1
|
Li Z, Han Q, Shao Y, Huang SB, Wang R, Rong XZ, Wang S, Liu Y. RPUSD1 enhances the expression of eIF4E through RluA catalytic domain, activates PI3K/AKT signaling pathway, and promotes the cell proliferation and invasion in non-small cell lung cancer. Int J Biol Macromol 2025; 306:141410. [PMID: 40010477 DOI: 10.1016/j.ijbiomac.2025.141410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 02/28/2025]
Abstract
RNA pseudouridylate synthase domain containing 1 (RPUSD1) is a pseudouridine synthase, and its role in human solid tumors remains unknown. We found that RPUSD1 showed enhanced cytoplasmic expression in non-small cell lung cancer (NSCLC) using immunohistochemistry and western blotting. Its increased expression is associated with tumor malignant phenotypes. The RluA catalytic domain of RPUSD1 activated the phosphoinositide 3-kinase (PI3K)/ Protein Kinase B (AKT) pathway, which promoted cell proliferation, migration, and invasion. Following transfection with full-length RPUSD1, the eukaryotic translation initiation factor 4E (eIF4E) mRNA enrichment increased. Moreover, full-length RPUSD1 enhanced eIF4E and Nijmegen breakage syndrome protein 1 (NBS1) proteins, whereas RPUSD1-ΔRluA did not significantly enhance eIF4E and NBS1 proteins. Moreover, NBS1 overexpression increased binding between NBS1 and PI3K-p110, whereas PI3K-p110/PI3K-p85 binding was diminished. RPUSD1 is an oncogene in NSCLC. RPUSD1 binds to eIF4E mRNA and stabilizes eIF4E mRNA through its RluA catalytic domain. EIF4E increases NBS1 expression, promoting its binding to PI3K p110. It competitively inhibits the interaction of PI3K p110 with PI3K p85, which leads to the dissociation of PI3K p85 from PI3K p110. This dissociation increases PI3K activity and activates downstream AKT. Thus, it promotes the ability of cell proliferation, migration, and invasion in NSCLC.
Collapse
Affiliation(s)
- Zhen Li
- Department of Pathology, the First Hospital of China Medical University, and College of Basic Medical Sciences of China Medical University, Shenyang 110122, PR China
| | - Qiang Han
- Department of Pathology, the First Hospital of China Medical University, and College of Basic Medical Sciences of China Medical University, Shenyang 110122, PR China
| | - Yang Shao
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Liaoning Clinical Research Center for Laboratory Medicine, Shenyang 110004, PR China
| | - Shao-Bing Huang
- Department of Pathology, the First Hospital of China Medical University, and College of Basic Medical Sciences of China Medical University, Shenyang 110122, PR China
| | - Rui Wang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang 110004, PR China
| | - Xue-Zhu Rong
- Department of Pathology, the First Hospital of China Medical University, Shenyang 110001, PR China.
| | - Si Wang
- Department of Medical Microbiology and Human Parasitology, College of Basic Medical Sciences, China Medical University, Shenyang 110122, PR China.
| | - Yang Liu
- Department of Pathology, the First Hospital of China Medical University, and College of Basic Medical Sciences of China Medical University, Shenyang 110122, PR China.
| |
Collapse
|
2
|
Liang L, Cai T, Li X, An J, Yu S, Zhang Y, Guo F, Wei F, He J, Xie K, Jiang T. Down-regulation of microRNA-23a promotes pancreatic ductal adenocarcinoma initiation and progression by up-regulation of FOXM1 expression. Genes Dis 2024; 11:101203. [PMID: 39022126 PMCID: PMC11252794 DOI: 10.1016/j.gendis.2023.101203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 10/19/2023] [Accepted: 11/19/2023] [Indexed: 07/20/2024] Open
Abstract
Transcriptional factor Forkhead box M1 (FOXM1) plays an important role in pancreatic ductal adenocarcinoma (PDAC) development and progression. The molecular mechanisms underlying its dysregulation remain unclear. We identified and functionally validated the microRNAs (miRNAs) that critically regulate FOXM1 expression in PDAC. The expression levels of miRNA-23a (miR-23a-3p and -5p) were altered in PDAC cell lines and their effects on FOXM1 signaling and cell proliferation and migration and tumorigenesis were examined in vitro and in vivo using mouse PDAC models. Compared with non-tumor pancreatic tissues, PDAC tissues and cell lines exhibited significantly reduced levels of miR-23a expression. Reduced miR-23a expression and concomitant increase in FOXM1 expression were also observed in acinar-to-ductal metaplasia and pancreatic intraepithelial neoplasia, the major premalignant lesions of PDAC. Transgenic expression of miR-23a reduced the expression of FOXM1 and suppressed cell proliferation and migration in PDAC cells, whereas the inhibitors of miR-23a did the opposite. Loss or reduced levels of miR-23a increased the levels of FOXM1 expression, while increased expression of FOXM1 down-regulated miR-23a expression, suggesting that miR-23a and FOXM1 were mutual negative regulators of their expression in PDAC cells. Therefore, the miR-23a/FOXM1 signaling axis is important in PDAC initiation and progression and could serve as an interventional or therapeutic target for patients with early or late stages of PDAC.
