1
|
Ashok G, AlAsmari AF, AlAsmari F, Livingstone P, Anbarasu A, Ramaiah S. Transcriptomic, mutational and structural bioinformatics approaches to explore the therapeutic role of FAP in predominant cancer types. Discov Oncol 2024; 15:699. [PMID: 39579201 PMCID: PMC11585531 DOI: 10.1007/s12672-024-01531-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 11/05/2024] [Indexed: 11/25/2024] Open
Abstract
Fibroblast activating protein (FAP) is a cell surface marker of cancer-associated fibroblasts with a distinct pro-tumorigenic role. The present study analyzed the pan-cancer expression; and clinical and mutational profiles of the FAP coding gene. Molecular dynamics simulation (MDS) deciphered the backbone dynamics and energetics of FAP. Virtual screening and subsequent pharmacokinetic-profiling (PK) filtered lead molecules, which were subjected to molecular docking. MDS projected a stable trajectory for the protein, as dynamics evidenced by low residue-level fluctuations, stable backbone dynamics, and energetics. Around five stabilization and deleterious mutations in the catalytic domain were identified. The low binding energy (BE) profiles from molecular docking studies screened the top five lead molecules for site-specific intermolecular interaction studies. Lead-16 (ZINC000245289699) exhibited a significant BE and inhibition constant of -6.87 kcal/mol and 12.27 μM, respectively, across FAP and its mutants. Interestingly, the docked complexes of Lead-16 interacted with the catalytic triad residues (S624, D702, and H734). The docked complexes of Lead-16 with FAP showed lower average root-mean-square fluctuations compared to the unbound protein, suggesting a stable ligand-protein complex. The tumor-specific expression and its critical overall survival suggest the inhibitors of FAP for potential cancer therapeutic intervention and hindering tumor microenvironment-driven cancer progression.
Collapse
Affiliation(s)
- Gayathri Ashok
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India
- Department of Bio-Sciences, SBST, VIT, Vellore, 632014, Tamil Nadu, India
| | - Abdullah F AlAsmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, 11451, 13 Riyadh, Saudi Arabia
| | - Fawaz AlAsmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, 11451, 13 Riyadh, Saudi Arabia
| | - Paul Livingstone
- School of Sports and Health Sciences, Cardiff Metropolitan University, Cardiff, CF5 2YB, UK
| | - Anand Anbarasu
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India
- Department of Biotechnology, SBST, VIT, Vellore, 632014, Tamil Nadu, India
| | - Sudha Ramaiah
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India.
- Department of Bio-Sciences, SBST, VIT, Vellore, 632014, Tamil Nadu, India.
| |
Collapse
|
2
|
Xia RJ, Du XY, Shen LW, Ma JG, Xu SM, Fan RF, Qin JW, Yan L. Roles of the tumor microenvironment in the resistance to programmed cell death protein 1 inhibitors in patients with gastric cancer. World J Gastrointest Oncol 2024; 16:3820-3831. [PMID: 39350980 PMCID: PMC11438768 DOI: 10.4251/wjgo.v16.i9.3820] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/27/2024] [Accepted: 08/09/2024] [Indexed: 09/09/2024] Open
Abstract
Despite the continuous developments and advancements in the treatment of gastric cancer (GC), which is one of the most prevalent types of cancer in China, the overall survival is still poor for most patients with advanced GC. In recent years, with the progress in tumor immunology research, attention has shifted toward immunotherapy as a therapeutic approach for GC. Programmed cell death protein 1 (PD-1) inhibitors, as novel immunosuppressive medications, have been widely utilized in the treatment of GC. However, many patients are still resistant to PD-1 inhibitors and experience recurrence in the advanced stages of PD-1 immunotherapy. To reduce the occurrence of drug resistance and recurrence in GC patients receiving PD-1 immunotherapy, to maximize the clinical activity of immunosuppressive drugs, and to elicit a lasting immune response, it is essential to research the tumor microenvironment mechanisms leading to PD-1 inhibitor resistance in GC patients. This article reviews the progress in studying the factors influencing the resistance to PD-1 inhibitors in the GC tumor microenvironment, aiming to provide insights and a basis for reducing resistance to PD-1 inhibitors for GC patients in the future.
Collapse
Affiliation(s)
- Ren-Jie Xia
- Department of General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
- Department of Medicine, Northwest Minzu University, Lanzhou 730050, Gansu Province, China
| | - Xiao-Yu Du
- Department of General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
- Department of Medicine, Northwest Minzu University, Lanzhou 730050, Gansu Province, China
| | - Li-Wen Shen
- Department of Medical Support Center, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Jian-Guo Ma
- Department of General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Shu-Mei Xu
- Department of General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Rui-Fang Fan
- Department of General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Jian-Wei Qin
- Department of General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Long Yan
- Department of General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| |
Collapse
|
3
|
Li P, Zhang H, Chen T, Zhou Y, Yang J, Zhou J. Cancer-associated fibroblasts promote proliferation, angiogenesis, metastasis and immunosuppression in gastric cancer. Matrix Biol 2024; 132:59-71. [PMID: 38936680 DOI: 10.1016/j.matbio.2024.06.004] [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: 03/19/2024] [Revised: 05/21/2024] [Accepted: 06/18/2024] [Indexed: 06/29/2024]
Abstract
Despite advances in surgery, radiotherapy and immunotherapy, the mortality rate for gastric cancer remains one of the highest in the world. A large body of evidence has demonstrated that cancer-associated fibroblasts (CAFs), as core members of the stroma, can secrete cytokines, proteins and exosomes to create a tumour microenvironment that is conducive to cancer cell survival. CAFs can also interact with cancer cells to form a complex signalling network, enabling cancer cells to more easily metastasise to other organs and tissues in the body and develop metastatic foci. In this review, we provide an overview of the CAFs concept and activators. We focus on elucidating their effects on immune cells, intratumoural vasculature, extracellular matrix, as well as cancer cell activity, metastatic power and metabolism, and on enhancing the metastatic ability of cancer cells through activation of JAK/STAT, NF/κB and CXCL12/CXCR4. Various therapeutic agents targeting CAFs are also under development and are expected to improve the prognosis of gastric cancer in combination with existing treatment options.
Collapse
Affiliation(s)
- Peiyuan Li
- Department of general surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, PR China
| | - Huan Zhang
- Department of general surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, PR China
| | - Tao Chen
- Department of general surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, PR China
| | - Yajing Zhou
- Department of general surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, PR China
| | - Jiaoyang Yang
- Department of general surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, PR China
| | - Jin Zhou
- Department of general surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, PR China.
| |
Collapse
|
4
|
Ozmen E, Demir TD, Ozcan G. Cancer-associated fibroblasts: protagonists of the tumor microenvironment in gastric cancer. Front Mol Biosci 2024; 11:1340124. [PMID: 38562556 PMCID: PMC10982390 DOI: 10.3389/fmolb.2024.1340124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/31/2024] [Indexed: 04/04/2024] Open
Abstract
Enhanced knowledge of the interaction of cancer cells with their environment elucidated the critical role of tumor microenvironment in tumor progression and chemoresistance. Cancer-associated fibroblasts act as the protagonists of the tumor microenvironment, fostering the metastasis, stemness, and chemoresistance of cancer cells and attenuating the anti-cancer immune responses. Gastric cancer is one of the most aggressive cancers in the clinic, refractory to anti-cancer therapies. Growing evidence indicates that cancer-associated fibroblasts are the most prominent risk factors for a poor tumor immune microenvironment and dismal prognosis in gastric cancer. Therefore, targeting cancer-associated fibroblasts may be central to surpassing resistance to conventional chemotherapeutics, molecular-targeted agents, and immunotherapies, improving survival in gastric cancer. However, the heterogeneity in cancer-associated fibroblasts may complicate the development of cancer-associated fibroblast targeting approaches. Although single-cell sequencing studies started dissecting the heterogeneity of cancer-associated fibroblasts, the research community should still answer these questions: "What makes a cancer-associated fibroblast protumorigenic?"; "How do the intracellular signaling and the secretome of different cancer-associated fibroblast subpopulations differ from each other?"; and "Which cancer-associated fibroblast subtypes predominate specific cancer types?". Unveiling these questions can pave the way for discovering efficient cancer-associated fibroblast targeting strategies. Here, we review current knowledge and perspectives on these questions, focusing on how CAFs induce aggressiveness and therapy resistance in gastric cancer. We also review potential therapeutic approaches to prevent the development and activation of cancer-associated fibroblasts via inhibition of CAF inducers and CAF markers in cancer.
Collapse
Affiliation(s)
- Ece Ozmen
- Koç University Graduate School of Health Sciences, Istanbul, Türkiye
| | - Tevriz Dilan Demir
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
| | - Gulnihal Ozcan
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
- Department of Medical Pharmacology, Koç University School of Medicine, Istanbul, Türkiye
| |
Collapse
|
5
|
Zarezadeh SM, Sharafi AM, Erabi G, Tabashiri A, Teymouri N, Mehrabi H, Golzan SA, Faridzadeh A, Abdollahifar Z, Sami N, Arabpour J, Rahimi Z, Ansari A, Abbasi MR, Azizi N, Tamimi A, Poudineh M, Deravi N. Natural STAT3 Inhibitors for Cancer Treatment: A Comprehensive Literature Review. Recent Pat Anticancer Drug Discov 2024; 19:403-502. [PMID: 37534488 DOI: 10.2174/1574892818666230803100554] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 06/05/2023] [Accepted: 06/09/2023] [Indexed: 08/04/2023]
Abstract
Cancer is one of the leading causes of mortality and morbidity worldwide, affecting millions of people physically and financially every year. Over time, many anticancer treatments have been proposed and studied, including synthetic compound consumption, surgical procedures, or grueling chemotherapy. Although these treatments have improved the daily life quality of patients and increased their survival rate and life expectancy, they have also shown significant drawbacks, including staggering costs, multiple side effects, and difficulty in compliance and adherence to treatment. Therefore, natural compounds have been considered a possible key to overcoming these problems in recent years, and thorough research has been done to assess their effectiveness. In these studies, scientists have discovered a meaningful interaction between several natural materials and signal transducer and activator of transcription 3 molecules. STAT3 is a transcriptional protein that is vital for cell growth and survival. Mechanistic studies have established that activated STAT3 can increase cancer cell proliferation and invasion while reducing anticancer immunity. Thus, inhibiting STAT3 signaling by natural compounds has become one of the favorite research topics and an attractive target for developing novel cancer treatments. In the present article, we intend to comprehensively review the latest knowledge about the effects of various organic compounds on inhibiting the STAT3 signaling pathway to cure different cancer diseases.
