1
|
Sun P, Huang H, Ma JC, Feng B, Zhang Y, Qin G, Zeng W, Cui ZK. Repurposing propofol for breast cancer therapy through promoting apoptosis and arresting cell cycle. Oncol Rep 2024; 52:155. [PMID: 39364744 PMCID: PMC11465104 DOI: 10.3892/or.2024.8814] [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: 02/23/2024] [Accepted: 06/17/2024] [Indexed: 10/05/2024] Open
Abstract
Breast cancer is the most prevalent cancer among women worldwide, characterized by a high mortality rate and propensity for metastasis. Although surgery is the standard treatment for breast cancer, there is still no effective method to inhibit tumor metastasis and improve the prognosis of patients with breast cancer after surgery. Propofol, one of the most widely used intravenous anesthetics in surgery, has exhibited a positive association with improved survival outcomes in patients with breast cancer post‑surgery. However, the underlying molecular mechanism remains to be elucidated. The present study revealed that triple negative breast cancer cells, MDA‑MB‑231 and 4T1, exposed to propofol exhibited a significant decrease in cell viability. Notably, propofol exhibited minimal cytotoxic effects on HUVECs under the same conditions. Furthermore, propofol significantly inhibited the migration and invasion ability of MDA‑MB‑231 and 4T1 cells. Propofol promoted apoptosis in 4T1 cells through upregulation of Bax and cleaved caspase 3, while downregulating B‑cell lymphoma‑extra large. Concomitantly, propofol induced cell cycle arrest of 4T1 cells by downregulating cyclin E2 and phosphorylated cell division cycle 6. Furthermore, propofol exhibited excellent anticancer efficacy in a 4T1 breast cancer allograft mouse model. The present study sheds light on the potential of propofol as an old medicine with a novel use for breast cancer treatment.
Collapse
Affiliation(s)
- Peng Sun
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Hanqing Huang
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jian-Chao Ma
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Binyang Feng
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yiqing Zhang
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Genggeng Qin
- Department of Radiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Weian Zeng
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Zhong-Kai Cui
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
2
|
Huang Y, Lei Y, Gong Z, Wang Y, Li M, Zhao D, Song L. Feeding Eucommia ulmoides extract enhances protection against high-temperature stress in chicks. Poult Sci 2024; 103:103827. [PMID: 38801811 PMCID: PMC11150962 DOI: 10.1016/j.psj.2024.103827] [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: 02/06/2024] [Revised: 04/18/2024] [Accepted: 05/02/2024] [Indexed: 05/29/2024] Open
Abstract
Chick's susceptibility to heat stress often leads to growth retardation, immune function impairment, disease, and mortality. This thesis explores the potential ameliorative effect of 0.8% Eucommia ulmoides extract (EUE) into the diet of heat-stressed chicks in a 15-d feeding trial. The investigation reveals that feeding EUE significantly enhances the BW, ADG, AFI, and F/G of chicks experiencing heat stress. Additionally, the EUE groups exhibited higher levels of T-AOC (at 7 and 15d), SOD (at 15 d), GSH-Px (at 15 d), as well as lower MDA concentrations (at 7 and 15d) in chick serum. Pathological changes and H&E staining revealed that EUE effectively improved tissue damage in the duodenum, heart, and stomach induced by heat stress in the chicks. The EUE groups also showed higher levels of IgA (at 7 d), IgG and IgM (at 7 and 15 d). RNA-seq and WGCNA analysis revealed that EUE mitigates cellular damage and losses in heat-stressed chicks primarily through pathways involving signal transduction, protein synthesis and degradation, as well as cell cycle regulation, particularly the latter. This investigation serves as a fundamental and cognitive framework for the development and application of Eucommia ulmoides feed additives aimed at safeguarding the well-being of chicks in adverse environmental conditions.
Collapse
Affiliation(s)
- Youwen Huang
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-Bioengineering, Guizhou University, Guiyang, Guizhou Province 550025, China; Guizhou Key Lab of Agro-Bioengineering, Guiyang, Guizhou Province 550025, China; National-Local Joint Engineering Research Center of Karst Region Plant Resources Utilization & Breeding (Guizhou), Guiyang, Guizhou Province 550025, China
| | - Yue Lei
- Guizhou Institute of Subtropical Crops, Xingyi, Guizhou Province 562400, China
| | - Zouxian Gong
- Clinical Medical College of Guizhou Medical University, Guiyang, Guizhou Province 550004, China
| | - Yifan Wang
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-Bioengineering, Guizhou University, Guiyang, Guizhou Province 550025, China; Guizhou Key Lab of Agro-Bioengineering, Guiyang, Guizhou Province 550025, China; National-Local Joint Engineering Research Center of Karst Region Plant Resources Utilization & Breeding (Guizhou), Guiyang, Guizhou Province 550025, China
| | - Minxue Li
- Agricultural and Rural Bureau, Shuicheng District, Liupanshui City, Guizhou Province 553040, China
| | - Degang Zhao
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-Bioengineering, Guizhou University, Guiyang, Guizhou Province 550025, China; National-Local Joint Engineering Research Center of Karst Region Plant Resources Utilization & Breeding (Guizhou), Guiyang, Guizhou Province 550025, China; Guizhou Academy of Agricultural Science, Guiyang, Guizhou Province 550006, China
| | - Li Song
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-Bioengineering, Guizhou University, Guiyang, Guizhou Province 550025, China; Guizhou Key Lab of Agro-Bioengineering, Guiyang, Guizhou Province 550025, China; National-Local Joint Engineering Research Center of Karst Region Plant Resources Utilization & Breeding (Guizhou), Guiyang, Guizhou Province 550025, China.
| |
Collapse
|
3
|
Jabarin A, Shtar G, Feinshtein V, Mazuz E, Shapira B, Ben-Shabat S, Rokach L. Eravacycline, an antibacterial drug, repurposed for pancreatic cancer therapy: insights from a molecular-based deep learning model. Brief Bioinform 2024; 25:bbae108. [PMID: 38647152 PMCID: PMC11033730 DOI: 10.1093/bib/bbae108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/04/2024] [Accepted: 02/25/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) remains a serious threat to health, with limited effective therapeutic options, especially due to advanced stage at diagnosis and its inherent resistance to chemotherapy, making it one of the leading causes of cancer-related deaths worldwide. The lack of clear treatment directions underscores the urgent need for innovative approaches to address and manage this deadly condition. In this research, we repurpose drugs with potential anti-cancer activity using machine learning (ML). METHODS We tackle the problem by using a neural network trained on drug-target interaction information enriched with drug-drug interaction information, which has not been used for anti-cancer drug repurposing before. We focus on eravacycline, an antibacterial drug, which was selected and evaluated to assess its anti-cancer effects. RESULTS Eravacycline significantly inhibited the proliferation and migration of BxPC-3 cells and induced apoptosis. CONCLUSION Our study highlights the potential of drug repurposing for cancer treatment using ML. Eravacycline showed promising results in inhibiting cancer cell proliferation, migration and inducing apoptosis in PDAC. These findings demonstrate that our developed ML drug repurposing models can be applied to a wide range of new oncology therapeutics, to identify potential anti-cancer agents. This highlights the potential and presents a promising approach for identifying new therapeutic options.
