1
|
Ruan F, Ruan Y, Gu H, Sun J, Chen Q. Clitocine enhances the drug sensitivity of colon cancer cells by promoting FBXW7-mediated MCL-1 degradation via inhibiting the A2B/cAMP/ERK axis. Am J Physiol Cell Physiol 2024; 327:C884-C900. [PMID: 39140602 DOI: 10.1152/ajpcell.00310.2024] [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: 05/10/2024] [Revised: 07/18/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024]
Abstract
Chemotherapy resistance to colon cancer is an unavoidable obstacle in the clinical management of the disease. Clitocine, an adenosine analog, played a significant role in the chemosensitivity of human colon cancer cells by promoting myeloid cell leukemia 1 (MCL-1) protein degradation. However, the detailed mechanism remains to be further elucidated. We found that clitocine upregulates the expression of F-box and WD repeat domain containing 7 (FBXW7), a ubiquitin ligase involved in the MCL-1 degradation. Transcriptome sequencing analysis revealed that clitocine significantly inhibits the cyclic adenosine monophosphate (cAMP) and extracellular regulated protein kinases (ERK) downstream signaling pathways in colon cancer cells, thereby enhancing FBXW7 expression and subsequently promoting the ubiquitination degradation of MCL-1 protein. We verified that clitocine regulated intracellular cAMP levels by competitive binding with the adenosine receptor A2B. A molecular docking assay also verified the binding relationship. By decreasing intracellular cAMP levels, clitocine blocks the activation of downstream signaling pathways, which ultimately enhances the drug sensitivity of colon cancer cells through increased FBXW7 expression due to the inhibition of its promoter DNA methylation. Both knockout of the adenosine receptor A2B and Br-cAMP treatment can effectively attenuate the function of clitocine in vitro and in vivo. This study clarified that clitocine enhanced the drug sensitivity of colon cancer cells by promoting FBXW7-mediated MCL-1 degradation via inhibiting the A2B/cAMP/ERK axis, providing further knowledge of the clinical application for clitocine.NEW & NOTEWORTHY Our study found that clitocine enhances the drug sensitivity of colon cancer cells by promoting FBXW7-mediated MCL-1 degradation via inhibiting the A2B/cAMP/ERK axis.
Collapse
Affiliation(s)
- Feng Ruan
- Department of Emergency Medicine, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Yanyun Ruan
- Precision Medicine Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, People's Republic of China
| | - Huamin Gu
- Department of Pathology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, People's Republic of China
| | - Jianguo Sun
- Precision Medicine Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, People's Republic of China
| | - Qi Chen
- Precision Medicine Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, People's Republic of China
| |
Collapse
|
2
|
Qi Y, Rezaeian AH, Wang J, Huang D, Chen H, Inuzuka H, Wei W. Molecular insights and clinical implications for the tumor suppressor role of SCF FBXW7 E3 ubiquitin ligase. Biochim Biophys Acta Rev Cancer 2024; 1879:189140. [PMID: 38909632 PMCID: PMC11390337 DOI: 10.1016/j.bbcan.2024.189140] [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: 04/12/2024] [Revised: 06/04/2024] [Accepted: 06/17/2024] [Indexed: 06/25/2024]
Abstract
FBXW7 is one of the most well-characterized F-box proteins, serving as substrate receptor subunit of SKP1-CUL1-F-box (SCF) E3 ligase complexes. SCFFBXW7 is responsible for the degradation of various oncogenic proteins such as cyclin E, c-MYC, c-JUN, NOTCH, and MCL1. Therefore, FBXW7 functions largely as a major tumor suppressor. In keeping with this notion, FBXW7 gene mutations or downregulations have been found and reported in many types of malignant tumors, such as endometrial, colorectal, lung, and breast cancers, which facilitate the proliferation, invasion, migration, and drug resistance of cancer cells. Therefore, it is critical to review newly identified FBXW7 regulation and tumor suppressor function under physiological and pathological conditions to develop effective strategies for the treatment of FBXW7-altered cancers. Since a growing body of evidence has revealed the tumor-suppressive activity and role of FBXW7, here, we updated FBXW7 upstream and downstream signaling including FBXW7 ubiquitin substrates, the multi-level FBXW7 regulatory mechanisms, and dysregulation of FBXW7 in cancer, and discussed promising cancer therapies targeting FBXW7 regulators and downstream effectors, to provide a comprehensive picture of FBXW7 and facilitate the study in this field.
Collapse
Affiliation(s)
- Yihang Qi
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Abdol-Hossein Rezaeian
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jingchao Wang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Daoyuan Huang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Hong Chen
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Hiroyuki Inuzuka
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
3
|
Tarr J, Salovich JM, Aichinger M, Jeon K, Veerasamy N, Sensintaffar JL, Arnhof H, Samwer M, Christov PP, Kim K, Wunberg T, Schweifer N, Trapani F, Arnold A, Martin F, Zhao B, Miriyala N, Sgubin D, Fogarty S, Moore WJ, Stott GM, Olejniczak ET, Engelhardt H, Rudolph D, Lee T, McConnell DB, Fesik SW. Discovery of a Myeloid Cell Leukemia 1 (Mcl-1) Inhibitor That Demonstrates Potent In Vivo Activities in Mouse Models of Hematological and Solid Tumors. J Med Chem 2024; 67:14370-14393. [PMID: 39102508 PMCID: PMC11345828 DOI: 10.1021/acs.jmedchem.4c01188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 08/07/2024]
Abstract
Myeloid cell leukemia 1 (Mcl-1) is a key regulator of the intrinsic apoptosis pathway. Overexpression of Mcl-1 is correlated with high tumor grade, poor survival, and both intrinsic and acquired resistance to cancer therapies. Herein, we disclose the structure-guided design of a small molecule Mcl-1 inhibitor, compound 26, that binds to Mcl-1 with subnanomolar affinity, inhibits growth in cell culture assays, and possesses low clearance in mouse and dog pharmacokinetic (PK) experiments. Evaluation of 26 as a single agent in Mcl-1 sensitive hematological and solid tumor xenograft models resulted in regressions. Co-treatment of Mcl-1-sensitive and Mcl-1 insensitive lung cancer derived xenografts with 26 and docetaxel or topotecan, respectively, resulted in an enhanced tumor response. These findings support the premise that pro-apoptotic priming of tumor cells by other therapies in combination with Mcl-1 inhibition may significantly expand the subset of cancers in which Mcl-1 inhibitors may prove beneficial.
Collapse
Affiliation(s)
- James
C. Tarr
- Department
of Biochemistry, Vanderbilt University School
of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - James M. Salovich
- Department
of Biochemistry, Vanderbilt University School
of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - Martin Aichinger
- Discovery
Research, Boehringer Ingelheim Regional
Center Vienna GmbH & Co KG, 1120 Vienna, Austria
| | - KyuOk Jeon
- Department
of Biochemistry, Vanderbilt University School
of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - Nagarathanam Veerasamy
- Department
of Biochemistry, Vanderbilt University School
of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - John L. Sensintaffar
- Department
of Biochemistry, Vanderbilt University School
of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - Heribert Arnhof
- Discovery
Research, Boehringer Ingelheim Regional
Center Vienna GmbH & Co KG, 1120 Vienna, Austria
| | - Matthias Samwer
- Discovery
Research, Boehringer Ingelheim Regional
Center Vienna GmbH & Co KG, 1120 Vienna, Austria
| | - Plamen P. Christov
- Molecular
Design and Synthesis Center, Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37323-0146, United States
| | - Kwangho Kim
- Molecular
Design and Synthesis Center, Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37323-0146, United States
| | - Tobias Wunberg
- Discovery
Research, Boehringer Ingelheim Regional
Center Vienna GmbH & Co KG, 1120 Vienna, Austria
| | - Norbert Schweifer
- Discovery
Research, Boehringer Ingelheim Regional
Center Vienna GmbH & Co KG, 1120 Vienna, Austria
| | - Francesca Trapani
- Discovery
Research, Boehringer Ingelheim Regional
Center Vienna GmbH & Co KG, 1120 Vienna, Austria
| | - Allison Arnold
- Department
of Biochemistry, Vanderbilt University School
of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - Florian Martin
- Discovery
Research, Boehringer Ingelheim Regional
Center Vienna GmbH & Co KG, 1120 Vienna, Austria
| | - Bin Zhao
- Department
of Biochemistry, Vanderbilt University School
of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - Nagaraju Miriyala
- Department
of Biochemistry, Vanderbilt University School
of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - Danielle Sgubin
- Department
of Biochemistry, Vanderbilt University School
of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - Stuart Fogarty
- Department
of Biochemistry, Vanderbilt University School
of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - William J. Moore
- Leidos
Biomedical Research, Frederick National
Laboratory for Cancer Research, Frederick, Maryland 21701-4907, United States
| | - Gordon M. Stott
- Leidos
Biomedical Research, Frederick National
Laboratory for Cancer Research, Frederick, Maryland 21701-4907, United States
| | - Edward T. Olejniczak
- Department
of Biochemistry, Vanderbilt University School
of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - Harald Engelhardt
- Discovery
Research, Boehringer Ingelheim Regional
Center Vienna GmbH & Co KG, 1120 Vienna, Austria
| | - Dorothea Rudolph
- Discovery
Research, Boehringer Ingelheim Regional
Center Vienna GmbH & Co KG, 1120 Vienna, Austria
| | - Taekyu Lee
- Department
of Biochemistry, Vanderbilt University School
of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - Darryl B. McConnell
- Discovery
Research, Boehringer Ingelheim Regional
Center Vienna GmbH & Co KG, 1120 Vienna, Austria
| | - Stephen W. Fesik
- Department
of Biochemistry, Vanderbilt University School
of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| |
Collapse
|
4
|
Boretto M, Geurts MH, Gandhi S, Ma Z, Staliarova N, Celotti M, Lim S, He GW, Millen R, Driehuis E, Begthel H, Smabers L, Roodhart J, van Es J, Wu W, Clevers H. Epidermal growth factor receptor (EGFR) is a target of the tumor-suppressor E3 ligase FBXW7. Proc Natl Acad Sci U S A 2024; 121:e2309902121. [PMID: 38483988 PMCID: PMC10962967 DOI: 10.1073/pnas.2309902121] [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: 06/12/2023] [Accepted: 01/08/2024] [Indexed: 03/19/2024] Open
Abstract
FBXW7 is an E3 ubiquitin ligase that targets proteins for proteasome-mediated degradation and is mutated in various cancer types. Here, we use CRISPR base editors to introduce different FBXW7 hotspot mutations in human colon organoids. Functionally, FBXW7 mutation reduces EGF dependency of organoid growth by ~10,000-fold. Combined transcriptomic and proteomic analyses revealed increased EGFR protein stability in FBXW7 mutants. Two distinct phosphodegron motifs reside in the cytoplasmic tail of EGFR. Mutations in these phosphodegron motifs occur in human cancer. CRISPR-mediated disruption of the phosphodegron motif at T693 reduced EGFR degradation and EGF growth factor dependency. FBXW7 mutant organoids showed reduced sensitivity to EGFR-MAPK inhibitors. These observations were further strengthened in CRC-derived organoid lines and validated in a cohort of patients treated with panitumumab. Our data imply that FBXW7 mutations reduce EGF dependency by disabling EGFR turnover.
Collapse
Affiliation(s)
- Matteo Boretto
- Organoid group, Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, 3584CTUtrecht, the Netherlands
| | - Maarten H. Geurts
- Organoid group, Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, 3584CTUtrecht, the Netherlands
| | - Shashank Gandhi
- Organoid group, Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, 3584CTUtrecht, the Netherlands
- Department of Molecular and Cellular Biology, Miller Institute for Basic Research in Science, University of California, Berkeley, CA94720
| | - Ziliang Ma
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore138648, Singapore
- Department of Pharmacy, National University of Singapore, Singapore117543, Singapore
- Department of Biomolecular Mass Spectrometry and Proteomics, Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CHUtrecht, the Netherlands
| | - Nadzeya Staliarova
- Department of Biomolecular Mass Spectrometry and Proteomics, Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CHUtrecht, the Netherlands
| | - Martina Celotti
- Organoid group, Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, 3584CTUtrecht, the Netherlands
| | - Sangho Lim
- Organoid group, Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, 3584CTUtrecht, the Netherlands
| | - Gui-Wei He
- Organoid group, Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, 3584CTUtrecht, the Netherlands
| | - Rosemary Millen
- Organoid group, Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, 3584CTUtrecht, the Netherlands
| | - Else Driehuis
- Organoid group, Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, 3584CTUtrecht, the Netherlands
| | - Harry Begthel
- Organoid group, Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, 3584CTUtrecht, the Netherlands
| | - Lidwien Smabers
- Department of Medical Oncology, University Medical Center Utrecht, 3584 CXUtrecht, the Netherlands
| | - Jeanine Roodhart
- Department of Medical Oncology, University Medical Center Utrecht, 3584 CXUtrecht, the Netherlands
| | - Johan van Es
- Organoid group, Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, 3584CTUtrecht, the Netherlands
| | - Wei Wu
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore138648, Singapore
- Department of Pharmacy, National University of Singapore, Singapore117543, Singapore
- Department of Biomolecular Mass Spectrometry and Proteomics, Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CHUtrecht, the Netherlands
| | - Hans Clevers
- Organoid group, Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, 3584CTUtrecht, the Netherlands
| |
Collapse
|
5
|
Cui Z, Cong M, Yin S, Li Y, Ye Y, Liu X, Tang J. Role of protein degradation systems in colorectal cancer. Cell Death Discov 2024; 10:141. [PMID: 38485957 PMCID: PMC10940631 DOI: 10.1038/s41420-023-01781-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 03/18/2024] Open
Abstract
Protein degradation is essential for maintaining protein homeostasis. The ubiquitin‒proteasome system (UPS) and autophagy-lysosome system are the two primary pathways responsible for protein degradation and directly related to cell survival. In malignant tumors, the UPS plays a critical role in managing the excessive protein load caused by cancer cells hyperproliferation. In this review, we provide a comprehensive overview of the dual roles played by the UPS and autolysosome system in colorectal cancer (CRC), elucidating their impact on the initiation and progression of this disease while also highlighting their compensatory relationship. Simultaneously targeting both protein degradation pathways offers new promise for enhancing treatment efficacy against CRC. Additionally, apoptosis is closely linked to ubiquitination and autophagy, and caspases degrade proteins. A thorough comprehension of the interplay between various protein degradation pathways is highly important for clarifying the mechanism underlying the onset and progression of CRC.