Collapse
Affiliation(s)
- Seyed Mahdi Zarezadeh
- Students' Scientific Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Mohammad Sharafi
- Students' Scientific Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Gisou Erabi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Arefeh Tabashiri
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Navid Teymouri
- Student Research Committee, Tabriz University of Medical Science, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hoda Mehrabi
- Student Research Committee, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Seyyed Amirhossein Golzan
- Student Research Committee, Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arezoo Faridzadeh
- Department of Immunology and Allergy, Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Abdollahifar
- Student Research Committee, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Nafiseh Sami
- Student Research Committee, Tehran Medical Sciences, Islamic Azad University Medical Branch of Tehran, Tehran, Iran
| | - Javad Arabpour
- Department of Microbiology, Faculty of New Sciences, Islamic Azad University Medical Branch of Tehran, Tehran, Iran
| | - Zahra Rahimi
- School of Medicine, Zanjan University of Medical Sciences Zanjan, Iran
| | - Arina Ansari
- Student Research Committee, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | | | - Nima Azizi
- Students' Scientific Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Niloofar Deravi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Ren Y, Wu H, Tan M, Chen J, Duan Z, Zhu B, Ruan X, Yu Q, Li S, Liu X, Liu Y, Si Y. Acetylation of MOB1 mediates polyphyllin II-reduced lysosome biogenesis in breast cancer by promoting the cytoplasmic retention of the YAP/TFEB coactivator complex. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 122:155152. [PMID: 37922793 DOI: 10.1016/j.phymed.2023.155152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/27/2023] [Accepted: 10/14/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND Autophagy‒lysosome abnormalities are associated with the malignant progression of cancer. Transcription factor EB (TFEB) is the master transcriptional regulator of the autophagy‒lysosome machinery, and its abnormal activity is associated with autophagy-lysosome dysfunction. Polyphyllin II (PPII), an active steroidal saponin isolated from the rhizomes of Paris polyphylla, has been demonstrated to have antitumor activity. PURPOSE Here, we explored the antitumor activity of PPII in breast cancer (BC) and further clarified its mechanism. METHODS Autophagosome was detected by transmission electron microscopy, an autophagy indicator system, and western blot. The effect of PPII on lysosomal activity was evaluated by flow cytometry, a lysosomal cathepsin activity assay, and acridine orange staining. The effect of PPII on the signaling pathway was evaluated by Western blot, gene expression measurement, gene alterations. The binding of PPII and MOB1 was examined through a drug affinity responsive target stability assay. The pharmacokinetic parameters of PPII were evaluated in Sprague-Dawley rats. RESULTS PPII exhibits therapeutic potential in BC by inducing the accumulation of autophagosome. PPII promotes the cytoplasmic retention of YAP/TFEB, which is responsible for the accumulation of autophagosome in BC. PPII activates Hippo signaling to promote cytoplasmic retention of YAP. PPII activates Hippo signaling by accelerating acetylation of MOB1 through a direct binding interaction. CONCLUSION Taken together, these results confirm that acetylation of MOB1 mediates PPII-induced autophagosome accumulation in BC by promoting cytoplasmic retention of the YAP/TFEB coactivator complex. PPII is expected to be a drug candidate for the treatment of BC based on lysosomal biosynthesis.
Collapse
Affiliation(s)
- Yuliang Ren
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Hui Wu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China; Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China
| | - Miao Tan
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Junjie Chen
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China
| | - Zhongqi Duan
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Bingxin Zhu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xuzhi Ruan
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Qingqing Yu
- Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China
| | - Shuzhen Li
- Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xuewen Liu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China; Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China.
| | - Ying Liu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China; Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China.
| | - Yuan Si
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China; Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China.
| |
Collapse
|
7
|
Zhang XL, Xiao W, Qian JP, Yang WJ, Xu H, Xu XD, Zhang GW. The Role and Application of Fibroblast Activating Protein. Curr Mol Med 2024; 24:1097-1110. [PMID: 37259211 DOI: 10.2174/1566524023666230530095305] [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: 08/07/2022] [Revised: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 06/02/2023]
Abstract
Fibroblast activation protein-α (FAP), a type-II transmembrane serine protease, is rarely expressed in normal tissues but highly abundant in pathological diseases, including fibrosis, arthritis, and cancer. Ever since its discovery, we have deciphered its structure and biological properties and continue to investigate its roles in various diseases while attempting to utilize it for targeted therapy. To date, no significant breakthroughs have been made in terms of efficacy. However, in recent years, several practical applications in the realm of imaging diagnosis have been discovered. Given its unique expression in a diverse array of pathological tissues, the fundamental biological characteristics of FAP render it a crucial target for disease diagnosis and immunotherapy. To obtain a more comprehensive understanding of the research progress of FAP, its biological characteristics, involvement in diseases, and recent targeted application research have been reviewed. Moreover, we explored its development trend in the direction of clinical diagnoses and treatment.
Collapse
Affiliation(s)
- Xiao-Lou Zhang
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wang Xiao
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jian-Ping Qian
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wan-Jun Yang
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hao Xu
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xing-da Xu
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guo-Wei Zhang
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
8
|
Bai Y, Li M, Geng D, Liu S, Chen Y, Li S, Zhang S, Wang H. Polyphyllins in cancer therapy: A systematic review and meta-analysis of animal studies. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 121:155096. [PMID: 37769554 DOI: 10.1016/j.phymed.2023.155096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 09/07/2023] [Accepted: 09/16/2023] [Indexed: 10/03/2023]
Abstract
BACKGROUND Polyphyllins are secondary metabolites that inhibit the growth of various tumours; however, clinical trials on their use are lacking. HYPOTHESIS/PURPOSE In this study, we aimed to evaluate the antitumour efficacy of polyphyllins in animal models. STUDY DESIGN Systematic review and meta-analysis. METHODS Electronic bibliographic databases including PubMed, Web of Science, China Science and Technology Journal Database, Wanfang Data, and China National Knowledge Infrastructure were searched for relevant articles. The Systematic Review Centre for Laboratory Animal Experimentation's Risk of Bias tool was used to assess methodological quality. RevMan V.5.4 (Cochrane) and Stata MP 17 software were used to perform a meta-analysis. RESULTS Thirty articles were analysed including 33 independent experiments and 452 animals in this paper. Overall, tumour volume (standardised mean difference [SMD]: -3.35; 95 % confidence interval [CI]: -4.27 to -2.43; p < 0.00001) and tumour weight (SMD: -3.79; 95% CI: -4.75 to -2.82; p < 0.00001) were reduced by polyphyllins, which showed a good cancer therapeutic effect; mouse weight (SMD: -0.22; 95% CI: -0.61 to -0.18; p = 0.28) was insignificantly different, which indicated that polyphyllins did not affect the growth of the mice within the test range. Moreover, the molecular mechanisms of the antitumour activity of polyphyllins were explained, including the P53, NF-kB, AMPK, and ERK signalling pathways. CONCLUSION Polyphyllins inhibit the growth of cancers within the experimental dose. However, due to heterogeneity of the results of the included studies, more studies are needed to support this conclusion.
Collapse
Affiliation(s)
- Yan Bai
- Zhejiang Provincial Key Laboratory of Resources Protection and Innovation of Traditional Chinese Medicine, Hangzhou 311300, China; College of Food and Health, Department of Traditional Chinese Medicine, Zhejiang Agriculture & Forestry University, Hangzhou 311300, China; State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, Hangzhou 311300, China.
| | - Mengmeng Li
- Zhejiang Provincial Key Laboratory of Resources Protection and Innovation of Traditional Chinese Medicine, Hangzhou 311300, China; College of Food and Health, Department of Traditional Chinese Medicine, Zhejiang Agriculture & Forestry University, Hangzhou 311300, China; State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, Hangzhou 311300, China
| | - Dongjie Geng
- Zhejiang Provincial Key Laboratory of Resources Protection and Innovation of Traditional Chinese Medicine, Hangzhou 311300, China; College of Food and Health, Department of Traditional Chinese Medicine, Zhejiang Agriculture & Forestry University, Hangzhou 311300, China; State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, Hangzhou 311300, China
| | - Shouzan Liu
- State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, Hangzhou 311300, China; Botanical Garden, Zhejiang Agricultural and Forestry University, Hangzhou 311300, China
| | - Ye Chen
- Zhejiang Provincial Key Laboratory of Resources Protection and Innovation of Traditional Chinese Medicine, Hangzhou 311300, China; College of Food and Health, Department of Traditional Chinese Medicine, Zhejiang Agriculture & Forestry University, Hangzhou 311300, China; State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, Hangzhou 311300, China
| | - Shan Li
- Zhejiang Provincial Key Laboratory of Resources Protection and Innovation of Traditional Chinese Medicine, Hangzhou 311300, China; College of Food and Health, Department of Traditional Chinese Medicine, Zhejiang Agriculture & Forestry University, Hangzhou 311300, China; State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, Hangzhou 311300, China
| | - Shaobo Zhang
- Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou 311300, China
| | - Hongzhen Wang
- Zhejiang Provincial Key Laboratory of Resources Protection and Innovation of Traditional Chinese Medicine, Hangzhou 311300, China; College of Food and Health, Department of Traditional Chinese Medicine, Zhejiang Agriculture & Forestry University, Hangzhou 311300, China; State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, Hangzhou 311300, China.
