1
|
Dong S, Alahari SK. Small molecule Mcl-1 inhibitor for triple negative breast cancer therapy. Front Cell Dev Biol 2024; 12:1408107. [PMID: 39372954 PMCID: PMC11449857 DOI: 10.3389/fcell.2024.1408107] [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: 03/27/2024] [Accepted: 08/23/2024] [Indexed: 10/08/2024] Open
Abstract
Apoptosis is an evolutionarily conserved cell death pathway that plays a crucial role in maintaining tissue homeostasis, orchestrating organismal development, and eliminating damaged cells. Dysregulation of apoptosis can contribute to the pathogenesis of malignant tumors and neurodegenerative diseases. Anticancer drugs typically possess the capacity to induce apoptosis in tumor cells. The Bcl-2 protein family, consisting of 27 members in humans, serves as the key regulator of mitochondrial function. This family can be divided into two functional groups: anti-apoptotic proteins (e.g., Bcl-2, Bcl-xl, Mcl-1) and pro-apoptotic proteins (e.g., Bad, Bax). Mcl-1 exerts its function by binding pro-apoptotic Bcl-2 proteins thereby preventing apoptosis induction. Overexpression of Mcl-1 not only correlates closely with tumorigenesis but also associates significantly with resistance towards targeted therapy and conventional chemotherapy. Effective induction of apoptosis can be achieved through inhibition or interference with Mcl-1. Thus, this mini review discusses existing Mcl-1 inhibitors.
Collapse
Affiliation(s)
| | - Suresh K. Alahari
- Department of Biochemistry and Molecular Biology, LSHSC School of Medicine, New Orleans, LA, United States
- Stanley S. Scott Cancer Center, LSUHSC School of Medicine, New Orleans, LA, United States
| |
Collapse
|
2
|
Di T, Luo QY, Song JT, Yan XL, Zhang L, Pan WT, Guo Y, Lu FT, Sun YT, Xia ZF, Yang LQ, Qiu MZ, Yang DJ, Sun J. APG-1252 combined with Cabozantinib inhibits hepatocellular carcinoma by suppressing MEK/ERK and CREB/Bcl-xl pathways. Int Immunopharmacol 2024; 139:112615. [PMID: 39032475 DOI: 10.1016/j.intimp.2024.112615] [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: 05/07/2024] [Revised: 06/30/2024] [Accepted: 06/30/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND AND PURPOSE Liver cancer is the fourth leading cause of cancer-related death worldwide, with hepatocellular carcinoma (HCC) being the most common type of primary liver cancer. APG-1252 is a small molecule inhibitor targeting Bcl-2 and Bcl-xl. However, its anti-tumor effects in HCC, alone or in combination with Cabozantinib, have not been extensively studied. EXPERIMENTAL Approach: TCGA database analysis was used to analysis the gene expression levels of Bcl-2 and Bcl-xl in HCC tissues. Western blot was employed to detect the protein expression levels. And the inhibitory effects of APG-1252 and Cabozantinib on the proliferation of HCC cell lines was detected by CCK-8. The effect on the migration and invasion of HCC cells was verified by transwell assay. Huh7 xenograft model in nude mice was used to investigate the combination antitumor effect in vivo. KEY RESULTS Our study demonstrated that APG-1252 monotherapy inhibited the proliferation and migration ability of HCC cells, and induced HCC cells apoptosis. The combination of APG-1252 and Cabozantinib showed significant synergistic antitumor effects. Furthermore, the in vivo experiment demonstrated that the combination therapy exerted a synergistic effect in delaying tumor growth, notably downregulating MEK/ERK phosphorylation levels. In terms of mechanism, Cabozantinib treatment caused an increase in the phosphorylation levels of CREB and Bcl-xl proteins, while the combination with APG-1252 mitigated this effect, thereby enhanced the antitumor effect of Cabozantinib. CONCLUSION AND IMPLICATIONS Our findings suggest that APG-1252 in combination with Cabozantinib offers a more effective treatment strategy for HCC patients, warranting further clinical investigation.
Collapse
Affiliation(s)
- Tian Di
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China; Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Qiu-Yun Luo
- Department of Clinical Research, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Jiang-Tao Song
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xiang-Lei Yan
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden; Center of Molecular Medicine, Stockholm, Sweden
| | - Lin Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China; Department of Clinical Laboratory, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Wen-Tao Pan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yu Guo
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Fei-Teng Lu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yu-Ting Sun
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China; Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Zeng-Fei Xia
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Li-Qiong Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China; Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Miao-Zhen Qiu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China; Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China; Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| | - Da-Jun Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China; Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| | - Jian Sun
- Department of Clinical Research, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China.
| |
Collapse
|
3
|
Kitai H, Choi PH, Yang YC, Boyer JA, Whaley A, Pancholi P, Thant C, Reiter J, Chen K, Markov V, Taniguchi H, Yamaguchi R, Ebi H, Evans J, Jiang J, Lee B, Wildes D, de Stanchina E, Smith JAM, Singh M, Rosen N. Combined inhibition of KRAS G12C and mTORC1 kinase is synergistic in non-small cell lung cancer. Nat Commun 2024; 15:6076. [PMID: 39025835 PMCID: PMC11258147 DOI: 10.1038/s41467-024-50063-z] [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: 12/21/2023] [Accepted: 06/28/2024] [Indexed: 07/20/2024] Open
Abstract
Current KRASG12C (OFF) inhibitors that target inactive GDP-bound KRASG12C cause responses in less than half of patients and these responses are not durable. A class of RASG12C (ON) inhibitors that targets active GTP-bound KRASG12C blocks ERK signaling more potently than the inactive-state inhibitors. Sensitivity to either class of agents is strongly correlated with inhibition of mTORC1 activity. We have previously shown that PI3K/mTOR and ERK-signaling pathways converge on key cellular processes and that inhibition of both pathways is required for inhibition of these processes and for significant antitumor activity. We find here that the combination of a KRASG12C inhibitor with a selective mTORC1 kinase inhibitor causes synergistic inhibition of Cyclin D1 expression and cap-dependent translation. Moreover, BIM upregulation by KRASG12C inhibition and inhibition of MCL-1 expression by the mTORC1 inhibitor are both required to induce significant cell death. In vivo, this combination causes deep, durable tumor regressions and is well tolerated. This study suggests that the ERK and PI3K/mTOR pathways each mitigate the effects of inhibition of the other and that combinatorial inhibition is a potential strategy for treating KRASG12C-dependent lung cancer.
Collapse
Affiliation(s)
- Hidenori Kitai
- Program in Molecular Pharmacology and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Philip H Choi
- Program in Molecular Pharmacology and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yu C Yang
- Department of Biology, Revolution Medicines Inc., Redwood City, CA, USA
| | - Jacob A Boyer
- Program in Molecular Pharmacology and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Adele Whaley
- Program in Molecular Pharmacology and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Priya Pancholi
- Program in Molecular Pharmacology and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Claire Thant
- Program in Molecular Pharmacology and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jason Reiter
- Program in Molecular Pharmacology and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kevin Chen
- Antitumor Assessment Core, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vladimir Markov
- Antitumor Assessment Core, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hirokazu Taniguchi
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rui Yamaguchi
- Division of Cancer Systems Biology, Aichi Cancer Center Research Institute, Nagoya, Aichi, 464-8681, Japan
| | - Hiromichi Ebi
- Division of Molecular Therapeutics, Aichi Cancer Center Research Institute, Nagoya, Aichi, 464-8681, Japan
| | - James Evans
- Department of Biology, Revolution Medicines Inc., Redwood City, CA, USA
| | - Jingjing Jiang
- Department of Biology, Revolution Medicines Inc., Redwood City, CA, USA
| | - Bianca Lee
- Department of Biology, Revolution Medicines Inc., Redwood City, CA, USA
| | - David Wildes
- Department of Biology, Revolution Medicines Inc., Redwood City, CA, USA
| | - Elisa de Stanchina
- Antitumor Assessment Core, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Mallika Singh
- Department of Biology, Revolution Medicines Inc., Redwood City, CA, USA.
| | - Neal Rosen
- Program in Molecular Pharmacology and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
4
|
Wang X, Cornish AE, Do MH, Brunner JS, Hsu TW, Xu Z, Malik I, Edwards C, Capistrano KJ, Zhang X, Ginsberg MH, Finley LWS, Lim MS, Horwitz SM, Li MO. Onco-Circuit Addiction and Onco-Nutrient mTORC1 Signaling Vulnerability in a Model of Aggressive T Cell Malignancy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.03.587917. [PMID: 38617314 PMCID: PMC11014592 DOI: 10.1101/2024.04.03.587917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
How genetic lesions drive cell transformation and whether they can be circumvented without compromising function of non-transformed cells are enduring questions in oncology. Here we show that in mature T cells-in which physiologic clonal proliferation is a cardinal feature- constitutive MYC transcription and Tsc1 loss in mice modeled aggressive human malignancy by reinforcing each other's oncogenic programs. This cooperation was supported by MYC-induced large neutral amino acid transporter chaperone SLC3A2 and dietary leucine, which in synergy with Tsc1 deletion overstimulated mTORC1 to promote mitochondrial fitness and MYC protein overexpression in a positive feedback circuit. A low leucine diet was therapeutic even in late-stage disease but did not hinder T cell immunity to infectious challenge, nor impede T cell transformation driven by constitutive nutrient mTORC1 signaling via Depdc5 loss. Thus, mTORC1 signaling hypersensitivity to leucine as an onco-nutrient enables an onco-circuit, decoupling pathologic from physiologic utilization of nutrient acquisition pathways.
Collapse
|
5
|
Bei Y, Bramé L, Kirchner M, Fritsche-Guenther R, Kunz S, Bhattacharya A, Rusu MC, Gürgen D, Dubios FPB, Köppke JKC, Proba J, Wittstruck N, Sidorova OA, Chamorro González R, Dorado Garcia H, Brückner L, Xu R, Giurgiu M, Rodriguez-Fos E, Yu Q, Spanjaard B, Koche RP, Schmitt CA, Schulte JH, Eggert A, Haase K, Kirwan J, Hagemann AIH, Mertins P, Dörr JR, Henssen AG. Passenger Gene Coamplifications Create Collateral Therapeutic Vulnerabilities in Cancer. Cancer Discov 2024; 14:492-507. [PMID: 38197697 PMCID: PMC10911929 DOI: 10.1158/2159-8290.cd-23-1189] [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: 10/10/2023] [Revised: 11/30/2023] [Accepted: 12/19/2023] [Indexed: 01/11/2024]
Abstract
DNA amplifications in cancer do not only harbor oncogenes. We sought to determine whether passenger coamplifications could create collateral therapeutic vulnerabilities. Through an analysis of >3,000 cancer genomes followed by the interrogation of CRISPR-Cas9 loss-of-function screens across >700 cancer cell lines, we determined that passenger coamplifications are accompanied by distinct dependency profiles. In a proof-of-principle study, we demonstrate that the coamplification of the bona fide passenger gene DEAD-Box Helicase 1 (DDX1) creates an increased dependency on the mTOR pathway. Interaction proteomics identified tricarboxylic acid (TCA) cycle components as previously unrecognized DDX1 interaction partners. Live-cell metabolomics highlighted that this interaction could impair TCA activity, which in turn resulted in enhanced mTORC1 activity. Consequently, genetic and pharmacologic disruption of mTORC1 resulted in pronounced cell death in vitro and in vivo. Thus, structurally linked coamplification of a passenger gene and an oncogene can result in collateral vulnerabilities. SIGNIFICANCE We demonstrate that coamplification of passenger genes, which were largely neglected in cancer biology in the past, can create distinct cancer dependencies. Because passenger coamplifications are frequent in cancer, this principle has the potential to expand target discovery in oncology. This article is featured in Selected Articles from This Issue, p. 384.
Collapse
Affiliation(s)
- Yi Bei
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Luca Bramé
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marieluise Kirchner
- Core Unit Proteomics, Berlin Institute of Health at Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Raphaela Fritsche-Guenther
- Core Unit Metabolomics, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Severine Kunz
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Technology Platform Electron Microscopy, Berlin, Germany
| | - Animesh Bhattacharya
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Mara-Camelia Rusu
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Technology Platform Electron Microscopy, Berlin, Germany
| | - Dennis Gürgen
- Experimental Pharmacology and Oncology (EPO), Berlin, Germany
| | - Frank P B Dubios
- Institute of pathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Julia K C Köppke
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jutta Proba
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Nadine Wittstruck
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Olga Alexandra Sidorova
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Rocío Chamorro González
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Heathcliff Dorado Garcia
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Lotte Brückner
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Technology Platform Electron Microscopy, Berlin, Germany
| | - Robin Xu
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Mădălina Giurgiu
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Elias Rodriguez-Fos
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Qinghao Yu
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Bastiaan Spanjaard
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Richard P Koche
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Clemens A Schmitt
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Johannes H Schulte
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Angelika Eggert
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kerstin Haase
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jennifer Kirwan
- Core Unit Metabolomics, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Anja I H Hagemann
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Philipp Mertins
- Core Unit Proteomics, Berlin Institute of Health at Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Jan R Dörr
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Berlin Institute of Health, Berlin, Germany
- Experimental and Clinical Research Center (ECRC) of the MDC and Charité Berlin, Berlin, Germany
| | - Anton G Henssen
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Technology Platform Electron Microscopy, Berlin, Germany
- Experimental and Clinical Research Center (ECRC) of the MDC and Charité Berlin, Berlin, Germany
| |
Collapse
|
6
|
Malvi P, Chava S, Cai G, Hu K, Zhu LJ, Edwards YJK, Green MR, Gupta R, Wajapeyee N. HOXC6 drives a therapeutically targetable pancreatic cancer growth and metastasis pathway by regulating MSK1 and PPP2R2B. Cell Rep Med 2023; 4:101285. [PMID: 37951219 PMCID: PMC10694669 DOI: 10.1016/j.xcrm.2023.101285] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 10/13/2023] [Accepted: 10/17/2023] [Indexed: 11/13/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers, which lacks effective therapies. Here, we demonstrate that the transcription factor, homeobox C6 (HOXC6), is overexpressed in most PDACs, and its inhibition blocks PDAC tumor growth and metastasis. HOXC6 transcriptionally activates tumor-promoting kinase MSK1 and suppresses tumor-inhibitory protein PPP2R2B in PDAC. HOXC6-induced PPP2R2B suppression causes mammalian target of rapamycin (mTOR) pathway activation, which facilitates PDAC growth. Also, MSK1 upregulation by HOXC6 is necessary for PDAC growth because of its ability to suppress apoptosis via its substrate DDX17. Combinatorial pharmacological inhibition of MSK1 and mTOR potently suppressed PDAC tumor growth and metastasis in PDAC mouse models. PDAC cells with acquired resistance to MSK1/mTOR-inhibitors displayed activated insulin-like growth factor 1 receptor (IGF1R) signaling and were successfully eradicated by IGF1R inhibitor. Furthermore, MEK inhibitor trametinib enhanced the efficacy of dual MSK1 and mTOR inhibition. Collectively, these results identify therapeutic vulnerabilities of PDAC and an approach to overcome acquired drug resistance to prolong therapeutic benefit.
