1
|
Wang Y, Wu Y, Liu B, Yang H, Qian H, Cheng Y, Li X, Yang G, Zheng X, Shen F. Binding domain peptide ameliorates alveolar hypercoagulation and fibrinolytic inhibition in mice with lipopolysaccharide-induced acute respiratory distress syndrome Via NF-κB signaling pathway. Am J Transl Res 2022; 14:3854-3863. [PMID: 35836863 PMCID: PMC9274609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/07/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Alveolar hypercoagulation and fibrinolytic inhibition are shown to be associated with refractory hypoxemia in acute respiratory distress syndrome (ARDS), and the NF-κB pathway is involved in this process. The purpose of this study is to explore the role of NEMO-binding domain peptide (NBDP) in alleviating alveolar hypercoagulation and fibrinolytic inhibition induced by lipopolysaccharide (LPS) in ARDS mice and its related mechanisms. MATERIALS AND METHODS ARDS was induced by inhalation of LPS (mg/L) in adult male BALB/c mice. Mice were treated with intratracheal inhalation of NBDP or saline aerosol at increased concentrations 30 minutes before LPS administration. Six hours after LPS treatment, bronchoalveolar lavage fluids (BALF) were collected and then all mice were euthanized. In addition, coagulation and fibrinolysis associated factors in lung tissues and BALF were detected, and the activation of NF-κB signaling pathway was observed. RESULTS NBDP pretreatment dose-dependently inhibited the expression of tissue factor (TF) and plasminogen activator inhibitor (PAI) 1 in lung tissues, reduced the secretions of TF, PAI-1, thrombin-antithrombin (TAT) complex, and promoted activated protein C (APC) secretion in BALF induced by LPS. LPS-induced high expression of pulmonary procollagen peptide type lll (PIIIP) was also reduced in a dose-dependent manner under NBDP pretreatment. Western blotting showed that NBDP pretreatment significantly attenuated LPS-induced activation of IKKα/β, Iκα and NF-κB p65. NBDP pretreatment also inhibited the DNA binding activity of p65 induced by LPS. We also noticed that NBDP protected mice against LPS-induced lung injury in a dose-dependent manner. CONCLUSIONS The experimental findings demonstrate that through inhibiting the NF-κB signaling pathway, NBDP dose-dependently ameliorates LPS-induced alveolar hypercoagulation and fibrinolytic inhibition, which is expected to be a new therapeutic target to correct the abnormalities of alveolar coagulation and fibrinolytic pathways in ARDS.
Collapse
Affiliation(s)
- Yahui Wang
- Department of Intensive Care Unit, Guizhou Medical University Affiliated HospitalGuiyang 550001, Guizhou, China
- Department of Intensive Care Unit, The People’s Hospital of Weining CountyWeining County 553100, Guizhou, China
| | - Yanqi Wu
- Department of Intensive Care Unit, Guizhou Medical University Affiliated HospitalGuiyang 550001, Guizhou, China
| | - Bo Liu
- Department of Intensive Care Unit, Guizhou Medical University Affiliated HospitalGuiyang 550001, Guizhou, China
| | - Huilin Yang
- Department of Intensive Care Unit, Guizhou Medical University Affiliated HospitalGuiyang 550001, Guizhou, China
| | - Hong Qian
- Department of Intensive Care Unit, Guizhou Medical University Affiliated HospitalGuiyang 550001, Guizhou, China
| | - Yumei Cheng
- Department of Intensive Care Unit, Guizhou Medical University Affiliated HospitalGuiyang 550001, Guizhou, China
| | - Xiang Li
- Department of Intensive Care Unit, Guizhou Medical University Affiliated HospitalGuiyang 550001, Guizhou, China
| | - Guixia Yang
- Department of Intensive Care Unit, Guizhou Medical University Affiliated HospitalGuiyang 550001, Guizhou, China
| | - Xinghao Zheng
- Department of Intensive Care Unit, Guizhou Medical University Affiliated HospitalGuiyang 550001, Guizhou, China
| | - Feng Shen
- Department of Intensive Care Unit, Guizhou Medical University Affiliated HospitalGuiyang 550001, Guizhou, China
| |
Collapse
|
2
|
Xu B, Dan W, Zhang X, Wang H, Cao L, Li S, Li J. Gene Differential Expression and Interaction Networks Illustrate the Biomarkers and Molecular Biological Mechanisms of Unsaponifiable Matter in Kanglaite Injection for Pancreatic Ductal Adenocarcinoma. BIOMED RESEARCH INTERNATIONAL 2022; 2022:6229462. [PMID: 35707377 PMCID: PMC9192213 DOI: 10.1155/2022/6229462] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 05/13/2022] [Indexed: 12/12/2022]
Abstract