Collapse
Affiliation(s)
- Adi Jabarin
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev (BGU), P.O.B. 653, Beer-Sheva 8410501, Israel
| | - Guy Shtar
- Department of Information Systems and Software Engineering, Ben-Gurion University of the Negev, P.O.B. 653, Beer-Sheva 8410501, Israel
| | - Valeria Feinshtein
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev (BGU), P.O.B. 653, Beer-Sheva 8410501, Israel
| | - Eyal Mazuz
- Department of Information Systems and Software Engineering, Ben-Gurion University of the Negev, P.O.B. 653, Beer-Sheva 8410501, Israel
| | - Bracha Shapira
- Department of Information Systems and Software Engineering, Ben-Gurion University of the Negev, P.O.B. 653, Beer-Sheva 8410501, Israel
| | - Shimon Ben-Shabat
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev (BGU), P.O.B. 653, Beer-Sheva 8410501, Israel
| | - Lior Rokach
- Department of Information Systems and Software Engineering, Ben-Gurion University of the Negev, P.O.B. 653, Beer-Sheva 8410501, Israel
| |
Collapse
|
4
|
Li F, Ling YP, Wang P, Gu SS, Jiang H, Zhu J. Downregulation of miR-503-5p Promotes the Development of Pancreatic Cancer by Targeting Cyclin E2. Crit Rev Immunol 2024; 44:51-60. [PMID: 38505921 DOI: 10.1615/critrevimmunol.2024051136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
This study aimed to elucidate the role of microRNA-503 (miR-503) in pancreatic cancer (PC) progression and the underlying regulatory mechanisms. We acquired miR-503-3p and miR-503-5p expression data along with survival times of PC and normal samples from the UCSC Xena database. Using the t-test, we compared the expression of miR-503-3p and miR-503-5p between PC and normal samples, and evaluated their prognostic significance via Kaplan-Meier survival analysis. The expression of miR-503-5p in PC cells was detected by quantitative PCR. We subsequently overexpressed miR-503-5p in PC cells and examined cell viability, apoptosis, and migration through CCK8 assay, flow cytometry, and Transwell assay, respectively. Potential functional targets were identified using miRTarBase and validated by dual-luciferase reporter assay. Both miR-503-3p and miR-503-5p expression were found to be downregulated in PC; however, only miR-503-5p was linked to cancer prognosis based on public data. In vitro experiments demonstrated that overexpression of miR-503-5p substantially decreased cell viability, induced apoptosis, caused G0/G1 arrest, and inhibited cell migration. miR-503-5p was found to target cyclin E2 (CCNE2), and overexpression of CCNE2 could counteract the effects of miR-503-5p on PC cells. Conclusion: The downregulation of miR-503-5p enhances the progression of PC by targeting CCNE2. The detection of miR-503-5p expression may provide valuable insights for the prevention and prognostic evaluation of PC.
Collapse
Affiliation(s)
- Fei Li
- Department of Clinical Laboratory, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Ying-Pei Ling
- Department of Clinical Laboratory, Taizhou Hospital of Traditional Chinese Medicine, Taizhou, Zhejiang, China
| | - Pan Wang
- Department of Clinical Laboratory, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Shi-Sheng Gu
- Department of Gastroenterology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Hao Jiang
- Department of Hepatobiliary Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Jie Zhu
- Taizhou Central Hospital(Taizhou University Hospital)
| |
Collapse
|
5
|
Koch DT, Yu H, Beirith I, Schirren M, Drefs M, Liu Y, Knoblauch M, Koliogiannis D, Sheng W, De Toni EN, Bazhin AV, Renz BW, Guba MO, Werner J, Ilmer M. Tigecycline causes loss of cell viability mediated by mitochondrial OXPHOS and RAC1 in hepatocellular carcinoma cells. J Transl Med 2023; 21:876. [PMID: 38041179 PMCID: PMC10693093 DOI: 10.1186/s12967-023-04615-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 10/11/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND Despite recent advances in locoregional, systemic, and novel checkpoint inhibitor treatment, hepatocellular carcinoma (HCC) is still associated with poor prognosis. The feasibility of potentially curative liver resection (LR) and transplantation (LT) is limited by the underlying liver disease and a shortage of organ donors. Especially after LR, high recurrence rates present a problem and circulating tumor cells are a major cause of extrahepatic recurrence. Tigecycline, a commonly used glycylcycline antibiotic, has been shown to have antitumorigenic effects and could be used as a perioperative and adjuvant therapeutic strategy to target circulating tumor cells. We aimed to investigate the effect of tigecycline on HCC cell lines and its mechanisms of action. METHODS Huh7, HepG2, Hep3B, and immortalized hepatocytes underwent incubation with clinically relevant tigecycline concentrations, and the influence on proliferation, migration, and invasion was assessed in two- and three-dimensional in vitro assays, respectively. Bioinformatic analysis was used to identify specific targets of tigecycline. The expression of RAC1 was detected using western blot, RT-PCR and RNA sequencing. ELISA and flow cytometry were utilized to measure reactive oxygen species (ROS) generation upon tigecycline treatment and flow cytometry to detect alterations in cell cycle. Changes in mitochondrial function were detected via seahorse analysis. RNA sequencing was performed to examine involved pathways. RESULTS Tigecycline treatment resulted in a significant reduction of mitochondrial function with concomitantly preserved mitochondrial size, which preceded the observed decrease in HCC cell viability. The sensitivity of HCC cells to tigecycline treatment was higher than that of immortalized non-cancerous THLE-2 hepatocytes. Tigecycline inhibited both migratory and invasive properties. Tigecycline application led to an increase of detected ROS and an S-phase cell cycle arrest. Bioinformatic analysis identified RAC1 as a likely target for tigecycline and the expression of this molecule was increased in HCC cells as a result of tigecycline treatment. CONCLUSION Our study provides evidence for the antiproliferative effect of tigecycline in HCC. We show for the first time that this effect, likely to be mediated by reduced mitochondrial function, is associated with increased expression of RAC1. The reported effects of tigecycline with clinically relevant and achievable doses on HCC cells lay the groundwork for a conceivable use of this agent in cancer treatment.
Collapse
Affiliation(s)
- Dominik T Koch
- Department of General, Visceral and Transplantation Surgery, LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, Marchioninistr. 15, 81377, Munich, Germany
- Transplantation Center Munich, LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
- Liver Center Munich, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Haochen Yu
- Department of General, Visceral and Transplantation Surgery, LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Iris Beirith
- Department of General, Visceral and Transplantation Surgery, LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Malte Schirren
- Department of General, Visceral and Transplantation Surgery, LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, Marchioninistr. 15, 81377, Munich, Germany
- Transplantation Center Munich, LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
- Liver Center Munich, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Moritz Drefs
- Department of General, Visceral and Transplantation Surgery, LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, Marchioninistr. 15, 81377, Munich, Germany
- Transplantation Center Munich, LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
- Liver Center Munich, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Yunfei Liu
- Department of General, Visceral and Transplantation Surgery, LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Mathilda Knoblauch
- Department of General, Visceral and Transplantation Surgery, LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Dionysios Koliogiannis
- Department of General, Visceral and Transplantation Surgery, LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, Marchioninistr. 15, 81377, Munich, Germany
- Transplantation Center Munich, LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
- Liver Center Munich, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Weiwei Sheng
- Department of Gastrointestinal Surgery, The First Hospital, China Medical University, Shenyang, 110001, China
| | - Enrico N De Toni
- Liver Center Munich, Ludwig-Maximilians-University Munich, Munich, Germany
- Department of Internal Medicine II, LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Alexandr V Bazhin
- Department of General, Visceral and Transplantation Surgery, LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, Marchioninistr. 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), DKTK Partner Site Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Bernhard W Renz
- Department of General, Visceral and Transplantation Surgery, LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, Marchioninistr. 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), DKTK Partner Site Munich, Munich, Germany
- Transplantation Center Munich, LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
- Liver Center Munich, Ludwig-Maximilians-University Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Markus O Guba
- Department of General, Visceral and Transplantation Surgery, LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, Marchioninistr. 15, 81377, Munich, Germany
- Transplantation Center Munich, LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
- Liver Center Munich, Ludwig-Maximilians-University Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Jens Werner
- Department of General, Visceral and Transplantation Surgery, LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, Marchioninistr. 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), DKTK Partner Site Munich, Munich, Germany
- Transplantation Center Munich, LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
- Liver Center Munich, Ludwig-Maximilians-University Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Matthias Ilmer
- Department of General, Visceral and Transplantation Surgery, LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, Marchioninistr. 15, 81377, Munich, Germany.
- German Cancer Consortium (DKTK), DKTK Partner Site Munich, Munich, Germany.
- Transplantation Center Munich, LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany.
- Liver Center Munich, Ludwig-Maximilians-University Munich, Munich, Germany.