Collapse
Affiliation(s)
- Parmanand Malvi
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Suresh Chava
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Guoping Cai
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Kai Hu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Lihua Julie Zhu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA; Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Yvonne J K Edwards
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Michael R Green
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Romi Gupta
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35233, USA; O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Narendra Wajapeyee
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35233, USA; O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
7
|
Yu J, Jin Y, Xu C, Fang C, Zhang Z, Chen L, Xu G. Downregulation of miR-125a-5p Promotes Endothelial Progenitor Cell Migration and Angiogenesis and Alleviates Deep Vein Thrombosis in Mice Via Upregulation of MCL-1. Mol Biotechnol 2023; 65:1664-1678. [PMID: 36738360 DOI: 10.1007/s12033-023-00676-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023]
Abstract
Endothelial progenitor cells (EPCs) contribute to recanalization of deep vein thrombosis (DVT). MicroRNAs (miRNAs) play regulatory roles in functions of EPCs, which is becoming a promising therapeutic choice for thrombus resolution. The main purpose of this study was to explore the effect of miR-125a-5p on EPC functions in deep vein thrombosis (DVT). EPCs were isolated from the peripheral blood of patients with DVT. In DVT mouse models, DVT was induced by stenosis of the inferior vena cava (IVC). The levels of miR-125a-5p and myeloid cell leukemia sequence 1 (MCL-1) in EPCs and thrombi of DVT mice were detected by RT-qPCR. EPC migration, angiogenesis, and apoptosis were estimated by Transwell assay, tube formation assay, and flow cytometry analysis. Luciferase reporter assay was utilized for detecting the binding of miR-125a-5p and MCL-1. The phosphorylation of PI3K and AKT was estimated by western blot. DVT formation in vivo was observed through hematoxylin-eosin (H&E) staining. The expression of thrombus resolution marker, CD34 molecule (CD34), in the thrombi was measured by immunofluorescence staining. MiR-125a-5p upregulation repressed EPC migration and angiogenesis and facilitated apoptosis. MiR-125a-5p downregulation showed the opposite effect. MCL-1 was targeted and negatively regulated by miR-125a-5p. Additionally, miR-125a-5p inhibited the PI3K/AKT pathway in EPCs. Inhibition of MCL-1 or PI3K/AKT pathway reversed the effect of miR-125a-5p knockdown on EPC functions. The in vivo experiments revealed that miR-125a-5p downregulation repressed thrombus formation and promoted the homing capability of EPCs to the thrombosis site, thereby alleviating DVT mice. Downregulation of miR-125a-5p promotes EPC migration and angiogenesis by upregulating MCL-1, thereby enhancing EPC homing to thrombi and facilitating thrombus resolution.
Collapse
Affiliation(s)
- Jingfan Yu
- Department of Vascular Surgery and Intervention, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, 188 Guangji Road, Gusu District, Suzhou, 215000, Jiangsu, China
| | - Yiqi Jin
- Department of Vascular Surgery and Intervention, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, 188 Guangji Road, Gusu District, Suzhou, 215000, Jiangsu, China
| | - Chen Xu
- Department of Vascular Surgery and Intervention, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, 188 Guangji Road, Gusu District, Suzhou, 215000, Jiangsu, China
| | - Changwen Fang
- Department of Vascular Surgery and Intervention, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, 188 Guangji Road, Gusu District, Suzhou, 215000, Jiangsu, China
| | - Zhixuan Zhang
- Department of Vascular Surgery and Intervention, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, 188 Guangji Road, Gusu District, Suzhou, 215000, Jiangsu, China
| | - Lei Chen
- Department of Vascular Surgery and Intervention, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, 188 Guangji Road, Gusu District, Suzhou, 215000, Jiangsu, China.
| | - Guoxiong Xu
- Department of Vascular Surgery and Intervention, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, 188 Guangji Road, Gusu District, Suzhou, 215000, Jiangsu, China.
| |
Collapse
|
8
|
Farmaki E, Nath A, Emond R, Karimi KL, Grolmusz VK, Cosgrove PA, Bild AH. ONC201/TIC10 enhances durability of mTOR inhibitor everolimus in metastatic ER+ breast cancer. eLife 2023; 12:e85898. [PMID: 37772709 PMCID: PMC10541180 DOI: 10.7554/elife.85898] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 08/17/2023] [Indexed: 09/30/2023] Open
Abstract
The mTOR inhibitor, everolimus, is an important clinical management component of metastatic ER+ breast cancer (BC). However, most patients develop resistance and progress on therapy, highlighting the need to discover strategies that increase mTOR inhibitor effectiveness. We developed ER+ BC cell lines, sensitive or resistant to everolimus, and discovered that combination treatment of ONC201/TIC10 with everolimus inhibited cell growth in 2D/3D in vitro studies. We confirmed increased therapeutic response in primary patient cells progressing on everolimus, supporting clinical relevance. We show that ONC201/TIC10 mechanism in metastatic ER+ BC cells involves oxidative phosphorylation inhibition and stress response activation. Transcriptomic analysis in everolimus resistant breast patient tumors and mitochondrial functional assays in resistant cell lines demonstrated increased mitochondrial respiration dependency, contributing to ONC201/TIC10 sensitivity. We propose that ONC201/TIC10 and modulation of mitochondrial function may provide an effective add-on therapy strategy for patients with metastatic ER+ BCs resistant to mTOR inhibitors.
Collapse
Affiliation(s)
- Elena Farmaki
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical CenterDuarteUnited States
| | - Aritro Nath
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical CenterDuarteUnited States
| | - Rena Emond
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical CenterDuarteUnited States
| | - Kimya L Karimi
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical CenterDuarteUnited States
| | - Vince K Grolmusz
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical CenterDuarteUnited States
| | - Patrick A Cosgrove
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical CenterDuarteUnited States
| | - Andrea H Bild
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical CenterDuarteUnited States
| |
Collapse
|
9
|
Ghosh A, Chakraborty P, Biswas D. Fine tuning of the transcription juggernaut: A sweet and sour saga of acetylation and ubiquitination. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2023; 1866:194944. [PMID: 37236503 DOI: 10.1016/j.bbagrm.2023.194944] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/26/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023]
Abstract
Among post-translational modifications of proteins, acetylation, phosphorylation, and ubiquitination are most extensively studied over the last several decades. Owing to their different target residues for modifications, cross-talk between phosphorylation with that of acetylation and ubiquitination is relatively less pronounced. However, since canonical acetylation and ubiquitination happen only on the lysine residues, an overlap of the same lysine residue being targeted for both acetylation and ubiquitination happens quite frequently and thus plays key roles in overall functional regulation predominantly through modulation of protein stability. In this review, we discuss the cross-talk of acetylation and ubiquitination in the regulation of protein stability for the functional regulation of cellular processes with an emphasis on transcriptional regulation. Further, we emphasize our understanding of the functional regulation of Super Elongation Complex (SEC)-mediated transcription, through regulation of stabilization by acetylation, deacetylation and ubiquitination and associated enzymes and its implication in human diseases.
Collapse
Affiliation(s)
- Avik Ghosh
- Laboratory of Transcription Biology Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Kolkata 32, India
| | - Poushali Chakraborty
- Laboratory of Transcription Biology Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Kolkata 32, India
| | - Debabrata Biswas
- Laboratory of Transcription Biology Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Kolkata 32, India.
| |
Collapse
|
10
|
Kazyken D, Lentz SI, Wadley M, Fingar DC. Alkaline intracellular pH (pHi) increases PI3K activity to promote mTORC1 and mTORC2 signaling and function during growth factor limitation. J Biol Chem 2023; 299:105097. [PMID: 37507012 PMCID: PMC10477693 DOI: 10.1016/j.jbc.2023.105097] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
The conserved protein kinase mTOR (mechanistic target of rapamycin) responds to diverse environmental cues to control cell metabolism and promote cell growth, proliferation, and survival as part of two multiprotein complexes, mTOR complex 1 (mTORC1) and mTORC2. Our prior work demonstrated that an alkaline intracellular pH (pHi) increases mTORC2 activity and cell survival in complete media in part by activating AMP-activated protein kinase, a kinase best known to sense energetic stress. It is important to note that an alkaline pHi represents an underappreciated hallmark of cancer cells that promotes their oncogenic behaviors. In addition, mechanisms that control mTORC1 and mTORC2 signaling and function remain incompletely defined, particularly in response to stress conditions. Here, we demonstrate that an alkaline pHi increases phosphatidylinositide 3-kinase (PI3K) activity to promote mTORC1 and mTORC2 signaling in the absence of serum growth factors. Alkaline pHi increases mTORC1 activity through PI3K-Akt signaling, which mediates inhibitory phosphorylation of the upstream proteins tuberous sclerosis complex 2 and proline-rich Akt substrate of 40 kDa and dissociates tuberous sclerosis complex from lysosomal membranes, thus enabling Rheb-mediated activation of mTORC1. Thus, alkaline pHi mimics growth factor-PI3K signaling. Functionally, we also demonstrate that an alkaline pHi increases cap-dependent protein synthesis through inhibitory phosphorylation of eIF4E binding protein 1 and suppresses apoptosis in a PI3K- and mTOR-dependent manner. We speculate that an alkaline pHi promotes a low basal level of cell metabolism (e.g., protein synthesis) that enables cancer cells within growing tumors to proliferate and survive despite limiting growth factors and nutrients, in part through elevated PI3K-mTORC1 and/or PI3K-mTORC2 signaling.
Collapse
Affiliation(s)
- Dubek Kazyken
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
| | - Stephen I Lentz
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Maxwell Wadley
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Diane C Fingar
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
| |
Collapse
|
11
|
Lee I, Doepner M, Weissenrieder J, Majer AD, Mercado S, Estell A, Natale CA, Sung PJ, Foskett JK, Carroll MP, Ridky TW. LNS8801 inhibits Acute Myeloid Leukemia by Inducing the Production of Reactive Oxygen Species and Activating the Endoplasmic Reticulum Stress Pathway. CANCER RESEARCH COMMUNICATIONS 2023; 3:1594-1606. [PMID: 37599786 PMCID: PMC10438922 DOI: 10.1158/2767-9764.crc-22-0478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 03/24/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023]
Abstract
Despite recent therapeutic advances, the 5-year survival rate for adults with acute myeloid leukemia (AML) is poor and standard-of-care chemotherapy is associated with significant toxicity, highlighting the need for new therapeutic approaches. Recent work from our group and others established that the G protein-coupled estrogen receptor (GPER) is tumor suppressive in melanoma and other solid tumors. We performed a preliminary screen of human cancer cell lines from multiple malignancies and found that LNS8801, a synthetic pharmacologic agonist of GPER currently in early phase clinical trials, promoted apoptosis in human AML cells. Using human AML cell lines and primary cells, we show that LNS8801 inhibits human AML in preclinical in vitro models, while not affecting normal mononuclear cells. Although GPER is broadly expressed in normal and malignant myeloid cells, this cancer-specific LNS8801-induced inhibition appeared to be independent of GPER signaling. LNS8801 induced AML cell death primarily through a caspase-dependent apoptosis pathway. This was independent of secreted classical death receptor ligands, and instead required induction of reactive oxygen species (ROS) and activation of endoplasmic reticulum (ER) stress response pathways including IRE1α. These studies demonstrate a novel activity of LNS8801 in AML cells and show that targeting ER stress with LNS8801 may be a useful therapeutic approach for AML. Significance Previous work demonstrated that LNS8801 inhibits cancer via GPER activation, especially in solid tumors. Here we show that LNS8801 inhibits AML via GPER-independent mechanisms that include ROS induction and ER activation.
Collapse
Affiliation(s)
- Inyoung Lee
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Miriam Doepner
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jillian Weissenrieder
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ariana D. Majer
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sophia Mercado
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Angela Estell
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Christopher A. Natale
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Linnaeus Therapeutics, Haddonfield, New Jersey
| | - Pamela J. Sung
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New Jersey
| | - J. Kevin Foskett
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Martin P. Carroll
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Todd W. Ridky
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
12
|
Dong S, Matossian MD, Yousefi H, Khosla M, Collins-Burow BM, Burow ME, Alahari SK. Targeting Mcl-1 by a small molecule NSC260594 for triple-negative breast cancer therapy. Sci Rep 2023; 13:11843. [PMID: 37481672 PMCID: PMC10363135 DOI: 10.1038/s41598-023-37058-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 06/15/2023] [Indexed: 07/24/2023] Open
Abstract
Triple-negative breast cancers (TNBCs) are aggressive forms of breast cancer and tend to grow and spread more quickly than most other types of breast cancer. TNBCs can neither be targeted by hormonal therapies nor the antibody trastuzumab that targets the HER2 protein. There are urgent unmet medical needs to develop targeted drugs for TNBCs. We identified a small molecule NSC260594 from the NCI diversity set IV compound library. NSC260594 exhibited dramatic cytotoxicity in multiple TNBCs in a dose-and time-dependent manner. NSC260594 inhibited the Myeloid cell leukemia-1 (Mcl-1) expression through downregulation of Wnt signaling proteins. Consistent with this, NSC260594 treatment increased apoptosis, which was confirmed by using an Annexin-V/PI assay. Interestingly, NSC260594 treatment reduced the cancer stem cell (CSC) population in TNBCs. To make NSC260594 more clinically relevant, we treated NSC260594 with TNBC cell derived xenograft (CDX) mouse model, and with patient-derived xenograft (PDX) organoids. NSC260594 significantly suppressed MDA-MB-231 tumor growth in vivo, and furthermore, the combination treatment of NSC260594 and everolimus acted synergistically to decrease growth of TNBC PDX organoids. Together, we found that NSC260594 might serve as a lead compound for triple-negative breast cancer therapy through targeting Mcl-1.
Collapse
Affiliation(s)
- Shengli Dong
- TYK Medicines Inc., Block D, No. 778 Huaxi Avenue, Changxing, 313100, Zhejiang, People's Republic of China.
- Department of Biochemistry and Molecular Biology, LSUHSC, CSRB 406, New Orleans, LA, 70112, USA.
| | | | - Hassan Yousefi
- Department of Biochemistry and Molecular Biology, LSUHSC, CSRB 406, New Orleans, LA, 70112, USA
| | - Maninder Khosla
- Department of Biochemistry and Molecular Biology, LSUHSC, CSRB 406, New Orleans, LA, 70112, USA
| | | | - Matthew E Burow
- Tulane University School of Medicine, New Orleans, LA, 70118, USA
| | - Suresh K Alahari
- Department of Biochemistry and Molecular Biology, LSUHSC, CSRB 406, New Orleans, LA, 70112, USA.
- Stanley S. Scott Cancer Center, LSUHSC School of Medicine, New Orleans, LA, 70112, USA.
| |
Collapse
|
13
|
Liang XW, Liu B, Chen JC, Cao Z, Chu FR, Lin X, Wang SZ, Wu JC. Characteristics and molecular mechanism of drug-tolerant cells in cancer: a review. Front Oncol 2023; 13:1177466. [PMID: 37483492 PMCID: PMC10360399 DOI: 10.3389/fonc.2023.1177466] [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: 03/01/2023] [Accepted: 06/23/2023] [Indexed: 07/25/2023] Open
Abstract
Drug resistance in tumours has seriously hindered the therapeutic effect. Tumour drug resistance is divided into primary resistance and acquired resistance, and the recent study has found that a significant proportion of cancer cells can acquire stable drug resistance from scratch. This group of cells first enters the drug tolerance state (DT state) under drug pressure, and gradually acquires stable drug resistance through adaptive mutations in this state. Although the specific mechanisms underlying the formation of drug tolerant cells (DTCs) remain unclear, various proteins and signalling pathways have been identified as being involved in the formation of DTCs. In the current review, we summarize the characteristics, molecular mechanisms and therapeutic strategies of DTCs in detail.
