunohistochemistry (mIHC) staining on pancreatic cancer tissue microarrays showed a reduced proportion of M1-type macrophages and an increase in PD-L1 positive cells in Gαi3-high pancreatic cancer tissues. Collectively, these findings highlight Gαi3's critical role in promoting pancreatic cancer cell growth, potentially through the modulation of the Akt-mTOR and PKA-Hippo-YAP pathways and its influence on the immune landscape.
Collapse
Affiliation(s)
- Jian-Zhuo Jiang
- Clinical Research and Lab Center, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Yin-Biao Qiao
- General Surgery, Cancer Center, Department of Colorectal Surgery, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Xiao-Ren Zhu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Qian-Hui Gu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Jing-Jing Lu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Zhen-Yu Ye
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Lu Xu
- Department of general surgery, The first affiliated hospital of Soochow university, Suzhou, China.
| | - Yuan-Yuan Liu
- Clinical Research and Lab Center, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China.
| |
Collapse
|
8
|
Zhang L, Chen Y, Pan Q, Fang S, Zhang Z, Wang J, Yang Y, Yang D, Sun X. Silencing of PCK1 mitigates the proliferation and migration of vascular smooth muscle cells and vascular intimal hyperplasia by suppressing STAT3/DRP1-mediated mitochondrial fission. Acta Biochim Biophys Sin (Shanghai) 2024; 57:633-645. [PMID: 39262325 PMCID: PMC12040600 DOI: 10.3724/abbs.2024154] [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: 05/20/2024] [Accepted: 07/30/2024] [Indexed: 09/13/2024] Open
Abstract
The pathological proliferation and migration of vascular smooth muscle cells (VSMCs) are key processes during vascular neointimal hyperplasia (NIH) and restenosis. Phosphoenolpyruvate carboxy kinase 1 (PCK1) is closely related to a variety of malignant proliferative diseases. However, the role of PCK1 in VSMCs has rarely been investigated. This study aims to examine the role of PCK1 in the proliferation and migration of VSMCs and vascular NIH after injury. In vivo, extensive NIH and increased expression of PCK1 within the neointima are observed in injured arteries. Interestingly, the administration of adeno-associated virus-9 (AAV-9) carrying Pck1 short hairpin RNA (sh Pck1) significantly attenuates NIH and stenosis of the vascular lumen. In vitro, Pck1 small interfering RNA (si Pck1)-induced PCK1 silencing inhibits VSMC proliferation and migration. Additionally, silencing of PCK1 leads to reduced expression of dynamin-related protein 1 (DRP1) and attenuated mitochondrial fission. Lentivirus-mediated DRP1 overexpression markedly reverses the inhibitory effects of PCK1 silencing on VSMC proliferation, migration, and mitochondrial fission. Finally, PCK1 inhibition attenuates the phosphorylation of signal transducer and activator of transcription 3 (STAT3). Activation of STAT3 abolishes the suppressive effects of PCK1 silencing on DRP1 expression, mitochondrial fission, proliferation, and migration in VSMCs. In conclusion, PCK1 inhibition attenuates the mitochondrial fission, proliferation, and migration of VSMCs by inhibiting the STAT3/DRP1 axis, thereby suppressing vascular NIH and restenosis.
Collapse
MESH Headings
- STAT3 Transcription Factor/metabolism
- STAT3 Transcription Factor/genetics
- Cell Proliferation/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/cytology
- Animals
- Cell Movement/genetics
- Mitochondrial Dynamics/genetics
- Dynamins/metabolism
- Dynamins/genetics
- Hyperplasia/metabolism
- Hyperplasia/genetics
- Hyperplasia/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Male
- Neointima/pathology
- Neointima/genetics
- Neointima/metabolism
- Gene Silencing
- Rats
- Rats, Sprague-Dawley
- Cells, Cultured
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Li Zhang
- Department of Cardiologythe General Hospital of Western Theater CommandChengdu610083China
| | - Yingmei Chen
- Department of Cardiologythe General Hospital of Western Theater CommandChengdu610083China
| | - Quanrong Pan
- Department of General Practicethe General Hospital of Western Theater CommandChengdu610083China
| | - Shizheng Fang
- Department of Critical Care Medicinethe General Hospital of Western Theater CommandChengdu610083China
| | - Zhongjian Zhang
- Department of Cardiologythe General Hospital of Western Theater CommandChengdu610083China
| | - Jia Wang
- Department of Cardiologythe General Hospital of Western Theater CommandChengdu610083China
| | - Yongjian Yang
- Department of Cardiologythe General Hospital of Western Theater CommandChengdu610083China
| | - Dachun Yang
- Department of Cardiologythe General Hospital of Western Theater CommandChengdu610083China
| | - Xiongshan Sun
- Department of Cardiologythe General Hospital of Western Theater CommandChengdu610083China
| |
Collapse
|
9
|
Yang R, Tang S, Xie X, Jin C, Tong Y, Huang W, Zan X. Enhanced Ocular Delivery of Beva via Ultra-Small Polymeric Micelles for Noninvasive Anti-VEGF Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2314126. [PMID: 38819852 DOI: 10.1002/adma.202314126] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 05/15/2024] [Indexed: 06/01/2024]
Abstract
Pathological ocular neovascularization resulting from retinal ischemia constitutes a major cause of vision loss. Current anti-VEGF therapies rely on burdensome intravitreal injections of Bevacizumab (Beva). Herein ultrasmall polymeric micelles encapsulating Beva (P@Beva) are developed for noninvasive topical delivery to posterior eye tissues. Beva is efficiently loaded into 11 nm micelles fabricated via self-assembly of hyperbranched amphiphilic copolymers. The neutral, brush-like micelles demonstrate excellent drug encapsulation and colloidal stability. In vitro, P@Beva enhances intracellular delivery of Beva in ocular cells versus free drug. Ex vivo corneal and conjunctival-sclera-choroidal tissues transport after eye drops are improved 23-fold and 7.9-fold, respectively. Anti-angiogenic bioactivity is retained with P@Beva eliciting greater inhibition of endothelial tube formation and choroid sprouting over Beva alone. Remarkably, in an oxygen-induced retinopathy (OIR) model, topical P@Beva matching efficacy of intravitreal Beva injection, is the clinical standard. Comprehensive biocompatibility verifies safety. Overall, this pioneering protein delivery platform holds promise to shift paradigms from invasive intravitreal injections toward simplified, noninvasive administration of biotherapeutics targeting posterior eye diseases.
Collapse
Affiliation(s)
- Ruhui Yang
- School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou, Zhejiang Province, 325035, China
- Wenzhou Key Laboratory of Perioperative Medicine, University of Chinese Academy of Sciences, Wenzhou, Zhejiang Province, 325001, China
| | - Sicheng Tang
- Wenzhou Key Laboratory of Perioperative Medicine, University of Chinese Academy of Sciences, Wenzhou, Zhejiang Province, 325001, China
| | - Xiaoling Xie
- School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou, Zhejiang Province, 325035, China
- Wenzhou Key Laboratory of Perioperative Medicine, University of Chinese Academy of Sciences, Wenzhou, Zhejiang Province, 325001, China
| | - Chaofan Jin
- Wenzhou Key Laboratory of Perioperative Medicine, University of Chinese Academy of Sciences, Wenzhou, Zhejiang Province, 325001, China
| | - Yuhua Tong
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang Province, 324000, China
| | - Wenjuan Huang
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, 317000, China
| | - Xingjie Zan
- School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou, Zhejiang Province, 325035, China
- Wenzhou Key Laboratory of Perioperative Medicine, University of Chinese Academy of Sciences, Wenzhou, Zhejiang Province, 325001, China
| |
Collapse
|
10
|
Yang WL, Zhang WF, Wang Y, Lou Y, Cai Y, Zhu J. Origin recognition complex 6 overexpression promotes growth of glioma cells. Cell Death Dis 2024; 15:485. [PMID: 38971772 PMCID: PMC11227543 DOI: 10.1038/s41419-024-06764-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 05/19/2024] [Accepted: 05/21/2024] [Indexed: 07/08/2024]
Abstract
The discovery of novel oncotargets for glioma is of immense significance. We here explored the expression patterns, biological functions, and underlying mechanisms associated with ORC6 (origin recognition complex 6) in glioma. Through the bioinformatics analyses, we found a significant increase in ORC6 expression within human glioma tissues, correlating with poorer overall survival, higher tumor grade, and wild-type isocitrate dehydrogenase status. Additionally, ORC6 overexpression is detected in glioma tissues obtained from locally-treated patients and across various primary/established glioma cells. Further bioinformatics scrutiny revealed that genes co-expressed with ORC6 are enriched in multiple signaling cascades linked to cancer. In primary and immortalized (A172) glioma cells, depleting ORC6 using specific shRNA or Cas9-sgRNA knockout (KO) significantly decreased cell viability and proliferation, disrupted cell cycle progression and mobility, and triggered apoptosis. Conversely, enhancing ORC6 expression via a lentiviral construct augmented malignant behaviors in human glioma cells. ORC6 emerged as a crucial regulator for the expression of key oncogenic genes, including Cyclin A2, Cyclin B2, and DNA topoisomerase II (TOP2A), within glioma cells. Silencing or KO of ORC6 reduced the mRNA and protein levels of these genes, while overexpression of ORC6 increased their expression in primary glioma cells. Bioinformatics analyses further identified RBPJ as a potential transcription factor of ORC6. RBPJ shRNA decreased ORC6 expression in primary glioma cells, while its overexpression increased it. Additionally, significantly enhanced binding between the RBPJ protein and the proposed ORC6 promoter region was detected in glioma tissues and cells. In vivo experiments demonstrated a significant reduction in the growth of patient-derived glioma xenografts in the mouse brain subsequent to ORC6 KO. ORC6 depletion, inhibited proliferation, decreased expression of Cyclin A2/B2/TOP2A, and increased apoptosis were detected within these ORC6 KO intracranial glioma xenografts. Altogether, RBPJ-driven ORC6 overexpression promotes glioma cell growth, underscoring its significance as a promising therapeutic target.
