1
|
Gao T, Magnano S, Quadros M, Barroeta PH, Zisterer DM. The pan-BH-3 mimetic, obatoclax, synergistically enhances cisplatin-induced apoptosis in oral squamous cell carcinoma through a mechanism that involves degradation of the pro-survival protein Mcl-1. Arch Oral Biol 2025; 174:106250. [PMID: 40194355 DOI: 10.1016/j.archoralbio.2025.106250] [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: 12/16/2024] [Revised: 03/29/2025] [Accepted: 03/31/2025] [Indexed: 04/09/2025]
Abstract
OBJECTIVES The purpose of the study was to elucidate the role of the Bcl-2 family of proteins in mediating cisplatin resistance in oral squamous cell carcinoma (OSCC). The value of the BH3-mimetics venetoclax and obatoclax as sensitisers for cisplatin treatment in OSCC was also evaluated. DESIGN In this study the expression levels of a series of pro- and anti-apoptotic members of the Bcl-2 family in paired cisplatin-sensitive (SCC4) and resistant (SCC4cisR) tongue squamous carcinoma cell lines were examined by western blotting. The apoptotic rate induced by cisplatin and BH3-mimetics venetoclax and obatoclax alone or in combination in OSCC was also evaluated by Annexin V/Propidium Iodide double-stained flow cytometric assays. RESULTS Obatoclax was shown to synergistically enhance cisplatin-induced apoptosis, and this enhancement was associated with a marked degradation in pro-survival Mcl-1 and upregulation in conformationally active form of pro-apoptotic Bak. CONCLUSIONS Our study presents novel insights into the relationship between the Bcl-2 family and cisplatin efficacy in OSCC. It also demonstrates that targeted therapy with BH-3 mimetics, such as obatoclax, may represent a new strategy for OSCC therapy.
Collapse
Affiliation(s)
- Tianyi Gao
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
| | - Stefania Magnano
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Molly Quadros
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Patricia Hannon Barroeta
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Daniela M Zisterer
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
2
|
Zeng H, Zeng X, Wang C, Wang G, Tian Q, Zhao J, Zhao L, Li R, Luo Y, Peng H, Zhang Z, Li X, Wu X. Combination therapy using Cel-CSO/Taxol NPs for reversing drug resistance in breast cancer through inhibiting PI3K/AKT/NF-κB/HIF-1α pathway. Drug Deliv Transl Res 2025; 15:992-1010. [PMID: 38922561 DOI: 10.1007/s13346-024-01653-3] [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] [Accepted: 06/13/2024] [Indexed: 06/27/2024]
Abstract
The resistance of malignant tumors to multiple drugs is a significant obstacle in cancer treatment and prognosis. Accordingly, we synthesized a celastrol (Cel) prodrug (Cel-CSO) by conjugating chitosan oligosaccharides (CSO) to Cel for reversing Taxol resistance in chemotherapy, followed by self-assembly with Taxol into a novel nanoplatform of Cel-CSO/Taxol nanoparticles (termed NPs). NPs showed a suitable size (about 153 nm), excellent stability and prolonged release of Cel and Taxol in a manner that depended on both pH and time. NPs effectively inhibited the overexpression of multidrug resistance-related protein P-gp, hypoxia inducible factor-1α (HIF-1α), and triggered the MCF-7/Taxol cell apoptosis through inhibiting the PI3K/AKT/NF-κB/HIF-1α pathway. In tumor-bearing mice, NPs exhibited significant curative effects in inducing apoptosis of MCF-7/Taxol tumors which showed a low expression level of P-gp, microtubule-related proteins TUBB3 and Tau. The results indicated that NPs may be a promising strategy to overcome drug resistance caused by P-gp, which improve the antitumor effects in drug-resistant breast cancer.
Collapse
Affiliation(s)
- Huahui Zeng
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, 450046, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou, 450046, China
| | - Xiaohu Zeng
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Can Wang
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, 450046, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou, 450046, China
| | - Guoqiang Wang
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Qikang Tian
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Junwei Zhao
- Department of Clinical Laboratory, Core Unit of National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450046, China
| | - Lingzhou Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Ruiqin Li
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Ying Luo
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou, 450046, China
| | - Haotian Peng
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Zhenqiang Zhang
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| | - Xiaofang Li
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| | - Xiangxiang Wu
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| |
Collapse
|
3
|
Zhou C, Wu K, Gu M, Yang Y, Tu J, Huang X. Reversal of chemotherapy resistance in gastric cancer with traditional Chinese medicine as sensitizer: potential mechanism of action. Front Oncol 2025; 15:1524182. [PMID: 40052129 PMCID: PMC11882405 DOI: 10.3389/fonc.2025.1524182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025] Open
Abstract
Gastric cancer (GC) remains one of the most common types of cancer, ranking fifth among cancer-related deaths worldwide. Chemotherapy is an effective treatment for advanced GC. However, the development of chemotherapy resistance, which involves the malfunction of several signaling pathways and is the consequence of numerous variables interacting, seriously affects patient treatment and leads to poor clinical outcomes. Therefore, in order to treat GC, it is imperative to find novel medications that will increase chemotherapy sensitivity and reverse chemotherapy resistance. Traditional Chinese medicine (TCM) has been extensively researched as an adjuvant medication in recent years. It has been shown to have anticancer benefits and to be crucial in enhancing chemotherapy sensitivity and reducing chemotherapy resistance. Given this, the mechanism of treatment resistance in GC is summed up in this work. The theoretical foundation for TCM as a sensitizer in adjuvant treatment of GC is established by introducing the primary signal pathways and possible targets implicated in improving chemotherapy sensitivity and reversing chemotherapy resistance of GC by TCM and active ingredients.
Collapse
Affiliation(s)
| | | | | | | | | | - Xuan Huang
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese
Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
4
|
Jia Y, Zhang L, Zhou W, Chen S, Zhang H, Liu L, Guo H, Wang Z, Cui Y, Niu R, Zhang F. Rack1 promotes breast cancer stemness properties and tumorigenesis through the E2F1-SOX2 axis. Cancer Cell Int 2025; 25:40. [PMID: 39939870 PMCID: PMC11823217 DOI: 10.1186/s12935-025-03678-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 02/04/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Breast cancer remains the most prevalent malignancy and the leading cause of cancer-related mortality among women worldwide. The primary factors contributing to the deterioration and death of patients with breast cancer are metastasis, recurrence, and drug resistance. These phenomena are closely related to the presence of breast cancer stem cells; however, the exact mechanisms regulating stemness remain to be elucidated. Rack1 (Receptor for Activated C Kinase 1), a well-known versatile scaffold protein, has been implicated in tumorigenesis and progression in numerous cancer types; however, its specific role in breast cancer stemness remains to be elucidated. METHODS Using bioinformatic and immunohistochemical approaches, we validated that the expression level of Rack1 is associated with cancer stemness and affects the prognosis of patients. Through a series of experimental methods including mammosphere formation assay, flow cytometry, qPCR, Western blotting, and CHX assays, we validated at the molecular and cellular levels the mechanism by which Rack1 influences cancer stemness via the E2F1/SOX2 axis. Furthermore, by designing and utilizing lentiviral constructs to establish xenograft tumor models in mice, we further confirmed in vivo the impact of the Rack1/E2F1/SOX2 axis on the tumorigenic capacity of breast cancer cells. RESULTS Our findings indicate that Rack1 plays a critical role in preserving the stemness characteristics of breast cancer cells. Mechanistically, the observed effects of Rack1 are achieved through the modulation of SOX2 expression, a master transcription factor that regulates cancer cell stemness and maintenance. We further demonstrate that Rack1 increases the stability of the E2F1 protein by inhibiting its ubiquitination and subsequent proteasome-mediated degradation, which in turn transcriptionally upregulates SOX2, thereby maintaining breast cancer cell stemness and tumorigenesis. CONCLUSION This study thus unveils a novel mechanism through which Rack1 executes its oncogenic function. This study also demonstrates that targeting the Rack1-E2F-SOX2 axis may be a potential strategy to inhibit breast cancer development and progression.
Collapse
Affiliation(s)
- Yidi Jia
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Luoming Zhang
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Wei Zhou
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Shuhua Chen
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - He Zhang
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Liming Liu
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Hui Guo
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Zhiyong Wang
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Yanfen Cui
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Ruifang Niu
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, China.
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China.
| | - Fei Zhang
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, China.
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China.
| |
Collapse
|
5
|
Salem MM, Gerges MN, Abd El Salam HA, Noser AA. New thiadiazolopyrimidine-ornamented pyrazolones as prospective anticancer candidates via suppressing VEGFR-2/PI3K/Akt signaling pathway: Synthesis, characterization, in-silico, and in-vitro studies. Int J Biol Macromol 2025; 289:138735. [PMID: 39689791 DOI: 10.1016/j.ijbiomac.2024.138735] [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: 11/12/2024] [Revised: 12/07/2024] [Accepted: 12/11/2024] [Indexed: 12/19/2024]
Abstract
New thiadiazolopyrimidine-ornamented pyrazolones (4a-8b) have been synthesized by a cyclocondensation reaction of 3a, b with different active methylene compounds. The structure of our products was confirmed via different physical and spectroscopic data. We assessed all newly thiadiazolopyrimidine-ornamented pyrazolones' potential to inhibit angiogenesis, metastasis, and cancer growth by utilizing in-silico investigations focused on the VEGFR-2 signaling pathway and elucidate their pharmacokinetic features using ADMET. Based on our results, compound 8b was chosen for in vitro evaluation since it had the highest binding energy of -10.252 kcal/mol using molecular docking. Compound 8b significantly damaged the T47D (IC50 = 33.01 ± 2.2 μM) cells, without any toxic effect on normal cells in comparison to chemotheraputic FDA approved drug cisplatin (Cis) (IC50 = 3.163 ± 1.7 μM). Additionally, compound 8b significantly suppressed the VEGFR-2 receptor protein that triggers the inhibition of PI3K/Akt genes which causes mitochondrial membrane malfunction resulting in Bax overexpression and Bcl-2 downregulation levels. Besides, compound 8b showed a notable decrease in the levels of nitric oxide (NO) production levels and arrested the cell cycle in the G0/G1 stage. These outcomes demonstrated that compound 8b adhered to Lipinski's rules and may serve as a potential candidate for future breast cancer treatments via obstructing the VEGFR-2/PI3K/Akt signaling pathway, which in turn prevents metastasis, angiogenesis, and proliferation.