| |
Collapse
|
9
|
Du H, Wu H, Kang Q, Liao M, Qin M, Chen N, Huang H, Huang D, Wang P, Tong G. Polyphyllin I attenuates the invasion and metastasis via downregulating GRP78 in drug-resistant hepatocellular carcinoma cells. Aging (Albany NY) 2023; 15:12251-12263. [PMID: 37934581 PMCID: PMC10683619 DOI: 10.18632/aging.205176] [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: 08/01/2023] [Accepted: 10/03/2023] [Indexed: 11/08/2023]
Abstract
Drug resistance to chemotherapy agents presents a major obstacle to the effective treatment of hepatocellular carcinoma (HCC), a common type of liver cancer. Increasing evidence indicates a link between drug resistance and the recurrence of HCC. Polyphyllin I (PPI), a promising pharmaceutical candidate, has shown potential therapeutic advantages in the treatment of sorafenib-resistant hepatocellular carcinoma (SR-HCC cells). In this study, we sought to investigate the mechanism underlying the inhibitory effect of PPI on the invasion and metastasis of SR-HCC cells. Our in vitro studies included scratch wound-healing migration assays and transwell assays to examine PPI's effect on HCC cell migration and invasion. Flow cytometry was employed to analyze the accumulation or efflux of chemotherapy drugs. The results of these experiments demonstrated that PPI increased the susceptibility of HCC to sorafenib while inhibiting SR-HCC cell growth, migration, and invasion. Molecular docking analysis revealed that PPI exhibited a higher binding affinity with GRP78. Western blot analysis and immunofluorescence experiments showed that PPI reduced the expression of GRP78, E-cadherin, N-cadherin, Vimentin, and ABCG2 in SR-HCC cells. Interference with and overproduction of GRP78 in vitro impacted the proliferation, migration, invasion, and metastasis of HCC cells. Further examination revealed that PPI hindered the expression of GRP78 protein, resulting in a suppressive effect on SR-HCC cell migration and invasion. Histological examination of tumor tissue substantiated that administering PPI via gavage to HepG2/S xenograft nude mice inhibited tumor growth and significantly reduced tumor size, as evidenced by xenograft experiments involving nude mice. Hematoxylin and eosin (HE) staining of tumor tissue specimens, along with immunohistochemistry (IHC), were conducted to evaluate the expression levels of Ki67, GRP78, N-cadherin, Vimentin, and ABCG2. The results indicated that PPI administration decreased the levels of proteins associated with metastasis and markers of drug resistance in tumor tissues, impeding tumor growth and spread. Overall, our findings demonstrated that PPI effectively suppressed the viability, proliferation, invasion, and metastasis of SR-HCC cells both in vitro and in vivo by modulating GRP78 activity. These findings provide new insights into the mechanism of PPI inhibition of SR-HCC cell invasion and metastasis, highlighting PPI as a potential treatment option for sorafenib-resistant HCC.
Collapse
Affiliation(s)
- Haiyan Du
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518000, Guangdong, China
| | - Haochen Wu
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518000, Guangdong, China
| | - Qinyang Kang
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518000, Guangdong, China
| | - Mianmian Liao
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518000, Guangdong, China
| | - Meirong Qin
- Shenzhen Institute for Drug Control, Shenzhen 518000, Guangdong, China
| | - Ning Chen
- Shenzhen Institute for Drug Control, Shenzhen 518000, Guangdong, China
| | - Houshuang Huang
- Shenzhen Institute for Drug Control, Shenzhen 518000, Guangdong, China
| | - Danping Huang
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518000, Guangdong, China
- Department of Integrated Traditional Chinese and Western Medicine, School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
| | - Ping Wang
- Shenzhen Institute for Drug Control, Shenzhen 518000, Guangdong, China
| | - Guangdong Tong
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518000, Guangdong, China
| |
Collapse
|
10
|
Yuan L, Miao H, Ding H, Zhang F, Lou ZK, Li XG. Polyphyllin I suppressed the apoptosis of intervertebral disc nucleus pulposus cells induced by IL-1β by miR-503-5p/Bcl-2 axis. J Orthop Surg Res 2023; 18:466. [PMID: 37380996 DOI: 10.1186/s13018-023-03947-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 06/22/2023] [Indexed: 06/30/2023] Open
Abstract
BACKGROUND There are no studies that have shown the role and underlying mechanism of Polyphyllin I (PPI)-mediated anti-apoptosis activity in nucleus pulposus cells (NPCs). The research aimed to evaluate the effects of PPI in interleukin (IL)-1β-induced NPCs apoptosis in vitro. METHODS Cell Counting Kit-8 (CCK-8) assay was used to detect cell viability, and cell apoptosis was evaluated by double-stained flow cytometry (FITC Annexin V/PI). The expression of miR-503-5p was quantified by real-time quantitative PCR (qRT-PCR), and the expression of Bcl-2, Bax, and cleaved caspase-3 was quantified by Western blot. Dual-luciferase reporter gene assay was used to detect the targeting relationship between miR-503-5p and Bcl-2. RESULTS PPI at 40 μg·mL-1 markedly promoted the viability of NPCs (P < 0.01). Also, PPI inhibited apoptosis and reduction in proliferative activity induced by IL-1β in the NPCs (P < 0.001, 0.01). PPI treatment significantly inhibited the expression of apoptosis-related protein Bax, cleaved caspase-3 (P < 0.05, 0.01), and enhanced the level of anti-apoptotic protein Bcl-2 (P < 0.01). The proliferative activity of NPCs was significantly decreased and the apoptosis rate of NPCs was increased under IL-1β treatment (P < 0.01, 0.001). Moreover, miR-503-5p was highly expressed in IL-1β-induced NPCs (P < 0.001). Furthermore, the effect of PPI on NPCs viability and apoptosis in IL-1β treatment was dramatically reversed by the overexpression of miR-503-5p (P < 0.01, 0.01). The targeted binding of miR-503-5p to the 3'UTR of Bcl-2 mRNA was confirmed by dual-luciferase reporter gene assays (P < 0.05). In further experiments, compared with miR-503-5p mimics, the effects of PPI on IL-1β-induced NPCs viability and apoptosis were greatly reversed by the co-overexpression of miR-503-5p and Bcl-2 (P < 0.05, 0.05). CONCLUSION PPI suppressed the apoptosis of intervertebral disk (IVD) NPCs induced by IL-1β via miR-503-5p/Bcl-2 molecular axis.
Collapse
Affiliation(s)
- Lei Yuan
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming City, 650031, Yunnan Province, China
| | - Hui Miao
- Rehabilitation Department, Yantai Yuhuangding Hospital, Yantai, 264001, Shandong, China
| | - Heng Ding
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming City, 650031, Yunnan Province, China
| | - Fan Zhang
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming City, 650031, Yunnan Province, China
| | - Zhen-Kai Lou
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming City, 650031, Yunnan Province, China
| | - Xing-Guo Li
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming City, 650031, Yunnan Province, China.
| |
Collapse
|
11
|
Zheng F, Wang Y, Zhang Q, Chen Q, Liang CL, Liu H, Qiu F, Chen Y, Huang H, Lu W, Dai Z. Polyphyllin I suppresses the gastric cancer growth by promoting cancer cell ferroptosis. Front Pharmacol 2023; 14:1145407. [PMID: 37081971 PMCID: PMC10110865 DOI: 10.3389/fphar.2023.1145407] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/27/2023] [Indexed: 04/07/2023] Open
Abstract
Background: Ferroptosis is a new form of regulated cell death characterized by the accumulation of iron-dependent lipid peroxides and membrane damages. Recent studies have identified an important role for cancer cell ferroptosis in antitumor therapy. On the other hand, polyphyllin I (PPI) has been reported to exert antitumor effects on some types of cancers. However, it remains unknown whether or not PPI regulates cancer cell ferroptosis.Methods: Two types of human gastric cancer cells (AGS and MKN-45) were used to establish tumor xenograft models in nude mice that were treated with polyphyllin I (PPI) to observe tumor growth, while cells also were cultured for in vitro studies. Ferroptosis, based on the intracellular ROS/lipid ROS production and accumulation of ferrous ions, was detected using a fluorescence microscope and flow cytometer, while the expression of NRF2/FTH1 was measured using Western blotting assays.Results: Here we found that PPI inhibited the gastric cancer growth in vivo and in vitro while increasing the intracellular reactive oxygen species (ROS)/lipid peroxides and ferrous ions in the gastric cancer cells. PPI also decreased the levels of nuclear factor erythroid 2-related factor 2 (NRF2) and ferritin heavy chain 1 (FTH1) in gastric cancer cells in vitro. Moreover, liproxstain-1, an inhibitor of cell ferroptosis, mostly reversed the cell ferroptosis and tumor growth arrest induced by PPI. Finally, the effects of PPI on cancer cell ferroptosis were diminished by the overexpression of NRF2.Conclusion: For the first time, our results have demonstrated that PPI exerts its antitumor activity on the gastric cancer by, at least partially, inducing cancer cell ferroptosis via regulating NRF2/FTH1 pathway. These findings may be implicated for clinical replacement therapy of the gastric cancer.
Collapse
Affiliation(s)
- Fang Zheng
- Section of Immunology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, China
| | - Yeshu Wang
- Section of Immunology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Qunfang Zhang
- Section of Immunology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, China
| | - Qiuyuan Chen
- Section of Immunology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Chun-Ling Liang
- Section of Immunology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Huazhen Liu
- Section of Immunology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Feifei Qiu
- Section of Immunology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yuchao Chen
- Section of Immunology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Haiding Huang
- Section of Immunology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, China
| | - Weihui Lu
- Section of Immunology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, China
- *Correspondence: Weihui Lu, ; Zhenhua Dai,
| | - Zhenhua Dai
- Section of Immunology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, China
- *Correspondence: Weihui Lu, ; Zhenhua Dai,
| |
Collapse
|
12
|
Butti R, Khaladkar A, Bhardwaj P, Prakasam G. Heterotypic signaling of cancer-associated fibroblasts in shaping the cancer cell drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:182-204. [PMID: 37065872 PMCID: PMC10099601 DOI: 10.20517/cdr.2022.72] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 09/28/2022] [Accepted: 11/22/2022] [Indexed: 03/29/2023]
Abstract
The context-dependent reciprocal interaction between the cancer cells and surrounding fibroblasts is imperative for regulating malignant potential, metabolic reprogramming, immunosuppression, and ECM deposition. However, recent evidence also suggests that cancer-associated fibroblasts induce chemoresistance in cancer cells to various anticancer regimens. Because of the protumorigenic function of cancer-associated fibroblasts, these stromal cell types have emerged as fascinating therapeutic targets for cancer. However, this notion was recently challenged by studies that targeted cancer-associated fibroblasts and highlighted the underlying heterogeneity by identifying a subset of these cells with tumor-restricting functions. Hence, it is imperative to understand the heterogeneity and heterotypic signaling of cancer-associated fibroblasts to target tumor-promoting signaling processes by sparing tumor-restricting ones. In this review, we discuss the heterogeneity and heterotypic signaling of cancer-associated fibroblasts in shaping drug resistance and also list the cancer-associated fibroblast-targeting therapeutics.
