1
|
Ayaz MO, Bhat AQ, Akhter Z, Badsera N, Hossain MM, Showket F, Parveen S, Dar MS, Tiwari H, Kumari N, Bhardwaj M, Hussain R, Sharma A, Kumar M, Singh U, Nargorta A, Kshatri AS, Nandi U, Monga SP, Ramajayan P, Singh PP, Dar MJ. Identification of a novel GSK3β inhibitor involved in abrogating KRas dependent pancreatic tumors in Wnt/beta-catenin and NF-kB dependent manner. Life Sci 2024; 351:122840. [PMID: 38876185 DOI: 10.1016/j.lfs.2024.122840] [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: 04/23/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 06/16/2024]
Abstract
Pancreatic cancer is an aggressive malignancy with a poor survival rate because it is difficult to diagnose the disease during its early stages. The currently available treatments, which include surgery, chemotherapy and radiation therapy, offer only limited survival benefit. Pharmacological interventions to inhibit Glycogen Synthase Kinase-3beta (GSK3β) activity is an important therapeutic strategy for the treatment of pancreatic cancer because GSK3β is one of the key factors involved in the onset, progression as well as in the acquisition of chemoresistance in pancreatic cancer. Here, we report the identification of MJ34 as a potent GSK3β inhibitor that significantly reduced growth and survival of human mutant KRas dependent pancreatic tumors. MJ34 mediated GSK3β inhibition was seen to induce apoptosis in a β-catenin dependent manner and downregulate NF-kB activity in MiaPaCa-2 cells thereby impeding cell survival and anti-apoptotic processes in these cells as well as in the xenograft model of pancreatic cancer. In vivo acute toxicity and in vitro cardiotoxicity studies indicate that MJ34 is well tolerated without any adverse effects. Taken together, we report the discovery of MJ34 as a potential drug candidate for the therapeutic treatment of mutant KRas-dependent human cancers through pharmacological inhibition of GSK3β.
Collapse
Affiliation(s)
- Mir Owais Ayaz
- Laboratory of Cell and Molecular Biology, Department of Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu, India; Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh 201002, India
| | - Aadil Qadir Bhat
- Laboratory of Cell and Molecular Biology, Department of Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu, India; Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh 201002, India
| | - Zaheen Akhter
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh 201002, India; Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, India
| | - Neetu Badsera
- Laboratory of Cell and Molecular Biology, Department of Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu, India; Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh 201002, India
| | - Md Mehedi Hossain
- Laboratory of Cell and Molecular Biology, Department of Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu, India; Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh 201002, India
| | - Farheen Showket
- Laboratory of Cell and Molecular Biology, Department of Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu, India; Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh 201002, India
| | - Sabra Parveen
- Laboratory of Cell and Molecular Biology, Department of Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu, India; Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh 201002, India
| | - Mohmmad Saleem Dar
- Laboratory of Cell and Molecular Biology, Department of Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu, India; Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh 201002, India
| | - Harshita Tiwari
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh 201002, India; Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, India
| | - Nedhi Kumari
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh 201002, India; Department of Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Mahir Bhardwaj
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh 201002, India; Department of Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Razak Hussain
- Department of Entomology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Ashutosh Sharma
- Division of Neuroscience and Aging Biology, CSIR-Central Drug Research Institute CDRI, Lucknow 226031, India
| | - Mukesh Kumar
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, India; Medicinal Product Chemistry, Sussex Drug Discovery Centre, School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9QG, United Kingdom of Great Britain and Northern Ireland
| | - Umed Singh
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, India; Department f Chemistry, E331 Chemistry Building, The University of Iowa, Iowa City, IA 52242-1294, USA
| | - Amit Nargorta
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh 201002, India; Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, India
| | - Aravind Singh Kshatri
- Division of Neuroscience and Aging Biology, CSIR-Central Drug Research Institute CDRI, Lucknow 226031, India
| | - Utpal Nandi
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh 201002, India; Department of Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India; Bose Institute, Unified Academic Campus, Kolkata 700032, India
| | - Satdarshan Pal Monga
- Pittsburgh Liver Research Center, School of Medicine, University of Pittsburgh, Pittsburgh, USA
| | - P Ramajayan
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh 201002, India; Department of Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Parvinder Pal Singh
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh 201002, India; Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, India.
| | - Mohd Jamal Dar
- Laboratory of Cell and Molecular Biology, Department of Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu, India; Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh 201002, India.
| |
Collapse
|
2
|
Zhang H, Read A, Cataisson C, Yang HH, Lee WC, Turk BE, Yuspa SH, Luo J. Protein phosphatase 6 activates NF-κB to confer sensitivity to MAPK pathway inhibitors in KRAS- and BRAF-mutant cancer cells. Sci Signal 2024; 17:eadd5073. [PMID: 38743809 PMCID: PMC11238902 DOI: 10.1126/scisignal.add5073] [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: 06/16/2022] [Accepted: 04/25/2024] [Indexed: 05/16/2024]
Abstract
The Ras-mitogen-activated protein kinase (MAPK) pathway is a major target for cancer treatment. To better understand the genetic pathways that modulate cancer cell sensitivity to MAPK pathway inhibitors, we performed a CRISPR knockout screen with MAPK pathway inhibitors on a colorectal cancer (CRC) cell line carrying mutant KRAS. Genetic deletion of the catalytic subunit of protein phosphatase 6 (PP6), encoded by PPP6C, rendered KRAS- and BRAF-mutant CRC and BRAF-mutant melanoma cells more resistant to these inhibitors. In the absence of MAPK pathway inhibition, PPP6C deletion in CRC cells decreased cell proliferation in two-dimensional (2D) adherent cultures but accelerated the growth of tumor spheroids in 3D culture and tumor xenografts in vivo. PPP6C deletion enhanced the activation of nuclear factor κB (NF-κB) signaling in CRC and melanoma cells and circumvented the cell cycle arrest and decreased cyclin D1 abundance induced by MAPK pathway blockade in CRC cells. Inhibiting NF-κB activity by genetic and pharmacological means restored the sensitivity of PPP6C-deficient cells to MAPK pathway inhibition in CRC and melanoma cells in vitro and in CRC cells in vivo. Furthermore, a R264 point mutation in PPP6C conferred loss of function in CRC cells, phenocopying the enhanced NF-κB activation and resistance to MAPK pathway inhibition observed for PPP6C deletion. These findings demonstrate that PP6 constrains the growth of KRAS- and BRAF-mutant cancer cells, implicates the PP6-NF-κB axis as a modulator of MAPK pathway output, and presents a rationale for cotargeting the NF-κB pathway in PPP6C-mutant cancer cells.
Collapse
Affiliation(s)
- Haibo Zhang
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Abigail Read
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
- Current affiliation: Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Christophe Cataisson
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Howard H. Yang
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Wei-Chun Lee
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Benjamin E. Turk
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA
| | - Stuart H. Yuspa
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Ji Luo
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
3
|
Zhang M, Xu W, Yang N, Li Z, Zhou S, Liu X, Wang J, Li H. PCV2 Induced Endothelial Derived IL-8 Affects MoDCs Maturation Mainly via NF-κB Signaling Pathway. Viruses 2024; 16:646. [PMID: 38675986 PMCID: PMC11053600 DOI: 10.3390/v16040646] [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: 02/05/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Porcine circovirus type 2 (PCV2) infection can cause immunosuppressive diseases in pigs. Vascular endothelial cells (VECs), as the target cells for PCV2, play an important role in the immune response and inflammatory regulation. Endothelial IL-8, which is produced by porcine hip artery endothelial cells (PIECs) infected with PCV2, can inhibit the maturation of monocyte-derived dendritic cells (MoDCs). Here, we established a co-culture system of MoDCs and different groups of PIECs to further investigate the PCV2-induced endothelial IL-8 signaling pathway that drives the inhibition of MoDC maturation. The differentially expressed genes related to MoDC maturation were mainly enriched in the NF-κB and JAK2-STAT3 signaling pathways. Both the NF-κB related factor RELA and JAK2-STAT3 signaling pathway related factors (IL2RA, JAK, STAT2, STAT5, IL23A, IL7, etc.) decreased significantly in the IL-8 up-regulated group, and increased significantly in the down-regulated group. The expression of NF-κB p65 in the IL-8 up-regulated group was reduced significantly, and the expression of IκBα was increased significantly. Nuclear translocation of NF-κB p65 was inhibited, while the nuclear translocation of p-STAT3 was increased in MoDCs in the PCV2-induced endothelial IL-8 group. The results of treatment with NF-κB signaling pathway inhibitors showed that the maturation of MoDCs was inhibited and the expression of IL-12 and GM-CSF at mRNA level were lower. Inhibition of the JAK2-STAT3 signaling pathway had no significant effect on maturation, and the expression of IL-12 and GM-CSF at mRNA level produced no significant change. In summary, the NF-κB signaling pathway is the main signaling pathway of MoDC maturation, and is inhibited by the PCV2-induced up-regulation of endothelial-derived IL-8.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jianfang Wang
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing 102206, China; (M.Z.); (W.X.); (N.Y.); (Z.L.); (S.Z.); (X.L.)
| | - Huanrong Li
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing 102206, China; (M.Z.); (W.X.); (N.Y.); (Z.L.); (S.Z.); (X.L.)
| |
Collapse
|
4
|
Li H, Hu J, Qiu L, Wu Y, Zhong B, Ye R, Xie B. Molecular mechanisms of HCG18 in the sorafenib resistance of hepatocellular carcinoma. Anticancer Drugs 2024; 35:55-62. [PMID: 37823256 DOI: 10.1097/cad.0000000000001539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
Sorafenib has been approved for advance hepatocellular carcinoma (HCC), however, drug resistance often occurred. Therefore, it is of great significance to clarify the underlying mechanisms of sorafenib resistance and to find out the effective strategies to overcome sorafenib resistance. The expression of HCG18 was detected by qPCR, MTT, colony formation, flow cytometry and TUNEL assay were used to explore the function of HCG18 on sorafenib resistance in HCC. RNA pull-down, RNA immunoprecipitation, immunofluorescence labeling, luciferase reporter assay, western blot and qPCR were used to investigate the mechanism of HCG18 regulating sorafenib resistance in HCC. Our results showed that HCG18 was significantly increased in HCC, which resulted in shorter 5-year survival for patients with HCC. Sorafenib can induce the expression of HCG18, suggesting HCG18 might be involved in sorafenib resistance in HCC. Further analysis showed that knockdown of HCG18 can reduce viability and increase apoptosis of HCC cells. Mechanistically, HCG18 can bind to USP15, further regulated the protein stability of p65, TAB2 and TAB3, and nuclear location of p65, which finally modulated the NF-κB signaling. Our findings showed that HCG18 played an important role in sorafenib resistance in HCC. And knockdown of HCG18 can promote the sensitivity of HCC cells to sorafenib, inferring that targeting HCG18 might be an effective strategy to overcome sorafenib resistance in HCC.
Collapse
Affiliation(s)
- Heping Li
- Department of Medical Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou
| | - Jie Hu
- Department of Medical Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou
| | - Lijie Qiu
- Department of Medical Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou
| | - Yijiang Wu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Gannan Medical University
| | - Baiyin Zhong
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Gannan Medical University
| | - Rong Ye
- Department of General surgery III, the First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Binhui Xie
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Gannan Medical University
| |
Collapse
|
5
|
Wang Y, Yao M, Li C, Yang K, Qin X, Xu L, Shi S, Yu C, Meng X, Xie C. Targeting ST8SIA6-AS1 counteracts KRAS G12C inhibitor resistance through abolishing the reciprocal activation of PLK1/c-Myc signaling. Exp Hematol Oncol 2023; 12:105. [PMID: 38104151 PMCID: PMC10724920 DOI: 10.1186/s40164-023-00466-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 12/03/2023] [Indexed: 12/19/2023] Open
Abstract
BACKGROUND KRASG12C inhibitors (KRASG12Ci) AMG510 and MRTX849 have shown promising efficacy in clinical trials and been approved for the treatment of KRASG12C-mutant cancers. However, the emergence of therapy-related drug resistance limits their long-term potential. This study aimed to identify the critical mediators and develop overcoming strategies. METHODS By using RNA sequencing, RT-qPCR and immunoblotting, we identified and validated the upregulation of c-Myc activity and the amplification of the long noncoding RNA ST8SIA6-AS1 in KRASG12Ci-resistant cells. The regulatory axis ST8SIA6-AS1/Polo-like kinase 1 (PLK1)/c-Myc was investigated by bioinformatics, RNA fluorescence in situ hybridization, RNA immunoprecipitation, RNA pull-down and chromatin immunoprecipitation. Gain/loss-of-function assays, cell viability assay, xenograft models, and IHC staining were conducted to evaluate the anti-cancer effects of co-inhibition of ST8SIA6-AS1/PLK1 pathway and KRAS both in vitro and in vivo. RESULTS KRASG12Ci sustainably decreased c-Myc levels in responsive cell lines but not in cell lines with intrinsic or acquired resistance to KRASG12Ci. PLK1 activation contributed to this ERK-independent c-Myc stability, which in turn directly induced PLK1 transcription, forming a positive feedback loop and conferring resistance to KRASG12Ci. ST8SIA6-AS1 was found significantly upregulated in resistant cells and facilitated the proliferation of KRASG12C-mutant cancers. ST8SIA6-AS1 bound to Aurora kinase A (Aurora A)/PLK1 and promoted Aurora A-mediated PLK1 phosphorylation. Concurrent targeting of KRAS and ST8SIA6-AS1/PLK1 signaling suppressed both ERK-dependent and -independent c-Myc expression, synergistically led to cell death and tumor regression and overcame KRASG12Ci resistance. CONCLUSIONS Our study deciphers that the axis of ST8SIA6-AS1/PLK1/c-Myc confers both intrinsic and acquired resistance to KRASG12Ci and represents a promising therapeutic target for combination strategies with KRASG12Ci in the treatment of KRASG12C-mutant cancers.