Collapse
Affiliation(s)
- Maha M Salem
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta 31527, Egypt.
| | - Marian N Gerges
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta 31527, Egypt.
| | - Hayam A Abd El Salam
- Green Chemistry Department, National Research Centre, Dokki, Giza 12622, Cairo, Egypt
| | - Ahmed A Noser
- Organic Chemistry, Chemistry Department, Faculty of Science, Tanta University, Tanta 31527, Egypt.
| |
Collapse
|
6
|
Zhang X, Li Z, Wang T. Etomidate suppresses proliferation, migration, invasion, and glycolysis in esophageal cancer cells via PI3K/AKT pathway inhibition. Cytotechnology 2025; 77:4. [PMID: 39583284 PMCID: PMC11579264 DOI: 10.1007/s10616-024-00661-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: 07/02/2024] [Accepted: 10/26/2024] [Indexed: 11/26/2024] Open
Abstract
Esophageal cancer remains a formidable challenge in oncology, characterized by its poor prognosis and limited therapeutic options. Recent investigations have unveiled the potential of repurposing existing drugs for cancer treatment. Notably, etomidate, an anesthetic agent traditionally used for inducing general anesthesia, has emerged as a promising candidate demonstrating significant anticancer properties across various tumor types. The present study aims to investigate the effects of etomidate on esophageal carcinoma cells, with a specific focus on its ability to modulate the PI3K/AKT signaling pathway and inhibit tumor proliferation. This study employed both in vitro and in vivo methodologies to assess the effects of etomidate on esophageal cancer cells. In vitro experiments evaluated the effects of etomidate on cell proliferation, migration, invasion, and glycolytic processes. An in vivo xenograft mouse model was established to investigate the therapeutic potential of etomidate on tumor growth and assess its impact on the PI3K/AKT signaling pathway in a physiologically relevant context. Etomidate demonstrated a significant inhibitory effect on the proliferation, migration, invasion, and glycolytic capacity of esophageal cancer cells. This multifaceted suppression of tumorigenic properties was closely associated with the inhibition of the PI3K/AKT pathway, as evidenced by reduced phosphorylation levels of PI3K and AKT. In vivo studies using a murine model of esophageal cancer corroborated these findings. Etomidate administration resulted in a substantial reduction in tumor volume and mass, accompanied by increased apoptotic activity and the inhibition of the PI3K/AKT pathway within the tumor tissue. This study demonstrates etomidate's potent inhibition of esophageal cancer progression through suppression of the PI3K/AKT pathway. These promising results warrant further clinical investigation of etomidate as a potential therapeutic strategy for esophageal cancer. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-024-00661-y.
Collapse
Affiliation(s)
- Xiangchao Zhang
- Department of Anesthesiology, Shenyang Chest Hospital, No. 11 Beihai Street, Dadong District, Shenyang City, 110044 Liaoning China
| | - Zhengjun Li
- Department of Thoracic Surgery, Shenyang Chest Hospital, No. 11 Beihai Street, Dadong District, Shenyang City, 110044 Liaoning China
| | - Tao Wang
- Department of Anesthesiology, Shenyang Chest Hospital, No. 11 Beihai Street, Dadong District, Shenyang City, 110044 Liaoning China
| |
Collapse
|
7
|
Zheng SX, Chen JP, Liang RS, Zhuang BB, Wang CH, Zhang GL, Shi SS, Chen J. Schizophyllum commune fruiting body polysaccharides inhibit glioma by mediating ARHI regulation of PI3K/AKT signalling pathway. Int J Biol Macromol 2024; 279:135326. [PMID: 39236963 DOI: 10.1016/j.ijbiomac.2024.135326] [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: 11/24/2023] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/07/2024]
Abstract
Glioma poses a serious threat to human health and has a high mortality rate. Therefore, developing natural anti-tumour drugs for cancer treatment is an urgent priority. Schizophyllum commune is an edible and medicinal fungus, with polysaccharides as its main active components, which may have anti-tumour properties. Herein, we characterised S. commune fruiting body polysaccharides (SCFP) structure and evaluated its anti-glioma activity in vitro and in vivo. UV and FTIR spectra, high-performance gel chromatography, and monosaccharide composition analyses demonstrated that SCFP was a heteropolysaccharide with a molecular weight of 290.92 kDa. Among the monosaccharide compositions, mannose, galactose, and glucose were the most abundant. SCFP significantly inhibited the survival of the glioma cell lines U251 and U-87MG. U251 xenograft tumours treated with SCFP via gavage showed a 47.39 % inhibition, with no significant toxic side effects observed. SCFP upregulated aplasia Ras homologue member I (ARHI) expression, thereby regulating PI3K/AKT signalling, inhibiting tumour migration, and inducing apoptosis, to inhibit tumour growth. Furthermore, SCFP treatment increased the relative abundance of beneficial bacteria, including Akkermansia muciniphila, Ligilactobacillus murinus, and Parabacteroides goldsteinii, in tumour-bearing mice and restored the gut microbiota structure to that of the normal group (NG group) mice without tumours. Thus, SCFP has the potential for application as a natural anticancer drug.
Collapse
Affiliation(s)
- Shi-Xing Zheng
- Department of Neurosurgery, Fujian Medical University Union Hospital, 29# Xinquan Road, Fuzhou 350001, Fujian, China
| | - Jian-Ping Chen
- Department of Neurosurgery, Fujian Medical University Union Hospital, 29# Xinquan Road, Fuzhou 350001, Fujian, China
| | - Ri-Sheng Liang
- Department of Neurosurgery, Fujian Medical University Union Hospital, 29# Xinquan Road, Fuzhou 350001, Fujian, China
| | - Bing-Bo Zhuang
- Department of Neurosurgery, Fujian Medical University Union Hospital, 29# Xinquan Road, Fuzhou 350001, Fujian, China
| | - Chun-Hua Wang
- Fujian Institute of Neurosurgery, 29# Xinquan Road, Fuzhou 350001, Fujian, China
| | - Guo-Liang Zhang
- Department of Neurosurgery, Fujian Medical University Union Hospital, 29# Xinquan Road, Fuzhou 350001, Fujian, China
| | - Song-Sheng Shi
- Department of Neurosurgery, Fujian Medical University Union Hospital, 29# Xinquan Road, Fuzhou 350001, Fujian, China
| | - Jing Chen
- Department of Neurosurgery, Fujian Medical University Union Hospital, 29# Xinquan Road, Fuzhou 350001, Fujian, China.
| |
Collapse
|
8
|
Shen W, Zeng X, Zeng X, Hu B, Ren C, Lin Z, Zhang L, Rui G, Yasen M, Chen X. Trifluoperazine activates AMPK / mTOR / ULK1 signaling pathway to induce mitophagy in osteosarcoma cells. Chem Biol Interact 2024; 392:110904. [PMID: 38360085 DOI: 10.1016/j.cbi.2024.110904] [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: 12/09/2023] [Revised: 01/26/2024] [Accepted: 02/06/2024] [Indexed: 02/17/2024]
Abstract
Osteosarcoma is a prevalent kind of primary bone malignancy. Trifluoperazine, as an antipsychotic drug, has anti-tumor activity against a variety of cancers. Nevertheless, the impact of trifluoperazine on osteosarcoma is unclear. Our investigation aimed to explore the mechanism of trifluoperazine's effect on osteosarcoma. We found that trifluoperazine inhibited 143B and U2-OS osteosarcoma cell proliferation in a method based on the dose. Furthermore, it was shown that trifluoperazine induced the accumulation of reactive oxygen species (ROS) to cause mitochondrial damage and induced mitophagy in osteosarcoma cells. Finally, combined with RNA-seq results, we first demonstrated the AMPK/mTOR/ULK1 signaling pathway as a potential mechanism of trifluoperazine-mediated mitophagy in osteosarcoma cells and can be suppressed by AMPK inhibitor Compound C.
Collapse
Affiliation(s)
- Wenhui Shen
- Department of Orthopedic Surgery, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, 361000, China
| | - Xiangchen Zeng
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Xianhui Zeng
- Department of Infectious Diseases, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, 570206, China
| | - Baoshan Hu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, 361001, China
| | - Chong Ren
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Zhiming Lin
- Department of Orthopedic Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, 361001, China
| | - Long Zhang
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Gang Rui
- Department of Orthopedic Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, 361001, China
| | - Miersalijiang Yasen
- Department of Orthopedic Surgery, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, 361000, China.
| | - Xiaohui Chen
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Xiamen University, Xiamen, 361102, China; Department of Orthopedic Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, 361001, China.
| |
Collapse
|
9
|
Gao L, Li L, Zhang D, Qiu J, Qian J, Liu H. TAPI-1 Exhibits Anti-tumor Efficacy in Human Esophageal Squamous Cell Carcinoma Cells via Suppression of NF-κB Signaling Pathway. Dig Dis Sci 2024; 69:81-94. [PMID: 38007701 PMCID: PMC10787672 DOI: 10.1007/s10620-023-08181-z] [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: 04/21/2023] [Accepted: 11/02/2023] [Indexed: 11/28/2023]
Abstract
BACKGROUND TNF-α processing inhibitor-1 (TAPI-1) is a known metalloproteinase inhibitor with potential anti-inflammatory effects. However, its anti-cancer effects on esophageal squamous cell carcinoma (ESCC) have not been uncovered. AIM In the present study, the effects of TAPI-1 on ESCC cell viability, migration, invasion, and cisplatin resistance and the underlying molecular mechanisms were investigated in TE-1 and Eca109 cells. METHODS To this end, TE-1 and Eca109 cells were exposed to TAPI-1 for indicated time intervals. Cell viability was assessed using cell counting kit-8 assay and apoptosis was evaluated using flow cytometry assay. Migration and invasion were assessed using Transwell assays. Gene expressions were analyzed using quantitative reverse transcription polymerase chain reaction. The activation of NF-κB signaling pathway was elucidated via Western blot and chromatin immunoprecipitation assay. RESULTS We observed that higher doses (10, 20 μM) of TAPI-1 inhibited ESCC cell viability, while a lower dose (5 μM) of TAPI-1 inhibited ESCC cell migration and invasion and enhanced the chemosensitivity of ESCC cells to cisplatin. Moreover, TAPI-1 suppressed the activation of NF-κB signaling and the target genes expression in the stage of transcription initiation. Furthermore, blocking NF-κB signaling in advance could abolish all the effects of TAPI-1 on ESCC cells. CONCLUSION Overall, these results indicated that TAPI-1 impairs ESCC cell viability, migration, and invasion and facilitates cisplatin-induced apoptosis via suppression of NF-κB signaling pathway. TAPI-1 may serve as a potential adjuvant agent with cisplatin for ESCC therapy.