Collapse
Affiliation(s)
- Ramesh Butti
- Kidney Cancer Program, Simmons Comprehensive Cancer Centre, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Ashwini Khaladkar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Bombay 400076, India
- Authors contributed equally
| | - Priya Bhardwaj
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi 110029, India
- Authors contributed equally
| | - Gopinath Prakasam
- Kidney Cancer Program, Simmons Comprehensive Cancer Centre, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| |
Collapse
|
13
|
Zhang X, Yang G, Zhao Y, Dai X, Liu W, Qu F, Huang Y. Selection and Identification of an ssDNA Aptamer for Fibroblast Activation Protein. Molecules 2023; 28:1682. [PMID: 36838669 PMCID: PMC9965484 DOI: 10.3390/molecules28041682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/16/2023] [Accepted: 01/31/2023] [Indexed: 02/12/2023] Open
Abstract
As a type II transmembrane serine protease, fibroblast activation protein (FAP) is specifically expressed on the surface of fibroblasts associated with a variety of epithelial-derived malignancies such as pancreatic cancer, breast cancer, and colon cancer. It participates in the processes of tumorigenesis, progression, and immunosuppression. FAP constitutes an important target for tumor treatment; however, the current studies on FAP are mainly related to structural characteristics, enzymatic properties, and biological functions, and aptamers of FAP have not been investigated. In this work, by using recombinant human FAP as the target, five candidate aptamers, which are AptFAP-A1, AptFAP-A2, AptFAP-A3, AptFAP-A4, and AptFAP-A5, were selected by capillary electrophoresis-systematic evolution of ligands by exponential enrichment (CE-SELEX), and their secondary structures were predicted to be mainly stem-loop. Moreover, the CE-laser-induced fluorescence (LIF) method was used to determine the equilibrium dissociation constant KD values between the FAP protein and candidate aptamers, and the KD value was in the low molar range. Finally, Cy5-labeled aptamers were co-incubated with human pancreatic cancer-associated fibroblasts highly expressing FAP protein, and confocal microscopy imaging showed that aptamer AptFAP-A4 had the highest affinities with the cells. The FAP aptamers screened in this study provide a promising direction for the development of rapid tumor diagnosis and targeted therapy.
Collapse
Affiliation(s)
- Xiaomin Zhang
- Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, School of Life Science, Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing 100081, China
| | - Ge Yang
- Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, School of Life Science, Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing 100081, China
- CAMS Key Laboratory of Antiviral Drug Research, Beijing Key Laboratory of Antimicrobial Agents, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yi Zhao
- Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, School of Life Science, Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing 100081, China
| | - Xuyan Dai
- Economic College, Hunan Agricultural University, Changsha 410128, China
| | - Wenjing Liu
- Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing 101125, China
| | - Feng Qu
- Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, School of Life Science, Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing 100081, China
| | - Yuanyu Huang
- Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, School of Life Science, Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
14
|
Song P, Pan Q, Sun Z, Zou L, Yang L. Fibroblast activation protein alpha: Comprehensive detection methods for drug target and tumor marker. Chem Biol Interact 2022; 354:109830. [PMID: 35104486 DOI: 10.1016/j.cbi.2022.109830] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/22/2021] [Accepted: 01/21/2022] [Indexed: 11/25/2022]
Abstract
Fibroblast activation protein alpha (FAP-α, EC3.4.2. B28), a type II transmembrane proteolytic enzyme for the serine protease peptidase family. It is underexpressed in normal tissues but increased significantly in disease states, especially in neoplasm, which is a potential biomarker to turmor diagnosis. The inhibition of FAP-α activity will retard tumor formation, which is expected to be a promising tumor therapeutic target. At present, although the FAP-α expression detection methods has diversification, a superlative detection means is necessary for the clinical diagnosis. This review covers the discovery and the latest advances in FAP-α, as well as the future research prospects. The tissue distribution, structural characteristics, small-molecule ligands and structure-activity relationship of major inhibitors of FAP-α were summarized in this review. Furthermore, a variety of detection methods including traditional detection methods and emerging probes detection were classified and compared, and the design strategy and kinetic parameters of these FAP-α probe substrates were summarized. In addition, these comprehensive information provides a series of practical and reliable assays for the optimal design principles of FAP-α probes, promoting the application of FAP-α as a disease marker in diagnosis, and a drug target in drug design.
Collapse
Affiliation(s)
- Peifang Song
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Quisha Pan
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | | | - Liwei Zou
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Ling Yang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
15
|
Chen C, Yu LT, Cheng BR, Xu JL, Cai Y, Jin JL, Feng RL, Xie L, Qu XY, Li D, Liu J, Li Y, Cui XY, Lu JJ, Zhou K, Lin Q, Wan J. Promising Therapeutic Candidate for Myocardial Ischemia/Reperfusion Injury: What Are the Possible Mechanisms and Roles of Phytochemicals? Front Cardiovasc Med 2022; 8:792592. [PMID: 35252368 PMCID: PMC8893235 DOI: 10.3389/fcvm.2021.792592] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 12/29/2021] [Indexed: 12/12/2022] Open
Abstract
Percutaneous coronary intervention (PCI) is one of the most effective reperfusion strategies for acute myocardial infarction (AMI) despite myocardial ischemia/reperfusion (I/R) injury, causing one of the causes of most cardiomyocyte injuries and deaths. The pathological processes of myocardial I/R injury include apoptosis, autophagy, and irreversible cell death caused by calcium overload, oxidative stress, and inflammation. Eventually, myocardial I/R injury causes a spike of further cardiomyocyte injury that contributes to final infarct size (IS) and bound with hospitalization of heart failure as well as all-cause mortality within the following 12 months. Therefore, the addition of adjuvant intervention to improve myocardial salvage and cardiac function calls for further investigation. Phytochemicals are non-nutritive bioactive secondary compounds abundantly found in Chinese herbal medicine. Great effort has been put into phytochemicals because they are often in line with the expectations to improve myocardial I/R injury without compromising the clinical efficacy or to even produce synergy. We summarized the previous efforts, briefly outlined the mechanism of myocardial I/R injury, and focused on exploring the cardioprotective effects and potential mechanisms of all phytochemical types that have been investigated under myocardial I/R injury. Phytochemicals deserve to be utilized as promising therapeutic candidates for further development and research on combating myocardial I/R injury. Nevertheless, more studies are needed to provide a better understanding of the mechanism of myocardial I/R injury treatment using phytochemicals and possible side effects associated with this approach.
Collapse
Affiliation(s)
- Cong Chen
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Lin-Tong Yu
- Department of Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bai-Ru Cheng
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Jiang-Lin Xu
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Yun Cai
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Jia-Lin Jin
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Ru-Li Feng
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Long Xie
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Xin-Yan Qu
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Dong Li
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Jing Liu
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Yan Li
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Xiao-Yun Cui
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Jin-Jin Lu
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Kun Zhou
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Qian Lin
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Qian Lin
| | - Jie Wan
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
- Jie Wan
| |
Collapse
|
16
|
Polyphyllin I combined with doxorubicin shows chemosensitization effect in vivo and reduces immunotoxicity of doxorubicin. Mol Cell Toxicol 2022. [DOI: 10.1007/s13273-021-00206-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
17
|
Elboga U, Sahin E, Kus T, Cayirli YB, Aktas G, Uzun E, Cinkir HY, Teker F, Sever ON, Aytekin A, Yilmaz L, Aytekin A, Cimen U, Mumcu V, Kilbas B, Çelen YZ. Superiority of 68Ga-FAPI PET/CT scan in detecting additional lesions compared to 18FDG PET/CT scan in breast cancer. Ann Nucl Med 2021; 35:1321-1331. [PMID: 34436740 DOI: 10.1007/s12149-021-01672-x] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/18/2021] [Indexed: 02/06/2023]
Abstract
PURPOSE We compared the ability of 68Ga-FAPI PET//CT with 18FDG PET/CT imaging techniques to detect additional lesions in breast cancer patients that may affect further chemotherapy options. METHODS A total of 48 patients with breast cancer underwent concurrent 68Ga-FAPI-04 and 18FDG PET/CT regardless of whether they had received chemotherapy or not in the last month before imaging. Both modalities were compared according to various parameters: clinical/pathological features, number of lesions detected, activity uptake (SUVmax), and the effect on the evaluation of response to treatment in the post-chemotherapy group. RESULTS This retrospective study included 48 patients with breast cancer (mean age 53.3 ± 11.7 years; IDC 89.6%; ILC 10.4%). In the comparison of both modalities, no statistical significance was obtained in terms of the pathological characteristics of the patients. More lesions were demonstrated in all categorized regions in 68Ga-FAPI PET/CT imaging with higher uptake values compared to 18FDG PET/CT in this study. In the treatment response evaluation of the post-chemotherapy group, 12 cases (12/24) who were evaluated as PMR, CMR, or SD according to 18FDG PET/CT results were later accepted as PD due to newly detected lesions in complementary 68Ga-FAPI PET/CT imaging and treatment of patients was managed accordingly by clinicians. CONCLUSION It was determined that 68Ga-FAPI PET/CT was superior to 18FDG PET/CT in terms of accuracy and it was thought that 68Ga-FAPI PET/CT could be utilized as an additional complementary imaging to 18FDG PET/CT. Moreover, 68Ga-FAPI PET/CT, with its significant theranostic potential, could become a key element in predicting the pathological response of breast cancer patients in further researches.