Collapse
Affiliation(s)
- Yafang Wang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 393 Middle Huaxia Road, Shanghai, 201210, People's Republic of China
| | - Mingyue Yao
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 393 Middle Huaxia Road, Shanghai, 201210, People's Republic of China
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), University of Science and Technology of China, Hefei, Anhui, China
- Drug Discovery and Development Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, People's Republic of China
| | - Cheng Li
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 393 Middle Huaxia Road, Shanghai, 201210, People's Republic of China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Kexin Yang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
- Lingang Laboratory, 319 Yueyang Road, Shanghai, 200031, China
| | - Xiaolong Qin
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 393 Middle Huaxia Road, Shanghai, 201210, People's Republic of China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Lansong Xu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 393 Middle Huaxia Road, Shanghai, 201210, People's Republic of China
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), University of Science and Technology of China, Hefei, Anhui, China
- Drug Discovery and Development Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, People's Republic of China
| | - Shangxuan Shi
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 393 Middle Huaxia Road, Shanghai, 201210, People's Republic of China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Chengcheng Yu
- Drug Discovery and Development Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, People's Republic of China
- Lingang Laboratory, 319 Yueyang Road, Shanghai, 200031, China
| | - Xiangjun Meng
- Gastroenterology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
- China Center for Digestive Diseases Research and Clinical Translation of Shanghai Jiao Tong University, Shanghai, 200001, China
- China Shanghai Key Laboratory of Gut Microecology and Associated Major Diseases Research, Shanghai, 200001, China
| | - Chengying Xie
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 393 Middle Huaxia Road, Shanghai, 201210, People's Republic of China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
- Lingang Laboratory, 319 Yueyang Road, Shanghai, 200031, China.
| |
Collapse
|
6
|
Mukherjee AG, Wanjari UR, Gopalakrishnan AV, Bradu P, Biswas A, Ganesan R, Renu K, Dey A, Vellingiri B, El Allali A, Alsamman AM, Zayed H, George Priya Doss C. Evolving strategies and application of proteins and peptide therapeutics in cancer treatment. Biomed Pharmacother 2023; 163:114832. [PMID: 37150032 DOI: 10.1016/j.biopha.2023.114832] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/18/2023] [Accepted: 04/30/2023] [Indexed: 05/09/2023] Open
Abstract
Several proteins and peptides have therapeutic potential and can be used for cancer therapy. By binding to cell surface receptors and other indicators uniquely linked with or overexpressed on tumors compared to healthy tissue, protein biologics enhance the active targeting of cancer cells, as opposed to the passive targeting of cells by conventional small-molecule chemotherapeutics. This study focuses on peptide medications that exist to slow or stop tumor growth and the spread of cancer, demonstrating the therapeutic potential of peptides in cancer treatment. As an alternative to standard chemotherapy, peptides that selectively kill cancer cells while sparing healthy tissue are developing. A mountain of clinical evidence supports the efficacy of peptide-based cancer vaccines. Since a single treatment technique may not be sufficient to produce favourable results in the fight against cancer, combination therapy is emerging as an effective option to generate synergistic benefits. One example of this new area is the use of anticancer peptides in combination with nonpeptidic cytotoxic drugs or the combination of immunotherapy with conventional therapies like radiation and chemotherapy. This review focuses on the different natural and synthetic peptides obtained and researched. Discoveries, manufacture, and modifications of peptide drugs, as well as their contemporary applications, are summarized in this review. We also discuss the benefits and difficulties of potential advances in therapeutic peptides.
Collapse
Affiliation(s)
- Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India.
| | - Pragya Bradu
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Antara Biswas
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Raja Ganesan
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 24252, South Korea
| | - Kaviyarasi Renu
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077 Tamil Nadu, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, West Bengal 700073, India
| | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda 151401, Punjab, India
| | - Achraf El Allali
- African Genome Center, Mohammed VI Polytechnic University, Ben Guerir, Morocco.
| | - Alsamman M Alsamman
- Department of Genome Mapping, Molecular Genetics, and Genome Mapping Laboratory, Agricultural Genetic Engineering Research Institute, Giza, Egypt
| | - Hatem Zayed
- Department of Biomedical Sciences College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - C George Priya Doss
- Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| |
Collapse
|
7
|
Spella M, Ntaliarda G, Skiadas G, Lamort AS, Vreka M, Marazioti A, Lilis I, Bouloukou E, Giotopoulou GA, Pepe MAA, Weiss SAI, Petrera A, Hauck SM, Koch I, Lindner M, Hatz RA, Behr J, Arendt KAM, Giopanou I, Brunn D, Savai R, Jenne DE, de Château M, Yull FE, Blackwell TS, Stathopoulos GT. Non-Oncogene Addiction of KRAS-Mutant Cancers to IL-1β via Versican and Mononuclear IKKβ. Cancers (Basel) 2023; 15:1866. [PMID: 36980752 PMCID: PMC10047096 DOI: 10.3390/cancers15061866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/10/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Kirsten rat sarcoma virus (KRAS)-mutant cancers are frequent, metastatic, lethal, and largely undruggable. While interleukin (IL)-1β and nuclear factor (NF)-κB inhibition hold promise against cancer, untargeted treatments are not effective. Here, we show that human KRAS-mutant cancers are addicted to IL-1β via inflammatory versican signaling to macrophage inhibitor of NF-κB kinase (IKK) β. Human pan-cancer and experimental NF-κB reporter, transcriptome, and proteome screens reveal that KRAS-mutant tumors trigger macrophage IKKβ activation and IL-1β release via secretory versican. Tumor-specific versican silencing and macrophage-restricted IKKβ deletion prevents myeloid NF-κB activation and metastasis. Versican and IKKβ are mutually addicted and/or overexpressed in human cancers and possess diagnostic and prognostic power. Non-oncogene KRAS/IL-1β addiction is abolished by IL-1β and TLR1/2 inhibition, indicating cardinal and actionable roles for versican and IKKβ in metastasis.
Collapse
Affiliation(s)
- Magda Spella
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Giannoula Ntaliarda
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Georgios Skiadas
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Anne-Sophie Lamort
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Malamati Vreka
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Antonia Marazioti
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Ioannis Lilis
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Eleni Bouloukou
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Georgia A. Giotopoulou
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Mario A. A. Pepe
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Stefanie A. I. Weiss
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Agnese Petrera
- Research Unit Protein Science-Core Facility Proteomics, Helmholtz Center Munich–German Research Center for Environmental Health, 80939 Munich, Germany
| | - Stefanie M. Hauck
- Research Unit Protein Science-Core Facility Proteomics, Helmholtz Center Munich–German Research Center for Environmental Health, 80939 Munich, Germany
| | - Ina Koch
- Center for Thoracic Surgery Munich, Ludwig-Maximilians-University of Munich and Asklepios Medical Center, 82131 Gauting, Germany
| | - Michael Lindner
- Center for Thoracic Surgery Munich, Ludwig-Maximilians-University of Munich and Asklepios Medical Center, 82131 Gauting, Germany
| | - Rudolph A. Hatz
- Center for Thoracic Surgery Munich, Ludwig-Maximilians-University of Munich and Asklepios Medical Center, 82131 Gauting, Germany
| | - Juergen Behr
- Department of Internal Medicine V, Ludwig-Maximilian-University of Munich, 81377 Munich, Germany
| | - Kristina A. M. Arendt
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Ioanna Giopanou
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - David Brunn
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Rajkumar Savai
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Frankfurt Cancer Institute (FCI), Goethe University, 60596 Frankfurt am Main, Germany
- Department of Internal Medicine and Institute for Lung Health (ILH), Justus Liebig University, 35392 Giessen, Germany
| | - Dieter E. Jenne
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
- Max-Planck-Institute of Neurobiology, 82152 Planegg, Germany
| | | | - Fiona E. Yull
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Timothy S. Blackwell
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Georgios T. Stathopoulos
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| |
Collapse
|
8
|
Abstract
C-Myc overexpression is a common finding in pancreatic cancer and predicts the aggressive behavior of cancer cells. It binds to the promoter of different genes, thereby regulating their transcription. C-Myc is downstream of KRAS and interacts with several oncogenic and proliferative pathways in pancreatic cancer. C-Myc enhances aerobic glycolysis in cancer cells and regulates glutamate biosynthesis from glutamine. It provides enough energy for cancer cells' metabolism and sufficient substrate for the synthesis of organic molecules. C-Myc overexpression is associated with chemoresistance, intra-tumor angiogenesis, epithelial-mesenchymal transition (EMT), and metastasis in pancreatic cancer. Despite its title, c-Myc is not "undruggable" and recent studies unveiled that it can be targeted, directly or indirectly. Small molecules that accelerate c-Myc ubiquitination and degradation have been effective in preclinical studies. Small molecules that hinder c-Myc-MAX heterodimerization or c-Myc/MAX/DNA complex formation can functionally inhibit c-Myc. In addition, c-Myc can be targeted through transcriptional, post-transcriptional, and translational modifications.
Collapse
Affiliation(s)
- Moein Ala
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
9
|
Du YB, Wang XF, Liu XJ, Li Y, Miao QF, Jiang M, Sheng WJ, Zhen YS. The recombinant defensin/HSA fusion protein that inhibits NF-κb associated with intensive macropinocytosis shows potent efficacy against pancreatic cancer. Biochem Pharmacol 2022; 201:115057. [PMID: 35489393 DOI: 10.1016/j.bcp.2022.115057] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/11/2022] [Accepted: 04/19/2022] [Indexed: 11/29/2022]
Abstract
KRAS mutation and NF-κB both play crucial role in pancreatic cancer; in addition, defensin, the peptide mediator in innate immunity, can inhibit NF-κB. Assuming a strategy that targets both NF-κB and concomitantly the mutated KRAS indirectly via intensive macropinocytosis, we designed and generated a recombinant protein DF2-HSA which consists of two molecules of human beta-defensin 2 (HBD2) and a moiety of human serum albumin (HSA). As shown, the recombinant protein DF2-HSA markedly down-regulated NF-κB in both KRAS mutant MIA PaCa-2 cells and wild type BxPC-3 cells. Determined by confocal microscopy, the uptake of DF2-HSA in MIA PaCa-2 cells was more intense than that in BxPC-3 cells. The uptake was blocked by the specific inhibitor EIPA, indicating that DF2-HSA internalized via macropinocytosis. DF2-HSA displayed more potent cytotoxicity to cancer cells than HBD2. DF2-HSA induced apoptosis in cancer cells. Notably, DF2-HSA inhibited tumor cell spheroid formation, an effect comparable to that of salinomycin. DF2-HSA inhibited tumor cell migration and invasion. As detected with scanning electron microscopy, DF2-HSA strongly depleted filopodia on cell surface; and salinomycin induced similar changes. By in vivo imaging, DF2-HSA displayed intense tumor-site accumulation and lasting retention for over 14 days; however, HBD2 showed much less tumor-site accumulation and a shorter retention time for only 24 h. DF2-HSA suppressed the growth of pancreatic cancer MIA PaCa-2 xenograft in athymic mice; and its combination with gemcitabine achieved higher antitumor efficacy. In summary, the recombinant defensin/HSA fusion protein that inhibits NF-κb associated with intensive macropinocytosis is highly effective against pancreatic cancer.
Collapse
Affiliation(s)
- Yi-Bo Du
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | | | - Xiu-Jun Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yi Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Qing-Fang Miao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Min Jiang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wei-Jin Sheng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Yong-Su Zhen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
10
|
Boyle AJ, Narvaez A, Chassé M, Vasdev N. PET imaging of glycogen synthase kinase-3 in pancreatic cancer xenograft mouse models. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2022; 12:1-14. [PMID: 35295885 PMCID: PMC8918402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 01/09/2022] [Indexed: 06/14/2023]
Abstract
Glycogen synthase kinase-3 (GSK-3) contributes to tumorigenesis in pancreatic cancer by modulating cell proliferation and survival. This study evaluated the lead GSK-3 targeted PET radiotracers for neuro-PET imaging, [11C]PF-367 and [11C]OCM-44, in pancreatic cancer xenograft mice. Immunohistochemistry showed that GSK-3α and GSK-3β were overexpressed in PANC-1 xenografts. In autoradiography studies, higher specific binding was observed for [3H]PF-367 compared to [3H]OCM-44 when co-incubated with unlabeled PF-367 (59.2±1.8% vs 22.6±3.75%, respectively). Co-incubation of [11C]OCM-44 with OCM-44 did not improve the specific binding (25.5±30.2%). In dynamic PET imaging of PANC-1 xenograft mouse models, tumors were not visualized with [11C]PF-367 but were well visualized with [11C]OCM-44. Time-activity curves revealed no difference in accumulation in PANC-1 tumor tissue compared to muscle tissue in [11C]PF-367 baseline studies, while a significant difference was observed for [11C]OCM-44 with a tumor-to-muscle ratio of 1.6. Tumor radioactivity accumulation following injection with [11C]OCM-44 was not displaced by pre-treatment with unlabeled PF-367. Radiometabolite analysis showed that intact [11C]PF-367 accounted for 7.5% of tumor radioactivity, with >30% in plasma, at 40 min post-injection of the radiotracer, and that intact [11C]OCM-44 accounted for 20% of tumor radioactivity, with >60% in plasma. [11C]OCM-44 is superior to [11C]PF-367 for detecting lesions in preclinical mouse models of pancreatic cancer, however, both radiotracers undergo rapid metabolism in vivo. GSK-3 PET radiotracers with improved in vivo stability are needed for clinical translation. To our knowledge this work represents the first PET imaging study of GSK-3 in oncology.
Collapse
Affiliation(s)
- Amanda J Boyle
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental HealthToronto, Ontario, M5T 1R8, Canada
- Department of Psychiatry, University of TorontoToronto, Ontario, M5T 1R8, Canada
| | - Andrea Narvaez
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental HealthToronto, Ontario, M5T 1R8, Canada
| | - Melissa Chassé
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental HealthToronto, Ontario, M5T 1R8, Canada
- Institute of Medical Sciences, University of TorontoToronto, Ontario, M5S 1A8, Canada
| | - Neil Vasdev
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental HealthToronto, Ontario, M5T 1R8, Canada
- Department of Psychiatry, University of TorontoToronto, Ontario, M5T 1R8, Canada
- Institute of Medical Sciences, University of TorontoToronto, Ontario, M5S 1A8, Canada
| |
Collapse
|
11
|
Stati G, Passaretta F, Gindraux F, Centurione L, Di Pietro R. The Role of the CREB Protein Family Members and the Related Transcription Factors in Radioresistance Mechanisms. Life (Basel) 2021; 11:life11121437. [PMID: 34947968 PMCID: PMC8706059 DOI: 10.3390/life11121437] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 12/02/2021] [Accepted: 12/16/2021] [Indexed: 02/05/2023] Open
Abstract
In the framework of space flight, the risk of radiation carcinogenesis is considered a "red" risk due to the high likelihood of occurrence as well as the high potential impact on the quality of life in terms of disease-free survival after space missions. The cyclic AMP response element-binding protein (CREB) is overexpressed both in haematological malignancies and solid tumours and its expression and function are modulated following irradiation. The CREB protein is a transcription factor and member of the CREB/activating transcription factor (ATF) family. As such, it has an essential role in a wide range of cell processes, including cell survival, proliferation, and differentiation. Among the CREB-related nuclear transcription factors, NF-κB and p53 have a relevant role in cell response to ionising radiation. Their expression and function can decide the fate of the cell by choosing between death or survival. The aim of this review was to define the role of the CREB/ATF family members and the related transcription factors in the response to ionising radiation of human haematological malignancies and solid tumours.