Collapse
Affiliation(s)
- Lin Gao
- Medical Research Center, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, 226001, People's Republic of China
| | - Li Li
- Department of Pathology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Shengli Road No. 666, Nantong, 226001, Jiangsu, People's Republic of China
| | - Dongmei Zhang
- Medical Research Center, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, 226001, People's Republic of China
| | - Jianwei Qiu
- Department of Gastroenterology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, 226001, People's Republic of China
| | - Junbo Qian
- Department of Gastroenterology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, 226001, People's Republic of China
| | - Hongbin Liu
- Department of Pathology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Shengli Road No. 666, Nantong, 226001, Jiangsu, People's Republic of China.
| |
Collapse
|
10
|
He HY, Shan HZ, Li SQ, Diao RG. Genistein attenuates renal ischemia-reperfusion injury via ADORA2A pathway. Hum Exp Toxicol 2023; 42:9603271231164913. [PMID: 36932924 DOI: 10.1177/09603271231164913] [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] [Indexed: 03/19/2023]
Abstract
BACKGROUND Studies have shown oxidative stress and apoptosis are the main pathogenic mechanisms of renal ischemia/reperfusion (IR) injury (IRI). Genistein, a polyphenolic non-steroidal compound, has been extensively explored in oxidative stress, inflammation and apoptosis. Our research aims to reveal the potential role of genistein on renal IRI and its potential molecular mechanism both in vivo and in vitro. METHODS In vivo experiments, mice were pretreated with or without genistein. Renal pathological changes and function, cell proliferation, oxidative stress and apoptosis were measured. In vitro experiments, overexpression of ADORA2A and knockout of ADORA2A cells were constructed. Cells proliferation, oxidative stress and apoptosis were analyzed. RESULTS Our results in vivo showed that the renal damage induced by IR was ameliorated by genistein pretreatment. Moreover, ADORA2A was activated by genistein, along with inhibition of oxidative stress and apoptosis. The results in vitro showed that genistein pretreatment and ADORA2A overexpression reversed the increase of apoptosis and oxidative stress in NRK-52E cells induced by H/R, while the knockdown of ADORA2A partially weakened this reversal from genistein treatment. CONCLUSIONS Our results demonstrated that genistein have a protective effect against renal IRI by inhibiting oxidative stress and apoptosis via activating ADORA2A, presenting its potential use for the treatment of renal IRI.
Collapse
Affiliation(s)
- H Y He
- Nephrology, 519688Yantaishan Hospital, Yantai, Shandong, China
| | - H Z Shan
- Department of Pharmacy, 155177Qingdao Traditional Chinese Medicine Hospital(Qingdao Hiser Hospital)Qingdao Hiser Hospital Affiliated of Qingdao University, Qingdao, Shandong, China
| | - S Q Li
- Department of Pharmacy, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - R G Diao
- Department of Pharmacy, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| |
Collapse
|
11
|
Miao X, Hu J, Chai C, Tang H, Zhao Z, Luo W, Zhou W, Xu H. Establishment and characterization of a new intrahepatic cholangiocarcinoma cell line derived from a Chinese patient. Cancer Cell Int 2022; 22:418. [PMID: 36578029 PMCID: PMC9795767 DOI: 10.1186/s12935-022-02840-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022] Open
Abstract
Patients with intrahepatic cholangiocarcinoma (ICC) require chemotherapy due to late detection, rapid disease progression, and low surgical resection rate. Tumor cell lines are extremely important in cancer research for drug discovery and development. Here, we established and characterized a new intrahepatic cholangiocarcinoma cell line, ICC-X1. STR testing confirmed the absence of cross-contamination and high similarity to the original tissue. ICC-X1 exhibited typical epithelial morphology and formed tumor spheres in the suspension culture. The population doubling time was approximately 48 h. The cell line had a complex hypotriploid karyotype. The cell line exhibited a strong migration ability in vitro and cell inoculation into BALB/c nude mice led to the formation of xenografts. Additionally, ICC-X1 cells were sensitive to gemcitabine and paclitaxel but resistant to 5-fluorouracil and oxaliplatin. RNA sequencing revealed that the upregulated cancer-related genes were mainly enriched in several signaling pathways, including the TNF signaling pathway, NOD-like receptor signaling pathway, and NF-κB signaling pathway. The downregulated cancer-related genes were mainly enriched in the Rap1 signaling pathway and Hippo signaling pathway among other pathways. In conclusion, we have created a new ICC cell line derived from Chinese patients. This cell line can be used as a preclinical model to study ICC, specifically tumor metastasis and drug resistance mechanisms.
Collapse
Affiliation(s)
- Xin Miao
- grid.410727.70000 0001 0526 1937State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Animal Virology of the Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000 China
| | - Jinjing Hu
- grid.412643.60000 0004 1757 2902The Forth Department of General Surgery, The First Hospital of Lanzhou University, No. 1, Donggang West Road, Lanzhou, 730000 Gansu China
| | - Changpeng Chai
- grid.412643.60000 0004 1757 2902The Forth Department of General Surgery, The First Hospital of Lanzhou University, No. 1, Donggang West Road, Lanzhou, 730000 Gansu China
| | - Huan Tang
- grid.412643.60000 0004 1757 2902The Forth Department of General Surgery, The First Hospital of Lanzhou University, No. 1, Donggang West Road, Lanzhou, 730000 Gansu China
| | - Zhenjie Zhao
- grid.412643.60000 0004 1757 2902The Forth Department of General Surgery, The First Hospital of Lanzhou University, No. 1, Donggang West Road, Lanzhou, 730000 Gansu China
| | - Wei Luo
- grid.412643.60000 0004 1757 2902The Forth Department of General Surgery, The First Hospital of Lanzhou University, No. 1, Donggang West Road, Lanzhou, 730000 Gansu China
| | - Wence Zhou
- grid.411294.b0000 0004 1798 9345Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou, 730000 China ,grid.32566.340000 0000 8571 0482The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000 China
| | - Hao Xu
- grid.412643.60000 0004 1757 2902The Forth Department of General Surgery, The First Hospital of Lanzhou University, No. 1, Donggang West Road, Lanzhou, 730000 Gansu China
| |
Collapse
|
12
|
Myostatin Mutation Enhances Bovine Myogenic Differentiation through PI3K/AKT/mTOR Signalling via Removing DNA Methylation of RACK1. Cells 2022; 12:cells12010059. [PMID: 36611855 PMCID: PMC9818849 DOI: 10.3390/cells12010059] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/26/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Myostatin (MSTN) is a negative regulator of skeletal muscle development and plays an important role in muscle development. Fluctuations in gene expression influenced by DNA methylation are critical for homeostatic responses in muscle. However, little is known about the mechanisms underlying this fluctuation regulation and myogenic differentiation of skeletal muscle. Here we report a genome-wide analysis of DNA methylation dynamics in bovine skeletal muscle myogenesis after myostatin editing. We show that, after myostatin editing, an increase in TETs (DNA demethylases) and a concomitant increase in the receptor for activated C kinase 1 (RACK1) control the myogenic development of skeletal muscle. Interestingly, enhancement of PI3K/AKT/mTOR signaling by RACK1 appears to be an essential driver of myogenic differentiation, as it was associated with an increase in myogenic differentiation marker factors (MyHC and MyoG) during muscle differentiation. Overall, our results suggest that loss of myostatin promotes the myogenic differentiation response in skeletal muscle by decreasing DNA methylation of RACK1.
Collapse
|
13
|
Qian S, Wei Z, Yang W, Huang J, Yang Y, Wang J. The role of BCL-2 family proteins in regulating apoptosis and cancer therapy. Front Oncol 2022; 12:985363. [PMID: 36313628 PMCID: PMC9597512 DOI: 10.3389/fonc.2022.985363] [Citation(s) in RCA: 340] [Impact Index Per Article: 113.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 09/22/2022] [Indexed: 11/27/2022] Open
Abstract
Apoptosis, as a very important biological process, is a response to developmental cues or cellular stress. Impaired apoptosis plays a central role in the development of cancer and also reduces the efficacy of traditional cytotoxic therapies. Members of the B-cell lymphoma 2 (BCL-2) protein family have pro- or anti-apoptotic activities and have been studied intensively over the past decade for their importance in regulating apoptosis, tumorigenesis, and cellular responses to anticancer therapy. Since the inflammatory response induced by apoptosis-induced cell death is very small, at present, the development of anticancer drugs targeting apoptosis has attracted more and more attention. Consequently, the focus of this review is to summarize the current research on the role of BCL-2 family proteins in regulating apoptosis and the development of drugs targeting BCL-2 anti-apoptotic proteins. Additionally, the mechanism of BCL-2 family proteins in regulating apoptosis was also explored. All the findings indicate the potential of BCL-2 family proteins in the therapy of cancer.
Collapse
Affiliation(s)
- Shanna Qian
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Zhong Wei
- Gastrointestinal Surgery, Anhui Provincial Hospital, Hefei, China
| | - Wanting Yang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Jinling Huang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Yinfeng Yang
- School of Medical Informatics Engineering, Anhui University of Chinese Medicine, Hefei, China
| | - Jinghui Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
14
|
Wang F, Bai J, Li F, Liu J, Wang Y, Li N, Wang Y, Xu J, Liu W, Xu L, Chen L. Investigation of the mechanism of the anti-cancer effects of Astragalus propinquus Schischkin and Pinellia pedatisecta Schott (A&P) on melanoma via network pharmacology and experimental verification. Front Pharmacol 2022; 13:895738. [PMID: 36034875 PMCID: PMC9411814 DOI: 10.3389/fphar.2022.895738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 07/15/2022] [Indexed: 11/13/2022] Open
Abstract
Melanoma is a commonly malignant cutaneous tumor in China. Astragalus propinquus Schischkin and Pinellia pedatisecta Schott (A&P) have been clinically used as adjunctive drugs in the treatment of malignant melanoma. However, the effect and mechanism of A&P on melanoma have yet to be explored. The current investigation seeks to characterize the active components of A&P and their potential roles in treating malignant melanoma using network pharmacology and in vitro and in vivo experiments. We first used the traditional Chinese medicine systems pharmacology (TCMSP) database and high-performance liquid chromatography-mass spectrometry (HPLC-MS/MS) to identify a total of 13 effective compounds within A&P. 70 common genes were obtained by matching 487 potential genes of A&P with 464 melanoma-related genes, and then we built up protein-protein interaction (PPI) network of these 70 genes, followed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. The results revealed that A&P might influence the pathobiology of melanoma through the PI3K/Akt pathway. Molecular docking also confirmed that higher content of ingredients in A&P, including hederagenin, quercetin, beta-sitosterol and stigmasterol, had a strong binding activity (affinity < −5 kcal/mol) with the core targets AKT1, MAPK3 and ESR1. Furthermore, we confirmed A&P could inhibit melanoma cells proliferation and induce cells apoptosis through suppressing the PI3K/Akt signaling pathway by in vitro and in vivo xenograft model experiments. These findings indicate that A&P may function as a useful therapy for melanoma through the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Fang Wang
- Department of Pharmacy, Xi’an International Medical Center Hospital, Xi’an, Shaanxi Province, China
| | - Juan Bai
- Department of Pharmacy, Xi’an International Medical Center Hospital, Xi’an, Shaanxi Province, China
| | - Feng Li
- Department of Pharmacy, Xi’an International Medical Center Hospital, Xi’an, Shaanxi Province, China
| | - Jing Liu
- Department of Pharmacy, Xi’an International Medical Center Hospital, Xi’an, Shaanxi Province, China
| | - Yanli Wang
- Department of Pharmacy, Xi’an International Medical Center Hospital, Xi’an, Shaanxi Province, China
| | - Ning Li
- Department of Pharmacy, Xi’an International Medical Center Hospital, Xi’an, Shaanxi Province, China
| | - Yaqi Wang
- Department of Pharmacy, Xi’an International Medical Center Hospital, Xi’an, Shaanxi Province, China
| | - Jin Xu
- Department of Pharmacy, Xi’an International Medical Center Hospital, Xi’an, Shaanxi Province, China
| | - Wanbao Liu
- Department of Pharmacy, Xi’an International Medical Center Hospital, Xi’an, Shaanxi Province, China
| | - Liting Xu
- Department of Pharmacy, Xi’an International Medical Center Hospital, Xi’an, Shaanxi Province, China
| | - Lin Chen
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi Province, China
- *Correspondence: Lin Chen,
| |
Collapse
|
15
|
Wang L, Liu X, Kang Q, Pan C, Zhang T, Feng C, Chen L, Wei S, Wang J. Nrf2 Overexpression Decreases Vincristine Chemotherapy Sensitivity Through the PI3K-AKT Pathway in Adult B-Cell Acute Lymphoblastic Leukemia. Front Oncol 2022; 12:876556. [PMID: 35646695 PMCID: PMC9134735 DOI: 10.3389/fonc.2022.876556] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/19/2022] [Indexed: 01/08/2023] Open
Abstract
Uncontrolled proliferation is an important cancer cell biomarker, which plays a critical role in carcinogenesis, progression and development of resistance to chemotherapy. An improved understanding of novel genes modulating cancer cell proliferation and mechanism will help develop new therapeutic strategies. The nuclear factor erythroid 2-related factor 2 (Nrf2), a transcription factor, decreases apoptosis when its expression is upregulated. However, the relationship between Nrf2 and Vincristine (VCR) chemotherapy resistance in B-cell acute lymphoblastic leukemia (B-ALL) is not yet established. Our results showed that Nrf2 levels could sufficiently modulate the sensitivity of B-ALL cells to VCRby regulating an apoptotic protein, i.e., the Bcl-2 agonist of cell death (BAD). Chemotherapeutic agents used for the treatment of B-ALL induced Nrf2 overactivation and PI3K-AKT pathway activation in the cells, independent of the resistance to chemotherapy; thus, a potential resistance loop during treatment for B-ALL with a drug combination is established. Therefore, B-ALL patients with a high expression of Nrf2 might mean induction chemotherapy with VCR effective little.