Collapse
Affiliation(s)
- Lixin Liang
- Center for Pancreatic Cancer Research, South China University of Technology School of Medicine, Guangzhou, Guangdong 510006, China
| | - Tian Cai
- Department of Laboratory Medicine, The Sixth Affiliated Hospital and Nanhai People's Hospital, South China University of Technology School of Medicine, Foshan, Guangdong 528200, China
| | - Xiaojia Li
- Center for Pancreatic Cancer Research, South China University of Technology School of Medicine, Guangzhou, Guangdong 510006, China
| | - Jianhong An
- Center for Pancreatic Cancer Research, South China University of Technology School of Medicine, Guangzhou, Guangdong 510006, China
| | - Sen Yu
- Center for Pancreatic Cancer Research, South China University of Technology School of Medicine, Guangzhou, Guangdong 510006, China
| | - Yang Zhang
- Center for Pancreatic Cancer Research, South China University of Technology School of Medicine, Guangzhou, Guangdong 510006, China
| | - Fengjie Guo
- Center for Pancreatic Cancer Research, South China University of Technology School of Medicine, Guangzhou, Guangdong 510006, China
| | - Fang Wei
- The Second Affiliated Hospital and Guangzhou First People's Hospital, South China University of Technology School of Medicine, Guangzhou, Guangdong 510006, China
| | - Jie He
- The Second Affiliated Hospital and Guangzhou First People's Hospital, South China University of Technology School of Medicine, Guangzhou, Guangdong 510006, China
| | - Keping Xie
- Center for Pancreatic Cancer Research, South China University of Technology School of Medicine, Guangzhou, Guangdong 510006, China
- The Second Affiliated Hospital and Guangzhou First People's Hospital, South China University of Technology School of Medicine, Guangzhou, Guangdong 510006, China
| | - Tingting Jiang
- Center for Pancreatic Cancer Research, South China University of Technology School of Medicine, Guangzhou, Guangdong 510006, China
| |
Collapse
|
3
|
Xu H, Luo Y, Li Q, Zhu H. Acupuncture influences multiple diseases by regulating gut microbiota. Front Cell Infect Microbiol 2024; 14:1371543. [PMID: 39040602 PMCID: PMC11260648 DOI: 10.3389/fcimb.2024.1371543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 06/13/2024] [Indexed: 07/24/2024] Open
Abstract
Acupuncture, an important green and side effect-free therapy in traditional Chinese medicine, is widely use both domestically and internationally. Acupuncture can interact with the gut microbiota and influence various diseases, including metabolic diseases, gastrointestinal diseases, mental disorders, nervous system diseases, and other diseases. This review presents a thorough analysis of these interactions and their impacts and examines the alterations in the gut microbiota and the potential clinical outcomes following acupuncture intervention to establish a basis for the future utilization of acupuncture in clinical treatments.
Collapse
Affiliation(s)
- Huimin Xu
- Department of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yingzhe Luo
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qiaoqi Li
- Department of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hong Zhu
- Department of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
4
|
Zhang X, Zhang F, Li Y, Fan N, Zhao K, Zhang A, Kang J, Lin Y, Xue X, Jiang X. Blockade of PI3K/AKT signaling pathway by Astragaloside IV attenuates ulcerative colitis via improving the intestinal epithelial barrier. J Transl Med 2024; 22:406. [PMID: 38689349 PMCID: PMC11061986 DOI: 10.1186/s12967-024-05168-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 04/05/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND The specific pathogenesis of UC is still unclear, but it has been clear that defects in intestinal barrier function play an important role in it. There is a temporary lack of specific drugs for clinical treatment. Astragaloside IV (AS-IV) is one of the main active ingredients extracted from Astragalus root and is a common Chinese herbal medicine for the treatment of gastrointestinal diseases. This study aimed to determine whether AS-IV has therapeutic value for DSS or LPS-induced intestinal epithelial barrier dysfunction in vivo and in vitro and its potential molecular mechanisms. METHODS The intestinal tissues from UC patients and colitis mice were collected, intestinal inflammation was observed by colonoscopy, and mucosal barrier function was measured by immunofluorescence staining. PI3K/AKT signaling pathway activator YS-49 and inhibitor LY-29 were administered to colitic mice to uncover the effect of this pathway on gut mucosal barrier modulation. Then, network pharmacology was used to screen Astragaloside IV (AS-IV), a core active component of the traditional Chinese medicine Astragalus membranaceus. The potential of AS-IV for intestinal barrier function repairment and UC treatment through blockade of the PI3K/AKT pathway was further confirmed by histopathological staining, FITC-dextran, transmission electron microscopy, ELISA, immunofluorescence, qRT-PCR, and western blotting. Finally, 16 S rRNA sequencing was performed to uncover whether AS-IV can ameliorate UC by regulating gut microbiota homeostasis. RESULTS Mucosal barrier function was significantly damaged in UC patients and murine colitis, and the activated PI3K/AKT signaling pathway was extensively involved. Both in vivo and vitro showed that the AS-IV-treated group significantly relieved inflammation and improved intestinal epithelial permeability by inhibiting the activation of the PI3K/AKT signaling pathway. In addition, microbiome data found that gut microbiota participates in AS-IV-mediated intestinal barrier recovery as well. CONCLUSIONS Our study highlights that AS-IV exerts a protective effect on the integrality of the mucosal barrier in UC based on the PI3K/AKT pathway, and AS-IV may serve as a novel AKT inhibitor to provide a potential therapy for UC.