Collapse
Affiliation(s)
- Wen-Lei Yang
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei-Feng Zhang
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yin Wang
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Yue Lou
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Yu Cai
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jun Zhu
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
11
|
Huan MJ, Fu PP, Chen X, Wang ZX, Ma ZR, Cai SZ, Jiang Q, Wang Q. Identification of the central role of RNA polymerase mitochondrial for angiogenesis. Cell Commun Signal 2024; 22:343. [PMID: 38907279 PMCID: PMC11191269 DOI: 10.1186/s12964-024-01712-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 06/10/2024] [Indexed: 06/23/2024] Open
Abstract
Mitochondria are central to endothelial cell activation and angiogenesis, with the RNA polymerase mitochondrial (POLRMT) serving as a key protein in regulating mitochondrial transcription and oxidative phosphorylation. In our study, we examined the impact of POLRMT on angiogenesis and found that its silencing or knockout (KO) in human umbilical vein endothelial cells (HUVECs) and other endothelial cells resulted in robust anti-angiogenic effects, impeding cell proliferation, migration, and capillary tube formation. Depletion of POLRMT led to impaired mitochondrial function, characterized by mitochondrial depolarization, oxidative stress, lipid oxidation, DNA damage, and reduced ATP production, along with significant apoptosis activation. Conversely, overexpressing POLRMT promoted angiogenic activity in the endothelial cells. In vivo experiments demonstrated that endothelial knockdown of POLRMT, by intravitreous injection of endothelial specific POLRMT shRNA adeno-associated virus, inhibited retinal angiogenesis. In addition, inhibiting POLRMT with a first-in-class inhibitor IMT1 exerted significant anti-angiogenic impact in vitro and in vivo. Significantly elevated expression of POLRMT was observed in the retinal tissues of streptozotocin-induced diabetic retinopathy (DR) mice. POLRMT endothelial knockdown inhibited pathological retinal angiogenesis and mitigated retinal ganglion cell (RGC) degeneration in DR mice. At last, POLRMT expression exhibited a substantial increase in the retinal proliferative membrane tissues of human DR patients. These findings collectively establish the indispensable role of POLRMT in angiogenesis, both in vitro and in vivo.
Collapse
Affiliation(s)
- Meng-Jia Huan
- Department of Ophthalmology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Ping-Ping Fu
- Department of Ophthalmology, Shanghai Eye Diseases Prevention & Treatment Center, Shanghai Eye Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xia Chen
- Department of Anesthesiology, Children's hospital of Soochow University, Suzhou, 215025, China
| | - Zhao-Xia Wang
- Department of Endocrinology, Fengcheng Hospital of Fengxian Distric, Shanghai, China
| | - Zhou-Rui Ma
- Department of Burn and Plastic Surgery, Children's Hospital of Soochow University, Suzhou, China
| | - Shi-Zhong Cai
- Department of Child and Adolescent Healthcare, Children's Hospital of Soochow University, Suzhou, China.
- Key Laboratory of Congenital Structural Malformations of Suzhou City, Suzhou, China.
| | - Qin Jiang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210029, China.
| | - Qian Wang
- Department of Anesthesiology, Children's hospital of Soochow University, Suzhou, 215025, China.
| |
Collapse
|
12
|
Tan M, Liu Y, Xu Y, Yan G, Zhou N, Chen H, Jiang Z, Peng L. Plant-Derived Exosomes as Novel Nanotherapeutics Contrive Glycolysis Reprogramming-Mediated Angiogenesis for Diabetic Ulcer Healing. Biomater Res 2024; 28:0035. [PMID: 38840655 PMCID: PMC11151174 DOI: 10.34133/bmr.0035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 04/26/2024] [Indexed: 06/07/2024] Open
Abstract
Reversal of endothelial cell (EC) dysfunction under high-glucose (HG) conditions to achieve angiogenesis has remained a big challenge in diabetic ulcers. Herein, exosomes derived from medicinal plant ginseng (GExos) were shown as excellent nanotherapeutics with biomimetic cell membrane-like structures to be able to efficiently transfer the encapsulated active substances to ECs, resulting in a marked reprogramming of glycolysis by up-regulating anaerobic glycolysis and down-regulating oxidative stress, which further restore the proliferation, migration, and tubule formation abilities of ECs under HG conditions. In vivo, GExos enhance the angiogenesis and nascent vessel network reconstruction in full-thickness diabetic complicated skin ulcer wounds in mice with high biosafety. GExos were shown as promising nanotherapeutics in stimulating glycolysis reprogramming-mediated angiogenesis in diabetic ulcers, possessing wide application potential for reversing hyperglycemic dysangiogenesis and stimulating vascular regeneration.
Collapse
Affiliation(s)
- Minhong Tan
- College of Pharmaceutical Sciences,
Zhejiang University, Hangzhou 310058, PR China
- School of Materials Science and Engineering,
Zhejiang University, Hangzhou 310058, PR China
| | - Yuda Liu
- College of Pharmaceutical Sciences,
Zhejiang University, Hangzhou 310058, PR China
| | - Yang Xu
- College of Pharmaceutical Sciences,
Zhejiang University, Hangzhou 310058, PR China
| | - Ge Yan
- College of Pharmaceutical Sciences,
Zhejiang University, Hangzhou 310058, PR China
| | - Nan Zhou
- College of Pharmaceutical Sciences,
Zhejiang University, Hangzhou 310058, PR China
| | - Haoran Chen
- College of Pharmaceutical Sciences,
Zhejiang University, Hangzhou 310058, PR China
| | - Zhihong Jiang
- State Key Laboratory of Quality Research in Chinese Medicine,
Macau University of Science and Technology, Macau, PR China
| | - Lihua Peng
- College of Pharmaceutical Sciences,
Zhejiang University, Hangzhou 310058, PR China
- State Key Laboratory of Quality Research in Chinese Medicine,
Macau University of Science and Technology, Macau, PR China
| |
Collapse
|
13
|
Shi X, Zhou XZ, Chen G, Luo WF, Zhou C, He TJ, Naik MT, Jiang Q, Marshall J, Cao C. Targeting the postsynaptic scaffolding protein PSD-95 enhances BDNF signaling to mitigate depression-like behaviors in mice. Sci Signal 2024; 17:eadn4556. [PMID: 38687826 PMCID: PMC11223518 DOI: 10.1126/scisignal.adn4556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 04/10/2024] [Indexed: 05/02/2024]
Abstract
Signaling mediated by brain-derived neurotrophic factor (BDNF), which is supported by the postsynaptic scaffolding protein PSD-95, has antidepressant effects. Conversely, clinical depression is associated with reduced BDNF signaling. We found that peptidomimetic compounds that bind to PSD-95 promoted signaling by the BDNF receptor TrkB in the hippocampus and reduced depression-like behaviors in mice. The compounds CN2097 and Syn3 both bind to the PDZ3 domain of PSD-95, and Syn3 also binds to an α-helical region of the protein. Syn3 reduced depression-like behaviors in two mouse models of stress-induced depression; CN2097 had similar but less potent effects. In hippocampal neurons, application of Syn3 enhanced the formation of TrkB-Gαi1/3-PSD-95 complexes and potentiated downstream PI3K-Akt-mTOR signaling. In mice subjected to chronic mild stress (CMS), systemic administration of Syn3 reversed the CMS-induced, depression-associated changes in PI3K-Akt-mTOR signaling, dendrite complexity, spine density, and autophagy in the hippocampus and reduced depression-like behaviors. Knocking out Gαi1/3 in hippocampal neurons prevented the therapeutic effects of Syn3, indicating dependence of these effects on the TrkB pathway. The findings suggest that compounds that induce the formation of PSD-95-TrkB complexes have therapeutic potential to alleviate depression.
Collapse
Affiliation(s)
- Xin Shi
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou 215123, China
| | - Xiao-zhong Zhou
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou 215123, China
- Department of Orthopedics, the Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Gang Chen
- Department of Neurosurgery, the First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Wei-feng Luo
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou 215123, China
| | - Chengyu Zhou
- Department of Neuroscience, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Tian-ju He
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou 215123, China
| | - Mandar T. Naik
- Department of Molecular Biology, Cell Biology & Biochemistry, Brown University, Providence, RI 02912, USA
| | - Qin Jiang
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| | - John Marshall
- Department of Molecular Biology, Cell Biology & Biochemistry, Brown University, Providence, RI 02912, USA
| | - Cong Cao
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou 215123, China
| |
Collapse
|
14
|
Bi HE, Zhang J, Yao Y, Wang S, Yao J, Shao Z, Jiang Q. Expression and functional significance of phosphoenolpyruvate carboxykinase 1 in uveal melanoma. Cell Death Discov 2024; 10:196. [PMID: 38670942 PMCID: PMC11053060 DOI: 10.1038/s41420-024-01963-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Uveal melanoma (UVM), an uncommon yet potentially life-threatening ocular cancer, arises from melanocytes in the uveal tract of the eye. The exploration of novel oncotargets for UVM is of paramount importance. In this study, we show that PCK1 (phosphoenolpyruvate carboxykinase 1) expression is upregulated in various UVM tissues as well as in primary UVM cells and immortalized lines. Furthermore, bioinformatics studies reveal that PCK1 overexpression in UVM correlates with advanced disease stages and poor patient survival. Genetic silencing (utilizing viral shRNA) or knockout (via CRISPR/Cas9) of PCK1 significantly curtailed cell viability, proliferation, cell cycle progression, and motility, while provoking apoptosis in primary and immortalized UVM cells. Conversely, ectopic overexpression of PCK1, achieved through a viral construct, bolstered UVM cell proliferation and migration. Gαi3 expression and Akt phosphorylation were reduced following PCK1 silencing or knockout, but increased after PCK1 overexpression in UVM cells. Restoring Akt phosphorylation through a constitutively active mutant Akt1 (S473D) ameliorated the growth inhibition, migration suppression, and apoptosis induced by PCK1 silencing in UVM cells. Additionally, ectopic expression of Gαi3 restored Akt activation and counteracted the anti-UVM cell effects by PCK1 silencing. In vivo, the growth of subcutaneous xenografts of primary human UVM cells was significantly inhibited following intratumoral injection of adeno-associated virus (aav) expressing PCK1 shRNA. PCK1 depletion, Gαi3 downregulation, Akt inhibition, proliferation arrest, and apoptosis were detected in PCK1-silenced UVM xenografts. Collectively, our findings demonstrate that PCK1 promotes UVM cell growth possibly by modulating the Gαi3-Akt signaling pathway.