Collapse
Affiliation(s)
- Li Wang
- Clinical Medical College, Guizhou Medical University, Guiyang, China
| | - Xin Liu
- Clinical Medical College, Guizhou Medical University, Guiyang, China
| | - Qian Kang
- Department of Hematology, Guizhou Province Institute of Hematology, Guizhou Province Laboratory of Haematopoietic Stem Cell Transplantation Centre, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Chengyun Pan
- Department of Hematology, Guizhou Province Institute of Hematology, Guizhou Province Laboratory of Haematopoietic Stem Cell Transplantation Centre, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Tianzhuo Zhang
- Clinical Medical College, Guizhou Medical University, Guiyang, China
| | - Cheng Feng
- Clinical Medical College, Guizhou Medical University, Guiyang, China
| | - Lu Chen
- Clinical Medical College, Guizhou Medical University, Guiyang, China
| | - Sixi Wei
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jishi Wang
- Department of Hematology, Guizhou Province Institute of Hematology, Guizhou Province Laboratory of Haematopoietic Stem Cell Transplantation Centre, Affiliated Hospital of Guizhou Medical University, Guiyang, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Jiangsu, China
| |
Collapse
|
16
|
Mascaraque-Checa M, Gallego-Rentero M, Nicolás-Morala J, Portillo-Esnaola M, Cuezva JM, González S, Gilaberte Y, Juarranz Á. Metformin overcomes metabolic reprogramming-induced resistance of skin squamous cell carcinoma to photodynamic therapy. Mol Metab 2022; 60:101496. [PMID: 35405370 PMCID: PMC9048115 DOI: 10.1016/j.molmet.2022.101496] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 04/04/2022] [Accepted: 04/05/2022] [Indexed: 11/25/2022] Open
Abstract
Objective Cancer metabolic reprogramming promotes resistance to therapies. In this study, we addressed the role of the Warburg effect in the resistance to photodynamic therapy (PDT) in skin squamous cell carcinoma (sSCC). Furthermore, we assessed the effect of metformin treatment, an antidiabetic type II drug that modulates metabolism, as adjuvant to PDT. Methods For that, we have used two human SCC cell lines: SCC13 and A431, called parental (P) and from these cell lines we have generated the corresponding PDT resistant cells (10GT). Results Here, we show that 10GT cells induced metabolic reprogramming to an enhanced aerobic glycolysis and reduced activity of oxidative phosphorylation, which could influence the response to PDT. This result was also confirmed in P and 10GT SCC13 tumors developed in mice. The treatment with metformin caused a reduction in aerobic glycolysis and an increase in oxidative phosphorylation in 10GT sSCC cells. Finally, the combination of metformin with PDT improved the cytotoxic effects on P and 10GT cells. The combined treatment induced an increase in the protoporphyrin IX production, in the reactive oxygen species generation and in the AMPK expression and produced the inhibition of AKT/mTOR pathway. The greater efficacy of combined treatments was also seen in vivo, in xenografts of P and 10GT SCC13 cells. Conclusions Altogether, our results reveal that PDT resistance implies, at least partially, a metabolic reprogramming towards aerobic glycolysis that is prevented by metformin treatment. Therefore, metformin may constitute an excellent adjuvant for PDT in sSCC. Cell resistant to Photodynamic therapy (PDT) is due to the metabolic reprogramming. Metformin modulates energetic metabolism in PDT-resistant cells, sensitizing to PDT. Metformin increases protoporphyrin IX and reactive oxygen species generation. Metformin+PDT is proposed as potential therapy against skin squamous cell carcinoma.
Collapse
|
17
|
Lohan-Codeço M, Barambo-Wagner ML, Nasciutti LE, Ribeiro Pinto LF, Meireles Da Costa N, Palumbo A. Molecular mechanisms associated with chemoresistance in esophageal cancer. Cell Mol Life Sci 2022; 79:116. [PMID: 35113247 PMCID: PMC11073146 DOI: 10.1007/s00018-022-04131-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 02/07/2023]
Abstract
Esophageal cancer (EC) is one of the most incident and lethal tumors worldwide. Although surgical resection is an important approach in EC treatment, late diagnosis, metastasis and recurrence after surgery have led to the management of adjuvant and neoadjuvant therapies over the past few decades. In this scenario, 5-fluorouracil (5-FU) and cisplatin (CISP), and more recently paclitaxel (PTX) and carboplatin (CBP), have been traditionally used in EC treatment. However, chemoresistance to these agents along EC therapeutic management represents the main obstacle to successfully treat this malignancy. In this sense, despite the fact that most of chemotherapy drugs were discovered several decades ago, in many cases, including EC, they still represent the most affordable and widely employed treatment approach for these tumors. Therefore, this review summarizes the main mechanisms through which the response to the most widely chemotherapeutic agents used in EC treatment is impaired, such as drug metabolism, apoptosis resistance, cancer stem cells (CSCs), cell cycle, autophagy, energetic metabolism deregulation, tumor microenvironment and epigenetic modifications.
Collapse
Affiliation(s)
- Matheus Lohan-Codeço
- Laboratório de Interações Celulares, Instituto de Ciências Biomédicas, Programa de Pesquisa em Biologia Celular e do Desenvolvimento, Universidade Federal do Rio de Janeiro, Prédio do Centro de Ciências da Saúde-Cidade Universitária, Ilha do Fundão, Rua César Pernetta, 1766 (LS.3.01), Rio de Janeiro, RJ, Brasil
| | - Maria Luísa Barambo-Wagner
- Programa de Carcinogênese Molecular Coordenação de Pesquisa, Instituto Nacional de Câncer-INCA, Rua André Cavalcanti, 37-6ºandar-Centro, Rio de Janeiro, RJ, 20231-050, Brazil
| | - Luiz Eurico Nasciutti
- Laboratório de Interações Celulares, Instituto de Ciências Biomédicas, Programa de Pesquisa em Biologia Celular e do Desenvolvimento, Universidade Federal do Rio de Janeiro, Prédio do Centro de Ciências da Saúde-Cidade Universitária, Ilha do Fundão, Rua César Pernetta, 1766 (LS.3.01), Rio de Janeiro, RJ, Brasil
| | - Luis Felipe Ribeiro Pinto
- Programa de Carcinogênese Molecular Coordenação de Pesquisa, Instituto Nacional de Câncer-INCA, Rua André Cavalcanti, 37-6ºandar-Centro, Rio de Janeiro, RJ, 20231-050, Brazil
| | - Nathalia Meireles Da Costa
- Programa de Carcinogênese Molecular Coordenação de Pesquisa, Instituto Nacional de Câncer-INCA, Rua André Cavalcanti, 37-6ºandar-Centro, Rio de Janeiro, RJ, 20231-050, Brazil.
| | - Antonio Palumbo
- Laboratório de Interações Celulares, Instituto de Ciências Biomédicas, Programa de Pesquisa em Biologia Celular e do Desenvolvimento, Universidade Federal do Rio de Janeiro, Prédio do Centro de Ciências da Saúde-Cidade Universitária, Ilha do Fundão, Rua César Pernetta, 1766 (LS.3.01), Rio de Janeiro, RJ, Brasil.
| |
Collapse
|
18
|
Wang Y, Shen SY, Liu L, Zhang XD, Liu DY, Liu N, Liu BH, Shen L. Jolkinolide B inhibits proliferation or migration and promotes apoptosis of MCF-7 or BT-474 breast cancer cells by downregulating the PI3K-Akt pathway. JOURNAL OF ETHNOPHARMACOLOGY 2022; 282:114581. [PMID: 34464697 DOI: 10.1016/j.jep.2021.114581] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 08/20/2021] [Accepted: 08/27/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The diterpenoids extracted from Euphorbia kansui S.L. Liou ex S.B.Ho, Euphorbia fischeriana Steud. have good antitumor effects. Jolkinolide B has anti-breast cancer effect, but it is unclear whether it has different therapeutic effects between luminal A subtype and luminal B subtype breast cancer. AIM OF THE STUDY This study investigated the Jolkinolide B has different therapeutic, important targets and pathways effects between luminal A subtype and luminal B subtype breast cancer. MATERIALS AND METHODS We used bioinformatics to predict the biological process and molecular mechanism of Jolkinolide B in treating two types of breast cancer. Then, in vitro, cultured MCF-7 cells and BT-474 cells were divided into control group, PI3K inhibitor + control group, Jolkinolide B group and PI3K inhibitor + Jolkinolide B group. The CCK-8 assay, Flow cytometric analysis and Transwell cell migration assay was used to detect the cell proliferation, apoptosis, and migration in each group, respectively. ELISA was used to measure the content of Akt and phosphorylated Akt (p-Akt) in cell lysis buffer. RESULTS Compared to luminal A breast cancer, Jolkinolide B had more targets, proliferation, migration processes and KEGG pathways when treating luminal B subtype breast cancer. Jolkinolide B significantly prolonged the survival time of luminal B subtype breast cancer patients. Compared to the control group, the cell proliferation absorbance value (A value) and migration number of the two kinds of breast cancer cells in the Jolkinolide B group were decreased (P < 0.01, n = 6), and the number of apoptotic cells was increased (P < 0.01, n = 6). Compared to the Jolkinolide B group, the A value and migration number of the two types of breast cancer cells were significantly decreased in the PI3K inhibitor + Jolkinolide B group (P < 0.01, n = 6), and the number of apoptotic cells was significantly increased (P < 0.01, n = 6). In addition, compared to MCF-7 cells, the A value and migration number of BT-474 cells stimulated with Jolkinolide B were significantly decreased (P < 0.01, n = 6), and the number of apoptotic cells was significantly increased (P < 0.01, n = 6). Akt and p-Akt protein levels in the two breast cancer cell lines in the Jolkinolide B group were all decreased (P < 0.01, n = 6), especially in BT-474 cells stimulated by Jolkinolide B. CONCLUSION Jolkinolide B regulates the luminal A and luminal B subtypes of breast cancer through PI3K-Akt, EGFR and other pathways. Jolkinolide B has more significant therapeutic effect on luminal B subtype breast cancer. In vitro, experiments verified that Jolkinolide B significantly inhibited the proliferation and migration activity of BT-474 breast cancer cells by downregulating the PI3K-Akt pathway.