Collapse
Affiliation(s)
- Xinhui Zhang
- Department of Pediatrics, Tangdu Hospital, Air Force Medical University, Xinsi Road, Baqiao District, 710038, Xi'an, Shaanxi, China
- The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, 620 West Chang'an Avenue, Chang 'a District, 710119, Xi'an, Shaanxi, China
| | - Fan Zhang
- Medical College, Yan'an University, 580 ShengDi Road, Baota District, 716099, Yan'an, Shaanxi, China
| | - Yan Li
- The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, 620 West Chang'an Avenue, Chang 'a District, 710119, Xi'an, Shaanxi, China
| | - Na Fan
- Medical College, Yan'an University, 580 ShengDi Road, Baota District, 716099, Yan'an, Shaanxi, China
| | - Ke Zhao
- The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, 620 West Chang'an Avenue, Chang 'a District, 710119, Xi'an, Shaanxi, China
- Department of Nutrition and Health, China Agriculture University, 100091, Beijing, China
| | - Anding Zhang
- Department of Pediatrics, Tangdu Hospital, Air Force Medical University, Xinsi Road, Baqiao District, 710038, Xi'an, Shaanxi, China
| | - Jiefang Kang
- The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, 620 West Chang'an Avenue, Chang 'a District, 710119, Xi'an, Shaanxi, China
| | - Yan Lin
- Department of Pediatrics, Tangdu Hospital, Air Force Medical University, Xinsi Road, Baqiao District, 710038, Xi'an, Shaanxi, China
| | - Xiaochang Xue
- The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, 620 West Chang'an Avenue, Chang 'a District, 710119, Xi'an, Shaanxi, China.
| | - Xun Jiang
- Department of Pediatrics, Tangdu Hospital, Air Force Medical University, Xinsi Road, Baqiao District, 710038, Xi'an, Shaanxi, China.
| |
Collapse
|
5
|
Lee YJ, Kim WR, Park EG, Lee DH, Kim JM, Shin HJ, Jeong HS, Roh HY, Kim HS. Exploring the Key Signaling Pathways and ncRNAs in Colorectal Cancer. Int J Mol Sci 2024; 25:4548. [PMID: 38674135 PMCID: PMC11050203 DOI: 10.3390/ijms25084548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/19/2024] [Accepted: 04/19/2024] [Indexed: 04/28/2024] Open
Abstract
Colorectal cancer (CRC) is the third most prevalent cancer to be diagnosed, and it has a substantial mortality rate. Despite numerous studies being conducted on CRC, it remains a significant health concern. The disease-free survival rates notably decrease as CRC progresses, emphasizing the urgency for effective diagnostic and therapeutic approaches. CRC development is caused by environmental factors, which mostly lead to the disruption of signaling pathways. Among these pathways, the Wingless/Integrated (Wnt) signaling pathway, Phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) signaling pathway, Mitogen-Activated Protein Kinase (MAPK) signaling pathway, Transforming Growth Factor-β (TGF-β) signaling pathway, and p53 signaling pathway are considered to be important. These signaling pathways are also regulated by non-coding RNAs (ncRNAs), including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs). They have emerged as crucial regulators of gene expression in CRC by changing their expression levels. The altered expression patterns of these ncRNAs have been implicated in CRC progression and development, suggesting their potential as diagnostic and therapeutic targets. This review provides an overview of the five key signaling pathways and regulation of ncRNAs involved in CRC pathogenesis that are studied to identify promising avenues for diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Yun Ju Lee
- Department of Integrated Biological Sciences, Pusan National University, Busan 46241, Republic of Korea; (Y.J.L.); (W.R.K.); (E.G.P.); (D.H.L.); (J.-m.K.); (H.J.S.); (H.-s.J.)
- Institute of Systems Biology, Pusan National University, Busan 46241, Republic of Korea;
| | - Woo Ryung Kim
- Department of Integrated Biological Sciences, Pusan National University, Busan 46241, Republic of Korea; (Y.J.L.); (W.R.K.); (E.G.P.); (D.H.L.); (J.-m.K.); (H.J.S.); (H.-s.J.)
- Institute of Systems Biology, Pusan National University, Busan 46241, Republic of Korea;
| | - Eun Gyung Park
- Department of Integrated Biological Sciences, Pusan National University, Busan 46241, Republic of Korea; (Y.J.L.); (W.R.K.); (E.G.P.); (D.H.L.); (J.-m.K.); (H.J.S.); (H.-s.J.)
- Institute of Systems Biology, Pusan National University, Busan 46241, Republic of Korea;
| | - Du Hyeong Lee
- Department of Integrated Biological Sciences, Pusan National University, Busan 46241, Republic of Korea; (Y.J.L.); (W.R.K.); (E.G.P.); (D.H.L.); (J.-m.K.); (H.J.S.); (H.-s.J.)
- Institute of Systems Biology, Pusan National University, Busan 46241, Republic of Korea;
| | - Jung-min Kim
- Department of Integrated Biological Sciences, Pusan National University, Busan 46241, Republic of Korea; (Y.J.L.); (W.R.K.); (E.G.P.); (D.H.L.); (J.-m.K.); (H.J.S.); (H.-s.J.)
- Institute of Systems Biology, Pusan National University, Busan 46241, Republic of Korea;
| | - Hae Jin Shin
- Department of Integrated Biological Sciences, Pusan National University, Busan 46241, Republic of Korea; (Y.J.L.); (W.R.K.); (E.G.P.); (D.H.L.); (J.-m.K.); (H.J.S.); (H.-s.J.)
- Institute of Systems Biology, Pusan National University, Busan 46241, Republic of Korea;
| | - Hyeon-su Jeong
- Department of Integrated Biological Sciences, Pusan National University, Busan 46241, Republic of Korea; (Y.J.L.); (W.R.K.); (E.G.P.); (D.H.L.); (J.-m.K.); (H.J.S.); (H.-s.J.)