- Bavarian Cancer Research Center (BZKF), Munich, Germany.
| |
Collapse
|
6
|
Zhang X, Dong Z, Yang Y, Liu C, Li J, Sun W, Zhu Y, Shen Y, Wang Z, Lü M, Cui H. Morusinol Extracted from Morus alba Inhibits Cell Proliferation and Induces Autophagy via FOXO3a Nuclear Accumulation-Mediated Cholesterol Biosynthesis Obstruction in Colorectal Cancer. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:16016-16031. [PMID: 37870273 DOI: 10.1021/acs.jafc.3c01244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
The incidence rate of colorectal cancer (CRC) has been increasing significantly in recent years, and it is urgent to develop novel drugs that have more effects for its treatment. It has been reported that many molecules extracted from the root bark of Morus alba L. (also known as Cortex Mori) have antitumor activities. In our study, we identified morusinol as a promising anticancer agent by selecting from 30 molecules extracted from Morus alba L. We found that morusinol treatment suppressed cell proliferation and promoted apoptosis of CRC cells in vitro. Besides this, we observed that morusinol induced cytoprotective autophagy. The GO analysis of differentially expressed genes from RNA-seq data showed that morusinol affected cholesterol metabolism. Then we found that key enzyme genes in the cholesterol biosynthesis pathway as well as the sterol regulatory element binding transcription factor 2 (SREBF2) were significantly downregulated. Furthermore, additional cholesterol treatment reversed the anti-CRC effect of morusinol. Interestingly, we also found that morusinol treatment could promote forkhead box O3 (FOXO3a) nuclear accumulation, which subsequently suppressed SREBF2 transcription. Then SREBF2-controlled cholesterol biosynthesis was blocked, resulting in the suppression of cell proliferation, promotion of apoptosis, and production of autophagy. The experiments in animal models also showed that morusinol significantly impeded tumor growth in mice models. Our results suggested that morusinol may be used as a candidate anticancer drug for the treatment of CRC.
Collapse
Affiliation(s)
- Xiaolin Zhang
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Zhen Dong
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400716, China
- Jinfeng Laboratory, Chongqing 401329, China
- Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Beibei, Chongqing 400716, China
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Beibei, Chongqing 400716, China
| | - Yuanmiao Yang
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Chaolong Liu
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Jisheng Li
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Wenli Sun
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Yikang Zhu
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Yang Shen
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Zhi Wang
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Muhan Lü
- Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Hongjuan Cui
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400716, China
- Jinfeng Laboratory, Chongqing 401329, China
- Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Beibei, Chongqing 400716, China
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Beibei, Chongqing 400716, China
| |
Collapse
|
7
|
Li J, Qin Y, Zhao C, Zhang Z, Zhou Z. Tetracycline antibiotics: Potential anticancer drugs. Eur J Pharmacol 2023; 956:175949. [PMID: 37541377 DOI: 10.1016/j.ejphar.2023.175949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/22/2023] [Accepted: 08/01/2023] [Indexed: 08/06/2023]
Abstract
In recent years, research on tetracycline antibiotics has gradually shifted from their antibacterial effects to anticancer effects. Doxycycline, minocycline, and tigecycline as the US Food and Drug Administration (FDA) approved tetracycline antibiotics have been the main subjects of studies. Evidence indicated that they have anticancer properties and are able to control cancer progression through different mechanisms, such as anti-proliferation, anti-metastasis, and promotion of autophagy or apoptosis. In addition, studies have shown that these three tetracycline antibiotics can be utilized in conjunction with chemotherapeutic and targeted drugs to inhibit cancer progression and improve the quality of patient survival. Therefore, doxycycline, minocycline, and tigecycline are taken as examples in this work. Their mechanisms of action in different cancers and related combination therapies are introduced. Their current roles in alleviating the suffering of patients undergoing chemotherapy when used as adjuvant drugs in clinical treatment are also described. Finally, the research gaps and potential research directions at this stage are briefly summarized.
Collapse
Affiliation(s)
- Jiayu Li
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yuan Qin
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China; College of Pharmacy, Nankai University, China
| | - Chenhao Zhao
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Zhi Zhang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Zhiruo Zhou
- School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou, China.
| |
Collapse
|
8
|
Guo L, Dong Z, Zhang X, Yang Y, Hu X, Ji Y, Li C, Wan S, Xu J, Liu C, Zhang Y, Liu L, Shi Y, Wu Z, Liu Y, Cui H. Morusinol extracted from Morus alba induces cell cycle arrest and apoptosis via inhibition of DNA damage response in melanoma by CHK1 degradation through the ubiquitin-proteasome pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 114:154765. [PMID: 37004403 DOI: 10.1016/j.phymed.2023.154765] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 03/06/2023] [Accepted: 03/12/2023] [Indexed: 06/19/2023]
Abstract
BACKGROUD Flavonoids have a variety of biological activities, such as anti-inflammation, anti-tumor, anti-thrombosis and so on. Morusinol, as a novel isoprene flavonoid extracted from Morus alba root barks, has the effects of anti-arterial thrombosis and anti-inflammatory in previous studies. However, the anti-cancer mechanism of morusinol remains unclear. PURPOSE In present study, we mainly studied the anti-tumor effect of morusinol and its mode of action in melanoma. METHODS The anti-cancer effect of morusinol on melanoma were evaluated by using the MTT, EdU, plate clone formation and soft agar assay. Flow cytometry was used for detecting cell cycle and apoptosis. The ɣ-H2AX immunofluorescence and the alkaline comet assay were used to detect DNA damage and the Western blotting analysis was used to investigate the expressions of DNA-damage related proteins. Ubiquitination and turnover of CHK1 were also detected by using the immunoprecipitation assay. The cell line-derived xenograft (CDX) mouse models were used in vivo to evaluate the effect of morusinol on tumorigenicity. RESULTS We demonstrated that morusinol not only had the ability to inhibit cell proliferation, but also induced cell cycle arrest at G0/G1 phase, caspase-dependent apoptosis and DNA damage in human melanoma cells. In addition, morusinol effectively inhibited the growth of melanoma xenografts in vivo. More strikingly, CHK1, which played an important role in maintaining the integrity of cell cycle, genomic stability and cell viability, was down-regulated in a dose- and time-dependent manner after morusinol treatment. Further research showed that CHK1 was degraded by the ubiquitin-proteasome pathway. Whereafter, morusinol-induced cell cycle arrest, apoptosis and DNA damage were partially salvaged by overexpressing CHK1 in melanoma cell lines. Herein, further experiments demonstrated that morusinol increased the sensitivity of dacarbazine (DTIC) to chemotherapy for melanoma in vitro and in vivo. CONCLUSION Morusinol induces CHK1 degradation through the ubiquitin-proteasome pathway, thereby inducing cell cycle arrest, apoptosis and DNA damage response in melanoma. Our study firstly provided a theoretical basis for morusinol to be a candidate drug for clinical treatment of cancer, such as melanoma, alone or combinated with dacarbazine.
Collapse
Affiliation(s)
- Leiyang Guo
- Department of Dermatology, The Third Hospital of Hebei Medical University, 050000, No.139 Ziqiang Road, Qiaoxi District, Shijiazhuang, Hebei 050051, China; State Key Laboratory of Resource Insects, Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Ministry of Education), Southwest University, Chongqing 400716, China
| | - Zhen Dong
- State Key Laboratory of Resource Insects, Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Ministry of Education), Southwest University, Chongqing 400716, China; Hospital of Southwest University, Medical Research Institute, Southwest University, Chongqing 400716, China; Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Beibei, Chongqing 400716, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Beibei, Chongqing 400716, China
| | - Xiaolin Zhang
- Hospital of Southwest University, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Yuanmiao Yang
- State Key Laboratory of Resource Insects, Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Ministry of Education), Southwest University, Chongqing 400716, China
| | - Xiaosong Hu
- State Key Laboratory of Resource Insects, Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Ministry of Education), Southwest University, Chongqing 400716, China
| | - Yacong Ji
- Department of Dermatology, The Third Hospital of Hebei Medical University, 050000, No.139 Ziqiang Road, Qiaoxi District, Shijiazhuang, Hebei 050051, China
| | - Chongyang Li
- State Key Laboratory of Resource Insects, Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Ministry of Education), Southwest University, Chongqing 400716, China
| | - Sicheng Wan
- State Key Laboratory of Resource Insects, Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Ministry of Education), Southwest University, Chongqing 400716, China
| | - Jie Xu
- State Key Laboratory of Resource Insects, Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Ministry of Education), Southwest University, Chongqing 400716, China
| | - Chaolong Liu
- State Key Laboratory of Resource Insects, Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Ministry of Education), Southwest University, Chongqing 400716, China
| | - Yanli Zhang
- Department of Dermatology, The Third Hospital of Hebei Medical University, 050000, No.139 Ziqiang Road, Qiaoxi District, Shijiazhuang, Hebei 050051, China
| | - Lichao Liu
- Department of Dermatology, The Third Hospital of Hebei Medical University, 050000, No.139 Ziqiang Road, Qiaoxi District, Shijiazhuang, Hebei 050051, China
| | - Yaqiong Shi
- Department of Dermatology, The Third Hospital of Hebei Medical University, 050000, No.139 Ziqiang Road, Qiaoxi District, Shijiazhuang, Hebei 050051, China
| | - Zonghui Wu
- Hospital of Southwest University, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Yaling Liu
- Department of Dermatology, The Third Hospital of Hebei Medical University, 050000, No.139 Ziqiang Road, Qiaoxi District, Shijiazhuang, Hebei 050051, China.