Collapse
Affiliation(s)
- Yang Wang
- Department of Physiology, Qiqihar Medical College, Qiqihar, 161006, PR China; Department of Research Section of Integrated Traditional Chinese and Western Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150040, PR China
| | - Shi-Yang Shen
- Grade 2019 of Acupuncture and Massage, Heilongjiang University of Chinese Medicine, Harbin, 150040, PR China
| | - Lei Liu
- Academy of Medical Sciences, Qiqihar Medical College, Qiqihar, 161006, PR China
| | - Xiao-Dong Zhang
- Department of Anatomy, Qiqihar Medical College, Qiqihar, 161006, PR China
| | - Dan-Yang Liu
- Department of Histology and Embryology, Qiqihar Medical College, Qiqihar, 161006, PR China
| | - Na Liu
- Department of Anatomy, Jiamusi University, Jiamusi, 154007, PR China
| | - Bing-Hua Liu
- Experimental Teaching of Clinical Skills, Qiqihar Medical College, Qiqihar, 161006, PR China
| | - Lei Shen
- Department of Anatomy, Qiqihar Medical College, Qiqihar, 161006, PR China.
| |
Collapse
|
19
|
Xu C, Li YM, Sun B, Zhong FJ, Yang LY. GNA14's interaction with RACK1 inhibits hepatocellular carcinoma progression through reducing MAPK/JNK and PI3K/AKT signaling pathway. Carcinogenesis 2021; 42:1357-1369. [PMID: 34657150 PMCID: PMC8598382 DOI: 10.1093/carcin/bgab098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 09/27/2021] [Accepted: 10/15/2021] [Indexed: 12/24/2022] Open
Abstract
Gαq subfamily proteins play critical roles in many biological functions including cardiovascular development, angiogenesis, and tumorigenesis of melanoma. However, the understanding of G Protein Subunit Alpha 14 (GNA14) in diseases, especially in cancers is limited. Here, we revealed that GNA14 was significantly low expression in Human hepatocellular carcinoma (HCC) samples. Low GNA14 expression was correlated with aggressive clinicopathological features. Moreover, the overall survival (OS) and disease-free survival (DFS) of high GNA14 expression HCC patients were much better than low GNA14 expression group. Lentivirus-mediated GNA14 knockdown significantly promoted the growth of liver cancer in vitro and in vivo. However, opposing results were observed when GNA14 is upregulated. Mechanistically, We identified receptor for activated C kinase 1 (RACK1) as a binding partner of GNA14 by co-immunoprecipitation and mass spectrometry (MS). Glutathione-S-transferase (GST) pull-down assay further verified the direct interaction between GNA14 and RACK1. RNA-Seq and loss- and gain-of-function assays also confirmed that GNA14 reduced the activity of both MAPK/JNK and PI3K/AKT signaling pathways through RACK1. GNA14 synergized with U73122 (PLC inhibitor) to enhance this effect. Further studies suggested that GNA14 potentially competed with protein kinase C (PKC) to bind with RACK1, consequently reducing the stability of PKC. Moreover, we also showed that GNA14’supression of p-AKT protein level depended on sufficient RACK1 expression. In conclusion, we indicated a different role of GNA14, which acted as a suppressor inhibiting liver cancer progression through MAPK/JNK and PI3K/AKT signaling pathways. Due to this, GNA14 served as a potentially valuable prognostic biomarker for liver cancer.
Collapse
Affiliation(s)
- Cong Xu
- Liver Cancer Laboratory, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Yi-Ming Li
- Liver Cancer Laboratory, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Bo Sun
- Liver Cancer Laboratory, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Fang-Jing Zhong
- Liver Cancer Laboratory, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Lian-Yue Yang
- Liver Cancer Laboratory, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- Department of Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- To whom correspondence should be addressed. Tel: +86-(0)731-84327365; Fax: (0)731-84327618;
| |
Collapse
|
20
|
Liu Y, Zheng C, Huang Y, He M, Xu WW, Li B. Molecular mechanisms of chemo- and radiotherapy resistance and the potential implications for cancer treatment. MedComm (Beijing) 2021; 2:315-340. [PMID: 34766149 PMCID: PMC8554658 DOI: 10.1002/mco2.55] [Citation(s) in RCA: 161] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 12/25/2020] [Accepted: 12/28/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer is a leading cause of death worldwide. Surgery is the primary treatment approach for cancer, but the survival rate is very low due to the rapid progression of the disease and presence of local and distant metastasis at diagnosis. Adjuvant chemotherapy and radiotherapy are important components of the multidisciplinary approaches for cancer treatment. However, resistance to radiotherapy and chemotherapy may result in treatment failure or even cancer recurrence. Radioresistance in cancer is often caused by the repair response to radiation-induced DNA damage, cell cycle dysregulation, cancer stem cells (CSCs) resilience, and epithelial-mesenchymal transition (EMT). Understanding the molecular alterations that lead to radioresistance may provide new diagnostic markers and therapeutic targets to improve radiotherapy efficacy. Patients who develop resistance to chemotherapy drugs cannot benefit from the cytotoxicity induced by the prescribed drug and will likely have a poor outcome with these treatments. Chemotherapy often shows a low response rate due to various drug resistance mechanisms. This review focuses on the molecular mechanisms of radioresistance and chemoresistance in cancer and discusses recent developments in therapeutic strategies targeting chemoradiotherapy resistance to improve treatment outcomes.
Collapse
Affiliation(s)
- Ya‐Ping Liu
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education InstitutesInstitute of Life and Health EngineeringJinan UniversityGuangzhouP. R. China
| | - Can‐Can Zheng
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education InstitutesInstitute of Life and Health EngineeringJinan UniversityGuangzhouP. R. China
| | - Yun‐Na Huang
- MOE Key Laboratory of Tumor Molecular Biology and Guangdong Provincial Key Laboratory of Bioengineering MedicineNational Engineering Research Center of Genetic MedicineInstitute of BiomedicineCollege of Life Science and TechnologyJinan UniversityGuangzhouP. R. China
| | - Ming‐Liang He
- Department of Biomedical SciencesCity University of Hong KongHong KongChina
| | - Wen Wen Xu
- MOE Key Laboratory of Tumor Molecular Biology and Guangdong Provincial Key Laboratory of Bioengineering MedicineNational Engineering Research Center of Genetic MedicineInstitute of BiomedicineCollege of Life Science and TechnologyJinan UniversityGuangzhouP. R. China
| | - Bin Li
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education InstitutesInstitute of Life and Health EngineeringJinan UniversityGuangzhouP. R. China
| |
Collapse
|
21
|
Liu Z, Hu Z, Cai X, Liu S. SLC39A5 promotes lung adenocarcinoma cell proliferation by activating PI3K/AKT signaling. Pathol Res Pract 2021; 224:153541. [PMID: 34252710 DOI: 10.1016/j.prp.2021.153541] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 07/01/2021] [Accepted: 07/01/2021] [Indexed: 12/13/2022]
Abstract
Lung cancer is the most common cancer and the primary cause of cancer-related deaths worldwide. Solute carrier family 39 member 5 (SLC39A5) regulates cellular zinc homeostasis and plays a vital role in several human cancers. However, the clinical significance and biological function of SLC39A5 in lung adenocarcinoma (LUAD) remain unclear. Hence, we sought to elucidate the role of SLC39A5 in LUAD pathophysiology in this study. The expression and clinical significance of SLC39A5 were evaluated using The Cancer Genome Atlas, the Gene Expression Omnibus, and tissue microarray data. We used the Cell Counting Kit-8, flow cytometry, western blotting, and quantitative reverse transcriptase-polymerase chain reaction analyses to determine the function of SLC39A5 in vitro. We also used a mouse xenograft model to evaluate the function of SLC39A5 in vivo. Our results indicate that SLC39A5 was upregulated in LUAD tissues compared with that in adjacent non-tumor lung tissues. SLC39A5 overexpression correlated with poor survival in patients with LUAD. SLC39A5 promoted LUAD cell proliferation by accelerating the G1-to-S phase transition and inhibiting apoptosis. SLC39A5 knockdown inhibited the tumorigenesis of LUAD cells in a nude mouse model of xenograft tumors. SLC39A5 promoted LUAD cell proliferation by activating the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling. SLC39A5 played an oncogenic role in LUAD by activating the PI3K/AKT signaling. Hence, SLC39A5 may serve as a novel prognostic biomarker and potential therapeutic target for LUAD.
Collapse
Affiliation(s)
- Zhaohui Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Department of Respiratory Medicine, Chenzhou No.1 People's Hospital, Chenzhou 423000, China
| | - Zheng Hu
- Translational Medicine Institute, the First People's Hospital of Chenzhou Affiliated to University of South China, Chenzhou 432000, China
| | - Xingdong Cai
- Department of Respiratory Medicine, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Shengming Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China.
| |
Collapse
|
22
|
MiR-325 Promotes Oxaliplatin-Induced Cytotoxicity Against Colorectal Cancer Through the HSPA12B/PI3K/AKT/Bcl-2 Pathway. Dig Dis Sci 2021; 66:2651-2660. [PMID: 32914380 DOI: 10.1007/s10620-020-06579-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 08/21/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Oxaliplatin is one of the most effective chemotherapeutic drugs used for the treatment of colorectal cancer (CRC). However, intervention that attenuates the resistance of oxaliplatin is still required in the treatment of CRC. AIMS To investigate the role of miR-325 in changing the oxaliplatin sensitivity to CRC cells. METHODS Expression of miR-325 in colorectal cancer tissues and cell lines was measured by using qRT-PCR analysis. Cytotoxicity of oxaliplatin to control or miR-325-overexpressed HT29 and SW480 cells was evaluated by CCK-8 assays. Luciferase reporter assay was used to confirm the regulation of miR-325 on HSPA12B. Flow cytometry was performed to detect the mitochondrial membrane potential and cell apoptosis. RESULTS Expression of miR-325 was decreased in colorectal cancer tissues and cell lines. However, overexpression of miR-325 can decrease the 50% inhibiting concentration of oxaliplatin to colorectal cancer cell lines HT29 and SW480. Mechanically, we confirmed that miR-325 targeted HSPA12B in colorectal cancer. Therefore, overexpression of miR-325 inhibited the phosphorylation of PI3K and AKT and decreased the expression of Bcl-2 to promote the oxaliplatin-induced mitochondrial apoptosis in colorectal cancer. CONCLUSIONS MiR-325 sensitizes the colorectal cancer cells to oxaliplatin-induced cytotoxicity through the HSPA12B/PI3K/AKT/Bcl-2 pathway.