Background Kanglaite injection (KLTi) has shown good clinical efficacy in the treatment of pancreatic ductal adenocarcinoma (PDAC). While previous studies have demonstrated the antitumor effects of the oil compounds in KLTi, it is unclear whether the unsaponifiable matter (USM) also has antitumor effects. This study used network pharmacology, molecular docking, and database verification methods to investigate the molecular biological mechanisms of USM. Methods Compounds of USM were obtained from GC-MS, and targets from DrugBank. Next, the GEO database was searched for differentially expressed genes in cancerous tissues and healthy tissues of PDAC to identify targets. Subsequently, the protein-protein interaction of USM and PDAC targets was constructed by BisoGenet to extract candidate genes. The candidate genes were enriched using GO and KEGG by Metascape, and the gene-pathway network was constructed to screen the key genes. Molecular docking and molecular dynamic simulations of core compound targets were finally performed and to explore the diagnostic, survival, and prognosis value of targets. Results A total of 10 active compounds and 36 drug targets were screened for USM, 919 genes associated with PDAC, and 139 USM candidate genes against PDAC were excavated. The enrichment predicted USM by acting on RELA, NFKB1, IKBKG, JUN, MAPK1, TP53, and AKT1. Molecular docking and dynamic simulations confirmed the screened core targets had good affinity and stability with the corresponding compounds. In diagnostic ROC validation, the above targets have certain accuracy for diagnosing PDAC, and the combined diagnosis is more advantageous. As the most diagnostic value of RELA, it is equally significant in predicting disease-specific survival and progression-free interval. Conclusions USM in KLTi plays an anti-PDAC role by intervening in the cell cycle, inducing apoptosis, and downregulating the NF-κB, MAPK, and PI3K-Akt pathways. It might participate in the pancreatic cancer pathway, and core target groups have diagnostic, survival, and prognosis value biomarker significance.
Collapse
Affiliation(s)
- Bowen Xu
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wenchao Dan
- Department of Dermatological, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Xiaoxiao Zhang
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Heping Wang
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Luchang Cao
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Shixin Li
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jie Li
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| |
Collapse
|
3
|
Napoleon JV, Sagar S, Kubica SP, Boghean L, Kour S, King HM, Sonawane YA, Crawford AJ, Gautam N, Kizhake S, Bialk PA, Kmiec E, Mallareddy JR, Patil PP, Rana S, Singh S, Prahlad J, Grandgenett PM, Borgstahl GEO, Ghosal G, Alnouti Y, Hollingsworth MA, Radhakrishnan P, Natarajan A. Small-molecule IKKβ activation modulator (IKAM) targets MAP3K1 and inhibits pancreatic tumor growth. Proc Natl Acad Sci U S A 2022; 119:e2115071119. [PMID: 35476515 PMCID: PMC9170026 DOI: 10.1073/pnas.2115071119] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 03/29/2022] [Indexed: 11/18/2022] Open
Abstract
Activation of inhibitor of nuclear factor NF-κB kinase subunit-β (IKKβ), characterized by phosphorylation of activation loop serine residues 177 and 181, has been implicated in the early onset of cancer. On the other hand, tissue-specific IKKβ knockout in Kras mutation-driven mouse models stalled the disease in the precancerous stage. In this study, we used cell line models, tumor growth studies, and patient samples to assess the role of IKKβ and its activation in cancer. We also conducted a hit-to-lead optimization study that led to the identification of 39-100 as a selective mitogen-activated protein kinase kinase kinase (MAP3K) 1 inhibitor. We show that IKKβ is not required for growth of Kras mutant pancreatic cancer (PC) cells but is critical for PC tumor growth in mice. We also observed elevated basal levels of activated IKKβ in PC cell lines, PC patient-derived tumors, and liver metastases, implicating it in disease onset and progression. Optimization of an ATP noncompetitive IKKβ inhibitor resulted in the identification of 39-100, an orally bioavailable inhibitor with improved potency and pharmacokinetic properties. The compound 39-100 did not inhibit IKKβ but inhibited the IKKβ kinase MAP3K1 with low-micromolar potency. MAP3K1-mediated IKKβ phosphorylation was inhibited by 39-100, thus we termed it IKKβ activation modulator (IKAM) 1. In PC models, IKAM-1 reduced activated IKKβ levels, inhibited tumor growth, and reduced metastasis. Our findings suggests that MAP3K1-mediated IKKβ activation contributes to KRAS mutation-associated PC growth and IKAM-1 is a viable pretherapeutic lead that targets this pathway.