- Institute of Systems Biology, Pusan National University, Busan 46241, Republic of Korea;
| | - Hyun-Young Roh
- Institute of Systems Biology, Pusan National University, Busan 46241, Republic of Korea;
- Department of Biological Sciences, College of Natural Sciences, Pusan National University, Busan 46241, Republic of Korea
| | - Heui-Soo Kim
- Institute of Systems Biology, Pusan National University, Busan 46241, Republic of Korea;
- Department of Biological Sciences, College of Natural Sciences, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
6
|
Su H, Peng C, Liu Y. Regulation of ferroptosis by PI3K/Akt signaling pathway: a promising therapeutic axis in cancer. Front Cell Dev Biol 2024; 12:1372330. [PMID: 38562143 PMCID: PMC10982379 DOI: 10.3389/fcell.2024.1372330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
The global challenge posed by cancer, marked by rising incidence and mortality rates, underscores the urgency for innovative therapeutic approaches. The PI3K/Akt signaling pathway, frequently amplified in various cancers, is central in regulating essential cellular processes. Its dysregulation, often stemming from genetic mutations, significantly contributes to cancer initiation, progression, and resistance to therapy. Concurrently, ferroptosis, a recently discovered form of regulated cell death characterized by iron-dependent processes and lipid reactive oxygen species buildup, holds implications for diseases, including cancer. Exploring the interplay between the dysregulated PI3K/Akt pathway and ferroptosis unveils potential insights into the molecular mechanisms driving or inhibiting ferroptotic processes in cancer cells. Evidence suggests that inhibiting the PI3K/Akt pathway may sensitize cancer cells to ferroptosis induction, offering a promising strategy to overcome drug resistance. This review aims to provide a comprehensive exploration of this interplay, shedding light on the potential for disrupting the PI3K/Akt pathway to enhance ferroptosis as an alternative route for inducing cell death and improving cancer treatment outcomes.
Collapse
Affiliation(s)
- Hua Su
- Xingyi People’s Hospital, Xinyi, China
| | - Chao Peng
- Xingyi People’s Hospital, Xinyi, China
| | - Yang Liu
- The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
7
|
Raghuwanshi S, Gartel AL. Small-molecule inhibitors targeting FOXM1: Current challenges and future perspectives in cancer treatments. Biochim Biophys Acta Rev Cancer 2023; 1878:189015. [PMID: 37913940 DOI: 10.1016/j.bbcan.2023.189015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 11/03/2023]
Abstract
Forkhead box (FOX) protein M1 (FOXM1) is a critical proliferation-associated transcription factor (TF) that is aberrantly overexpressed in the majority of human cancers and has also been implicated in poor prognosis. A comprehensive understanding of various aspects of this molecule has revealed its role in, cell proliferation, cell migration, invasion, angiogenesis and metastasis. The FOXM1 as a TF directly or indirectly regulates the expression of several target genes whose dysregulation is associated with almost all hallmarks of cancer. Moreover, FOXM1 expression is associated with chemoresistance to different anti-cancer drugs. Several studies have confirmed that suppression of FOXM1 enhanced the drug sensitivity of various types of cancer cells. Current data suggest that small molecule inhibitors targeting FOXM1 in combination with anticancer drugs may represent a novel therapeutic strategy for chemo-resistant cancers. In this review, we discuss the clinical utility of FOXM1, further, we summarize and discuss small-molecule inhibitors targeting FOXM1 and categorize them according to their mechanisms of targeting FOXM1. Despite great progress, small-molecule inhibitors targeting FOXM1 face many challenges, and we present here all small-molecule FOXM1 inhibitors in different stages of development. We discuss the current challenges and provide insights on the future application of FOXM1 inhibition to the clinic.
Collapse
Affiliation(s)
- Sanjeev Raghuwanshi
- University of Illinois at Chicago, Department of Medicine, Chicago, IL 60612, USA
| | - Andrei L Gartel
- University of Illinois at Chicago, Department of Medicine, Chicago, IL 60612, USA.
| |
Collapse
|
8
|
Elgazar A, El-Domany RA, Eldehna WM, Badria FA. 3-Acetyl-11-keto-β-boswellic Acid-Based Hybrids Alleviate Acetaminophen-Induced Hepatotoxicity in HepG2 by the Regulation of Inflammatory and Oxidative Stress Pathways: An Integrated Approach. ACS OMEGA 2023; 8:39490-39510. [PMID: 37901542 PMCID: PMC10601058 DOI: 10.1021/acsomega.3c05247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/28/2023] [Indexed: 10/31/2023]
Abstract
In an effort to develop new compounds for managing drug-induced liver injury, we prepared 23 novel hybrids based on 3-acetyl-11-keto-β-boswellic acid (AKBA) using various biocompatible linkers. A bioguided approach was employed to identify the most promising hybrid. Eight compounds exhibited superior anti-inflammatory activity compared to the parent compound. Two of these hybrids (5b and 18) were able to reduce gene expression of TNF-α in LPS-induced inflammation in RAW 264.7 cells, similar to dexamethasone. Subsequently, the hepatoprotective potential of these hybrids was evaluated against acetaminophen (APAP) toxicity in HepG2 cells at doses of 1 and 10 μM. Both hybrids effectively restored cytokine levels, which had been elevated by APAP, to normal levels. Furthermore, they normalized depleted superoxide dismutase and reduced glutathione levels while significantly reducing malondialdehyde (MDA) levels. Network pharmacology analysis suggested that AKBA-based hybrids exert their action by regulating PI3K and EGFR pathways, activating anti-inflammatory mechanisms, and initiating tissue repair and regeneration. Molecular docking studies provided insights into the interaction of the hybrids with PI3K. Additionally, the hybrids demonstrated good stability at different pH levels, following first-order kinetics, with relatively long half-lives, suggesting potential for absorption into circulation without significant degradation.
