1
|
Wang T, Fu J, Huang Y, Fu C. Mechanism of APC truncation involved in colorectal cancer tumorigenesis (Review). Oncol Lett 2025; 29:2. [PMID: 39526304 PMCID: PMC11544694 DOI: 10.3892/ol.2024.14748] [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: 03/25/2024] [Accepted: 09/26/2024] [Indexed: 11/16/2024] Open
Abstract
Adenomatous polyposis coli (APC) is widely recognized as a heavily mutated gene that suppresses tumor growth in colorectal cancer (CRC). Its mutation is considered to be the primary and early event that occurs in the development of CRC. In addition, APC has a crucial role in inhibiting the canonical Wnt signaling pathway. APC mutations in CRC result in the production of shortened gene products. This impairment of β-catenin destruction complexes causes an accumulation of active β-catenin in the cytoplasm and nucleus. In these compartments, β-catenin can bind with DNA-binding proteins of the transcription factor/lymphoid enhancer-binding factor family, thereby activating the Wnt signaling pathway. Consequently, the balance of numerous cellular processes is disrupted, ultimately driving the formation of tumors. There is a growing body of evidence indicating the prevalent occurrence of APC truncation in the majority of CRC cases. Furthermore, it has been observed that these truncated proteins have a crucial role in the activation of the Wnt signaling pathway and the subsequent loss of tumor inhibitory function. This review aimed to provide an overview of the recent advancements in understanding the mechanism behind APC truncation and its association with the onset and progression of CRC.
Collapse
Affiliation(s)
- Tuya Wang
- Department of Medicine, Hetao College, Bayannur, Inner Mongolia Autonomous Region 015000, P.R. China
| | - Jing Fu
- Department of Medicine, Hetao College, Bayannur, Inner Mongolia Autonomous Region 015000, P.R. China
| | - Ye Huang
- Department of Medicine, Hetao College, Bayannur, Inner Mongolia Autonomous Region 015000, P.R. China
| | - Chun Fu
- Department of Medicine, Hetao College, Bayannur, Inner Mongolia Autonomous Region 015000, P.R. China
| |
Collapse
|
2
|
Huang M, Li Y, Li Y, Liu S. C-Terminal Binding Protein: Regulator between Viral Infection and Tumorigenesis. Viruses 2024; 16:988. [PMID: 38932279 PMCID: PMC11209466 DOI: 10.3390/v16060988] [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: 05/23/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
C-terminal binding protein (CtBP), a transcriptional co-repressor, significantly influences cellular signaling, impacting various biological processes including cell proliferation, differentiation, apoptosis, and immune responses. The CtBP family comprises two highly conserved proteins, CtBP1 and CtBP2, which have been shown to play critical roles in both tumorigenesis and the regulation of viral infections. Elevated CtBP expression is noted in various tumor tissues, promoting tumorigenesis, invasiveness, and metastasis through multiple pathways. Additionally, CtBP's role in viral infections varies, exhibiting differing or even opposing effects depending on the virus. This review synthesizes the advances in CtBP's function research in viral infections and virus-associated tumorigenesis, offering new insights into potential antiviral and anticancer strategies.
Collapse
Affiliation(s)
- Meihui Huang
- Xiangya School of Medicine, Central South University, Changsha 410013, China; (M.H.); (Y.L.); (Y.L.)
| | - Yucong Li
- Xiangya School of Medicine, Central South University, Changsha 410013, China; (M.H.); (Y.L.); (Y.L.)
| | - Yuxiao Li
- Xiangya School of Medicine, Central South University, Changsha 410013, China; (M.H.); (Y.L.); (Y.L.)
| | - Shuiping Liu
- Xiangya School of Medicine, Central South University, Changsha 410013, China; (M.H.); (Y.L.); (Y.L.)