Collapse
Affiliation(s)
- John Victor Napoleon
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE 68198
| | - Satish Sagar
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE 68198
| | - Sydney P. Kubica
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE 68198
| | - Lidia Boghean
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE 68198
| | - Smit Kour
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE 68198
| | - Hannah M. King
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE 68198
| | - Yogesh A. Sonawane
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE 68198
| | - Ayrianne J. Crawford
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE 68198
| | - Nagsen Gautam
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198
| | - Smitha Kizhake
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE 68198
| | - Pawel A. Bialk
- Gene Editing Institute, Christiana Care, Newark, DE 19713
| | - Eric Kmiec
- Gene Editing Institute, Christiana Care, Newark, DE 19713
| | | | - Prathamesh P. Patil
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE 68198
| | - Sandeep Rana
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE 68198
| | - Sarbjit Singh
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE 68198
| | - Janani Prahlad
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE 68198
| | - Paul M. Grandgenett
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE 68198
| | - Gloria E. O. Borgstahl
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE 68198
| | - Gargi Ghosal
- Department of Genetics Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198
| | - Yazen Alnouti
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198
| | - Michael A. Hollingsworth
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE 68198
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198
| | - Prakash Radhakrishnan
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE 68198
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198
| | - Amarnath Natarajan
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE 68198
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198
- Department of Genetics Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198
| |
Collapse
|
4
|
Xu H, Liu T, Li J, Chen F, Xu J, Hu L, Jiang L, Xiang Z, Wang X, Sheng J. Roburic Acid Targets TNF to Inhibit the NF-κB Signaling Pathway and Suppress Human Colorectal Cancer Cell Growth. Front Immunol 2022; 13:853165. [PMID: 35222445 PMCID: PMC8864141 DOI: 10.3389/fimmu.2022.853165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 01/24/2022] [Indexed: 12/24/2022] Open
Abstract
Tumor necrosis factor (TNF)-stimulated nuclear factor-kappa B (NF-κB) signaling plays very crucial roles in cancer development and progression, and represents a potential target for drug discovery. Roburic acid is a newly discovered tetracyclic triterpene acid isolated from oak galls and exhibits anti-inflammatory activity. However, whether roburic acid exerts antitumor effects through inhibition of TNF-induced NF-κB signaling remains unknown. Here, we demonstrated that roburic acid bound directly to TNF with high affinity (KD = 7.066 μM), blocked the interaction between TNF and its receptor (TNF-R1), and significantly inhibited TNF-induced NF-κB activation. Roburic acid exhibited antitumor activity in numerous cancer cells and could effectively induce G0/G1 cell cycle arrest and apoptosis in colorectal cancer cells. Importantly, roburic acid inhibited the TNF-induced phosphorylation of IKKα/β, IκBα, and p65, degradation of IκBα, nuclear translocation of p65, and NF-κB-target gene expression, including that of XIAP, Mcl-1, and Survivin, in colorectal cancer cells. Moreover, roburic acid suppressed tumor growth by blocking NF-κB signaling in a xenograft nude mouse model of colorectal cancer. Taken together, our findings showed that roburic acid directly binds to TNF with high affinity, thereby disrupting its interaction with TNF-R1 and leading to the inhibition of the NF-κB signaling pathway, both in vitro and in vivo. The results indicated that roburic acid is a novel TNF-targeting therapeutics agent in colorectal cancer as well as other cancer types.