Collapse
Affiliation(s)
- Xian-Wen Liang
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Bing- Liu
- Department of Gastrointestinal Surgery, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, China
| | - Jia-Cheng Chen
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Zhi Cao
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Feng-ran Chu
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Xiong Lin
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Sheng-Zhong Wang
- Department of Gastrointestinal Surgery, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, China
| | - Jin-Cai Wu
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| |
Collapse
|
14
|
Meng D, Zhao X, Yang YC, Navickas A, Helland C, Goodarzi H, Singh M, Bandyopadhyay S. A bi-steric mTORC1-selective inhibitor overcomes drug resistance in breast cancer. Oncogene 2023:10.1038/s41388-023-02737-z. [PMID: 37264081 DOI: 10.1038/s41388-023-02737-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 04/30/2023] [Accepted: 05/18/2023] [Indexed: 06/03/2023]
Abstract
Activation of the PI3K-mTOR pathway is central to breast cancer pathogenesis including resistance to many targeted therapies. The mTOR kinase forms two distinct complexes, mTORC1 and mTORC2, and understanding which is required for the survival of malignant cells has been limited by tools to selectively and completely impair either subcomplex. To address this, we used RMC-6272, a bi-steric molecule with a rapamycin-like moiety linked to an mTOR active-site inhibitor that displays >25-fold selectivity for mTORC1 over mTORC2 substrates. Complete suppression of mTORC1 by RMC-6272 causes apoptosis in ER+/HER2- breast cancer cell lines, particularly in those that harbor mutations in PIK3CA or PTEN, due to inhibition of the rapamycin resistant, mTORC1 substrate 4EBP1 and reduction of the pro-survival protein MCL1. RMC-6272 reduced translation of ribosomal mRNAs, MYC target genes, and components of the CDK4/6 pathway, suggesting enhanced impairment of oncogenic pathways compared to the partial mTORC1 inhibitor everolimus. RMC-6272 maintained efficacy in hormone therapy-resistant acquired cell lines and patient-derived xenografts (PDX), showed increased efficacy in CDK4/6 inhibitor treated acquired resistant cell lines versus their parental counterparts, and was efficacious in a PDX from a patient experiencing resistance to CDK4/6 inhibition. Bi-steric mTORC1-selective inhibition may be effective in overcoming multiple forms of therapy-resistance in ER+ breast cancers.
Collapse
Affiliation(s)
- Delong Meng
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Xin Zhao
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Yu Chi Yang
- Department of Biology, Revolution Medicines Inc., Redwood City, CA, USA
| | - Albertas Navickas
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, 94158, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
- Institut Curie, PSL Research University, CNRS UMR3348, INSERM U1278, Orsay, France
| | - Ciara Helland
- Department of Biology, Revolution Medicines Inc., Redwood City, CA, USA
| | - Hani Goodarzi
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, 94158, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Mallika Singh
- Department of Biology, Revolution Medicines Inc., Redwood City, CA, USA
| | - Sourav Bandyopadhyay
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
15
|
Tantawy SI, Sarkar A, Hubner S, Tan Z, Wierda WG, Eldeib A, Zhang S, Kornblau S, Gandhi V. Mechanisms of MCL-1 Protein Stability Induced by MCL-1 Antagonists in B-Cell Malignancies. Clin Cancer Res 2023; 29:446-457. [PMID: 36346691 PMCID: PMC9852224 DOI: 10.1158/1078-0432.ccr-22-2088] [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: 07/03/2022] [Revised: 09/19/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022]
Abstract
PURPOSE Several MCL-1 inhibitors (MCL-1i), including AMG-176 and AZD5991, have shown promise in preclinical studies and are being tested for the treatment of hematologic malignancies. A unique feature of these agents is induction and stability of Mcl-1 protein; however, the precise mechanism is unknown. We aim to study the mechanism of MCL-1i-induced Mcl-1 protein stability. EXPERIMENTAL DESIGN Using several B-cell leukemia and lymphoma cell lines and primary chronic lymphocytic leukemia (CLL) lymphocytes, we evaluated molecular events associated with Mcl-1 protein stability including protein half-life, reverse-phase protein array, protein-protein interaction, phosphorylation, ubiquitination, and de-ubiquitination, followed by molecular simulation and modeling. RESULTS Using both in vivo and in vitro analysis, we demonstrate that MCL-1i-induced Mcl-1 protein stability is predominantly associated with defective Mcl-1 ubiquitination and concurrent apoptosis induction in both cell lines and primary CLL subjects. These MCL1i also induced ERK-mediated Mcl-1Thr163 phosphorylation, which partially contributed to Mcl-1 stability. Disruption of Mcl-1:Noxa interaction followed by Noxa degradation, enhanced Mcl-1 de-ubiquitination by USP9x, and Mule destabilization are the major effects of these inhibitors. However, unlike other BH3 proteins, Mule:Mcl-1 interaction was unaffected by MCL-1i. WP1130, a global deubiquitinase (DUB) inhibitor, abrogated Mcl-1 induction reaffirming a critical role of DUBs in the observed Mcl-1 protein stability. Further, in vitro ubiquitination studies of Mcl-1 showed distinct difference among these inhibitors. CONCLUSIONS We conclude that MCL-1i blocked Mcl-1 ubiquitination via enhanced de-ubiquitination and dissociation of Mcl-1 from Noxa, Bak and Bax, and Mule de-stabilization. These are critical events associated with increased Mcl-1 protein stability with AMG-176 and AZD5991.
Collapse
Affiliation(s)
- Shady I. Tantawy
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas,Department of Internal Medicine, College of Medicine, Suez Canal University, Ismailia, Egypt
| | - Aloke Sarkar
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stefan Hubner
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zhi Tan
- Center for Drug Discovery, Department of Pathology and Immunology, Department of Pharmacology and Chemical Biology, Baylor College of Medicine
| | - William G. Wierda
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Abdelraouf Eldeib
- Center for Drug Discovery, Department of Pathology and Immunology, Department of Pharmacology and Chemical Biology, Baylor College of Medicine
| | - Shuxing Zhang
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Steven Kornblau
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Varsha Gandhi
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas,Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
16
|
Al-Odat OS, Guirguis DA, Schmalbach NK, Yao G, Budak-Alpdogan T, Jonnalagadda SC, Pandey MK. Autophagy and Apoptosis: Current Challenges of Treatment and Drug Resistance in Multiple Myeloma. Int J Mol Sci 2022; 24:ijms24010644. [PMID: 36614089 PMCID: PMC9820338 DOI: 10.3390/ijms24010644] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022] Open
Abstract
Over the past two decades, the natural history of multiple myeloma (MM) has evolved dramatically, owing primarily to novel agents targeting MM in the bone marrow microenvironment (BMM) pathways. However, the mechanisms of resistance acquisition remain a mystery and are poorly understood. Autophagy and apoptosis are tightly controlled processes and play a critical role in the cell growth, development, and survival of MM. Genetic instability and abnormalities are two hallmarks of MM. During MM progression, plasma malignant cells become genetically unstable and activate various signaling pathways, resulting in the overexpression of abnormal proteins that disrupt autophagy and apoptosis biological processes. Thus, achieving a better understanding of the autophagy and apoptosis processes and the proteins that crosslinked both pathways, could provide new insights for the MM treatment and improve the development of novel therapeutic strategies to overcome resistance. This review presents a sufficient overview of the roles of autophagy and apoptosis and how they crosslink and control MM progression and drug resistance. Potential combination targeting of both pathways for improving outcomes in MM patients also has been addressed.
Collapse
Affiliation(s)
- Omar S. Al-Odat
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
- Department of Chemistry and Biochemistry, Rowan University, Glassboro, NJ 08028, USA
| | - Daniel A. Guirguis
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| | - Nicole K. Schmalbach
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| | - Gabriella Yao
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| | | | | | - Manoj K. Pandey
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
- Correspondence: ; Tel.: +1-856-956-2751
| |
Collapse
|
17
|
Targeting anti-apoptotic pathways eliminates senescent melanocytes and leads to nevi regression. Nat Commun 2022; 13:7923. [PMID: 36564381 PMCID: PMC9789033 DOI: 10.1038/s41467-022-35657-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Human melanocytic nevi (moles) result from a brief period of clonal expansion of melanocytes. As a cellular defensive mechanism against oncogene-induced hyperplasia, nevus-resident melanocytes enter a senescent state of stable cell cycle arrest. Senescent melanocytes can persist for months in mice and years in humans with a risk to escape the senescent state and progress to melanoma. The mechanisms providing prolonged survival of senescent melanocytes remain poorly understood. Here, we show that senescent melanocytes in culture and in nevi express high level of the anti-apoptotic BCL-2 family member BCL-W but remain insensitive to the pan-BCL-2 inhibitor ABT-263. We demonstrate that resistance to ABT-263 is driven by mTOR-mediated enhanced translation of another anti-apoptotic member, MCL-1. Strikingly, the combination of ABT-263 and MCL-1 inhibitors results in synthetic lethality to senescent melanocytes, and its topical application sufficient to eliminate nevi in male mice. These data highlight the important role of redundant anti-apoptotic mechanisms for the survival advantage of senescent melanocytes, and the proof-of-concept for a non-invasive combination therapy for nevi removal.
Collapse
|
18
|
McNamara MC, Hosios AM, Torrence ME, Zhao T, Fraser C, Wilkinson M, Kwiatkowski DJ, Henske EP, Wu CL, Sarosiek KA, Valvezan AJ, Manning BD. Reciprocal effects of mTOR inhibitors on pro-survival proteins dictate therapeutic responses in tuberous sclerosis complex. iScience 2022; 25:105458. [PMID: 36388985 PMCID: PMC9663903 DOI: 10.1016/j.isci.2022.105458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 09/30/2022] [Accepted: 10/23/2022] [Indexed: 11/13/2022] Open
Abstract
mTORC1 is aberrantly activated in cancer and in the genetic tumor syndrome tuberous sclerosis complex (TSC), which is caused by loss-of-function mutations in the TSC complex, a negative regulator of mTORC1. Clinically approved mTORC1 inhibitors, such as rapamycin, elicit a cytostatic effect that fails to eliminate tumors and is rapidly reversible. We sought to determine the effects of mTORC1 on the core regulators of intrinsic apoptosis. In TSC2-deficient cells and tumors, we find that mTORC1 inhibitors shift cellular dependence from MCL-1 to BCL-2 and BCL-XL for survival, thereby altering susceptibility to BH3 mimetics that target specific pro-survival BCL-2 proteins. The BCL-2/BCL-XL inhibitor ABT-263 synergizes with rapamycin to induce apoptosis in TSC-deficient cells and in a mouse tumor model of TSC, resulting in a more complete and durable response. These data expose a therapeutic vulnerability in regulation of the apoptotic machinery downstream of mTORC1 that promotes a cytotoxic response to rapamycin.
Collapse
Affiliation(s)
- Molly C. McNamara
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Aaron M. Hosios
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Margaret E. Torrence
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, USA
| | - Ting Zhao
- Department of Urology, Massachusetts General Hospital, Boston, MA, USA
- Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Cameron Fraser
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA 02215, USA
| | - Meghan Wilkinson
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - David J. Kwiatkowski
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Elizabeth P. Henske
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Chin-Lee Wu
- Department of Urology, Massachusetts General Hospital, Boston, MA, USA
- Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Kristopher A. Sarosiek
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA 02215, USA
| | - Alexander J. Valvezan
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, USA
| | - Brendan D. Manning
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
19
|
Dunn S, Eberlein C, Yu J, Gris-Oliver A, Ong SH, Yelland U, Cureton N, Staniszewska A, McEwen R, Fox M, Pilling J, Hopcroft P, Coker EA, Jaaks P, Garnett MJ, Isherwood B, Serra V, Davies BR, Barry ST, Lynch JT, Yusa K. AKT-mTORC1 reactivation is the dominant resistance driver for PI3Kβ/AKT inhibitors in PTEN-null breast cancer and can be overcome by combining with Mcl-1 inhibitors. Oncogene 2022; 41:5046-5060. [PMID: 36241868 PMCID: PMC9652152 DOI: 10.1038/s41388-022-02482-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 09/12/2022] [Accepted: 09/21/2022] [Indexed: 11/08/2022]
Abstract
The PI3K pathway is commonly activated in breast cancer, with PI3K-AKT pathway inhibitors used clinically. However, mechanisms that limit or enhance the therapeutic effects of PI3K-AKT inhibitors are poorly understood at a genome-wide level. Parallel CRISPR screens in 3 PTEN-null breast cancer cell lines identified genes mediating resistance to capivasertib (AKT inhibitor) and AZD8186 (PI3Kβ inhibitor). The dominant mechanism causing resistance is reactivated PI3K-AKT-mTOR signalling, but not other canonical signalling pathways. Deletion of TSC1/2 conferred resistance to PI3Kβi and AKTi through mTORC1. However, deletion of PIK3R2 and INPPL1 drove specific PI3Kβi resistance through AKT. Conversely deletion of PIK3CA, ERBB2, ERBB3 increased PI3Kβi sensitivity while modulation of RRAGC, LAMTOR1, LAMTOR4 increased AKTi sensitivity. Significantly, we found that Mcl-1 loss enhanced response through rapid apoptosis induction with AKTi and PI3Kβi in both sensitive and drug resistant TSC1/2 null cells. The combination effect was BAK but not BAX dependent. The Mcl-1i + PI3Kβ/AKTi combination was effective across a panel of breast cancer cell lines with PIK3CA and PTEN mutations, and delivered increased anti-tumor benefit in vivo. This study demonstrates that different resistance drivers to PI3Kβi and AKTi converge to reactivate PI3K-AKT or mTOR signalling and combined inhibition of Mcl-1 and PI3K-AKT has potential as a treatment strategy for PI3Kβi/AKTi sensitive and resistant breast tumours.
Collapse
Affiliation(s)
- Shanade Dunn
- Wellcome Sanger Institute, Cambridge, UK
- Bioscience, Early Oncology, AstraZeneca, Cambridge, UK
| | - Cath Eberlein
- Bioscience, Early Oncology, AstraZeneca, Alderley Park, UK
| | - Jason Yu
- Wellcome Sanger Institute, Cambridge, UK
- Molecular Biology of Metabolism Lab, The Francis Crick Institute, London, UK
| | | | | | - Urs Yelland
- Bioscience, Early Oncology, AstraZeneca, Alderley Park, UK
| | | | | | - Robert McEwen
- Bioscience, Early Oncology, AstraZeneca, Cambridge, UK
| | - Millie Fox
- Bioscience, Early Oncology, AstraZeneca, Cambridge, UK
| | | | | | | | | | | | | | - Violeta Serra
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | | | - Simon T Barry
- Bioscience, Early Oncology, AstraZeneca, Cambridge, UK.
| | - James T Lynch
- Bioscience, Early Oncology, AstraZeneca, Cambridge, UK
| | - Kosuke Yusa
- Wellcome Sanger Institute, Cambridge, UK.