Collapse
|
23
|
Bao F, Hao P, An S, Yang Y, Liu Y, Hao Q, Ejaz M, Guo XX, Xu TR. Akt scaffold proteins: the key to controlling specificity of Akt signaling. Am J Physiol Cell Physiol 2021; 321:C429-C442. [PMID: 34161152 DOI: 10.1152/ajpcell.00146.2020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The phosphatidylinositol 3-kinase-Akt signaling pathway plays an essential role in regulating cell proliferation and apoptosis. Akt kinase is at the center of this signaling pathway and interacts with a variety of proteins. Akt is overexpressed in almost 80% of tumors. However, inhibiting Akt has serious clinical side effects so is not a suitable treatment for cancer. During recent years, Akt scaffold proteins have received increasing attention for their ability to regulate Akt signaling and have emerged as potential targets for cancer therapy. In this paper, we categorize Akt kinase scaffold proteins into four groups based on their cellular location: membrane-bound activator and inhibitor, cytoplasm, and endosome. We describe how these scaffolds interact with Akt kinase, how they affect Akt activity, and how they regulate the specificity of Akt signaling. We also discuss the clinical application of Akt scaffold proteins as targets for cancer therapy.
Collapse
Affiliation(s)
- Fan Bao
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China.,Center of Stomatology, The First People's Hospital of Yunnan Province, Kunming, China.,The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Peiqi Hao
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Su An
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yang Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Ying Liu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Qian Hao
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Mubashir Ejaz
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Xiao-Xi Guo
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Tian-Rui Xu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| |
Collapse
|
24
|
Trendafilova A, Moujir LM, Sousa PMC, Seca AML. Research Advances on Health Effects of Edible Artemisia Species and Some Sesquiterpene Lactones Constituents. Foods 2020; 10:E65. [PMID: 33396790 PMCID: PMC7823681 DOI: 10.3390/foods10010065] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/22/2020] [Accepted: 12/25/2020] [Indexed: 12/20/2022] Open
Abstract
The genus Artemisia, often known collectively as "wormwood", has aroused great interest in the scientific community, pharmaceutical and food industries, generating many studies on the most varied aspects of these plants. In this review, the most recent evidence on health effects of edible Artemisia species and some of its constituents are presented and discussed, based on studies published until 2020, available in the Scopus, Web of Sciences and PubMed databases, related to food applications, nutritional and sesquiterpene lactones composition, and their therapeutic effects supported by in vivo and clinical studies. The analysis of more than 300 selected articles highlights the beneficial effect on health and the high clinical relevance of several Artemisia species besides some sesquiterpene lactones constituents and their derivatives. From an integrated perspective, as it includes therapeutic and nutritional properties, without ignoring some adverse effects described in the literature, this review shows the great potential of Artemisia plants and some of their constituents as dietary supplements, functional foods and as the source of new, more efficient, and safe medicines. Despite all the benefits demonstrated, some gaps need to be filled, mainly related to the use of raw Artemisia extracts, such as its standardization and clinical trials on adverse effects and its health care efficacy.
Collapse
Affiliation(s)
- Antoaneta Trendafilova
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 9, 1113 Sofia, Bulgaria
| | - Laila M. Moujir
- Department of Biochemistry, Microbiology, Genetics and Cell Biology, Facultad de Farmacia, Universidad de La Laguna, 38206 La Laguna, Tenerife, Spain;
| | - Pedro M. C. Sousa
- Faculty of Sciences and Technology, University of Azores, 9500-321 Ponta Delgada, Portugal;
| | - Ana M. L. Seca
- cE3c—Centre for Ecology, Evolution and Environmental Changes/Azorean Biodiversity Group & Faculty of Sciences and Technology, University of Azores, Rua Mãe de Deus, 9500-321 Ponta Delgada, Portugal
- LAQV-REQUIMTE, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
25
|
Huang D, Chen X, Chen X, Qu Y, Wang Y, Yang Y, Cheng Y. WDR5 Promotes Proliferation and Correlates with Poor Prognosis in Oesophageal Squamous Cell Carcinoma. Onco Targets Ther 2020; 13:10525-10534. [PMID: 33116631 PMCID: PMC7573303 DOI: 10.2147/ott.s234773] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 09/11/2020] [Indexed: 12/12/2022] Open
Abstract
Background The WD40 protein family member WD repeat domain 5 (WDR5) plays significant roles in the tumorigenesis and development of multiple organ tumours. However, the correlation between WDR5 expression and oesophageal squamous cell carcinoma (ESCC) has not been elucidated. Methods WDR5 mRNA expression data were acquired from The Cancer Genome Atlas (TCGA) database, and the expression and prognostic potential of WDR5 were assessed by immunohistochemistry (IHC) and Western blot. The cell counting kit-8 (CCK-8), colony formation assay and cell cycle evaluation were performed to verify the WDR5 function in vitro. The xenograft model was used to verify WDR5 function in vivo. Results The mRNA and protein expression levels of WDR5 were significantly upregulated in ESCC tissues compared with expression in adjacent normal tissues. Kaplan-Meier analysis showed that high WDR5 expression in ESCC patients was associated with poor overall survival (P=0.004). Multivariate analysis revealed that WDR5 overexpression emerged as an independent predictor of poor overall survival (P=0.013) in ESCC. The in vitro and in vivo experiments revealed that downregulation of WDR5 expression blocked cell proliferation of ESCC. Mechanistically, we found that WDR5 may influence ESCC proliferation by targeting the PI3K/AKT/mTOR signalling pathway. Conclusion Our data demonstrate that overexpression of WDR5 was associated with poor prognosis in patients with ESCC and that WDR5 may act as a potential novel prognostic biomarker for ESCC.
Collapse
Affiliation(s)
- Di Huang
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Xue Chen
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Xuan Chen
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Yan Qu
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Yuanyuan Wang
- Department of Oncology, People's Hospital of Linyi County, Dezhou, Shandong 251500, People's Republic of China
| | - Yafei Yang
- Department of Hepatobiliary Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Yufeng Cheng
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, People's Republic of China
| |
Collapse
|
26
|
The chemosensitizer ferulic acid enhances epirubicin-induced apoptosis in MDA-MB-231 cells. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.104130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
27
|
PI3K/AKT pathway as a key link modulates the multidrug resistance of cancers. Cell Death Dis 2020; 11:797. [PMID: 32973135 PMCID: PMC7515865 DOI: 10.1038/s41419-020-02998-6] [Citation(s) in RCA: 510] [Impact Index Per Article: 102.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 08/17/2020] [Accepted: 08/27/2020] [Indexed: 12/13/2022]
Abstract
Multidrug resistance (MDR) is the dominant challenge in the failure of chemotherapy in cancers. Phosphatidylinositol 3-kinase (PI3K) is a lipid kinase that spreads intracellular signal cascades and regulates a variety of cellular processes. PI3Ks are considered significant causes of chemoresistance in cancer therapy. Protein kinase B (AKT) is also a significant downstream effecter of PI3K signaling, and it modulates several pathways, including inhibition of apoptosis, stimulation of cell growth, and modulation of cellular metabolism. This review highlights the aberrant activation of PI3K/AKT as a key link that modulates MDR. We summarize the regulation of numerous major targets correlated with the PI3K/AKT pathway, which is further related to MDR, including the expression of apoptosis-related protein, ABC transport and glycogen synthase kinase-3 beta (GSK-3β), synergism with nuclear factor kappa beta (NF-κB) and mammalian target of rapamycin (mTOR), and the regulation of glycolysis.
Collapse
|
28
|
Huang Z, Gao Y, Hou D. Interleukin-22 enhances chemoresistance of lung adenocarcinoma cells to paclitaxel. Hum Cell 2020; 33:850-858. [PMID: 32452013 DOI: 10.1007/s13577-020-00373-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 05/07/2020] [Indexed: 12/16/2022]
Abstract
The chemoresistance of tumors is the main barrier to cancer treatment. Interleukin-22 (IL-22) plays an important role in the chemoresistance of multi-cancers; however, the roles of IL-22 in the paclitaxel resistance of lung adenocarcinoma cells remain to be investigated. The present study aims to investigate the potential mechanisms of IL-22 enhancing the chemoresistance of lung adenocarcinoma cells to paclitaxel. We cultured A549, H358, and A549/PTX cell lines. qRT-PCR and western blot assays were performed to examine the mRNA and/or protein levels of IL-22 in A549, A549/PTX, H358, and H358/PTX. Moreover, cells were transfected with IL-22 siRNA1, IL-22 siRNA2, and siRNA NC, and treated with paclitaxel, and the proliferation rate of lung adenocarcinoma cells was evaluated by MTT assay. Flow cytometry was conducted to determine the apoptosis rate of lung adenocarcinoma cells. The results showed that the expression of IL-22 in lung adenocarcinoma tissues was higher than that in normal tissues, and the expression of IL-22 was higher in A549/PTX and H358/PTX compared with A549 and H358 cells. Meanwhile, the expression of IL-22 was strongly correlated with smoking history and TMN stage, as well. Furthermore, IL-22 siRNA inhibited the proliferation and promoted the apoptosis of A549/PTX and H358/PTX cells, and IL-22 siRNA also suppressed the expression levels of AKT and Bcl-2 and increased the expression levels of Bax and cleaved caspase 3. To sum up, IL-22 may mediate the chemosensitivity of lung adenocarcinoma cells to paclitaxel through inhibiting the AKT signaling pathways.
Collapse
Affiliation(s)
- Zhiliang Huang
- Thoracic Cardiovascular Surgery, Inner Mongolia Forestry General Hospital, Yakeshi, 022150, China
- The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, No. 168 Hong Kong Road, Jiang'an District, Wuhan, 430015, Hubei Province, China
| | - Yu Gao
- The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, No. 168 Hong Kong Road, Jiang'an District, Wuhan, 430015, Hubei Province, China
- Geriatrics, Inner Mongolia Forestry General Hospital, Yakeshi, 022150, China
| | - Dianchen Hou
- Thoracic Cardiovascular Surgery, Inner Mongolia Forestry General Hospital, Yakeshi, 022150, China.
| |
Collapse
|
29
|
Javid H, Asadi J, Zahedi Avval F, Afshari AR, Hashemy SI. The role of substance P/neurokinin 1 receptor in the pathogenesis of esophageal squamous cell carcinoma through constitutively active PI3K/Akt/NF-κB signal transduction pathways. Mol Biol Rep 2020; 47:2253-2263. [PMID: 32072401 DOI: 10.1007/s11033-020-05330-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 02/13/2020] [Indexed: 10/25/2022]
Abstract
One of the most prevalent malignancies is esophageal squamous cell carcinoma (ESCC), which is associated with high morbidity and mortality. Substance P (SP), as one of the peptides released from sensory nerves, causes the enhancement of cellular excitability through the activation of the neurokinin-1 (NK1) receptor in several human tumor cells. Aprepitant, a specific, potent, and long-acting NK1 receptor antagonist, is considered as a novel agent to inhibit proliferation and induce apoptosis in malignant cells. Since the antitumor mechanism of aprepitant in ESCC is not completely understood, we conducted this study and found that aprepitant induced growth inhibition of KYSE-30 cells and arrested cells in the G2/M phase of the cell cycle. Aprepitant also caused apoptotic cell death and inhibited activation of the PI3K/Akt axis and its downstream effectors, including NF-κB in KYSE-30 cells. Besides, quantitative real-time (qRT)-PCR analysis showed a significant down-regulation of NF-κB target genes in KYSE-30 cells, indicating a probable NF-κB-dependent mechanism involved in aprepitant cytotoxicity. Thus, the present study recommends that SP/NK1R system might, therefore, be considered as an emerging and promising therapeutic strategy against ESCC.