| | - Hongjuan Cui
- State Key Laboratory of Resource Insects, Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Ministry of Education), Southwest University, Chongqing 400716, China; Hospital of Southwest University, Medical Research Institute, Southwest University, Chongqing 400716, China; Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Beibei, Chongqing 400716, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Beibei, Chongqing 400716, China.
| |
Collapse
|
9
|
Ruiz-Malagón AJ, Hidalgo-García L, Rodríguez-Sojo MJ, Molina-Tijeras JA, García F, Diez-Echave P, Vezza T, Becerra P, Marchal JA, Redondo-Cerezo E, Hausmann M, Rogler G, Garrido-Mesa J, Rodríguez-Cabezas ME, Rodríguez-Nogales A, Gálvez J. Tigecycline reduces tumorigenesis in colorectal cancer via inhibition of cell proliferation and modulation of immune response. Biomed Pharmacother 2023; 163:114760. [PMID: 37119741 DOI: 10.1016/j.biopha.2023.114760] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/13/2023] [Accepted: 04/20/2023] [Indexed: 05/01/2023] Open
Abstract
BACKGROUND and Purpose: Colorectal cancer (CRC) is one of the cancers with the highest incidence in which APC gene mutations occur in almost 80% of patients. This mutation leads to β-catenin aberrant accumulation and an uncontrolled proliferation. Apoptosis evasion, changes in the immune response and microbiota composition are also events that arise in CRC. Tetracyclines are drugs with proven antibiotic and immunomodulatory properties that have shown cytotoxic activity against different tumor cell lines. EXPERIMENTAL APPROACH The effect of tigecycline was evaluated in vitro in HCT116 cells and in vivo in a colitis-associated colorectal cancer (CAC) murine model. 5-fluorouracil was assayed as positive control in both studies. KEY RESULTS Tigecycline showed an antiproliferative activity targeting the Wnt/β-catenin pathway and downregulating STAT3. Moreover, tigecycline induced apoptosis through extrinsic, intrinsic and endoplasmic reticulum pathways converging on an increase of CASP7 levels. Furthermore, tigecycline modulated the immune response in CAC, reducing the cancer-associated inflammation through downregulation of cytokines expression. Additionally, tigecycline favored the cytotoxic activity of cytotoxic T lymphocytes (CTLs), one of the main immune defenses against tumor cells. Lastly, the antibiotic reestablished the gut dysbiosis in CAC mice increasing the abundance of bacterial genera and species, such as Akkermansia and Parabacteroides distasonis, that act as protectors against tumor development. These findings resulted in a reduction of the number of tumors and an amelioration of the tumorigenesis process in CAC. CONCLUSION AND IMPLICATIONS Tigecycline exerts a beneficial effect against CRC supporting the use of this antibiotic for the treatment of this disease.
Collapse
Affiliation(s)
- Antonio Jesús Ruiz-Malagón
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain
| | - Laura Hidalgo-García
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain
| | - María Jesús Rodríguez-Sojo
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain
| | - José Alberto Molina-Tijeras
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain
| | - Federico García
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain; Servicio Microbiología, Hospital Universitario Clínico San Cecilio, 18100 Granada, Spain; Ciber de Enfermedades Infecciosas, CiberInfecc, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Patricia Diez-Echave
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain
| | - Teresa Vezza
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain
| | - Patricia Becerra
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain; Servicio de Anatomía Patológica, Hospital Universitario Clínico San Cecilio, 18014 Granada, Spain
| | - Juan Antonio Marchal
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain; Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, E-18100 Granada, Spain; Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada E-18016, Spain
| | - Eduardo Redondo-Cerezo
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain; Servicio de Aparato Digestivo. Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Martin Hausmann
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Gerhard Rogler
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091, Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057, Zurich, Switzerland
| | - José Garrido-Mesa
- The William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London, UK.
| | - María Elena Rodríguez-Cabezas
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain.
| | - Alba Rodríguez-Nogales
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain
| | - Julio Gálvez
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
10
|
Araújo D, Ribeiro E, Amorim I, Vale N. Repurposed Drugs in Gastric Cancer. MOLECULES (BASEL, SWITZERLAND) 2022; 28:molecules28010319. [PMID: 36615513 PMCID: PMC9822219 DOI: 10.3390/molecules28010319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 12/21/2022] [Accepted: 12/25/2022] [Indexed: 01/04/2023]
Abstract
Gastric cancer (GC) is one of the major causes of death worldwide, ranking as the fifth most incident cancer in 2020 and the fourth leading cause of cancer mortality. The majority of GC patients are in an advanced stage at the time of diagnosis, presenting a poor prognosis and outcome. Current GC treatment approaches involve endoscopic detection, gastrectomy and chemotherapy or chemoradiotherapy in an adjuvant or neoadjuvant setting. Drug development approaches demand extreme effort to identify molecular mechanisms of action of new drug candidates. Drug repurposing is based on the research of new therapeutic indications of drugs approved for other pathologies. In this review, we explore GC and the different drugs repurposed for this disease.
Collapse
Affiliation(s)
- Diana Araújo
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal
| | - Eduarda Ribeiro
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Irina Amorim
- Institute of Biomedical Sciences Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Correspondence: ; Tel.: +351-220426537
| |
Collapse
|
11
|
Yu J, Yin Y, Yu Y, Cheng M, Zhang S, Jiang S, Dong M. Effect of concomitant antibiotics use on patient outcomes and adverse effects in patients treated with ICIs. Immunopharmacol Immunotoxicol 2022; 45:386-394. [PMID: 36382735 DOI: 10.1080/08923973.2022.2145966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Jiuhang Yu
- College of Pharmacy, Jiamusi University, Jiamusi, China
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yichuang Yin
- College of Pharmacy, Jiamusi University, Jiamusi, China
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yang Yu
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Mengfei Cheng
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shuo Zhang
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shuai Jiang
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Mei Dong
- College of Pharmacy, Jiamusi University, Jiamusi, China
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
12
|
Hu X, Zhang K, Pan G, Wang Y, Shen Y, Peng C, Deng L, Cui H. Cortex Mori extracts induce apoptosis and inhibit tumor invasion via blockage of the PI3K/AKT signaling in melanoma cells. Front Pharmacol 2022; 13:1007279. [PMID: 36339598 PMCID: PMC9627489 DOI: 10.3389/fphar.2022.1007279] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 10/05/2022] [Indexed: 08/22/2023] Open
Abstract
Melanoma, the most aggressive and deadliest form of skin cancer, has attracted increased attention due to its increasing incidence worldwide. The Cortex Mori (CM) has long been used as a classical traditional Chinese medicine (TCM) to treat various diseases, including cancer. The bioactive components and underlying mechanisms, however, remain largely unknown. The current study aims to investigate the anti-melanoma effects of CM and potential mechanisms through combined network pharmacology and bioinformatic analyses, and validated by in vitro and in vivo experiments. We report here that CM has anti-melanoma activity both in vitro and in vivo. Furthermore, 25 bioactive compounds in CM were found to share 142 melanoma targets, and network pharmacology and enrichment analyses suggested that CM inhibits melanoma through multiple biological processes and signaling pathways, particularly the PI3K-AKT signaling inhibition and activation of apoptotic pathways, which were further confirmed by biochemical and histological examinations. Finally, partial CM-derived bioactive compounds were found to show anti-melanoma effects, validating the anti-melanoma potential of bioactive ingredients of CM. Taken together, these results reveal bioactive components and mechanisms of CM in inhibiting melanoma, providing them as potential anti-cancer natural products for the treatment of melanoma.