Collapse
Affiliation(s)
- Gianmarco Stati
- Department of Medicine and Ageing Sciences, G. d’Annunzio University of Chieti-Pescara, 66100 Chieti, Italy; (F.P.); (L.C.); (R.D.P.)
- Correspondence: ; Tel.: +39-08713554567
| | - Francesca Passaretta
- Department of Medicine and Ageing Sciences, G. d’Annunzio University of Chieti-Pescara, 66100 Chieti, Italy; (F.P.); (L.C.); (R.D.P.)
| | - Florelle Gindraux
- Laboratoire de Nanomédecine, Imagerie, Thérapeutique EA 4662, Université Bourgogne Franche-Comté, 25030 Besançon, France;
- Service de Chirurgie Orthopédique, Traumatologique et Plastique, CHU, 25030 Besançon, France
| | - Lucia Centurione
- Department of Medicine and Ageing Sciences, G. d’Annunzio University of Chieti-Pescara, 66100 Chieti, Italy; (F.P.); (L.C.); (R.D.P.)
| | - Roberta Di Pietro
- Department of Medicine and Ageing Sciences, G. d’Annunzio University of Chieti-Pescara, 66100 Chieti, Italy; (F.P.); (L.C.); (R.D.P.)
| |
Collapse
|
12
|
Bansod S, Dodhiawala PB, Lim KH. Oncogenic KRAS-Induced Feedback Inflammatory Signaling in Pancreatic Cancer: An Overview and New Therapeutic Opportunities. Cancers (Basel) 2021; 13:cancers13215481. [PMID: 34771644 PMCID: PMC8582583 DOI: 10.3390/cancers13215481] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/25/2021] [Accepted: 10/27/2021] [Indexed: 12/20/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains highly refractory to treatment. While the KRAS oncogene is present in almost all PDAC cases and accounts for many of the malignant feats of PDAC, targeting KRAS or its canonical, direct effector cascades remains unsuccessful in patients. The recalcitrant nature of PDAC is also heavily influenced by its highly fibro-inflammatory tumor microenvironment (TME), which comprises an acellular extracellular matrix and various types of non-neoplastic cells including fibroblasts, immune cells, and adipocytes, underscoring the critical need to delineate the bidirectional signaling interplay between PDAC cells and the TME in order to develop novel therapeutic strategies. The impact of tumor-cell KRAS signaling on various cell types in the TME has been well covered by several reviews. In this article, we critically reviewed evidence, including work from our group, on how the feedback inflammatory signals from the TME impact and synergize with oncogenic KRAS signaling in PDAC cells, ultimately augmenting their malignant behavior. We discussed past and ongoing clinical trials that target key inflammatory pathways in PDAC and highlight lessons to be learned from outcomes. Lastly, we provided our perspective on the future of developing therapeutic strategies for PDAC through understanding the breadth and complexity of KRAS and the inflammatory signaling network.
Collapse
Affiliation(s)
- Sapana Bansod
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA; (S.B.); (P.B.D.)
| | - Paarth B. Dodhiawala
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA; (S.B.); (P.B.D.)
- Medical Scientist Training Program, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Kian-Huat Lim
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA; (S.B.); (P.B.D.)
- Correspondence: ; Tel.: +1-314-362-6157
| |
Collapse
|
13
|
Mathien S, Tesnière C, Meloche S. Regulation of Mitogen-Activated Protein Kinase Signaling Pathways by the Ubiquitin-Proteasome System and Its Pharmacological Potential. Pharmacol Rev 2021; 73:263-296. [PMID: 34732541 DOI: 10.1124/pharmrev.120.000170] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Mitogen-activated protein kinase (MAPK) cascades are evolutionarily conserved signaling pathways that play essential roles in transducing extracellular environmental signals into diverse cellular responses to maintain homeostasis. These pathways are classically organized into an architecture of three sequentially acting protein kinases: a MAPK kinase kinase that phosphorylates and activates a MAPK kinase, which in turn phosphorylates and activates the effector MAPK. The activity of MAPKs is tightly regulated by phosphorylation of their activation loop, which can be modulated by positive and negative feedback mechanisms to control the amplitude and duration of the signal. The signaling outcomes of MAPK pathways are further regulated by interactions of MAPKs with scaffolding and regulatory proteins. Accumulating evidence indicates that, in addition to these mechanisms, MAPK signaling is commonly regulated by ubiquitin-proteasome system (UPS)-mediated control of the stability and abundance of MAPK pathway components. Notably, the biologic activity of some MAPKs appears to be regulated mainly at the level of protein turnover. Recent studies have started to explore the potential of targeted protein degradation as a powerful strategy to investigate the biologic functions of individual MAPK pathway components and as a new therapeutic approach to overcome resistance to current small-molecule kinase inhibitors. Here, we comprehensively review the mechanisms, physiologic importance, and pharmacological potential of UPS-mediated protein degradation in the control of MAPK signaling. SIGNIFICANCE STATEMENT: Accumulating evidence highlights the importance of targeted protein degradation by the ubiquitin-proteasome system in regulating and fine-tuning the signaling output of mitogen-activated protein kinase (MAPK) pathways. Manipulating protein levels of MAPK cascade components may provide a novel approach for the development of selective pharmacological tools and therapeutics.
Collapse
Affiliation(s)
- Simon Mathien
- Institute for Research in Immunology and Cancer, Montreal, Quebec, Canada (S.Ma., C.T., S.Me.); and Molecular Biology Program, Faculty of Medicine (C.T., S.Me.) and Department of Pharmacology and Physiology (S.Me.), Université de Montréal, Montreal, Quebec, Canada
| | - Chloé Tesnière
- Institute for Research in Immunology and Cancer, Montreal, Quebec, Canada (S.Ma., C.T., S.Me.); and Molecular Biology Program, Faculty of Medicine (C.T., S.Me.) and Department of Pharmacology and Physiology (S.Me.), Université de Montréal, Montreal, Quebec, Canada
| | - Sylvain Meloche
- Institute for Research in Immunology and Cancer, Montreal, Quebec, Canada (S.Ma., C.T., S.Me.); and Molecular Biology Program, Faculty of Medicine (C.T., S.Me.) and Department of Pharmacology and Physiology (S.Me.), Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
14
|
Osaku D, Oishi T, Kawamura N, Iida Y, Komatsu H, Kudoh A, Chikumi J, Sato S, Harada T. Differential expression of estrogen receptor subtypes in ovarian high-grade serous carcinoma and clear cell carcinoma. Reprod Med Biol 2021; 20:467-476. [PMID: 34646075 PMCID: PMC8499597 DOI: 10.1002/rmb2.12402] [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: 05/29/2021] [Revised: 06/14/2021] [Accepted: 06/20/2021] [Indexed: 11/09/2022] Open
Abstract
PURPOSE To investigate the role of estrogen receptors (ERs) in high-grade serous carcinoma (HGSC) and clear cell carcinoma (CCC) of the ovary and evaluate ERs as prognostic biomarkers for ovarian cancer. METHODS This study included 79 patients with HGSC (n = 38) or CCC (n = 41) treated at our institution between 2005 and 2014. Immunohistochemistry examined protein expression of ERα, ERβ, and G protein-coupled estrogen receptor-1 (GPER-1); relationships between ERα, ERβ, and GPER-1 with patient survival were evaluated. Additionally, cell proliferation assay and phosphokinase proteome profiling were performed. RESULTS In HGSC patients, expression of ERα, cytoplasmic GPER-1, or nuclear GPER-1 was associated with poor progression-free survival (PFS) (P = .041, P = .010, or P = .013, respectively). Cytoplasmic GPER-1 was an independent prognostic factor for PFS in HGSC patients (HR = 2.83, 95% CI = 1.03-9.16, P = .007). ER expressions were not associated with prognosis in CCC patients. GPER-1 knockdown by siRNA reduced the cells number to 60% of siRNA-control-treated cells (P < .05), and GPER-1 antagonist, G-15 inhibited two HGSC cell lines proliferation (KF and UWB1.289) in a dose-dependent manner. Phosphoprotein array revealed that GPER-1 silencing decreased relative phosphorylation of glycogen synthase kinase-3. CONCLUSIONS High GPER-1 expression is an independent prognostic factor for PFS in HGSC patients, and GPER-1 may play a role in HGSC cell proliferation.
Collapse
Affiliation(s)
- Daiken Osaku
- Department of Obstetrics and GynecologyTottori University School of MedicineYonagoJapan
| | - Tetsuro Oishi
- Department of Obstetrics and GynecologyTottori University School of MedicineYonagoJapan
| | - Naoshi Kawamura
- Department of Obstetrics and GynecologyTottori University School of MedicineYonagoJapan
| | - Yuki Iida
- Department of Obstetrics and GynecologyTottori University School of MedicineYonagoJapan
| | - Hiroaki Komatsu
- Department of Obstetrics and GynecologyTottori University School of MedicineYonagoJapan
| | - Akiko Kudoh
- Department of Obstetrics and GynecologyTottori University School of MedicineYonagoJapan
| | - Jun Chikumi
- Department of Obstetrics and GynecologyTottori University School of MedicineYonagoJapan
| | - Shinya Sato
- Department of Obstetrics and GynecologyTottori University School of MedicineYonagoJapan
| | - Tasuku Harada
- Department of Obstetrics and GynecologyTottori University School of MedicineYonagoJapan
| |
Collapse
|
15
|
Silke J, O’Reilly LA. NF-κB and Pancreatic Cancer; Chapter and Verse. Cancers (Basel) 2021; 13:4510. [PMID: 34572737 PMCID: PMC8469693 DOI: 10.3390/cancers13184510] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 02/07/2023] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is one of the world's most lethal cancers. An increase in occurrence, coupled with, presently limited treatment options, necessitates the pursuit of new therapeutic approaches. Many human cancers, including PDAC are initiated by unresolved inflammation. The transcription factor NF-κB coordinates many signals that drive cellular activation and proliferation during immunity but also those involved in inflammation and autophagy which may instigate tumorigenesis. It is not surprising therefore, that activation of canonical and non-canonical NF-κB pathways is increasingly recognized as an important driver of pancreatic injury, progression to tumorigenesis and drug resistance. Paradoxically, NF-κB dysregulation has also been shown to inhibit pancreatic inflammation and pancreatic cancer, depending on the context. A pro-oncogenic or pro-suppressive role for individual components of the NF-κB pathway appears to be cell type, microenvironment and even stage dependent. This review provides an outline of NF-κB signaling, focusing on the role of the various NF-κB family members in the evolving inflammatory PDAC microenvironment. Finally, we discuss pharmacological control of NF-κB to curb inflammation, focussing on novel anti-cancer agents which reinstate the process of cancer cell death, the Smac mimetics and their pre-clinical and early clinical trials.
Collapse
Affiliation(s)
- John Silke
- Inflammation Division, Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC 3052, Australia;
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Lorraine Ann O’Reilly
- Inflammation Division, Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC 3052, Australia;
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
16
|
Pecoraro C, Faggion B, Balboni B, Carbone D, Peters GJ, Diana P, Assaraf YG, Giovannetti E. GSK3β as a novel promising target to overcome chemoresistance in pancreatic cancer. Drug Resist Updat 2021; 58:100779. [PMID: 34461526 DOI: 10.1016/j.drup.2021.100779] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/02/2021] [Accepted: 08/09/2021] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer is an aggressive malignancy with increasing incidence and poor prognosis due to its late diagnosis and intrinsic chemoresistance. Most pancreatic cancer patients present with locally advanced or metastatic disease characterized by inherent resistance to chemotherapy. These features pose a series of therapeutic challenges and new targets are urgently needed. Glycogen synthase kinase 3 beta (GSK3β) is a conserved serine/threonine kinase, which regulates key cellular processes including cell proliferation, DNA repair, cell cycle progression, signaling and metabolic pathways. GSK3β is implicated in non-malignant and malignant diseases including inflammation, neurodegenerative diseases, diabetes and cancer. GSK3β recently emerged among the key factors involved in the onset and progression of pancreatic cancer, as well as in the acquisition of chemoresistance. Intensive research has been conducted on key oncogenic functions of GSK3β and its potential as a druggable target; currently developed GSK3β inhibitors display promising results in preclinical models of distinct tumor types, including pancreatic cancer. Here, we review the latest findings about GSK-3β biology and its role in the development and progression of pancreatic cancer. Moreover, we discuss therapeutic agents targeting GSK3β that could be administered as monotherapy or in combination with other drugs to surmount chemoresistance. Several studies are also defining potential gene signatures to identify patients who might benefit from GSK3β-based therapeutic intervention. This detailed overview emphasizes the urgent need of additional molecular studies on the impact of GSK3β inhibition as well as structural analysis of novel compounds and omics studies of predictive biomarkers.
Collapse
Affiliation(s)
- Camilla Pecoraro
- Department of Medical Oncology, Amsterdam University Medical Center, VU University, 1081 HV Amsterdam, the Netherlands; Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy
| | - Beatrice Faggion
- Department of Medical Oncology, Amsterdam University Medical Center, VU University, 1081 HV Amsterdam, the Netherlands
| | - Beatrice Balboni
- Department of Medical Oncology, Amsterdam University Medical Center, VU University, 1081 HV Amsterdam, the Netherlands; Computational and Chemical Biology, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy, and Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Daniela Carbone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy
| | - Godefridus J Peters
- Department of Medical Oncology, Amsterdam University Medical Center, VU University, 1081 HV Amsterdam, the Netherlands; Department of Biochemistry, Medical University of Gdansk, Poland
| | - Patrizia Diana
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Elisa Giovannetti
- Department of Medical Oncology, Amsterdam University Medical Center, VU University, 1081 HV Amsterdam, the Netherlands; Cancer Pharmacology Lab, Fondazione Pisana per la Scienza, Via Ferruccio Giovannini 13, 56017 San Giuliano Terme (Pisa), Italy.
| |
Collapse
|
17
|
Important role of Nfkb2 in the Kras G12D-driven carcinogenesis in the pancreas. Pancreatology 2021; 21:912-919. [PMID: 33824054 DOI: 10.1016/j.pan.2021.03.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/11/2021] [Accepted: 03/17/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Oncogenic Kras initiates and drives carcinogenesis in the pancreas by complex signaling networks, including activation of the NFκB pathway. Although recent evidence has shown that oncogenic gains in Nfκb2 collaborate with Kras in the carcinogenesis, no data at the level of genetics for the contribution of Nfκb2 is available so far. METHODS We used Nfkb2 knock-out mice to decipher the role of the gene in Kras-driven carcinogenesis in vivo. RESULTS We show that the Nfkb2 gene is needed for cancer initiation and progression in KrasG12D-driven models and this requirement of Nfkb2 is mechanistically connected to proliferative pathways. In contrast, Nfκb2 is dispensable in aggressive pancreatic ductal adenocarcinoma (PDAC) models relying on the simultaneous expression of the Kras oncogene and the mutated tumor suppressor p53. CONCLUSIONS Our data add to the understanding of context-dependent requirements of oncogenic Kras signaling during pancreatic carcinogenesis.