Collapse
Affiliation(s)
- Zihan Cui
- Department of Pathology, Harbin Medical University, Harbin, 150081, China
| | - Mingqi Cong
- Department of Pathology, Harbin Medical University, Harbin, 150081, China
| | - Shengjie Yin
- Department of Oncology, Chifeng City Hospital, Chifeng, 024000, China
| | - Yuqi Li
- Department of Pathology, Harbin Medical University, Harbin, 150081, China
| | - Yuguang Ye
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| | - Xi Liu
- Cardiovascular Center, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia, 010017, China.
| | - Jing Tang
- Department of Pathology, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
6
|
Chen D, Ermine K, Wang YJ, Chen X, Lu X, Wang P, Beer-Stolz D, Yu J, Zhang L. PUMA/RIP3 Mediates Chemotherapy Response via Necroptosis and Local Immune Activation in Colorectal Cancer. Mol Cancer Ther 2024; 23:354-367. [PMID: 37992761 PMCID: PMC10932881 DOI: 10.1158/1535-7163.mct-23-0162] [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: 03/19/2023] [Revised: 10/02/2023] [Accepted: 11/21/2023] [Indexed: 11/24/2023]
Abstract
Induction of programmed cell death (PCD) is a key cytotoxic effect of anticancer therapies. PCD is not confined to caspase-dependent apoptosis, but includes necroptosis, a regulated form of necrotic cell death controlled by receptor-interacting protein (RIP) kinases 1 and 3, and mixed lineage kinase domain-like (MLKL) pseudokinase. Necroptosis functions as a defense mechanism against oncogenic mutations and pathogens and can be induced by a variety of anticancer agents. However, the functional role and regulatory mechanisms of necroptosis in anticancer therapy are poorly understood. In this study, we found that RIP3-dependent but RIP1-independent necroptosis is engaged by 5-fluorouracil (5-FU) and other widely used antimetabolite drugs, and functions as a major mode of cell death in a subset of colorectal cancer cells that express RIP3. We identified a novel 5-FU-induced necroptosis pathway involving p53-mediated induction of the BH3-only Bcl-2 family protein, p53 upregulated modulator of apoptosis (PUMA), which promotes cytosolic release of mitochondrial DNA and stimulates its sensor z-DNA-binding protein 1 (ZBP1) to activate RIP3. PUMA/RIP3-dependent necroptosis mediates the in vitro and in vivo antitumor effects of 5-FU and promotes a robust antitumor immune response. Our findings provide a rationale for stimulating necroptosis to enhance tumor cell killing and antitumor immune response leading to improved colorectal cancer treatments.
Collapse
Affiliation(s)
- Dongshi Chen
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, CA
| | - Kaylee Ermine
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Yi-Jun Wang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Xiaojun Chen
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Xinyan Lu
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, CA
- Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, CA
| | - Peng Wang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Donna Beer-Stolz
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Jian Yu
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, CA
- Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, CA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Lin Zhang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, CA
- Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, CA
| |
Collapse
|
7
|
Xiao Y, Liu R, Li N, Li Y, Huang X. Role of the ubiquitin-proteasome system on macrophages in the tumor microenvironment. J Cell Physiol 2024; 239:e31180. [PMID: 38219045 DOI: 10.1002/jcp.31180] [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: 09/06/2023] [Revised: 11/14/2023] [Accepted: 12/12/2023] [Indexed: 01/15/2024]
Abstract
Tumor-associated macrophages (TAMs) are key components of the tumor microenvironment, and their different polarization states play multiple roles in tumors by secreting cytokines, chemokines, and so on, which are closely related to tumor development. In addition, the enrichment of TAMs is often associated with poor prognosis of tumors. Thus, targeting TAMs is a potential tumor treatment strategy, in which therapeutic approaches such as reducing TAMs numbers, remodeling TAMs phenotypes, and altering their functions are being extensively investigated. Meanwhile, the ubiquitin-proteasome system (UPS), an important mechanism of protein hydrolysis in eukaryotic cells, participates in cellular processes by regulating the activity and stability of key proteins. Interestingly, UPS plays a dual role in the process of tumor development, and its role in TAMs deserve to be investigated in depth. This review builds on this foundation to further explore the multiple roles of UPS on TAMs and identifies a promising approach to treat tumors by targeting TAMs with UPS.
Collapse
Affiliation(s)
- Yue Xiao
- First School of Clinical Medicine, Nanchang University, Nanchang, China
| | - Ruiqian Liu
- School of Future Technology, Nanchang University, Nanchang, China
| | - Na Li
- School of Future Technology, Nanchang University, Nanchang, China
| | - Yong Li
- Department of Anesthesiology, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xuan Huang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, China
| |
Collapse
|
8
|
Di Gregorio J, Appignani M, Flati V. Role of the Mitochondrial E3 Ubiquitin Ligases as Possible Therapeutic Targets in Cancer Therapy. Int J Mol Sci 2023; 24:17176. [PMID: 38139010 PMCID: PMC10743160 DOI: 10.3390/ijms242417176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 11/27/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023] Open
Abstract
Ubiquitination is a post-translational modification that targets specific proteins on their lysine residues. Depending on the type of ubiquitination, this modification ultimately regulates the stability or degradation of the targeted proteins. Ubiquitination is mediated by three different classes of enzymes: the E1 ubiquitin-activating enzymes, the E2 ubiquitin-conjugating enzymes and, most importantly, the E3 ubiquitin ligases. E3 ligases are responsible for the final step of the ubiquitin cascade, interacting directly with the target proteins. E3 ligases can also be involved in DNA repair, cell cycle regulation and response to stress; alteration in their levels can be involved in oncogenic transformation and cancer progression. Of all the six hundred E3 ligases of the human genome, only three of them are specific to the mitochondrion: MARCH5, RNF185 and MUL1. Their alterations (that reflect on the alteration of the mitochondria functions) can be related to cancer progression, as underlined by the increasing research performed in recent years on these three mitochondrial enzymes. This review will focus on the function and mechanisms of the mitochondrial E3 ubiquitin ligases, as well as their important targets, in cancer development and progression, also highlighting their potential use for cancer therapy.
Collapse
Affiliation(s)
| | | | - Vincenzo Flati
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (J.D.G.); (M.A.)
| |
Collapse
|
9
|
Wang W, Jiang K, Liu X, Li J, Zhou W, Wang C, Cui J, Liang T. FBXW7 and human tumors: mechanisms of drug resistance and potential therapeutic strategies. Front Pharmacol 2023; 14:1278056. [PMID: 38027013 PMCID: PMC10680170 DOI: 10.3389/fphar.2023.1278056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
Drug therapy, including chemotherapy, targeted therapy, immunotherapy, and endocrine therapy, stands as the foremost therapeutic approach for contemporary human malignancies. However, increasing drug resistance during antineoplastic therapy has become a substantial barrier to favorable outcomes in cancer patients. To enhance the effectiveness of different cancer therapies, an in-depth understanding of the unique mechanisms underlying tumor drug resistance and the subsequent surmounting of antitumor drug resistance is required. Recently, F-box and WD Repeat Domain-containing-7 (FBXW7), a recognized tumor suppressor, has been found to be highly associated with tumor therapy resistance. This review provides a comprehensive summary of the underlying mechanisms through which FBXW7 facilitates the development of drug resistance in cancer. Additionally, this review elucidates the role of FBXW7 in therapeutic resistance of various types of human tumors. The strategies and challenges implicated in overcoming tumor therapy resistance by targeting FBXW7 are also discussed.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Tingting Liang
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
10
|
Jiang Y, Ni S, Xiao B, Jia L. Function, mechanism and drug discovery of ubiquitin and ubiquitin-like modification with multiomics profiling for cancer therapy. Acta Pharm Sin B 2023; 13:4341-4372. [PMID: 37969742 PMCID: PMC10638515 DOI: 10.1016/j.apsb.2023.07.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 05/21/2023] [Accepted: 07/17/2023] [Indexed: 11/17/2023] Open
Abstract
Ubiquitin (Ub) and ubiquitin-like (Ubl) pathways are critical post-translational modifications that determine whether functional proteins are degraded or activated/inactivated. To date, >600 associated enzymes have been reported that comprise a hierarchical task network (e.g., E1-E2-E3 cascade enzymatic reaction and deubiquitination) to modulate substrates, including enormous oncoproteins and tumor-suppressive proteins. Several strategies, such as classical biochemical approaches, multiomics, and clinical sample analysis, were combined to elucidate the functional relations between these enzymes and tumors. In this regard, the fundamental advances and follow-on drug discoveries have been crucial in providing vital information concerning contemporary translational efforts to tailor individualized treatment by targeting Ub and Ubl pathways. Correspondingly, emphasizing the current progress of Ub-related pathways as therapeutic targets in cancer is deemed essential. In the present review, we summarize and discuss the functions, clinical significance, and regulatory mechanisms of Ub and Ubl pathways in tumorigenesis as well as the current progress of small-molecular drug discovery. In particular, multiomics analyses were integrated to delineate the complexity of Ub and Ubl modifications for cancer therapy. The present review will provide a focused and up-to-date overview for the researchers to pursue further studies regarding the Ub and Ubl pathways targeted anticancer strategies.
Collapse
Affiliation(s)
| | | | - Biying Xiao
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Lijun Jia
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| |
Collapse
|
11
|
Wang C, Huang M, Lin Y, Zhang Y, Pan J, Jiang C, Cheng M, Li S, He W, Li Z, Tu Z, Fan J, Zeng H, Lin J, Wang Y, Yao N, Liu T, Qi Q, Liu X, Zhang Z, Chen M, Xia L, Zhang D, Ye W. ENO2-derived phosphoenolpyruvate functions as an endogenous inhibitor of HDAC1 and confers resistance to antiangiogenic therapy. Nat Metab 2023; 5:1765-1786. [PMID: 37667133 DOI: 10.1038/s42255-023-00883-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/31/2023] [Indexed: 09/06/2023]
Abstract
Metabolic reprogramming is associated with resistance to antiangiogenic therapy in cancer. However, its molecular mechanisms have not been clearly elucidated. Here, we identify the glycolytic enzyme enolase 2 (ENO2) as a driver of resistance to antiangiogenic therapy in colorectal cancer (CRC) mouse models and human participants. ENO2 overexpression induces neuroendocrine differentiation, promotes malignant behaviour in CRC and desensitizes CRC to antiangiogenic drugs. Mechanistically, the ENO2-derived metabolite phosphoenolpyruvate (PEP) selectively inhibits histone deacetylase 1 (HDAC1) activity, which increases the acetylation of β-catenin and activates the β-catenin pathway in CRC. Inhibition of ENO2 with enolase inhibitors AP-III-a4 or POMHEX synergizes the efficacy of antiangiogenic drugs in vitro and in mice bearing drug-resistant CRC xenograft tumours. Together, our findings reveal that ENO2 constitutes a useful predictive biomarker and therapeutic target for resistance to antiangiogenic therapy in CRC, and uncover a previously undefined and metabolism-independent role of PEP in regulating resistance to antiangiogenic therapy by functioning as an endogenous HDAC1 inhibitor.
Collapse
Affiliation(s)
- Chenran Wang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- The First Affiliated Hospital of Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Maohua Huang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Yuning Lin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Yiming Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Jinghua Pan
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Chang Jiang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Minjing Cheng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Shenrong Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Wenzhuo He
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Zhengqiu Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Zhengchao Tu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Jun Fan
- School of Medicine, Jinan University, Guangzhou, China
| | - Huhu Zeng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Jiahui Lin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Yongjin Wang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Nan Yao
- School of Medicine, Jinan University, Guangzhou, China
| | - Tongzheng Liu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Qi Qi
- School of Medicine, Jinan University, Guangzhou, China
| | - Xiangning Liu
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Zhimin Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Minfeng Chen
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China.
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China.
| | - Liangping Xia
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| | - Dongmei Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China.
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China.
| | - Wencai Ye
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China.
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China.
| |
Collapse
|
12
|
Lu X, Liu R, Liao Y, Cui L, Sun H, Zhang D, Wang B, Fang L, Guan X, Yao Y, Liu C, Zhang Y. ACVRL1 drives resistance to multitarget tyrosine kinase inhibitors in colorectal cancer by promoting USP15-mediated GPX2 stabilization. BMC Med 2023; 21:366. [PMID: 37743483 PMCID: PMC10518977 DOI: 10.1186/s12916-023-03066-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 09/04/2023] [Indexed: 09/26/2023] Open
Abstract
BACKGROUND Multitarget tyrosine kinase inhibitors (mTKIs) such as Regorafenib and Sorafenib have already been approved for the treatment of many solid tumours. However, the efficacy of mTKIs in colorectal cancer (CRC) is limited; the underlined mechanism remains largely elusive. Our study was aimed to find out the resistance mechanism of mTKIs in CRC. METHODS RNA sequencing was used to identify the expression of Activin A receptor-like type 1 (ACVRL1) under the treatment of mTKIs. Gain/loss-of-function experiments were performed to assess the biological function of ACVRL1 in resistance to mTKIs. The underlying mechanisms of ACVRL1-mediated mTKI resistance were investigated by using liquid chromatography-mass spectrometry assays (LC-MS), co-immunoprecipitation assays (Co-IP), chromatin immunoprecipitation assays, ubiquitination assays, dual luciferase reporter assays, etc. RESULTS: RNA sequencing identified the activation of ACVRL1 under the treatment of mTKIs in CRC cells. ACVRL1 knockdown and overexpression significantly affects the sensitivity of CRC cells to mTKIs both in vitro and vivo. Mechanistically, we found the β-catenin/TCF-1-KCNQ1OT1/miR-7-5p axis mediated the activation of ACVRL1. Furthermore, LC-MS assays indicated the interaction between ACVRL1 and glutathione peroxidase 2(GPX2) protein. IP assay defined ACVRL1 truncation (282-503aa) could be responsible for interacting with GPX2, and rescue experiments with ACVRL1 truncations confirmed the importance of this interaction in driving mTKI resistance. Co-IP assays confirmed that ACVRL1 associates with ubiquitin-specific peptidase 15(USP15) which directly deubiquinates GPX2 at the K187(K, lysine) site, leading to the accumulation of GPX2 protein. Rescue experiments performed with the lysine mutants in GPX2 CRISPR knockout cell model confirmed the importance of GPX2 K187 mutant. As a result, the increased ROS clearance and decreased cell apoptosis eventually lead to mTKI resistance in CRC. CONCLUSIONS Our results demonstrate that the Wnt/β-catenin/KCNQ1OT1/miR-7-5p/ACVRL1/GPX2 biological axis plays a vital role in CRC, targeting which may be an effective approach for overcoming mTKI resistance.
Collapse
Affiliation(s)
- Xiaolin Lu
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China
- China Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China
- Department of Orthopedic Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ruiqi Liu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yuanyu Liao
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Luying Cui
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Haoxiu Sun
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Dongzhi Zhang
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Bojun Wang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lin Fang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xin Guan
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuanfei Yao
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China.
- China Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China.
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China.
| | - Chao Liu
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China.
- China Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China.
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China.
| | - Yanqiao Zhang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China.
- China Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China.