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.
| |
Collapse
|
20
|
Laukkanen S, Veloso A, Yan C, Oksa L, Alpert EJ, Do D, Hyvärinen N, McCarthy K, Adhikari A, Yang Q, Iyer S, Garcia SP, Pello A, Ruokoranta T, Moisio S, Adhikari S, Yoder JA, Gallagher K, Whelton L, Allen JR, Jin AH, Loontiens S, Heinäniemi M, Kelliher M, Heckman CA, Lohi O, Langenau DM. Therapeutic targeting of LCK tyrosine kinase and mTOR signaling in T-cell acute lymphoblastic leukemia. Blood 2022; 140:1891-1906. [PMID: 35544598 PMCID: PMC10082361 DOI: 10.1182/blood.2021015106] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 04/19/2022] [Indexed: 11/20/2022] Open
Abstract
Relapse and refractory T-cell acute lymphoblastic leukemia (T-ALL) has a poor prognosis, and new combination therapies are sorely needed. Here, we used an ex vivo high-throughput screening platform to identify drug combinations that kill zebrafish T-ALL and then validated top drug combinations for preclinical efficacy in human disease. This work uncovered potent drug synergies between AKT/mTORC1 (mammalian target of rapamycin complex 1) inhibitors and the general tyrosine kinase inhibitor dasatinib. Importantly, these same drug combinations effectively killed a subset of relapse and dexamethasone-resistant zebrafish T-ALL. Clinical trials are currently underway using the combination of mTORC1 inhibitor temsirolimus and dasatinib in other pediatric cancer indications, leading us to prioritize this therapy for preclinical testing. This combination effectively curbed T-ALL growth in human cell lines and primary human T-ALL and was well tolerated and effective in suppressing leukemia growth in patient-derived xenografts (PDX) grown in mice. Mechanistically, dasatinib inhibited phosphorylation and activation of the lymphocyte-specific protein tyrosine kinase (LCK) to blunt the T-cell receptor (TCR) signaling pathway, and when complexed with mTORC1 inhibition, induced potent T-ALL cell killing through reducing MCL-1 protein expression. In total, our work uncovered unexpected roles for the LCK kinase and its regulation of downstream TCR signaling in suppressing apoptosis and driving continued leukemia growth. Analysis of a wide array of primary human T-ALLs and PDXs grown in mice suggest that combination of temsirolimus and dasatinib treatment will be efficacious for a large fraction of human T-ALLs.
Collapse
Affiliation(s)
- Saara Laukkanen
- Tampere Center for Child, Adolescent, and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Alexandra Veloso
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Chuan Yan
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Laura Oksa
- Tampere Center for Child, Adolescent, and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Eric J. Alpert
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Daniel Do
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Noora Hyvärinen
- Tampere Center for Child, Adolescent, and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Karin McCarthy
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Abhinav Adhikari
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Qiqi Yang
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Sowmya Iyer
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Sara P. Garcia
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Annukka Pello
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Tanja Ruokoranta
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Sanni Moisio
- The Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Sadiksha Adhikari
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Jeffrey A. Yoder
- Department of Molecular Biomedical Sciences, Comparative Medicine Institute, and Center for Human Health and the Environment, North Carolina State University, Raleigh, NC
| | - Kayleigh Gallagher
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA
| | - Lauren Whelton
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - James R. Allen
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Alex H. Jin
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Siebe Loontiens
- Cancer Research Institute Ghent and Center for Medical Genetics, Ghent, Belgium
| | - Merja Heinäniemi
- The Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Michelle Kelliher
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA
| | - Caroline A. Heckman
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Olli Lohi
- Tampere Center for Child, Adolescent, and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tampere University Hospital, Tays Cancer Center, Tampere, Finland
| | - David M. Langenau
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
21
|
Sancho M, Leiva D, Lucendo E, Orzáez M. Understanding MCL1: from cellular function and regulation to pharmacological inhibition. FEBS J 2022; 289:6209-6234. [PMID: 34310025 PMCID: PMC9787394 DOI: 10.1111/febs.16136] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/09/2021] [Accepted: 07/22/2021] [Indexed: 12/30/2022]
Abstract
Myeloid cell leukemia-1 (MCL1), an antiapoptotic member of the BCL2 family characterized by a short half-life, functions as a rapid sensor that regulates cell death and other relevant processes that include cell cycle progression and mitochondrial homeostasis. In cancer, MCL1 overexpression contributes to cell survival and resistance to diverse chemotherapeutic agents; for this reason, several MCL1 inhibitors are currently under preclinical and clinical development for cancer treatment. However, the nonapoptotic functions of MCL1 may influence their therapeutic potential. Overall, the complexity of MCL1 regulation and function represent challenges to the clinical application of MCL1 inhibitors. We now summarize the current knowledge regarding MCL1 structure, regulation, and function that could impact the clinical success of MCL1 inhibitors.
Collapse
Affiliation(s)
- Mónica Sancho
- Targeted Therapies on Cancer and Inflammation LaboratoryCentro de Investigación Príncipe FelipeValenciaSpain
| | - Diego Leiva
- Targeted Therapies on Cancer and Inflammation LaboratoryCentro de Investigación Príncipe FelipeValenciaSpain
| | - Estefanía Lucendo
- Targeted Therapies on Cancer and Inflammation LaboratoryCentro de Investigación Príncipe FelipeValenciaSpain
| | - Mar Orzáez
- Targeted Therapies on Cancer and Inflammation LaboratoryCentro de Investigación Príncipe FelipeValenciaSpain
| |
Collapse
|
22
|
Petsri K, Thongsom S, Racha S, Chamni S, Jindapol S, Kaekratoke N, Zou H, Chanvorachote P. Novel mechanism of napabucasin, a naturally derived furanonaphthoquinone: apoptosis and autophagy induction in lung cancer cells through direct targeting on Akt/mTOR proteins. BMC Complement Med Ther 2022; 22:250. [PMID: 36180880 PMCID: PMC9524025 DOI: 10.1186/s12906-022-03727-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 09/15/2022] [Indexed: 11/22/2022] Open
Abstract
Background Akt and mTOR are aberrantly activated in cancers and targeting these proteins are interesting for cancer drug discovery. Napabucasin (NB), a phytochemical compound, has been reported as potential anti-cancer agent, however, Akt and mTOR targeting mechanisms remain unclear. Method Apoptosis induction was investigated by Hoechst 33342/PI double staining and annexin V/PI staining with flowcytometry. Autophagy was evaluated by monodansylcadaverine staining and Western blot analysis. Binding affinity of NB and essential signaling proteins (PI3K, Akt, and mTOR) was investigated using molecular docking and confirmed by Western blot analysis. Result A structure modification from changing methyl moiety of acetyl group of NB to hydroxyl moiety of carboxyl group of NB derivative (napabucasin-acid or NB-acid) greatly affected the compound activities. NB showed more potent anti-cancer activity. NB reduced cell viability with an approximately 20 times lower IC50 and inhibited the colony formation capacity much more than NB-acid treated cells. NB induced cell apoptosis, which was accompanied by decrease Bcl‑2 and Mcl-1 and clevage of PARP, while NB-acid show lesser effect on Mcl-1. NB was found to strongly induce autophagy indicated by acidic vesicle staining and the LC3B conversion. Interestingly, computational molecular docking analysis further demonstrated that NB directly bound to Akt and mTOR (complex 1 and 2) proteins at their critical sites indicating that NB targets the upstream regulators of apoptosis and autophagy. The docking results were confirmed by decrease of p-Akt/Akt, p-mTOR/mTOR, and c-Myc a downstream target of Akt protein levels. Conclusion Results show for the first time that NB exerts an anti-cancer activity through the direct interaction to Akt and mTOR proteins. The methyl moiety of acetyl group of NB is required for its potent anti-cancer activities. These data encourage further development of NB compounds for Akt and mTOR driven cancers. Supplementary Information The online version contains supplementary material available at 10.1186/s12906-022-03727-6.
Collapse
|
23
|
Hong J, Zheng W, Cai X. Small-molecule high-throughput screening identifies a MEK inhibitor PD1938306 that enhances sorafenib efficacy via MCL-1 and BIM in hepatocellular carcinoma cells. Comb Chem High Throughput Screen 2022; 26:1364-1374. [PMID: 36043792 PMCID: PMC9971357 DOI: 10.2174/1386207325666220830145026] [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: 02/06/2022] [Revised: 07/03/2022] [Accepted: 07/07/2022] [Indexed: 11/22/2022]
Abstract
Background Sorafenib is the most widely used systematic therapy drug for treating unresectable hepatocellular carcinoma (HCC) but showed dissatisfactory efficacy in clinical applications. Objective We conducted a combinational quantitative small-molecule high-throughput screening (qHTS) to identify potential candidates to enhance the treatment effectiveness of sorafenib. Methods First, using a Hep3B human HCC cell line, 7051 approved drugs and bioactive compounds were screened, then the primary hits were tested with/ without 0.5 μM sorafenib respectively, the compound has the half maximal inhibitory concentration (IC50) shift value greater than 1.5 was thought to have the synergistic effect with sorafenib. Furthermore, the MEK inhibitor PD198306 was selected for further mechanistic study. Results 12 effective compounds were identified, including kinase inhibitors that target MEK, AURKB, CAMK, ROCK2, BRAF, PI3K, AKT and EGFR, as well as a μ-opioid receptor agonist and a L-type calcium channel blocker. The mechanistic research of the combination of sorafenib plus PD198306 showed that the two compounds synergistically inhibited MEK-ERK and mTORC1-4EBP1, and induced apoptosis in HCC cells, which can be attributed to the transcriptional and posttranslational regulation of MCL-1 and BIM. Conclusion Small-molecule qHTS identifies MEK inhibitor PD1938306 as a potent sorafenib enhancer, together with several novel combination strategies that are valuable for further studies.
Collapse
Affiliation(s)
- Junjie Hong
- Department of General Surgery, Key Laboratory of Laparoscopic Technique Research of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310016, China,National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Xiujun Cai
- Department of General Surgery, Key Laboratory of Laparoscopic Technique Research of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310016, China,Correspondence to: Xiujun Cai, 3 East Qingchun Road, Jianggan District, Hangzhou 310000, China. Tel: +86-0571-8600-6617; Fax: +86-0571-8604-4817;
| |
Collapse
|
24
|
Manni S, Pesavento M, Spinello Z, Saggin L, Arjomand A, Fregnani A, Quotti Tubi L, Scapinello G, Gurrieri C, Semenzato G, Trentin L, Piazza F. Protein Kinase CK2 represents a new target to boost Ibrutinib and Venetoclax induced cytotoxicity in mantle cell lymphoma. Front Cell Dev Biol 2022; 10:935023. [PMID: 36035991 PMCID: PMC9403710 DOI: 10.3389/fcell.2022.935023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
Mantle cell lymphoma (MCL) is an incurable B cell non-Hodgkin lymphoma, characterized by frequent relapses. In the last decade, the pro-survival pathways related to BCR signaling and Bcl-2 have been considered rational therapeutic targets in B cell derived lymphomas. The BTK inhibitor Ibrutinib and the Bcl-2 inhibitor Venetoclax are emerging as effective drugs for MCL. However, primary and acquired resistance also to these agents may occur. Protein Kinase CK2 is a S/T kinase overexpressed in many solid and blood-derived tumours. CK2 promotes cancer cell growth and clonal expansion, sustaining pivotal survival signaling cascades, such as the ones dependent on AKT, NF-κB, STAT3 and others, counteracting apoptosis through a “non-oncogene” addiction mechanism. We previously showed that CK2 is overexpressed in MCL and regulates the levels of activating phosphorylation on S529 of the NF-κB family member p65/RelA. In the present study, we investigated the effects of CK2 inactivation on MCL cell proliferation, survival and apoptosis and this kinase’s involvement in the BCR and Bcl-2 related signaling. By employing CK2 loss of function MCL cell models, we demonstrated that CK2 sustains BCR signaling (such as BTK, NF-κB and AKT) and the Bcl-2-related Mcl-1 expression. CK2 inactivation enhanced Ibrutinib and Venetoclax-induced cytotoxicity. The demonstration of a CK2-dependent upregulation of pathways that may antagonize the effect of these drugs may offer a novel strategy to overcome primary and secondary resistance.
Collapse
Affiliation(s)
- Sabrina Manni
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
- *Correspondence: Sabrina Manni, ; Francesco Piazza,
| | - Maria Pesavento
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Zaira Spinello
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Lara Saggin
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Arash Arjomand
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Anna Fregnani
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Laura Quotti Tubi
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Greta Scapinello
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Carmela Gurrieri
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Gianpietro Semenzato
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Livio Trentin
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Francesco Piazza
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
- *Correspondence: Sabrina Manni, ; Francesco Piazza,
| |
Collapse
|
25
|
Narayan S, Raza A, Mahmud I, Koo N, Garrett TJ, Law ME, Law BK, Sharma AK. Sensitization of FOLFOX-resistant colorectal cancer cells via the modulation of a novel pathway involving protein phosphatase 2A. iScience 2022; 25:104518. [PMID: 35754740 PMCID: PMC9218363 DOI: 10.1016/j.isci.2022.104518] [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: 08/16/2021] [Revised: 05/16/2022] [Accepted: 05/30/2022] [Indexed: 12/04/2022] Open
Abstract
The treatment of colorectal cancer (CRC) with FOLFOX shows some efficacy, but these tumors quickly develop resistance to this treatment. We have observed increased phosphorylation of AKT1/mTOR/4EBP1 and levels of p21 in FOLFOX-resistant CRC cells. We have identified a small molecule, NSC49L, that stimulates protein phosphatase 2A (PP2A) activity, downregulates the AKT1/mTOR/4EBP1-axis, and inhibits p21 translation. We have provided evidence that NSC49L- and TRAIL-mediated sensitization is synergistically induced in p21-knockdown CRC cells, which is reversed in p21-overexpressing cells. p21 binds with procaspase 3 and prevents the activation of caspase 3. We have shown that TRAIL induces apoptosis through the activation of caspase 3 by NSC49L-mediated downregulation of p21 translation, and thereby cleavage of procaspase 3 into caspase 3. NSC49L does not affect global protein synthesis. These studies provide a mechanistic understanding of NSC49L as a PP2A agonist, and how its combination with TRAIL sensitizes FOLFOX-resistant CRC cells.
Collapse
Affiliation(s)
- Satya Narayan
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL 32610, USA
| | - Asif Raza
- Department of Pharmacology, Penn State University College of Medicine, Penn State Cancer Institute, Hershey, PA 17033, USA
| | - Iqbal Mahmud
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Nayeong Koo
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL 32610, USA
| | - Timothy J. Garrett
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Mary E. Law
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA
| | - Brian K. Law
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA
| | - Arun K. Sharma
- Department of Pharmacology, Penn State University College of Medicine, Penn State Cancer Institute, Hershey, PA 17033, USA
| |
Collapse
|
26
|
Winder ML, Campbell KJ. MCL-1 is a clinically targetable vulnerability in breast cancer. Cell Cycle 2022; 21:1439-1455. [PMID: 35349392 PMCID: PMC9278428 DOI: 10.1080/15384101.2022.2054096] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 03/03/2022] [Accepted: 03/11/2022] [Indexed: 11/03/2022] Open
Abstract
Pro-survival members of the BCL-2 family, including MCL-1, are emerging as important proteins during the development and therapeutic response of solid tumors. Notably, high levels of MCL-1 occur in breast cancer, where functional dependency has been demonstrated using cell lines and mouse models. The utility of restoring apoptosis in cancer cells through inhibition of pro-survival BCL-2 proteins has been realized in the clinic, where the first specific inhibitor of BCL-2 is approved for use in leukemia. A variety of MCL-1 inhibitors are now undergoing clinical trials for blood cancer treatment and application of this new class of drugs is also being tested in solid cancers. On-target compounds specific to MCL-1 have demonstrated promising efficacy in preclinical models of breast cancer and show potential to enhance the anti-tumor effect of conventional therapies. Taken together, this makes MCL-1 an extremely attractive target for clinical evaluation in the context of breast cancer.Abbreviations: ADC (antibody-drug conjugate); AML (Acute myeloid leukemia); APAF1 (apoptotic protease activating factor 1); bCAFs (breast cancer associated fibroblasts); BCL-2 (B-cell lymphoma 2); BH (BCL-2 homology); CLL (chronic lymphocytic leukemia); EGF (epidermal growth factor); EMT (epithelial to mesenchymal transition); ER (estrogen receptor); FDA (food and drug administration); GEMM (genetically engineered mouse model); HER2 (human epidermal growth factor 2); IL6 (interleukin 6); IMM (inner mitochondrial membrane); IMS (intermembrane space); MCL-1 (myeloid cell leukemia-1); MOMP (mitochondrial outer membrane permeabilisation); MM (multiple myeloma); PDX (patient-derived xenograft); OMM (outer mitochondrial membrane); PROTAC (proteolysis-targeting chimeras) TNBC (triple negative breast cancer); UPS (ubiquitin mediated proteolysis system).