Collapse
|
18
|
Hibino S, Kawazoe T, Kasahara H, Itoh S, Ishimoto T, Sakata-Yanagimoto M, Taniguchi K. Inflammation-Induced Tumorigenesis and Metastasis. Int J Mol Sci 2021; 22:ijms22115421. [PMID: 34063828 PMCID: PMC8196678 DOI: 10.3390/ijms22115421] [Citation(s) in RCA: 147] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 02/07/2023] Open
Abstract
Inflammation, especially chronic inflammation, plays a pivotal role in tumorigenesis and metastasis through various mechanisms and is now recognized as a hallmark of cancer and an attractive therapeutic target in cancer. In this review, we discuss recent advances in molecular mechanisms of how inflammation promotes tumorigenesis and metastasis and suppresses anti-tumor immunity in various types of solid tumors, including esophageal, gastric, colorectal, liver, and pancreatic cancer as well as hematopoietic malignancies.
Collapse
Affiliation(s)
- Sana Hibino
- Research Center for Advanced Science and Technology, Department of Inflammology, The University of Tokyo, Tokyo 153-0041, Japan;
| | - Tetsuro Kawazoe
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan;
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan;
| | - Hidenori Kasahara
- National Center for Global Health and Medicine, Department of Stem Cell Biology, Research Institute, Tokyo 162-8655, Japan;
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Shinji Itoh
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan;
| | - Takatsugu Ishimoto
- Gastrointestinal Cancer Biology, International Research Center of Medical Sciences (IRCMS), Kumamoto University, Kumamoto 860-0811, Japan;
| | | | - Koji Taniguchi
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan;
- Department of Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
- Correspondence: ; Tel.: +81-11-706-5050
| |
Collapse
|
19
|
TAK1 signaling is a potential therapeutic target for pathological angiogenesis. Angiogenesis 2021; 24:453-470. [PMID: 33973075 DOI: 10.1007/s10456-021-09787-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 03/29/2021] [Indexed: 02/07/2023]
Abstract
Angiogenesis plays a critical role in both physiological responses and disease pathogenesis. Excessive angiogenesis can promote neoplastic diseases and retinopathies, while inadequate angiogenesis can lead to aberrant perfusion and impaired wound healing. Transforming growth factor β activated kinase 1 (TAK1), a member of the mitogen-activated protein kinase kinase kinase family, is a key modulator involved in a range of cellular functions including the immune responses, cell survival and death. TAK1 is activated in response to various stimuli such as proinflammatory cytokines, hypoxia, and oxidative stress. Emerging evidence has recently suggested that TAK1 is intimately involved in angiogenesis and mediates pathogenic processes related to angiogenesis. Several detailed mechanisms by which TAK1 regulates pathological angiogenesis have been clarified, and potential therapeutics targeting TAK1 have emerged. In this review, we summarize recent studies of TAK1 in angiogenesis and discuss the crosstalk between TAK1 and signaling pathways involved in pathological angiogenesis. We also discuss the approaches for selectively targeting TAK1 and highlight the rationales of therapeutic strategies based on TAK1 inhibition for the treatment of pathological angiogenesis.
Collapse
|
20
|
Xia S, Ji L, Tao L, Pan Y, Lin Z, Wan Z, Pan H, Zhao J, Cai L, Xu J, Cai X. TAK1 Is a Novel Target in Hepatocellular Carcinoma and Contributes to Sorafenib Resistance. Cell Mol Gastroenterol Hepatol 2021; 12:1121-1143. [PMID: 33962073 PMCID: PMC8350196 DOI: 10.1016/j.jcmgh.2021.04.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Identifying novel and actionable targets in hepatocellular carcinoma (HCC) remains an unmet medical need. TAK1 was originally identified as a transforming growth factor-β-activated kinase and was further proved to phosphorylate and activate numerous downstream targets and promote cancer progression. However, the role of TAK1 in developed HCC progression and targeted therapy resistance is poorly understood. METHODS The expression of TAK1 or MTDH in HCC cell lines, tumor tissues, and sorafenib-resistant models was analyzed by in silico analysis, quantitative real-time polymerase chain reaction, Western blotting, and immunohistochemistry. In vivo and in vitro experiments were introduced to examine the function of TAK1 or MTDH in HCC and sorafenib resistance using small interfering RNA and pharmacologic inhibitors in combination with or without sorafenib. Co-immunoprecipitation and RNA immunoprecipitation were carried out to determine the binding between TAK1 and FBXW2 or between MTDH and FBXW2 mRNA. Protein half-life and in vitro ubiquitination experiment was performed to validate whether FBXW2 regulates TAK1 degradation. RESULTS Our findings unraveled the clinical significance of TAK1 in promoting HCC and sorafenib resistance. We identified a novel E3 ubiquitin ligase, FBXW2, targeting TAK1 for K48-linked polyubiquitylation and subsequent degradation. We also found that MTDH contributes to TAK1 up-regulation in HCC and sorafenib resistance through binding to FBXW2 mRNA and accelerates its degradation. Moreover, combination of TAK1 inhibitor and sorafenib suppressed the growth of sorafenib-resistant HCCLM3 xenograft in mouse models. CONCLUSIONS These results revealed novel mechanism underlying TAK1 protein degradation and highlighted the therapeutic value of targeting TAK1 in suppressing HCC and overcoming sorafenib resistance.
Collapse
Affiliation(s)
- Shunjie Xia
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, China; Zhejiang University Cancer Center, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
| | - Lin Ji
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, China; Zhejiang University Cancer Center, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
| | - Liye Tao
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, China; Zhejiang University Cancer Center, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
| | - Yu Pan
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, China; Zhejiang University Cancer Center, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
| | - Zhongjie Lin
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, China; Zhejiang University Cancer Center, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
| | - Zhe Wan
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, China; Zhejiang University Cancer Center, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
| | - Haoqi Pan
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, China; Zhejiang University Cancer Center, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
| | - Jie Zhao
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, China; Zhejiang University Cancer Center, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
| | - Liuxin Cai
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, China; Zhejiang University Cancer Center, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
| | - Junjie Xu
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, China; Zhejiang University Cancer Center, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China.
| | - Xiujun Cai
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, China; Zhejiang University Cancer Center, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China.
| |
Collapse
|
21
|
GSK-3β Can Regulate the Sensitivity of MIA-PaCa-2 Pancreatic and MCF-7 Breast Cancer Cells to Chemotherapeutic Drugs, Targeted Therapeutics and Nutraceuticals. Cells 2021; 10:cells10040816. [PMID: 33917370 PMCID: PMC8067414 DOI: 10.3390/cells10040816] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/04/2021] [Accepted: 04/04/2021] [Indexed: 02/06/2023] Open
Abstract
Glycogen synthase kinase-3 (GSK-3) is a regulator of signaling pathways. KRas is frequently mutated in pancreatic cancers. The growth of certain pancreatic cancers is KRas-dependent and can be suppressed by GSK-3 inhibitors, documenting a link between KRas and GSK-3. To further elucidate the roles of GSK-3β in drug-resistance, we transfected KRas-dependent MIA-PaCa-2 pancreatic cells with wild-type (WT) and kinase-dead (KD) forms of GSK-3β. Transfection of MIA-PaCa-2 cells with WT-GSK-3β increased their resistance to various chemotherapeutic drugs and certain small molecule inhibitors. Transfection of cells with KD-GSK-3β often increased therapeutic sensitivity. An exception was observed with cells transfected with WT-GSK-3β and sensitivity to the BCL2/BCLXL ABT737 inhibitor. WT-GSK-3β reduced glycolytic capacity of the cells but did not affect the basal glycolysis and mitochondrial respiration. KD-GSK-3β decreased both basal glycolysis and glycolytic capacity and reduced mitochondrial respiration in MIA-PaCa-2 cells. As a comparison, the effects of GSK-3 on MCF-7 breast cancer cells, which have mutant PIK3CA, were examined. KD-GSK-3β increased the resistance of MCF-7 cells to chemotherapeutic drugs and certain signal transduction inhibitors. Thus, altering the levels of GSK-3β can have dramatic effects on sensitivity to drugs and signal transduction inhibitors which may be influenced by the background of the tumor.
Collapse
|
22
|
Silva-García O, Cortés-Vieyra R, Mendoza-Ambrosio FN, Ramírez-Galicia G, Baizabal-Aguirre VM. GSK3α: An Important Paralog in Neurodegenerative Disorders and Cancer. Biomolecules 2020; 10:E1683. [PMID: 33339170 PMCID: PMC7765659 DOI: 10.3390/biom10121683] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 11/29/2020] [Accepted: 11/30/2020] [Indexed: 12/18/2022] Open
Abstract
The biological activity of the enzyme glycogen synthase kinase-3 (GSK3) is fulfilled by two paralogs named GSK3α and GSK3β, which possess both redundancy and specific functions. The upregulated activity of these proteins is linked to the development of disorders such as neurodegenerative disorders (ND) and cancer. Although various chemical inhibitors of these enzymes restore the brain functions in models of ND such as Alzheimer's disease (AD), and reduce the proliferation and survival of cancer cells, the particular contribution of each paralog to these effects remains unclear as these molecules downregulate the activity of both paralogs with a similar efficacy. Moreover, given that GSK3 paralogs phosphorylate more than 100 substrates, the simultaneous inhibition of both enzymes has detrimental effects during long-term inhibition. Although the GSK3β kinase function has usually been taken as the global GSK3 activity, in the last few years, a growing interest in the study of GSK3α has emerged because several studies have recognized it as the main GSK3 paralog involved in a variety of diseases. This review summarizes the current biological evidence on the role of GSK3α in AD and various types of cancer. We also provide a discussion on some strategies that may lead to the design of the paralog-specific inhibition of GSK3α.
Collapse
Affiliation(s)
- Octavio Silva-García
- Departamento de Química Teórica, Universidad del Papaloapan, Oaxaca 68301, Mexico; (F.N.M.-A.); (G.R.-G.)
| | - Ricarda Cortés-Vieyra
- Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Michoacán 58000, Mexico;
| | | | - Guillermo Ramírez-Galicia
- Departamento de Química Teórica, Universidad del Papaloapan, Oaxaca 68301, Mexico; (F.N.M.-A.); (G.R.-G.)
| | - Víctor M. Baizabal-Aguirre
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, Michoacán 58893, Mexico
| |
Collapse
|
23
|
Khurana N, Dodhiawala PB, Bulle A, Lim KH. Deciphering the Role of Innate Immune NF-ĸB Pathway in Pancreatic Cancer. Cancers (Basel) 2020; 12:cancers12092675. [PMID: 32961746 PMCID: PMC7564842 DOI: 10.3390/cancers12092675] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Chronic inflammation is a major mechanism that underlies the aggressive nature and treatment resistance of pancreatic cancer. In many ways, the molecular mechanisms that drive chronic inflammation in pancreatic cancer are very similar to our body’s normal innate immune response to injury or invading microorganisms. Therefore, during cancer development, pancreatic cancer cells hijack the innate immune pathway to foster a chronically inflamed tumor environment that helps shield them from immune attack and therapeutics. While blocking the innate immune pathway is theoretically reasonable, untoward side effects must also be addressed. In this review, we comprehensively summarize the literature that describe the role of innate immune signaling in pancreatic cancer, emphasizing the specific role of this pathway in different cell types. We review the interaction of the innate immune pathway and cancer-driving signaling in pancreatic cancer and provide an updated overview of novel therapeutic opportunities against this mechanism. Abstract Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers with no effective treatment option. A predominant hallmark of PDAC is the intense fibro-inflammatory stroma which not only physically collapses vasculature but also functionally suppresses anti-tumor immunity. Constitutive and induced activation of the NF-κB transcription factors is a major mechanism that drives inflammation in PDAC. While targeting this pathway is widely supported as a promising therapeutic strategy, clinical success is elusive due to a lack of safe and effective anti-NF-κB pathway therapeutics. Furthermore, the cell type-specific contribution of this pathway, specifically in neoplastic cells, stromal fibroblasts, and immune cells, has not been critically appraised. In this article, we highlighted seminal and recent literature on molecular mechanisms that drive NF-κB activity in each of these major cell types in PDAC, focusing specifically on the innate immune Toll-like/IL-1 receptor pathway. We reviewed recent evidence on the signaling interplay between the NF-κB and oncogenic KRAS signaling pathways in PDAC cells and their collective contribution to cancer inflammation. Lastly, we reviewed clinical trials on agents that target the NF-κB pathway and novel therapeutic strategies that have been proposed in preclinical studies.
Collapse
Affiliation(s)
- Namrata Khurana
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Paarth B Dodhiawala
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ashenafi Bulle
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kian-Huat Lim
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
24
|
Glycogen Synthase Kinase 3β in Cancer Biology and Treatment. Cells 2020; 9:cells9061388. [PMID: 32503133 PMCID: PMC7349761 DOI: 10.3390/cells9061388] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/28/2020] [Accepted: 06/01/2020] [Indexed: 12/15/2022] Open
Abstract
Glycogen synthase kinase (GSK)3β is a multifunctional serine/threonine protein kinase with more than 100 substrates and interacting molecules. GSK3β is normally active in cells and negative regulation of GSK3β activity via phosphorylation of its serine 9 residue is required for most normal cells to maintain homeostasis. Aberrant expression and activity of GSK3β contributes to the pathogenesis and progression of common recalcitrant diseases such as glucose intolerance, neurodegenerative disorders and cancer. Despite recognized roles against several proto-oncoproteins and mediators of the epithelial–mesenchymal transition, deregulated GSK3β also participates in tumor cell survival, evasion of apoptosis, proliferation and invasion, as well as sustaining cancer stemness and inducing therapy resistance. A therapeutic effect from GSK3β inhibition has been demonstrated in 25 different cancer types. Moreover, there is increasing evidence that GSK3β inhibition protects normal cells and tissues from the harmful effects associated with conventional cancer therapies. Here, we review the evidence supporting aberrant GSK3β as a hallmark property of cancer and highlight the beneficial effects of GSK3β inhibition on normal cells and tissues during cancer therapy. The biological rationale for targeting GSK3β in the treatment of cancer is also discussed at length.