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China.
| |
Collapse
|
13
|
Tong J, Tan X, Hao S, Ermine K, Lu X, Liu Z, Jha A, Yu J, Zhang L. Inhibition of multiple CDKs potentiates colon cancer chemotherapy via p73-mediated DR5 induction. Oncogene 2023; 42:869-880. [PMID: 36721000 PMCID: PMC10364554 DOI: 10.1038/s41388-023-02598-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 02/01/2023]
Abstract
Targeting cyclin-dependent kinases (CDKs) has recently emerged as a promising therapeutic approach against cancer. However, the anticancer mechanisms of different CDK inhibitors (CDKIs) are not well understood. Our recent study revealed that selective CDK4/6 inhibitors sensitize colorectal cancer (CRC) cells to therapy-induced apoptosis by inducing Death Receptor 5 (DR5) via the p53 family member p73. In this study, we investigated if this pathway is involved in anticancer effects of different CDKIs. We found that less-selective CDKIs, including flavopiridol, roscovitine, dinaciclib, and SNS-032, induced DR5 via p73-mediated transcriptional activation. The induction of DR5 by these CDKIs was mediated by dephosphorylation of p73 at Threonine 86 and p73 nuclear translocation. Knockdown of a common target of these CDKIs, including CDK1, 2, or 9, recapitulated p73-mediated DR5 induction. CDKIs strongly synergized with 5-fluorouracil (5-FU), the most commonly used CRC chemotherapy agent, in vitro and in vivo to promote growth suppression and apoptosis, which required DR5 and p73. Together, these findings indicate p73-mediated DR5 induction as a potential tumor suppressive mechanism and a critical target engaged by different CDKIs in potentiating therapy-induced apoptosis in CRC cells. These findings help better understand the anticancer mechanisms of CDKIs and may help facilitate their clinical development and applications in CRC.
Collapse
Affiliation(s)
- Jingshan Tong
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Xiao Tan
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Suisui Hao
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Kaylee Ermine
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Xinyan Lu
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Zhaojin Liu
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Anupma Jha
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Jian Yu
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Lin Zhang
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA.
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
14
|
Chai C, Ji P, Xu H, Tang H, Wang Z, Zhang H, Zhou W. Targeting cancer drug resistance utilizing organoid technology. Biomed Pharmacother 2023; 158:114098. [PMID: 36528918 DOI: 10.1016/j.biopha.2022.114098] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/04/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Cancer organoids generated from 3D in vitro cell cultures have contributed to the study of drug resistance. Maintenance of genomic and transcriptomic similarity between organoids and parental cancer allows organoids to have the ability of accurate prediction in drug resistance testing. Protocols of establishing therapy-sensitive and therapy-resistant organoids are concluded in two aspects, which are generated directly from respective patients' cancer and by induction of anti-cancer drug. Genomic and transcriptomic analyses and gene editing have been applied to organoid studies to identify key targets in drug resistance and FGFR3, KHDRBS3, lnc-RP11-536 K7.3 and FBN1 were found to be key targets. Furthermore, mechanisms contributing to resistance have been identified, including metabolic adaptation, activation of DNA damage response, defects in apoptosis, reduced cellular senescence, cellular plasticity, subpopulation interactions and gene fusions. Additionally, cancer stem cells (CSCs) have been verified to be involved in drug resistance utilizing organoid technology. Reversal of drug resistance can be achieved by targeting key genes and CSCs in cancer organoids. In this review, we summarize applications of organoids to cancer drug resistance research, indicating prospects and limitations.
Collapse
Affiliation(s)
- Changpeng Chai
- The First Hospital of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu, China; The Forth Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, Gansu, China.
| | - Pengfei Ji
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, Gansu, China.
| | - Hao Xu
- The First Hospital of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu, China.
| | - Huan Tang
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, Gansu, China.
| | - Zhengfeng Wang
- The First Hospital of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu, China.
| | - Hui Zhang
- The Second Hospital of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu, China.
| | - Wence Zhou
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou 730000, Gansu, China; The Second Hospital of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu, China.
| |
Collapse
|
15
|
Shahidi S, Rostamizadeh K, Fathi M, Nedaei K, Ramazani A. Combination of Quercetin or/and siRNA-loaded DDAB-mPEG-PCL hybrid nanoparticles reverse resistance to Regorafenib in colon cancer cells. BMC Complement Med Ther 2022; 22:340. [PMID: 36575448 PMCID: PMC9793538 DOI: 10.1186/s12906-022-03787-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 11/10/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the second leading cause of cancer death. Although Regorafenib showed survival benefits in patients with CRC, reports imply the recurrence of malignant phenotype resulting from chemotherapy. Evidence demonstrated that a5β1 integrin plays an important role in the Regorafenib treatment, which, may be led to resistance. In this study, the effects of /siRNA or/ and Quercetin loaded DDAB-mPEG-PCLnanoparticles could reverse this resistance phenotype in colon cancer cells in vitro. METHODS Regorafenib-resistant Ls-180 colon cancer cell line was developed by long-term exposure to Regorafenib. Quercetin and Regorafenib were separately encapsulated into mPEG-PCL micelles through the nano-precipitation method and characterized by DLS. Optimized doses of Quercetin and Regorafenib were used for combination therapy of resistant cells followed cytotoxicity study using MTT. Gene expression levels of the β1 subunit of integrin were determined by the real-time method of RT-PCR. RESULTS Developed Regorafenib resistant LS-180 showed to have Regorafenib IC50 of 38.96 ± 1.72 µM whereas IC50 in non-resistant cells were 8.51 ± 0.29 µM, which meaningful was lower statistically compared to that of a resistant one. The β1 mRNA level of whole α5β1 integrin was significantly higher in the resistant cells compared to those of non-resistant ones. Gene expression levels in each siRNA-loaded nanoparticle and Quercetin-loaded one were lower than that in mock experiments. Finally, when these two types of nanoparticles were used to treat resistant cells, gene expression decrease of integrin indicated a greater effect that could be capable of reverse resistancy. CONCLUSION Results of this study demonstrated another confirmation of involving integrins in cancer resistance following chemotherapy using Regorafenib. Also, it indicated how using siRNA targeting integrin could enhance the plant derivatives like Quercetin effects to reverse resistance in vitro.
Collapse
Affiliation(s)
- Shabnam Shahidi
- grid.469309.10000 0004 0612 8427Department of Clinical Biochemistry, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Kobra Rostamizadeh
- grid.469309.10000 0004 0612 8427Department of Pharmaceutical Biomaterial, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran ,grid.469309.10000 0004 0612 8427Zanjan Pharmaceutical Nanotechnology Research Center, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mojtaba Fathi
- grid.469309.10000 0004 0612 8427Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran ,grid.412606.70000 0004 0405 433XDepartment of Biochemistry and Genetics, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Keivan Nedaei
- grid.469309.10000 0004 0612 8427Department of Medical Biotechnology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Ramazani
- grid.469309.10000 0004 0612 8427Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran ,grid.469309.10000 0004 0612 8427Department of Pharmaceutical Biotechnology, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
16
|
Kuang C, Tong J, Ermine K, Cai M, Dai F, Hao S, Giles F, Huang Y, Yu J, Zhang L. Dual inhibition of BET and HAT/p300 suppresses colorectal cancer via DR5- and p53/PUMA-mediated cell death. Front Oncol 2022; 12:1018775. [PMID: 36313707 PMCID: PMC9599411 DOI: 10.3389/fonc.2022.1018775] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 09/26/2022] [Indexed: 01/30/2023] Open
Abstract
Background Colorectal cancer (CRC) frequently has a dysregulated epigenome causing aberrant up-regulation of oncogenes such as c-MYC. Bromodomain and extra-terminal domain (BET) proteins and histone acetyltransferases (HAT) are epigenetic regulatory proteins that create and maintain epigenetic states supporting oncogenesis. BET inhibitors and HAT inhibitors are currently being investigated as cancer therapeutics due to their ability to suppress cancer-promoting epigenetic modifiers. Due to the extensive molecular crosstalk between BET proteins and HAT proteins, we hypothesized that dual inhibition of BET and HAT could more potently inhibit CRC cells than inhibition of each individual protein. Methods We investigated the activity and mechanisms of a dual BET and HAT inhibitor, NEO2734, in CRC cell lines and mouse xenografts. MTS, flow cytometry, and microscopy were used to assess cell viability. qPCR, Western blotting, and immunofluorescent staining were used to assess mechanisms of action. Results We found that NEO2734 more potently suppresses CRC cell growth than first generation BET inhibitors, regardless of the status of common CRC driver mutations. We previously showed that BET inhibitors upregulate DR5 to induce extrinsic apoptosis. In the current study, we show that NEO2734 treatment induces CRC cell apoptosis via both the intrinsic and extrinsic apoptosis pathways. NEO2734 increases p53 expression and subsequently increased expression of the p53-upregulated mediator of apoptosis (PUMA), which is a critical mechanism for activating intrinsic apoptosis. We demonstrate that inhibition of either the intrinsic or extrinsic branches of apoptosis partially rescues CRC cells from NEO2734 treatment, while the dual inhibition of both branches of apoptosis more strongly rescues CRC cells from NEO2734 treatment. Finally, we show that NEO2734 monotherapy is able to suppress tumor growth in CRC xenografts by inducing apoptosis. Conclusions Our study demonstrates NEO2734 potently suppresses CRC cells in vitro and in vivo by simultaneously upregulating PUMA and DR5 to induce cell death. Further studies of NEO2734 for treating CRC are warranted.
Collapse
Affiliation(s)
- Chaoyuan Kuang
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jingshan Tong
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kaylee Ermine
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Manbo Cai
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Fujun Dai
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Suisui Hao
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Francis Giles
- Developmental Therapeutics Consortium, Chicago, IL, United States
| | - Yi Huang
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jian Yu
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Lin Zhang
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
17
|
Pan S, Zhang X, Guo Y, Li Y. DPCPX induces Bim-dependent apoptosis in nasopharyngeal carcinoma cells. Cell Biol Int 2022; 46:2050-2059. [PMID: 35989488 DOI: 10.1002/cbin.11887] [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: 12/26/2021] [Revised: 07/08/2022] [Accepted: 08/05/2022] [Indexed: 11/07/2022]
Abstract
ADORA1 promotes tumor growth and development in multiple cancers. DPCPX (a selective adenosine A1 receptor antagonist), a specific ADORA1 antagonist, has shown antitumor effects in many cancer types. Nevertheless, the function of DPCPX in nasopharyngeal carcinoma (NPC) still remains to be unraveled. In this study, we investigated the functional role of DPCPX on NPC cells. We found that DPCPX promotes NPC cells growth inhibition. DPCPX induced Bim-dependent apoptosis in NPC cells irrespective of p53 status via the FoxO3a pathway following PI3K/AKT inhibition. Furthermore, DPCPX enhanced the antitumor effect of cisplatin, 5-FU and Paclitaxel in NPC. Xenograft experiment revealed that deficiency of Bim in vivo stalls apoptosis and antitumor activity of DPCPX. In conclusion, the PI3K/AKT/FoxO3a/Bim axis plays a critical role in the anticancer effects of DPCPX in NPC.
Collapse
Affiliation(s)
- Suming Pan
- Department of Radiation Oncology, Yue Bei People's Hospital, Shaoguan, China
| | - Xiangguo Zhang
- Department of Radiation Oncology, Yue Bei People's Hospital, Shaoguan, China
| | - Yugan Guo
- Department of Radiation Oncology, Yue Bei People's Hospital, Shaoguan, China
| | - Yin Li
- Faculty of education, Shaoguan University, Shaoguan, China
| |
Collapse
|
18
|
Sulkshane P, Teni T. Myeloid cell leukemia-1: a formidable barrier to anticancer therapeutics and the quest of targeting it. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2022; 3:278-296. [PMID: 36045907 PMCID: PMC9400788 DOI: 10.37349/etat.2022.00083] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 03/16/2022] [Indexed: 11/22/2022] Open
Abstract
The antiapoptotic B cell lymphoma-2 (Bcl-2) family members are apical regulators of the intrinsic pathway of apoptosis that orchestrate mitochondrial outer membrane permeabilization (MOMP) through interactions with their proapoptotic counterparts. Overexpression of antiapoptotic Bcl-2 family proteins has been linked to therapy resistance and poor prognosis in diverse cancers. Among the antiapoptotic Bcl-2 family members, predominant overexpression of the prosurvival myeloid cell leukemia-1 (Mcl-1) has been reported in a myriad of hematological malignancies and solid tumors, contributing to therapy resistance and poor outcomes, thus making it a potential druggable target. The unique structure of Mcl-1 and its complex regulatory mechanism makes it an adaptive prosurvival switch that ensures tumor cell survival despite therapeutic intervention. This review focusses on diverse mechanisms adopted by tumor cells to maintain sustained elevated levels of Mcl-1 and how high Mcl-1 levels contribute to resistance in conventional as well as targeted therapies. Moreover, recent developments in the Mcl-1-targeted therapeutics and the underlying challenges and considerations in designing novel Mcl-1 inhibitors are also discussed.
Collapse
Affiliation(s)
- Prasad Sulkshane
- Glickman Laboratory, Faculty of Biology, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Tanuja Teni
- Teni Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India; Homi Bhabha National Institute, Training School Complex, Mumbai 400094, India
| |
Collapse
|
19
|
Shen W, Zhou Q, Peng C, Li J, Yuan Q, Zhu H, Zhao M, Jiang X, Liu W, Ren C. FBXW7 and the Hallmarks of Cancer: Underlying Mechanisms and Prospective Strategies. Front Oncol 2022; 12:880077. [PMID: 35515121 PMCID: PMC9063462 DOI: 10.3389/fonc.2022.880077] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 03/15/2022] [Indexed: 12/13/2022] Open
Abstract
FBXW7, a member of the F-box protein family within the ubiquitin–proteasome system, performs an indispensable role in orchestrating cellular processes through ubiquitination and degradation of its substrates, such as c-MYC, mTOR, MCL-1, Notch, and cyclin E. Mainly functioning as a tumor suppressor, inactivation of FBXW7 induces the aberrations of its downstream pathway, resulting in the occurrence of diseases especially tumorigenesis. Here, we decipher the relationship between FBXW7 and the hallmarks of cancer and discuss the underlying mechanisms. Considering the interplay of cancer hallmarks, we propose several prospective strategies for circumventing the deficits of therapeutic resistance and complete cure of cancer patients.