Collapse
Affiliation(s)
- Huanhuan Xu
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, China.,College of Science, Yunnan Agricultural University, Kunming, China
| | - Titi Liu
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, China.,College of Science, Yunnan Agricultural University, Kunming, China
| | - Jin Li
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, China.,College of Science, Yunnan Agricultural University, Kunming, China
| | - Fei Chen
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, China.,College of Science, Yunnan Agricultural University, Kunming, China
| | - Jing Xu
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, China
| | - Lihong Hu
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, China
| | - Li Jiang
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, China
| | - Zemin Xiang
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, China.,College of Science, Yunnan Agricultural University, Kunming, China
| | - Xuanjun Wang
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, China.,College of Science, Yunnan Agricultural University, Kunming, China.,State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
| | - Jun Sheng
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, China.,State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
| |
Collapse
|
5
|
Yuanyuan L, Yakun L, Zhongyao L, Le Y, Weisong D, Moran G, Hui B, Chunyan L. Myeloid NEMO deficiency promotes tumor immunosuppression partly via MCP1-CCR2 axis. Exp Cell Res 2021; 399:112467. [PMID: 33428904 DOI: 10.1016/j.yexcr.2020.112467] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 12/21/2020] [Accepted: 12/27/2020] [Indexed: 11/19/2022]
Abstract
Tumor-associated macrophages (TAM), which are found in the tumor microenvironment of solid tumors, not only mediate cancer immune evasion but also promote tumor growth. The transcription factor NF-κB, which is a crucial link between inflammation and tumors, can accelerate tumor occurrence and development. NEMO, the regulatory subunit of the IKK complex, plays a pivotal role in activating the NF-κB signaling pathway. However, the function of myeloid NEMO in the tumor microenvironment remains unclear. Here, we found that conditional knockout of NEMO in myeloid cells promoted tumor growth in a transplanted cancer mouse model. In Nemofl/fl lyz-cre+/- mice, the deletion of Nemo in myeloid cells increased the recruitment of M2 macrophages and myeloid-derived suppressor cells (MDSCs) into the tumor, reduced the expression of apoptosis-related proteins, and upregulated the expression of the chemokine receptor CCR2, thereby promoting tumor growth in vivo. Then, we showed that blocking the MCP1-CCR2 pathway could inhibit tumor growth, especially in mice with myeloid NEMO deletion. In this study, we examined the mechanism of NEMO in myeloid cells and explored the role of NEMO in the prevention and treatment of cancer.
Collapse
Affiliation(s)
- Li Yuanyuan
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People's Republic of China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, 050000, People's Republic of China
| | - Liu Yakun
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People's Republic of China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, 050000, People's Republic of China
| | - Li Zhongyao
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People's Republic of China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, 050000, People's Republic of China
| | - Yi Le
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People's Republic of China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, 050000, People's Republic of China
| | - Duan Weisong
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People's Republic of China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, 050000, People's Republic of China; Institute of Cardiocerebrovascular Disease, Shijiazhuang, Hebei, 050000, People's Republic of China
| | - Guo Moran
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People's Republic of China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, 050000, People's Republic of China
| | - Bu Hui
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People's Republic of China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, 050000, People's Republic of China; Institute of Cardiocerebrovascular Disease, Shijiazhuang, Hebei, 050000, People's Republic of China
| | - Li Chunyan
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People's Republic of China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, 050000, People's Republic of China; Institute of Cardiocerebrovascular Disease, Shijiazhuang, Hebei, 050000, People's Republic of China.