Collapse
|
25
|
Ding L, Billadeau DD. Glycogen synthase kinase-3β: a novel therapeutic target for pancreatic cancer. Expert Opin Ther Targets 2020; 24:417-426. [PMID: 32178549 DOI: 10.1080/14728222.2020.1743681] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of cancer death in the United States with a single-digit 5-year survival rate despite advances in understanding the genetics and biology of the disease. Glycogen synthase kinase-3α (GSK-3α) and GSK-3β are serine/threonine kinases that localize to the cytoplasm, mitochondria and nucleus. Although they are highly homologous within their kinase domains and phosphorylate an overlapping set of target proteins, genetic studies have shown that GSK-3β regulates the activity of several proteins that promote neoplastic transformation. Significantly, GSK-3β is progressively overexpressed during PDAC development where it participates in tumor progression, survival and chemoresistance. Thus, GSK-3β has become an attractive target for treating PDAC.Areas covered: This review summarizes the mechanisms regulating GSK-3β activity, including upstream translational and post-translational regulation, as well as the downstream targets and their functions in PDAC cell growth, metastasis and chemoresistance.Expert opinion: The activity of GSK-3 kinases are considered cell- and context-specific. In PDAC, oncogenic KRas drives the transcriptional expression of the GSK-3β gene, which has been shown to regulate cancer cell proliferation and survival, as well as resistance to chemotherapy. Thus, the combination of GSK-3 inhibitors with chemotherapeutic drugs could be a promising strategy for PDAC.
Collapse
Affiliation(s)
- Li Ding
- The Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Daniel D Billadeau
- The Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
26
|
Ren X, Chen C, Luo Y, Liu M, Li Y, Zheng S, Ye H, Fu Z, Li M, Li Z, Chen R. lncRNA-PLACT1 sustains activation of NF-κB pathway through a positive feedback loop with IκBα/E2F1 axis in pancreatic cancer. Mol Cancer 2020; 19:35. [PMID: 32085715 PMCID: PMC7033942 DOI: 10.1186/s12943-020-01153-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 02/13/2020] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND The activation of NF-κB signaling pathway is regarded as the dominant process that correlates with tumorigenesis. Recently, increasing evidence shows that long noncoding RNAs (lncRNAs) play crucial roles in sustaining the NF-κB signaling pathway. However, the underlying mechanisms have not yet been elucidated. METHODS The expression and clinical features of PLACT1 were analyzed in a 166-case cohort of PDAC by qRT-PCR and in situ hybridization. The functional role of PLACT1 was evaluated by both in vitro and in vivo experiments. Chromatin isolation by RNA purification assays were utilized to examine the interaction of PLACT1 with IκBα promoter. RESULTS We identified a novel lncRNA-PLACT1, which was significantly upregulated in tumor tissues and correlated with progression and poor survival in PDAC patients. Moreover, PLACT1 promoted the proliferation and invasion of PDAC cells in vitro. Consistently, PLACT1 overexpression fostered the progression of PDAC both in orthotopic and lung metastasis mice models. Mechanistically, PLACT1 suppressed IκBα expression by recruiting hnRNPA1 to IκBα promoter, which led to increased H3K27me3 that decreased the transcriptional level of IκBα. Furthermore, E2F1-mediated overexpression of PLACT1 modulated the progression of PDAC by sustained activation of NF-κB signaling pathway through forming a positive feedback loop with IκBα. Importantly, administration of the NF-κB signaling pathway inhibitor significantly suppressed PLACT1-induced sustained activation of NF-κB signaling pathway, leading to reduced tumorigenesis in vivo. CONCLUSIONS Our findings suggest that PLACT1 provides a novel epigenetic mechanism involved in constitutive activation of NF-κB signaling pathway and may represent a new therapeutic target of PDAC.
Collapse
Affiliation(s)
- Xiaofan Ren
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, 107th Yanjiangxi Road, Yuexiu District, Guangzhou, Guangdong province, 510120, People's Republic of China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, 107th Yanjiangxi Road, Yuexiu District, Guangzhou, 510120, Guangdong province, People's Republic of China
| | - Changhao Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, 107th Yanjiangxi Road, Yuexiu District, Guangzhou, 510120, Guangdong province, People's Republic of China. .,Department of Urology, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, 107th Yanjiangxi Road, Yuexiu District, Guangzhou, Guangdong province, 510120, People's Republic of China.
| | - Yuming Luo
- Department of Hepatopancreatobiliary Surgery, Sun Yat-sen Memorial Hospital, 107th Yanjiangxi Road, Yuexiu District, Guangzhou, 510120, Guangdong province, People's Republic of China
| | - Mingyang Liu
- Department of Medicine, Department of Surgery, the University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1262A, Oklahoma City, OK, 73104, USA
| | - Yuting Li
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, 107th Yanjiangxi Road, Yuexiu District, Guangzhou, Guangdong province, 510120, People's Republic of China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, 107th Yanjiangxi Road, Yuexiu District, Guangzhou, 510120, Guangdong province, People's Republic of China
| | - Shangyou Zheng
- Department of Hepatopancreatobiliary Surgery, Sun Yat-sen Memorial Hospital, 107th Yanjiangxi Road, Yuexiu District, Guangzhou, 510120, Guangdong province, People's Republic of China
| | - Huilin Ye
- Department of Hepatopancreatobiliary Surgery, Sun Yat-sen Memorial Hospital, 107th Yanjiangxi Road, Yuexiu District, Guangzhou, 510120, Guangdong province, People's Republic of China
| | - Zhiqiang Fu
- Department of Hepatopancreatobiliary Surgery, Sun Yat-sen Memorial Hospital, 107th Yanjiangxi Road, Yuexiu District, Guangzhou, 510120, Guangdong province, People's Republic of China
| | - Min Li
- Department of Medicine, Department of Surgery, the University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1262A, Oklahoma City, OK, 73104, USA.
| | - Zhihua Li
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, 107th Yanjiangxi Road, Yuexiu District, Guangzhou, Guangdong province, 510120, People's Republic of China. .,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, 107th Yanjiangxi Road, Yuexiu District, Guangzhou, 510120, Guangdong province, People's Republic of China.
| | - Rufu Chen
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106th of 2nd Zhongshan Road, Yuexiu District, Guangzhou, Guangdong Province, 510080, People's Republic of China.
| |
Collapse
|
27
|
Santoro R, Zanotto M, Simionato F, Zecchetto C, Merz V, Cavallini C, Piro G, Sabbadini F, Boschi F, Scarpa A, Melisi D. Modulating TAK1 Expression Inhibits YAP and TAZ Oncogenic Functions in Pancreatic Cancer. Mol Cancer Ther 2019; 19:247-257. [PMID: 31562256 DOI: 10.1158/1535-7163.mct-19-0270] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 07/19/2019] [Accepted: 09/18/2019] [Indexed: 11/16/2022]
Abstract
YAP and TAZ are central determinants of malignancy; however, their functions remain still undruggable. We identified TGFβ-activated kinase 1 (TAK1) as a central hub integrating the most relevant signals sustaining pancreatic cancer aggressiveness and chemoresistance. Glycogen synthase kinase (GSK)3 is known to stabilize TAK1, and its inhibition causes a reduction in TAK1 levels. Here, we hypothesized that TAK1 could sustain YAP/TAZ program, and thus, modulation of TAK1 expression through the inhibition of GSK3 could impair YAP/TAZ functions in pancreatic cancer.Differentially expressed transcripts between pancreatic cancer cells expressing scramble or TAK1-specific shRNA were annotated for functional interrelatedness by ingenuity pathway analysis. TAK1 expression was modulated by using different GSK3 inhibitors, including LY2090314. In vivo activity of LY2090314 alone or in combination with nab-paclitaxel was evaluated in an orthotopic nude mouse model.Differential gene expression profiling revealed significant association of TAK1 expression with HIPPO and ubiquitination pathways. We measured a significant downregulation of YAP/TAZ and their regulated genes in shTAK1 cells. TAK1 prevented YAP/TAZ proteasomal degradation in a kinase independent manner, through a complex with TRAF6, thereby fostering their K63-ubiquitination versus K48-ubiquitination. Pharmacologic modulation of TAK1 by using GSK3 inhibitors significantly decreased YAP/TAZ levels and suppressed their target genes and oncogenic functions. In vivo, LY2090314 plus nab-paclitaxel significantly prolonged mice survival duration.Our study demonstrates a unique role for TAK1 in controlling YAP/TAZ in pancreatic cancer. LY2090314 is a novel agent that warrants further clinical development in combination with nab-paclitaxel for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Raffaela Santoro
- Digestive Molecular Clinical Oncology Research Unit, Department of Medicine, Università degli studi di Verona, Verona, Italy
| | - Marco Zanotto
- Digestive Molecular Clinical Oncology Research Unit, Department of Medicine, Università degli studi di Verona, Verona, Italy
| | - Francesca Simionato
- Medical Oncology Unit, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Camilla Zecchetto
- Medical Oncology Unit, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Valeria Merz
- Medical Oncology Unit, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Chiara Cavallini
- Research Center LURM, Interdepartmental Laboratory of Medical Research, Università degli studi di Verona, Verona, Italy
| | - Geny Piro
- Digestive Molecular Clinical Oncology Research Unit, Department of Medicine, Università degli studi di Verona, Verona, Italy
| | - Fabio Sabbadini
- Digestive Molecular Clinical Oncology Research Unit, Department of Medicine, Università degli studi di Verona, Verona, Italy
| | - Federico Boschi
- Department of Computer Science, Università degli studi di Verona, Verona, Italy
| | - Aldo Scarpa
- ARC-Net Research Centre, University and Hospital Trust of Verona and Department of Diagnostics and Public Health, Section of Anatomical Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Davide Melisi
- Digestive Molecular Clinical Oncology Research Unit, Department of Medicine, Università degli studi di Verona, Verona, Italy. .,Medical Oncology Unit, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| |
Collapse
|
28
|
Wagner FF, Benajiba L, Campbell AJ, Weïwer M, Sacher JR, Gale JP, Ross L, Puissant A, Alexe G, Conway A, Back M, Pikman Y, Galinsky I, DeAngelo DJ, Stone RM, Kaya T, Shi X, Robers MB, Machleidt T, Wilkinson J, Hermine O, Kung A, Stein AJ, Lakshminarasimhan D, Hemann MT, Scolnick E, Zhang YL, Pan JQ, Stegmaier K, Holson EB. Exploiting an Asp-Glu "switch" in glycogen synthase kinase 3 to design paralog-selective inhibitors for use in acute myeloid leukemia. Sci Transl Med 2019. [PMID: 29515000 DOI: 10.1126/scitranslmed.aam8460] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Glycogen synthase kinase 3 (GSK3), a key regulatory kinase in the wingless-type MMTV integration site family (WNT) pathway, is a therapeutic target of interest in many diseases. Although dual GSK3α/β inhibitors have entered clinical trials, none has successfully translated to clinical application. Mechanism-based toxicities, driven in part by the inhibition of both GSK3 paralogs and subsequent β-catenin stabilization, are a concern in the translation of this target class because mutations and overexpression of β-catenin are associated with many cancers. Knockdown of GSK3α or GSK3β individually does not increase β-catenin and offers a conceptual resolution to targeting GSK3: paralog-selective inhibition. However, inadequate chemical tools exist. The design of selective adenosine triphosphate (ATP)-competitive inhibitors poses a drug discovery challenge due to the high homology (95% identity and 100% similarity) in this binding domain. Taking advantage of an Asp133→Glu196 "switch" in their kinase hinge, we present a rational design strategy toward the discovery of paralog-selective GSK3 inhibitors. These GSK3α- and GSK3β-selective inhibitors provide insights into GSK3 targeting in acute myeloid leukemia (AML), where GSK3α was identified as a therapeutic target using genetic approaches. The GSK3α-selective compound BRD0705 inhibits kinase function and does not stabilize β-catenin, mitigating potential neoplastic concerns. BRD0705 induces myeloid differentiation and impairs colony formation in AML cells, with no apparent effect on normal hematopoietic cells. Moreover, BRD0705 impairs leukemia initiation and prolongs survival in AML mouse models. These studies demonstrate feasibility of paralog-selective GSK3α inhibition, offering a promising therapeutic approach in AML.
Collapse
Affiliation(s)
- Florence F Wagner
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA.
| | - Lina Benajiba
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA.,Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA.,INSERM U1163 and CNRS 8254, Imagine Institute, Université Paris Saclay, 91190 Paris, France
| | - Arthur J Campbell
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - Michel Weïwer
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - Joshua R Sacher
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - Jennifer P Gale
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - Linda Ross
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Alexandre Puissant
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA.,INSERM U944, Institute of Hematology, St. Louis Hospital, 75010 Paris, France
| | - Gabriela Alexe
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA.,Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA.,Bioinformatics Graduate Program, Boston University, Boston, MA 02215, USA
| | - Amy Conway
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Morgan Back
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Yana Pikman
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA.,Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Ilene Galinsky
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Daniel J DeAngelo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Richard M Stone
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Taner Kaya
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - Xi Shi
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - Matthew B Robers
- Promega Corporation, 2800 Woods Hollow Road, Madison, WI 53711, USA
| | - Thomas Machleidt
- Promega Corporation, 2800 Woods Hollow Road, Madison, WI 53711, USA
| | | | - Olivier Hermine
- INSERM U1163 and CNRS 8254, Imagine Institute, Université Sorbonne Paris Cité, Paris, France.,Department of Hematology, Hôpital Necker, Assistance Publique Hôpitaux de Paris, University Paris Descartes, 75006 Paris, France
| | - Andrew Kung
- Department of Pediatrics, Columbia University Medical Center, New York, NY 10032, USA
| | | | | | - Michael T Hemann
- Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Edward Scolnick
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - Yan-Ling Zhang
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - Jen Q Pan
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - Kimberly Stegmaier
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA. .,Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Edward B Holson
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| |
Collapse
|
29
|
Kabacaoglu D, Ruess DA, Ai J, Algül H. NF-κB/Rel Transcription Factors in Pancreatic Cancer: Focusing on RelA, c-Rel, and RelB. Cancers (Basel) 2019; 11:E937. [PMID: 31277415 PMCID: PMC6679104 DOI: 10.3390/cancers11070937] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 06/26/2019] [Accepted: 07/02/2019] [Indexed: 02/07/2023] Open
Abstract
Regulation of Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB)/Rel transcription factors (TFs) is extremely cell-type-specific owing to their ability to act disparately in the context of cellular homeostasis driven by cellular fate and the microenvironment. This is also valid for tumor cells in which every single component shows heterogenic effects. Whereas many studies highlighted a per se oncogenic function for NF-κB/Rel TFs across cancers, recent advances in the field revealed their additional tumor-suppressive nature. Specifically, pancreatic ductal adenocarcinoma (PDAC), as one of the deadliest malignant diseases, shows aberrant canonical-noncanonical NF-κB signaling activity. Although decades of work suggest a prominent oncogenic activity of NF-κB signaling in PDAC, emerging evidence points to the opposite including anti-tumor effects. Considering the dual nature of NF-κB signaling and how it is closely linked to many other cancer related signaling pathways, it is essential to dissect the roles of individual Rel TFs in pancreatic carcinogenesis and tumor persistency and progression. Here, we discuss recent knowledge highlighting the role of Rel TFs RelA, RelB, and c-Rel in PDAC development and maintenance. Next to providing rationales for therapeutically harnessing Rel TF function in PDAC, we compile strategies currently in (pre-)clinical evaluation.