Collapse
Affiliation(s)
- Wenyue Shen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Quanwei Zhou
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Chenxi Peng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jiaheng Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qizhi Yuan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hecheng Zhu
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China.,Changsha Kexin Cancer Hospital, Changsha, China
| | - Ming Zhao
- Changsha Kexin Cancer Hospital, Changsha, China
| | - Xingjun Jiang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Weidong Liu
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, School of Basic Medicine, Central South University, Changsha, China
| | - Caiping Ren
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, School of Basic Medicine, Central South University, Changsha, China
| |
Collapse
|
20
|
Tong J, Tan X, Song X, Gao M, Risnik D, Hao S, Ermine K, Wang P, Li H, Huang Y, Yu J, Zhang L. CDK4/6 Inhibition Suppresses p73 Phosphorylation and Activates DR5 to Potentiate Chemotherapy and Immune Checkpoint Blockade. Cancer Res 2022; 82:1340-1352. [PMID: 35149588 PMCID: PMC8983601 DOI: 10.1158/0008-5472.can-21-3062] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 01/12/2022] [Accepted: 02/08/2022] [Indexed: 11/16/2022]
Abstract
Targeting cyclin-dependent kinases 4 and 6 (CDK4/6) is a successful therapeutic approach against breast and other solid tumors. Inhibition of CDK4/6 halts cell cycle progression and promotes antitumor immunity. However, the mechanisms underlying the antitumor activity of CDK4/6 inhibitors are not fully understood. We found that CDK4/6 bind and phosphorylate the p53 family member p73 at threonine 86, which sequesters p73 in the cytoplasm. Inhibition of CDK4/6 led to dephosphorylation and nuclear translocation of p73, which transcriptionally activated death receptor 5 (DR5), a cytokine receptor and key component of the extrinsic apoptotic pathway. p73-mediated induction of DR5 by CDK4/6 inhibitors promoted immunogenic cell death of cancer cells. Deletion of DR5 in cancer cells in vitro and in vivo abrogated the potentiating effects of CDK4/6 inhibitors on immune cytokine TRAIL, 5-fluorouracil chemotherapy, and anti-PD-1 immunotherapy. Together, these results reveal a previously unrecognized consequence of CDK4/6 inhibition, which may be critical for potentiating the killing and immunogenic effects on cancer cells. SIGNIFICANCE This work demonstrates how inhibition of CDK4/6 sensitizes cancer cells to chemotherapy and immune checkpoint blockade and may provide a new molecular marker for improving CDK4/6-targeted cancer therapies. See related commentary by Frank, p. 1170.
Collapse
Affiliation(s)
- Jingshan Tong
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Xiao Tan
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Xiangping Song
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Man Gao
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213. USA
| | - Denise Risnik
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Suisui Hao
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Kaylee Ermine
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Peng Wang
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Hua Li
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Yi Huang
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jian Yu
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213. USA
| | - Lin Zhang
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
21
|
Roberto M, Arrivi G, Pilozzi E, Montori A, Balducci G, Mercantini P, Laghi A, Ierinò D, Panebianco M, Marinelli D, Tomao S, Marchetti P, Mazzuca F. The Potential Role of Genomic Signature in Stage II Relapsed Colorectal Cancer (CRC) Patients: A Mono-Institutional Study. Cancer Manag Res 2022; 14:1353-1369. [PMID: 35418781 PMCID: PMC9000544 DOI: 10.2147/cmar.s342612] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Purpose The absolute benefit of adjuvant chemotherapy in stage II CRC is only 3–4%. The identification of biomarkers through molecular profiling could identify patients who will more benefit from adjuvant chemotherapy. Patients and Methods This retrospective analysis examined tissue blocks from 17 patients affected by relapsed stage II CRC, whose comprehensive genomic profiling of tumors was conducted through next-generation sequencing (NGS) via Roche-FoundationOne®. Results Mutations were found in APC (76.5%), TP53 (58.8%) and KRAS (52.9%). Only KRAS wild-type samples showed FBXW7. APC frameshift mutations and MLH1 splice variant were conversely significant correlated (7% v 93%, P = 0.014). The median number of gene mutations reported was 6 (range 2–14). The TP53 mutation was associated most frequently with lung metastasis (P = 0.07) and high tumor budding (P = 0.03). Despite no statistical significance, lung recurrence, LVI/Pni, MSI and more than 6 genetic mutations were correlated to worse DFS and OS. Patients carried co-mutations of TP53-FBXW7 reported the worse DFS (4 v 14 months) and OS (4 v 65 months) compared to the other patients. Conclusion According to the present analysis, the setting of relapsed CRC emerges as one of the fields of greatest utility for NGS, looking at personalized cancer care.
Collapse
Affiliation(s)
- Michela Roberto
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, Medical Oncology Unit A, Policlinico Umberto I, “Sapienza” University of Rome, Rome, Italy
| | - Giulia Arrivi
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Oncology Unit, Sant’ Andrea University Hospital, Rome, Italy
- Correspondence: Giulia Arrivi, Department of Clinical and Molecular Medicine, Sapienza University of Rome, Oncology Unit, Sant’ Andrea University Hospital, Via di Grottarossa 1035-1039, Rome, 00189, Italy, Tel +39 3387231524, Fax +39 0633776629, Email
| | - Emanuela Pilozzi
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Anatomia Patologica Unit, Sant’ Andrea University Hospital, Rome, Italy
| | - Andrea Montori
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Anatomia Patologica Unit, Sant’ Andrea University Hospital, Rome, Italy
| | - Genoveffa Balducci
- Department of Medical-Surgical Sciences and Translation Medicine, Sapienza University of Rome, Gastro-intestinal Surgery Unit, Sant’ Andrea University Hospital, Rome, Italy
| | - Paolo Mercantini
- Department of Medical-Surgical Sciences and Translation Medicine, Sapienza University of Rome, Gastro-intestinal Surgery Unit, Sant’ Andrea University Hospital, Rome, Italy
| | - Andrea Laghi
- Department of Medical-Surgical Sciences and Translation Medicine, Sapienza University of Rome, Radiology Unit, Sant’ Andrea University Hospital, Rome, Italy
| | - Debora Ierinò
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Oncology Unit, Sant’ Andrea University Hospital, Rome, Italy
| | - Martina Panebianco
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Oncology Unit, Sant’ Andrea University Hospital, Rome, Italy
| | - Daniele Marinelli
- Medical Oncology Unit B, Policlinico Umberto I, Sapienza University, Rome, Italy
| | - Silverio Tomao
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, Medical Oncology Unit A, Policlinico Umberto I, “Sapienza” University of Rome, Rome, Italy
| | - Paolo Marchetti
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Oncology Unit, Sant’ Andrea University Hospital, Rome, Italy
| | - Federica Mazzuca
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Oncology Unit, Sant’ Andrea University Hospital, Rome, Italy
| |
Collapse
|
22
|
Fan J, Bellon M, Ju M, Zhao L, Wei M, Fu L, Nicot C. Clinical significance of FBXW7 loss of function in human cancers. Mol Cancer 2022; 21:87. [PMID: 35346215 PMCID: PMC8962602 DOI: 10.1186/s12943-022-01548-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/22/2022] [Indexed: 12/13/2022] Open
Abstract
FBXW7 (F-Box and WD Repeat Domain Containing 7) (also referred to as FBW7 or hCDC4) is a component of the Skp1-Cdc53 / Cullin-F-box-protein complex (SCF/β-TrCP). As a member of the F-box protein family, FBXW7 serves a role in phosphorylation-dependent ubiquitination and proteasome degradation of oncoproteins that play critical role(s) in oncogenesis. FBXW7 affects many regulatory functions involved in cell survival, cell proliferation, tumor invasion, DNA damage repair, genomic instability and telomere biology. This thorough review of current literature details how FBXW7 expression and functions are regulated through multiple mechanisms and how that ultimately drives tumorigenesis in a wide array of cell types. The clinical significance of FBXW7 is highlighted by the fact that FBXW7 is frequently inactivated in human lung, colon, and hematopoietic cancers. The loss of FBXW7 can serve as an independent prognostic marker and is significantly correlated with the resistance of tumor cells to chemotherapeutic agents and poorer disease outcomes. Recent evidence shows that genetic mutation of FBXW7 differentially affects the degradation of specific cellular targets resulting in a distinct and specific pattern of activation/inactivation of cell signaling pathways. The clinical significance of FBXW7 mutations in the context of tumor development, progression, and resistance to therapies as well as opportunities for targeted therapies is discussed.
Collapse
Affiliation(s)
- Jingyi Fan
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute; Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China.,Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China.,Liaoning Province, China Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, 110122, Liaoning Province, China
| | - Marcia Bellon
- Department of Pathology and Laboratory Medicine, Center for Viral Pathogenesis, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - Mingyi Ju
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China.,Liaoning Province, China Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, 110122, Liaoning Province, China
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China.,Liaoning Province, China Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, 110122, Liaoning Province, China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China.,Liaoning Province, China Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, 110122, Liaoning Province, China
| | - Liwu Fu
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute; Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China.
| | - Christophe Nicot
- Department of Pathology and Laboratory Medicine, Center for Viral Pathogenesis, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA.
| |
Collapse
|
23
|
Tian H, Zhao H, Qu B, Chu X, Xin X, Zhang Q, Li W, Yang S. TRIM24 promotes colorectal cancer cell progression via the Wnt/β-catenin signaling pathway activation. Am J Transl Res 2022; 14:831-848. [PMID: 35273688 PMCID: PMC8902576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 10/12/2021] [Indexed: 06/14/2023]
Abstract
Overexpression of TRIM24 is observed in several human cancers and is correlated with an increase in the progression and metastasis of tumors. In this study, we investigated the changes in activity and biochemical events that occur after overexpression of TRIM24 in a colorectal cancer (CRC) mouse model. We observed upregulated TRIM24 expression in CRC tissues compared to that in nonneoplastic adjacent tissues. Enhanced expression of TRIM24 was significantly associated with the status of lymph nodes and poor recurrence-free survival of patients with CRC. The role of TRIM24 in CRC tumor growth was investigated using an orthotopic model of MC38 mouse colon cancer cells overexpressing TRIM24, and CRC tumor growth was found to increase dramatically by TRIM24 overexpression. Moreover, angiogenesis was stimulated by TRIM24 overexpression via the upregulation of vascular endothelial growth factor (VEGF) expression. Overexpression of TRIM24 in MC38 cells led to an increase in the protein levels of ALDH1 and other stem cell markers. In addition, we observed that Wnt/β-catenin signaling is required for the function of TRIM24 in CRC cells. Tumor-associated macrophages (TAMs) were found to be recruited by tumor cells overexpressing TRIM24 via the increased expression of CCL2/5, CSF-1, and VEGF, further enhancing CRC tumor growth. In conclusion, overexpression of TRIM24 facilitates the growth of CRC and the remodeling of the tumor stroma via angiogenesis stimulation and TAM recruitment. The Wnt/β-catenin pathway is a possible crucial link in the TRIM24-associated progression of tumors, which may provide opportunities for pharmacological intervention.
Collapse
Affiliation(s)
- Hong Tian
- Oncology Department, The 4th People’s Hospital of ShenyangShenyang 110013, Liaoning, China
| | - Hongmei Zhao
- Department of Laboratory Medicine, The People’s Hospital of China Medical University (The People’s Hospital of Liaoning Province)Shenyang 110016, Liaoning, China
| | - Bo Qu
- Department of Laboratory Medicine, The People’s Hospital of China Medical University (The People’s Hospital of Liaoning Province)Shenyang 110016, Liaoning, China
| | - Xiaoli Chu
- Oncology Department, The 4th People’s Hospital of ShenyangShenyang 110013, Liaoning, China
| | - Xing Xin
- Oncology Department, The 4th People’s Hospital of ShenyangShenyang 110013, Liaoning, China
| | - Qingwei Zhang
- General Surgery Dept. VI Ward (Biliary-Pancreatic Surgery), The People’s Hospital of China Medical University (The People’s Hospital of Liaoning Province)Shenyang 110016, Liaoning, China
| | - Weizhou Li
- Department of Laboratory Medicine, The People’s Hospital of China Medical University (The People’s Hospital of Liaoning Province)Shenyang 110016, Liaoning, China
| | - Shida Yang
- Department of Laboratory Medicine, The People’s Hospital of China Medical University (The People’s Hospital of Liaoning Province)Shenyang 110016, Liaoning, China
| |
Collapse
|
24
|
Comprehensive Study of Human FBXW7 Deleterious nsSNP's Functional Inference and Susceptibility to Gynaecological Cancer. Appl Biochem Biotechnol 2021; 194:407-433. [PMID: 34817806 DOI: 10.1007/s12010-021-03759-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 11/08/2021] [Indexed: 10/19/2022]
Abstract
Cancer is one of the world's major causes of mortality, and it plays a most important role in the world's declining life expectancy. F-box and WD-40 domain protein 7 (FBXW7), a typical participant of the F-box family of proteins, has been considered as an anti-tumor protein and one of the maximum deregulated ubiquitin-proteasome system proteins in uterine carcinosarcoma, endometrial clear cell carcinoma and cervical carcinoma with the greatest prevalence of alterations. FBXW7 variants with known clinical significance, as well as non-synonymous single nucleotide polymorphisms (nsSNPs) in the F-Box and WD40 domains, were evaluated using functionality prediction web resources. Upon analysing the seventy-three deleterious nsSNP's impact on protein stability and function, we identified that forty-one nsSNPs of WD40 domain and three of F-Box domain imply decreased stability of the FBXW7 structure. Next to TP53 and PTEN, FBXW7 was reported with the highest percentage of arginine substitution among mutations related to cancer. The current research concentrated on two arginine residue locations (Arg465, Arg505) within the WD40-repeat domain, which is vital for substrate binding. Computational analysis revealed significant deviation in stability and structural configuration of mutants R505L, R465H, R465P, R505G, R505C, R465C, R505S and R505L structures. Protein-protein interaction network of FBXW7 populated with promising hub proteins NOTCH1, c-Myc, CCNE1, STYX, KLG5, SREB1, NFKB2, SKP1 and CUL1; thus, alteration in the FBXW7 leads to aberration in their signalling pathways as well as their substrate binding ability makes this protein as attractive target for personalized therapeutic intervention.
Collapse
|
25
|
Liu J, Tokheim C, Lee JD, Gan W, North BJ, Liu XS, Pandolfi PP, Wei W. Genetic fusions favor tumorigenesis through degron loss in oncogenes. Nat Commun 2021; 12:6704. [PMID: 34795215 PMCID: PMC8602260 DOI: 10.1038/s41467-021-26871-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 10/27/2021] [Indexed: 12/12/2022] Open
Abstract
Chromosomal rearrangements can generate genetic fusions composed of two distinct gene sequences, many of which have been implicated in tumorigenesis and progression. Our study proposes a model whereby oncogenic gene fusions frequently alter the protein stability of the resulting fusion products, via exchanging protein degradation signal (degron) between gene sequences. Computational analyses of The Cancer Genome Atlas (TCGA) identify 2,406 cases of degron exchange events and reveal an enrichment of oncogene stabilization due to loss of degrons from fusion. Furthermore, we identify and experimentally validate that some recurrent fusions, such as BCR-ABL, CCDC6-RET and PML-RARA fusions, perturb protein stability by exchanging internal degrons. Likewise, we also validate that EGFR or RAF1 fusions can be stabilized by losing a computationally-predicted C-terminal degron. Thus, complementary to enhanced oncogene transcription via promoter swapping, our model of degron loss illustrates another general mechanism for recurrent fusion proteins in driving tumorigenesis.