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410013, China
| |
Collapse
|
3
|
Filograna A, De Tito S, Monte ML, Oliva R, Bruzzese F, Roca MS, Zannetti A, Greco A, Spano D, Ayala I, Liberti A, Petraccone L, Dathan N, Catara G, Schembri L, Colanzi A, Budillon A, Beccari AR, Del Vecchio P, Luini A, Corda D, Valente C. Identification and characterization of a new potent inhibitor targeting CtBP1/BARS in melanoma cells. J Exp Clin Cancer Res 2024; 43:137. [PMID: 38711119 PMCID: PMC11071220 DOI: 10.1186/s13046-024-03044-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 04/10/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND The C-terminal-binding protein 1/brefeldin A ADP-ribosylation substrate (CtBP1/BARS) acts both as an oncogenic transcriptional co-repressor and as a fission inducing protein required for membrane trafficking and Golgi complex partitioning during mitosis, hence for mitotic entry. CtBP1/BARS overexpression, in multiple cancers, has pro-tumorigenic functions regulating gene networks associated with "cancer hallmarks" and malignant behavior including: increased cell survival, proliferation, migration/invasion, epithelial-mesenchymal transition (EMT). Structurally, CtBP1/BARS belongs to the hydroxyacid-dehydrogenase family and possesses a NAD(H)-binding Rossmann fold, which, depending on ligands bound, controls the oligomerization of CtBP1/BARS and, in turn, its cellular functions. Here, we proposed to target the CtBP1/BARS Rossmann fold with small molecules as selective inhibitors of mitotic entry and pro-tumoral transcriptional activities. METHODS Structured-based screening of drug databases at different development stages was applied to discover novel ligands targeting the Rossmann fold. Among these identified ligands, N-(3,4-dichlorophenyl)-4-{[(4-nitrophenyl)carbamoyl]amino}benzenesulfonamide, called Comp.11, was selected for further analysis. Fluorescence spectroscopy, isothermal calorimetry, computational modelling and site-directed mutagenesis were employed to define the binding of Comp.11 to the Rossmann fold. Effects of Comp.11 on the oligomerization state, protein partners binding and pro-tumoral activities were evaluated by size-exclusion chromatography, pull-down, membrane transport and mitotic entry assays, Flow cytometry, quantitative real-time PCR, motility/invasion, and colony assays in A375MM and B16F10 melanoma cell lines. Effects of Comp.11 on tumor growth in vivo were analyzed in mouse tumor model. RESULTS We identify Comp.11 as a new, potent and selective inhibitor of CtBP1/BARS (but not CtBP2). Comp.11 directly binds to the CtBP1/BARS Rossmann fold affecting the oligomerization state of the protein (unlike other known CtBPs inhibitors), which, in turn, hinders interactions with relevant partners, resulting in the inhibition of both CtBP1/BARS cellular functions: i) membrane fission, with block of mitotic entry and cellular secretion; and ii) transcriptional pro-tumoral effects with significantly hampered proliferation, EMT, migration/invasion, and colony-forming capabilities. The combination of these effects impairs melanoma tumor growth in mouse models. CONCLUSIONS: This study identifies a potent and selective inhibitor of CtBP1/BARS active in cellular and melanoma animal models revealing new opportunities to study the role of CtBP1/BARS in tumor biology and to develop novel melanoma treatments.
Collapse
Affiliation(s)
- Angela Filograna
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Stefano De Tito
- Molecular Cell Biology of Autophagy, The Francis Crick Institute, London, UK. The Study Has Been Previously Performed at IEOS-CNR, Naples, Italy
| | - Matteo Lo Monte
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Rosario Oliva
- Department of Chemical Sciences, University of Naples Federico II, 80126, Naples, Italy
| | - Francesca Bruzzese
- Animal Facility Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Maria Serena Roca
- Experimental Pharmacology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, 80131, Italy
| | - Antonella Zannetti
- Institute of Biostructures and Bioimaging (IBB), National Research Council (CNR), Naples, 80145, Italy
| | - Adelaide Greco
- Interdepartmental Service Center of Veterinary Radiology, University of Naples Federico II, 80137, Naples, Italy
| | - Daniela Spano
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Inmaculada Ayala
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Assunta Liberti
- National Research Council (CNR), Piazzale Aldo Moro, 700185, Rome, Italy
- Biology and Evolution of Marine Organisms (BEOM), Stazione Zoologica Anton Dohrn, Naples, Italy
| | - Luigi Petraccone
- Department of Chemical Sciences, University of Naples Federico II, 80126, Naples, Italy
| | - Nina Dathan
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Giuliana Catara
- Institute of Biochemistry and Cell Biology, National Research Council (CNR), 80131, Naples, Italy
| | - Laura Schembri
- National Research Council (CNR), Piazzale Aldo Moro, 700185, Rome, Italy
- Department of Pharmacy, University of Naples Federico II, 80131, Naples, Italy
| | - Antonino Colanzi
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Alfredo Budillon
- Scientific Directorate, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | | | - Pompea Del Vecchio
- Department of Chemical Sciences, University of Naples Federico II, 80126, Naples, Italy
| | - Alberto Luini
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Daniela Corda
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy.