Collapse
Affiliation(s)
- Matthew L Winder
- CRUK Beatson Institute, Garscube Estate,Switchback Road, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - Kirsteen J Campbell
- CRUK Beatson Institute, Garscube Estate,Switchback Road, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| |
Collapse
|
27
|
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
|
28
|
Thomas AF, Kelly GL, Strasser A. Of the many cellular responses activated by TP53, which ones are critical for tumour suppression? Cell Death Differ 2022; 29:961-971. [PMID: 35396345 PMCID: PMC9090748 DOI: 10.1038/s41418-022-00996-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 03/24/2022] [Accepted: 03/28/2022] [Indexed: 12/12/2022] Open
Abstract
The tumour suppressor TP53 is a master regulator of several cellular processes that collectively suppress tumorigenesis. The TP53 gene is mutated in ~50% of human cancers and these defects usually confer poor responses to therapy. The TP53 protein functions as a homo-tetrameric transcription factor, directly regulating the expression of ~500 target genes, some of them involved in cell death, cell cycling, cell senescence, DNA repair and metabolism. Originally, it was thought that the induction of apoptotic cell death was the principal mechanism by which TP53 prevents the development of tumours. However, gene targeted mice lacking the critical effectors of TP53-induced apoptosis (PUMA and NOXA) do not spontaneously develop tumours. Indeed, even mice lacking the critical mediators for TP53-induced apoptosis, G1/S cell cycle arrest and cell senescence, namely PUMA, NOXA and p21, do not spontaneously develop tumours. This suggests that TP53 must activate additional cellular responses to mediate tumour suppression. In this review, we will discuss the processes by which TP53 regulates cell death, cell cycling/cell senescence, DNA damage repair and metabolic adaptation, and place this in context of current understanding of TP53-mediated tumour suppression.
Collapse
Affiliation(s)
- Annabella F Thomas
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,The Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Gemma L Kelly
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,The Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia. .,The Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
29
|
Narla ST, Bushnell DS, Duara JL, Bates CM. AKT Signaling Downstream of KGF Is Necessary and Sufficient for Blocking Cyclophosphamide Bladder Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:604-612. [PMID: 35063403 PMCID: PMC8961277 DOI: 10.1016/j.ajpath.2022.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/23/2021] [Accepted: 01/04/2022] [Indexed: 01/01/2023]
Abstract
Keratinocyte growth factor (KGF) drives phosphorylated (activated) AKT (pAKT) in bladder urothelium, which correlates with cytoprotection from cyclophosphamide. The current study determined whether: i) KGF modifies AKT targets [B-cell lymphoma protein 2-associated agonist of cell death (BAD) and mammalian target of rapamycin complex (mTORC)-1] that could block apoptosis; ii) AKT signaling is required for KGF cytoprotection; iii) direct AKT activation drives cytoprotection; iv) co-administration of KGF and an AKT inhibitor blocks urothelial cytoprotection and AKT and AKT-target activation; and v) an AKT agonist prevents cyclophosphamide-induced urothelial apoptosis. Mice were given KGF and cyclophosphamide (or sham injury), and pBAD (readout of BAD inhibition) or p-p70S6k (pS6, readout of mTORC1 signaling) was assessed. KGF induced pBAD urothelial staining and prevented cyclophosphamide-induced loss of urothelial pS6 staining (likely stabilizing mTORC1 activity). Co-administration of KGF and AKT inhibitor blocked KGF-driven urothelial cytoprotection from cyclophosphamide and prevented pAKT, pBAD, and pS6 urothelial expression. Conversely, systemic AKT agonist blocked cyclophosphamide-induced urothelial apoptosis and induced pAKT, pBAD, and pS6, similar to KGF. Thus, the KGF-AKT signaling axis appeared to phosphorylate (suppress) BAD and prevent cyclophosphamide-induced loss of mTORC1 signaling, both of which likely suppress apoptosis. Additionally, AKT signaling was required for KGF-driven cytoprotection, and direct AKT activation was sufficient for blocking apoptosis. Thus, AKT may be a therapeutic target for blocking urothelial apoptosis from cyclophosphamide.
Collapse
Affiliation(s)
- Sridhar T Narla
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Daniel S Bushnell
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Joanne L Duara
- Division of Neonatology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Carlton M Bates
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Division of Nephrology, Children's Hospital, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.
| |
Collapse
|
30
|
Narla ST, Rice L, Ostrov D, Swarts SG, Siemann D, Bushnell DS, Holden JG, Duara J, Bates CM. FGF7 peptide (FGF7p) mimetic mitigates bladder urothelial injury from cyclophosphamide. Physiol Rep 2022; 10:e15241. [PMID: 35388988 PMCID: PMC8988121 DOI: 10.14814/phy2.15241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/04/2022] [Accepted: 03/08/2022] [Indexed: 04/17/2023] Open
Abstract
Although full-length fibroblast growth factor 7 (FGF7) blocks cyclophosphamide-induced urothelial apoptosis in mice, limitations include high production costs because of its large size. We previously identified a small peptide derived from FGF2 that mitigated acute radiation syndrome as well as full-length FGF2. Based on the sequence of the FGF2 peptide, we synthesized a corresponding 19 amino acid FGF7 peptide (FGF7p). Our objectives were to determine if systemic FGF7p triggered the downstream targets and protected against cyclophosphamide bladder injury similar to full-length FGF7. We administered FGF7p or vehicle subcutaneously (SQ) to mice subjected to no injury or intraperitoneal (IP) cyclophosphamide and harvested bladders 1 day after injury. We then performed hematoxylin and eosin, TUNEL and immunofluorescence (IF) staining. In uninjured mice, a 20 mg/kg threshold FGF7p dose induced expression of phosphorylated (activated) FRS2α (pFRS2α), and pAKT in urothelium (consistent with cytoprotective effects of FGF7). We then gave FGF7p (20 mg/kg) or vehicle at 72 and 48 h prior to cyclophosphamide. One day after injury, TUNEL staining revealed many more apoptotic urothelial cells with vehicle treatment versus FGF7p treatment. IF for pAKT and readouts of two anti-apoptotic AKT targets (BAD and mTORC1) revealed minimal staining with vehicle treatment, but strong urothelial expression for all markers with FGF7p treatment. In conclusion, FGF7p appears to block bladder urothelial apoptosis via AKT and its targets, similar to FGF7. FGF7p is much more inexpensive to make and has a longer shelf life and higher purity than FGF7.
Collapse
Affiliation(s)
- Sridhar Tatarao Narla
- Division of NephrologyDepartment of PediatricsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Lori Rice
- Department of Radiation OncologyCollege of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - David Ostrov
- Department of Pathology, Immunology and Laboratory MedicineUniversity of Florida College of MedicineGainesvilleFloridaUSA
| | - Steven G. Swarts
- Department of Radiation OncologyCollege of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Dietmar W. Siemann
- Department of Radiation OncologyCollege of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Daniel Scott Bushnell
- Division of NephrologyDepartment of PediatricsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Jacqueline G. Holden
- Division of NephrologyDepartment of PediatricsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Joanne Lindsey Duara
- Division of NeonatologyDepartment of PediatricsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Carlton Matthew Bates
- Division of NephrologyDepartment of PediatricsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Division of NephrologyUPMC Children’s Hospital of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
31
|
Rapaka D, Bitra VR, Challa SR, Adiukwu PC. mTOR signaling as a molecular target for the alleviation of Alzheimer's disease pathogenesis. Neurochem Int 2022; 155:105311. [PMID: 35218870 DOI: 10.1016/j.neuint.2022.105311] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/12/2022] [Accepted: 02/20/2022] [Indexed: 10/19/2022]
Abstract
Mechanistic/mammalian target of rapamycin (mTOR) belongs to the phosphatidylinositol kinase-related kinase (PIKK) family. mTOR signaling is required for the commencement of essential cell functions including autophagy. mTOR primarily governs cell growth in response to favourable nutrients and other growth stimuli. However, it also influences aging and other aspects of nutrient-related physiology such as protein synthesis, ribosome biogenesis, and cell proliferation in adults with very limited growth. The major processes for survival such as synaptic plasticity, memory storage and neuronal recovery involve a significant mTOR activity. mTOR dysregulation is becoming a prevalent motif in a variety of human diseases, including cancer, neurological disorders, and other metabolic syndromes. The use of rapamycin to prolong life in different animal models may be attributable to the multiple roles played by mTOR signaling in various processes involved in ageing, protein translation, autophagy, stem cell pool turnover, inflammation, and cellular senescence. mTOR activity was found to be altered in AD brains and rodent models, supporting the notion that aberrant mTOR activity is one of the key events contributing to the onset and progression of AD hallmarks This review assesses the molecular association between the mTOR signaling pathway and pathogenesis of Alzheimer's disease. The research data supporting this theme are also reviewed.
Collapse
Affiliation(s)
- Deepthi Rapaka
- A.U. College of Pharmaceutical Sciences, Andhra University, Visakhapatnam, 530003, India.
| | | | - Siva Reddy Challa
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL, 61614, USA.
| | - Paul C Adiukwu
- School of Pharmacy, University of Botswana, Gaborone, 0022, Botswana.
| |
Collapse
|
32
|
Legrand N, Pradier A, Poulain L, Mouche S, Birsen R, Larrue C, Simonetta F, Tamburini J. AMP-Activated Protein Kinase Contributes to Apoptosis Induced by the Bcl-2 Inhibitor Venetoclax in Acute Myeloid Leukemia. Cancers (Basel) 2021; 13:cancers13235966. [PMID: 34885077 PMCID: PMC8656606 DOI: 10.3390/cancers13235966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary The Bcl2 inhibitor venetoclax is a breakthrough therapy in acute myeloid leukemia (AML). We show that venetoclax induces caspase-dependent degradation of AMPK, a central regulator of cellular energy metabolism, with implications in the anti-Leukemic activity of venetoclax in AML. Abstract The treatment of acute myeloid leukemia (AML) remains a challenge especially among the elderly. The Bcl-2 inhibitor venetoclax recently showed significant survival benefits in AML patients when combined to low-dose cytarabine or azacitidine. Bcl-2 inhibition initiate mitochondrial apoptosis, but also respiration and cellular ATP production in AML. AMP-Activated Protein Kinase (AMPK) is a central energy sensor activated by increased AMP:ATP ratio to restore the cellular energy balance. Unexpectedly, we observed that venetoclax inhibited AMPK activity through caspase-dependent degradation of AMPK subunits in AML cells. On the other hand, genetic models of AMPK invalidation and re-expression suggested that AMPK participated to the early stages of apoptotic response through a negative regulation of multi-domain anti-apoptotic effectors such as Mcl-1 or Bcl-xL. Together our results suggested a new link between AMPK and Bcl-2-dependent mitochondrial apoptosis that participated to the anti-leukemic activity of venetoclax in AML.
Collapse
Affiliation(s)
- Noémie Legrand
- Translational Research Centre in Onco-Hematology, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; (N.L.); (A.P.); (L.P.); (S.M.); (R.B.); (C.L.); (F.S.)
- Geneva University Medical Center, 1205 Geneva, Switzerland
| | - Amandine Pradier
- Translational Research Centre in Onco-Hematology, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; (N.L.); (A.P.); (L.P.); (S.M.); (R.B.); (C.L.); (F.S.)
- Geneva University Medical Center, 1205 Geneva, Switzerland
| | - Laury Poulain
- Translational Research Centre in Onco-Hematology, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; (N.L.); (A.P.); (L.P.); (S.M.); (R.B.); (C.L.); (F.S.)
- Geneva University Medical Center, 1205 Geneva, Switzerland
| | - Sarah Mouche
- Translational Research Centre in Onco-Hematology, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; (N.L.); (A.P.); (L.P.); (S.M.); (R.B.); (C.L.); (F.S.)
- Geneva University Medical Center, 1205 Geneva, Switzerland
| | - Rudy Birsen
- Translational Research Centre in Onco-Hematology, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; (N.L.); (A.P.); (L.P.); (S.M.); (R.B.); (C.L.); (F.S.)
- Geneva University Medical Center, 1205 Geneva, Switzerland
- Institut Cochin, Université de Paris, INSERM U1016, F-75014 Paris, France
| | - Clément Larrue
- Translational Research Centre in Onco-Hematology, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; (N.L.); (A.P.); (L.P.); (S.M.); (R.B.); (C.L.); (F.S.)
- Geneva University Medical Center, 1205 Geneva, Switzerland
| | - Federico Simonetta
- Translational Research Centre in Onco-Hematology, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; (N.L.); (A.P.); (L.P.); (S.M.); (R.B.); (C.L.); (F.S.)
- Geneva University Medical Center, 1205 Geneva, Switzerland
| | - Jerome Tamburini
- Translational Research Centre in Onco-Hematology, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; (N.L.); (A.P.); (L.P.); (S.M.); (R.B.); (C.L.); (F.S.)
- Geneva University Medical Center, 1205 Geneva, Switzerland
- Institut Cochin, Université de Paris, INSERM U1016, F-75014 Paris, France
- Correspondence:
| |
Collapse
|
33
|
Bock FJ, Sedov E, Koren E, Koessinger AL, Cloix C, Zerbst D, Athineos D, Anand J, Campbell KJ, Blyth K, Fuchs Y, Tait SWG. Apoptotic stress-induced FGF signalling promotes non-cell autonomous resistance to cell death. Nat Commun 2021; 12:6572. [PMID: 34772930 PMCID: PMC8590049 DOI: 10.1038/s41467-021-26613-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/08/2021] [Indexed: 11/13/2022] Open
Abstract
Damaged or superfluous cells are typically eliminated by apoptosis. Although apoptosis is a cell-autonomous process, apoptotic cells communicate with their environment in different ways. Here we describe a mechanism whereby cells under apoptotic stress can promote survival of neighbouring cells. We find that upon apoptotic stress, cells release the growth factor FGF2, leading to MEK-ERK-dependent transcriptional upregulation of pro-survival BCL-2 proteins in a non-cell autonomous manner. This transient upregulation of pro-survival BCL-2 proteins protects neighbouring cells from apoptosis. Accordingly, we find in certain cancer types a correlation between FGF-signalling, BCL-2 expression and worse prognosis. In vivo, upregulation of MCL-1 occurs in an FGF-dependent manner during skin repair, which regulates healing dynamics. Importantly, either co-treatment with FGF-receptor inhibitors or removal of apoptotic stress restores apoptotic sensitivity to cytotoxic therapy and delays wound healing. These data reveal a pathway by which cells under apoptotic stress can increase resistance to cell death in surrounding cells. Beyond mediating cytotoxic drug resistance, this process also provides a potential link between tissue damage and repair.