Collapse
Affiliation(s)
- Abdullah
A. Elgazar
- Department
of Pharmacognosy, Faculty of Pharmacy, Kafrelsheikh
University, Kafrelsheikh 33516, Egypt
| | - Ramadan A. El-Domany
- Department
of Microbiology and Immunology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Wagdy M. Eldehna
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Farid A. Badria
- Department
of Pharmacognosy, Faculty of Pharmacy, Mansoura
University, Mansoura 35516, Egypt
| |
Collapse
|
9
|
Guo M, Wang X. Pathological mechanism and targeted drugs of ulcerative colitis: A review. Medicine (Baltimore) 2023; 102:e35020. [PMID: 37713856 PMCID: PMC10508406 DOI: 10.1097/md.0000000000035020] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/09/2023] [Indexed: 09/17/2023] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory disease of the colon with abdominal pain, diarrhea, and mucopurulent stools as the main symptoms. Its incidence is increasing worldwide, and traditional treatments have problems such as immunosuppression and metabolic disorders. In this article, the etiology and pathogenesis of ulcerative colitis are reviewed to clarify the targeted drugs of UC in the latest research. Our aim is to provide more ideas for the clinical treatment and new drug development of UC, mainly by analyzing and sorting out the relevant literature on PubMed, summarizing and finding that it is related to the main genetic, environmental, immune and other factors, and explaining its pathogenesis from the NF-κB pathway, PI3K/Akt signaling pathway, and JAK/STAT signaling pathway, and obtaining anti-TNF-α monoclonal antibodies, integrin antagonists, IL-12/IL-23 antagonists, novel UC-targeted drugs such as JAK inhibitors and SIP receptor agonists. We believe that rational selection of targeted drugs and formulation of the best dosing strategy under the comprehensive consideration of clinical evaluation is the best way to treat UC.
Collapse
Affiliation(s)
- Meitong Guo
- Changchun University of Chinese Medicine, Changchun City, China
| | - Xiaoyan Wang
- Jilin Academy of Chinese Medicine, Chaoyang District, China
| |
Collapse
|
10
|
Tsoi H, Tsang WC, Man EPS, Leung MH, You CP, Chan SY, Chan WL, Khoo US. Checkpoint Kinase 2 Inhibition Can Reverse Tamoxifen Resistance in ER-Positive Breast Cancer. Int J Mol Sci 2022; 23:ijms232012290. [PMID: 36293165 PMCID: PMC9604393 DOI: 10.3390/ijms232012290] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/11/2022] [Accepted: 10/13/2022] [Indexed: 11/06/2022] Open
Abstract
Breast cancer is a heterogeneous disease. Tamoxifen is frequently used to treat ER-positive breast cancer. Our team has identified a novel splice variant of NCOR2, BQ323636.1 (BQ), that mediates tamoxifen resistance. However, the upstream factors that modulate BQ expression are not apparent. This study reveals that tamoxifen treatment causes induction of DNA damage which can enhance BQ expression. We show that DNA damage can activate the ATM/CHK2 and ATR/CHK1 signalling cascades and confirm that ATM/CHK2 signalling is responsible for enhancing the protein stability of BQ. siRNA or a small inhibitor targeting CHK2 resulted in the reduction in BQ expression through reduced phosphorylation and enhanced poly-ubiquitination of BQ. Inhibition of CHK2 by CCT241533 could reverse tamoxifen resistance in vitro and in vivo. Using clinical samples in the tissue microarray, we confirmed that high p-CHK2 expression was significantly associated with high nuclear BQ expression, tamoxifen resistance and poorer overall and disease-specific survival. In conclusion, tamoxifen treatment can enhance BQ expression in ER-positive breast cancer by activating the ATM/CHK2 axis. Targeting CHK2 is a promising approach to overcoming tamoxifen resistance in ER-positive breast cancer.