Collapse
Affiliation(s)
- Hui-E Bi
- The Affiliated Eye Hospital, The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- Department of Ophthalmology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Jie Zhang
- Obstetrics and Gynecology Department, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China
| | - Yujia Yao
- The Affiliated Eye Hospital, The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Suyu Wang
- The Affiliated Eye Hospital, The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Jin Yao
- The Affiliated Eye Hospital, The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China.
| | - Zhijiang Shao
- Department of Ophthalmology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China.
| | - Qin Jiang
- The Affiliated Eye Hospital, The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
15
|
Sun HJ, Ni ZR, Liu Y, Fu X, Liu SY, Hu JY, Sun QY, Li YC, Hou XH, Zhang JR, Zhu XX, Lu QB. Deficiency of neutral cholesterol ester hydrolase 1 (NCEH1) impairs endothelial function in diet-induced diabetic mice. Cardiovasc Diabetol 2024; 23:138. [PMID: 38664801 PMCID: PMC11046792 DOI: 10.1186/s12933-024-02239-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Neutral cholesterol ester hydrolase 1 (NCEH1) plays a critical role in the regulation of cholesterol ester metabolism. Deficiency of NCHE1 accelerated atherosclerotic lesion formation in mice. Nonetheless, the role of NCEH1 in endothelial dysfunction associated with diabetes has not been explored. The present study sought to investigate whether NCEH1 improved endothelial function in diabetes, and the underlying mechanisms were explored. METHODS The expression and activity of NCEH1 were determined in obese mice with high-fat diet (HFD) feeding, high glucose (HG)-induced mouse aortae or primary endothelial cells (ECs). Endothelium-dependent relaxation (EDR) in aortae response to acetylcholine (Ach) was measured. RESULTS Results showed that the expression and activity of NCEH1 were lower in HFD-induced mouse aortae, HG-exposed mouse aortae ex vivo, and HG-incubated primary ECs. HG exposure reduced EDR in mouse aortae, which was exaggerated by endothelial-specific deficiency of NCEH1, whereas NCEH1 overexpression restored the impaired EDR. Similar results were observed in HFD mice. Mechanically, NCEH1 ameliorated the disrupted EDR by dissociating endothelial nitric oxide synthase (eNOS) from caveolin-1 (Cav-1), leading to eNOS activation and nitric oxide (NO) release. Moreover, interaction of NCEH1 with the E3 ubiquitin-protein ligase ZNRF1 led to the degradation of Cav-1 through the ubiquitination pathway. Silencing Cav-1 and upregulating ZNRF1 were sufficient to improve EDR of diabetic aortas, while overexpression of Cav-1 and downregulation of ZNRF1 abolished the effects of NCEH1 on endothelial function in diabetes. Thus, NCEH1 preserves endothelial function through increasing NO bioavailability secondary to the disruption of the Cav-1/eNOS complex in the endothelium of diabetic mice, depending on ZNRF1-induced ubiquitination of Cav-1. CONCLUSIONS NCEH1 may be a promising candidate for the prevention and treatment of vascular complications of diabetes.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Aorta/enzymology
- Aorta/physiopathology
- Aorta/metabolism
- Aorta/drug effects
- Aorta/pathology
- Caveolin 1/metabolism
- Caveolin 1/deficiency
- Caveolin 1/genetics
- Cells, Cultured
- Diabetes Mellitus, Experimental/enzymology
- Diabetes Mellitus, Experimental/physiopathology
- Diet, High-Fat
- Endothelial Cells/enzymology
- Endothelial Cells/metabolism
- Endothelial Cells/drug effects
- Endothelium, Vascular/physiopathology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/drug effects
- Mice, Inbred C57BL
- Mice, Knockout
- Nitric Oxide/metabolism
- Nitric Oxide Synthase Type III/metabolism
- Obesity/enzymology
- Obesity/physiopathology
- Obesity/metabolism
- Signal Transduction
- Sterol Esterase/metabolism
- Sterol Esterase/genetics
- Ubiquitination
- Vasodilation/drug effects
Collapse
Affiliation(s)
- Hai-Jian Sun
- Department of Physiology, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China.
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China.
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China.
| | - Zhang-Rong Ni
- Department of Physiology, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Yao Liu
- Department of Cardiac Ultrasound, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, China
| | - Xiao Fu
- Department of Physiology, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Shi-Yi Liu
- Department of Physiology, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Jin-Yi Hu
- Department of Physiology, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Qing-Yi Sun
- Department of Physiology, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Yu-Chao Li
- Department of Physiology, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Xiao-Hui Hou
- Department of Cardiac Ultrasound, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, China
| | - Ji-Ru Zhang
- Department of Physiology, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China.
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China.
| | - Xue-Xue Zhu
- Department of Physiology, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China.
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China.
| | - Qing-Bo Lu
- Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, 214125, China.
| |
Collapse
|
16
|
Xiong QW, Jiang K, Shen XW, Ma ZR, Yan XM, Xia H, Cao X. The requirement of the mitochondrial protein NDUFS8 for angiogenesis. Cell Death Dis 2024; 15:253. [PMID: 38594244 PMCID: PMC11004167 DOI: 10.1038/s41419-024-06636-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/11/2024]
Abstract
Mitochondria are important for the activation of endothelial cells and the process of angiogenesis. NDUFS8 (NADH:ubiquinone oxidoreductase core subunit S8) is a protein that plays a critical role in the function of mitochondrial Complex I. We aimed to investigate the potential involvement of NDUFS8 in angiogenesis. In human umbilical vein endothelial cells (HUVECs) and other endothelial cell types, we employed viral shRNA to silence NDUFS8 or employed the CRISPR/Cas9 method to knockout (KO) it, resulting in impaired mitochondrial functions in the endothelial cells, causing reduction in mitochondrial oxygen consumption and Complex I activity, decreased ATP production, mitochondrial depolarization, increased oxidative stress and reactive oxygen species (ROS) production, and enhanced lipid oxidation. Significantly, NDUFS8 silencing or KO hindered cell proliferation, migration, and capillary tube formation in cultured endothelial cells. In addition, there was a moderate increase in apoptosis within NDUFS8-depleted endothelial cells. Conversely, ectopic overexpression of NDUFS8 demonstrated a pro-angiogenic impact, enhancing cell proliferation, migration, and capillary tube formation in HUVECs and other endothelial cells. NDUFS8 is pivotal for Akt-mTOR cascade activation in endothelial cells. Depleting NDUFS8 inhibited Akt-mTOR activation, reversible with exogenous ATP in HUVECs. Conversely, NDUFS8 overexpression boosted Akt-mTOR activation. Furthermore, the inhibitory effects of NDUFS8 knockdown on cell proliferation, migration, and capillary tube formation were rescued by Akt re-activation via a constitutively-active Akt1. In vivo experiments using an endothelial-specific NDUFS8 shRNA adeno-associated virus (AAV), administered via intravitreous injection, revealed that endothelial knockdown of NDUFS8 inhibited retinal angiogenesis. ATP reduction, oxidative stress, and enhanced lipid oxidation were detected in mouse retinal tissues with endothelial knockdown of NDUFS8. Lastly, we observed an increase in NDUFS8 expression in retinal proliferative membrane tissues obtained from human patients with proliferative diabetic retinopathy. Our findings underscore the essential role of the mitochondrial protein NDUFS8 in regulating endothelial cell activation and angiogenesis.
Collapse
Affiliation(s)
- Qian-Wei Xiong
- Department of Urology Surgery, Children's Hospital of Soochow University, Suzhou, China
| | - Kun Jiang
- Vascular Surgery Department, Kunshan Traditional Chinese Medicine Hospital, Kunshan, China
| | - Xiao-Wei Shen
- Department of General Surgery, QingPu Branch of Zhongshan Hospital Affiliated to Fudan University, QingPu District Central Hospital Shanghai, Shanghai, China
| | - Zhou-Rui Ma
- Department of Burns and Plastic Surgery, Children's Hospital of Soochow University, Suzhou, China
| | - Xiang-Ming Yan
- Department of Urology Surgery, Children's Hospital of Soochow University, Suzhou, China.
| | - Hao Xia
- Department of Pediatric Emergency and Critical Care Medicine, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xu Cao
- Department of Urology Surgery, Children's Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
17
|
Bai CW, Lu L, Zhang JN, Zhou C, Ni YC, Li KR, Yao J, Zhou XZ, Lan CG, Cao C. G protein subunit alpha i2's pivotal role in angiogenesis. Theranostics 2024; 14:2190-2209. [PMID: 38505600 PMCID: PMC10945342 DOI: 10.7150/thno.92909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/26/2024] [Indexed: 03/21/2024] Open
Abstract
Here we explored the potential role of Gαi2 (G protein subunit alpha i2) in endothelial cell function and angiogenesis. Methods: Genetic methodologies such as shRNA, CRISPR/Cas9, dominant negative mutation, and overexpression were utilized to modify Gαi2 expression or regulate its function. Their effects on endothelial cell functions were assessed in vitro. In vivo, the endothelial-specific Gαi2 shRNA adeno-associated virus (AAV) was utilized to silence Gαi2 expression. The impact of this suppression on retinal angiogenesis in control mice and streptozotocin (STZ)-induced diabetic retinopathy (DR) mice was analyzed. Results: Analysis of single-cell RNA sequencing data revealed Gαi2 (GNAI2) was predominantly expressed in retinal endothelial cells and expression was increased in retinal endothelial cells following oxygen-induced retinopathy (OIR) in mice. Moreover, transcriptome analysis linking Gαi2 to angiogenesis-related processes/pathways, supported by increased Gαi2 expression in experimental OIR mouse retinas, highlighted its possible role in angiogenesis. In various endothelial cell types, shRNA-induced silencing and CRISPR/Cas9-mediated knockout (KO) of Gαi2 resulted in substantial reductions in cell proliferation, migration, invasion, and capillary tube formation. Conversely, Gαi2 over-expression in endothelial cells induced pro-angiogenic activities, enhancing cell proliferation, migration, invasion, and capillary tube formation. Furthermore, our investigation revealed a crucial role of Gαi2 in NFAT (nuclear factor of activated T cells) activation, as evidenced by the down-regulation of NFAT-luciferase reporter activity and pro-angiogenesis NFAT-targeted genes (Egr3, CXCR7, and RND1) in Gαi2-silenced or -KO HUVECs, which were up-regulated in Gαi2-overexpressing endothelial cells. Expression of a dominant negative Gαi2 mutation (S48C) also down-regulated NFAT-targeted genes, slowing proliferation, migration, invasion, and capillary tube formation in HUVECs. Importantly, in vivo experiments revealed that endothelial Gαi2 knockdown inhibited retinal angiogenesis in mice, with a concomitant down-regulation of NFAT-targeted genes in mouse retinal tissue. In contrast, Gαi2 over-expression in endothelial cells enhanced retinal angiogenesis in mice. Single-cell RNA sequencing data confirmed increased levels of Gαi2 specifically in retinal endothelial cells of mice with streptozotocin (STZ)-induced diabetic retinopathy (DR). Importantly, endothelial Gαi2 silencing ameliorated retinal pathological angiogenesis in DR mice. Conclusion: Our study highlights a critical role for Gαi2 in NFAT activation, endothelial cell activation and angiogenesis, offering valuable insights into potential therapeutic strategies for modulating these processes.