Collapse
Affiliation(s)
- Florian J Bock
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK.
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK.
- Department of Radiotherapy (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University, 6229 ER, Maastricht, The Netherlands.
| | - Egor Sedov
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Elle Koren
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Anna L Koessinger
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - Catherine Cloix
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - Désirée Zerbst
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - Dimitris Athineos
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Jayanthi Anand
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Kirsteen J Campbell
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - Karen Blyth
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - Yaron Fuchs
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Stephen W G Tait
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK.
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK.
| |
Collapse
|
34
|
Shimizu K, Gi M, Suzuki S, North BJ, Watahiki A, Fukumoto S, Asara JM, Tokunaga F, Wei W, Inuzuka H. Interplay between protein acetylation and ubiquitination controls MCL1 protein stability. Cell Rep 2021; 37:109988. [PMID: 34758305 PMCID: PMC8621139 DOI: 10.1016/j.celrep.2021.109988] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 06/02/2021] [Accepted: 10/21/2021] [Indexed: 01/29/2023] Open
Abstract
The anti-apoptotic myeloid cell leukemia 1 (MCL1) protein belongs to the pro-survival BCL2 family and is frequently amplified or elevated in human cancers. MCL1 is highly unstable, with its stability being regulated by phosphorylation and ubiquitination. Here, we identify acetylation as another critical post-translational modification regulating MCL1 protein stability. We demonstrate that the lysine acetyltransferase p300 targets MCL1 at K40 for acetylation, which is counteracted by the deacetylase sirtuin 3 (SIRT3). Mechanistically, acetylation enhances MCL1 interaction with USP9X, resulting in deubiquitination and subsequent MCL1 stabilization. Therefore, ectopic expression of acetylation-mimetic MCL1 promotes apoptosis evasion of cancer cells, enhances colony formation potential, and facilitates xenografted tumor progression. We further demonstrate that elevated MCL1 acetylation sensitizes multiple cancer cells to pharmacological inhibition of USP9X. These findings reveal that acetylation of MCL1 is a critical post-translational modification enhancing its oncogenic function and provide a rationale for developing innovative therapeutic strategies for MCL1-dependent tumors. MCL1, an anti-apoptotic BCL2 family protein, is frequently overexpressed in a variety of cancers, and its oncogenic function is finely regulated by post-translational modifications such as phosphorylation and ubiquitination. Shimizu et al. dissect the molecular mechanism of acetylation-mediated MCL1 stability control, providing insights into potential therapeutic intervention targeting the MCL1 protein.
Collapse
Affiliation(s)
- Kouhei Shimizu
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Center for Advanced Stem Cell and Regenerative Research, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan; Department of Pathobiochemistry, Graduate School of Medicine, Osaka City University, Osaka 545-8585, Japan.
| | - Min Gi
- Department of Molecular Pathology, Graduate School of Medicine, Osaka City University, Osaka 545-8585, Japan; Department of Environmental Risk Assessment, Graduate School of Medicine, Osaka City University, Osaka 545-8585, Japan
| | - Shugo Suzuki
- Department of Molecular Pathology, Graduate School of Medicine, Osaka City University, Osaka 545-8585, Japan
| | - Brian J North
- Department of Biomedical Sciences, Creighton University, Omaha, NE 68178, USA
| | - Asami Watahiki
- Center for Advanced Stem Cell and Regenerative Research, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan
| | - Satoshi Fukumoto
- Center for Advanced Stem Cell and Regenerative Research, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan; Division of Pediatric Dentistry, Department of Oral Health and Development Sciences, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan; Section of Pediatric Dentistry, Division of Oral Health, Growth and Development, Kyushu University Faculty of Dental Science, Fukuoka 812-8582, Japan
| | - John M Asara
- Division of Signal Transduction, Beth Israel Deaconess Medical Center and Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Fuminori Tokunaga
- Department of Pathobiochemistry, Graduate School of Medicine, Osaka City University, Osaka 545-8585, Japan
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Hiroyuki Inuzuka
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
35
|
Jeong M, Jeong MH, Kim JE, Cho S, Lee KJ, Park S, Sohn J, Park YG. TCTP protein degradation by targeting mTORC1 and signaling through S6K, Akt, and Plk1 sensitizes lung cancer cells to DNA-damaging drugs. Sci Rep 2021; 11:20812. [PMID: 34675258 PMCID: PMC8531033 DOI: 10.1038/s41598-021-00247-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 10/08/2021] [Indexed: 11/14/2022] Open
Abstract
Translationally controlled tumor protein (TCTP) is expressed in many tissues, particularly in human tumors. It plays a role in malignant transformation, apoptosis prevention, and DNA damage repair. The signaling mechanisms underlying TCTP regulation in cancer are only partially understood. Here, we investigated the role of mTORC1 in regulating TCTP protein levels, thereby modulating chemosensitivity, in human lung cancer cells and an A549 lung cancer xenograft model. The inhibition of mTORC1, but not mTORC2, induced ubiquitin/proteasome-dependent TCTP degradation without a decrease in the mRNA level. PLK1 activity was required for TCTP ubiquitination and degradation and for its phosphorylation at Ser46 upon mTORC1 inhibition. Akt phosphorylation and activation was indispensable for rapamycin-induced TCTP degradation and PLK1 activation, and depended on S6K inhibition, but not mTORC2 activation. Furthermore, the minimal dose of rapamycin required to induce TCTP proteolysis enhanced the efficacy of DNA-damaging drugs, such as cisplatin and doxorubicin, through the induction of apoptotic cell death in vitro and in vivo. This synergistic cytotoxicity of these drugs was induced irrespective of the functional status of p53. These results demonstrate a new mechanism of TCTP regulation in which the mTORC1/S6K pathway inhibits a novel Akt/PLK1 signaling axis and thereby induces TCTP protein stabilization and confers resistance to DNA-damaging agents. The results of this study suggest a new therapeutic strategy for enhancing chemosensitivity in lung cancers regardless of the functional status of p53.
Collapse
Affiliation(s)
- Mini Jeong
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, 73 Koryodae-ro, Sungbuk-gu, Seoul, 02841, Republic of Korea
- Korean Institute of Molecular Medicine and Nutrition, Korea University College of Medicine, Seoul, Republic of Korea
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, ASAN Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Mi Hyeon Jeong
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, 73 Koryodae-ro, Sungbuk-gu, Seoul, 02841, Republic of Korea
- Korean Institute of Molecular Medicine and Nutrition, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jung Eun Kim
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, 73 Koryodae-ro, Sungbuk-gu, Seoul, 02841, Republic of Korea
- Korean Institute of Molecular Medicine and Nutrition, Korea University College of Medicine, Seoul, Republic of Korea
| | - Serin Cho
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, 73 Koryodae-ro, Sungbuk-gu, Seoul, 02841, Republic of Korea
- Korean Institute of Molecular Medicine and Nutrition, Korea University College of Medicine, Seoul, Republic of Korea
| | - Kyoung Jin Lee
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, 73 Koryodae-ro, Sungbuk-gu, Seoul, 02841, Republic of Korea
- Korean Institute of Molecular Medicine and Nutrition, Korea University College of Medicine, Seoul, Republic of Korea
| | - Serkin Park
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, 73 Koryodae-ro, Sungbuk-gu, Seoul, 02841, Republic of Korea
- Korean Institute of Molecular Medicine and Nutrition, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jeongwon Sohn
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, 73 Koryodae-ro, Sungbuk-gu, Seoul, 02841, Republic of Korea
- Korean Institute of Molecular Medicine and Nutrition, Korea University College of Medicine, Seoul, Republic of Korea
| | - Yun Gyu Park
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, 73 Koryodae-ro, Sungbuk-gu, Seoul, 02841, Republic of Korea.
- Korean Institute of Molecular Medicine and Nutrition, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
36
|
Sénéchal P, Robert F, Cencic R, Yanagiya A, Chu J, Sonenberg N, Paquet M, Pelletier J. Assessing eukaryotic initiation factor 4F subunit essentiality by CRISPR-induced gene ablation in the mouse. Cell Mol Life Sci 2021; 78:6709-6719. [PMID: 34559254 PMCID: PMC11073133 DOI: 10.1007/s00018-021-03940-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/31/2021] [Accepted: 09/10/2021] [Indexed: 01/16/2023]
Abstract
Eukaryotic initiation factor (eIF) 4F plays a central role in the ribosome recruitment phase of cap-dependent translation. This heterotrimeric complex consists of a cap binding subunit (eIF4E), a DEAD-box RNA helicase (eIF4A), and a large bridging protein (eIF4G). In mammalian cells, there are two genes encoding eIF4A (eIF4A1 and eIF4A2) and eIF4G (eIF4G1 and eIF4G3) paralogs that can assemble into eIF4F complexes. To query the essential nature of the eIF4F subunits in normal development, we used CRISPR/Cas9 to generate mouse strains with targeted ablation of each gene encoding the different eIF4F subunits. We find that Eif4e, Eif4g1, and Eif4a1 are essential for viability in the mouse, whereas Eif4g3 and Eif4a2 are not. However, Eif4g3 and Eif4a2 do play essential roles in spermatogenesis. Crossing of these strains to the lymphoma-prone Eμ-Myc mouse model revealed that heterozygosity at the Eif4e or Eif4a1 loci significantly delayed tumor onset. Lastly, tumors derived from Eif4e∆38 fs/+/Eμ-Myc or Eif4a1∆5 fs/+/Eμ-Myc mice show increased sensitivity to the chemotherapeutic agent doxorubicin, in vivo. Our study reveals that eIF4A2 and eIF4G3 play non-essential roles in gene expression regulation during embryogenesis; whereas reductions in eIF4E or eIF4A1 levels are protective against tumor development in a murine Myc-driven lymphoma setting.
Collapse
Affiliation(s)
- Patrick Sénéchal
- Department of Biochemistry, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Francis Robert
- Department of Biochemistry, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Regina Cencic
- Department of Biochemistry, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Akiko Yanagiya
- Department of Biochemistry, McGill University, Montreal, QC, H3G 1Y6, Canada
- Cell Signal Unit, Okinawa Institute of Science and Technology, Okinawa, 904-0495, Japan
| | - Jennifer Chu
- Department of Biochemistry, McGill University, Montreal, QC, H3G 1Y6, Canada
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02138, USA
| | - Nahum Sonenberg
- Department of Biochemistry, McGill University, Montreal, QC, H3G 1Y6, Canada
- Rosalind and Morris Goodman Cancer Research Center, McGill University, Montreal, QC, H3A 1A3, Canada
| | - Marilène Paquet
- Département de Pathologie et Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Jerry Pelletier
- Department of Biochemistry, McGill University, Montreal, QC, H3G 1Y6, Canada.
- Department of Oncology, McGill University, Montreal, QC, H3A 1G5, Canada.
- Rosalind and Morris Goodman Cancer Research Center, McGill University, Montreal, QC, H3A 1A3, Canada.
| |
Collapse
|
37
|
Chin HS, Fu NY. Physiological Functions of Mcl-1: Insights From Genetic Mouse Models. Front Cell Dev Biol 2021; 9:704547. [PMID: 34336857 PMCID: PMC8322662 DOI: 10.3389/fcell.2021.704547] [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: 05/05/2021] [Accepted: 06/14/2021] [Indexed: 01/27/2023] Open
Abstract
The ability to regulate the survival and death of a cell is paramount throughout the lifespan of a multicellular organism. Apoptosis, a main physiological form of programmed cell death, is regulated by the Bcl-2 family proteins that are either pro-apoptotic or pro-survival. The in vivo functions of distinct Bcl-2 family members are largely unmasked by genetically engineered murine models. Mcl-1 is one of the two Bcl-2 like pro-survival genes whose germline deletion causes embryonic lethality in mice. Its requisite for the survival of a broad range of cell types has been further unraveled by using conditional and inducible deletion murine model systems in different tissues or cell lineages and at distinct developmental stages. Moreover, genetic mouse cancer models have also demonstrated that Mcl-1 is essential for the survival of multiple tumor types. The MCL-1 locus is commonly amplified across various cancer types in humans. Small molecule inhibitors with high affinity and specificity to human MCL-1 have been developed and explored for the treatment of certain cancers. To facilitate the pre-clinical studies of MCL-1 in cancer and other diseases, transgenic mouse models over-expressing human MCL-1 as well as humanized MCL-1 mouse models have been recently engineered. This review discusses the current advances in understanding the physiological roles of Mcl-1 based on studies using genetic murine models and its critical implications in pathology and treatment of human diseases.
Collapse
Affiliation(s)
- Hui San Chin
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Nai Yang Fu
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore.,Department of Physiology, National University of Singapore, Singapore, Singapore
| |
Collapse
|
38
|
Zhang L, Liu G, Kong M, Li T, Wu D, Zhou X, Yang C, Xia L, Yang Z, Chen L. Revealing dynamic regulations and the related key proteins of myeloma-initiating cells by integrating experimental data into a systems biological model. Bioinformatics 2021; 37:1554-1561. [PMID: 31350562 DOI: 10.1093/bioinformatics/btz542] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 06/17/2019] [Accepted: 07/19/2019] [Indexed: 12/24/2022] Open
Abstract
MOTIVATION The growth and survival of myeloma cells are greatly affected by their surrounding microenvironment. To understand the molecular mechanism and the impact of stiffness on the fate of myeloma-initiating cells (MICs), we develop a systems biological model to reveal the dynamic regulations by integrating reverse-phase protein array data and the stiffness-associated pathway. RESULTS We not only develop a stiffness-associated signaling pathway to describe the dynamic regulations of the MICs, but also clearly identify three critical proteins governing the MIC proliferation and death, including FAK, mTORC1 and NFκB, which are validated to be related with multiple myeloma by our immunohistochemistry experiment, computation and manually reviewed evidences. Moreover, we demonstrate that the systematic model performs better than widely used parameter estimation algorithms for the complicated signaling pathway. AVAILABILITY AND IMPLEMENTATION We can not only use the systems biological model to infer the stiffness-associated genetic signaling pathway and locate the critical proteins, but also investigate the important pathways, proteins or genes for other type of the cancer. Thus, it holds universal scientific significance. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Le Zhang
- College of Computer Science.,Medical Big Data Center, Sichuan University, Chengdu 610065, China.,Chongqqing Zhongdi Medical Information Technology Co., Ltd, Chongqing 401320, China
| | - Guangdi Liu
- College of Computer and Information Science, Southwest University, Chongqing 400715, China.,Library of Chengdu University, Chengdu University, Chengdu 610106, China
| | - Meijing Kong
- College of Computer and Information Science, Southwest University, Chongqing 400715, China
| | - Tingting Li
- College of Mathematics and Statistics, Southwest University, Chongqing 400715, China
| | - Dan Wu
- Department of Radiology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Xiaobo Zhou
- Department of Radiology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Chuanwei Yang
- Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lei Xia
- Cancer Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Zhenzhou Yang
- Cancer Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Luonan Chen
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China.,Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai 201210, China
| |
Collapse
|
39
|
Domostegui A, Peddigari S, Mercer CA, Iannizzotto F, Rodriguez ML, Garcia-Cajide M, Amador V, Diepstraten ST, Kelly GL, Salazar R, Kozma SC, Kusnadi EP, Kang J, Gentilella A, Pearson RB, Thomas G, Pelletier J. Impaired ribosome biogenesis checkpoint activation induces p53-dependent MCL-1 degradation and MYC-driven lymphoma death. Blood 2021; 137:3351-3364. [PMID: 33512431 PMCID: PMC8212515 DOI: 10.1182/blood.2020007452] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 12/24/2020] [Indexed: 12/11/2022] Open
Abstract
MYC-driven B-cell lymphomas are addicted to increased levels of ribosome biogenesis (RiBi), offering the potential for therapeutic intervention. However, it is unclear whether inhibition of RiBi suppresses lymphomagenesis by decreasing translational capacity and/or by p53 activation mediated by the impaired RiBi checkpoint (IRBC). Here we generated Eμ-Myc lymphoma cells expressing inducible short hairpin RNAs to either ribosomal protein L7a (RPL7a) or RPL11, the latter an essential component of the IRBC. The loss of either protein reduced RiBi, protein synthesis, and cell proliferation to similar extents. However, only RPL7a depletion induced p53-mediated apoptosis through the selective proteasomal degradation of antiapoptotic MCL-1, indicating the critical role of the IRBC in this mechanism. Strikingly, low concentrations of the US Food and Drug Administration-approved anticancer RNA polymerase I inhibitor Actinomycin D (ActD) dramatically prolonged the survival of mice harboring Trp53+/+;Eμ-Myc but not Trp53-/-;Eμ-Myc lymphomas, which provides a rationale for treating MYC-driven B-cell lymphomas with ActD. Importantly, the molecular effects of ActD on Eμ-Myc cells were recapitulated in human B-cell lymphoma cell lines, highlighting the potential for ActD as a therapeutic avenue for p53 wild-type lymphoma.