Collapse
Affiliation(s)
- Umut Elboga
- Department of Nuclear Medicine, Gaziantep University, 27310, Sahinbey, Gaziantep, Turkey.
| | - Ertan Sahin
- Department of Nuclear Medicine, Gaziantep University, 27310, Sahinbey, Gaziantep, Turkey
| | - Tulay Kus
- Department of Oncology, Gaziantep University, Gaziantep, Turkey
| | - Yusuf Burak Cayirli
- Department of Nuclear Medicine, Gaziantep University, 27310, Sahinbey, Gaziantep, Turkey
| | - Gokmen Aktas
- Department of Oncology, Medical Park Private Hospital, Gaziantep, Turkey
| | - Evren Uzun
- Department of Pathology, Gaziantep University, Gaziantep, Turkey
| | | | - Fatih Teker
- Department of Oncology, Gaziantep University, Gaziantep, Turkey
| | | | - Alper Aytekin
- Department of General Surgery, Gaziantep University, Gaziantep, Turkey
| | - Latif Yilmaz
- Department of General Surgery, Gaziantep University, Gaziantep, Turkey
| | - Aydin Aytekin
- Department of Oncology, Gaziantep University, Gaziantep, Turkey
| | - Ufuk Cimen
- Department of Nuclear Medicine, Gaziantep University, 27310, Sahinbey, Gaziantep, Turkey
| | - Vuslat Mumcu
- Department of Nuclear Medicine, Gaziantep University, 27310, Sahinbey, Gaziantep, Turkey
| | - Benan Kilbas
- Department of Chemistry, Moltek Inc., İstanbul, Turkey
| | - Y Zeki Çelen
- Department of Nuclear Medicine, Gaziantep University, 27310, Sahinbey, Gaziantep, Turkey
| |
Collapse
|
18
|
Xin L, Gao J, Zheng Z, Chen Y, Lv S, Zhao Z, Yu C, Yang X, Zhang R. Fibroblast Activation Protein-α as a Target in the Bench-to-Bedside Diagnosis and Treatment of Tumors: A Narrative Review. Front Oncol 2021; 11:648187. [PMID: 34490078 PMCID: PMC8416977 DOI: 10.3389/fonc.2021.648187] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 07/28/2021] [Indexed: 12/13/2022] Open
Abstract
Fibroblast activation protein-α (FAP) is a type II integral serine protease that is specifically expressed by activated fibroblasts. Cancer-associated fibroblasts (CAFs) in the tumor stroma have an abundant and stable expression of FAP, which plays an important role in promoting tumor growth, invasion, metastasis, and immunosuppression. For example, in females with a high incidence of breast cancer, CAFs account for 50–70% of the cells in the tumor’s microenvironment. CAF overexpression of FAP promotes tumor development and metastasis by influencing extracellular matrix remodeling, intracellular signaling, angiogenesis, epithelial-to-mesenchymal transition, and immunosuppression. This review discusses the basic biological characteristics of FAP and its applications in the diagnosis and treatment of various cancers. We review the emerging basic and clinical research data regarding the use of nanomaterials that target FAP.
Collapse
Affiliation(s)
- Lei Xin
- Department of Radiology, Affiliated Tumor Hospital of Shanxi Medical University, Taiyuan, China
| | - Jinfang Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Ziliang Zheng
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Yiyou Chen
- Department of Radiology, Affiliated Tumor Hospital of Shanxi Medical University, Taiyuan, China
| | - Shuxin Lv
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Zhikai Zhao
- Department of Radiology, Affiliated Tumor Hospital of Shanxi Medical University, Taiyuan, China
| | - Chunhai Yu
- Department of Radiology, Affiliated Tumor Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaotang Yang
- Department of Radiology, Affiliated Tumor Hospital of Shanxi Medical University, Taiyuan, China
| | - Ruiping Zhang
- Department of Radiology, Affiliated Tumor Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
19
|
Zhu H, Liu X. Advances of Tumorigenesis, Diagnosis at Early Stage, and Cellular Immunotherapy in Gastrointestinal Malignancies. Front Oncol 2021; 11:666340. [PMID: 34434889 PMCID: PMC8381364 DOI: 10.3389/fonc.2021.666340] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 07/19/2021] [Indexed: 01/10/2023] Open
Abstract
Globally, in 2018, 4.8 million new patients have a diagnosis of gastrointestinal (GI) cancers, while 3.4 million people died of such disorders. GI malignancies are tightly relevant to 26% of the world-wide cancer incidence and occupies 35% of all cancer-associated deaths. In this article, we principally investigated molecular and cellular mechanisms of tumorigenesis in five major GI cancers occurring at esophagus, stomach, liver, pancreas, and colorectal region that illustrate high morbidity in Eastern and Western countries. Moreover, through this investigation, we not only emphasize importance of the tumor microenvironment in development and treatment of malignant tumors but also identify significance of M2PK, miRNAs, ctDNAs, circRNAs, and CTCs in early detection of GI cancers, as well as systematically evaluate contribution of personalized precision medicine including cellular immunotherapy, new antigen and vaccine therapy, and oncolytic virotherapy in treatment of GI cancers.
Collapse
Affiliation(s)
- Haipeng Zhu
- Precision and Personalized Cancer Treatment Center, Division of Cancer Diagnosis & Therapy, Ciming Boao International Hospital, Boao Lecheng International Medical Tourism Pilot Zone, Qionghai, China.,Stem Cell and Biotherapy Technology Research Center, Xinxiang Medical College, Xinxiang, China
| | - Xiaojun Liu
- Division of Cellular & Biomedical Science, Ciming Boao International Hospital, Boao Lecheng International Medical Tourism Pilot Zone, Qionghai, China
| |
Collapse
|
20
|
Yu S, Gao W, Zeng P, Chen C, Liu Z, Zhang Z, Liu J. Exploring the effect of Polyphyllin I on hepatitis B virus-related liver cancer through network pharmacology and in vitro experiments. Comb Chem High Throughput Screen 2021; 25:934-944. [PMID: 34397325 DOI: 10.2174/1386207324666210816141436] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 06/12/2021] [Accepted: 06/15/2021] [Indexed: 11/22/2022]
Abstract
AIM AND OBJECTIVE To investigate the effect of Polyphyllin I (PPI) on HBV-related liver cancer through network pharmacology and in vitro experiments, and to explore its mechanism of action. MATERIALS AND METHODS Use bioinformatics software to predict the active ingredient target of PPI and the disease target of liver cancer, and perform active ingredient-disease target analysis. The results of network pharmacology through molecular docking and in vitro experiments can be further verified. The HepG2 receptor cells (HepG2. 2. 15) were transfected with HBV plasmid for observation, with the human liver cancer HepG2 being used as the control. RESULTS Bioinformatics analysis found that PPI had totally 161 protein targets, and the predicted target and liver cancer targets were combined to obtain 13 intersection targets. The results of molecular docking demonstrated that PPI had good affinity with STAT3, PTP1B, IL2, and BCL2L1. The results of the in vitro experiments indicated that the PPI inhibited cell proliferation and metastasis in a concentration-dependent manner (P<0.01). Compared with the vehicle group, the PPI group of 1.5, 3, and 6 μmol/L can promote the apoptosis of liver cancer to different degrees (P<0.01). CONCLUSION The present study revealed the mechanism of PPI against liver cancer through network pharmacology and in vitro experiments. Its mechanism of action is related to the inhibition of PPI on the proliferation of HBV-related liver cancer through promoting the apoptosis of liver cancer cells. Additionally, in vitro experiments have also verified that PPI can promote the apoptosis of HepG2 and HepG2.2.15 cells.
Collapse
Affiliation(s)
- Shuxian Yu
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Wenhui Gao
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Puhua Zeng
- Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha 410006, China
| | - Chenglong Chen
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Zhuo Liu
- Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha 410006, China
| | - Zhen Zhang
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Jiyong Liu
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| |
Collapse
|
21
|
Abstract
Fibroblast activation protein-α (FAP) is a type-II transmembrane serine protease expressed almost exclusively to pathological conditions including fibrosis, arthritis, and cancer. Across most cancer types, elevated FAP is associated with worse clinical outcomes. Despite the clear association between FAP and disease severity, the biological reasons underlying these clinical observations remain unclear. Here we review basic FAP biology and FAP's role in non-oncologic and oncologic disease. We further explore how FAP may worsen clinical outcomes via its effects on extracellular matrix remodeling, intracellular signaling regulation, angiogenesis, epithelial-to-mesenchymal transition, and immunosuppression. Lastly, we discuss the potential to exploit FAP biology to improve clinical outcomes.
Collapse
Affiliation(s)
- Allison A Fitzgerald
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3870 Reservoir Road NW, Washington, DC, 20057, USA
| | - Louis M Weiner
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3870 Reservoir Road NW, Washington, DC, 20057, USA.
| |
Collapse
|
22
|
Li HS, Xu Y. Inhibition of EZH2 via the STAT3/HOTAIR signalling axis contributes to cell cycle arrest and apoptosis induced by polyphyllin I in human non-small cell lung cancer cells. Steroids 2020; 164:108729. [PMID: 32941921 DOI: 10.1016/j.steroids.2020.108729] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 08/20/2020] [Accepted: 09/06/2020] [Indexed: 02/08/2023]
Abstract
OBJECTIVE To explore the potential mechanism of polyphyllin I (PPI)-induced apoptosis in lung cancer cells. METHODS AND MATERIALS The pathological changes in lung cancer tissues and paracancerous tissues were first analysed by H&E staining and IHC staining. After PPI treatment, cell viability and apoptosis were detected by MTT assays, cell cycle analyses and flow cytometry. The expression levels of EZH2 and apoptosis-related molecules were evaluated by qRT-PCR and Western blotting. RESULTS EZH2 overexpression decreased proapoptotic proteins, and this effect was reversed by PPI. Knockdown of HOTAIR downregulated EZH2 expression, upregulated proapoptotic proteins, and enhanced the effect of PPI treatment. Moreover, knockdown of STAT3 could counteract the effect of HOTAIR overexpression, which significantly increased the expression of EZH2, thus facilitating cell apoptosis in lung cancer. CONCLUSIONS PPI induced cell cycle arrest and apoptosis in lung cancer by inhibiting EZH2 through the STAT3/HOTAIR signalling pathway.