Collapse
Affiliation(s)
- Ho Tsoi
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Wai-Chung Tsang
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Ellen P. S. Man
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Man-Hong Leung
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Chan-Ping You
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Sum-Yin Chan
- Department of Clinical Oncology, Queen Mary Hospital, Hong Kong SAR, China
| | - Wing-Lok Chan
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Ui-Soon Khoo
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Correspondence: ; Tel.: +852-2255-2664; Fax: +852-2218-5205
| |
Collapse
|
11
|
Zhu CL, Xie J, Zhao ZZ, Li P, Liu Q, Guo Y, Meng Y, Wan XJ, Bian JJ, Deng XM, Wang JF. PD-L1 maintains neutrophil extracellular traps release by inhibiting neutrophil autophagy in endotoxin-induced lung injury. Front Immunol 2022; 13:949217. [PMID: 36016930 PMCID: PMC9396256 DOI: 10.3389/fimmu.2022.949217] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
Programmed death ligand 1 (PD-L1) is not only an important molecule in mediating tumor immune escape, but also regulates inflammation development. Here we showed that PD-L1 was upregulated on neutrophils in lipopolysaccharide (LPS)-induced acute respiratory distress syndrome (ARDS). Neutrophil specific knockout of PD-L1 reduced lung injury in ARDS model induced by intratracheal LPS injection. The level of NET release was reduced and autophagy is elevated by PD-L1 knockout in ARDS neutrophils both in vivo and in vitro. Inhibition of autophagy could reverse the inhibitory effect of PD-L1 knockout on NET release. PD-L1 interacted with p85 subunit of PI3K at the endoplasmic reticulum (ER) in neutrophils from ARDS patients, activating the PI3K/Akt/mTOR pathway. An extrinsic neutralizing antibody against PD-L1 showed a protective effect against ARDS. Together, PD-L1 maintains the release of NETs by regulating autophagy through the PI3K/Akt/mTOR pathway in ARDS. Anti-PD-L1 therapy may be a promising measure in treating ARDS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jin-jun Bian
- *Correspondence: Jin-jun Bian, ; Xiao-ming Deng, ; Jia-feng Wang,
| | - Xiao-ming Deng
- *Correspondence: Jin-jun Bian, ; Xiao-ming Deng, ; Jia-feng Wang,
| | - Jia-feng Wang
- *Correspondence: Jin-jun Bian, ; Xiao-ming Deng, ; Jia-feng Wang,
| |
Collapse
|
12
|
Guo P, Li R, Piao TH, Wang CL, Wu XL, Cai HY. Pathological Mechanism and Targeted Drugs of COPD. Int J Chron Obstruct Pulmon Dis 2022; 17:1565-1575. [PMID: 35855746 PMCID: PMC9288175 DOI: 10.2147/copd.s366126] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 07/04/2022] [Indexed: 01/17/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) includes chronic bronchitis, emphysema, and small airway obstruction. Incompletely reversible airflow limitation, inflammation, excessive mucus secretion and bronchial mucosal epithelial lesions are the main pathological basis of the disease. The prevalence of COPD is increasingly worldwide, which has caused the burden on individuals and society. This paper summarizes the pathogenesis of COPD and clarifies the effect and mechanism of the latest targeted drugs for COPD. Besides, we focus on NOD-like receptor thermal protein domain associated protein 3 inflammasome (NLRP3 inflammasome). NLRP3 can promote production of interleukin-1β (IL-1β) and interleukin-18 (IL-18). NLRP3 is an important factor in the migratory aggregation of macrophages and neutrophils and the generation of oxidative stress. Inhibition of NLRP3 inflammasome indirectly blocks the inflammatory effects of IL-1β and IL-18, which may be regarded as an ideal target for COPD treatment.
Collapse
Affiliation(s)
- Peng Guo
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Changchun, 130000, People's Republic of China
| | - Rui Li
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100000, People's Republic of China
| | - Tie Hua Piao
- Pulmonology Department, The First Clinical Hospital of Jilin Academy of Traditional Chinese Medicine, Changchun, 130000, People's Republic of China
| | - Chun Lan Wang
- Pulmonology Department, The First Clinical Hospital of Jilin Academy of Traditional Chinese Medicine, Changchun, 130000, People's Republic of China
| | - Xiao Lu Wu
- Pulmonology Department, The First Clinical Hospital of Jilin Academy of Traditional Chinese Medicine, Changchun, 130000, People's Republic of China
| | - Hong Yan Cai
- Pulmonology Department, The First Clinical Hospital of Jilin Academy of Traditional Chinese Medicine, Changchun, 130000, People's Republic of China
| |
Collapse
|
13
|
Huang X, Rao W, Wang C, Lu J, Li Z, Kong W, Feng Y, Xu T, Apaer R, Gao F. Kaposi's sarcoma-associated herpes virus-derived microRNA K12-1 over-activates the PI3K/Akt pathway to facilitate cancer progression in HIV-related gastrointestinal Kaposi's sarcoma. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2022; 27:258-265. [PMID: 35413475 DOI: 10.1016/j.slasd.2022.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/22/2022] [Accepted: 04/05/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Kaposi's sarcoma-associated herpes virus (KSHV) initiate and accelerate the development of Kaposi's sarcoma (KS), and KSHV possesses many cancer-associated genes, including KSHV-derived microRNA miR-K12-1, which has been identified to be closely associated with KS progression. However, the detailed mechanisms by which miR-K12-1 facilitates HIV-related gastrointestinal KS development are still not fully delineated. OBJECTIVES This study strived to evaluate the effect of miR-K12-1 on the progression of HIV-related gastrointestinal KS. MATERIALS AND METHODS The expression levels of miR-K12-1 in HIV-related gastrointestinal KS tissues were determined by RT-qPCR. Proliferation and apoptosis were assessed by colony formation, CCK-8 and flow cytometry, respectively. The expression of all proteins was detected by Western blot. The in vivo effect of miR-K12-1 on the formation of a tumor was explored by using the mouse xenograft model. RESULTS In this study, we uncovered that KSHV-miR-K12-1 was upregulated in HIV-related gastrointestinal KS tissues and associated with poor outcome in HIV-related gastrointestinal KS patients. Compared with the control group, after miR-K12-1 inhibitor transfection, BCBL-1 cell viability was decreased, and the cell apoptosis was significantly increased, whereas transfection of miR-K12-1 mimics promoted cell proliferation and mitosis. In addition, our rescuing experiments verified that miR-K12-1 promoted cell proliferation via activating the PI3K/Akt pathway, and inhibition of the PI3K/Akt pathway by LY294002 abrogated the tumor-promoting effects of miR-K12-1 in HIV-related gastrointestinal KS. CONCLUSIONS In summary, we concluded that KSHV-derived miR-K12-1 activate the PI3K/Akt pathway to initiate and accelerate the development of KS, which convinces us that miR-K12-1 can be used as potential biomarkers for KS diagnosis, treatment and prognosis.