Collapse
Affiliation(s)
- Chao-wen Bai
- Department of Orthopedics, Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou, China
| | - Lu Lu
- Department of Joint Surgery and Geriatric Orthopedics, Affiliated Hospital of YouJiang Medical University for Nationalities, Guangxi Key Laboratory of Basic and Translational Research of Bone and Joint Degenerative Diseases, Guangxi Biomedical Materials Engineering Research Center for Bone and Joint Degenerative Diseases, Baise City, China
| | - Jia-nan Zhang
- Department of Orthopedics, Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou, China
| | - Chengyu Zhou
- Department of Neuroscience, Case Western Reserve University, Cleveland, USA
| | - Yi-chao Ni
- Department of Orthopedics, Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou, China
| | - Ke-ran Li
- The Fourth Medical School, Eye hospital, Nanjing Medical University, Nanjing, China
| | - Jin Yao
- The Fourth Medical School, Eye hospital, Nanjing Medical University, Nanjing, China
| | - Xiao-zhong Zhou
- Department of Orthopedics, Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou, China
| | - Chang-gong Lan
- Department of Joint Surgery and Geriatric Orthopedics, Affiliated Hospital of YouJiang Medical University for Nationalities, Guangxi Key Laboratory of Basic and Translational Research of Bone and Joint Degenerative Diseases, Guangxi Biomedical Materials Engineering Research Center for Bone and Joint Degenerative Diseases, Baise City, China
| | - Cong Cao
- Department of Orthopedics, Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou, China
| |
Collapse
|
18
|
Li L, Hua S, You L, Zhong T. Secretome Derived from Mesenchymal Stem/Stromal Cells: A Promising Strategy for Diabetes and its Complications. Curr Stem Cell Res Ther 2024; 19:1328-1350. [PMID: 37711134 DOI: 10.2174/1574888x19666230913154544] [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] [Received: 02/10/2023] [Revised: 07/06/2023] [Accepted: 07/24/2023] [Indexed: 09/16/2023]
Abstract
Diabetes is a complex metabolic disease with a high global prevalence. The health and quality of life of patients with diabetes are threatened by many complications, including diabetic foot ulcers, diabetic kidney diseases, diabetic retinopathy, and diabetic peripheral neuropathy. The application of mesenchymal stem/stromal cells (MSCs) in cell therapies has been recognized as a potential treatment for diabetes and its complications. MSCs were originally thought to exert biological effects exclusively by differentiating and replacing specific impaired cells. However, the paracrine function of factors secreted by MSCs may exert additional protective effects. MSCs secrete multiple compounds, including proteins, such as growth factors, chemokines, and other cytokines; nucleic acids, such as miRNAs; and lipids, extracellular vesicles (EVs), and exosomes (Exos). Collectively, these secreted compounds are called the MSC secretome, and usage of these chemicals in cell-free therapies may provide stronger effects with greater safety and convenience. Recent studies have demonstrated positive effects of the MSC secretome, including improved insulin sensitivity, reduced inflammation, decreased endoplasmic reticulum stress, enhanced M2 polarization of macrophages, and increased angiogenesis and autophagy; however, the mechanisms leading to these effects are not fully understood. This review summarizes the current research regarding the secretome derived from MSCs, including efforts to quantify effectiveness and uncover potential molecular mechanisms in the treatment of diabetes and related disorders. In addition, limitations and challenges are also discussed so as to facilitate applications of the MSC secretome as a cell-free therapy for diabetes and its complications.
Collapse
Affiliation(s)
- Ling Li
- Nanjing Maternity and Child Health Care Institute, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China
| | - Siyu Hua
- Nanjing Maternity and Child Health Care Institute, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China
| | - Lianghui You
- Nanjing Maternity and Child Health Care Institute, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China
| | - Tianying Zhong
- Nanjing Maternity and Child Health Care Institute, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China
| |
Collapse
|
19
|
Yin DP, Zhang H, Teng H, Zhang D, Chen P, Xie L, Liu JS. Overexpressed Gαi1 exerts pro-tumorigenic activity in nasopharyngeal carcinoma. Cell Death Dis 2023; 14:792. [PMID: 38049415 PMCID: PMC10696052 DOI: 10.1038/s41419-023-06308-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 11/04/2023] [Accepted: 11/14/2023] [Indexed: 12/06/2023]
Abstract
The current study tested the expression and potential functions of Gαi1 in nasopharyngeal carcinoma (NPC). The Cancer Genome Atlas (TCGA) database results demonstrate that Gαi1 transcripts' number in NPC tissues is significantly higher than that in the normal nasal epithelial tissues. Its overexpression correlates with poor survival in certain NPC patients. Moreover, Gαi1 is significantly upregulated in NPC tissues of local primary patients and in different primary human NPC cells. Whereas its expression is relatively low in cancer-surrounding normal tissues and in primary nasal epithelial cells. Genetic silencing (via shRNA strategy) or knockout (via CRISPR-sgRNA method) of Gαi1 substantially suppressed viability, proliferation, cell cycle progression, and migration in primary NPC cells, causing significant caspase-apoptosis activation. Contrarily, ectopic Gαi1 expression exerted pro-tumorigenic activity and strengthened cell proliferation and migration in primary NPC cells. Gαi1 is important for Akt-mTOR activation in NPC cells. Akt-S6K phosphorylation was downregulated after Gαi1 shRNA or KO in primary NPC cells, but strengthened following Gαi1 overexpression. In Gαi1-silenced primary NPC cells, a S473D constitutively-active mutant Akt1 (caAkt1) restored Akt-S6K phosphorylation and ameliorated Gαi1 shRNA-induced proliferation inhibition, migration reduction and apoptosis. Bioinformatics analyses proposed zinc finger protein 384 (ZNF384) as a potential transcription factor of Gαi1. In primary NPC cells, ZNF384 shRNA or knockout (via CRISPR-sgRNA method) decreased Gαi1 mRNA and protein expression, whereas ZNF384 overexpression upregulated it. Importantly, there was an increased binding between ZNF384 protein and the Gαi1 promoter in human NPC tissues and different NPC cells. In vivo studies showed that intratumoral injection of Gαi1-shRNA-expressing adeno-associated virus (AAV) impeded subcutaneous NPC xenograft growth in nude mice. Gαi1 downregulation, Akt-mTOR inactivation, and apoptosis induction were detected in Gαi1-silenced NPC xenograft tissues. Gαi1 KO also effectively inhibited the growth of NPC xenografts in nude mice. Together, overexpressed Gαi1 exerts pro-tumorigenic activity in NPC possibly by promoting Akt-mTOR activation.
Collapse
Affiliation(s)
- De-Pei Yin
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Otorhinolaryngology Head and Neck Surgery, Children's Hospital of Soochow University, Suzhou, China
| | - Huanle Zhang
- Department of Radiotherapy, Suzhou Ninth People's Hospital, Suzhou, China
| | - Hua Teng
- Department of Otorhinolaryngology Head and Neck Surgery, Jiangsu Province Hospital on Integration of Chinese and Western Medicine, Nanjing, China
| | - Dan Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Peipei Chen
- Department of Otorhinolaryngology Head and Neck Surgery, Children's Hospital of Soochow University, Suzhou, China.
| | - Lixiao Xie
- Department of Otorhinolaryngology Head and Neck Surgery, Children's Hospital of Soochow University, Suzhou, China.
| | - Ji-Sheng Liu
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
20
|
Li J, Chen K, Li X, Zhang X, Zhang L, Yang Q, Xia Y, Xie C, Wang X, Tong J, Shen Y. Mechanistic insights into the alterations and regulation of the AKT signaling pathway in diabetic retinopathy. Cell Death Discov 2023; 9:418. [PMID: 37978169 PMCID: PMC10656479 DOI: 10.1038/s41420-023-01717-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 11/05/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023] Open
Abstract
In the early stages of diabetic retinopathy (DR), diabetes-related hyperglycemia directly inhibits the AKT signaling pathway by increasing oxidative stress or inhibiting growth factor expression, which leads to retinal cell apoptosis, nerve proliferation and fundus microvascular disease. However, due to compensatory vascular hyperplasia in the late stage of DR, the vascular endothelial growth factor (VEGF)/phosphatidylinositol 3 kinase (PI3K)/AKT cascade is activated, resulting in opposite levels of AKT regulation compared with the early stage. Studies have shown that many factors, including insulin, insulin-like growth factor-1 (IGF-1), VEGF and others, can regulate the AKT pathway. Disruption of the insulin pathway decreases AKT activation. IGF-1 downregulation decreases the activation of AKT in DR, which abrogates the neuroprotective effect, upregulates VEGF expression and thus induces neovascularization. Although inhibiting VEGF is the main treatment for neovascularization in DR, excessive inhibition may lead to apoptosis in inner retinal neurons. AKT pathway substrates, including mammalian target of rapamycin (mTOR), forkhead box O (FOXO), glycogen synthase kinase-3 (GSK-3)/nuclear factor erythroid 2-related factor 2 (Nrf2), and nuclear factor kappa-B (NF-κB), are a research focus. mTOR inhibitors can delay or prevent retinal microangiopathy, whereas low mTOR activity can decrease retinal protein synthesis. Inactivated AKT fails to inhibit FOXO and thus causes apoptosis. The GSK-3/Nrf2 cascade regulates oxidation and inflammation in DR. NF-κB is activated in diabetic retinas and is involved in inflammation and apoptosis. Many pathways or vital activities, such as the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) and mitogen-activated protein kinase (MAPK) signaling pathways, interact with the AKT pathway to influence DR development. Numerous regulatory methods can simultaneously impact the AKT pathway and other pathways, and it is essential to consider both the connections and interactions between these pathways. In this review, we summarize changes in the AKT signaling pathway in DR and targeted drugs based on these potential sites.