Collapse
Affiliation(s)
- Hok Shing Li
- R&D Department of AtaGenix Laboratories Co., Ltd. (Wuhan), Wuhan 430000, Hubei Province, PR China
| | - Yao Xu
- R&D Department of AtaGenix Laboratories Co., Ltd. (Wuhan), Wuhan 430000, Hubei Province, PR China.
| |
Collapse
|
23
|
Song C, Pan B, Yang X, Tang W. Polyphyllin VII suppresses cell proliferation, the cell cycle and cell migration in colorectal cancer. Oncol Lett 2020; 21:25. [PMID: 33240431 PMCID: PMC7681227 DOI: 10.3892/ol.2020.12286] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 08/18/2020] [Indexed: 12/13/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common types of human cancer. However, there is still an urgent need to identify novel treatment strategies for CRC. The present study aimed to validate the potential antitumor effects of polyphyllin VII in CRC. The present study revealed that polyphyllin VII could significantly inhibit CRC proliferation and induce cell cycle arrest and apoptosis. Moreover, the anti-metastatic effect of polyphyllin VII in CRC cells was implicated. Microarray analysis identified that polyphyllin VII could affect multiple protein coding genes and non-coding RNAs. Bioinformatics analysis revealed that polyphyllin VII regulated multiple pathways in CRC, including ‘ER to Golgi vesicle-mediated transport’, ‘response to cAMP’, ‘Ras protein signal transduction’, ‘metabolic pathways’, ‘MAPK signaling pathway’ and ‘cell cycle’. Protein-Protein Interaction network analysis identified a series of key polyphyllin VII-regulating genes in CRC, including ribonucleoside-diphosphate reductase subunit M2, structural maintenance of chromosomes protein 4 and DNA replication licensing factor MCM4. Finally, the present results demonstrated that these key polyphyllin VII-regulating genes were dysregulated in CRC. Taken together, these results indicated that polyphyllin VII could be a novel antitumor drug for the treatment of CRC.
Collapse
Affiliation(s)
- Cheng Song
- Department of Chinese Medicine, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, P.R. China
| | - Bo Pan
- First Department of Oncology, Hunan Academy of Traditional Chinese Medicine Affiliated Hospital, Changsha, Hunan 410013, P.R. China
| | - Xiao Yang
- Department of Oncology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410013, P.R. China
| | - Wei Tang
- First Department of Oncology, Hunan Academy of Traditional Chinese Medicine Affiliated Hospital, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
24
|
Zhang J, Gu C, Song Q, Zhu M, Xu Y, Xiao M, Zheng W. Identifying cancer-associated fibroblasts as emerging targets for hepatocellular carcinoma. Cell Biosci 2020; 10:127. [PMID: 33292459 PMCID: PMC7603733 DOI: 10.1186/s13578-020-00488-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 10/23/2020] [Indexed: 02/07/2023] Open
Abstract
The tumor microenvironment (TME) is a complex multicellular functional compartment that includes fibroblasts, myofibroblasts, endothelial cells, immune cells, and extracellular matrix (ECM) elements. The microenvironment provides an optimum condition for the initiation, growth, and dissemination of hepatocellular carcinoma (HCC). As one of the critical and abundant components in tumor microenvironment, cancer-associated fibroblasts (CAFs) have been implicated in the progression of HCC. Through secreting various growth factors and cytokines, CAFs contribute to the ECM remodeling, stem features, angiogenesis, immunosuppression, and vasculogenic mimicry (VM), which reinforce the initiation and development of HCC. In order to restrain the CAFs-initiated HCC progression, current strategies include targeting specific markers, engineering CAFs with tumor-suppressive phenotype, depleting CAFs’ precursors, and repressing the secretions or downstream signaling. In this review, we update the emerging understanding of CAFs in HCC, with particular emphasis on cellular origin, phenotypes, biological functions and targeted strategies. It provides insights into the targeting CAFs for HCC treatment.
Collapse
Affiliation(s)
- Jie Zhang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, Jiangsu, China
| | - Chaoyu Gu
- School of Medicine, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Qianqian Song
- Department of Radiology, Wake Forest School of Medicine, One Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Mengqi Zhu
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, Jiangsu, China
| | - Yuqing Xu
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, Jiangsu, China
| | - Mingbing Xiao
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, Jiangsu, China.
| | - Wenjie Zheng
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
25
|
Li M, Cheng X, Rong R, Gao Y, Tang X, Chen Y. High expression of fibroblast activation protein (FAP) predicts poor outcome in high-grade serous ovarian cancer. BMC Cancer 2020; 20:1032. [PMID: 33109151 PMCID: PMC7590670 DOI: 10.1186/s12885-020-07541-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 10/19/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND High-grade serous ovarian cancer (HGSOC) is a fatal form of ovarian cancer. Previous studies indicated some potential biomarkers for clinical evaluation of HGSOC prognosis. However, there is a lack of systematic analysis of different expression genes (DEGs) to screen and detect significant biomarkers of HGSOC. METHODS TCGA database was conducted to analyze relevant genes expression in HGSOC. Outcomes of candidate genes expression, including overall survival (OS) and progression-free survival (PFS), were calculated by Cox regression analysis for hazard rates (HR). Histopathological investigation of the identified genes was carried out in 151 Chinese HGSOC patients to validate gene expression in different stages of HGSOC. RESULTS Of all 57,331 genes that were analyzed, FAP was identified as the only novel gene that significantly contributed to both OS and PFS of HGSOC. In addition, FAP had a consistent expression profile between carcinoma-paracarcinoma and early-advanced stages of HGSOC. Immunological tests in paraffin section also confirmed that up-regulation of FAP was present in advanced stage HGSOC patients. Prediction of FAP network association suggested that FN1 could be a potential downstream gene which further influenced HGSOC survival. CONCLUSIONS High-level expression of FAP was associated with poor prognosis of HGSOC via FN1 pathway.
Collapse
Affiliation(s)
- Min Li
- Department of Gynecology & Obstetrics, the First Affiliated Hospital of Soochow University, No.188, Shizi Street, Suzhou, Jiangsu Province, 215006, China
| | - Xue Cheng
- Department of Pathology, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Rong Rong
- Department of Pathology, the First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Yan Gao
- Institute of Suzhou Biobank, Suzhou Center for Disease Prevention and Control, Suzhou, 215004, China.,School of Public Health, Medical College of Soochow University, Suzhou, 215123, China
| | - Xiuwu Tang
- Department of Gynecology & Obstetrics, the First Affiliated Hospital of Soochow University, No.188, Shizi Street, Suzhou, Jiangsu Province, 215006, China.
| | - Youguo Chen
- Department of Gynecology & Obstetrics, the First Affiliated Hospital of Soochow University, No.188, Shizi Street, Suzhou, Jiangsu Province, 215006, China.
| |
Collapse
|
26
|
Alternative Options for Skin Cancer Therapy via Regulation of AKT and Related Signaling Pathways. Int J Mol Sci 2020; 21:ijms21186869. [PMID: 32962182 PMCID: PMC7560163 DOI: 10.3390/ijms21186869] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/11/2020] [Accepted: 09/17/2020] [Indexed: 12/25/2022] Open
Abstract
Global environmental pollution has led to human exposure to ultraviolet (UV) radiation due to the damaged ozone layer, thereby increasing the incidence and death rate of skin cancer including both melanoma and non-melanoma. Overexpression and activation of V-akt murine thymoma viral oncogene homolog (AKT, also known as protein kinase B) and related signaling pathways are major factors contributing to many cancers including lung cancer, esophageal squamous cell carcinoma and skin cancer. Although BRAF inhibitors are used to treat melanoma, further options are needed due to treatment resistance and poor efficacy. Depletion of AKT expression and activation, and related signaling cascades by its inhibitors, decreases the growth of skin cancer and metastasis. Here we have focused the effects of AKT and related signaling (PI3K/AKT/mTOR) pathways by regulators derived from plants and suggest the need for efficient treatment in skin cancer therapy.
Collapse
|
27
|
Huang R, Shu J, Dai X, Liu Y, Yu F, Shi G. The protective effect of polyphyllin I on myocardial ischemia/reperfusion injury in rats. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:644. [PMID: 32566581 PMCID: PMC7290651 DOI: 10.21037/atm-20-3371] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Myocardial ischemia/reperfusion (I/R) injury has become a global public health concern. An increasing amount of evidence has shown that polyphyllin I (PPI) has anti-apoptotic and antioxidant functions. This study was performed to evaluate the cardioprotective effects of PPI in a rat model of myocardial I/R injury and the underlying mechanism. Methods We exposed induced a rat model of I/R injury by exposing rat hearts to left anterior descending coronary artery ligation for 30 min, followed by 24 h of reperfusion. Cardiac function was analyzed by echocardiography and HE staining. Myocardial apoptosis, inflammation, and oxidative stress were detected to analyze the PPI's role in I/R injury. Results The results showed that pretreatment with PPI improved impaired histological morphology, as shown by histopathological examination. Echocardiography analysis showed that PPI increased the levels of HR, left ventricular ejection fraction (LVEF), and left ventricular wall thickness (LVWT), accompanied by decreased left ventricular end-systolic volume (LVESV). Also, PPI decreased the expression of CK-MB, Mb, cTnI, and LDH. Specifically, PPI also changed the expression of apoptotic makers (Caspase-3, Bax, and Bcl-2), inflammatory cytokines (TNF-α, IL-6, iNOS, and IL-10) and oxidative stress markers (SOD, GSH, ROS, and MDA). Notably, western blot (WB) showed that PPI treatment inhibited the phosphorylation activity of NF-κB p65. Conclusions The findings showed that PPI exerted a favorable protective effect on I/R injury by inhibiting the inflammatory response and oxidative stress. It offered new drug candidates for the treatment of myocardial I/R injury.