| | - Carmen Valente
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy.
- Present address: Dompé Farmaceutici S.P.A, L'Aquila, Italy.
| |
Collapse
|
4
|
Dcona MM, Chougoni KK, Dcona DT, West JL, Singh SJ, Ellis KC, Grossman SR. Combined Targeting of NAD Biosynthesis and the NAD-dependent Transcription Factor C-terminal Binding Protein as a Promising Novel Therapy for Pancreatic Cancer. CANCER RESEARCH COMMUNICATIONS 2023; 3:2003-2013. [PMID: 37707363 PMCID: PMC10549224 DOI: 10.1158/2767-9764.crc-22-0521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 08/07/2023] [Accepted: 09/06/2023] [Indexed: 09/15/2023]
Abstract
Cancer therapies targeting metabolic derangements unique to cancer cells are emerging as a key strategy to address refractory solid tumors such as pancreatic ductal adenocarcinomas (PDAC) that exhibit resistance to extreme nutrient deprivation in the tumor microenvironment. Nicotinamide adenine dinucleotide (NAD) participates in multiple metabolic pathways and nicotinamide phosphoribosyl transferase (NAMPT) is one of the key intracellular enzymes that facilitate the synthesis of NAD. C-terminal binding proteins 1 and 2 (CtBP) are paralogous NAD-dependent oncogenic transcription factors and dehydrogenases that nucleate an epigenetic complex regulating a cohort of genes responsible for cancer proliferation and metastasis. As adequate intracellular NAD is required for CtBP to oligomerize and execute its oncogenic transcriptional coregulatory activities, we hypothesized that NAD depletion would synergize with CtBP inhibition, improving cell inhibitory efficacy. Indeed, depletion of cellular NAD via the NAMPT inhibitor GMX1778 enhanced growth inhibition induced by either RNAi-mediated CtBP1/2 knockdown or the CtBP dehydrogenase inhibitor 4-chlorophenyl-2-hydroxyimino propanoic acid as much as 10-fold in PDAC cells, while untransformed pancreatic ductal cells were unaffected. The growth inhibitory effects of the NAMPT/CtBP inhibitor combination correlated pharmacodynamically with on-target disruption of CtBP1/2 dimerization, CtBP2 interaction with the CoREST epigenetic regulator, and transcriptional activation of the oncogenic target gene TIAM1. Moreover, this same therapeutic combination strongly attenuated growth of PDAC cell line xenografts in immunodeficient mice, with no observable toxicity. Collectively, our data demonstrate that targeting CtBP in combination with NAD depletion represents a promising therapeutic strategy for PDAC. SIGNIFICANCE Effective precision therapies are lacking in PDAC. We demonstrate that simultaneous inhibition of NAD metabolism and the oncoprotein CtBP is potently effective at blocking growth of both PDAC cells in culture and human PDAC-derived tumors in mice and should be explored further as a potential therapy for patients with PDAC.