Collapse
Affiliation(s)
- Xiaoling Huang
- Department of Gastroenterology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China; Xinjiang Clinical Research Center of Digestive System Diseases, Urumqi, 830001, China.
| | - Wei Rao
- Department of Gastroenterology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China; Xinjiang Clinical Research Center of Digestive System Diseases, Urumqi, 830001, China.
| | - Chun Wang
- Department of Pathology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China.
| | - Jiajie Lu
- Department of Gastroenterology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China; Xinjiang Clinical Research Center of Digestive System Diseases, Urumqi, 830001, China.
| | - Ziqiong Li
- Department of Gastroenterology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China; Xinjiang Clinical Research Center of Digestive System Diseases, Urumqi, 830001, China.
| | - Wenjie Kong
- Department of Gastroenterology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China; Xinjiang Clinical Research Center of Digestive System Diseases, Urumqi, 830001, China.
| | - Yan Feng
- Department of Gastroenterology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China; Xinjiang Clinical Research Center of Digestive System Diseases, Urumqi, 830001, China.
| | - Tian Xu
- Department of Gastroenterology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China; Xinjiang Clinical Research Center of Digestive System Diseases, Urumqi, 830001, China.
| | - Rziya Apaer
- Department of Gastroenterology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China; Xinjiang Clinical Research Center of Digestive System Diseases, Urumqi, 830001, China.
| | - Feng Gao
- Department of Gastroenterology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China; Xinjiang Clinical Research Center of Digestive System Diseases, Urumqi, 830001, China.
| |
Collapse
|
14
|
He QD, Guo JJ, Zhang Q, Yau YM, Yu Y, Zhong ZH, Tong ZY, Yang ZB, Chen M. Effects of Electroacupuncture on the Gut Microbiome in Cisplatin-Induced Premature Ovarian Failure Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:9352833. [PMID: 35321505 PMCID: PMC8938064 DOI: 10.1155/2022/9352833] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 01/24/2022] [Accepted: 02/08/2022] [Indexed: 12/17/2022]
Abstract
Growing evidence showed that the gut microbiota was associated with premature ovarian failure (POF). Many clinical types of research had shown that electroacupuncture was effective in the treatment of POF. However, there was little research on regulating the gut microbiome of POF mice by electroacupuncture. Therefore, this study attempted to verify whether electroacupuncture could regulate the gut microbiome in POF mice. POF mice were established by being injected intraperitoneally with cisplatin (2 mg/kg) for 2 weeks. Guanyuan (CV4) and Sanyinjiao (SP6) were selected in the electroacupuncture-at-the-acupoints group (EA group). Nonacupoints around CV4 and SP6 were selected in the electroacupuncture-at-the-nonacupoints group (EN group). The EA group and EN group were treated for 3 weeks. The ovarian function was evaluated by histopathological and molecular assays. Meanwhile, the gut microbiome of all mice was detected by 16S rDNA sequencing. The results showed that EA could restore the estrous cycle and reduce the number of atresia follicles in POF mice. The levels of serum follicle-stimulating hormone and luteinizing hormone were decreased by EA. As well, the levels of serum estradiol, anti-Mullerian hormone, and β-glucuronidase were increased by EA. The relative expressions of PI3K, AKT, and mTOR were increased to promote the proliferation of ovarian cells in the EA group. According to the results of 16S rDNA sequencing, the abundance and diversity of the gut microbiome could be regulated by EA. The relative abundance of beneficial bacteria was increased by EA. The KEGG pathway analysis showed that the gut microbiome associated with the estrogen signaling pathway, oocyte maturation, and PI3K-AKT signaling pathway was regulated by EA.
Collapse
Affiliation(s)
- Qi-da He
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, China
| | - Jing-jing Guo
- Department of Traditional Chinese Medicine, Xiamen University, Xiamen 361000, China
| | - Qi Zhang
- Department of Traditional Chinese Medicine, Xiamen University, Xiamen 361000, China
| | - Yuen-ming Yau
- Department of Traditional Chinese Medicine, Xiamen University, Xiamen 361000, China
| | - Yue Yu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, China
| | - Zheng-hong Zhong
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, China
| | - Zi-yan Tong
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, China
| | - Zong-bao Yang
- Department of Traditional Chinese Medicine, Xiamen University, Xiamen 361000, China
| | - Min Chen
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, China
| |
Collapse
|
15
|
Zhang YL, Ma Y, Zeng YQ, Liu Y, He EP, Liu YT, Qiao FL, Yu R, Wang YS, Wu XY, Leng P. A narrative review of research progress on FoxM1 in breast cancer carcinogenesis and therapeutics. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1704. [PMID: 34988213 PMCID: PMC8667115 DOI: 10.21037/atm-21-5271] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 10/29/2021] [Indexed: 02/05/2023]
Abstract
OBJECTIVE The purpose of this review is to clarify the potential roles of forkhead box transcription factor M1 (FoxM1) in the occurrence and progression of breast cancer, as well as the predictive value of FoxM1 as a prognostic biomarker and potential therapeutic target for breast cancer. BACKGROUND Breast cancer, well-known as a molecularly heterogeneous cancer, is still one of the most frequently diagnosed malignant tumors among females worldwide. Tumor recurrence and metastasis are the central causes of high mortality in breast cancer patients. Many factors contribute to the occurrence and progression of breast cancer, including FoxM1. FoxM1, widely regarded as a classic proliferation-related transcription factor, plays pivotal roles in the occurrence, proliferation, invasion, migration, drug resistance, and epithelial-mesenchymal transition (EMT) processes of multiple human tumors including breast cancer. METHODS The PubMed database was searched for articles published in English from February 2008 to May 2021 using related keywords such as "forkhead box transcription factor M1", "human breast cancer", "FoxM1", and "human tumor". About 90 research papers and reports written in English were identified, most of which were published after 2015. These papers mainly concentrated on the functions of FoxM1 in the occurrence, development, drug resistance, and treatment of human breast cancer. CONCLUSIONS Considering that the abnormal expression of FoxM1 plays a significant role in the proliferation, invasion, metastasis, and chemotherapy drug resistance of breast cancer, and its overexpression is closely correlated with the unfavorable clinicopathological characteristics of breast tumor patients, it is considerably important to comprehend the regulatory mechanism of FoxM1 in breast cancer. This will provide strong evidence for FoxM1 as a potential biomarker for the targeted treatment and prognostic evaluation of breast cancer patients.