Collapse
Affiliation(s)
- Derya Kabacaoglu
- Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Dietrich A Ruess
- Department of Surgery, Faculty of Medicine, Medical Center, University of Freiburg, 79106 Freiburg, Germany
| | - Jiaoyu Ai
- Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Hana Algül
- Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany.
| |
Collapse
|
30
|
Wang Y, Dou X, Jiang L, Jin H, Zhang L, Zhang L, Liu Z. Discovery of novel glycogen synthase kinase-3α inhibitors: Structure-based virtual screening, preliminary SAR and biological evaluation for treatment of acute myeloid leukemia. Eur J Med Chem 2019; 171:221-234. [PMID: 30925338 DOI: 10.1016/j.ejmech.2019.03.039] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 03/14/2019] [Accepted: 03/16/2019] [Indexed: 12/26/2022]
Abstract
Glycogen synthase kinase 3α (GSK-3α) plays a constitutive role in various physiological processes and has been proved to be a therapeutic target for acute myeloid leukemia (AML). In this paper, by means of computer-aided drug design, we discovered a novel chemical series of GSK-3α inhibitors with an IC50 value of 0.033-2.804 μM. The preliminary structure-activity relationship was concluded and, notably, the most potent and isoform-selective compound G28_14 was identified with IC50 values of 33 nM and 218 nM against GSK-3α and -3β, respectively, exhibiting a nearly ten-fold isoform-selectivity. Further cell viability assays and colony formation assays revealed that G28_14 suppressed cell survival by impairing cell proliferation by up to 90% in two AML cell lines. Moreover, surface marker expression analysis demonstrated that G28_14 induced terminal differentiation with a high level of CD11b, CD11c, and CD14. Western immunoblotting showed that G28_14 isoform-selectively inhibited the phosphorylation of GSK-3α in-cell without activating Wnt/β-catenin signaling. In addition, to elucidate its structure-activity relationship, the binding mode of this chemical series was proposed using molecular docking and molecular dynamics simulations. Taken together, this chemical series is worth developing as differentiation therapies for the treatment of AML.
Collapse
Affiliation(s)
- Yanxing Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Xiaodong Dou
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Lan Jiang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Hongwei Jin
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Lihe Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China.
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China.
| |
Collapse
|
31
|
Zhang N, Dou Y, Liu L, Zhang X, Liu X, Zeng Q, Liu Y, Yin M, Liu X, Deng H, Song D. SA-49, a novel aloperine derivative, induces MITF-dependent lysosomal degradation of PD-L1. EBioMedicine 2019; 40:151-162. [PMID: 30711516 PMCID: PMC6414307 DOI: 10.1016/j.ebiom.2019.01.054] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/25/2019] [Accepted: 01/25/2019] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Programmed death-ligand 1 (PD-L1) is a T-cell inhibitory checkpoint molecule that suppresses antitumor immunity. Anti-PD-L1 antibodies have shown remarkable promise in treating tumors, but the patient response rate is low. Therefore, small-molecule checkpoint inhibitors blocking PD-L1 function are urgently needed. METHODS Changes of protein expression and phosphorylation levels were determined by immunoblotting. The level of Membrane PD-L1 was examined by flow cytometer. Cytotoxicity of T cells and NK cells toward tumor cells were detected using LDH and cell index assays. Lysosome function was investigated by NAG assay. Changes in lysosomal-related genes were measured by RT-PCR. In vivo anti-NSCLC cancer effects were assessed using C57BL/6 mice bearing Lewis tumor xenografts. FINDINGS We identified SA-49 as a new regulator of PD-L1 expression from a series of novel aloperine derivatives. SA-49 decreased the expression of PD-L1 in NSCLC cells and enhanced the cytotoxicity of co-cultured T and NK cells toward tumor cells. Importantly, lysosomal pathway contributed to SA-49-mediated down-regulation of PD-L1. SA-49 increased the biogenesis of lysosome and promoted translocation of PD-L1 to lysosome for proteolysis, which was associated with nuclear translocation of MITF. SA-49-induced MITF translocation acted through activation of PKCα and subsequently suppression of GSK3β activity. Furthermore, SA-49 suppressed Lewis tumor xenograft growth by activating immune microenvironment in C57BL/6 mice. INTERPRETATION Our data demonstrate that SA-49 can be used to regulate PD-L1 in cancer cells and trigger its degradation by activating lysosome function.
Collapse
Affiliation(s)
- Na Zhang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yueying Dou
- Pharmacy Department, Hefei BOE Hospital Co., Ltd., Hefei 230011, China
| | - Lu Liu
- Qingdao Women and Children's Hospital, Qingdao University, Qingdao 266034, China
| | - Xin Zhang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xiaojia Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Qingxuan Zeng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yang Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Mingxiao Yin
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xiujun Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Hongbin Deng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| | - Danqing Song
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
32
|
Kuenzi BM, Remsing Rix LL, Kinose F, Kroeger JL, Lancet JE, Padron E, Rix U. Off-target based drug repurposing opportunities for tivantinib in acute myeloid leukemia. Sci Rep 2019; 9:606. [PMID: 30679640 PMCID: PMC6345777 DOI: 10.1038/s41598-018-37174-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 11/20/2018] [Indexed: 02/07/2023] Open
Abstract
GSK3α has been identified as a new target in the treatment of acute myeloid leukemia (AML). However, most GSK3 inhibitors lack specificity for GSK3α over GSK3β and other kinases. We have previously shown in lung cancer cells that GSK3α and to a lesser extent GSK3β are inhibited by the advanced clinical candidate tivantinib (ARQ197), which was designed as a MET inhibitor. Thus, we hypothesized that tivantinib would be an effective therapy for the treatment of AML. Here, we show that tivantinib has potent anticancer activity across several AML cell lines and primary patient cells. Tivantinib strongly induced apoptosis, differentiation and G2/M cell cycle arrest and caused less undesirable stabilization of β-catenin compared to the pan-GSK3 inhibitor LiCl. Subsequent drug combination studies identified the BCL-2 inhibitor ABT-199 to synergize with tivantinib while cytarabine combination with tivantinib was antagonistic. Interestingly, the addition of ABT-199 to tivantinib completely abrogated tivantinib induced β-catenin stabilization. Tivantinib alone, or in combination with ABT-199, downregulated anti-apoptotic MCL-1 and BCL-XL levels, which likely contribute to the observed synergy. Importantly, tivantinib as single agent or in combination with ABT-199 significantly inhibited the colony forming capacity of primary patient AML bone marrow mononuclear cells. In summary, tivantinib is a novel GSK3α/β inhibitor that potently kills AML cells and tivantinib single agent or combination therapy with ABT-199 may represent attractive new therapeutic opportunities for AML.
Collapse
Affiliation(s)
- Brent M Kuenzi
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, 33612, United States.,Cancer Biology Ph.D. Program, University of South Florida, Tampa, Florida, 33620, United States
| | - Lily L Remsing Rix
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, 33612, United States
| | - Fumi Kinose
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, 33612, United States
| | - Jodi L Kroeger
- Flow Cytometry Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, 33612, United States
| | - Jeffrey E Lancet
- Department of Hematologic Malignancies, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, 33612, United States
| | - Eric Padron
- Department of Hematologic Malignancies, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, 33612, United States
| | - Uwe Rix
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, 33612, United States.
| |
Collapse
|
33
|
Pramanik KC, Makena MR, Bhowmick K, Pandey MK. Advancement of NF-κB Signaling Pathway: A Novel Target in Pancreatic Cancer. Int J Mol Sci 2018; 19:ijms19123890. [PMID: 30563089 PMCID: PMC6320793 DOI: 10.3390/ijms19123890] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 11/21/2018] [Accepted: 11/29/2018] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers and is the third highest among cancer related deaths. Despite modest success with therapy such as gemcitabine, pancreatic cancer incidence remains virtually unchanged in the past 25 years. Among the several driver mutations for PDAC, Kras mutation contributes a central role for its development, progression and therapeutic resistance. In addition, inflammation is implicated in the development of most human cancer, including pancreatic cancer. Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) is recognized as a key mediator of inflammation and has been frequently observed to be upregulated in PDAC. Several lines of evidence suggest that NF-κB pathways play a crucial role in PDAC development, progression and resistance. In this review, we focused on emphasizing the recent advancements in the involvement of NF-κB in PADC’s progression and resistance. We also highlighted the interaction of NF-κB with other signaling pathways. Lastly, we also aim to discuss how NF-κB could be an excellent target for PDAC prevention or therapy. This review could provide insight into the development of novel therapeutic strategies by considering NF-κB as a target to prevent or treat PDAC.
Collapse
Affiliation(s)
- Kartick C Pramanik
- Department of Basic Sciences, Kentucky College of Osteopathic Medicine, University of Pikeville, Pikeville, KY 41501, USA.
| | - Monish Ram Makena
- Department of Physiology, The Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA.
| | - Kuntal Bhowmick
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA.
| | - Manoj K Pandey
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA.
| |
Collapse
|
34
|
GSK3 suppression upregulates β-catenin and c-Myc to abrogate KRas-dependent tumors. Nat Commun 2018; 9:5154. [PMID: 30514931 PMCID: PMC6279809 DOI: 10.1038/s41467-018-07644-6] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 11/15/2018] [Indexed: 01/05/2023] Open
Abstract
Mutant KRas is a significant driver of human oncogenesis and confers resistance to therapy, underscoring the need to develop approaches that disable mutant KRas-driven tumors. Because targeting KRas directly has proven difficult, identifying vulnerabilities specific for mutant KRas tumors is an important alternative approach. Here we show that glycogen synthase kinase 3 (GSK3) is required for the in vitro and in vivo growth and survival of human mutant KRas-dependent tumors but is dispensable for mutant KRas-independent tumors. Further, inhibiting phosphorylation of GSK3 substrates c-Myc on T58 and β-catenin on S33/S37/T41 and their subsequent upregulation contribute to the antitumor activity of GSK3 inhibition. Importantly, GSK3 blockade inhibits the in vivo growth of G12D, G12V, and G12C mutant KRas primary and metastatic patient-derived xenografts from pancreatic cancer patients who progressed on chemo- and radiation therapies. This discovery opens new avenues to target mutant KRas-dependent cancers. Direct targeting of mutant KRas is challenging and alternative approaches are needed. Here they show glycogen synthase kinase 3 (GSK3) to be required for the growth and survival of human mutant KRas-dependent tumors but dispensable for mutant KRas-independent tumors and show GSK3 inhibition to inhibit in vivo growth of Kras mutant patient-derived pancreatic tumors.
Collapse
|
35
|
Zoranovic T, Manent J, Willoughby L, Matos de Simoes R, La Marca JE, Golenkina S, Cuiping X, Gruber S, Angjeli B, Kanitz EE, Cronin SJF, Neely GG, Wernitznig A, Humbert PO, Simpson KJ, Mitsiades CS, Richardson HE, Penninger JM. A genome-wide Drosophila epithelial tumorigenesis screen identifies Tetraspanin 29Fb as an evolutionarily conserved suppressor of Ras-driven cancer. PLoS Genet 2018; 14:e1007688. [PMID: 30325918 PMCID: PMC6203380 DOI: 10.1371/journal.pgen.1007688] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 10/26/2018] [Accepted: 09/11/2018] [Indexed: 12/15/2022] Open
Abstract
Oncogenic mutations in the small GTPase Ras contribute to ~30% of human cancers. However, Ras mutations alone are insufficient for tumorigenesis, therefore it is paramount to identify cooperating cancer-relevant signaling pathways. We devised an in vivo near genome-wide, functional screen in Drosophila and discovered multiple novel, evolutionarily-conserved pathways controlling Ras-driven epithelial tumorigenesis. Human gene orthologs of the fly hits were significantly downregulated in thousands of primary tumors, revealing novel prognostic markers for human epithelial tumors. Of the top 100 candidate tumor suppressor genes, 80 were validated in secondary Drosophila assays, identifying many known cancer genes and multiple novel candidate genes that cooperate with Ras-driven tumorigenesis. Low expression of the confirmed hits significantly correlated with the KRASG12 mutation status and poor prognosis in pancreatic cancer. Among the novel top 80 candidate cancer genes, we mechanistically characterized the function of the top hit, the Tetraspanin family member Tsp29Fb, revealing that Tsp29Fb regulates EGFR signaling, epithelial architecture and restrains tumor growth and invasion. Our functional Drosophila screen uncovers multiple novel and evolutionarily conserved epithelial cancer genes, and experimentally confirmed Tsp29Fb as a key regulator of EGFR/Ras induced epithelial tumor growth and invasion. Cancer involves the cooperative interaction of many gene mutations. The Ras signaling pathway is upregulated in many human cancers, but upregulated Ras signaling alone is not sufficient to induce malignant tumors. We have undertaken a genome-wide genetic screen using a transgenic RNAi library in the vinegar fly, Drosophila melanogaster, to identify tumor suppressor genes that cooperate with the Ras oncogene (RasV12) in conferring overgrown invasive tumors. We stratified the hits by analyzing the expression of human orthologs of these genes in human epithelial cancers, revealing genes that were strongly downregulated in human cancer. By conducting secondary genetic interaction tests, we validated 80 of the top 100 genes. Pathway analysis of these genes revealed that 55 fell into known pathways involved in human cancer, whereas 25 were unique genes. We then confirmed the tumor suppressor properties of one of these genes, Tsp29Fb, encoding a Tetraspanin membrane protein, and showed that Tsp29Fb functions as a tumor suppressor by inhibiting Ras signaling and by maintaining epithelial cell polarity. Altogether, our study has revealed novel Ras-cooperating tumor suppressors in Drosophila and suggests that these genes may also be involved in human cancer.