Collapse
Affiliation(s)
- Jing Liu
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Collin Tokheim
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Jonathan D Lee
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
- Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Wenjian Gan
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Brian J North
- Department of Biomedical Sciences, Creighton University, Omaha, NE, 68178, USA
| | - X Shirley Liu
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA.
| | - Pier Paolo Pandolfi
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA.
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10124, Italy.
- Renown Institute for Cancer, Nevada System of Higher Education, Reno, NV, 89502, USA.
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
26
|
Hu X, Liu Z, Duan X, Han X, Yuan M, Liu L, Xia X, Li N, Qin J, Wang Y. Blocking MCT4 SUMOylation inhibits the growth of breast cancer cells. Mol Carcinog 2021; 60:702-714. [PMID: 34347919 DOI: 10.1002/mc.23336] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/22/2021] [Accepted: 07/22/2021] [Indexed: 12/20/2022]
Abstract
Monocarboxylate transporter 4 (MCT4) is highly expressed in various types of solid neoplasms including breast cancer (BC); however, the pro-tumor functions underlying its increased expression have not been explained. Here, we examined the roles of posttranslational modifications to MCT4 in BC, particularly SUMOylation. Our findings revealed that SUMOylation of MCT4 inhibited its degradation and stabilized MCT4 protein levels, while ubiquitination facilitated MCT4 degradation. The E3 ubiquitin ligases β-TRCP and FBW7 interacted with MCT4 at the DSG-box and TPETS sequences, respectively, and Lys448 (K448) of MCT4 could be modified by SUMO chains. Our key finding was that K448 was crucial for MCT4 SUMOylation. Moreover, mutations of K448 abolished MCT4 expression, delaying the growth of BC. This study suggested that SUMOylation of K448 increased MCT4 levels, and mutations of K448 in MCT4 could have therapeutic significance in BC.
Collapse
Affiliation(s)
- Xiao Hu
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
| | - Zhanzhao Liu
- Department of Immunology, School of Medicine, Nankai University, Tianjin, China
| | - Xianxian Duan
- Department of Immunology, School of Medicine, Nankai University, Tianjin, China
| | - Xiao Han
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
| | - Mengci Yuan
- Department of Immunology, School of Medicine, Nankai University, Tianjin, China
| | - Lingyan Liu
- Institute of Elemento-Organic Chemistry, College of Chemistry, Nankai University, Tianjin, China
| | - Xiaojun Xia
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou, China
| | - Ning Li
- Institue of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Junfang Qin
- Department of Immunology, School of Medicine, Nankai University, Tianjin, China
| | - Yue Wang
- Department of Immunology, School of Medicine, Nankai University, Tianjin, China.,Department of Immunology, Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Hospital of Stomatology, Nankai University, Tianjin, China
| |
Collapse
|
27
|
Kong S, Ge X, Li X, Liu Z, Zhang R, Yang M, Wang Z, Li Z. SD-36 promotes growth inhibition and induces apoptosis via suppression of Mcl-1 in glioma. J Cell Mol Med 2021; 25:8261-8270. [PMID: 34291563 PMCID: PMC8419162 DOI: 10.1111/jcmm.16754] [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: 12/05/2020] [Revised: 05/02/2021] [Accepted: 06/15/2021] [Indexed: 12/14/2022] Open
Abstract
Glioma is one of the most commonly observed tumours, representing approximately 75% of brain tumours in the adult population. Generally, glioma therapy includes surgical resection followed by radiotherapy and chemotherapy. The transcription factor STAT3 (signal transducer and activator of transcription 3) is a promising target for the treatment of cancer and several other diseases. At nanomolar concentrations, SD‐36 induces rapid cellular degradation of STAT3 but cannot degrade other STAT proteins. The current study demonstrates the therapeutic efficacies of the STAT3 degraders SD‐36 against glioma, as well as understanding the elucidating mechanisms and identifying molecular markers that determine cell sensitivity to STAT3 degraders. Glioma cell lines possessed similar response patterns to SD‐36 but different responses to the STAT3 inhibitor Stattic. SD‐36 potently induced apoptosis in glioma cells along with a reduction in Mcl‐1 levels, which are critical for mediating the induction of apoptosis and enhancing TMZ‐induced apoptosis. Accordingly, SD‐36 sensitizes the antitumour effect of TMZ in patient‐derived xenograft. In addition, the downregulation of Mcl‐1 expression‐mediated antitumour effect of SD‐36 was analysed in cell‐derived xenograft. These observations need to be validated clinically to confirm the efficacy of STAT3 degraders in glioma.
Collapse
Affiliation(s)
- Shiqi Kong
- Department of Neurosurgery, Xingtai People's Hospital, Hebei Province, China
| | - Xinbo Ge
- Department of Neurosurgery, Xingtai People's Hospital, Hebei Province, China
| | - Xin Li
- Department of Neurosurgery, The First People's Hospital of Shenyang, Shenyang, China
| | - Zhenbo Liu
- Department of Neurosurgery, Xingtai People's Hospital, Hebei Province, China
| | - Rui Zhang
- Department of Neurosurgery, Xingtai People's Hospital, Hebei Province, China
| | - Ming Yang
- Department of Neurosurgery, Xingtai People's Hospital, Hebei Province, China
| | - Zhenhai Wang
- Department of Neurosurgery, Xingtai People's Hospital, Hebei Province, China
| | - Zhenzhong Li
- Department of Neurosurgery, Xingtai People's Hospital, Hebei Province, China
| |
Collapse
|
28
|
Michel M, Kaps L, Maderer A, Galle PR, Moehler M. The Role of p53 Dysfunction in Colorectal Cancer and Its Implication for Therapy. Cancers (Basel) 2021; 13:2296. [PMID: 34064974 PMCID: PMC8150459 DOI: 10.3390/cancers13102296] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/28/2021] [Accepted: 05/03/2021] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common and fatal cancers worldwide. The carcinogenesis of CRC is based on a stepwise accumulation of mutations, leading either to an activation of oncogenes or a deactivation of suppressor genes. The loss of genetic stability triggers activation of proto-oncogenes (e.g., KRAS) and inactivation of tumor suppression genes, namely TP53 and APC, which together drive the transition from adenoma to adenocarcinoma. On the one hand, p53 mutations confer resistance to classical chemotherapy but, on the other hand, they open the door for immunotherapy, as p53-mutated tumors are rich in neoantigens. Aberrant function of the TP53 gene product, p53, also affects stromal and non-stromal cells in the tumor microenvironment. Cancer-associated fibroblasts together with other immunosuppressive cells become valuable assets for the tumor by p53-mediated tumor signaling. In this review, we address the manifold implications of p53 mutations in CRC regarding therapy, treatment response and personalized medicine.
Collapse
Affiliation(s)
- Maurice Michel
- I. Department of Medicine, University Medical Center Mainz, 55131 Mainz, Germany; (M.M.); (L.K.); (A.M.); (P.R.G.)
| | - Leonard Kaps
- I. Department of Medicine, University Medical Center Mainz, 55131 Mainz, Germany; (M.M.); (L.K.); (A.M.); (P.R.G.)
- Institute of Translational Immunology and Research Center for Immune Therapy, University Medical Center Mainz, 55131 Mainz, Germany
| | - Annett Maderer
- I. Department of Medicine, University Medical Center Mainz, 55131 Mainz, Germany; (M.M.); (L.K.); (A.M.); (P.R.G.)
| | - Peter R. Galle
- I. Department of Medicine, University Medical Center Mainz, 55131 Mainz, Germany; (M.M.); (L.K.); (A.M.); (P.R.G.)
| | - Markus Moehler
- I. Department of Medicine, University Medical Center Mainz, 55131 Mainz, Germany; (M.M.); (L.K.); (A.M.); (P.R.G.)
| |
Collapse
|
29
|
Pan S, Liang S, Wang X. ADORA1 promotes nasopharyngeal carcinoma cell progression through regulation of PI3K/AKT/GSK-3β/β-catenin signaling. Life Sci 2021; 278:119581. [PMID: 33961854 DOI: 10.1016/j.lfs.2021.119581] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/11/2021] [Accepted: 04/26/2021] [Indexed: 11/18/2022]
Abstract
AIMS For most human cancers, the expression pattern and biological function of ADORA1 (Adenosine A1 Receptor) are largely unknown. This study has been designed to explore the clinical significance and the mechanism of ADORA1 in nasopharyngeal carcinoma (NPC) cells. MATERIALS AND METHODS The level of ADORA1 in NPC and its adjacent tissues was analyzed by IHC, real-time PCR and western blotting. MTT and colony formation assays were used to determine the cell viability post ADORA1 overexpression or knockdown. Wound-healing assay and Transwell assay were used to analyze the effect of ADORA1 on migration and invasion. Moreover, the effect of ADORA1 on tumor growth was also studied in vivo by using xenograft mouse model. The regulation of ADORA1 on PI3K/AKT/GSK-3β/β-catenin pathway was determined by western blotting and TOP-Flash luciferase assay. KEY FINDINGS Primary NPC exhibits overexpression of ADORA1, which is related to the overexpression of its mRNA. Ectopic expression of ADORA1 promotes the proliferation, invasion and migration in NPC cells. The apoptosis, however, is suppressed. ADORA1 silencing was found to exert opposite effects in in vitro studies and produced a significant inhibitory effect on murine xenograft tumor growth in vivo experiments. Besides, ADORA1 also triggers the PI3K/AKT/GSK-3β/β-catenin intracellular oncogenic pathway for signal transduction. Inhibition of this pathway by PI3K inhibitor LY294002 obstructed the impact of ADORA1 on tumor development in cells with ADORA1-overexpression. SIGNIFICANCE ADORA1 has been identified as an important oncoprotein, promoting tumor cell proliferation via PI3K/AKT/GSK-3β/β-catenin signaling pathway in NPC.
Collapse
Affiliation(s)
- Suming Pan
- Department of Radiation Oncology, Yue Bei People's Hospital, Shaoguan, Guangdong, China.
| | - Sixian Liang
- Department of Radiation Oncology, Yue Bei People's Hospital, Shaoguan, Guangdong, China
| | - Xianyan Wang
- Department of Radiation Oncology, Yue Bei People's Hospital, Shaoguan, Guangdong, China
| |
Collapse
|
30
|
Wang H, Guo M, Wei H, Chen Y. Targeting MCL-1 in cancer: current status and perspectives. J Hematol Oncol 2021; 14:67. [PMID: 33883020 PMCID: PMC8061042 DOI: 10.1186/s13045-021-01079-1] [Citation(s) in RCA: 147] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/14/2021] [Indexed: 12/13/2022] Open
Abstract
Myeloid leukemia 1 (MCL-1) is an antiapoptotic protein of the BCL-2 family that prevents apoptosis by binding to the pro-apoptotic BCL-2 proteins. Overexpression of MCL-1 is frequently observed in many tumor types and is closely associated with tumorigenesis, poor prognosis and drug resistance. The central role of MCL-1 in regulating the mitochondrial apoptotic pathway makes it an attractive target for cancer therapy. Significant progress has been made with regard to MCL-1 inhibitors, some of which have entered clinical trials. Here, we discuss the mechanism by which MCL-1 regulates cancer cell apoptosis and review the progress related to MCL-1 small molecule inhibitors and their role in cancer therapy.
Collapse
Affiliation(s)
- Haolan Wang
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Ming Guo
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Hudie Wei
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Yongheng Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
31
|
Zhiani M, Mousavi MA, Rostamizadeh K, Pirizadeh R, Osali A, Mennati A, Motlagh B, Fathi M. Apoptosis induction by siRNA targeting integrin-β1 and regorafenib/DDAB-mPEG-PCL hybrid nanoparticles in regorafenib-resistant colon cancer cells. Am J Cancer Res 2021; 11:1170-1184. [PMID: 33948352 PMCID: PMC8085858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 11/02/2020] [Indexed: 06/12/2023] Open
Abstract
Colorectal cancer (CRC) is regarded as the third most common cancer worldwide. Although Regorafenib as a receptor tyrosine kinase inhibitor (RTKI) disrupts tumor growth and angiogenesis in metastatic CRC (mCRC) patients, drug resistance leads to poor prognosis and survival. Integrin-β1 overexpression has been proposed to be the major player in this regard. Herein, the Regorafenib-resistant human colon cancer cell line (SW-48) was induced, and the Integrin-β1 gene expression, as well as apoptosis, was assessed through the combination of small interfering RNA (siRNA) targeting Integrin-β1 and Regorafenib/Dimethyldioctadecylammonium bromide (DDAB)-methoxy poly (ethylene glycol) (mPEG)-poly-ε-caprolactone (PCL) hybrid nanoparticles (HNPs). In the current study, Regorafenib-resistant SW-48 cell line was generated in which the Regorafenib half-maximal inhibitory concentration (IC50) for non-resistant and resistant cells was 13.5±1.5 µM and 55.1±0.8 µM, respectively. The results of DLS also demonstrated that the size and the charge of the HNPs were equal to 66.56±0.5 nm and +29.5±1.2 mv, respectively. In addition, the Integrin-β1 gene expression was significantly higher in resistant cells than in non-resistant ones (P<0.05). The siRNA/HNP complexes in combination with Regorafenib/HNPs were accordingly identified as the most effective treatment to decrease the Integrin-β1 gene expression and to enhance the apoptosis rate in resistant cells (P<0.001). Overall, the study indicated that combination therapy using siRNA/HNP and Regorafenib/HNPs complex could down-regulate the Integrin-β1 gene expression and consequently trigger apoptosis, and this may potentially induce drug sensitivity.