Collapse
Affiliation(s)
- Jiayuan Li
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Kuangqi Chen
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiang Li
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Xuhong Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Liyue Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Qianjie Yang
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Yutong Xia
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Chen Xie
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiawei Wang
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianping Tong
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China.
| | - Ye Shen
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
21
|
Liu TT, Shi X, Hu HW, Chen JP, Jiang Q, Zhen YF, Cao C, Liu XW, Liu JG. Endothelial cell-derived RSPO3 activates Gαi1/3-Erk signaling and protects neurons from ischemia/reperfusion injury. Cell Death Dis 2023; 14:654. [PMID: 37805583 PMCID: PMC10560285 DOI: 10.1038/s41419-023-06176-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 09/19/2023] [Accepted: 09/26/2023] [Indexed: 10/09/2023]
Abstract
The current study explores the potential function and the underlying mechanisms of endothelial cell-derived R-spondin 3 (RSPO3) neuroprotection against ischemia/reperfusion-induced neuronal cell injury. In both neuronal cells (Neuro-2a) and primary murine cortical neurons, pretreatment with RSPO3 ameliorated oxygen and glucose deprivation (OGD)/re-oxygenation (OGD/R)-induced neuronal cell death and oxidative injury. In neurons RSPO3 activated the Akt, Erk and β-Catenin signaling cascade, but only Erk inhibitors reversed RSPO3-induced neuroprotection against OGD/R. In mouse embryonic fibroblasts (MEFs) and neuronal cells, RSPO3-induced LGR4-Gab1-Gαi1/3 association was required for Erk activation, and either silencing or knockout of Gαi1 and Gαi3 abolished RSPO3-induced neuroprotection. In mice, middle cerebral artery occlusion (MCAO) increased RSPO3 expression and Erk activation in ischemic penumbra brain tissues. Endothelial knockdown or knockout of RSPO3 inhibited Erk activation in the ischemic penumbra brain tissues and increased MCAO-induced cerebral ischemic injury in mice. Conversely, endothelial overexpression of RSPO3 ameliorated MCAO-induced cerebral ischemic injury. We conclude that RSPO3 activates Gαi1/3-Erk signaling to protect neuronal cells from ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Ting-Tao Liu
- Shandong University, Department of Neurology, Shandong Provincial Hospital, Jinan, China
- Department of Neurology, Shouguang Hospital of T.C.M, Shouguang, China
| | - Xin Shi
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Hong-Wei Hu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ju-Ping Chen
- Department of Neurology, Changshu Hospital of Traditional Chinese Medicine, Changshu, China
| | - Qin Jiang
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Yun-Fang Zhen
- Department of Orthopedics, Children's hospital of Soochow University, Suzhou, China.
| | - Cong Cao
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Xue-Wu Liu
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| | - Jian-Gang Liu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
22
|
Han Q, Yan P, Song R, Liu F, Tian Q. HOXC13-driven TIMM13 overexpression promotes osteosarcoma cell growth. Cell Death Dis 2023; 14:398. [PMID: 37407582 DOI: 10.1038/s41419-023-05910-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 06/11/2023] [Accepted: 06/19/2023] [Indexed: 07/07/2023]
Abstract
TIMM13 (translocase of inner mitochondrial membrane 13) located at the mitochondrial intermembrane space is vital for the integrity and function of mitochondria. We found that the mitochondrial protein TIMM13 is upregulated in human OS tissues and cells. In patient-derived primary OS cells and established cell lines, TIMM13 shRNA or knockout provoked mitochondrial dysfunction, causing mitochondrial depolarization, reactive oxygen species production, and oxidative injury, as well as lipid peroxidation, DNA damage, and ATP depletion. Moreover, TIMM13 depletion provoked OS cell apoptosis and inhibited cell proliferation and migration. Conversely, ectopic TIMM13 overexpression increased ATP contents, enhancing OS cell proliferation and migration. Moreover, we discovered that Akt-mTOR activation was inhibited with TIMM13 depletion in primary OS cells. Further studies revealed that HOXC13 (Homeobox C13)-dependent TIMM13 transcription was significantly increased in OS tissues and cells. Whereas TIMM13 transcription and expression were decreased following HOXC13 silencing in primary OS cells. In vivo, TIMM13 KO potently inhibited OS xenograft growth in the proximal tibia of nude mice. TIMM13 KO also induced Akt-mTOR inactivation, ATP depletion, oxidative injury, and apoptosis in the in situ OS tumors. Together, upregulation of the mitochondrial protein TIMM13 is important for OS cell growth, representing a novel and promising therapeutic target.
Collapse
Affiliation(s)
- Qicai Han
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Penghui Yan
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruipeng Song
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Feifei Liu
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qing Tian
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
23
|
Li L, Shen FR, Cheng Q, Sun J, Li H, Sun HT, Cai X, Chen M, Yang B, Wang L, Xu L. SLC5A3 is important for cervical cancer cell growth. Int J Biol Sci 2023; 19:2787-2802. [PMID: 37324953 PMCID: PMC10266070 DOI: 10.7150/ijbs.84570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/16/2023] [Indexed: 06/17/2023] Open
Abstract
Novel molecular targets for cervical cancer must be identified. This study examined the role of SLC5A3, a myo-inositol transporter, in the pathogenesis of cervical cancer. Through boinformatics analysis, we showed that the SLC5A3 mRNA levels were upregulated in cervical cancer tissues. The upregulated SLC5A3 mRNA levels were negatively correlated with survival and progression-free interval. Genes co-expressed with SLC5A3 were enriched in multiple signaling cascades involved in cancer progression. In primary/established cervical cancer cells, SLC5A3 shRNA/knockout (KO) exerted growth-inhibitory effects and promoted cell death/apoptosis. Furthermore, SLC5A3 knockdown or KO downregulated myo-inositol levels, induced oxidative injury, and decreased Akt-mTOR activation in cervical cancer cells. In contrast, supplementation of myo-inositol or n-acetyl-L-cysteine or transduction of a constitutively active Akt1 construct mitigated SLC5A3 KO-induced cytotoxicity in cervical cancer cells. Lentiviral SLC5A3 overexpression construct transduction upregulated the cellular myo-inositol level and promoted Akt-mTOR activation, enhancing cervical cancer cell proliferation and migration. The binding of TonEBP to the SLC5A3 promoter was upregulated in cervical cancer. In vivo studies showed that intratumoral injection of SLC5A3 shRNA-expressing virus arrested cervical cancer xenograft growth in mice. SLC5A3 KO also inhibited pCCa-1 cervical cancer xenograft growth. The SLC5A3-depleted xenograft tissues exhibited myo-inositol downregulation, Akt-mTOR inactivation, and oxidative injury. Transduction of sh-TonEBP AAV construct downregulated SLC5A3 expression and inhibited pCCa-1 cervical cancer xenograft growth. Together, overexpressed SLC5A3 promotes growth of cervical cancer cells, representing as a novel therapeutic oncotarget for the devastating disease.
Collapse
Affiliation(s)
- Li Li
- Department of Obstetrics and Gynecology, Minhang Hospital, Fudan University, Shanghai, China
| | - Fang-rong Shen
- Obstetrics and Gynecology Department, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qunxian Cheng
- Department of Obstetrics and Gynecology, Minhang Hospital, Fudan University, Shanghai, China
| | - Jing Sun
- Department of Obstetrics and Gynecology, Minhang Hospital, Fudan University, Shanghai, China
| | - Hang Li
- Department of Obstetrics and Gynecology, Minhang Hospital, Fudan University, Shanghai, China
| | - Hua-ting Sun
- Obstetrics and Gynecology Department, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xia Cai
- Department of Obstetrics and Gynecology, Minhang Hospital, Fudan University, Shanghai, China
| | - Mengting Chen
- Department of Obstetrics and Gynecology, Minhang Hospital, Fudan University, Shanghai, China
| | - Baohua Yang
- Department of Obstetrics and Gynecology, Minhang Hospital, Fudan University, Shanghai, China
| | - Lifeng Wang
- Department of Obstetrics and Gynecology, Minhang Hospital, Fudan University, Shanghai, China
| | - Ling Xu
- Department of Obstetrics and Gynecology, Minhang Hospital, Fudan University, Shanghai, China
| |
Collapse
|
24
|
Ma ZR, Li HP, Cai SZ, Du SY, Chen X, Yao J, Cao X, Zhen YF, Wang Q. The mitochondrial protein TIMM44 is required for angiogenesis in vitro and in vivo. Cell Death Dis 2023; 14:307. [PMID: 37147302 PMCID: PMC10163060 DOI: 10.1038/s41419-023-05826-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/07/2023]
Abstract
The mitochondrial integrity and function in endothelial cells are essential for angiogenesis. TIMM44 (translocase of inner mitochondrial membrane 44) is essential for integrity and function of mitochondria. Here we explored the potential function and the possible mechanisms of TIMM44 in angiogenesis. In HUVECs, human retinal microvascular endothelial cells and hCMEC/D3 brain endothelial cells, silence of TIMM44 by targeted shRNA largely inhibited cell proliferation, migration and in vitro capillary tube formation. TIMM44 silencing disrupted mitochondrial functions in endothelial cells, causing mitochondrial protein input arrest, ATP reduction, ROS production, and mitochondrial depolarization, and leading to apoptosis activation. TIMM44 knockout, by Cas9-sgRNA strategy, also disrupted mitochondrial functions and inhibited endothelial cell proliferation, migration and in vitro capillary tube formation. Moreover, treatment with MB-10 ("MitoBloCK-10"), a TIMM44 blocker, similarly induced mitochondrial dysfunction and suppressed angiogenic activity in endothelial cells. Contrarily, ectopic overexpression of TIMM44 increased ATP contents and augmented endothelial cell proliferation, migration and in vitro capillary tube formation. In adult mouse retinas, endothelial knockdown of TIMM44, by intravitreous injection of endothelial specific TIMM44 shRNA adenovirus, inhibited retinal angiogenesis, causing vascular leakage, acellular capillary growth, and retinal ganglion cells degeneration. Significant oxidative stress was detected in TIMM44-silenced retinal tissues. Moreover, intravitreous injection of MB-10 similarly induced oxidative injury and inhibited retinal angiogenesis in vivo. Together, the mitochondrial protein TIMM44 is important for angiogenesis in vitro and in vivo, representing as a novel and promising therapeutic target of diseases with abnormal angiogenesis.
Collapse
Affiliation(s)
- Zhou-Rui Ma
- Department of Burns and Plastic Surgery, Children's hospital of Soochow University, Suzhou, China
- Suzhou Key Laboratory of Children's Structural Deformities, Suzhou, China
| | - Hong-Peng Li
- Kunshan Hospital of Chinese Medicine, Affiliated Hospital of Yangzhou University, Kunshan, China
| | - Shi-Zhong Cai
- Suzhou Key Laboratory of Children's Structural Deformities, Suzhou, China
- Department of Child and Adolescent Healthcare, Children's Hospital of Soochow University, Suzhou, China
| | - Sheng-Yang Du
- Department of Orthopedics, Xuzhou First People's Hospital, Xuzhou, China
| | - Xia Chen
- Department of Anesthesiology, Children's hospital of Soochow University, Suzhou, China
| | - Jin Yao
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China.
| | - Xu Cao
- Suzhou Key Laboratory of Children's Structural Deformities, Suzhou, China.