Collapse
Affiliation(s)
- Xin Hu
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing, China
- Southwest University Engineering Research Center for Cancer Biomedical and Translational Medicine, Chongqing, China
| | - Kui Zhang
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing, China
- Southwest University Engineering Research Center for Cancer Biomedical and Translational Medicine, Chongqing, China
| | - Guangzhao Pan
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing, China
- Southwest University Engineering Research Center for Cancer Biomedical and Translational Medicine, Chongqing, China
| | - Yinggang Wang
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing, China
- Southwest University Engineering Research Center for Cancer Biomedical and Translational Medicine, Chongqing, China
| | - Yue Shen
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing, China
- Southwest University Engineering Research Center for Cancer Biomedical and Translational Medicine, Chongqing, China
| | - Cheng Peng
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing, China
- Southwest University Engineering Research Center for Cancer Biomedical and Translational Medicine, Chongqing, China
| | - Longfei Deng
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing, China
- Southwest University Engineering Research Center for Cancer Biomedical and Translational Medicine, Chongqing, China
| |
Collapse
|
13
|
Deng L, Jiang A, Zeng H, Peng X, Song L. Comprehensive analyses of PDHA1 that serves as a predictive biomarker for immunotherapy response in cancer. Front Pharmacol 2022; 13:947372. [PMID: 36003495 PMCID: PMC9393251 DOI: 10.3389/fphar.2022.947372] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 06/27/2022] [Indexed: 01/10/2023] Open
Abstract
Recent studies have proposed that pyruvate dehydrogenase E1 component subunit alpha (PDHA1), a cuproptosis-key gene, is crucial to the glucose metabolism reprogram of tumor cells. However, the functional roles and regulated mechanisms of PDHA1 in multiple cancers are largely unknown. The Cancer Genome Atlas (TCGA), GEPIA2, and cBioPortal databases were utilized to elucidate the function of PDHA1 in 33 tumor types. We found that PDHA1 was aberrantly expressed in most cancer types. Lung adenocarcinoma (LUAD) patients with high PDHA1 levels were significantly correlated with poor prognosis of overall survival (OS) and first progression (FP). Kidney renal clear cell carcinoma (KIRC) patients with low PDHA1 levels displayed poor OS and disease-free survival (DFS). However, for stomach adenocarcinoma (STAD), the downregulated PDHA1 expression predicted a good prognosis in patients. Moreover, we evaluated the mutation diversity of PDHA1 in cancers and their association with prognosis. We also analyzed the protein phosphorylation and DNA methylation of PDHA1 in various tumors. The PDHA1 expression was negatively correlated with tumor-infiltrating immune cells, such as myeloid dendritic cells (DCs), B cells, and T cells in pan-cancers. Mechanically, we used single-cell sequencing to discover that the PDHA1 expression had a close link with several cancer-associated signaling pathways, such as DNA damage, cell invasion, and angiogenesis. At last, we conducted a co-expressed enrichment analysis and showed that aberrantly expressed PDHA1 participated in the regulation of mitochondrial signaling pathways, including oxidative phosphorylation, cellular respiration, and electron transfer activity. In summary, PDHA1 could be a prognostic and immune-associated biomarker in multiple cancers.
Collapse
Affiliation(s)
- Langmei Deng
- Department of Emergency, The Third Xiangya Hospital, Central South University, Changsha, HN, China
| | - Anqi Jiang
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, HN, China
| | - Hanqing Zeng
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, HN, China
| | - Xiaoji Peng
- Department of Pharmacy, Yueyang Hospital of Traditional Chinese Medicine, Yueyang, HN, China
| | - Liying Song
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, HN, China
- *Correspondence: Liying Song,
| |
Collapse
|
14
|
Feng Z, Chen Y, Cai C, Tan J, Liu P, Chen Y, Shen H, Zeng S, Han Y. Pan-Cancer and Single-Cell Analysis Reveals CENPL as a Cancer Prognosis and Immune Infiltration-Related Biomarker. Front Immunol 2022; 13:916594. [PMID: 35844598 PMCID: PMC9279617 DOI: 10.3389/fimmu.2022.916594] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 05/24/2022] [Indexed: 01/22/2023] Open
Abstract
Background Centromere protein L (CENPL) is an important member of the centromere protein (CENP) family. However, the correlation between CENPL expression and cancer development and immune infiltration has rarely been studied. Here, we studied the role of CENPL in pan-cancer and further verified the results in lung adenocarcinoma (LUAD) through in vitro experiments. Methods The CENPL expression level was studied with TIMER 2.0 and Oncomine databases. The potential value of CENPL as a diagnostic and prognostic biomarker in pan-cancer was evaluated with the TCGA database and GEPIA. The CENPL mutation character was analyzed using the cBioPortal database. The LinkedOmics and CancerSEA databases were used to carry out the function analysis of CENPL. The role of CENPL in immune infiltration was studied using the TIMER and TISIDB websites. Moreover, the expression of CENPL was detected through RT-qPCR and Western blotting. Immunohistochemistry was used to evaluate the infiltration level of CD8+ T cells. Cell proliferation was detected by EdU and CCK8. A flow cytometer was used to analyze the influence of CENPL in cell cycle and apoptosis. Results CENPL was increased in most of the cancers. The upregulation and mutation of CENPL were associated with a poorer prognosis in many cancers. The results showed a significant positive correlation between CENPL and myeloid-derived suppressor cell (MDSC) infiltration and a negative correlation between CENPL and T-cell NK infiltration in most of the cancers. CENPL regulated cell proliferation and cell cycle, and was negatively correlated with the inflammation level of LUAD. The in vitro experiments suggested that CENPL was increased in LUAD tissue and cell lines. There was a negative correlation between CENPL expression and CD8+ T-cell infiltration. The knockdown of CENPL significantly suppressed the expression of CDK2 and CCNE2, and induced G0/G1 arrest and apoptosis of LUAD. Conclusions CENPL may function as a potential biomarker and oncogene in pan-cancer, especially LUAD. Furthermore, CENPL was associated with immune cell infiltration in pan-cancer, providing a potential immune therapy target for tumor treatment.
Collapse
Affiliation(s)
- Ziyang Feng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Yu Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Changjing Cai
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Jun Tan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Ping Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Yihong Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Hong Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Shan Zeng, ; Ying Han,
| | - Ying Han
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Shan Zeng, ; Ying Han,
| |
Collapse
|
15
|
Tan P, Li M, Liu Z, Li T, Zhao L, Fu W. Glycolysis-Related LINC02432/Hsa-miR-98–5p/HK2 Axis Inhibits Ferroptosis and Predicts Immune Infiltration, Tumor Mutation Burden, and Drug Sensitivity in Pancreatic Adenocarcinoma. Front Pharmacol 2022; 13:937413. [PMID: 35795552 PMCID: PMC9251347 DOI: 10.3389/fphar.2022.937413] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/02/2022] [Indexed: 11/30/2022] Open
Abstract
Pancreatic adenocarcinoma (PAAD) is a malignant cancer with high incidence and mortality. Glycometabolic rearrangements (aerobic glycolysis) is a hallmark of PAAD and contributes to tumorigenesis and progression through numerous mechanisms. This study aimed to identify a novel glycolysis-related lncRNA-miRNA-mRNA ceRNA signature in PAAD and explore its potential molecular function. We first calculated the glycolysis score for each PAAD patient by the ssGSEA algorithm and found that patients with higher hallmark glycolysis scores had poorer prognosis. Subsequently, we obtained a novel glycolysis-related LINC02432/hsa-miR-98–5p/HK2 axis from the TCGA and GEO databases using comprehensive bioinformatics analysis and developed a nomogram to predict overall survival. Furthermore, functional characterization analysis revealed that LINC02432/hsa-miR-98–5p/HK2 axis risk score was negatively correlated with ferroptosis. The tumor immune infiltration analysis suggested positive correlations between ceRNA risk score and infiltrated M0 macrophage levels in PAAD. Correlation analysis found that ceRNA risk scores were positively correlated with four chemokines (CXCL3, CXCL5, CXCL8 and CCL20) and one immune checkpoint gene (SIGLEC15). Meanwhile, tumor mutation burden (TMB), an indicator for predicting response to immunotherapy, was positively correlated with ceRNA risk score. Finally, the drug sensitivity analysis showed that the high-risk score patients might be more sensitive to EGFR, MEK and ERK inhibitors than low-risk score patients. In conclusion, our study suggested that LINC02432/hsa-miR-98–5p/HK2 axis may serve as a novel diagnostic, prognostic, and therapeutic target in PAAD treatment.