Collapse
Affiliation(s)
- Tamara Zoranovic
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Science, Campus Vienna BioCentre, Vienna, Austria
| | - Jan Manent
- Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Department of Biochemistry & Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Lee Willoughby
- Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Ricardo Matos de Simoes
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - John E. La Marca
- Department of Biochemistry & Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Sofya Golenkina
- Department of Biochemistry & Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Xia Cuiping
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Science, Campus Vienna BioCentre, Vienna, Austria
| | - Susanne Gruber
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Science, Campus Vienna BioCentre, Vienna, Austria
| | - Belinda Angjeli
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Science, Campus Vienna BioCentre, Vienna, Austria
| | - Elisabeth Eva Kanitz
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Science, Campus Vienna BioCentre, Vienna, Austria
| | - Shane J. F. Cronin
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Science, Campus Vienna BioCentre, Vienna, Austria
| | - G. Gregory Neely
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Science, Campus Vienna BioCentre, Vienna, Austria
- The Charles Perkins Centre, School of Life & Environmental Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | | | - Patrick O. Humbert
- Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Department of Biochemistry & Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, Department of Anatomy & Neuroscience, Department of Biochemistry & Molecular Biology, and Department of Clinical Pathology, University of Melbourne, Melbourne, Victoria, Australia
| | - Kaylene J. Simpson
- Sir Peter MacCallum Department of Oncology, Department of Anatomy & Neuroscience, Department of Biochemistry & Molecular Biology, and Department of Clinical Pathology, University of Melbourne, Melbourne, Victoria, Australia
- Victorian Center for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Constantine S. Mitsiades
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Helena E. Richardson
- Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Department of Biochemistry & Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, Department of Anatomy & Neuroscience, Department of Biochemistry & Molecular Biology, and Department of Clinical Pathology, University of Melbourne, Melbourne, Victoria, Australia
- * E-mail: (HER); (JMP)
| | - Josef M. Penninger
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Science, Campus Vienna BioCentre, Vienna, Austria
- * E-mail: (HER); (JMP)
| |
Collapse
|
36
|
Liu L, Liu Y, Liu X, Zhang N, Mao G, Zeng Q, Yin M, Song D, Deng H. Resibufogenin suppresses transforming growth factor-β-activated kinase 1-mediated nuclear factor-κB activity through protein kinase C-dependent inhibition of glycogen synthase kinase 3. Cancer Sci 2018; 109:3611-3622. [PMID: 30168902 PMCID: PMC6215888 DOI: 10.1111/cas.13788] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/26/2018] [Accepted: 08/28/2018] [Indexed: 12/17/2022] Open
Abstract
Resibufogenin (RB), one of the major active compounds of the traditional Chinese medicine Chansu, has received considerable attention for its potency in cancer therapy. However, the anticancer effects and the underlying mechanisms of RB on pancreatic cancer remain elusive. Here, we found that RB inhibited the viability and induces caspase‐dependent apoptosis in human pancreatic cancer cells Panc‐1 and Aspc. Resibufogenin‐induced apoptosis was through inhibition of constitutive nuclear factor‐κB (NF‐κB) activity and its target genes’ expression, which was caused by downregulation of transforming growth factor‐β‐activated kinase 1 (TAK1) levels and suppression of IκB kinase activity in Panc‐1 and Aspc cells. This induction of TAK1‐mediated NF‐κB inactivation by RB was associated with increased glycogen synthase kinase‐3 (GSK‐3) phosphorylation and subsequent suppression of its activity. Moreover, RB‐induced GSK‐3 phosphorylation/inactivation acted through activation of protein kinase C but not Akt. Finally, RB suppressed human pancreatic tumor xenograft growth in athymic nude mice. Thus, our findings reveal a novel mechanism by which RB suppresses TAK1‐mediated NF‐κB activity through protein kinase C‐dependent inhibition of GSK‐3. Our findings provide a rationale for the potential application of RB in pancreatic cancer therapy.
Collapse
Affiliation(s)
- Lu Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Qingdao Women and Children's Hospital, Qingdao University, Qingdao, China
| | - Yang Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiaojia Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Na Zhang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Genxiang Mao
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou, China
| | - Qingxuan Zeng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Mingxiao Yin
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Danqing Song
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Hongbin Deng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
37
|
Dasatinib sensitises KRAS -mutant cancer cells to mitogen-activated protein kinase kinase inhibitor via inhibition of TAZ activity. Eur J Cancer 2018; 99:37-48. [DOI: 10.1016/j.ejca.2018.05.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/08/2018] [Accepted: 05/10/2018] [Indexed: 12/30/2022]
|
38
|
Noncanonical NF-κB in Cancer. Biomedicines 2018; 6:biomedicines6020066. [PMID: 29874793 PMCID: PMC6027307 DOI: 10.3390/biomedicines6020066] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 05/31/2018] [Accepted: 06/04/2018] [Indexed: 12/31/2022] Open
Abstract
The NF-κB pathway is a critical regulator of immune responses and is often dysregulated in cancer. Two NF-κB pathways have been described to mediate these responses, the canonical and the noncanonical. While understudied compared to the canonical NF-κB pathway, noncanonical NF-κB and its components have been shown to have effects, usually protumorigenic, in many different cancer types. Here, we review noncanonical NF-κB pathways and discuss its important roles in promoting cancer. We also discuss alternative NF-κB-independent functions of some the components of noncanonical NF-κB signaling. Finally, we discuss important crosstalk between canonical and noncanonical signaling, which blurs the two pathways, indicating that understanding the full picture of NF-κB regulation is critical to deciphering how this broad pathway promotes oncogenesis.
Collapse
|
39
|
Alam SK, Astone M, Liu P, Hall SR, Coyle AM, Dankert EN, Hoffman DK, Zhang W, Kuang R, Roden AC, Mansfield AS, Hoeppner LH. DARPP-32 and t-DARPP promote non-small cell lung cancer growth through regulation of IKKα-dependent cell migration. Commun Biol 2018; 1:43. [PMID: 29782621 PMCID: PMC5959014 DOI: 10.1038/s42003-018-0050-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related death worldwide. Here we demonstrate that elevated expression of dopamine and cyclic adenosine monophosphate-regulated phosphoprotein, Mr 32000 (DARPP-32) and its truncated splice variant t-DARPP promote lung tumor growth, while abrogation of DARPP-32 expression in human non-small cell lung cancer (NSCLC) cells reduces tumor growth in orthotopic mouse models. We observe a novel physical interaction between DARPP-32 and inhibitory kappa B kinase-α (IKKα) that promotes NSCLC cell migration through non-canonical nuclear factor kappa-light-chain-enhancer of activated B cells 2 (NF-κB2) signaling. Bioinformatics analysis of 513 lung adenocarcinoma patients reveals elevated t-DARPP isoform expression is associated with poor overall survival. Histopathological investigation of 62 human lung adenocarcinoma tissues also shows that t-DARPP expression is elevated with increasing tumor (T) stage. Our data suggest that DARPP-32 isoforms serve as a negative prognostic marker associated with increasing stages of NSCLC and may represent a novel therapeutic target.
Collapse
Affiliation(s)
- Sk Kayum Alam
- The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
| | - Matteo Astone
- The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
| | - Ping Liu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA.,Department of Oncology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Stephanie R Hall
- The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
| | - Abbygail M Coyle
- The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
| | - Erin N Dankert
- The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
| | - Dane K Hoffman
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Wei Zhang
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Rui Kuang
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Anja C Roden
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Aaron S Mansfield
- Department of Oncology, Division of Medical Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Luke H Hoeppner
- The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA.
| |
Collapse
|
40
|
Shuang T, Wang M, Zhou Y, Shi C, Wang D. NF-κB1, c-Rel, and ELK1 inhibit miR-134 expression leading to TAB1 upregulation in paclitaxel-resistant human ovarian cancer. Oncotarget 2018; 8:24853-24868. [PMID: 28206956 PMCID: PMC5421894 DOI: 10.18632/oncotarget.15267] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 01/16/2017] [Indexed: 02/07/2023] Open
Abstract
The mechanism by which the transcription factors inhibit the miRNA expression in ovarian cancer chemoresistance is unclear. The present study investigated the mechanism underlying the transcriptional repression of miR-134 in chemoresistant serous epithelial ovarian cancer. The results demonstrate that NF-κB1, c-Rel, and ELK1 are involved as transcription factors in repressing miR-134 expression in paclitaxel-resistant ovarian cancer cells. Knockdown of these transcription factors led to increased miR-134 expression, resulting in increased apoptosis and inhibition of proliferation in SKOV3-TR30 cells. NF-κB1, c-Rel, and ELK1 mRNA expression was upregulated in chemoresistant specimens and negatively correlated with miR-134 expression. Kaplan-Meier analysis revealed that high nuclear expressions of NF-κB1, c-Rel, ELK1 were significantly associated with short survival in serous epithelial ovarian cancer patients. Finally, TAB1 was identified as a functional target of miR-134, and the expression of TAB1 was increased by the transcription factors of NF-κB1, c-Rel, and ELK1 via miR-134. Taken together, these results provide an insight into the mechanism of repressed miR-134 expression in chemoresistance of serous epithelial ovarian cancer.
Collapse
Affiliation(s)
- Ting Shuang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, China.,Department of Obstetrics and Gynecology, Xijing Hospital, The Fourth Military Medical University, Xi'an 710033, China
| | - Min Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yingying Zhou
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Cong Shi
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Dandan Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| |
Collapse
|
41
|
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: 72] [Impact Index Per Article: 10.3] [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
|
42
|
Wang K, Huynh N, Wang X, Baldwin G, Nikfarjam M, He H. Inhibition of p21 activated kinase enhances tumour immune response and sensitizes pancreatic cancer to gemcitabine. Int J Oncol 2017; 52:261-269. [PMID: 29115428 DOI: 10.3892/ijo.2017.4193] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 10/18/2017] [Indexed: 12/17/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is one of the major types of cancer that exhibit high mortality worldwide because of the late diagnosis and the lack of effective treatment. Immunotherapy appears to be ineffective in PDA treatment due to the existence of a unique immune-suppressive microenvironment in PDA. Gemcitabine-based therapy is still the most commonly used chemotherapy to treat PDA patients with only marginal increased survival rates. This prompted us to continue the search for more effective therapy for PDA treatment. The effects of p21 activated kinases (PAKs) on tumour immune response and gemcitabine response were examined in PDA. An orthotopic murine PDA model, in which pancreatic cancer cells were injected to the tail of pancreas, was used. The mice were treated with PAK inhibitor, PF‑3758309, plus or minus gemcitabine. Tumour growth was measured by volume and weight. Tumour immune response was determined by flow cytometry analysis of splenic cells and immunohistochemical staining of intratumoural lymphocytes. Inhibition of PAKs by PF‑3758309, not only suppressed tumour growth, but also stimulated tumour immune response by increasing the numbers of splenic and intratumoural T lymphocytes. Furthermore, inhibition of PAKs decreased PDA cell growth synergistically with gemcitabine in vitro and in vivo. The dual effects of inhibition of PAKs make PAK-targeted therapy more potent for the treatment of PDA. The combination of PAK inhibitors with gemcitabine may be a more effective therapeutic approach in PDA treatment.
Collapse
Affiliation(s)
- Kai Wang
- Department of Surgery, University of Melbourne, Austin Health, Heidelberg, Victoria 3048, Australia
| | - Nhi Huynh
- Department of Surgery, University of Melbourne, Austin Health, Heidelberg, Victoria 3048, Australia
| | - Xiao Wang
- Department of Surgery, University of Melbourne, Austin Health, Heidelberg, Victoria 3048, Australia
| | - Graham Baldwin
- Department of Surgery, University of Melbourne, Austin Health, Heidelberg, Victoria 3048, Australia
| | - Mehrdad Nikfarjam
- Department of Surgery, University of Melbourne, Austin Health, Heidelberg, Victoria 3048, Australia
| | - Hong He
- Department of Surgery, University of Melbourne, Austin Health, Heidelberg, Victoria 3048, Australia
| |
Collapse
|
43
|
Santoro R, Carbone C, Piro G, Chiao PJ, Melisi D. TAK -ing aim at chemoresistance: The emerging role of MAP3K7 as a target for cancer therapy. Drug Resist Updat 2017; 33-35:36-42. [DOI: 10.1016/j.drup.2017.10.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 10/11/2017] [Accepted: 10/21/2017] [Indexed: 01/08/2023]
|
44
|
Siddiqui I, Erreni M, Kamal MA, Porta C, Marchesi F, Pesce S, Pasqualini F, Schiarea S, Chiabrando C, Mantovani A, Allavena P. Differential role of Interleukin-1 and Interleukin-6 in K-Ras-driven pancreatic carcinoma undergoing mesenchymal transition. Oncoimmunology 2017; 7:e1388485. [PMID: 29308316 DOI: 10.1080/2162402x.2017.1388485] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 09/29/2017] [Accepted: 09/30/2017] [Indexed: 02/07/2023] Open
Abstract
K-Ras mutations are a hallmark of human pancreatic adenocarcinoma (PDAC) and epithelial-mesenchymal-transition (EMT) is a driver of progression. Oncogenic K-Ras causes the constitutive activation of NF-kB and the switch-on of an inflammatory program, which further fuels NF-kB and STAT3 activation. In this study we investigated how inflammatory pathways triggered by oncogenic K-Ras are regulated in human pancreatic cancer cells with distict epithelial or mesenchymal phenotype. Our results demonstrate that in cells with epithelial features, K-Ras driven inflammation is under the control of IL-1, while in cells undergoing EMT, is IL-1 independent. In pancreatic tumor cells with EMT phenotype, treatment with IL-1R antagonist (Anakinra) did not inhibit inflammatory cytokine production and tumor growth in mice. In these cells IL-6 is actively transcribed by the EMT transcription factor TWIST. Targeting of mesenchymal pancreatic tumors in vivo with anti-IL-6RmAb (RoActemra) successfully decreased tumor growth in immunodeficient mice, inhibited the inflammatory stroma and NF-kB-p65 and STAT3 phosphorylation in cancer cells. The results confirm that IL-1 is an important driver of inflammation in epithelial pancreatic tumors; however, tumor cells undergoing EMT will likely escape IL-1R inhibition, as IL-6 is continuously transcribed by TWIST. These findings have implications for the rational targeting of inflammatory pathways in human pancreatic cancer.