- Department of Urology, Children's Hospital of Soochow University, Suzhou, China.
| | - Yun-Fang Zhen
- Department of Orthopedics, Children's hospital of Soochow University, Suzhou, China.
| | - Qian Wang
- Department of Anesthesiology, Children's hospital of Soochow University, Suzhou, China.
| |
Collapse
|
25
|
Shan HJ, Jiang K, Zhao MZ, Deng WJ, Cao WH, Li JJ, Li KR, She C, Luo WF, Yao J, Zhou XZ, Zhang D, Cao C. SCF/c-Kit-activated signaling and angiogenesis require Gαi1 and Gαi3. Int J Biol Sci 2023; 19:1910-1924. [PMID: 37063428 PMCID: PMC10092767 DOI: 10.7150/ijbs.82855] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 03/03/2023] [Indexed: 04/18/2023] Open
Abstract
The stem cell factor (SCF) binds to c-Kit in endothelial cells, thus activating downstream signaling and angiogenesis. Herein, we examined the role of G protein subunit alpha inhibitory (Gαi) proteins in this process. In MEFs and HUVECs, Gαi1/3 was associated with SCF-activated c-Kit, promoting c-Kit endocytosis, and binding of key adaptor proteins, subsequently transducing downstream signaling. SCF-induced Akt-mTOR and Erk activation was robustly attenuated by Gαi1/3 silencing or knockout (KO), or due to dominant negative mutations but was strengthened substantially following ectopic overexpression of Gαi1/3. SCF-induced HUVEC proliferation, migration, and capillary tube formation were suppressed after Gαi1/3 silencing or KO, or due to dominant negative mutations. In vivo, endothelial knockdown of Gαi1/3 by intravitreous injection of endothelial-specific shRNA adeno-associated virus (AAV) potently reduced SCF-induced signaling and retinal angiogenesis in mice. Moreover, mRNA and protein expressions of SCF increased significantly in the retinal tissues of streptozotocin-induced diabetic retinopathy (DR) mice. SCF silencing, through intravitreous injection of SCF shRNA AAV, inhibited pathological retinal angiogenesis and degeneration of retinal ganglion cells in DR mice. Finally, the expression of SCF and c-Kit increased in proliferative retinal tissues of human patients with proliferative DR. Taken together, Gαi1/3 mediate SCF/c-Kit-activated signaling and angiogenesis.
Collapse
Affiliation(s)
- Hua-jian Shan
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou, China
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Kun Jiang
- Vascular Surgery Department, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, China
| | - Ming-zhi Zhao
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou, China
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Wen-jing Deng
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou, China
| | - Wen-hao Cao
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou, China
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jia-jun Li
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Ke-ran Li
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Chang She
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou, China
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Wei-feng Luo
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou, China
| | - Jin Yao
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Xiao-zhong Zhou
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou, China
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Dan Zhang
- Department of Otorhinolaryngology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Cong Cao
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou, China
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
26
|
Zhang H, Si P, Kong Q, Ma J. Transcriptome reveals the toxicity and genetic response of zebrafish to naphthenic acids and benzo[a]pyrene at ambient concentrations. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 253:114700. [PMID: 36863161 DOI: 10.1016/j.ecoenv.2023.114700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 02/14/2023] [Accepted: 02/26/2023] [Indexed: 06/18/2023]
Abstract
Naphthenic acids (NAs) are typical contaminants in heavily crude oil. Benzo[a]pyrene (B[a]P) is also a component of crude oil, but their combined effects have not been systematically explored. In this study, zebrafish (Danio rerio) were used as the test organisms, and behavioral indicators and enzyme activities were used as toxicity indicators. Combined with the effects of environmental concentrations, the toxic effects of low concentrations of commercially available NAs (0.5 mg/LNA) and benzo[a]pyrene (0.8 μg/LBaP) at single and compound exposures (0.5 mg/LNA and 0.8 μg/LBaP) were assayed in zebrafish, and transcriptome sequencing technology was used to explore the molecular mechanism of the two compounds affecting zebrafish from the molecular biology level. Sensitive molecular markers that could indicate the presence of contaminants were screened. The results showed that (1) zebrafish in the NA and BaP exposure groups exhibited increased locomotor behavior, and the mixed exposure group exhibited inhibition of locomotor behavior. Oxidative stress biomarkers showed increased activity under single exposure and decreased activity under the mixed exposure. (2) NA stress led to changes in the activity of transporters and the intensity of energy metabolism; BaP directly stimulates the pathway of actin production. When the two compounds are combined, the excitability of neurons in the central nervous system is decreased, and the actin-related genes are down-regulated. (3) After BaP and Mix treatments, genes were enriched in the cytokine-receptor interaction and actin signal pathway, while NA increased the toxic effect on the mixed treatment group. In general, the interaction between NA and BaP has a synergistic effect on the transcription of zebrafish nerve and motor behavior-related genes, resulting in increased toxicity under combined exposure. The changes in expression of various zebrafish genes are manifested in the changes in the normal movement behavior of zebrafish and the intensification of oxidative stress in the apparent behavior and physiological indicators. CAPSULE ABSTRACT: We investigated the toxicity and genetic alterations caused by NA, B[a]P, and their mixtures in zebrafish in an aquatic environment using transcriptome sequencing technology and comprehensive behavioral analysis. These changes involved energy metabolism, the generation of muscle cells, and the nervous system.
Collapse
Affiliation(s)
- Huanxin Zhang
- College of Geography and Environment, Shandong Normal University, Jinan 250000, China.
| | - Panpan Si
- College of Geography and Environment, Shandong Normal University, Jinan 250000, China
| | - Qiang Kong
- College of Geography and Environment, Shandong Normal University, Jinan 250000, China
| | - Jinyue Ma
- College of Geography and Environment, Shandong Normal University, Jinan 250000, China
| |
Collapse
|
27
|
Peng SQ, Zhu XR, Zhao MZ, Zhang YF, Wang AR, Chen MB, Ye ZY. Identification of matrix-remodeling associated 5 as a possible molecular oncotarget of pancreatic cancer. Cell Death Dis 2023; 14:157. [PMID: 36828810 PMCID: PMC9958022 DOI: 10.1038/s41419-023-05684-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/11/2023] [Accepted: 02/14/2023] [Indexed: 02/26/2023]
Abstract
Pancreatic cancer has an extremely poor prognosis. Here we examined expression, potential functions and underlying mechanisms of MXRA5 (matrix remodeling associated 5) in pancreatic cancer. Bioinformatics studies revealed that MXRA5 transcripts are significantly elevated in pancreatic cancer tissues, correlating with the poor overall survival, high T-stage, N1 and pathologic stage of the patients. MXRA5 mRNA and protein expression is significantly elevated in microarray pancreatic cancer tissues and different pancreatic cancer cells. In primary and immortalized (BxPC-3 and PANC-1 lines) pancreatic cancer cells, shRNA-induced MXRA5 silencing or CRISPR/Cas9-mediated MXRA5 knockout suppressed cell survival, proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT), while provoking cell apoptosis. Conversely, forced overexpression of MXRA5 further promoted pancreatic cancer cell progression and EMT. Bioinformatics studies and the protein chip analyses revealed that differentially expressed genes (DEGs) and differentially expressed proteins (DEPs) in MXRA5-overexpressed primary pancreatic cancer cells were enriched in the PI3K-Akt-mTOR cascade. Indeed, Akt-mTOR activation in primary human pancreatic cancer cells was inhibited by MXRA5 shRNA or knockout, but was augmented following MXRA5 overexpression. In vivo, the growth of MXRA5 KO PANC-1 xenografts was largely inhibited in nude mice. Moreover, intratumoral injection of adeno-associated virus-packed MXRA5 shRNA potently inhibited primary pancreatic cancer cell growth in nude mice. Akt-mTOR activation was also largely inhibited in the MXRA5-depleted pancreatic cancer xenografts. Contrarily MXRA5 overexpression promoted primary pancreatic cancer cell growth in nude mice. Together, overexpressed MXRA5 is important for pancreatic cancer cell growth possibly through promoting EMT and Akt-mTOR activation. MXRA5 could be a potential therapeutic oncotarget for pancreatic cancer.
Collapse
Affiliation(s)
- Shi-Qing Peng
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Xiao-Ren Zhu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Ming-Zhi Zhao
- Clinical Research Center of Neurological Disease and Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yi-Fan Zhang
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - An-Ran Wang
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Min-Bin Chen
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China.
| | - Zhen-Yu Ye
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
28
|
Chen M, Li Z, Gu C, Zheng H, Chen Y, Cheng L. Identification of G protein subunit alpha i2 as a promising therapeutic target of hepatocellular carcinoma. Cell Death Dis 2023; 14:143. [PMID: 36805440 PMCID: PMC9941495 DOI: 10.1038/s41419-023-05675-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 02/22/2023]
Abstract
Hepatocellular carcinoma (HCC) is a global health problem. Its incidence and mortality are increasing. Exploring novel therapeutic targets against HCC is important and urgent. We here explored the expression and potential function of Gαi2 (G protein subunit alpha i2) in HCC. The Cancer Genome Atlas Liver Hepatocellular Carcinoma (TCGA-LIHC) database shows that the number of Gαi2 transcripts in HCC tissues is significantly higher than that in the normal liver tissues. Moreover, Gαi2 overexpression in HCC correlates with poor prognosis of the patients. Gαi2 mRNA and protein expression are also elevated in local HCC tissues and different human HCC cells. In patient-derived primary HCC cells and immortalized HepG2 cells, Gαi2 silencing (by targeted shRNA) or knockout (KO, by the dCas9-sgRNA method) largely suppressed cell proliferation and motility, while inducing cell cycle arrest and caspase-apoptosis activation. Moreover, Gαi2 silencing or KO-induced reactive oxygen species (ROS) production and oxidative injury in primary and HepG2 HCC cells. Whereas different antioxidants ameliorated Gαi2-shRNA-induced anti-HCC cell activity. Using a lentiviral construct, Gαi2 overexpression further augmented proliferation and motility of primary and immortalized HCC cells. Further studies revealed that the binding between the transcription factor early growth response zinc finger transcription factor 1 (EGR1) and Gαi2 DNA promoter was significantly increased in HCC tissues and cells. In vivo, intratumoral injection of Gαi2 shRNA adeno-associated virus significantly hindered HCC xenograft growth in nude mice. Moreover, the growth of Gαi2-KO HCC xenografts in the nude mice was remarkably slow. Gαi2 depletion, oxidative injury, and apoptosis induction were detected in Gαi2-silenced or Gαi2-KO HCC xenografts. Together, overexpressed Gαi2 is required for HCC cell growth in vitro and in vivo, representing as a novel and promising diagnosis marker and therapeutic target of HCC.