Collapse
Affiliation(s)
- Peng Tan
- Department of Cell Biology and Genetics / Institute of Genetics and Developmental Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Mo Li
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Zhuoran Liu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Tongxi Li
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Lingyu Zhao
- Department of Cell Biology and Genetics / Institute of Genetics and Developmental Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- *Correspondence: Lingyu Zhao, ; Wenguang Fu,
| | - Wenguang Fu
- Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, China
- *Correspondence: Lingyu Zhao, ; Wenguang Fu,
| |
Collapse
|
16
|
Wang S, Lv T, Chen Q, Yang Y, Xu L, Zhang X, Wang E, Hu X, Liu Y. Transcriptome sequencing and lncRNA-miRNA-mRNA network construction in cardiac fibrosis and heart failure. Bioengineered 2022; 13:7118-7133. [PMID: 35235759 PMCID: PMC8974171 DOI: 10.1080/21655979.2022.2045839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Cardiac fibrosis (CF) and heart failure (HF) are common heart diseases, and severe CF can lead to HF. In this study, we tried to find their common potential molecular markers, which may help the diagnosis and treatment of CF and HF. RNA library construction and high-throughput sequencing were performed. The DESeq2 package in R was used to screen differentially expressed mRNAs (DEmRNAs), differentially expressed lncRNA (DElncRNAs) and differentially expressed miRNA (DEmiRNAs) between different samples. The common DEmRNAs, DElncRNAs and DEmiRNAs for the two diseases were obtained. The ConsensusPathDB (CPDB) was used to perform biological function enrichment for common DEmRNAs. Gene interaction network was constructed to screen out key genes. Subsequently, real-time polymerase chain reaction (RT-PCR) verification was performed. Lastly, GSE104150 and GSE21125 data sets were utilized for expression validation and diagnostic analysis. There were 1477 DEmRNAs, 502 DElncRNAs and 36 DEmiRNAs between CF and healthy control group. There were 607 DEmRNAs, 379DElncRNAs,s and 42 DEmiRNAs between HF and healthy control group. CH and FH shared 146 DEmRNAs, 80 DElncRNAs, and 6 DEmiRNAs. Hsa-miR-144-3p, CCNE2, C9orf72, MAP3K20-AS1, LEF1-AS1, AC243772.2, FLJ46284, and AC239798.2 were key molecules in lncRNA-miRNA-mRNA network. In addition, hsa-miR-144-3p and CCNE2 may be considered as potential diagnostic gene biomarkers in HF. In this study, the identification of common biomarkers of CF and HF may help prevent CF to HF transition as early as possible.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhaung, HB, China
| | - Tianjie Lv
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhaung, HB, China
| | - Qincong Chen
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhaung, HB, China
| | - Yan Yang
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhaung, HB, China
| | - Lei Xu
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhaung, HB, China
| | - Xiaolei Zhang
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhaung, HB, China
| | - Enmao Wang
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhaung, HB, China
| | - Xitian Hu
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhaung, HB, China
| | - Yuying Liu
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhaung, HB, China
| |
Collapse
|
17
|
Li Y, Su Y, Zhao Y, Hu X, Zhao G, He J, Wan S, Lü M, Cui H. Demethylzeylasteral inhibits proliferation, migration, and invasion through FBXW7/c-Myc axis in gastric cancer. MedComm (Beijing) 2021; 2:467-480. [PMID: 34766156 PMCID: PMC8554662 DOI: 10.1002/mco2.73] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/05/2021] [Accepted: 04/07/2021] [Indexed: 12/14/2022] Open
Abstract
Gastric cancer (GC) is one of the most familiar malignancy in the digestive system. Demethylzeylasteral (Dem), a natural functional monomer extracted from Tripterygium wilfordii Hook F, shows anti‐tumor effects in a variety of cancers, including GC, however, with the underlying mechanism poorly understood. In our study, we show that Dem inhibits the proliferation, migration, and invasion of GC cells, which are mediated by down‐regulating c‐Myc protein levels. Mechanistically, Dem reduces the stability of c‐Myc by up‐regulating FBXW7, an E3 ubiquitin ligase. Moreover, in xenograft tumor model experiment, Dem also inhibits GC, which depends on suppressing c‐Myc expression. Finally, Dem enhances GC cell chemosensitivity to the combination treatment of 5‐Fluorouracil (5‐Fu) and doxorubicin (DOX) in vitro. Together, Dem exerts anti‐neoplastic activities through destabilizing and suppressing c‐Myc, establishing a theory foundation for using it in future treatment of GC.
Collapse
Affiliation(s)
- Yongsen Li
- State Key Laboratory of Silkworm Genome Biology College of Sericulture Textile and Biomass sciences Southwest University Chongqing China
| | - Yongyue Su
- Department of Orthopaedic 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army Kunming China
| | - Yuzu Zhao
- State Key Laboratory of Silkworm Genome Biology College of Sericulture Textile and Biomass sciences Southwest University Chongqing China
| | - Xiaosong Hu
- State Key Laboratory of Silkworm Genome Biology College of Sericulture Textile and Biomass sciences Southwest University Chongqing China
| | - Gaichao Zhao
- State Key Laboratory of Silkworm Genome Biology College of Sericulture Textile and Biomass sciences Southwest University Chongqing China
| | - Jiang He
- State Key Laboratory of Silkworm Genome Biology College of Sericulture Textile and Biomass sciences Southwest University Chongqing China
| | - Sicheng Wan
- State Key Laboratory of Silkworm Genome Biology College of Sericulture Textile and Biomass sciences Southwest University Chongqing China
| | - Muhan Lü
- Department of Gastroenterology The Affiliated Hospital of Southwest Medical University Luzhou China
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology College of Sericulture Textile and Biomass sciences Southwest University Chongqing China.,Department of Gastroenterology The Affiliated Hospital of Southwest Medical University Luzhou China.,Cancer Centre Medical Research Institute Southwest University Chongqing China
| |
Collapse
|
18
|
Liao R, Xie B, Cui J, Qi Z, Xue S, Wang Y. E2F transcription factor 1 (E2F1) promotes the transforming growth factor TGF-β1 induced human cardiac fibroblasts differentiation through promoting the transcription of CCNE2 gene. Bioengineered 2021; 12:6869-6877. [PMID: 34521301 PMCID: PMC8806588 DOI: 10.1080/21655979.2021.1972194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The differentiation of cardiac fibroblast to myofibroblast is the key process of cardiac fibrosis. In the study, we aimed to determine the function of E2F Transcription Factor 1 (E2F1) in human cardiac fibroblasts (HCFs) differentiation, search for its downstream genes and elucidate the function of them in HCFs differentiation. As a result, we found that E2F1 was up-regulated in TGF-β1-induced HCFs differentiation. Silencing the expression of E2F1 by siRNA in HCFs, we found that the expression of differentiation-related genes (Collagen-1, α-Smooth muscle actin, and Fibronectin-1) was significantly suppressed, combining with proliferation and migration assay, we determined that HCFs differentiation was decreased. Luciferase report assay and immunoprecipitation proved that the oncogene CCNE2 was a direct target gene of E2F1, overexpression of CCNE2 was found in differentiated HCFs, silencing the expression of CCNE2 by siRNA decreased HCFs differentiation. Our research suggested that E2F1 and its downstream target gene CCNE2 play a vital role in TGF-β1-induced HCFs differentiation, thus E2F1 and CCNE2 may be a potential therapeutic target for cardiac fibrosis.