Collapse
Affiliation(s)
- Yan-Ling Zhang
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yan Ma
- Emergency Department of West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China.,Institute of Disaster Medicine, Sichuan University, Chengdu, China
| | - You-Qin Zeng
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yan Liu
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - En-Ping He
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chengdu Medical College-Nuclear Industry 416 Hospital, Chengdu, China
| | - Yi-Tong Liu
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Feng-Ling Qiao
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rong Yu
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ying-Shuang Wang
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xin-Yu Wu
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ping Leng
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
16
|
Liu C, Barger CJ, Karpf AR. FOXM1: A Multifunctional Oncoprotein and Emerging Therapeutic Target in Ovarian Cancer. Cancers (Basel) 2021; 13:3065. [PMID: 34205406 PMCID: PMC8235333 DOI: 10.3390/cancers13123065] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/11/2021] [Accepted: 06/16/2021] [Indexed: 02/08/2023] Open
Abstract
Forkhead box M1 (FOXM1) is a member of the conserved forkhead box (FOX) transcription factor family. Over the last two decades, FOXM1 has emerged as a multifunctional oncoprotein and a robust biomarker of poor prognosis in many human malignancies. In this review article, we address the current knowledge regarding the mechanisms of regulation and oncogenic functions of FOXM1, particularly in the context of ovarian cancer. FOXM1 and its associated oncogenic transcriptional signature are enriched in >85% of ovarian cancer cases and FOXM1 expression and activity can be enhanced by a plethora of genomic, transcriptional, post-transcriptional, and post-translational mechanisms. As a master transcriptional regulator, FOXM1 promotes critical oncogenic phenotypes in ovarian cancer, including: (1) cell proliferation, (2) invasion and metastasis, (3) chemotherapy resistance, (4) cancer stem cell (CSC) properties, (5) genomic instability, and (6) altered cellular metabolism. We additionally discuss the evidence for FOXM1 as a cancer biomarker, describe the rationale for FOXM1 as a cancer therapeutic target, and provide an overview of therapeutic strategies used to target FOXM1 for cancer treatment.
Collapse
Affiliation(s)
| | | | - Adam R. Karpf
- Eppley Institute and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68918-6805, USA; (C.L.); (C.J.B.)
| |
Collapse
|
17
|
Klinhom-On N, Seubwai W, Sawanyawisuth K, Lert-Itthiporn W, Waraasawapati S, Detarya M, Wongkham S. FOXM1c is the predominant FOXM1 isoform expressed in cholangiocarcinoma that associated with metastatic potential and poor prognosis of patients. Heliyon 2021; 7:e06846. [PMID: 33997388 PMCID: PMC8093466 DOI: 10.1016/j.heliyon.2021.e06846] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/16/2021] [Accepted: 04/14/2021] [Indexed: 12/22/2022] Open
Abstract
Forkhead box M1 (FOXM1) is a transcriptional factor which plays an important role in oncogenesis. Four FOXM1 isoforms, FOXM1a, FOXM1b, FOXM1c and FOXM1d, are known so far. Different FOXM1 isoforms influence progression of cancer in different cancer types. In this study, the FOXM1c isoform and its impact in cholangiocarcinoma (CCA) was identified. FOXM1c was found to be the predominant isoform in patient-CCA tissues and cell lines. Detection of FOXM1c expression in CCA tissues reflected the worse prognosis of the patients, namely the advanced stage and shorter survival. Suppression of FOXM1 expression using siRNA considerably reduced migration and invasion abilities of CCA cell lines. RNA sequencing analysis revealed claudin-1 as a target of FOXM1. FOXM1 exhibited a negative correlation with claudin-1 expression which was demonstrated in patient CCA tissues and cell lines. FOXM1 may be a potential target for therapeutic treatment of the metastatic CCA.
Collapse
Affiliation(s)
- Nathakan Klinhom-On
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, 40002, Thailand
| | - Wunchana Seubwai
- Department of Forensic Medicine, Faculty of Medicine, Khon Kaen University, 40002, Thailand.,Cholangiocarcinoma Research Institute, Khon Kaen University, 40002, Thailand
| | - Kanlayanee Sawanyawisuth
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, 40002, Thailand.,Cholangiocarcinoma Research Institute, Khon Kaen University, 40002, Thailand
| | - Worachart Lert-Itthiporn
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, 40002, Thailand.,Cholangiocarcinoma Research Institute, Khon Kaen University, 40002, Thailand
| | - Sakda Waraasawapati
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand.,Cholangiocarcinoma Research Institute, Khon Kaen University, 40002, Thailand
| | - Marutpong Detarya
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, 40002, Thailand
| | - Sopit Wongkham
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, 40002, Thailand.,Cholangiocarcinoma Research Institute, Khon Kaen University, 40002, Thailand
| |
Collapse
|