Collapse
Affiliation(s)
- Minbin Chen
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, China
| | - Zhifei Li
- Department of Interventional and Vascular surgery, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou, China
| | - Chengtao Gu
- Department of Interventional and Vascular surgery, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou, China
| | - Hao Zheng
- Department of Interventional and Vascular surgery, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou, China
| | - Yan Chen
- General Surgery Department, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Long Cheng
- Department of Interventional and Vascular surgery, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou, China.
| |
Collapse
|
29
|
Mi YY, Ji Y, Zhang L, Sun CY, Wei BB, Yang DJ, Wan HY, Qi XW, Wu S, Zhu LJ. A first-in-class HBO1 inhibitor WM-3835 inhibits castration-resistant prostate cancer cell growth in vitro and in vivo. Cell Death Dis 2023; 14:67. [PMID: 36709328 PMCID: PMC9884225 DOI: 10.1038/s41419-023-05606-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/30/2023]
Abstract
The prognosis and overall survival of castration-resistant prostate cancer (CRPC) patients are poor. The search for novel and efficient anti-CRPC agents is therefore extremely important. WM-3835 is a cell-permeable, potent and first-in-class HBO1 (KAT7 or MYST2) inhibitor. Here in primary human prostate cancer cells-derived from CRPC patients, WM-3835 potently inhibited cell viability, proliferation, cell cycle progression and in vitro cell migration. The HBO1 inhibitor provoked apoptosis in the prostate cancer cells. It failed to induce significant cytotoxicity and apoptosis in primary human prostate epithelial cells. shRNA-induced silencing of HBO1 resulted in robust anti-prostate cancer cell activity as well, and adding WM-3835 failed to induce further cytotoxicity in the primary prostate cancer cells. Conversely, ectopic overexpression of HBO1 further augmented primary prostate cancer cell proliferation and migration. WM-3835 inhibited H3-H4 acetylation and downregulated several pro-cancerous genes (CCR2, MYLK, VEGFR2, and OCIAD2) in primary CRPC cells. Importantly, HBO1 mRNA and protein levels are significantly elevated in CRPC tissues and cells. In vivo, daily intraperitoneal injection of WM-3835 potently inhibited pPC-1 xenograft growth in nude mice, and no apparent toxicities detected. Moreover, intratumoral injection of HBO1 shRNA adeno-associated virus (AAV) suppressed the growth of primary prostate cancer xenografts in nude mice. H3-H4 histone acetylation and HBO1-dependent genes (CCR2, MYLK, VEGFR2, and OCIAD2) were remarkably decreased in WM-3835-treated or HBO1-silenced xenograft tissues. Together, targeting HBO1 by WM-3835 robustly inhibits CRPC cell growth.
Collapse
Affiliation(s)
- Yuan-Yuan Mi
- Department of Urology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yu Ji
- Department of Pathology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Lifeng Zhang
- Department of Urology, Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Chuan-Yu Sun
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Bing-Bing Wei
- Department of Urology, Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Dong-Jie Yang
- Department of Pathology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Hong-Yuan Wan
- Department of Urology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xiao-Wei Qi
- Department of Pathology, Affiliated Hospital of Jiangnan University, Wuxi, China.
| | - Sheng Wu
- Department of Urology, Affiliated Hospital of Jiangnan University, Wuxi, China.
| | - Li-Jie Zhu
- Department of Urology, Affiliated Hospital of Jiangnan University, Wuxi, China.
| |
Collapse
|
30
|
Wang Y, Liu F, Wu J, Zhang MQ, Chai JL, Cao C. G protein inhibitory α subunit 2 is a molecular oncotarget of human glioma. Int J Biol Sci 2023; 19:865-879. [PMID: 36778118 PMCID: PMC9909998 DOI: 10.7150/ijbs.79355] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 12/27/2022] [Indexed: 01/11/2023] Open
Abstract
Identification of novel therapeutic oncotargets for human glioma is extremely important. Here we tested expression, potential functions and underlying mechanisms of G protein inhibitory α subunit 2 (Gαi2) in glioma. Bioinformatics analyses revealed that Gαi2 expression is significantly elevated in human glioma, correlating with poor patients' survival, higher tumor grade and wild-type IDH status. Moreover, increased Gαi2 expression was also in local glioma tissues and different glioma cells. In primary and immortalized (A172) glioma cells, Gαi2 shRNA or knockout (KO, by Cas9-sgRNA) potently suppressed viability, proliferation, and mobility, and induced apoptosis. Ectopic Gαi2 overexpression, using a lentiviral construct, further augmented malignant behaviors in glioma cells. p65 phosphorylation, NFκB activity and expression of NFκB pathway genes were decreased in Gαi2-depleted primary glioma cells, but increased following Gαi2 overexpression. There was an increased binding between Gαi2 promoter and Sp1 (specificity protein 1) transcription factor in glioma tissues and different glioma cells. In primary glioma cells Gαi2 expression was significantly reduced following Sp1 silencing, KO or inhibition. In vivo studies revealed that Gαi2 shRNA-expressing AAV intratumoral injection hindered growth of subcutaneous glioma xenografts in nude mice. Moreover, Gαi2 KO inhibited intracranial glioma xenograft in nude mice. Gαi2 depletion, NFκB inhibition and apoptosis induction were observed in subcutaneous and intracranial glioma xenografts with Gαi2 depletion. Together, overexpressed Gαi2 is important for glioma cell growth possibly by promoting NFκB cascade activation.
Collapse
Affiliation(s)
- Yin Wang
- Institute of Neuroscience, Soochow University, Institute for Excellence in Clinical Medicine of Kunshan First People's Hospital and Soochow University, Suzhou, China
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Fang Liu
- Department of Neurosurgery, The affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Jiang Wu
- Department of Neurosurgery, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Mei-qing Zhang
- Institute of Neuroscience, Soochow University, Institute for Excellence in Clinical Medicine of Kunshan First People's Hospital and Soochow University, Suzhou, China
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Jin-long Chai
- Institute of Neuroscience, Soochow University, Institute for Excellence in Clinical Medicine of Kunshan First People's Hospital and Soochow University, Suzhou, China
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Cong Cao
- Institute of Neuroscience, Soochow University, Institute for Excellence in Clinical Medicine of Kunshan First People's Hospital and Soochow University, Suzhou, China
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
31
|
Li Y, Chai JL, Shi X, Feng Y, Li JJ, Zhou LN, Cao C, Li KR. Gαi1/3 mediate Netrin-1-CD146-activated signaling and angiogenesis. Theranostics 2023; 13:2319-2336. [PMID: 37153740 PMCID: PMC10157725 DOI: 10.7150/thno.80749] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 04/08/2023] [Indexed: 05/10/2023] Open
Abstract
Netrin-1 binds to the high-affinity receptor CD146 to activate downstream signaling and angiogenesis. Here, we examine the role and underlying mechanisms of G protein subunit alpha i1 (Gαi1) and Gαi3 in Netrin-1-induced signaling and pro-angiogenic activity. In mouse embryonic fibroblasts (MEFs) and endothelial cells, Netrin-1-induced Akt-mTOR (mammalian target of rapamycin) and Erk activation was largely inhibited by silencing or knockout of Gαi1/3, whereas signaling was augmented following Gαi1/3 overexpression. Netrin-1 induced Gαi1/3 association with CD146, required for CD146 internalization, Gab1 (Grb2 associated binding protein 1) recruitment and downstream Akt-mTOR and Erk activation. Netrin-1-induced signaling was inhibited by CD146 silencing, Gab1 knockout, or Gαi1/3 dominant negative mutants. Netrin-1-induced human umbilical vein endothelial cell (HUVEC) proliferation, migration and tube formation were inhibited by Gαi1/3 short hairpin RNA (shRNA), but were potentiated by ectopic Gαi1/3 overexpression. In vivo, intravitreous injection of Netrin-1 shRNA adeno-associated virus (AAV) significantly inhibited Akt-mTOR and Erk activation in murine retinal tissues and reduced retinal angiogenesis. Endothelial knockdown of Gαi1/3 significantly inhibited Netrin1-induced signaling and retinal angiogenesis in mice. Netrin-1 mRNA and protein expression were significantly elevated in retinal tissues of diabetic retinopathy (DR) mice. Importantly, silence of Netrin-1, by intravitreous Netrin-1 shRNA AAV injection, inhibited Akt-Erk activation, pathological retinal angiogenesis and retinal ganglion cells degeneration in DR mice. Lastly, Netrin-1 and CD146 expression is significantly increased in the proliferative retinal tissues of human proliferative diabetic retinopathy patients. Together, Netrin-1 induces CD146-Gαi1/3-Gab1 complex formation to mediate downstream Akt-mTOR and Erk activation, important for angiogenesis in vitro and in vivo.