Collapse
Affiliation(s)
- Mina Zhiani
- Department of Clinical Biochemistry, School of Medicine, Zanjan University of Medical SciencesZanjan, Iran
- Cancer Gene Therapy Research Center, Zanjan University of Medical SciencesZanjan, Iran
- Student Research Committee, School of Medicine, Zanjan University of Medical SciencesZanjan, Iran
| | - Mir Ali Mousavi
- Department of Clinical Biochemistry, School of Medicine, Zanjan University of Medical SciencesZanjan, Iran
- Cancer Gene Therapy Research Center, Zanjan University of Medical SciencesZanjan, Iran
- Student Research Committee, School of Medicine, Zanjan University of Medical SciencesZanjan, Iran
| | - Kobra Rostamizadeh
- Zanjan Pharmaceutical Nanotechnology Research Center, School of Pharmacy, Zanjan University of Medical SciencesZanjan, Iran
- Department of Pharmaceutical Biomaterial, School of Pharmacy, Zanjan University of Medical SciencesZanjan, Iran
| | - Reza Pirizadeh
- Department of Clinical Biochemistry, School of Medicine, Zanjan University of Medical SciencesZanjan, Iran
- Cancer Gene Therapy Research Center, Zanjan University of Medical SciencesZanjan, Iran
- Student Research Committee, School of Medicine, Zanjan University of Medical SciencesZanjan, Iran
| | - Abdolreza Osali
- Department of Immunology, School of Medicine, Zanjan University of Medical SciencesZanjan, Iran
| | - Afsaneh Mennati
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical SciencesZanjan, Iran
| | - Behrouz Motlagh
- Department of Clinical Biochemistry, School of Medicine, Zanjan University of Medical SciencesZanjan, Iran
| | - Mojtaba Fathi
- Department of Clinical Biochemistry, School of Medicine, Zanjan University of Medical SciencesZanjan, Iran
- Cancer Gene Therapy Research Center, Zanjan University of Medical SciencesZanjan, Iran
| |
Collapse
|
32
|
Liu Y, Duan C, Zhang C. E3 Ubiquitin Ligase in Anticancer Drugdsla Resistance: Recent Advances and Future Potential. Front Pharmacol 2021; 12:645864. [PMID: 33935743 PMCID: PMC8082683 DOI: 10.3389/fphar.2021.645864] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 02/24/2021] [Indexed: 12/31/2022] Open
Abstract
Drug therapy is the primary treatment for patients with advanced cancer. The use of anticancer drugs will inevitably lead to drug resistance, which manifests as tumor recurrence. Overcoming chemoresistance may enable cancer patients to have better therapeutic effects. However, the mechanisms underlying drug resistance are poorly understood. E3 ubiquitin ligases (E3s) are a large class of proteins, and there are over 800 putative functional E3s. E3s play a crucial role in substrate recognition and catalyze the final step of ubiquitin transfer to specific substrate proteins. The diversity of the set of substrates contributes to the diverse functions of E3s, indicating that E3s could be desirable drug targets. The E3s MDM2, FBWX7, and SKP2 have been well studied and have shown a relationship with drug resistance. Strategies targeting E3s to combat drug resistance include interfering with their activators, degrading the E3s themselves and influencing the interaction between E3s and their substrates. Research on E3s has led to the discovery of possible therapeutic methods to overcome the challenging clinical situation imposed by drug resistance. In this article, we summarize the role of E3s in cancer drug resistance from the perspective of drug class.
Collapse
Affiliation(s)
- Yuanqi Liu
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, China.,Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis & Treatment, Changsha, China
| | - Chaojun Duan
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, China.,Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis & Treatment, Changsha, China.,Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Chunfang Zhang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, China.,Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis & Treatment, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China
| |
Collapse
|
33
|
Fletcher R, Tong J, Risnik D, Leibowitz BJ, Wang YJ, Concha-Benavente F, DeLiberty JM, Stolz DB, Pai RK, Ferris RL, Schoen RE, Yu J, Zhang L. Non-steroidal anti-inflammatory drugs induce immunogenic cell death in suppressing colorectal tumorigenesis. Oncogene 2021; 40:2035-2050. [PMID: 33603166 PMCID: PMC7981263 DOI: 10.1038/s41388-021-01687-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 01/19/2021] [Accepted: 01/27/2021] [Indexed: 01/30/2023]
Abstract
Use of non-steroidal anti-inflammatory drugs (NSAIDs) is associated with reduced risk of colorectal cancer (CRC). However, the mechanism by which NSAIDs suppress colorectal tumorigenesis remains unclear. We previously showed that NSAIDs selectively kill emerging tumor cells via death receptor (DR) signaling and a synthetic lethal interaction mediated by the proapoptotic Bcl-2 family protein BID. In this study, we found NSAIDs induce endoplasmic reticulum (ER) stress to activate DR signaling and BID in tumor suppression. Importantly, our results unveiled an ER stress- and BID-dependent immunogenic effect of NSAIDs, which may be critical for tumor suppression. NSAID treatment induced hallmarks of immunogenic cell death (ICD) in CRC cells and colonic epithelial cells upon loss of APC tumor suppressor, and elevated tumor-infiltrating lymphocytes (TILs) in the polyps of APCMin/+ mice. ER stress inhibition or BID deletion abrogated the antitumor and immunogenic effects of NSAIDs. Furthermore, increased ER stress and TILs were detected in human advanced adenomas from NSAID-treated patients. Together, our results suggest that NSAIDs induce ER stress- and BID-mediated ICD to restore immunosurveillance and suppress colorectal tumor formation.
Collapse
Affiliation(s)
- Rochelle Fletcher
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jingshan Tong
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Denise Risnik
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Brian J Leibowitz
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yi-Jun Wang
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Fernando Concha-Benavente
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Departments of Otolaryngology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jonathan M DeLiberty
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Donna B Stolz
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Reet K Pai
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Robert L Ferris
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Departments of Otolaryngology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Robert E Schoen
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Departments of Medicine and Epidemiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jian Yu
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lin Zhang
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
34
|
Li S, Guo W, Wu H. The role of post-translational modifications in the regulation of MCL1. Cell Signal 2021; 81:109933. [PMID: 33508399 DOI: 10.1016/j.cellsig.2021.109933] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 01/22/2021] [Accepted: 01/22/2021] [Indexed: 12/25/2022]
Abstract
Apoptosis is an evolutionarily conserved form of programed cell death (PCD) that has a vital effect on early embryonic development, tissue homeostasis and clearance of damaged cells. Dysregulation of apoptosis can lead to many diseases, such as Alzheimer's disease, cancer, AIDS and heart disease. The anti-apoptotic protein MCL1, a member of the BCL2 family, plays important roles in these physiological and pathological processes. Its high expression is closely related to drug resistances in the treatment of tumor. This review summarizes the structure and function of MCL1, the types of post-translational modifications of MCL1 and their effects on the functions of MCL1, as well as the treatment strategies targeting MCL1 in cancer therapy. The research on the fine regulation of MCL1 will be favorable to the provision of a promising future for the design and screening of MCL1 inhibitors.
Collapse
Affiliation(s)
- Shujing Li
- School of Bioengineering & Province Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology, China
| | - Wanping Guo
- School of Bioengineering & Province Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology, China
| | - Huijian Wu
- School of Bioengineering & Province Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology, China.
| |
Collapse
|
35
|
Kim YJ, Kim Y, Kumar A, Kim CW, Toth Z, Cho NH, Lee HR. Kaposi's sarcoma-associated herpesvirus latency-associated nuclear antigen dysregulates expression of MCL-1 by targeting FBW7. PLoS Pathog 2021; 17:e1009179. [PMID: 33471866 PMCID: PMC7816990 DOI: 10.1371/journal.ppat.1009179] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 11/22/2020] [Indexed: 12/11/2022] Open
Abstract
Primary effusion lymphoma (PEL) is an aggressive B cell lymphoma that is etiologically linked to Kaposi’s sarcoma-associated herpesvirus (KSHV). Despite standard multi-chemotherapy treatment, PEL continues to cause high mortality. Thus, new strategies to control PEL are needed urgently. Here, we show that a phosphodegron motif within the KSHV protein, latency-associated nuclear antigen (LANA), specifically interacts with E3 ubiquitin ligase FBW7, thereby competitively inhibiting the binding of the anti-apoptotic protein MCL-1 to FBW7. Consequently, LANA-FBW7 interaction enhances the stability of MCL-1 by preventing its proteasome-mediated degradation, which inhibits caspase-3-mediated apoptosis in PEL cells. Importantly, MCL-1 inhibitors markedly suppress colony formation on soft agar and tumor growth of KSHV+PEL/BCBL-1 in a xenograft mouse model. These results strongly support the conclusion that high levels of MCL-1 expression enable the oncogenesis of PEL cells and thus, MCL-1 could be a potential drug target for KSHV-associated PEL. This work also unravels a mechanism by which an oncogenic virus perturbs a key component of the ubiquitination pathway to induce tumorigenesis. Primary effusion lymphoma (PEL), a highly aggressive B cell lymphoma, is associated with Kaposi’s sarcoma-associated herpesvirus (KSHV). However, the underlying mechanisms that govern the aggressiveness of KSHV-associated PEL are poorly understood. Here, we demonstrate that KSHV LANA interacts with cellular ubiquitin E3 ligase FBW7, sequestering MCL-1 from FBW7, which reduces MCL-1 ubiquitination. As such, LANA potently stabilizes and increases MCL-1 protein, leading to inhibition of caspase-3-mediated apoptosis in PEL cells. Furthermore, MCL-1 inhibitors efficiently blocked PEL progression in mouse xenograft model. These results suggest that LANA acts as a proto-oncogene via deregulating tumor suppressor FBW7, which upregulates anti-apoptotic MCL-1 expression. This study suggests drugs that target MCL-1 may serve as an effective therapy against KSHV+ PEL.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, Viral/genetics
- Antigens, Viral/metabolism
- Apoptosis
- Cell Proliferation
- F-Box-WD Repeat-Containing Protein 7/genetics
- F-Box-WD Repeat-Containing Protein 7/metabolism
- Female
- Herpesvirus 8, Human/physiology
- Humans
- Lymphoma, Primary Effusion/genetics
- Lymphoma, Primary Effusion/metabolism
- Lymphoma, Primary Effusion/pathology
- Lymphoma, Primary Effusion/virology
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Myeloid Cell Leukemia Sequence 1 Protein/genetics
- Myeloid Cell Leukemia Sequence 1 Protein/metabolism
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Phosphorylation
- Sarcoma, Kaposi/genetics
- Sarcoma, Kaposi/metabolism
- Sarcoma, Kaposi/pathology
- Sarcoma, Kaposi/virology
- Tumor Cells, Cultured
- Ubiquitination
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Yeong Jun Kim
- Department of Biotechnology and Bioinformatics, College of Science and Technology, Korea University, Sejong, South Korea
| | - Yuri Kim
- Department of Microbiology and Immunology, Seoul National University college of Medicine, Seoul, South Korea
| | - Abhishek Kumar
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, United States of America
| | - Chan Woo Kim
- Department of Biotechnology and Bioinformatics, College of Science and Technology, Korea University, Sejong, South Korea
| | - Zsolt Toth
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, United States of America
| | - Nam Hyuk Cho
- Department of Microbiology and Immunology, Seoul National University college of Medicine, Seoul, South Korea
- Department of Biomedical Sciences, Seoul National University college of Medicine, Seoul, South Korea
| | - Hye-Ra Lee
- Department of Biotechnology and Bioinformatics, College of Science and Technology, Korea University, Sejong, South Korea
- Department of Lab Medicine, College of Medicine, Korea University, Seoul, South Korea
- * E-mail:
| |
Collapse
|
36
|
Asadi Z, Fathi M, Rismani E, Bigdelou Z, Johari B. Application of decoy oligodeoxynucleotides strategy for inhibition of cell growth and reduction of metastatic properties in nonresistant and erlotinib-resistant SW480 cell line. Cell Biol Int 2021; 45:1001-1014. [PMID: 33377576 DOI: 10.1002/cbin.11543] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/28/2020] [Accepted: 12/25/2020] [Indexed: 12/19/2022]
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a critical regulator for angiogenesis, cell cycle progression, apoptosis, and drug resistance. Resistance toward EGF receptor (EGFR) inhibitors is a significant clinical concern for metastatic colon cancer patients. The present study aimed to evaluate the blocking influences of STAT3 decoy oligodeoxynucleotides (ODNs) on the STAT3 survival signaling pathway in nonresistant and erlotinib-resistant SW480 colon cancer cells. First, STAT3 decoy and scramble ODNs were designed according to STAT3 elements in the promoter region of MYCT1 gene and tested for the interaction of STAT3 protein with designed ODNs via in silico molecular docking study. Then, the efficiency of transfection and subcellular localization of ODNs were assessed using flow cytometry and fluorescence microscopy, respectively. Cell viability, cell cycle, and apoptosis tests, scratch and colony formation assays, and real-time PCR were also used to study the cancerous properties of cells. A considerable decrease in proliferation of colon cancer cells was observed with blockade of STAT3 signaling due to cell cycle arrest and induced apoptosis via downregulation of cyclin D1 and Bcl-XL, respectively. Furthermore, upon transfecting STAT3 decoy ODNs, colony formation potential and migration activity in both SW480 colon cancer cell lines were decreased compared to the control groups. From this study, it could be concluded that STAT3 is critical for cell growth inhibition and metastatic properties reduction of resistant SW480 colon cancer cells; therefore, STAT3 decoy ODNs could be considered as potential therapeutics along with current remedies for treating drug-resistant colon cancer.
Collapse
Affiliation(s)
- Zoleykha Asadi
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mojtaba Fathi
- Department of Clinical Biochemistry, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.,Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Elham Rismani
- Molecular Medicine Department, Pasteur Institute of Iran, Tehran, Iran
| | - Zahra Bigdelou
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Behrooz Johari
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.,Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
37
|
Wang D, Fan C, Tang J. Regorafenib Suppresses Migration of and Induces Cell Cycle Arrest and Apoptosis in MCF-7 Cells. BRAZ J PHARM SCI 2021. [DOI: 10.1590/s2175-979020200004181122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Dawei Wang
- Wuhan Huaxia University of Technology, China
| | - Ceji Fan
- Wuhan Huaxia University of Technology, China
| | - Jun Tang
- Wuhan Huaxia University of Technology, China
| |
Collapse
|
38
|
Tan X, Zhang Z, Liu P, Yao H, Shen L, Tong JS. Inhibition of EZH2 enhances the therapeutic effect of 5-FU via PUMA upregulation in colorectal cancer. Cell Death Dis 2020; 11:1061. [PMID: 33311453 PMCID: PMC7733595 DOI: 10.1038/s41419-020-03266-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 02/07/2023]
Abstract
Although the survival rate of patients with cancer have increased due to the use of current chemotherapeutic agents, adverse effects of cancer therapy remain a concern. The reversal of drug resistance, reduction in harmful side effects and accelerated increase in efficiency have often been addressed in the development of combination therapeutics. Tazemetostat (EPZ-6438), a histone methyltransferase EZH2 selective inhibitor, was approved by the FDA for the treatment of advanced epithelioid sarcoma. However, the effect of tazemetostat on colorectal cancer (CRC) and 5-FU sensitivity remains unclear. In this study, the enhancement of tazemetostat on 5-FU sensitivity was examined in CRC cells. Our findings demonstrated that tazemetostat combined with 5-FU exhibits synergistic antitumor function in vitro and in vivo in CRC cells. In addition, tazemetostat promotes PUMA induction through the ROS/ER stress/CHOP axis. PUMA depletion attenuates the antitumor effect of the combination therapy. Therefore, tazemetostat may be a novel treatment to improve the sensitivity of tumors to 5-FU in CRC therapy. In conclusion, the combination of 5-FU and tazemetostat shows high therapeutic possibility with reduced unfavorable effects.