Collapse
Affiliation(s)
- M. Michael Dcona
- USC Norris Comprehensive Cancer Center and Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Kranthi Kumar Chougoni
- USC Norris Comprehensive Cancer Center and Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Diana T. Dcona
- USC Norris Comprehensive Cancer Center and Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Jacqueline L. West
- Department of Medicinal Chemistry and Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, Virginia
| | - Sahib J. Singh
- VCU Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | - Keith C. Ellis
- Department of Medicinal Chemistry and Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, Virginia
- VCU Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | - Steven R. Grossman
- USC Norris Comprehensive Cancer Center and Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
5
|
Kellogg GE, Cen Y, Dukat M, Ellis KC, Guo Y, Li J, May AE, Safo MK, Zhang S, Zhang Y, Desai UR. Merging cultures and disciplines to create a drug discovery ecosystem at Virginia commonwealth university: Medicinal chemistry, structural biology, molecular and behavioral pharmacology and computational chemistry. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2023; 28:255-269. [PMID: 36863508 PMCID: PMC10619687 DOI: 10.1016/j.slasd.2023.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/10/2023] [Accepted: 02/21/2023] [Indexed: 03/04/2023]
Abstract
The Department of Medicinal Chemistry, together with the Institute for Structural Biology, Drug Discovery and Development, at Virginia Commonwealth University (VCU) has evolved, organically with quite a bit of bootstrapping, into a unique drug discovery ecosystem in response to the environment and culture of the university and the wider research enterprise. Each faculty member that joined the department and/or institute added a layer of expertise, technology and most importantly, innovation, that fertilized numerous collaborations within the University and with outside partners. Despite moderate institutional support with respect to a typical drug discovery enterprise, the VCU drug discovery ecosystem has built and maintained an impressive array of facilities and instrumentation for drug synthesis, drug characterization, biomolecular structural analysis and biophysical analysis, and pharmacological studies. Altogether, this ecosystem has had major impacts on numerous therapeutic areas, such as neurology, psychiatry, drugs of abuse, cancer, sickle cell disease, coagulopathy, inflammation, aging disorders and others. Novel tools and strategies for drug discovery, design and development have been developed at VCU in the last five decades; e.g., fundamental rational structure-activity relationship (SAR)-based drug design, structure-based drug design, orthosteric and allosteric drug design, design of multi-functional agents towards polypharmacy outcomes, principles on designing glycosaminoglycans as drugs, and computational tools and algorithms for quantitative SAR (QSAR) and understanding the roles of water and the hydrophobic effect.
Collapse
Affiliation(s)
- Glen E Kellogg
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, 23298-0540, USA.
| | - Yana Cen
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, 23298-0540, USA
| | - Malgorzata Dukat
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, 23298-0540, USA
| | - Keith C Ellis
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, 23298-0540, USA
| | - Youzhong Guo
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, 23298-0540, USA
| | - Jiong Li
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, 23298-0540, USA
| | - Aaron E May
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, 23298-0540, USA
| | - Martin K Safo
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, 23298-0540, USA
| | - Shijun Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, 23298-0540, USA
| | - Yan Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, 23298-0540, USA
| | - Umesh R Desai
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, 23298-0540, USA.
| |
Collapse
|
6
|
Wang CJ, Jia MZ, Deng LP, Li WJ, Zhang Q, Zhang TJ, Li SY, Cui L, Li ZG. Interaction between CASP8AP2 and ZEB2-CtBP2 Regulates the Expression of LEF1. Pediatr Hematol Oncol 2022; 39:549-560. [PMID: 35139734 DOI: 10.1080/08880018.2022.2033369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Low expression of CTBP2 and CASP8AP2 correlated with poor outcome and predicted risk of relapse in pediatric B-cell acute lymphoblastic leukemia (B-ALL). This study aimed to investigate the molecular mechanism by which CASP8AP2 regulates LEF1 expression by interacting with CtBP2 and ZEB2 in Acute lymphoblastic lymphoma (ALL). There was an interaction between CASP8AP2, ZEB2, and CtBP2, and then the interaction between CtBP2 and ZEB2 was observed after downregulating the expression of CASP8AP2. The wild type (containing the ZEB2 binding site) or mutant (containing a mutant binding site) LEF1 gene promoter sequence was inserted into the pGL3-basic plasmid, and a dual-luciferase reporter gene detection system was used to observe how CASP8AP2, ZEB2, and CtBP2 regulate the transcription of the LEF1 gene. We conclude that CASP8AP2, CtBP2, and ZEB2 can all bind to the LEF1 gene promoter region and reduce the luciferase activity of the LEF1 promoter. Meanwhile, the interaction of ZEB2 and the LEF1 promoter was significantly weakened after downregulation of CASP8AP2. Knockdown of CASP8AP2 in the 697 cell lines resulted in the significant upregulation of the mRNA expression levels of the stemness-related genes CD44, JAG1, and SALL4. In conclusion, CASP8AP2 is vital for the interaction between CtBP2 and ZEB2, inhibiting LEF1 and stemness-related genes expression ALL.Supplemental data for this article is available online at https://doi.org/10.1080/08880018.2022.2033369 .