Collapse
Affiliation(s)
- Ya Li
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University and North District, The Municipal Hospital of Suzhou, Gusu School, Nanjing Medical University, Suzhou, China
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Jin-long Chai
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University and North District, The Municipal Hospital of Suzhou, Gusu School, Nanjing Medical University, Suzhou, China
| | - Xin Shi
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University and North District, The Municipal Hospital of Suzhou, Gusu School, Nanjing Medical University, Suzhou, China
| | - Yu Feng
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jia-jun Li
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Li-na Zhou
- Department of Radiotherapy and Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, China
| | - Cong Cao
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University and North District, The Municipal Hospital of Suzhou, Gusu School, Nanjing Medical University, Suzhou, China
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
- ✉ Corresponding authors: Prof. Cong Cao, Clinical Research Center of Neurological Disease of the Second Affiliated Hospital of Soochow University.199Ren-ai Road, Suzhou, Jiangsu 215123, China. E-mail: . Prof. Ke-ran Li, The Affiliated Eye Hospital, Nanjing Medical University,138 Hanzhong Rd, Nanjing, Jiangsu, 210029, China. E-mail:
| | - Ke-ran Li
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
- ✉ Corresponding authors: Prof. Cong Cao, Clinical Research Center of Neurological Disease of the Second Affiliated Hospital of Soochow University.199Ren-ai Road, Suzhou, Jiangsu 215123, China. E-mail: . Prof. Ke-ran Li, The Affiliated Eye Hospital, Nanjing Medical University,138 Hanzhong Rd, Nanjing, Jiangsu, 210029, China. E-mail:
| |
Collapse
|
32
|
Lu J, Liu Q, Zhu L, Liu Y, Zhu X, Peng S, Chen M, Li P. Endothelial cell-specific molecule 1 drives cervical cancer progression. Cell Death Dis 2022; 13:1043. [PMID: 36522312 PMCID: PMC9755307 DOI: 10.1038/s41419-022-05501-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 12/04/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022]
Abstract
The expression, biological functions and underlying molecular mechanisms of endothelial cell-specific molecule 1 (ESM1) in human cervical cancer remain unclear. Bioinformatics analysis revealed that ESM1 expression was significantly elevated in human cervical cancer tissues, correlating with patients' poor prognosis. Moreover, ESM1 mRNA and protein upregulation was detected in local cervical cancer tissues and various cervical cancer cells. In established and primary cervical cancer cells, ESM1 shRNA or CRISPR/Cas9-induced ESM1 KO hindered cell proliferation, cell cycle progression, in vitro cell migration and invasion, and induced significant apoptosis. Whereas ESM1 overexpression by a lentiviral construct accelerated proliferation and migration of cervical cancer cells. Further bioinformatics studies and RNA sequencing data discovered that ESM1-assocaited differentially expressed genes (DEGs) were enriched in PI3K-Akt and epithelial-mesenchymal transition (EMT) cascades. Indeed, PI3K-Akt cascade and expression of EMT-promoting proteins were decreased after ESM1 silencing in cervical cancer cells, but increased following ESM1 overexpression. Further studies demonstrated that SYT13 (synaptotagmin 13) could be a primary target gene of ESM1. SYT13 silencing potently inhibited ESM1-overexpression-induced PI3K-Akt cascade activation and cervical cancer cell migration/invasion. In vivo, ESM1 knockout hindered SiHa cervical cancer xenograft growth in mice. In ESM1-knockout xenografts tissues, PI3K-Akt inhibition, EMT-promoting proteins downregulation and apoptosis activation were detected. In conclusion, overexpressed ESM1 is important for cervical cancer growth in vitro and in vivo, possibly by promoting PI3K-Akt activation and EMT progression. ESM1 represents as a promising diagnostic marker and potential therapeutic target of cervical cancer.
Collapse
Affiliation(s)
- Jingjing Lu
- grid.452273.50000 0004 4914 577XDepartment of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Qin Liu
- grid.452273.50000 0004 4914 577XDepartment of Gynaecology and Obstetrics, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Lixia Zhu
- grid.452273.50000 0004 4914 577XDepartment of Gynaecology and Obstetrics, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Yuanyuan Liu
- grid.452273.50000 0004 4914 577XClinical Research and Lab Center, Affiliated Kunshan Hospital of Jiangsu University, 215300 Kunshan, China
| | - Xiaoren Zhu
- grid.452273.50000 0004 4914 577XDepartment of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Shiqing Peng
- grid.452273.50000 0004 4914 577XDepartment of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Minbin Chen
- grid.452273.50000 0004 4914 577XDepartment of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Ping Li
- grid.452273.50000 0004 4914 577XDepartment of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| |
Collapse
|
33
|
Wei X, Zhuang P, Liu K, Hou W, Zhao Y, Wei W, Tu R, Li H, Dai H. Mesoporous bioglass capsule composite injectable hydrogels with antibacterial and vascularization promotion properties for chronic wound repair. J Mater Chem B 2022; 10:10139-10149. [PMID: 36472313 DOI: 10.1039/d2tb01777f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Building an angiogenesis microenvironment and inhibiting wound infection are of great significance for chronic wound repair. In this paper, polydopamine-encapsulated mesoporous bioglass (MBG@PDA) capsules were constructed to realize the integration of angiogenesis and infection inhibition through the formation of a composite hydrogel with modified hyaluronic acid (HAMA) to promote wound healing. The experiments showed that the composite hydrogel had good adhesion and toughness and promoted the migration of fibroblasts to accelerate the epithelialization process. In addition, in the composite hydrogel, MBG@PDA could release Mg2+ to promote the proliferation and migration of vascular endothelial cells for angiogenesis. At the same time, MBG@PDA in the composite hydrogel could facilitate the long-term release of drugs to inhibit the growth of Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus) for reducing the possibility of wound infection. Finally, the results of in vivo experiments showed that a multifunctional dressing could repair wounds more quickly by promoting angiogenesis and reducing the pathological areas. In summary, the construction of these composite hydrogels can provide a repair method in the wound-repair field.
Collapse
Affiliation(s)
- Xuejie Wei
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China.
| | - Pengzhen Zhuang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China.
| | - Kun Liu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China.
| | - Wen Hou
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China.
| | - Yanan Zhao
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China.
| | - Wenying Wei
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China.
| | - Rong Tu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China.
| | - Haiwen Li
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China.
| | - Honglian Dai
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China. .,Foshan Xianhu Laboratory of the Advanced Energy Science and Technology Guangdong Laboratory, Xianhu Hydrogen Valley, Foshan 528200, China
| |
Collapse
|
34
|
Guo YZ, Chen G, Huang M, Wang Y, Liu YY, Jiang Q, Cao C, Liu F. TIMM44 is a potential therapeutic target of human glioma. Theranostics 2022; 12:7586-7602. [PMID: 36438483 PMCID: PMC9691352 DOI: 10.7150/thno.78616] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 10/23/2022] [Indexed: 11/24/2022] Open
Abstract
TIMM44 (translocase of inner mitochondrial membrane 44) is essential for the maintenance of mitochondrial functions. Bioinformatics studies and results from the local high-grade glioma tissues showed that TIMM44 mRNA and protein levels are elevated in glioma, correlating with poor overall survival. Mitochondrial TIMM44 upregulation was also detected in patient-derived primary glioma cells and immortalized cell line. In primary and established glioma cells, TIMM44 depletion, using the lentiviral shRNA strategy or the CRISPR/Cas9 knockout (KO) method, robustly inhibited cell viability, proliferation and migration. Moreover, TIMM44 silencing/KO resulted in mitochondrial complex I inhibition, ATP depletion, mitochondrial membrane potential reduction, oxidative stress and DNA damage, and eventually provoked apoptosis. Conversely, ectopic overexpression of TIMM44 augmented glioma cell proliferation and migration. TIMM44 upregulation in glioma is possibly due to increased TIMM44 transcriptional machinery by the transcription factor GATA3 in a YME1L (YME1 Like 1 ATPase)-dependent manner. In vivo, the growth of subcutaneous glioma xenografts was suppressed after intratumoral injection of TIMM44 shRNA adeno-associated virus (AAV). TIMM44 depletion, ATP reduction, oxidative injury and apoptosis were detected in TIMM44 shRNA AAV-injected glioma xenografts. Moreover, the intracranial growth of TIMM44 KO glioma cells in the mouse brain was largely inhibited. Together, overexpressed TIMM44 could be a novel and promising therapeutic target of human glioma.
Collapse
Affiliation(s)
- Yi-zhuo Guo
- Institute of Neuroscience, Soochow University, Suzhou, and Department of Neurosurgery, The affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Gang Chen
- Department of Neurosurgery, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Man Huang
- Department of Oncology, Dushu Lake Hospital Affiliated of Soochow University, Suzhou, China
| | - Yin Wang
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Yuan-yuan Liu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Qin Jiang
- The Fourth School of Clinical Medicine, The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Cong Cao
- Institute of Neuroscience, Soochow University, Suzhou, and Department of Neurosurgery, The affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
- The Fourth School of Clinical Medicine, The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Fang Liu
- Institute of Neuroscience, Soochow University, Suzhou, and Department of Neurosurgery, The affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| |
Collapse
|
35
|
Zhang J, Yin DP, Zhang Y, Zhang JN, Yang Y, Zhang ZQ, Zhou L, Lv Y, Huang HW, Cao C. Identification of Gαi3 as a novel molecular therapeutic target of cervical cancer. Int J Biol Sci 2022; 18:5667-5680. [PMID: 36263185 PMCID: PMC9576524 DOI: 10.7150/ijbs.77126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 08/25/2022] [Indexed: 01/12/2023] Open
Abstract
Here we studied expression and potential functions of Gαi3 in cervical cancer. The bioinformatics analysis together with the results from local patients' tissues revealed that Gαi3 expression was remarkably elevated in human cervical cancer tissues and different cervical cancer cells, and was associated with poor overall survival and poor disease-specific survival of patients. Gαi3 depletion resulted in profound anti-cervical cancer activity. In primary or immortalized cervical cancer cells, Gαi3 shRNA or CRISPR/Cas9-caused Gαi3 knockout/KO largely hindered cell proliferation and migration, and provoked apoptosis. On the contrast, ectopic Gαi3 overexpression further enhanced cervical cancer proliferation and migration. Akt-mTOR activation in primary cervical cancer cells was significantly reduced after Gαi3 silencing or KO, but was augmented following Gαi3 overexpression. Further studies revealed that the transcription factor GATA4 binding to Gαi3 promoter region was significantly enhanced in cervical cancer tissues and cells. Gαi3 expression was decreased by GATA4 shRNA, but upregulated following GATA4 overexpression. In vivo, the growth of cervical cancer xenografts was robustly suppressed after Gαi3 silencing or KO. Gαi3 depletion and Akt-mTOR inactivation were detected in Gαi3-silenced/-KO cervical cancer xenograft tissues. Together, upregulated Gαi3 is a valuable oncotarget of cervical cancer.
Collapse
Affiliation(s)
- Jie Zhang
- Obstetrics and Gynecology Department, The Affiliated Zhangjiagang Hospital of Soochow University, Institute of Neuroscience, Soochow University, Suzhou, China
| | - De-pei Yin
- Department of Otorhinolaryngology Head and Neck Surgery, Children's Hospital of Soochow University, Suzhou, China
| | - Yan Zhang
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, China
| | - Jia-nan Zhang
- Obstetrics and Gynecology Department, The Affiliated Zhangjiagang Hospital of Soochow University, Institute of Neuroscience, Soochow University, Suzhou, China
| | - Yan Yang
- Obstetrics and Gynecology Department, The Affiliated Zhangjiagang Hospital of Soochow University, Institute of Neuroscience, Soochow University, Suzhou, China
| | - Zhi-qing Zhang
- Obstetrics and Gynecology Department, The Affiliated Zhangjiagang Hospital of Soochow University, Institute of Neuroscience, Soochow University, Suzhou, China
| | - Li Zhou
- Center of Translational Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China
| | - Yan Lv
- Center of Translational Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China
| | - Hai-wei Huang
- Obstetrics and Gynecology Department, The Affiliated Zhangjiagang Hospital of Soochow University, Institute of Neuroscience, Soochow University, Suzhou, China
| | - Cong Cao
- Obstetrics and Gynecology Department, The Affiliated Zhangjiagang Hospital of Soochow University, Institute of Neuroscience, Soochow University, Suzhou, China
| |
Collapse
|