Collapse
Affiliation(s)
- Imran Siddiqui
- Department of Immunology, IRCCS Clinical and Research Institute Humanitas, Rozzano (Milano), Italy
| | - Marco Erreni
- Department of Immunology, IRCCS Clinical and Research Institute Humanitas, Rozzano (Milano), Italy
| | - Mohammad Azhar Kamal
- Department of Immunology, IRCCS Clinical and Research Institute Humanitas, Rozzano (Milano), Italy
| | - Chiara Porta
- Department of Pharmaceutical Sciences, Università Piemonte Orientale, Novara, Italy
| | - Federica Marchesi
- Department of Immunology, IRCCS Clinical and Research Institute Humanitas, Rozzano (Milano), Italy.,Department of Medical Biotechnology and Translational Medicine, University of Milan, Italy
| | - Samantha Pesce
- Department of Immunology, IRCCS Clinical and Research Institute Humanitas, Rozzano (Milano), Italy
| | - Fabio Pasqualini
- Department of Immunology, IRCCS Clinical and Research Institute Humanitas, Rozzano (Milano), Italy
| | - Silvia Schiarea
- Department of Environmental Health Sciences, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Chiara Chiabrando
- Department of Environmental Health Sciences, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Alberto Mantovani
- Department of Immunology, IRCCS Clinical and Research Institute Humanitas, Rozzano (Milano), Italy.,Humanitas University, Rozzano (Milano), Italy
| | - Paola Allavena
- Department of Immunology, IRCCS Clinical and Research Institute Humanitas, Rozzano (Milano), Italy.,Humanitas University, Rozzano (Milano), Italy
| |
Collapse
|
45
|
Asai S, Miura N, Sawada Y, Noda T, Kikugawa T, Tanji N, Saika T. Silencing of ECHDC1 inhibits growth of gemcitabine-resistant bladder cancer cells. Oncol Lett 2017; 15:522-527. [PMID: 29391886 DOI: 10.3892/ol.2017.7269] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 04/04/2017] [Indexed: 02/06/2023] Open
Abstract
Combined gemcitabine and cisplatin (GC) treatment is a first line chemotherapy for bladder cancer. However, acquired resistance to GC has been a major problem. To address the mechanism of gemcitabine resistance, and to identify potential biomarkers or target proteins for its therapy, we aimed to identify candidate proteins associated with gemcitabine resistance using proteomic analysis. We established gemcitabine-resistant human bladder cancer cell lines (UMUC3GR and HT1376GR) from gemcitabine-sensitive human bladder cancer cell lines (UMUC3 and HT1376). We compared the protein expression of parental and gemcitabine-resistant cell lines using isobaric tags for relative and absolute quantification (iTRAQ) and liquid chromatography tandem mass spectrometry. Among the identified proteins, ethylmalonyl-CoA decarboxylase (ECHDC1) expression was significantly increased in both of the gemcitabine-resistant cell lines compared to the respective parental cell lines. Silencing of ECHDC1 reduced ECHDC1 expression and significantly inhibited the proliferation of UMUC3GR cells. Furthermore, silencing of ECHDC1 induced upregulation of p27, which is critical for cell cycle arrest in the G1 phase, and induced G1 arrest. In conclusion, ECHDC1 expression is increased in gemcitabine-resistant bladder cancer cells, and is involved in their cell growth. ECHDC1, which is a metabolite proofreading enzyme, may be a novel potential target for gemcitabine-resistant bladder cancer therapy.
Collapse
Affiliation(s)
- Seiji Asai
- Department of Urology, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| | - Noriyoshi Miura
- Department of Urology, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| | - Yuichiro Sawada
- Department of Urology, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| | - Terutaka Noda
- Department of Urology, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| | - Tadahiko Kikugawa
- Department of Urology, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| | - Nozomu Tanji
- Department of Urology, Houshasen-Daiichi Hospital, Imabari, Ehime 794-0054, Japan
| | - Takashi Saika
- Department of Urology, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| |
Collapse
|
46
|
Kim J, Hyun J, Wang S, Lee C, Lee JW, Moon EY, Cha H, Diehl AM, Jung Y. Thymosin beta-4 regulates activation of hepatic stellate cells via hedgehog signaling. Sci Rep 2017; 7:3815. [PMID: 28630423 PMCID: PMC5476572 DOI: 10.1038/s41598-017-03782-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 05/03/2017] [Indexed: 12/29/2022] Open
Abstract
The molecular mechanisms of thymosin beta-4 (TB4) involved in regulating hepatic stellate cell (HSC) functions remain unclear. Therefore, we hypothesize that TB4 influences HSC activation through hedgehog (Hh) pathway. HSC functions declined in a TB4 siRNA-treated LX-2. TB4 suppression down-regulated both integrin linked kinase (ILK), an activator of smoothened, and phosphorylated glycogen synthase kinase 3 beta (pGSK-3B), an inactive form of GSK-3B degrading glioblastoma 2 (GLI2), followed by the decreased expression of both smoothened and GLI2. A TB4 CRISPR also blocked the activation of primary HSCs, with decreased expression of smoothened, GLI2 and ILK compared with cells transfected with nontargeting control CRISPR. Double immunostaining and an immunoprecipitation assay revealed that TB4 interacted with either smoothened at the cytoplasm or GLI2 at the nucleus in LX-2. Smoothened suppression in primary HSCs using a Hh antagonist or adenovirus transduction decreased TB4 expression with the reduced activation of HSCs. Tb4-overexpressing transgenic mice treated with CCl4 were susceptible to the development hepatic fibrosis with higher levels of ILK, pGSK3b, and Hh activity, as compared with wild-type mice. These findings demonstrate that TB4 regulates HSC activation by influencing the activity of Smoothened and GLI2, suggesting TB4 as a novel therapeutic target in liver disease.
Collapse
Affiliation(s)
- Jieun Kim
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, 63-2 Pusandaehak-ro, Geumjeong-gu, Pusan, 46241, Republic of Korea
| | - Jeongeun Hyun
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, 63-2 Pusandaehak-ro, Geumjeong-gu, Pusan, 46241, Republic of Korea
| | - Sihyung Wang
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, 63-2 Pusandaehak-ro, Geumjeong-gu, Pusan, 46241, Republic of Korea
| | - Chanbin Lee
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, 63-2 Pusandaehak-ro, Geumjeong-gu, Pusan, 46241, Republic of Korea
| | - Jae-Wook Lee
- Department of Bioscience and Biotechnology, Sejong University, 209 Neungdong-ro, Gwangjin-gu, Seoul, 05006, Republic of Korea
| | - Eun-Yi Moon
- Department of Bioscience and Biotechnology, Sejong University, 209 Neungdong-ro, Gwangjin-gu, Seoul, 05006, Republic of Korea
| | - Heejae Cha
- Department of Parasitology and Genetics, Kosin University College of Medicine, 262 Gamcheon-ro, Seogu, Pusan, 49267, Republic of Korea
| | - Anna Mae Diehl
- Division of Gastroenterology, Department of Medicine, Duke University, 595 LaSalle Street, Durham, NC, 27710, USA
| | - Youngmi Jung
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, 63-2 Pusandaehak-ro, Geumjeong-gu, Pusan, 46241, Republic of Korea.
- Department of Biological Sciences, College of Natural Science, Pusan National University, 63-2 Pusandaehak-ro, Geumjeong-gu, Pusan, 46241, Republic of Korea.
| |
Collapse
|
47
|
Guo C, Huang Y, Yu J, Liu L, Gong X, Huang M, Jiang C, Liao Y, Huang L, Yang G, Li J. The impacts of single nucleotide polymorphisms in genes of cell cycle and NF-kB pathways on the efficacy and acute toxicities of radiotherapy in patients with nasopharyngeal carcinoma. Oncotarget 2017; 8:25334-25344. [PMID: 28445979 PMCID: PMC5421934 DOI: 10.18632/oncotarget.15835] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 02/06/2017] [Indexed: 12/12/2022] Open
Abstract
Radiotherapy is one of the primary choices for the treatment of nasopharyngeal carcinoma (NPC) and may result in severe radiotoxicities on normal tissues. Single nucleotide polymorphisms (SNPs) in genes of cell cycle and NF-κB pathways have been linked with the prognoses of various cancers. The aim of this study was to explore whether SNPs of genes involved in cell cycle and NF-κB pathways are associated with responses to radiotherapy in NPC patients. We selected 3 SNPs in cell cycle pathway and 5 SNPs in NF-κB pathway and genotyped them in 154 NPC patients treated with radiotherapy. Multivariate logistic regression was used to determine the association of these 8 SNPs with the responses to radiotherapy. We observed that cyclin-dependent kinase inhibitor gene CDKN2A rs3088440 was significantly related with a poorer treatment efficacy on the primary tumor and cervical lymph node after radiotherapy, and also with a decreased risk of grade 3-4 acute radiation-induced myelosuppression. In some subgroups, cyclin D1 gene CCND1 rs9344 and inhibitor of κB kinase gene IKBKB rs12676482 were related with the grade 3-4 acute radiation-induced myelosuppression, and CCND1 rs9344 was also associated with grade 3-4 acute radiation-induced oral mucositis. The current results reveal that SNPs in genes of cell cycle pathwayand NF-κB pathway have the potential to predict the clinical responses to radiotherapy for NPC patients.
Collapse
Affiliation(s)
- Chengxian Guo
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Yuling Huang
- Department of Radiation Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi 330029, China
| | - Jingjing Yu
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Lijuan Liu
- Department of Pharmacy, Jiangxi Cancer Hospital, Nanchang, Jiangxi 330029, China
| | - Xiaochang Gong
- Department of Radiation Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi 330029, China
| | - Min Huang
- Department of Radiation Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi 330029, China
| | - Chunling Jiang
- Department of Radiation Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi 330029, China
| | - Yulu Liao
- Department of Radiation Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi 330029, China
| | - Lihua Huang
- Center for Medical Experiments, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Guoping Yang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Jingao Li
- Department of Radiation Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi 330029, China
| |
Collapse
|
48
|
Durand JK, Baldwin AS. Targeting IKK and NF-κB for Therapy. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2017; 107:77-115. [PMID: 28215229 DOI: 10.1016/bs.apcsb.2016.11.006] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In addition to regulating immune responses, the NF-κB family of transcription factors also promotes cellular proliferation and survival. NF-κB and its activating kinase, IKK, have become appealing therapeutic targets because of their critical roles in the progression of many diseases including chronic inflammation and cancer. Here, we discuss the conditions that lead to pathway activation, the effects of constitutive activation, and some of the strategies used to inhibit NF-κB signaling.
Collapse
Affiliation(s)
- J K Durand
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC, United States; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States
| | - A S Baldwin
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States.
| |
Collapse
|
49
|
Zhang D, Li L, Jiang H, Knolhoff BL, Lockhart AC, Wang-Gillam A, DeNardo DG, Ruzinova MB, Lim KH. Constitutive IRAK4 Activation Underlies Poor Prognosis and Chemoresistance in Pancreatic Ductal Adenocarcinoma. Clin Cancer Res 2016; 23:1748-1759. [PMID: 27702822 DOI: 10.1158/1078-0432.ccr-16-1121] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 09/19/2016] [Accepted: 09/28/2016] [Indexed: 12/19/2022]
Abstract
Purpose: Aberrant activation of the NF-κB transcription factors underlies the aggressive behavior and poor outcome of pancreatic ductal adenocarcinoma (PDAC). However, clinically effective and safe NF-κB inhibitors are not yet available. Because NF-κB transcription factors can be activated by the interleukin-1 receptor-associated kinases (IRAKs) downstream of the Toll-like receptors (TLRs), but has not been explored in PDAC, we sought to investigate the role of IRAKs in the pathobiology of PDAC.Experimental Design: We examined the phosphorylation status of IRAK4 (p-IRAK4), the master regulator of TLR signaling, in PDAC cell lines, in surgical samples and commercial tissue microarray. We then performed functional studies using small-molecule IRAK1/4 inhibitor, RNA-interference, and CRISPR/Cas9n techniques to delineate the role of IRAK4 in NF-κB activity, chemoresistance, cytokine production, and growth of PDAC cells in vitro and in vivoResults: p-IRAK4 staining was detectable in the majority of PDAC lines and about 60% of human PDAC samples. The presence of p-IRAK4 strongly correlated with phospho-NF-κB/p65 staining in PDAC samples and is predictive of postoperative relapse and poor overall survival. Inhibition of IRAK4 potently reduced NF-κB activity, anchorage-independent growth, chemoresistance, and secretion of proinflammatory cytokines from PDAC cells. Both pharmacologic suppression and genetic ablation of IRAK4 greatly abolished PDAC growth in mice and augmented the therapeutic effect of gemcitabine by promoting apoptosis, reducing tumor cell proliferation and tumor fibrosis.Conclusions: Our data established IRAK4 as a novel therapeutic target for PDAC treatment. Development of potent IRAK4 inhibitors is needed for clinical testing. Clin Cancer Res; 23(7); 1748-59. ©2016 AACR.
Collapse
Affiliation(s)
- Daoxiang Zhang
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, Saint Louis, Missouri
| | - Lin Li
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, Saint Louis, Missouri
| | - Hongmei Jiang
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, Saint Louis, Missouri
| | - Brett L Knolhoff
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, Saint Louis, Missouri
| | - Albert C Lockhart
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, Saint Louis, Missouri
| | - Andrea Wang-Gillam
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, Saint Louis, Missouri
| | - David G DeNardo
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, Saint Louis, Missouri
| | - Marianna B Ruzinova
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri
| | - Kian-Huat Lim
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, Saint Louis, Missouri.
| |
Collapse
|
50
|
The PRKD1 promoter is a target of the KRas-NF-κB pathway in pancreatic cancer. Sci Rep 2016; 6:33758. [PMID: 27649783 PMCID: PMC5030668 DOI: 10.1038/srep33758] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 09/01/2016] [Indexed: 12/15/2022] Open
Abstract
Increased expression of PRKD1 and its gene product protein kinase D1 (PKD1) are linked to oncogenic signaling in pancreatic ductal adenocarcinoma, but a direct functional relationship to oncogenic KRas has not been established so far. We here describe the PRKD1 gene promoter as a target for oncogenic KRas signaling. We demonstrate that KRas-induced activation of the canonical NF-κB pathway is one mechanism of how PRKD1 expression is increased and identify the binding sites for NF-κB in the PRKD1 promoter. Altogether, these results describe a novel mechanism governing PRKD1 gene expression in PDA and provide a functional link between oncogenic KRas, NF-κB and expression of PRKD1.
Collapse
|