Collapse
Affiliation(s)
- Ana Domostegui
- Laboratory of Cancer Metabolism, Molecular Mechanisms and Experimental Therapy in Oncology Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Suresh Peddigari
- Division of Hematology Oncology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH
| | - Carol A Mercer
- Division of Hematology Oncology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH
| | - Flavia Iannizzotto
- Laboratory of Cancer Metabolism, Molecular Mechanisms and Experimental Therapy in Oncology Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Marta L Rodriguez
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Marta Garcia-Cajide
- Laboratory of Cancer Metabolism, Molecular Mechanisms and Experimental Therapy in Oncology Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Virginia Amador
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Sarah T Diepstraten
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Gemma L Kelly
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Ramón Salazar
- Catalan Institute of Oncology, Molecular Mechanisms and Experimental Therapy in Oncology Program, IDIBELL, Barcelona, Spain
| | - Sara C Kozma
- Laboratory of Cancer Metabolism, Molecular Mechanisms and Experimental Therapy in Oncology Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Eric P Kusnadi
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Jian Kang
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Antonio Gentilella
- Laboratory of Cancer Metabolism, Molecular Mechanisms and Experimental Therapy in Oncology Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
- Department of Biochemistry and Physiology, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain
| | - Richard B Pearson
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia; and
| | - George Thomas
- Laboratory of Cancer Metabolism, Molecular Mechanisms and Experimental Therapy in Oncology Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
- Department of Physiological Sciences, Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain
| | - Joffrey Pelletier
- Laboratory of Cancer Metabolism, Molecular Mechanisms and Experimental Therapy in Oncology Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| |
Collapse
|
40
|
Lin YH, Wang H, Fiore A, Förster M, Tung LT, Belle JI, Robert F, Pelletier J, Langlais D, Nijnik A. Loss of MYSM1 inhibits the oncogenic activity of cMYC in B cell lymphoma. J Cell Mol Med 2021; 25:7089-7094. [PMID: 34114734 PMCID: PMC8278115 DOI: 10.1111/jcmm.16554] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 03/15/2021] [Accepted: 03/26/2021] [Indexed: 12/18/2022] Open
Abstract
MYSM1 is a chromatin‐binding protein, widely investigated for its functions in haematopoiesis in human and mouse; however, its role in haematologic malignancies remains unexplored. Here, we investigate the cross‐talk between MYSM1 and oncogenic cMYC in the transcriptional regulation of genes encoding ribosomal proteins, and the implications of these mechanisms for cMYC‐driven carcinogenesis. We demonstrate that in cMYC‐driven B cell lymphoma in mouse models, MYSM1‐loss represses ribosomal protein gene expression and protein synthesis. Importantly, the loss of MYSM1 also strongly inhibits cMYC oncogenic activity and protects against B cell lymphoma onset and progression in the mouse models. This advances the understanding of the molecular and transcriptional mechanisms of lymphomagenesis, and suggests MYSM1 as a possible drug target for cMYC‐driven malignancies.
Collapse
Affiliation(s)
- Yun Hsiao Lin
- Department of Physiology, McGill University, Montreal, QC, Canada.,McGill University Research Centre on Complex Traits, McGill University, QC, Canada
| | - HanChen Wang
- Department of Physiology, McGill University, Montreal, QC, Canada.,McGill University Research Centre on Complex Traits, McGill University, QC, Canada.,Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Amanda Fiore
- Department of Physiology, McGill University, Montreal, QC, Canada.,McGill University Research Centre on Complex Traits, McGill University, QC, Canada
| | - Michael Förster
- Department of Physiology, McGill University, Montreal, QC, Canada.,McGill University Research Centre on Complex Traits, McGill University, QC, Canada
| | - Lin Tze Tung
- Department of Physiology, McGill University, Montreal, QC, Canada.,McGill University Research Centre on Complex Traits, McGill University, QC, Canada
| | - Jad I Belle
- Department of Physiology, McGill University, Montreal, QC, Canada.,McGill University Research Centre on Complex Traits, McGill University, QC, Canada
| | - Francis Robert
- Department of Biochemistry, McGill University, Montreal, QC, Canada.,The Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada
| | - Jerry Pelletier
- Department of Biochemistry, McGill University, Montreal, QC, Canada.,The Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada
| | - David Langlais
- McGill University Research Centre on Complex Traits, McGill University, QC, Canada.,Department of Human Genetics, McGill University, Montreal, QC, Canada.,McGill University Genome Centre, McGill University, Montreal, QC, Canada.,Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Anastasia Nijnik
- Department of Physiology, McGill University, Montreal, QC, Canada.,McGill University Research Centre on Complex Traits, McGill University, QC, Canada
| |
Collapse
|
41
|
Jung HR, Oh Y, Na D, Min S, Kang J, Jang D, Shin S, Kim J, Lee SE, Jeong EM, An JY, Sung CO, Lee WS, Lee C, Cho SY. CRISPR screens identify a novel combination treatment targeting BCL-X L and WNT signaling for KRAS/BRAF-mutated colorectal cancers. Oncogene 2021; 40:3287-3302. [PMID: 33846570 DOI: 10.1038/s41388-021-01777-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 03/12/2021] [Accepted: 03/29/2021] [Indexed: 02/01/2023]
Abstract
Metastatic or recurrent colorectal cancer (CRC) patients require systemic chemotherapy, but the therapeutic options of targeted agents remain limited. CRC patients with KRAS or BRAF gene mutations exhibit a worse prognosis and are resistant to anti-EGFR treatment. Previous studies have shown that the expression of anti-apoptotic protein BCL-XL is increased in CRC patients with KRAS/BRAF mutations, suggesting BCL-XL as a therapeutic target for this subgroup. Here, we performed genome-wide CRISPR/Cas9 screens of cell lines with KRAS mutations to investigate the factors required for sensitivity to BCL-XL inhibitor ABT-263 using single-guide RNAs (sgRNAs) that induce loss-of-function mutations. In the presence of ABT-263, sgRNAs targeting negative regulators of WNT signaling (resulting in WNT activation) were enriched, whereas sgRNAs targeting positive regulators of WNT signaling (resulting in WNT inhibition) were depleted in ABT-263-resistant cells. The activation of WNT signaling was highly associated with an increased expression ratio of anti- to pro-apoptotic BCL-2 family genes in CRC samples. Genetic and pharmacologic inhibition of WNT signaling using β-catenin short hairpin RNA or TNIK inhibitor NCB-0846, respectively, augmented ABT-263-induced cell death in KRAS/BRAF-mutated cells. Inhibition of WNT signaling resulted in transcriptional repression of the anti-apoptotic BCL-2 family member, MCL1, via the functional inhibition of the β-catenin-containing complex at the MCL1 promoter. In addition, the combination of ABT-263 and NCB-0846 exhibited synergistic effects in in vivo patient-derived xenograft (PDX) models with KRAS mutations. Our data provide a novel targeted combination treatment strategy for the CRC patient subgroup with KRAS or BRAF mutations.
Collapse
Affiliation(s)
- Hae Rim Jung
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea
- Medical Research Center, Genomic Medicine Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Yumi Oh
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea
- Medical Research Center, Genomic Medicine Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Deukchae Na
- Ewha Institute of Convergence Medicine, Ewha Womans University Mokdong Hospital, Seoul, Korea
| | - Seoyeon Min
- Department of Life Science, Ewha Womans University, Seoul, Korea
| | - Jinjoo Kang
- Department of Life Science, Ewha Womans University, Seoul, Korea
| | - Dongjun Jang
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Seungjae Shin
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Jiwon Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Sang Eun Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Eui Man Jeong
- Department of Pharmacy, College of Pharmacy, Jeju National University, Jeju Special Self-Governing Province, Korea
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju Special Self-Governing Province, Korea
- Bio-Health Materials Core-Facility Center, Jeju National University, Jeju Special Self-Governing Province, Korea
- Practical Translational Research Center, Jeju National University, Jeju Special Self-Governing Province, Korea
| | - Joon Yong An
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul, Korea
| | - Chang Ohk Sung
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Won-Suk Lee
- Department of Surgery, Gil Medical Center, Gachon University, Incheon, Korea
| | - Charles Lee
- Department of Life Science, Ewha Womans University, Seoul, Korea
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Sung-Yup Cho
- Medical Research Center, Genomic Medicine Institute, Seoul National University College of Medicine, Seoul, Korea.
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea.
- Cancer Research Institute, Seoul National University, Seoul, Korea.
| |
Collapse
|
42
|
The self-renewal dental pulp stem cell microtissues challenged by a toxic dental monomer. Biosci Rep 2021; 40:225156. [PMID: 32495822 PMCID: PMC7303350 DOI: 10.1042/bsr20200210] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 05/12/2020] [Accepted: 05/19/2020] [Indexed: 01/19/2023] Open
Abstract
Dental pulp stem cells (DPSCs) regenerate injured/diseased pulp tissue and deposit tertiary dentin. DPSCs stress response can be activated by exposing cells to the monomer triethyleneglycol dimethacrylate (TEGDMA) and inducing the DNA-damage inducible transcript 4 (DDIT4) protein expression. The goal of the present study was to determine the impact of TEGDMA on the ability of DPSCs to maintain their self-renewal capabilities, develop and preserve their 3D structures and deposit the mineral. Human primary and immortalized DPSCs were cultured in extracellular matrix/basement membrane (ECM/BM) to support stemness and to create multicellular interacting layers (microtissues). The microtissues were exposed to the toxic concentrations of TEGDMA (0.5 and 1.5 mmol/l). The DPSCs spatial architecture was assessed by confocal microscopy. Mineral deposition was detected by alizarin red staining and visualized by stereoscopy. Cellular self-renewal transcription factor SOX2 was determined by immunocytochemistry. The microtissue thicknesses/vertical growth, surface area of the mineralizing microtissues, the percentage of area covered by the deposited mineral, and the fluorescence intensity of the immunostained cells were quantified ImageJ. DDIT4 expression was determined by a single molecule RNA-FISH technique and the cell phenotype was determined morphologically. DDIT4 expression was correlated with the cytotoxic phenotype. TEGDMA affected the structures of developing and mature microtissues. It inhibited the deposition of the mineral in the matrix while not affecting the SOX2 expression. Our data demonstrate that DPSCs retained their self-renewal capacity although their other functions were impeded. Since the DPSCs pool remained preserved, properties effected by the irritant should be restored by a proper rescue therapy.
Collapse
|
43
|
Saoudaoui S, Bernard M, Cardin GB, Malaquin N, Christopoulos A, Rodier F. mTOR as a senescence manipulation target: A forked road. Adv Cancer Res 2021; 150:335-363. [PMID: 33858600 DOI: 10.1016/bs.acr.2021.02.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Cellular senescence, cancer and aging are highly interconnected. Among many important molecular machines that lie at the intersection of this triad, the mechanistic (formerly mammalian) target of rapamycin (mTOR) is a central regulator of cell metabolism, proliferation, and survival. The mTOR signaling cascade is essential to maintain cellular homeostasis in normal biological processes or in response to stress, and its dysregulation is implicated in the progression of many disorders, including age-associated diseases. Accordingly, the pharmacological implications of mTOR inhibition using rapamycin or others rapalogs span the treatment of various human diseases from immune disorders to cancer. Importantly, rapamycin is one of the only known pan-species drugs that can extend lifespan. The molecular and cellular mechanisms explaining the phenotypic consequences of mTOR are vast and heavily studied. In this review, we will focus on the potential role of mTOR in the context of cellular senescence, a tumor suppressor mechanism and a pillar of aging. We will explore the link between senescence, autophagy and mTOR and discuss the opportunities to exploit senescence-associated mTOR functions to manipulate senescence phenotypes in age-associated diseases and cancer treatment.
Collapse
Affiliation(s)
- Sarah Saoudaoui
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada; Institut du cancer de Montréal, Montreal, QC, Canada
| | - Monique Bernard
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada; Institut du cancer de Montréal, Montreal, QC, Canada
| | - Guillaume B Cardin
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada; Institut du cancer de Montréal, Montreal, QC, Canada
| | - Nicolas Malaquin
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada; Institut du cancer de Montréal, Montreal, QC, Canada
| | - Apostolos Christopoulos
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada; Institut du cancer de Montréal, Montreal, QC, Canada; Otolaryngology-Head and Neck Surgery Service, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - Francis Rodier
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada; Institut du cancer de Montréal, Montreal, QC, Canada; Université de Montréal, Département de radiologie, radio-oncologie et médicine nucléaire, Montreal, QC, Canada.
| |
Collapse
|
44
|
Herzog LO, Walters B, Buono R, Lee JS, Mallya S, Fung A, Chiu H, Nguyen N, Li B, Pinkerton AB, Jackson MR, Schneider RJ, Ronai ZA, Fruman DA. Targeting eIF4F translation initiation complex with SBI-756 sensitises B lymphoma cells to venetoclax. Br J Cancer 2021; 124:1098-1109. [PMID: 33318657 PMCID: PMC7960756 DOI: 10.1038/s41416-020-01205-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 09/30/2020] [Accepted: 11/20/2020] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND The BCL2 inhibitor venetoclax has shown efficacy in several hematologic malignancies, with the greatest response rates in indolent blood cancers such as chronic lymphocytic leukaemia. There is a lower response rate to venetoclax monotherapy in diffuse large B-cell lymphoma (DLBCL). METHODS We tested inhibitors of cap-dependent mRNA translation for the ability to sensitise DLBCL and mantle cell lymphoma (MCL) cells to apoptosis by venetoclax. We compared the mTOR kinase inhibitor (TOR-KI) MLN0128 with SBI-756, a compound targeting eukaryotic translation initiation factor 4G1 (eIF4G1), a scaffolding protein in the eIF4F complex. RESULTS Treatment of DLBCL and MCL cells with SBI-756 synergised with venetoclax to induce apoptosis in vitro, and enhanced venetoclax efficacy in vivo. SBI-756 prevented eIF4E-eIF4G1 association and cap-dependent translation without affecting mTOR substrate phosphorylation. In TOR-KI-resistant DLBCL cells lacking eIF4E binding protein-1, SBI-756 still sensitised to venetoclax. SBI-756 selectively reduced translation of mRNAs encoding ribosomal proteins and translation factors, leading to a reduction in protein synthesis rates in sensitive cells. When normal lymphocytes were treated with SBI-756, only B cells had reduced viability, and this correlated with reduced protein synthesis. CONCLUSIONS Our data highlight a novel combination for treatment of aggressive lymphomas, and establishes its efficacy and selectivity using preclinical models.