Collapse
Affiliation(s)
- Rongheng Liao
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bo Xie
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Cui
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zhen Qi
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Song Xue
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yongyi Wang
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
19
|
Barman S, Fatima I, Singh AB, Dhawan P. Pancreatic Cancer and Therapy: Role and Regulation of Cancer Stem Cells. Int J Mol Sci 2021; 22:ijms22094765. [PMID: 33946266 PMCID: PMC8124621 DOI: 10.3390/ijms22094765] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/20/2021] [Accepted: 04/20/2021] [Indexed: 12/21/2022] Open
Abstract
Despite significant improvements in clinical management, pancreatic cancer (PC) remains one of the deadliest cancer types, as it is prone to late detection with extreme metastatic properties. The recent findings that pancreatic cancer stem cells (PaCSCs) contribute to the tumorigenesis, progression, and chemoresistance have offered significant insight into the cancer malignancy and development of precise therapies. However, the heterogeneity of cancer and signaling pathways that regulate PC have posed limitations in the effective targeting of the PaCSCs. In this regard, the role for K-RAS, TP53, Transforming Growth Factor-β, hedgehog, Wnt and Notch and other signaling pathways in PC progression is well documented. In this review, we discuss the role of PaCSCs, the underlying molecular and signaling pathways that help promote pancreatic cancer development and metastasis with a specific focus on the regulation of PaCSCs. We also discuss the therapeutic approaches that target different PaCSCs, intricate mechanisms, and therapeutic opportunities to eliminate heterogeneous PaCSCs populations in pancreatic cancer.
Collapse
Affiliation(s)
- Susmita Barman
- Department of Biochemistry and Molecular Biology, Omaha, NE 68198, USA; (S.B.); (I.F.); (A.B.S.)
| | - Iram Fatima
- Department of Biochemistry and Molecular Biology, Omaha, NE 68198, USA; (S.B.); (I.F.); (A.B.S.)
| | - Amar B. Singh
- Department of Biochemistry and Molecular Biology, Omaha, NE 68198, USA; (S.B.); (I.F.); (A.B.S.)
- VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
- Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | - Punita Dhawan
- Department of Biochemistry and Molecular Biology, Omaha, NE 68198, USA; (S.B.); (I.F.); (A.B.S.)
- VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
- Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68105, USA
- Correspondence:
| |
Collapse
|
20
|
Li C, Zhang K, Pan G, Ji H, Li C, Wang X, Hu X, Liu R, Deng L, Wang Y, Yang L, Cui H. Dehydrodiisoeugenol inhibits colorectal cancer growth by endoplasmic reticulum stress-induced autophagic pathways. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:125. [PMID: 33838688 PMCID: PMC8035743 DOI: 10.1186/s13046-021-01915-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 03/16/2021] [Indexed: 12/11/2022]
Abstract
Background Dehydrodiisoeugenol (DEH), a novel lignan component extracted from nutmeg, which is the seed of Myristica fragrans Houtt, displays noticeable anti-inflammatory and anti-allergic effects in digestive system diseases. However, the mechanism of its anticancer activity in gastrointestinal cancer remains to be investigated. Methods In this study, the anticancer effect of DEH on human colorectal cancer and its underlying mechanism were evaluated. Assays including MTT, EdU, Plate clone formation, Soft agar, Flow cytometry, Electron microscopy, Immunofluorescence and Western blotting were used in vitro. The CDX and PDX tumor xenograft models were used in vivo. Results Our findings indicated that treatment with DEH arrested the cell cycle of colorectal cancer cells at the G1/S phase, leading to significant inhibition in cell growth. Moreover, DEH induced strong cellular autophagy, which could be inhibited through autophagic inhibitors, with a rction in the DEH-induced inhibition of cell growth in colorectal cancer cells. Further analysis indicated that DEH also induced endoplasmic reticulum (ER) stress and subsequently stimulated autophagy through the activation of PERK/eIF2α and IRE1α/XBP-1 s/CHOP pathways. Knockdown of PERK or IRE1α significantly decreased DEH-induced autophagy and retrieved cell viability in cells treated with DEH. Furthermore, DEH also exhibited significant anticancer activities in the CDX- and PDX-models. Conclusions Collectively, our studies strongly suggest that DEH might be a potential anticancer agent against colorectal cancer by activating ER stress-induced inhibition of autophagy. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-01915-9.
Collapse
Affiliation(s)
- Changhong Li
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass sciences, Southwest University, #2, Tiansheng Rd., Beibei District, Chongqing, 400716, China.,Cancer Centre, Medical Research Institute, Southwest University, Chongqing, 400716, China.,Affiliated Hospital of Southwest University (the Ninth People's Hospital of Chongqing), Chongqing, 400716, China
| | - Kui Zhang
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass sciences, Southwest University, #2, Tiansheng Rd., Beibei District, Chongqing, 400716, China.,Cancer Centre, Medical Research Institute, Southwest University, Chongqing, 400716, China
| | - Guangzhao Pan
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass sciences, Southwest University, #2, Tiansheng Rd., Beibei District, Chongqing, 400716, China.,Cancer Centre, Medical Research Institute, Southwest University, Chongqing, 400716, China
| | - Haoyan Ji
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass sciences, Southwest University, #2, Tiansheng Rd., Beibei District, Chongqing, 400716, China.,Cancer Centre, Medical Research Institute, Southwest University, Chongqing, 400716, China
| | - Chongyang Li
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass sciences, Southwest University, #2, Tiansheng Rd., Beibei District, Chongqing, 400716, China.,Cancer Centre, Medical Research Institute, Southwest University, Chongqing, 400716, China
| | - Xiaowen Wang
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass sciences, Southwest University, #2, Tiansheng Rd., Beibei District, Chongqing, 400716, China.,Cancer Centre, Medical Research Institute, Southwest University, Chongqing, 400716, China
| | - Xin Hu
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass sciences, Southwest University, #2, Tiansheng Rd., Beibei District, Chongqing, 400716, China.,Cancer Centre, Medical Research Institute, Southwest University, Chongqing, 400716, China
| | - Ruochen Liu
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass sciences, Southwest University, #2, Tiansheng Rd., Beibei District, Chongqing, 400716, China.,Cancer Centre, Medical Research Institute, Southwest University, Chongqing, 400716, China
| | - Longfei Deng
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass sciences, Southwest University, #2, Tiansheng Rd., Beibei District, Chongqing, 400716, China.,Cancer Centre, Medical Research Institute, Southwest University, Chongqing, 400716, China
| | - Yi Wang
- Affiliated Hospital of Southwest University (the Ninth People's Hospital of Chongqing), Chongqing, 400716, China
| | - Liqun Yang
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass sciences, Southwest University, #2, Tiansheng Rd., Beibei District, Chongqing, 400716, China. .,Cancer Centre, Medical Research Institute, Southwest University, Chongqing, 400716, China. .,Affiliated Hospital of Southwest University (the Ninth People's Hospital of Chongqing), Chongqing, 400716, China.