Collapse
Affiliation(s)
- Hossein Javid
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jahanbakhsh Asadi
- Department of Clinical Biochemistry, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Farnaz Zahedi Avval
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir R Afshari
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Seyed Isaac Hashemy
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
30
|
Shen C, Hua H, Gu L, Cao S, Cai H, Yao X, Chen X. Overexpression of RACK1 Predicts Poor Prognosis in Melanoma. J Cancer 2020; 11:795-803. [PMID: 31949482 PMCID: PMC6959021 DOI: 10.7150/jca.36905] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 09/22/2019] [Indexed: 12/13/2022] Open
Abstract
Melanoma is a highly malignant skin cancer with limited treatment options, the mechanism of the occurrence and development of melanoma is still unclear till now. Receptor for activated C kinase 1 (RACK1) is a scaffolding protein that mediates multiple signaling pathways; it interconnects distinct signaling pathways to control essential cellular processes. RACK1 was reported as an oncogene in human tumorigenesis, but little is known about its role in melanoma. This study aimed to investigate the expression of RACK1 in patients with melanoma and to reveal its possible functions in melanoma cells. The expression profiles of RACK1 detected in tumor tissues from melanoma patients showed that RACK1 was higher in tumor tissues, and its expression level was well associated with the clinical progression of melanoma (TNM stage, P=0.009). Furthermore, RNA interfering (RNAi) knockdown of RACK1 could efficiently suppress the proliferation, migration and invasion of A375 and A875 cells and promote their apoptosis. Taken together, these results suggest that RACK1 may be a poor prognostic factor in human melanoma, and it may be a new therapeutic target for melanoma treatment.
Collapse
Affiliation(s)
- Congcong Shen
- Department of Dermatology, Affiliated Hospital of Nantong University, Nantong, 226001, P.R. China
| | - Hui Hua
- Department of Dermatology, The Third People's Hospital of Nantong, Nantong, 226001, P.R. China
| | - Lixiong Gu
- Department of Dermatology, Affiliated Hospital of Nantong University, Nantong, 226001, P.R. China
| | - Shuanglin Cao
- Department of Dermatology, Affiliated Hospital of Nantong University, Nantong, 226001, P.R. China
| | - Hengji Cai
- Department of Dermatology, Affiliated Hospital of Nantong University, Nantong, 226001, P.R. China
| | - Xiaodong Yao
- Department of Dermatology, Affiliated Hospital of Nantong University, Nantong, 226001, P.R. China
| | - Xiaodong Chen
- Department of Dermatology, Affiliated Hospital of Nantong University, Nantong, 226001, P.R. China
| |
Collapse
|
31
|
Fan Y, Si W, Ji W, Wang Z, Gao Z, Tian R, Song W, Zhang H, Niu R, Zhang F. Rack1 mediates tyrosine phosphorylation of Anxa2 by Src and promotes invasion and metastasis in drug-resistant breast cancer cells. Breast Cancer Res 2019; 21:66. [PMID: 31113450 PMCID: PMC6530024 DOI: 10.1186/s13058-019-1147-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 04/29/2019] [Indexed: 12/20/2022] Open
Abstract
Background Acquirement of resistance is always associated with a highly aggressive phenotype of tumor cells. Recent studies have revealed that Annexin A2 (Anxa2) is a key protein that links drug resistance and cancer metastasis. A high level of Anxa2 in cancer tissues is correlated to a highly aggressive phenotype. Increased Anxa2 expression appears to be specific in many drug-resistant cancer cells. The functional activity of Anxa2 is regulated by tyrosine phosphorylation at the Tyr23 site. Nevertheless, the accurate molecular mechanisms underlying the regulation of Anxa2 tyrosine phosphorylation and whether phosphorylation is necessary for the enhanced invasive phenotype of drug-resistant cells remain unknown. Methods Small interfering RNAs, small molecule inhibitors, overexpression, loss of function or gain of function, rescue experiments, Western blot, wound healing assays, transwell assays, and in vivo metastasis mice models were used to investigate the functional effects of Rack1 and Src on the tyrosine phosphorylation of Anxa2 and the invasion and metastatic potential of drug-resistant breast cancer cells. The interaction among Rack1, Src, and Anxa2 in drug-resistant cells was verified by co-immunoprecipitation assay. Results We demonstrated that Anxa2 Tyr23 phosphorylation is necessary for multidrug-resistant breast cancer invasion and metastasis. Rack1 is required for the invasive and metastatic potential of drug-resistant breast cancer cells through modulating Anxa2 phosphorylation. We provided evidence that Rack1 acts as a signal hub and mediates the interaction between Src and Anxa2, thereby facilitating Anxa2 phosphorylation by Src kinase. Conclusions Our findings suggest a convergence point role of Rack1/Src/Anxa2 complex in the crosstalk between drug resistance and cancer aggressiveness. The interaction between Anxa2 and Rack1/Src is responsible for the association between drug resistance and invasive/metastatic potential in breast cancer cells. Thus, our findings provide novel insights on the mechanism underlying the functional linkage between drug resistance and cancer aggressiveness. Electronic supplementary material The online version of this article (10.1186/s13058-019-1147-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yanling Fan
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin, 300060, China
| | - Weiyao Si
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin, 300060, China
| | - Wei Ji
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin, 300060, China
| | - Zhiyong Wang
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin, 300060, China
| | - Zicong Gao
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin, 300060, China
| | - Ran Tian
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin, 300060, China
| | - Weijie Song
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin, 300060, China
| | - He Zhang
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin, 300060, China
| | - Ruifang Niu
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China. .,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China. .,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China. .,Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin, 300060, China.
| | - Fei Zhang
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China. .,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China. .,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China. .,Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin, 300060, China.
| |
Collapse
|
32
|
Ren Z, Chen L, Wang Y, Wei X, Zeng S, Zheng Y, Gao C, Liu H. Activation of the Omega-3 Fatty Acid Receptor GPR120 Protects against Focal Cerebral Ischemic Injury by Preventing Inflammation and Apoptosis in Mice. THE JOURNAL OF IMMUNOLOGY 2018; 202:747-759. [PMID: 30598514 DOI: 10.4049/jimmunol.1800637] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 11/22/2018] [Indexed: 12/28/2022]
Abstract
G protein-coupled receptor 120 (GPR120) has been shown to negatively regulate inflammation and apoptosis, but its role in cerebral ischemic injury remains unclear. Using an in vivo model of middle cerebral artery occlusion (MCAO) and an in vitro model of oxygen-glucose deprivation (OGD), we investigated the potential role and molecular mechanisms of GPR120 in focal cerebral ischemic injury. Increased GPR120 expression was observed in microglia and neurons following MCAO-induced ischemia in wild type C57BL/6 mice. Treatment with docosahexaenoic acid (DHA) inhibited OGD-induced inflammatory response in primary microglia and murine microglial BV2 cells, whereas silencing of GPR120 strongly exacerbated the inflammation induced by OGD and abolished the anti-inflammatory effects of DHA. Mechanistically, DHA inhibited OGD-induced inflammation through GPR120 interacting with β-arrestin2. In addition to its anti-inflammatory function, GPR120 also played a role in apoptosis as its knockdown impaired the antiapoptotic effect of DHA in OGD-induced rat pheochromocytoma (PC12) cells. Finally, using MCAO mouse model, we demonstrated that GPR120 activation protected against focal cerebral ischemic injury by preventing inflammation and apoptosis. Our study indicated that pharmacological targeting of GPR120 may provide a novel approach for the treatment of patients with ischemic stroke.
Collapse
Affiliation(s)
- Zhiping Ren
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Lin Chen
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Yimeng Wang
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Xinbing Wei
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Shenglan Zeng
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Yi Zheng
- State Key Laboratory of Microbial Technology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China.,Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China; and.,Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Chengjiang Gao
- State Key Laboratory of Microbial Technology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China; .,Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China; and.,Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Huiqing Liu
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China;
| |
Collapse
|
33
|
Cao JX, Lu Y. Targeting CDC7 improves sensitivity to chemotherapy of esophageal squamous cell carcinoma. Onco Targets Ther 2018; 12:63-74. [PMID: 30588031 PMCID: PMC6304257 DOI: 10.2147/ott.s183629] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
PURPOSE The cell division cycle 7 (CDC7) is a serine/threonine kinase that is essential for DNA replication in human cells which has been identified to play a critical role in multiple cancer types. However, the expression and clinical significance of CDC7 in ESCC has never been reported. PATIENTS AND METHODS CDC7 expression was detected in 30 ESCC and matched adjacent normal tissues, and a series of loss-of-function and gain-of-function assays were performed to evaluate the effects of CDC7 on the proliferation, migration and invasion, and chemoresistance of ESCC cells. RESULTS The results showed that CDC7 was highly expressed in ESCC tissues compared with matched adjacent normal tissues. Functional studies demonstrated that knockdown of CDC7 inhibited proliferation by arresting ESCC cells in the G0/G1 phase and inducing apoptosis. Knockdown of CDC7 also inhibited cell migration and invasion in ESCC cells. Furthermore, knockdown of CDC7 sensitized ESCC cells to Cis and 5-FU. CONCLUSION Our results suggest that CDC7 is highly expressed in ESCC tissues, and silencing CDC7 enhances chemosensitivity of ESCC cells, providing a new avenue for ESCC therapy.