Collapse
Affiliation(s)
- Ruizhen Huang
- Department of Cardiovascular, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Jia Shu
- Functional Inspection Division, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Xiaoqin Dai
- Department of Traditional Chinese Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu 610072, China
| | - Yanru Liu
- Department of Cardiovascular, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Fang Yu
- Functional Inspection Division, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Gang Shi
- Department of Pharmacy Service, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| |
Collapse
|
28
|
Feng FF, Cheng P, Sun C, Wang H, Wang W. Inhibitory effects of polyphyllins I and VII on human cisplatin-resistant NSCLC via p53 upregulation and CIP2A/AKT/mTOR signaling axis inhibition. Chin J Nat Med 2020; 17:768-777. [PMID: 31703757 DOI: 10.1016/s1875-5364(19)30093-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Indexed: 01/28/2023]
Abstract
Cancerous inhibitor of protein phosphatase 2A (CIP2A) is a human oncoprotein that is overexpressed in multiple kinds of cancers including non-small cell lung cancer (NSCLC). CIP2A plays an 'oncogenic nexus' to participate in the tumorigenesis and chemoresistance in several cancer types. AKT and mTORC1 overactivation are detected in NSCLC and many other cancers. Previous studies found that the CIP2A/AKT/mTOR pathway controls cell growth, apoptosis, autophagy process. Polyphyllin I (PPI) and polyphyllin VII (PPVII) are natural components extracted from Paris polyphylla that display anti-cancer properties. In the present study, we investigated whether PPI and PPVII can be used in the cisplatin (DDP)-resistant human NSCLC cell line A549/DDP. Results demonstrated that PPI and PPVII treatment significantly suppressed A549/DDP cell proliferation, migration, invasion and EMT, induced apoptosis and autophagy. Further examination of the mechanism revealed that the PPI and PPVII significantly upregulated the p53, induced caspase-dependent apoptosis and suppressed the CIP2A/AKT/mTOR pathway. The activation of autophagy was mediated through PPI and PPVII induced inhibition of mTOR. We propose that PPI and PPVII might be developed as candidate drugs for DDP-resistant NSCLC.
Collapse
Affiliation(s)
- Fei-Fei Feng
- Department of Respiratory Medicine, The Second Hospital of Shandong University, Jinan 250033, China
| | - Peng Cheng
- Department of Neural Medicine, The Second Hospital of Shandong University, Jinan 250033, China
| | - Chao Sun
- Department of Central Laboratory, The Second Hospital of Shandong University, Jinan 250033, China
| | - Hui Wang
- Department of Respiratory Medicine, The Second Hospital of Shandong University, Jinan 250033, China
| | - Wei Wang
- Department of Respiratory Medicine, The Second Hospital of Shandong University, Jinan 250033, China.
| |
Collapse
|
29
|
Wu Y, Si Y, Xiang Y, Zhou T, Liu X, Wu M, Li W, Zhang T, Xiang K, Zhang L, Zhao H, Liu Y. Polyphyllin I activates AMPK to suppress the growth of non-small-cell lung cancer via induction of autophagy. Arch Biochem Biophys 2020; 687:108285. [PMID: 32074500 DOI: 10.1016/j.abb.2020.108285] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 01/07/2020] [Accepted: 01/29/2020] [Indexed: 12/14/2022]
Abstract
Polyphyllin I (PPI), a bioactive constituent extracted from the rhizomes of Paris polyphylla, is cytotoxic to several cancer types. This study was designed to explore whether PPI prevents non-small-cell lung cancer (NSCLC) growth and to investigate the molecular mechanism. AMP-activated protein kinase (AMPK) has been implicated in the activation of autophagy in distinct tissues. In cultured human NSCLC cell lines, PPI induces autophagy by activating AMPK and then inhibiting mTOR signaling in a concentration-dependent manner. Furthermore, the activation of autophagy induced by PPI was reversed by the AMPK inhibitor compound C. Computational docking showed that PPI directly interacted with the allosteric drug and metabolite site of AMPK to stabilize its activation. Microscale thermophoresis and Drug Affinity Responsive Targeting Stability (DARTS) assay further confirmed the high affinity between PPI and AMPK. In vivo studies indicated that PPI suppressed the growth of NSCLC and increased the levels of LC3-II and phosphorylated AMPK in tumors isolated from a xenograft model of NSCLC in mice. Moreover, PPI exhibited favorable pharmacokinetics in rats. In summary, PPI conclusively acts as a direct AMPK activator to induce cell autophagy which inhibits the growth of NSCLC cells. In the future, PPI therapy should be applied to treat patients with NSCLC.
Collapse
Affiliation(s)
- Yezi Wu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yuan Si
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yuchen Xiang
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China
| | - Tong Zhou
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xuewen Liu
- Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China
| | - Mingwei Wu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China
| | - Wenjuan Li
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research and Institute of Medicinal Chemistry, Hubei University of Medicine, Shiyan, Hubei, China
| | - Te Zhang
- Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research and Institute of Medicinal Chemistry, Hubei University of Medicine, Shiyan, Hubei, China
| | - Ke Xiang
- Department of Science and Education, Gucheng People's Hospital, Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Liang Zhang
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China
| | - Huzi Zhao
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China
| | - Ying Liu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research and Institute of Medicinal Chemistry, Hubei University of Medicine, Shiyan, Hubei, China.
| |
Collapse
|
30
|
Tian Y, Gong GY, Ma LL, Wang ZQ, Song D, Fang MY. Anti-cancer effects of Polyphyllin I: An update in 5 years. Chem Biol Interact 2020; 316:108936. [DOI: 10.1016/j.cbi.2019.108936] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 11/17/2019] [Accepted: 12/19/2019] [Indexed: 02/06/2023]
|
31
|
Feng F, Cheng P, Wang C, Wang Y, Wang W. Polyphyllin I and VII potentiate the chemosensitivity of A549/DDP cells to cisplatin by enhancing apoptosis, reversing EMT and suppressing the CIP2A/AKT/mTOR signaling axis. Oncol Lett 2019; 18:5428-5436. [PMID: 31612051 PMCID: PMC6781722 DOI: 10.3892/ol.2019.10895] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 08/14/2019] [Indexed: 11/23/2022] Open
Abstract
Poor response and resistance to cisplatin (DDP)-based chemotherapy frequently leads to treatment failure in advanced non-small cell lung cancer (NSCLC). The underlying molecular mechanism is extremely complex and currently remains unclear. The overexpression of cancerous inhibitor of protein phosphatase 2A (CIP2A) indicates poor prognosis and promotes the epithelial-to-mesenchymal transition (EMT) and metastasis. The EMT has been reported to promote drug resistance in numerous previous studies. CIP2A and its downstream protein kinase B (AKT)/mammalian target of rapamycin (mTOR) pathway also plays a role in mediating DDP resistance. Polyphyllin I (PPI) and polyphyllin VII (PPVII) are natural components extracted from Paris polyphylla that display anti-cancer properties. In the present study, the chemosensitizing effects of PPI and PPVII were investigated in the DDP-resistant NSCLC cell line A549/DDP, as well as the underlying molecular mechanisms. The results demonstrated that PPI and PPVII could significantly inhibit cell proliferation and enhance the sensitivities of A549/DDP cells to DDP. When assessing the underlying molecular mechanism, it was revealed that PPI and PPVII enhanced DDP-induced apoptosis in A549/DDP cells via p53 upregulation and the caspase-dependent pathway. Furthermore, PPI and PPVII reversed the EMT and suppressed CIP2A and its downstream AKT/mTOR signaling cascade in A549/DDP cells. Overall, the results from the present study demonstrated that PPI and PPVII may function as chemosensitizers by enhancing apoptosis via the p53 pathway, reversing EMT and suppressing the CIP2A/AKT/mTOR signaling axis, and the combination with DDP may be a promising strategy for the development of new therapeutic agents.
Collapse
Affiliation(s)
- Feifei Feng
- Department of Respiratory Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Peng Cheng
- Department of Neural Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Chaochao Wang
- Department of Respiratory Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Yongbin Wang
- Department of Respiratory Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Wei Wang
- Department of Respiratory Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| |
Collapse
|
32
|
He J, Yu S, Guo C, Tan L, Song X, Wang M, Wu J, Long Y, Gong D, Zhang R, Cao Z, Li Y, Peng C. Polyphyllin I induces autophagy and cell cycle arrest via inhibiting PDK1/Akt/mTOR signal and downregulating cyclin B1 in human gastric carcinoma HGC-27 cells. Biomed Pharmacother 2019; 117:109189. [PMID: 31387191 DOI: 10.1016/j.biopha.2019.109189] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 06/17/2019] [Accepted: 06/28/2019] [Indexed: 12/25/2022] Open
Abstract
Paris polyphylla. is a traditional medicinal herb that has long been used to prevent cancer in many Asian countries. Polyphyllin I (PPI), an important bioactive constituent of Paris polyphylla, has been found to exhibit a wide variety of anticancer activities in many types of cancer cells. However, the effects of PPI on human gastric carcinoma cells and its mechanism of action remain unclear. In this study, we examined the effective anti-gastric carcinoma activity of PPI and its underlying mechanism of action in HGC-27 cells. In vitro, sub-micromolar concentrations of PPI inhibited HGC-27 cell proliferation with an IC50 of 0.34 ± 0.06 μM after a 72-h treatment. In vivo, 3 mg/kg PPI significantly inhibited proliferation of HGC-27 tumor cells, with a 78.8% inhibition rate compared to paclitaxel, and demonstrated higher safety. Analysis of MDC and mGFP-LC3 fluorescence, Western blotting and flow cytometry indicated that PPI induced cell cycle arrest in HGC-27 cells by promoting the conversion of LC3-I to LC3-II and by downregulating cyclin B1. Furthermore, Western blotting showed that PPI inhibited the autophagy-regulating PDK1/Akt/mTOR signaling pathway in vitro and in vivo. In addition, immunohistochemistry and TUNEL staining revealed that PPI decreased Ki67 expression and increased the percentage of apoptotic cells in HGC-27 xenograft tumors. These data indicate that PPI is an PDK1/Akt/mTOR signaling inhibitor and of therapeutic relevance for gastric cancer treatment and that the rhizome of Paris polyphylla deserves further clinical investigation as an alternative therapy for gastric cancer.