Collapse
Affiliation(s)
- Xiao Tan
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, People's Republic of China.
| | - Zhongqiang Zhang
- Department of Liver Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, 410011, People's Republic of China
| | - Ping Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, People's Republic of China
| | - Hongliang Yao
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, 410011, People's Republic of China
| | - Liangfang Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, People's Republic of China
| | - Jing-Shan Tong
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
39
|
Bolomsky A, Vogler M, Köse MC, Heckman CA, Ehx G, Ludwig H, Caers J. MCL-1 inhibitors, fast-lane development of a new class of anti-cancer agents. J Hematol Oncol 2020; 13:173. [PMID: 33308268 PMCID: PMC7731749 DOI: 10.1186/s13045-020-01007-9] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 11/22/2020] [Indexed: 12/24/2022] Open
Abstract
Cell death escape is one of the most prominent features of tumor cells and closely linked to the dysregulation of members of the Bcl-2 family of proteins. Among those, the anti-apoptotic family member myeloid cell leukemia-1 (MCL-1) acts as a master regulator of apoptosis in various human malignancies. Irrespective of its unfavorable structure profile, independent research efforts recently led to the generation of highly potent MCL-1 inhibitors that are currently evaluated in clinical trials. This offers new perspectives to target a so far undruggable cancer cell dependency. However, a detailed understanding about the tumor and tissue type specific implications of MCL-1 are a prerequisite for the optimal (i.e., precision medicine guided) use of this novel drug class. In this review, we summarize the major functions of MCL-1 with a special focus on cancer, provide insights into its different roles in solid vs. hematological tumors and give an update about the (pre)clinical development program of state-of-the-art MCL-1 targeting compounds. We aim to raise the awareness about the heterogeneous role of MCL-1 as drug target between, but also within tumor entities and to highlight the importance of rationale treatment decisions on a case by case basis.
Collapse
Affiliation(s)
- Arnold Bolomsky
- Wilhelminen Cancer Research Institute, Wilhelminenspital, Vienna, Austria
| | - Meike Vogler
- Department of Clinical Hematology, GIGA-I3, University of Liège, CHU De Liège, 35, Dom Univ Sart Tilman B, 4000, Liège, Belgium
| | - Murat Cem Köse
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Frankfurt, Germany
| | - Caroline A Heckman
- Institute for Molecular Medicine Finland-FIMM, HiLIFE-Helsinki Institute of Life Science, iCAN Digital Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Grégory Ehx
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Frankfurt, Germany
| | - Heinz Ludwig
- Wilhelminen Cancer Research Institute, Wilhelminenspital, Vienna, Austria
| | - Jo Caers
- Department of Clinical Hematology, GIGA-I3, University of Liège, CHU De Liège, 35, Dom Univ Sart Tilman B, 4000, Liège, Belgium.
| |
Collapse
|
40
|
Qu H, Song X, Song Z, Jiang X, Gao X, Bai L, Wu J, Na L, Yao Z. Berberine reduces temozolomide resistance by inducing autophagy via the ERK1/2 signaling pathway in glioblastoma. Cancer Cell Int 2020; 20:592. [PMID: 33298057 PMCID: PMC7727240 DOI: 10.1186/s12935-020-01693-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/01/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The ability to treat glioblastoma (GBM) using the chemotherapeutic agent temozolomide (TMZ) has been hampered by the development of therapeutic resistance. In this study, we assessed the ability of the isoquinoline alkaloid berberine to alter GBM TMZ resistance using two different TMZ-resistant cell lines to mimic a physiologically relevant GBM experimental system. METHODS By treating these resistant cell lines with berberine followed by TMZ, we were able to assess the chemosensitivity of these cells and their parental strains, based on their performance in the MTT and colony formation assays, as well as on the degree of detectable apoptosis that was detected in the strains. Furthermore, we used Western blotting to assess autophagic responses in these cell lines, and we extended this work into a xenograft mouse model to assess the in vivo efficacy of berberine. RESULTS Through these experiments, our findings indicated that berberine enhanced autophagy and apoptosis in TMZ-resistant cells upon TMZ treatment in a manner that was linked with ERK1/2 signaling. Similarly, when used in vivo, berberine increased GBM sensitivity to TMZ through ERK1/2 signaling pathways. CONCLUSIONS These findings demonstrate that berberine is an effective method of increasing the sensitization of GBM cells to TMZ treatment in a manner that is dependent upon the ERK1/2-mediated induction of autophagy, thus making berberine a potentially viable therapeutic agent for GBM treatment.
Collapse
Affiliation(s)
- Huiling Qu
- Department of Neurology, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110016, Liaoning, China
| | - Xiaofu Song
- Department of Neurology, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110016, Liaoning, China
| | - Zhuyin Song
- Department of Neurology, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110016, Liaoning, China
| | - Xin Jiang
- Department of Neurology, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110016, Liaoning, China
| | - Xin Gao
- Department of Laboratory Medicine, The People's Hospital of Liaoning Province, Shenyang, Liaoning, China
| | - Lijuan Bai
- Department of Neurology, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110016, Liaoning, China
| | - Jiao Wu
- Department of Neurology, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110016, Liaoning, China
| | - Li Na
- Department of Neurology, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110016, Liaoning, China
| | - Zhicheng Yao
- Department of Neurology, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110016, Liaoning, China.
| |
Collapse
|
41
|
Zhao P, Song Z, Wang Y, Cai H, Du X, Li C, Lv J, Liu X, Guo M, Chen Z. The endothelial nitric oxide synthase/cyclic guanosine monophosphate/protein kinase G pathway activates primordial follicles. Aging (Albany NY) 2020; 13:1096-1119. [PMID: 33291075 PMCID: PMC7835019 DOI: 10.18632/aging.202235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 10/20/2020] [Indexed: 01/18/2023]
Abstract
In mammals, the well-organized activation of quiescent primordial follicles is pivotal for female reproductive reserve. In the present study, we examined the mechanisms underlying primordial follicle activation in mice. We found that endothelial nitric oxide synthase (eNOS) and its downstream effectors, cyclic guanosine monophosphate (cGMP) and cGMP-dependent protein kinase G (PKG), were expressed in pre-granulosa cells and promoted primordial follicle activation, oocyte growth and granulosa cell proliferation in neonatal ovaries. Mammalian target of rapamycin (mTOR) colocalized with PKG in pre-granulosa cells and was essential for eNOS/cGMP/PKG pathway-induced primordial follicle activation. The eNOS/cGMP/PKG pathway was found to stabilize mTOR protein. The mRNA levels of F-box and WD repeat domain containing 7 (FBXW7), an E3 ubiquitin ligase, correlated negatively with mTOR protein levels in neonatal ovaries. FBXW7 bound to and destabilized mTOR protein in pre-granulosa cells in a ubiquitin/proteasome-dependent manner. However, agonists of the eNOS/cGMP/PKG pathway reduced FBXW7 mRNA levels. FBXW7 overexpression suppressed primordial follicle activation and prevented the eNOS/cGMP/PKG pathway from activating primordial follicles and stabilizing mTOR protein. These findings demonstrate that the eNOS/cGMP/PKG pathway activates primordial follicles by suppressing FBXW7-induced ubiquitination of mTOR in mice.
Collapse
Affiliation(s)
- Peikun Zhao
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Zidai Song
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Yan Wang
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Han Cai
- Fujian Provincial Key Laboratory of Reproductive Health Research, Medical College of Xiamen University, Xiamen, Fujian, China
| | - Xiaoyan Du
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Changlong Li
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Jianyi Lv
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Xin Liu
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Meng Guo
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Zhenwen Chen
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
42
|
Chen M, Gao YT, Li WX, Wang JC, He YP, Li ZW, Gan GS, Yuan B. FBW7 protects against spinal cord injury by mitigating inflammation-associated neuronal apoptosis in mice. Biochem Biophys Res Commun 2020; 532:576-583. [PMID: 32900488 DOI: 10.1016/j.bbrc.2020.08.071] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 08/20/2020] [Indexed: 01/18/2023]
Abstract
Spinal cord injury (SCI) leads to severe and long-lasting neurological disability. Presently, the lack of effective therapies for SCI is largely attributable to an incomplete understanding of its pathogenesis. F-box and WD repeat domain-containing protein 7 (FBW7, also known as FBXW7) is a type of E3 ubiquitin ligase complex, and plays essential roles in regulating different pathological and physiological processes. In this study, we attempted to explore the effects of FBW7 on SCI progression by the in vivo and in vitro experiments. SCI mice showed significantly reduced expression of FBW7 in spinal cord tissues. Promoting FBW7 expression via intrathecal injection of AAV9/FBW7 effectively improved locomotor function in SCI mice. Neuronal death in spinal cords of SCI mice was obviously ameliorated by FBW7 over-expression, along with greatly decreased expression of cleaved Caspase-3. In addition, microglial activation in spinal cord specimens was detected in SCI mice through increasing Iba-1 expression levels, which was, however, attenuated in SCI mice injected with AAV9/FBW7. Additionally, FBW7 over-expression dramatically restrained inflammatory response in spinal cord tissues of SCI mice, as evidenced by the down-regulated expression of tumor necrosis factor-α (TNF-α) and interleukin 1β (IL-1β) through blocking the activation of nuclear factor-κB (NF-κB) signaling. These anti-inflammatory effects of FBW7 were confirmed in LPS-stimulated mouse microglial BV2 cells. Finally, our in vitro studies showed that conditional medium (CM) collected from LPS-incubated BV2 cells markedly induced apoptosis in the isolated primary spinal neurons; However, this effect was overtly ameliorated by CM from LPS-exposed BV2 cells over-expressing FBW7. Thus, FBW7-regulated inflammation in microglial cells was involved in the amelioration of neuronal apoptosis during SCI development. Collectively, these findings illustrated that FBW7 expression was down-regulated in spinal cords of SCI mice, and promoting its expression could effectively mitigate SCI progression by repressing microglial inflammation and neuronal death.
Collapse
Affiliation(s)
- Min Chen
- Department of Anesthesiology, Shenzhen Samii Medical Center, Shenzhen City, 518118, China
| | - Yu-Ting Gao
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Wei-Xin Li
- Department of Neurosugery, Shenzhen Samii Medical Center, Shenzhen City, 518118, China
| | - Jian-Chun Wang
- Experimental Center, Shenzhen Samii Medical Center, Shenzhen City, 518118, China
| | - Yun-Peng He
- Department of Anesthesiology, Shenzhen Samii Medical Center, Shenzhen City, 518118, China
| | - Zhi-Wen Li
- Shenzhen Samii Medical Center, Shenzhen City, 518118, China
| | - Guo-Sheng Gan
- Department of Anesthesiology, General Hospital of Central Cheater Command of People's Liberation Army of China, Wuhan, 430070, China.
| | - Bo Yuan
- Department of Neurology, Shenzhen Samii Medical Center, Shenzhen City, 518118, China.
| |
Collapse
|
43
|
Yan J, Yang S, Tian H, Zhang Y, Zhao H. Copanlisib promotes growth inhibition and apoptosis by modulating the AKT/FoxO3a/PUMA axis in colorectal cancer. Cell Death Dis 2020; 11:943. [PMID: 33139695 PMCID: PMC7606528 DOI: 10.1038/s41419-020-03154-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 10/13/2020] [Accepted: 10/15/2020] [Indexed: 12/30/2022]
Abstract
Colorectal cancer (CRC) is the type of cancer with the third highest incidence and is associated with high mortality and low 5-year survival rates. We observed that copanlisib, an inhibitor of PI3K (pan-class I phosphoinositide 3-kinase) that preferentially inhibits PI3Kδ and PI3Kα, impedes the growth of CRC cells by inducing apoptosis via PUMA. There was a marked increase in the expression of PUMA independent of p53 after treatment with copanlisib. The response of CRC cells to copanlisib could be predicted by PUMA expression. Copanlisib was found to induce PUMA expression through FoxO3a by directly binding to the PUMA promoter after inhibiting AKT signaling. PUMA deficiency mitigated the apoptosis induced by copanlisib. Caspase activation and mitochondrial dysfunction led to copanlisib resistance, as observed through a clonogenic assay, whereas enhanced expression of PUMA increased the copanlisib-induced susceptibility to apoptosis. Moreover, the antitumor effects of copanlisib were suppressed by a deficiency of PUMA in a xenograft model, and caspase activation and reduced apoptosis were also observed in vivo. Copanlisib-mediated chemosensitization seemed to involve the concurrent induction of PUMA expression via mechanisms that were both dependent and independent of p53. These observations indicate that apoptosis mediated by PUMA is crucial for the anticancer effects of copanlisib and that manipulation of PUMA may aid in enhancing anticancer activities.
Collapse
Affiliation(s)
- Ji Yan
- Department of Medicine Laboratory, The 4th People's Hospital of Shenyang, Shenyang, Liaoning, China
| | - Shida Yang
- Department of Laboratory Medicine, The People's Hospital of China Medical University (The People's Hospital of Liaoning Province), Shenyang, Liaoning, China
| | - Hong Tian
- Oncology Department, The 4th People's Hospital of Shenyang, Shenyang, Liaoning, China
| | - Yang Zhang
- Department of Pathology, The 4th People's Hospital of Shenyang, Shenyang, Liaoning, China
| | - Hongmei Zhao
- Department of Laboratory Medicine, The People's Hospital of China Medical University (The People's Hospital of Liaoning Province), Shenyang, Liaoning, China.
| |
Collapse
|
44
|
Yu Z, Du J, Zhao Y, Gao Y, Li Y, Zhao K, Lu N. A novel kinase inhibitor, LZT-106, downregulates Mcl-1 and sensitizes colorectal cancer cells to BH3 mimetic ABT-199 by targeting CDK9 and GSK-3β signaling. Cancer Lett 2020; 498:31-41. [PMID: 33129955 DOI: 10.1016/j.canlet.2020.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/10/2020] [Accepted: 10/01/2020] [Indexed: 12/27/2022]
Abstract
Colorectal cancer (CRC) is one of the most common malignancies worldwide and is associated with poor prognosis and high mortality. Despite advances in treatment with chemotherapy, CRC remains a major cause of drug resistance-related cancer deaths. One of the main reasons for such resistance is dysregulation of Mcl-1 expression. In this study, we identified LZT-106 as a novel kinase inhibitor that was able to bind to CDK9 with potent inhibitory ability, and indirectly regulate the expression of Mcl-1. However, different regulatory profiles were observed between LZT-106 and the well-studied CDK9 inhibitor flavopiridol with regards to Mcl-1 inhibition. Via Western blotting, real-time PCR and immunoprecipitation, we confirmed that LZT-106 was also able to target GSK-3β signaling and facilitate the degradation of Mcl-1. And LZT-106 was shown to synergize with ABT-199 to induce apoptosis even in the RKO cell line that overexpressed Mcl-1. Finally, LZT-106 significantly inhibited tumor growth in a xenograft mouse model with minimal toxicity. Overall, our findings suggest that LZT-106 is a promising candidate drug for the treatment of patients with CRC.