| |
Collapse
|
6
|
Lin T, Gu J, Qu K, Zhang X, Ma X, Miao R, Xiang X, Fu Y, Niu W, She J, Liu C. A new risk score based on twelve hepatocellular carcinoma-specific gene expression can predict the patients' prognosis. Aging (Albany NY) 2019; 10:2480-2497. [PMID: 30243023 PMCID: PMC6188480 DOI: 10.18632/aging.101563] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 09/14/2018] [Indexed: 12/31/2022]
Abstract
A large panel of molecular biomarkers have been identified to predict the prognosis of hepatocellular carcinoma (HCC), yet with limited clinical application due to difficult extrapolation. We here generated a genetic risk score system comprised of 12 HCC-specific genes to better predict the prognosis of HCC patients. Four genomics profiling datasets (GSE5851, GSE28691, GSE15765 and GSE14323) were searched to seek HCC-specific genes by comparisons between cancer samples and normal liver tissues and between different subtypes of hepatic neoplasms. Univariate survival analysis screened HCC-specific genes associated with overall survival (OS) in the training dataset for next-step risk model construction. The prognostic value of the constructed HCC risk score system was then validated in the TCGA dataset. Stratified analysis indicated this scoring system showed better performance in elderly male patients with HBV infection and preoperative lower levels of creatinine, alpha-fetoprotein and platelet and higher level of albumin. Functional annotation of this risk model in high-risk patients revealed that pathways associated with cell cycle, cell migration and inflammation were significantly enriched. In summary, our constructed HCC-specific gene risk model demonstrated robustness and potentiality in predicting the prognosis of HCC patients, especially among elderly male patients with HBV infection and relatively better general conditions.
Collapse
Affiliation(s)
- Ting Lin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'a, Shaanxi 710061, China
| | - Jingxian Gu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'a, Shaanxi 710061, China
| | - Kai Qu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'a, Shaanxi 710061, China
| | - Xing Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'a, Shaanxi 710061, China
| | - Xiaohua Ma
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'a, Shaanxi 710061, China
| | - Runchen Miao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'a, Shaanxi 710061, China
| | - Xiaohong Xiang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'a, Shaanxi 710061, China
| | - Yunong Fu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'a, Shaanxi 710061, China
| | - Wenquan Niu
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, China
| | - Junjun She
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'a, Shaanxi 710061, China
| | - Chang Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'a, Shaanxi 710061, China
| |
Collapse
|
7
|
Cao ZQ, Wang XX, Lu L, Xu JW, Li XB, Zhang GR, Ma ZJ, Shi AC, Wang Y, Song YJ. β-Sitosterol and Gemcitabine Exhibit Synergistic Anti-pancreatic Cancer Activity by Modulating Apoptosis and Inhibiting Epithelial-Mesenchymal Transition by Deactivating Akt/GSK-3β Signaling. Front Pharmacol 2019; 9:1525. [PMID: 30670971 PMCID: PMC6331459 DOI: 10.3389/fphar.2018.01525] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 12/12/2018] [Indexed: 12/13/2022] Open
Abstract
β-sitosterol (BS), a major bioactive constituent present in plants, has shown potent anti-cancer activity against many human cancer cells, but its activity in pancreatic cancer (PC) cells has rarely been reported. Gemcitabine (GEM) is one of the first-line drugs for PC therapy, however, the treatment effect is not sustained due to prolonged drug resistance. In this study, we firstly studied the anti-PC activity and the mechanism of BS alone and in combination with GEM in vitro and in vivo. BS effectively inhibited the growth of PC cell lines by inhibiting proliferation, inducing G0/G1 phase arrest and apoptosis, suppressed the NF- kB activity, and increased expression of the protein Bax but decreased expression of the protein Bcl-2. Moreover, BS inhibited migration and invasion and downregulated epithelial–mesenchymal transition (EMT) markers and AKT/GSK-3β signaling pathways. Furthermore, the combination of BS and GEM exhibited a significant synergistic effect in MIAPaCa-2 and BXPC-3 cells. More importantly, the combined treatment with BS and GEM lead to significant growth inhibition of PC xenografts. Overall, our data revealed a promising treatment option for PC by the combination therapy of BS and GEM.