Collapse
Affiliation(s)
- Ji-Xiang Cao
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Science, Peking University, Beijing 100871, People's Republic of China,
- Department of Pathology, Zhongshan Hospital Xiamen University, Xiamen 361004, People's Republic of China,
| | - Yao Lu
- Department of Rehabilitation Medicine, Peking University 3rd Hospital, Beijing 100191, People's Republic of China
| |
Collapse
|
34
|
Chen YT, Xie JY, Sun Q, Mo WJ. Novel drug candidates for treating esophageal carcinoma: A study on differentially expressed genes, using connectivity mapping and molecular docking. Int J Oncol 2018; 54:152-166. [PMID: 30387840 PMCID: PMC6254996 DOI: 10.3892/ijo.2018.4618] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 10/23/2018] [Indexed: 12/14/2022] Open
Abstract
Patients with esophageal carcinoma (ESCA) have a poor prognosis and high mortality rate. Although standard therapies have had effect, there is an urgent requirement to develop novel options, as increasing drug tolerance has been identified in clinical practice. In the present study, differentially expressed genes (DEGs) of ESCA were identified in The Cancer Genome Atlas and Genotype-Tissue Expression databases. Functional and protein-protein interaction (PPI) analyses were performed. The Connectivity Map (CMAP) was selected to predict drugs for the treatment of ESCA, and their target genes were acquired from the Search Tool for Interactions of Chemicals (STITCH) by uploading the Simplified Molecular-Input Line-Entry System structure. Additionally, significant target genes and ESCA-associated hub genes were extracted using another PPI analysis, and the corresponding drugs were added to construct a network. Furthermore, the binding affinity between predicted drug candidates and ESCA-associated hub genes was calculated using molecular docking. Finally, 827 DEGs (|log2 fold-change|≥2; q-value <0.05), which are principally involved in protein digestion and absorption (P<0.005), the plasminogen-activating cascade (P<0.01), as well as the ‘biological regulation’ of the Biological Process, ‘membrane’ of the Cellular Component and ‘protein binding’ of the Molecular Function categories, were obtained. Additionally, 11 hub genes were obtained from the PPI network (all degrees ≥30). Furthermore, the 15 first screen drugs were extracted from CMAP (score <−0.85) and the 9 second screen drugs with 70 target genes were extracted from STITCH. Furthermore, another PPI analysis extracted 51 genes, and apigenin, baclofen, Prestwick-685, menadione, butyl hydroxybenzoate, gliclazide and valproate were selected as drug candidates for ESCA. Those molecular docking results with a docking score of >5.52 indicated the significance of apigenin, Prestwick-685 and menadione. The results of the present study may lead to novel drug candidates for ESCA, among which Prestwick-685 and menadione were identified to be significant new drug candidates.
Collapse
Affiliation(s)
- Yu-Ting Chen
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jia-Yi Xie
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Qi Sun
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Wei-Jia Mo
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
35
|
Zhang L, Xu Y, Wang L, Liu H. Role of RACK1 on cell proliferation, adhesion, and bortezomib-induced apoptosis in multiple myeloma. Int J Biol Macromol 2018; 121:1077-1085. [PMID: 30315883 DOI: 10.1016/j.ijbiomac.2018.10.034] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 09/16/2018] [Accepted: 10/09/2018] [Indexed: 12/30/2022]
Abstract
Receptor for activated C kinase 1 (RACK1), a scaffold protein, plays a crucial role in the progression of various cancers. However, the biological function and underlying mechanism of RACK1 in multiple myeloma (MM) cells remain unclear. The present study aimed to explore the function of RACK1 on the cell proliferation, adhesion, and bortezomib-induced apoptosis in MM. We found that RACK1 was significantly overexpressed in myeloma cell lines and primary myeloma cells compared with normal bone marrow plasma cells. Moreover, immunofluorescence revealed that RACK1 was primarily expressed in the cytoplasm of MM cells. Knockdown of RACK1 impaired growth of MM cells, blocked entry into the S-phase of the cell cycle, and resulted in reduced cell adhesion rates. More importantly, knockdown of RACK1 decreased the proliferation of MM cells by activating P-P38 and P-ERK in the MAPK/ERK signaling pathway. We also found that altered expression of RACK1 is associated with bortezomib-mediated MM cell apoptosis. In summary, these results may provide a possible target for therapy in MM.
Collapse
Affiliation(s)
- Linlin Zhang
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong University, Nantong, People's Republic of China
| | - Ya Xu
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong University, Nantong, People's Republic of China
| | - Li Wang
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong University, Nantong, People's Republic of China
| | - Hong Liu
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong University, Nantong, People's Republic of China.
| |
Collapse
|
36
|
Shi Q, Shen LY, Dong B, Fu H, Kang XZ, Yang YB, Dai L, Yan WP, Xiong HC, Liang Z, Chen KN. The identification of the ATR inhibitor VE-822 as a therapeutic strategy for enhancing cisplatin chemosensitivity in esophageal squamous cell carcinoma. Cancer Lett 2018; 432:56-68. [PMID: 29890208 DOI: 10.1016/j.canlet.2018.06.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 05/12/2018] [Accepted: 06/06/2018] [Indexed: 12/11/2022]
Abstract
Inducing DNA damage is known to be one of the mechanisms of cytotoxic chemotherapy agents for cancer such as cisplatin. The endogenous DNA damage response confers chemoresistance to these agents by repairing DNA damage. The initiation and transduction of the DNA damage response (DDR) signaling pathway, which is dependent on the activation of ATM (ataxia-telangiectasia mutated) and ATR (ataxia telangiectasia and Rad3-related), is essential for DNA damage repair, the maintenance of genomic stability and cell survival. Therefore, ATM or ATR inhibition is considered as a promising strategy for sensitizing cancer cells to chemotherapy. This study is aimed to explore the effect of ATR inhibitor on sensitizing ESCC (esophageal squamous cell carcinoma) cells to cisplatin, and whether ATM deficiency could impact the sensitization. We found that 21.5% of ESCC cases had ATM deficiency and that patients with ATR activation after neoadjuvant chemotherapy had worse chemotherapy response and poorer overall survival than that without ATR activation (32 mons vs. >140mons). Then, it was shown that VE-822 inhibited ATR-CHK1 pathway activation, leading to the accumulation of cisplatin-modified DNA. And it inhibited cell proliferation, induced cell cycle arrest in G1 phase and enhanced cell apoptosis. Moreover, VE-822 significantly sensitized ESCC cells to cisplatin, and these two drugs had synergistic effects, especially in ATM-deficient cells, in vitro and in vivo. Our results suggest that ATR inhibition combining with cisplatin is a new strategy for managing patients with ESCC, especially those with ATM-deficiency. However, this is an idea that requires further validation.
Collapse
Affiliation(s)
- Qi Shi
- Department of Thoracic Surgery I, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Lu-Yan Shen
- Department of Thoracic Surgery I, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Bin Dong
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Hao Fu
- Department of Thoracic Surgery I, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Xiao-Zheng Kang
- Department of Thoracic Surgery I, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Yong-Bo Yang
- Department of Thoracic Surgery I, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Liang Dai
- Department of Thoracic Surgery I, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Wan-Pu Yan
- Department of Thoracic Surgery I, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Hong-Chao Xiong
- Department of Thoracic Surgery I, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Zhen Liang
- Department of Thoracic Surgery I, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Ke-Neng Chen
- Department of Thoracic Surgery I, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People's Republic of China.
| |
Collapse
|
37
|
Shi H, Mao Y, Ju Q, Wu Y, Bai W, Wang P, Zhang Y, Jiang M. C-terminal binding protein‑2 mediates cisplatin chemoresistance in esophageal cancer cells via the inhibition of apoptosis. Int J Oncol 2018; 53:167-176. [PMID: 29658564 DOI: 10.3892/ijo.2018.4367] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 03/09/2018] [Indexed: 11/05/2022] Open
Abstract
C-terminal binding protein‑2 (CtBP2) is a transcriptional co-repressor that is associated with tumorigenesis and tumor progression. It has been reported to predict a poor prognosis in several human cancers, including esophageal squamous cell carcinoma (ESCC). The present study aimed to investigate the involvement of CtBP2 in the cisplatin (DDP) resistance of the ECA109 ESCC cell line and its effect on the expression of apoptosis-associated proteins. Constructed recombinant lentiviruses were used for the knockdown or overexpression of CtBP2 in ECA109 cells, and the expression of CtBP2 was measured using reverse transcription-quantitative polymerase chain reaction and western blotting following transfection. MTT assays, Hoechst 33342 staining and flow cytometry (FCM) were applied to detect the influence of CtBP2 on the DDP-induced viability and apoptosis of the transfected ECA109 cells. In addition, the levels of apoptosis-associated proteins, including p53, B‑cell lymphoma 2 (Bcl‑2), Bcl‑2‑associated X protein (Bax) and activated caspase-3 were investigated in the transfected ECA109 cells. Stable ECA109 cells with CtBP2 overexpression or knockdown were successfully established. The results of the MTT, Hoechst 33342 and FCM assays demonstrated that overexpression of CtBP2 attenuated the reduction of cell viability and inhibited the cell apoptosis induced by DDP. Furthermore, the western blotting results indicated that CtBP2 overexpression inhibited the DDP-induced apoptosis of ECA109 cells via the reduction of p53, activated caspase-3 and Bax expression, and promotion of Bcl‑2 expression. Therefore, the present study indicated that CtBP2 reduced the susceptibility of ECA109 cells to DDP by regulating the expression of apoptosis-related proteins, suggesting that it may be a promising therapeutic target in ESCC in the future.
Collapse
Affiliation(s)
- Hui Shi
- Laboratory Animals Center, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yinting Mao
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Qianqian Ju
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yingcheng Wu
- Medical College of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Wen Bai
- Medical College of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Peiwen Wang
- Medical College of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yudong Zhang
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Maorong Jiang
- Laboratory Animals Center, Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
38
|
Konstat-Korzenny E, Ascencio-Aragón JA, Niezen-Lugo S, Vázquez-López R. Artemisinin and Its Synthetic Derivatives as a Possible Therapy for Cancer. ACTA ACUST UNITED AC 2018; 6:medsci6010019. [PMID: 29495461 PMCID: PMC5872176 DOI: 10.3390/medsci6010019] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 02/15/2018] [Accepted: 02/22/2018] [Indexed: 12/11/2022]
Abstract
To assess the possibility of using the antimalarial drug artemisinin and its synthetic derivatives as antineoplastic drugs. A Pubmed and Google Scholar (1983–2018) search was performed using the terms artemisinin, cancer, artesunate and Artemisia annua. Case reports and original research articles, review articles, and clinical trials in both humans and animals were evaluated. Both in vitro and in vivo clinical trials and case reports have shown promising activity of the artemisinin drug derivatives in treating certain types of cancer. However, the reported articles are few, and therefore not statistically significant. The minimal toxicity shown in clinical trials and case reports, along with the selective cytotoxic activity of the compounds, make them possible cancer therapies due to the emerging evidence of the drug’s effectiveness.
Collapse
Affiliation(s)
- Enrique Konstat-Korzenny
- Departamento de Microbiología del Centro de Investigación en Ciencias de la Salud (CICSA) Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte. Av. Universidad Anáhuac 46 Col. Lomas Anáhuac Huixquilucan, Estado de México 52786, México.
| | - Jorge Alberto Ascencio-Aragón
- Departamento de Microbiología del Centro de Investigación en Ciencias de la Salud (CICSA) Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte. Av. Universidad Anáhuac 46 Col. Lomas Anáhuac Huixquilucan, Estado de México 52786, México.
| | - Sebastian Niezen-Lugo
- Departamento de Microbiología del Centro de Investigación en Ciencias de la Salud (CICSA) Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte. Av. Universidad Anáhuac 46 Col. Lomas Anáhuac Huixquilucan, Estado de México 52786, México.
| | - Rosalino Vázquez-López
- Departamento de Microbiología del Centro de Investigación en Ciencias de la Salud (CICSA) Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte. Av. Universidad Anáhuac 46 Col. Lomas Anáhuac Huixquilucan, Estado de México 52786, México.
| |
Collapse
|