Collapse
Affiliation(s)
- Junlin He
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, China
| | - Si Yu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, China; Chengdu Medical College, Chengdu 610500, China
| | - Chuanjie Guo
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, China
| | - Lu Tan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, China
| | - Xiaominting Song
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, China
| | - Miao Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, China
| | - Jing Wu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, China
| | - Yuling Long
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, China
| | - Daoyin Gong
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, China
| | - Ruoqi Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, China
| | - Zhixing Cao
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, China
| | - Yuzhi Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, China.
| | - Cheng Peng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, China.
| |
Collapse
|
33
|
Ham IH, Lee D, Hur H. Role of Cancer-Associated Fibroblast in Gastric Cancer Progression and Resistance to Treatments. JOURNAL OF ONCOLOGY 2019; 2019:6270784. [PMID: 31281359 PMCID: PMC6590541 DOI: 10.1155/2019/6270784] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/23/2019] [Indexed: 12/21/2022]
Abstract
Although the survival of gastric cancer (GC) patients has gradually improved, the outcomes of advanced GC patients remain unsatisfactory despite standard treatment with conventional chemotherapy or targeted agents. Several studies have shown that cancer-associated fibroblasts (CAFs), a major component of tumor stroma in GC, may have significant roles in GC progression and resistance to treatments. CAFs are a major source of various secreted molecules in the tumor microenvironment, which stimulate cancer cells and other noncancerous components of GC. Surprisingly, these factors could be involved in gastric carcinogenesis. Cytokines, including interleukin-6 and interleukin-11, or growth factors, such as fibroblast growth factor produced from CAFs, can directly activate GC cells and consequently lead to the development of an aggressive phenotype. Galectin-1 or hepatocyte growth factor can be involved in CAF-derived neovascularization in GC. In addition, recent studies showed that CAFs can affect tumor immunity through M2 polarization of tumor-associated macrophages. Finally, the current study aimed to introduce several inhibitory agents and evaluate their suppressive effects on CAFs in patients with GC progression. However, further studies are required to evaluate their safety and select appropriate patients for application in clinical settings.
Collapse
Affiliation(s)
- In-Hye Ham
- Department of Surgery, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Dagyeong Lee
- Department of Surgery, Ajou University School of Medicine, Suwon, Republic of Korea
- Brain Korea 21 Plus Research Center for Biomedical Sciences, Ajou University, Suwon, Republic of Korea
- Department of Biomedical Science, Graduated School of Ajou University, Suwon, Republic of Korea
| | - Hoon Hur
- Department of Surgery, Ajou University School of Medicine, Suwon, Republic of Korea
- Brain Korea 21 Plus Research Center for Biomedical Sciences, Ajou University, Suwon, Republic of Korea
- Department of Biomedical Science, Graduated School of Ajou University, Suwon, Republic of Korea
| |
Collapse
|
34
|
Abstract
Propionibacterium acnes (P. acnes) has been implicated in the progression of acne inflammation. Because current acne medications have various side effects, it is necessary to explore alternative medications possessing anti-inflammatory activity against P. acnes. We investigated the inhibitory effects of polyphyllin I (PPI) on P. acnes-induced inflammation in vitro. In this study, we examined the effects of PPI on the production of inflammatory cytokines in HaCaT keratinocytes treated with heat-killed P. acnes. These treated HaCaT keratinocytes showed increased expression of Toll-like receptor 2 (TLR2) and production of inflammatory cytokines. PPI significantly suppressed the secretion of inflammatory cytokines, including interleukin (IL)-6, IL-8, and tumor necrosis factor (TNF)-α, and the expression of TLR2 in P. acnes-treated cells. Moreover, we studied the influence of PPI on the nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways in P. acnes-treated keratinocytes. PPI diminished the activation of NF-κB. Phosphorylated p38 levels were markedly increased after treatment with heat-killed P. acnes but were decreased after treatment with PPI, while the effect of PPI on ERK phosphorylation was not significant. Heat-killed P. acnes and PPI did not have any effect on JNK phosphorylation. Furthermore, we confirmed that NF-κB p65 inhibitor (BAY11-7082), p38 MAPK inhibitor (SB203580), and PPI blocked the expression of IL-8 in heat-killed P. acnes-treated cells. These results demonstrated that PPI has potential for development as a treatment for acne inflammation.
Collapse
|
35
|
Chen Y, Liu F, Wu K, Wu W, Wu H, Zhang W. Targeting dipeptidyl peptidase 8 genes inhibits proliferation, migration and invasion by inhibition of cyclin D1 and MMP2MMP9 signal pathway in cervical cancer. J Gene Med 2018; 20:e3056. [PMID: 30225951 DOI: 10.1002/jgm.3056] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 08/19/2018] [Accepted: 09/12/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND DPP8 is a member of the dipeptidyl peptidase IV family, which belongs to the S9b protease subfamily. It regulates cell proliferation, apoptosis, migration and invasion during cancer progression. METHODS To investigate the role of DPP8 in cervical cancer, we examined DPP8 levels in cervical cancer tissues and cells. The localization of DPP8 was determined by immunofluorescence staining. Subsequently, SiHa and HeLa cells were treated with small interfering RNA (siRNA)-DPP8. We used cell cycle analysis, an 5-ethyl-2'-deoxyuridine assay proliferation assay and a cellular apoptosis assay to determine the effect of DPP8 on the proliferation and apoptosis of cervical cancer cells. We used a Transwell assay to assess the number of transfection cancer cells migrating through the matrix. A real-time polymerase chain reaction and western blot analysis were used to analyze the expression of related proteins and to determine the phenotype caused by the depletion or overexpression of DPP8 in cervical cancer cells. RESULTS We observed that DPP8 was highly expressed in cervical cancer tissues and cells. DPP8 expression was observed in the cytosol and in the perinuclear area, as well as in the nuclei of cervical cancer cells. Notably, when cells were treated with siRNA-DPP8, the expression of BAX increased, and the expression of cyclin D1, Bcl-2, MMP2 and MMP9 was downregulated. In cervical cancer cell lines, silencing the expression of DPP8 not only suppressed the proliferation, migration and invasion of the cervical cancer cells, but also promoted cervical cancer cell apoptosis. CONCLUSIONS The data obtained in the present study reveal that DPP8 promotes the progression of cervical cancer.
Collapse
Affiliation(s)
- Yurou Chen
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Fulin Liu
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Kejia Wu
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Wanrong Wu
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Hanshu Wu
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Wei Zhang
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| |
Collapse
|
36
|
Chai Y, Si Y, Xu J, Xiang Y, Zhao H, Si Y, Zhang T, Liu Y. Polyphyllin I Inhibits Proliferation and Induces Apoptosis by Downregulating AML1-ETO and Suppressing C-KIT/Akt Signaling in t(8;21) Acute Myeloid Leukemia. Chem Biodivers 2018; 15:e1800314. [PMID: 30194712 DOI: 10.1002/cbdv.201800314] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 09/03/2018] [Indexed: 11/08/2022]
Abstract
Polyphyllin I (PPI), a bioactive constituent extracted from traditional medicinal herbs, is cytotoxic to several cancer types. However, whether PPI can be used to treat t(8;21) acute myeloid leukemia (AML) cells requires further investigation. Here, we determined the inhibitory effects of PPI on t(8;21) AML cells by Cell Counting Kit-8 (CCK-8) and the trypan blue dye exclusion assay. DAPI staining and Wright-Giemsa staining were performed to check for apoptosis. Detection of apoptotic protein and AML1-ETO signaling protein expression were conducted by Western blot analysis. Our results suggested that PPI decreased growth and induced apoptosis in a dosage-dependent manner in the t(8;21) AML cell line Kasumi-1. PPI significantly downregulated AML1-ETO expression in a dosage- and time-dependent manner. PPI also upregulated P21 and downregulated survivin expression by reducing AML1-ETO. Mechanistically, PPI significantly reduced the expression of C-KIT, another therapeutic target for AML with t(8;21), followed by inhibition of Akt signaling. These results suggest that PPI can suppress growth and induce apoptosis of t(8;21) AML by suppressing the AML1-ETO and C-KIT/Akt signaling pathways. Therefore, PPI may be an anticancer therapeutic to treat t(8;21) AML.
Collapse
MESH Headings
- Antineoplastic Agents, Phytogenic/chemistry
- Antineoplastic Agents, Phytogenic/pharmacology
- Apoptosis/drug effects
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Chromosomes, Human, Pair 21/genetics
- Chromosomes, Human, Pair 8/genetics
- Core Binding Factor Alpha 2 Subunit/antagonists & inhibitors
- Core Binding Factor Alpha 2 Subunit/metabolism
- Diosgenin/analogs & derivatives
- Diosgenin/chemistry
- Diosgenin/pharmacology
- Dose-Response Relationship, Drug
- Down-Regulation/drug effects
- Drug Screening Assays, Antitumor
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Medicine, Chinese Traditional
- Molecular Conformation
- Proto-Oncogene Proteins c-akt/antagonists & inhibitors
- Proto-Oncogene Proteins c-akt/metabolism
- Proto-Oncogene Proteins c-kit/antagonists & inhibitors
- Proto-Oncogene Proteins c-kit/metabolism
- RUNX1 Translocation Partner 1 Protein/antagonists & inhibitors
- RUNX1 Translocation Partner 1 Protein/metabolism
- Signal Transduction/drug effects
- Structure-Activity Relationship
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Yanting Chai
- Child Health Center, Shiyan Maternal and Child Health Hospital, Shiyan, 442000, P. R. China
| | - Ying Si
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, 30 Renmin Road, Shiyan, 442000, P. R. China
- Department of Rehabilitation Medicine, Gucheng People's Hospital, Hubei University of Arts and Science, Xiangyang, 441700, P. R. China
| | - Jiaxin Xu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, 30 Renmin Road, Shiyan, 442000, P. R. China
| | - Yuchen Xiang
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, 30 Renmin Road, Shiyan, 442000, P. R. China
| | - Hongyan Zhao
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, 30 Renmin Road, Shiyan, 442000, P. R. China
| | - Yuan Si
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, 30 Renmin Road, Shiyan, 442000, P. R. China
| | - Te Zhang
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, 30 Renmin Road, Shiyan, 442000, P. R. China
| | - Ying Liu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, 30 Renmin Road, Shiyan, 442000, P. R. China
| |
Collapse
|