Collapse
Affiliation(s)
- Zhang-Qi Cao
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xue-Xi Wang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Li Lu
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Jing-Wen Xu
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xiao-Bin Li
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Guang-Ru Zhang
- Qinghai Hospital of Traditional Chinese Medicine, Xining, China
| | - Zhan-Jun Ma
- The Second Clinical School, Lanzhou University, Lanzhou, China
| | - An-Chen Shi
- The Second Clinical School, Lanzhou University, Lanzhou, China
| | - Yan Wang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Yu-Jun Song
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| |
Collapse
|
8
|
Yang X, Song S. Silencing of Astrocyte elevated gene-1 (AEG-1) inhibits proliferation, migration, and invasiveness, and promotes apoptosis in pancreatic cancer cells. Biochem Cell Biol 2018; 97:165-175. [PMID: 30359541 DOI: 10.1139/bcb-2018-0181] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
To investigate the role of Astrocyte elevated gene-1 (AEG-1) in the development and progress of pancreatic cancer, short hairpin RNA (shRNA) was inserted into the RNA interference vector to knock-down the endogenous AEG-1 in two pancreatic cancer cell lines: AsPC-1 and PANC-1. Our results showed that silencing of AEG-1 suppressed the proliferation, colony formation ability, and cell stemness of AsPC-1 and PANC-1 cells, and inhibited their G1-to-S phase transition. Results from apoptosis assay showed that knock-down of AEG-1 led to cell apoptosis. The expression of anti-apoptotic Bcl-2 was downregulated and that of the pro-apoptotic Bax and cleaved caspase-3 was upregulated in AEG-1-silenced pancreatic cancer cells. Further, the capability of AEG-1-silenced cells to migrate and to invade through the Matrigel-coated membrane was weaker, and the expression of matrix metallopeptidase 2 (MMP-2) and MMP-9 were decreased. Moreover, the AKT-β-catenin signaling pathway was inhibited in the cells with knock-down of AEG-1. In addition, the growth of xenograft tumors formed by AsPC-1 and PANC-1 cells was suppressed by AEG-1 shRNA. In conclusion, our study demonstrates that pancreatic cancer cells require AEG-1 to maintain their survival and metastasis, suggesting AEG-1 as a potential target for the treatment of pancreatic cancers.
Collapse
Affiliation(s)
- Xing Yang
- Department of Pancreatobiliary Surgery, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China.,Department of Pancreatobiliary Surgery, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Shaowei Song
- Department of Pancreatobiliary Surgery, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China.,Department of Pancreatobiliary Surgery, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| |
Collapse
|
9
|
Li Q, Yang G, Feng M, Zheng S, Cao Z, Qiu J, You L, Zheng L, Hu Y, Zhang T, Zhao Y. NF-κB in pancreatic cancer: Its key role in chemoresistance. Cancer Lett 2018; 421:127-134. [PMID: 29432846 DOI: 10.1016/j.canlet.2018.02.011] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 01/18/2018] [Accepted: 02/06/2018] [Indexed: 12/15/2022]
Abstract
Pancreatic cancer is considered a lethal disease with a high mortality and an extremely low five-year survival rate. Chemotherapy plays a pivotal role in pancreatic cancer treatment both in an adjuvant setting after complete resection and in the case of unresectable metastatic disease. However, none of the available combination chemotherapy regimens has resulted in satisfactory survival outcomes. Recent studies have revealed that both constitutive and induced activation of nuclear factor kappa B (NF-κB) in pancreatic cancer cells are closely associated with cell proliferation, invasion, anti-apoptosis, inflammation, angiogenesis and chemotherapeutic resistance. Therefore, NF-κB inhibitors in combination with cytotoxic compounds have been reported as novel agents that improve chemotherapy sensitivity in pancreatic cancer. In this review, we outline recent developments in the understanding of the role of the NF-κB signaling pathway and its associated genes in the progression of pancreatic cancer and highlight some potentially effective strategies for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Quanxiao Li
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; Second Affiliated Hospital, Harbin Medical University, Harbin, 150086, China.
| | - Gang Yang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Mengyu Feng
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Suli Zheng
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Zhe Cao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Jiangdong Qiu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Lianfang Zheng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Ya Hu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; Clinical Immunology Center, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|