Collapse
Affiliation(s)
- Zhou Yu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China
| | - Jiaying Du
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China
| | - Yue Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China
| | - Yuan Gao
- Pharmaceutical Animal Experimental Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China
| | - Yongxu Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China
| | - Kai Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China.
| | - Na Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
45
|
Tian T, Guo T, Zhen W, Zou J, Li F. BET degrader inhibits tumor progression and stem-like cell growth via Wnt/β-catenin signaling repression in glioma cells. Cell Death Dis 2020; 11:900. [PMID: 33093476 PMCID: PMC7582157 DOI: 10.1038/s41419-020-03117-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 12/23/2022]
Abstract
Based on their histological appearance, gliomas are a very common primary tumor type of the brain and are classified into grades, Grade I to Grade IV, of the World Health Organization. Treatment failure is due to the cancer stem cells (CSC) phenotype maintenance and self-renewal. BET degraders such as ZBC260 represents a novel class of BET inhibitors that act by inducing BET proteins degradation. This study explores the mode of action and effects of ZBC260 in vivo and in vitro against glioma. By inhibiting cell proliferation and inducting cell cycle arrest, the fact that glioma cell lines show sensitivity to ZBC260. Notably, ZBC260 targeted glioma without side effects in vivo. In addition, the stem cell-like properties of glioma cells were inhibited upon ZBC260 treatment. When the mechanism was examined, our findings indicated that Wnt/β-catenin pathway repression is required for ZBC260-induced stem cell-like properties and tumor growth suppression. In conclusion, the growth of tumors and stem cell-like properties were inhibited by ZBC260 via Wnt/β-catenin repression, which suggests ZBC260 as a potential therapeutic agent for glioma.
Collapse
Affiliation(s)
- Tao Tian
- Department of Oncology, Shandong Zaozhuang Municipal Hospital, Zaozhuang City, Shandong Province, China
| | - Tongqi Guo
- Department of Neurosurgery, The People's Hospital of China Medical University (The People's Hospital of Liaoning Province), Shenyang, Liaoning Province, China
| | - Wei Zhen
- Department of Neurosurgery, The People's Hospital of China Medical University (The People's Hospital of Liaoning Province), Shenyang, Liaoning Province, China
| | - Jianjun Zou
- Department of Neurosurgery, The People's Hospital of China Medical University (The People's Hospital of Liaoning Province), Shenyang, Liaoning Province, China
| | - Fuyong Li
- Department of Neurosurgery, The People's Hospital of China Medical University (The People's Hospital of Liaoning Province), Shenyang, Liaoning Province, China.
| |
Collapse
|
46
|
Li J, Zheng Y, Li X, Dong X, Chen W, Guan Z, Zhang C. UCHL3 promotes proliferation of colorectal cancer cells by regulating SOX12 via AKT/mTOR signaling pathway. Am J Transl Res 2020; 12:6445-6454. [PMID: 33194042 PMCID: PMC7653583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 09/15/2020] [Indexed: 06/11/2023]
Abstract
OBJECTIVE The dysregulation of deubiquitinating enzymes is important in the development of many cancers, including colorectal cancer (CRC). However, the precise function and potential mode of action of the deubiquitinating enzyme UCHL3 in CRC progression are poorly elucidated. METHODS The expression levels of UCHL3 in patient samples were analyzed by western blotting, real-time PCR and immunohistochemistry and its association with overall survival was analyzed using Kaplan-Meier method. Colony formation, CCK-8 and Transwell were used to examine the effects of UCHL3 knockdown or over-expression on CRC cells growth, invasion and migration. The functional effects of UCHL3 and SOX12 on tumor growth were further examined using xenograft tumor mouse models in vivo. RESULTS Here, we found high expression of UCHL3 in CRC tissues which showed an association with the development of tumor and CRC patient survival. Studies conducted in vitro showed that UCHL3 overexpression facilitates proliferation, invasion, migration, and EMT (epithelial-mesenchymal transition) in cells of CRC, and a knockdown of UCHL3 had a reverse effect. Likewise, experiments conducted in vivo also showed enhanced tumor growth due to UCHL3 overexpression. In addition, UCHL3 was found regulates SOX12 expression in CRC cells. PI3K/AKT/mTOR pathway is required for UCHL3-mediated SOX12 expression. Mechanically, UCHL3 regulates SOX12 via AKT/mTOR signaling pathway and facilitated tumor progression. CONCLUSION UCHL3 plays an oncogenic role through the AKT/mTOR/SOX12 axis and can be considered as a potential target for therapy and CRC prognostic biomarker.
Collapse
Affiliation(s)
- Jiangning Li
- Department of Laboratory Medicine, The First People’s Hospital of ShenyangShenyang, Liaoning, P. R. China
| | - Yang Zheng
- Department of Laboratory Medicine, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and InstituteShenyang, Liaoning, P. R. China
| | - Xiaofeng Li
- Institute of Transfusion Medicine, Liaoning Blood CenterShenyang, Liaoning, P. R. China
| | - Xue Dong
- Microbiological Laboratory Center, Shenyang Center for Disease Control and PreventionShenyang, Liaoning, P. R. China
| | - Weiyan Chen
- Department of Pathology, Affiliated Hospital of Liaoning University of Traditional Chinese MedicineShenyang, Liaoning, P. R. China
| | - Zhongying Guan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Liaoning University of Traditional Chinese MedicineShenyang, Liaoning, P. R. China
| | - Chong Zhang
- Department of Plastic Surgery, Beijing Weiyan Medical Cosmetology ClinicBeijing, P. R. China
| |
Collapse
|
47
|
Han S, Zhen W, Guo T, Zou J, Li F. SETDB1 promotes glioblastoma growth via CSF-1-dependent macrophage recruitment by activating the AKT/mTOR signaling pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:218. [PMID: 33059737 PMCID: PMC7560339 DOI: 10.1186/s13046-020-01730-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 10/08/2020] [Indexed: 12/11/2022]
Abstract
Background Glioblastoma is a common disease of the central nervous system (CNS), with high morbidity and mortality. In the infiltrate in the tumor microenvironment, tumor-associated macrophages (TAMs) are abundant, which are important factors in glioblastoma progression. However, the exact details of TAMs in glioblastoma progression have yet to be determined. Methods The clinical relevance of SET domain bifurcated 1 (SETDB1) was analyzed by immunohistochemistry, real-time PCR and Western blotting of glioblastoma tissues. SETDB1-induced cell proliferation, migration and invasion were investigated by CCK-8 assay, colony formation assay, wound healing and Transwell assay. The relationship between SETDB1 and colony stimulating factor 1 (CSF-1), as well as TAMs recruitment was examined by Western blotting, real-time PCR and syngeneic mouse model. Results Our findings showed that SETDB1 upregulated in glioblastoma and relative to poor progression. Gain and loss of function approaches showed the SETDB1 overexpression promotes cell proliferation, migration and invasion in glioblastoma cells. However, knockdown SETDB1 exerted opposite effects in vitro. Moreover, SETDB1 promotes AKT/mTOR-dependent CSF-1 induction and secretion, which leads to macrophage recruitment in the tumor, resulted in tumor growth. Conclusion Our research clarified that SETDB1 regulates of tumor microenvironment and hence presents a potential therapeutic target for treating glioblastoma.
Collapse
Affiliation(s)
- Shuai Han
- Department of Neurosurgery, the First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Wei Zhen
- Department of Neurosurgery, The People's Hospital of China Medical University (The People's Hospital of Liaoning Province), No.33, Wenyi Road, Shenhe District, Shenyang, 110016, Liaoning Province, PR China
| | - Tongqi Guo
- Department of Neurosurgery, The People's Hospital of China Medical University (The People's Hospital of Liaoning Province), No.33, Wenyi Road, Shenhe District, Shenyang, 110016, Liaoning Province, PR China
| | - Jianjun Zou
- Department of Neurosurgery, The People's Hospital of China Medical University (The People's Hospital of Liaoning Province), No.33, Wenyi Road, Shenhe District, Shenyang, 110016, Liaoning Province, PR China
| | - Fuyong Li
- Department of Neurosurgery, The People's Hospital of China Medical University (The People's Hospital of Liaoning Province), No.33, Wenyi Road, Shenhe District, Shenyang, 110016, Liaoning Province, PR China.
| |
Collapse
|
48
|
Wang Z, Zhao Y, Xu H, Liang F, Zou Q, Wang C, Jiang J, Lin F. CtBP1 promotes tumour-associated macrophage infiltration and progression in non-small-cell lung cancer. J Cell Mol Med 2020; 24:11445-11456. [PMID: 32910558 PMCID: PMC7576280 DOI: 10.1111/jcmm.15751] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/17/2020] [Accepted: 07/31/2020] [Indexed: 12/14/2022] Open
Abstract
The progression of lung cancer is majorly facilitated by TAMs (tumour-associated macrophages). However, how the TAMs infiltrate the NSCLC microenvironment and the associated biochemical are not fully elaborated. Research has revealed that changes in CtBP1 modulates innate immunity. Here, we investigated if CtBP1 facilitates infiltration of TAM and the subsequent progression of NSCLC. Immunohistochemical analysis was carried out in 96 NSCLC patients to estimate the clinicopathological importance of CtBP1 in the disease. CtBP1 overexpression and knockdown were carried out to assess the activity of CtBP1 in NSCLC cells. Elevated expression of CtBP1 correlated positively with TAMs infiltration into NSCLC tissues, induced EMT (epithelial-mesenchymal transition) in NSCLC cells and modulated the activated NF-κB signalling pathway leading to increase in CCL2 secretion from NSCLC cells, thus promoting TAM recruitment and polarization. TAM induction and polarization reduced significantly on exhausting p65 in NSCLC cells with CtBP1. Moreover, infiltration of TMAs was reduced remarkably on antagonist-mediated blocking of CCR2 and impeded the progression of NSCLC in a mouse model. These findings thus show a novel insight into the process of CtBP1-regulated TAM infiltration in NSCLC.
Collapse
Affiliation(s)
- Zhenxing Wang
- Department of Thoracic SurgeryChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Yan Zhao
- Physical Examination CenterThe Second Hospital of Jilin UniversityChangchunChina
| | - Hongyan Xu
- Department of Medical OncologyThe Tumor Hospital of Jilin CityJilinChina
| | - Feihai Liang
- Department of Cardiovascular thoracic SurgeryThe Second Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Qingxu Zou
- Department of Thoracic SurgeryChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Chen Wang
- Department of Thoracic SurgeryChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Jingyuan Jiang
- Department of Thoracic SurgeryChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Fengwu Lin
- Department of Thoracic SurgeryChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| |
Collapse
|
49
|
Li Y, Zhang H, Li Q, Zou P, Huang X, Wu C, Tan L. CDK12/13 inhibition induces immunogenic cell death and enhances anti-PD-1 anticancer activity in breast cancer. Cancer Lett 2020; 495:12-21. [PMID: 32941949 DOI: 10.1016/j.canlet.2020.09.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 01/12/2023]
Abstract
Immunogenic cell death (ICD) improves the T cell response against different tumors, indicating that ICD can enhance the antitumor immunity elicited by the anti-checkpoint antibody anti-programmed death 1 (anti-PD-1). In the present study, we reported a synergistic and durable immune-mediated antitumor response elicited by the combined treatment of SR-4835, a CDK12/13 specific inhibitor, with PD-1 blockade in a syngeneic mouse model. The developed combination therapy elicited antitumor activity in immunocompetent mouse tumor models. Furthermore, the SR-4835-treated tumor cells exhibited characteristics of ICD, including the release of high mobility group box 1 (HMGB1) and ATP and calreticulin (CRT) translocation. This activity led to a significant T-cell-dependent tumor suppression. The enhanced dendritic cell (DC) and infiltration of T cells activation in the tumors treated with both SR-4835 and anti-PD-1 indicate that this combination treatment promotes an improved immune response. Therefore, the results of the present study demonstrate the potential of CDK12/13 inhibition combined with checkpoint inhibition in breast cancer treatment.
Collapse
Affiliation(s)
- Yi Li
- Department of Breast Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Hui Zhang
- Department of Ultrasound, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Qin Li
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Pingjin Zou
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Xingxiang Huang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Chihua Wu
- Department of Breast Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Li Tan
- Department of Ultrasound, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
50
|
Song X, Shen L, Tong J, Kuang C, Zeng S, Schoen RE, Yu J, Pei H, Zhang L. Mcl-1 inhibition overcomes intrinsic and acquired regorafenib resistance in colorectal cancer. Theranostics 2020; 10:8098-8110. [PMID: 32724460 PMCID: PMC7381732 DOI: 10.7150/thno.45363] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 06/19/2020] [Indexed: 12/11/2022] Open
Abstract
Intrinsic and acquired resistance to targeted therapies is a significant clinical problem in cancer. We previously showed that resistance to regorafenib, a multi-kinase inhibitor for treating colorectal cancer (CRC) patients, can be caused by mutations in the tumor suppressor FBW7, which block degradation of the pro-survival Bcl-2 family protein Mcl-1. We tested if Mcl-1 inhibition can be used to develop a precision combination therapy for overcoming regorafenib resistance. METHODS Small-molecule Mcl-1 inhibitors were tested on CRC cells with knock-in (KI) of a non-degradable Mcl-1. Effects of Mcl-1 inhibitors on regorafenib sensitivity were determined in FBW7-mutant and -wild-type (WT) CRC cells and tumors, and in those with acquired regorafenib resistance due to enriched FBW7 mutations. Furthermore, translational potential was explored by establishing and analyzing FBW7-mutant and -WT patient-derived organoid (PDO) and xenograft (PDX) tumor models. RESULTS We found that highly potent and specific Mcl-1 inhibitors such as S63845 overcame regorafenib resistance by restoring apoptosis in multiple regorafenib-resistant CRC models. Mcl-1 inhibition re-sensitized CRC tumors with intrinsic and acquired regorafenib resistance in vitro and in vivo, including those with FBW7 mutations. Importantly, Mcl-1 inhibition also sensitized FBW7-mutant PDO and PDX models to regorafenib. In contrast, Mcl-1 inhibition had no effect in FBW7-WT CRCs. CONCLUSIONS Our results demonstrate that Mcl-1 inhibitors can overcome intrinsic and acquired regorafenib resistance in CRCs by restoring apoptotic response. FBW7 mutations might be a potential biomarker predicting for response to the regorafenib/Mcl-1 inhibitor combination.
Collapse
Affiliation(s)
- Xiangping Song
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Gastrointestinal Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Lin Shen
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Jingshan Tong
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Robert E. Schoen
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213. USA
| | - Jian Yu
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213. USA
| | - Haiping Pei
- Department of Gastrointestinal Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Lin Zhang
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|