Collapse
Affiliation(s)
- Lee-or Herzog
- grid.266093.80000 0001 0668 7243Department of Molecular Biology & Biochemistry, University of California, Irvine, CA 92697 USA
| | - Beth Walters
- grid.137628.90000 0004 1936 8753New York University School of Medicine, New York, NY USA
| | - Roberta Buono
- grid.266093.80000 0001 0668 7243Department of Molecular Biology & Biochemistry, University of California, Irvine, CA 92697 USA
| | - J. Scott Lee
- grid.266093.80000 0001 0668 7243Department of Molecular Biology & Biochemistry, University of California, Irvine, CA 92697 USA ,grid.418185.10000 0004 0627 6737Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121 USA
| | - Sharmila Mallya
- grid.266093.80000 0001 0668 7243Department of Molecular Biology & Biochemistry, University of California, Irvine, CA 92697 USA
| | - Amos Fung
- grid.266093.80000 0001 0668 7243Department of Molecular Biology & Biochemistry, University of California, Irvine, CA 92697 USA
| | - Honyin Chiu
- grid.266093.80000 0001 0668 7243Department of Molecular Biology & Biochemistry, University of California, Irvine, CA 92697 USA ,grid.416879.50000 0001 2219 0587Benaroya Research Institute, Seattle, WA 98101 USA
| | - Nancy Nguyen
- grid.266093.80000 0001 0668 7243Department of Molecular Biology & Biochemistry, University of California, Irvine, CA 92697 USA
| | - Boyang Li
- grid.266093.80000 0001 0668 7243Department of Molecular Biology & Biochemistry, University of California, Irvine, CA 92697 USA
| | - Anthony B. Pinkerton
- grid.479509.60000 0001 0163 8573Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037 USA
| | - Michael R. Jackson
- grid.479509.60000 0001 0163 8573Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037 USA
| | - Robert J. Schneider
- grid.137628.90000 0004 1936 8753New York University School of Medicine, New York, NY USA
| | - Ze’ev A. Ronai
- grid.479509.60000 0001 0163 8573Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037 USA
| | - David A. Fruman
- grid.266093.80000 0001 0668 7243Department of Molecular Biology & Biochemistry, University of California, Irvine, CA 92697 USA
| |
Collapse
|
45
|
Tang Y, Yang Y, Luo J, Liu S, Zhan Y, Zang H, Zheng H, Zhang Y, Feng J, Fan S, Wen Q. Overexpression of HSP10 correlates with HSP60 and Mcl-1 levels and predicts poor prognosis in non-small cell lung cancer patients. Cancer Biomark 2021; 30:85-94. [PMID: 32986659 PMCID: PMC7990427 DOI: 10.3233/cbm-200410] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND HSP60 and its partner HSP10 are members of heat shock proteins (HSPs) family, which help mitochondrial protein to fold correctly. Mcl-1, a member of the Bcl-2 family, plays a crucial role in regulation of cell apoptosis. Aberrant expression of HSP10, HSP60 and Mcl-1 is involved in the development of many tumors. OBJECTIVE To examine the association between expression of HSP10, HSP60 and Mcl-1 and clinicopathological features of non-small cell lung cancer (NSCLC). METHODS Tissue microarrays including 53 non-cancerous lung tissues (Non-CLT) and 354 surgically resected NSCLC were stained with anti-HSP10, anti-HSP60 and anti-Mcl-1 antibodies respectively by immunohistochemistry. RESULTS Higher expression of HSP10, HSP60 and Mcl-1 was found in NSCLC compared with Non-CLT. Both individual and combined HSP10 and HSP60 expression in patients with clinical stage III was higher than that in stage I ∼ II. Expression of HSP10 showed a positive correlation with HSP60 and Mcl-1. Overall survival time of NSCLC patients was remarkably shorter with elevated expression of HSP10, HSP60 and Mcl-1 alone and in combination. Moreover overexpression of HSP10 and Mcl-1 was poor independent prognostic factor for lung adenocarcinoma patients. CONCLUSIONS High expression of HSP10, HSP60 and Mcl-1 might act as novel biomarker of poor prognosis for NSCLC patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Qiuyuan Wen
- Corresponding author: Qiuyuan Wen, Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China. E-mail:
| |
Collapse
|
46
|
Birkinshaw RW. Challenges in small-molecule target identification: a commentary on "BDA-366, a putative Bcl-2 BH4 domain antagonist, induces apoptosis independently of Bcl-2 in a variety of cancer cell models". Cell Death Differ 2021; 28:1130-1132. [PMID: 33469228 DOI: 10.1038/s41418-020-00717-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/06/2020] [Accepted: 12/16/2020] [Indexed: 01/19/2023] Open
Affiliation(s)
- Richard W Birkinshaw
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Melbourne, VIC, 3052, Australia. .,Department of Medical Biology, University of Melbourne, Melbourne, VIC, 3010, Australia.
| |
Collapse
|
47
|
Oh Y, Jung HR, Min S, Kang J, Jang D, Shin S, Kim J, Lee SE, Sung CO, Lee WS, Lee C, Jeong EM, Cho SY. Targeting antioxidant enzymes enhances the therapeutic efficacy of the BCL-X L inhibitor ABT-263 in KRAS-mutant colorectal cancers. Cancer Lett 2020; 497:123-136. [PMID: 33068701 DOI: 10.1016/j.canlet.2020.10.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/27/2020] [Accepted: 10/12/2020] [Indexed: 12/27/2022]
Abstract
Cancer chemotherapeutic drugs exert cytotoxic effects by modulating intracellular reactive oxygen species (ROS) levels. However, whether ROS modulates the efficacy of targeted therapeutics remains poorly understood. Previously, we reported that upregulation of the anti-apoptotic protein, BCL-XL, by KRAS activating mutations was a potential target for KRAS-mutant colorectal cancer (CRC) treatment. Here, we demonstrated that the BCL-XL targeting agent, ABT-263, increased intracellular ROS levels and targeting antioxidant pathways augmented the therapeutic efficacy of this BH3 mimetic. ABT-263 induced expression of genes associated with ROS response and increased intracellular ROS levels by enhancing mitochondrial superoxide generation. The superoxide dismutase inhibitor, 2-methoxyestradiol (2-ME), exhibited synergism with ABT-263 in KRAS-mutant CRC cell lines. This synergistic effect was attributed to the inhibition of mTOR-dependent translation of the anti-apoptotic MCL-1 protein via caspase 3-mediated cleavage of AKT and S6K. In addition, combination treatment of ABT-263 and 2-ME demonstrated a synergistic effect in in vivo patient-derived xenografts harboring KRAS mutations. Our data suggest a novel role for ROS in BH3 mimetic-based targeted therapy and provide a novel strategy for treatment of CRC patients with KRAS mutations.
Collapse
Affiliation(s)
- Yumi Oh
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea; Medical Research Center, Genomic Medicine Institute (GMI), Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Hae Rim Jung
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea; Medical Research Center, Genomic Medicine Institute (GMI), Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Seoyeon Min
- Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Jinjoo Kang
- Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Dongjun Jang
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Seungjae Shin
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Jiwon Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Sang Eun Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Chang Ohk Sung
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Won-Suk Lee
- Department of Surgery, Gil Medical Center, Gachon University, Incheon, 21565, Republic of Korea
| | - Charles Lee
- Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea; The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Eui Man Jeong
- Department of Pharmacy, College of Pharmacy, Jeju National University, Jeju Special Self-Governing Province, 63243, Republic of Korea; Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju Special Self-Governing Province, 63243, Republic of Korea; Bio-Health Materials Core-Facility Center, Jeju National University, Jeju Special Self-Governing Province, 63243, Republic of Korea; Practical Translational Research Center, Jeju National University, Jeju Special Self-Governing Province, 63243, Republic of Korea.
| | - Sung-Yup Cho
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea; Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea; Medical Research Center, Genomic Medicine Institute (GMI), Seoul National University College of Medicine, Seoul, 03080, Korea.
| |
Collapse
|
48
|
BDA-366, a putative Bcl-2 BH4 domain antagonist, induces apoptosis independently of Bcl-2 in a variety of cancer cell models. Cell Death Dis 2020; 11:769. [PMID: 32943617 PMCID: PMC7498462 DOI: 10.1038/s41419-020-02944-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 08/10/2020] [Accepted: 08/24/2020] [Indexed: 12/14/2022]
Abstract
Several cancer cell types, including chronic lymphocytic leukemia (CLL) and diffuse large B-cell lymphoma (DLBCL) upregulate antiapoptotic Bcl-2 to cope with oncogenic stress. BH3 mimetics targeting Bcl-2's hydrophobic cleft have been developed, including venetoclax as a promising anticancer precision medicine for treating CLL patients. Recently, BDA-366 was identified as a small molecule BH4-domain antagonist that could kill lung cancer and multiple myeloma cells. BDA-366 was proposed to switch Bcl-2 from an antiapoptotic into a proapoptotic protein, thereby activating Bax and inducing apoptosis. Here, we scrutinized the therapeutic potential and mechanism of action of BDA-366 in CLL and DLBCL. Although BDA-366 displayed selective toxicity against both cell types, the BDA-366-induced cell death did not correlate with Bcl-2-protein levels and also occurred in the absence of Bcl-2. Moreover, although BDA-366 provoked Bax activation, it did neither directly activate Bax nor switch Bcl-2 into a Bax-activating protein in in vitro Bax/liposome assays. Instead, in primary CLL cells and DLBCL cell lines, BDA-366 inhibited the activity of the PI3K/AKT pathway, resulted in Bcl-2 dephosphorylation and reduced Mcl-1-protein levels without affecting the levels of Bcl-2 or Bcl-xL. Hence, our work challenges the current view that BDA-366 is a BH4-domain antagonist of Bcl-2 that turns Bcl-2 into a pro-apoptotic protein. Rather, our results indicate that other mechanisms beyond switching Bcl-2 conformation underlie BDA-366's cell-death properties that may implicate Mcl-1 downregulation and/or Bcl-2 dephosphorylation.
Collapse
|
49
|
Ye H, Liu Y, Wu K, Luo H, Cui L. AMPK activation overcomes anti-EGFR antibody resistance induced by KRAS mutation in colorectal cancer. Cell Commun Signal 2020; 18:115. [PMID: 32703218 PMCID: PMC7376720 DOI: 10.1186/s12964-020-00584-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 04/21/2020] [Indexed: 12/13/2022] Open
Abstract
Background Colorectal cancer (CRC) is associated with resistance to anti-epidermal growth factor receptor (EGFR) antibodies (both acquired and intrinsic), owing to the amplification or mutation of the KRAS oncogene. However, the mechanism underlying this resistance is incompletely understood. Methods DLD1 cells with WT (+/−) or KRAS G13D mutant allele were treated with different concentrations of Cetuximab (Cet) or panitumumab (Pab) to study the mechanism underlying the KRAS mutation-induced resistance to anti-EGFR antibodies. The function of AMPK in KRAS mutation-induced resistance to anti-EGFR antibodies in CRC cells, and the regulatory role of Bcl-2 family proteins in DLD1 cells with WT or mutated KRAS upon AMPK activation were investigated. In addition, xenograft tumor models with the nude mouse using DLD1 cells with WT or mutated KRAS were established to examine the effects of AMPK activation on KRAS mutation-mediated anti-EGFR antibody resistance. Results Higher levels of AMPK activity in CRC cells with wild-type KRAS treated with anti-EGFR antibody resulted in apoptosis induction. In contrast, CRC cells with mutated KRAS showed lower AMP-activated protein kinase (AMPK) activity and decreased sensitivity to the inhibitory effect of anti-EGFR antibody. CRC cells with mutated KRAS showed high levels of glycolysis and produced an excessive amount of ATP, which suppressed AMPK activation. The knockdown of AMPK expression in CRC cells with WT KRAS produced similar effects to those observed in cells with mutated KRAS and decreased their sensitivity to cetuximab. On the contrary, the activation of AMPK by metformin (Met) or 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) could overcome the KRAS-induced resistance to the anti-EGFR antibody in vivo and in vitro. The activation of AMPK resulted in the inhibition of myeloid cell leukemia 1 (Mcl-1) translation through the suppression of the mammalian target of rapamycin (mTOR) pathway. Conclusion The results established herein indicate that targeting AMPK is a potentially promising and effective CRC treatment strategy. Video abstract
Collapse
Affiliation(s)
- Hua Ye
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, 524023, Guangdong Province, China. .,Institute of Marine Biomedical Research, Guangdong Medical University, No.2 Wenming East Road, Zhanjiang, 524023, Guangdong Province, China. .,Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, 524023, Guangdong Province, China.
| | - Yi Liu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, 524023, Guangdong Province, China.,Institute of Marine Biomedical Research, Guangdong Medical University, No.2 Wenming East Road, Zhanjiang, 524023, Guangdong Province, China
| | - Kefeng Wu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, 524023, Guangdong Province, China.,Institute of Marine Biomedical Research, Guangdong Medical University, No.2 Wenming East Road, Zhanjiang, 524023, Guangdong Province, China
| | - Hui Luo
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, 524023, Guangdong Province, China.,Institute of Marine Biomedical Research, Guangdong Medical University, No.2 Wenming East Road, Zhanjiang, 524023, Guangdong Province, China
| | - Liao Cui
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, 524023, Guangdong Province, China.,Institute of Marine Biomedical Research, Guangdong Medical University, No.2 Wenming East Road, Zhanjiang, 524023, Guangdong Province, China.,Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, 524023, Guangdong Province, China
| |
Collapse
|
50
|
Inde Z, Forcina GC, Denton K, Dixon SJ. Kinetic Heterogeneity of Cancer Cell Fractional Killing. Cell Rep 2020; 32:107845. [PMID: 32640215 PMCID: PMC7409774 DOI: 10.1016/j.celrep.2020.107845] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/09/2020] [Accepted: 06/11/2020] [Indexed: 01/18/2023] Open
Abstract
Lethal drugs can induce incomplete cell death in a population of cancer cells, a phenomenon referred to as fractional killing. Here, we show that high-throughput population-level time-lapse imaging can be used to quantify fractional killing in response to hundreds of different drug treatments in parallel. We find that stable intermediate levels of fractional killing are uncommon, with many drug treatments resulting in complete or near-complete eradication of all cells, if given enough time. The kinetics of fractional killing over time vary substantially as a function of drug, drug dose, and genetic background. At the molecular level, the antiapoptotic protein MCL1 is an important determinant of the kinetics of fractional killing in response to MAPK pathway inhibitors but not other lethal stimuli. These studies suggest that fractional killing is governed by diverse lethal stimulus-specific mechanisms.
Collapse
Affiliation(s)
- Zintis Inde
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | | | - Kyle Denton
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|