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass sciences, Southwest University, #2, Tiansheng Rd., Beibei District, Chongqing, 400716, China.,Cancer Centre, Medical Research Institute, Southwest University, Chongqing, 400716, China.,Affiliated Hospital of Southwest University (the Ninth People's Hospital of Chongqing), Chongqing, 400716, China
| |
Collapse
|
21
|
Shi S, Li C, Zhang Y, Deng C, Liu W, Du J, Li Q, Ji Y, Guo L, Liu L, Hu H, Liu Y, Cui H. Dihydrocapsaicin Inhibits Cell Proliferation and Metastasis in Melanoma via Down-regulating β-Catenin Pathway. Front Oncol 2021; 11:648052. [PMID: 33833997 PMCID: PMC8023049 DOI: 10.3389/fonc.2021.648052] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/04/2021] [Indexed: 11/13/2022] Open
Abstract
Dihydrocapsaicin (DHC) is one of the main components of capsaicinoids in Capsicum. It has been reported that DHC exerts anti-cancer effects on diverse malignant tumors, such as colorectal cancer, breast cancer, and glioma. However, studies focused on the effect of DHC upon melanoma have rarely been done. In the present study, melanoma A375 and MV3 cell lines were treated with DHC and the cell proliferation, migration, and invasion were significantly suppressed. Furthermore, DHC effectively inhibited xenograft tumor growth and pulmonary metastasis of melanoma cells in NOD/SCID mice model. It was identified that β-catenin, which plays significant roles in cell proliferation and epithelial-mesenchymal transition, was down-regulated after DHC treatment. In addition, cyclin D1, c-Myc, MMP2, and MMP7, which are critical in diverse cellular process regulation as downstream proteins of β-catenin, were all decreased. Mechanistically, DHC accelerates ubiquitination of β-catenin and up-regulates the beta-transducin repeat containing E3 ubiquitin protein ligase (BTRC) in melanoma cells. The DHC induced suppression of cell proliferation, migration, and invasion were partly rescued by exogenous β-catenin overexpression, both in vitro and in vivo. Taken together, DHC may serve as a candidate natural compound for human melanoma treatment through β-catenin pathway.
Collapse
Affiliation(s)
- Shaomin Shi
- Department of Dermatology, Third Hospital of Hebei Medical University, Shijiazhuang, China
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
- Department of Dermatology, Fifth Hospital of Shijiazhuang, Shijiazhuang, China
| | - Chongyang Li
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
| | - Yanli Zhang
- Department of Dermatology, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chaowei Deng
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
| | - Wei Liu
- Department of Dermatology, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Juan Du
- Department of Dermatology, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Qian Li
- Department of Dermatology, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yacong Ji
- Department of Dermatology, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Leiyang Guo
- Department of Dermatology, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lichao Liu
- Department of Dermatology, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Huanrong Hu
- Department of Dermatology, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yaling Liu
- Department of Dermatology, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
| |
Collapse
|
22
|
Yao B, Zhou Z, Zhang M, Leng X, Zhao D. Investigating the molecular control of deer antler extract on articular cartilage. J Orthop Surg Res 2021; 16:8. [PMID: 33407721 PMCID: PMC7788833 DOI: 10.1186/s13018-020-02148-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 12/02/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Deer antler is considered as a precious traditional Chinese medicinal material and has been widely used to reinforce kidney's yang, nourish essence, and strengthen bone function. The most prominent bioactive components in deer antler are water-soluble proteins that play potential roles in bone formation and repair. The aim of this study was to explore the molecular control and therapeutic targets of deer antler extract (DAE) on articular cartilage. METHODS DAE was prepared as previously described. All rats were randomly divided into Blank group and DAE group (10 rats per group) after 7-day adaptive feeding. The rats in DAE group were orally administrated with DAE at a dose of 0.2 g/kg per day for 3 weeks, and the rats in Blank group were fed with drinking water. Total RNA was isolated from the articular cartilage of knee joints. RNA sequencing (RNA-seq) experiment combined with quantitative real-time polymerase chain reaction (qRT-PCR) verification assay was carried out to explore the molecular control and therapeutic targets of DAE on articular cartilage. RESULTS We demonstrated that DAE significantly increased the expression levels of functional genes involved in cartilage formation, growth, and repair and decreased the expression levels of susceptibility genes involved in the pathophysiology of osteoarthritis. CONCLUSIONS DAE might serve as a candidate supplement for maintaining cartilage homeostasis and preventing cartilage degeneration and inflammation. These effects were possibly achieved by accelerating the expression of functional genes involved in chondrocyte commitment, survival, proliferation, and differentiation and suppressing the expression of susceptibility genes involved in the pathophysiology of osteoarthritis. Thus, our findings will contribute towards deepening the knowledge about the molecular control and therapeutic targets of DAE on the treatment of cartilage-related diseases.
Collapse
Affiliation(s)
- Baojin Yao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117 China
| | - Zhenwei Zhou
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117 China
| | - Mei Zhang
- Innovation Practice Center, Changchun University of Chinese Medicine, Changchun, 130117 China
| | - Xiangyang Leng
- The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130117 China
| | - Daqing Zhao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117 China
| |
Collapse
|
23
|
Yang J, Dong Z, Ren A, Fu G, Zhang K, Li C, Wang X, Cui H. Antibiotic tigecycline inhibits cell proliferation, migration and invasion via down-regulating CCNE2 in pancreatic ductal adenocarcinoma. J Cell Mol Med 2020; 24:4245-4260. [PMID: 32141702 PMCID: PMC7171345 DOI: 10.1111/jcmm.15086] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 10/17/2019] [Accepted: 01/15/2020] [Indexed: 12/17/2022] Open
Abstract
Recently, many researches have reported that antibiotic tigecycline has significant effect on cancer treatment. However, biomedical functions and molecular mechanisms of tigecycline in human pancreatic ductal adenocarcinoma (PDAC) remain unclear. In the current study, we tried to assess the effect of tigecycline in PDAC cells. AsPC‐1 and HPAC cells were treated with indicated concentrations of tigecycline for indicated time, and then, MTT, BrdU and soft agar assay were used to test cell proliferation. The effect of tigecycline on cell cycle and cellular apoptosis was tested by cytometry. Migration and invasion were detected by wound healing assay and transwell migration/invasion assay. Expressions of cell cycle‐related and migration/invasion‐related protein were determined by using Western blot. The results revealed that tigecycline observably suppressed cell proliferation by inducing cell cycle arrest at G0/G1 phase and blocked cell migration/invasion via holding back the epithelial‐mesenchymal transition (EMT) process in PDAC. In addition, tigecycline also remarkably blocked tumorigenecity in vivo. Furthermore, the effects of tigecycline alone or combined with gemcitabine in vitro or on PDAC xenografts were also performed. The results showed that tigecycline enhanced the chemosensitivity of PDAC cells to gemcitabine. Interestingly, we found CCNE2 expression was declined distinctly after tigecycline treatment. Then, CCNE2 was overexpressed to rescue tigecycline‐induced effect. The results showed that CCNE2 overexpression significantly rescued tigecycline‐inhibited cell proliferation and migration/invasion. Collectively, we showed that tigecycline inhibits cell proliferation, migration and invasion via down‐regulating CCNE2, and tigecycline might be used as a potential drug for PDAC treatment alone or combined with gemcitabine.
Collapse
Affiliation(s)
- Jie Yang
- State Key Laboratory of Silkworm Genome Biology, Institute of Sericulture and Systems Biology, Southwest University, Beibei, Chongqing, China.,Cancer Center, Medical Research Institute, Southwest University, Beibei, Chongqing, China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Beibei, Chongqing, China.,Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Beibei, Chongqing, China
| | - Zhen Dong
- State Key Laboratory of Silkworm Genome Biology, Institute of Sericulture and Systems Biology, Southwest University, Beibei, Chongqing, China.,Cancer Center, Medical Research Institute, Southwest University, Beibei, Chongqing, China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Beibei, Chongqing, China.,Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Beibei, Chongqing, China
| | - Aishu Ren
- College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Gang Fu
- College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Kui Zhang
- State Key Laboratory of Silkworm Genome Biology, Institute of Sericulture and Systems Biology, Southwest University, Beibei, Chongqing, China.,Cancer Center, Medical Research Institute, Southwest University, Beibei, Chongqing, China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Beibei, Chongqing, China.,Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Beibei, Chongqing, China
| | - Changhong Li
- State Key Laboratory of Silkworm Genome Biology, Institute of Sericulture and Systems Biology, Southwest University, Beibei, Chongqing, China.,Cancer Center, Medical Research Institute, Southwest University, Beibei, Chongqing, China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Beibei, Chongqing, China.,Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Beibei, Chongqing, China
| | - Xiangwei Wang
- Department of Urology, Carson International Cancer Center, Shenzhen University General Hospital & Shenzhen University Clinical Medical Academy Center, Shenzhen University, Shenzhen, China
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Institute of Sericulture and Systems Biology, Southwest University, Beibei, Chongqing, China.,Cancer Center, Medical Research Institute, Southwest University, Beibei, Chongqing, China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Beibei, Chongqing, China.,Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Beibei, Chongqing, China
| |
Collapse
|