Collapse
Affiliation(s)
- Chan-Juan Wang
- Hematologic Diseases Laboratory, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics, Capital Medical University, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Ming-Zhu Jia
- Hematologic Diseases Laboratory, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics, Capital Medical University, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Li-Ping Deng
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Wei-Jing Li
- Hematologic Diseases Laboratory, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics, Capital Medical University, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Qing Zhang
- Hematologic Diseases Laboratory, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics, Capital Medical University, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Tong-Jia Zhang
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Science, Peking University Health Science Centre, Beijing, China
| | - Shu-Yan Li
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Science, Peking University Health Science Centre, Beijing, China
| | - Lei Cui
- Hematologic Diseases Laboratory, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics, Capital Medical University, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Zhi-Gang Li
- Hematologic Diseases Laboratory, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics, Capital Medical University, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| |
Collapse
|
7
|
C-terminal binding protein 2 promotes high-glucose-triggered cell proliferation, angiogenesis and cellular adhesion of human retinal endothelial cell line. Int Ophthalmol 2022; 42:2975-2985. [PMID: 35353294 DOI: 10.1007/s10792-022-02283-9] [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: 10/21/2021] [Accepted: 03/12/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE The proliferation and angiogenesis of human retinal endothelial cells (HRECs) are critical for the pathophysiology of diabetic retinopathy (DR). C-terminal binding protein 2 (CtBP2) has multiple biologic functions, but its effect on HRECs under high-glucose (HG) conditions is unclear. METHODS The cell viability, angiogenesis, cellular adhesion and CtBP2 expression levels of HRECs were measured following treatment with different concentrations of glucose. Small interfering CtBP2-targeting RNA, wide-type and function mutant plasmid of CtBP2 were constructed and then were transfected into HRECs to evaluate the effects of CtBP2 on cell functions of HRECs. RESULTS The expression of CtBP2 in HRECs was increased after HG treatment. HG treatment significantly increased cell proliferation, angiogenesis, and decreased relative gene expressions in gap junctions, tight junctions and adherens junctions. After CtBP2 was inhibited via siRNA, the changes induced by HG were partially restored. Conversely, only wild-type CtBP2 could increase cell proliferation and angiogenesis under HG condition. Mechanistically, we also found that CtBP2 exerted its functions to effect HG-induced changes via Akt signaling pathway. CONCLUSION This study implicates that CtBP2 promotes HG-induced cell proliferation, angiogenesis and cellular adhesion, and CtBP2 might be a potential target in the prevention of DR.
Collapse
|
8
|
Zhang W, Kong L, Zhu H, Sun D, Han Q, Yan B, Cui Z, Zhang W, Zhang S, Kang X, Dai G, Qian N, Yan W. Retinoic Acid-Induced 2 (RAI2) Is a Novel Antagonist of Wnt/β-Catenin Signaling Pathway and Potential Biomarker of Chemosensitivity in Colorectal Cancer. Front Oncol 2022; 12:805290. [PMID: 35299743 PMCID: PMC8922473 DOI: 10.3389/fonc.2022.805290] [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: 10/30/2021] [Accepted: 02/04/2022] [Indexed: 11/24/2022] Open
Abstract
Objective Aberrant activation of Wnt/β-catenin signaling contributes to the maintenance of cancer stem cells and chemoresistance in colorectal cancer (CRC). Retinoic acid-induced 2 (RAI2) was proved to be a tumor suppressor in CRC in our previous report. In this study, the role of RAI2 in Wnt/β-catenin signaling was further investigated. Methods As a transcriptional co-regulator, C-terminal Binding Protein 2 (CtBP2) was reported to be involved in Wnt signaling in multiple and complex ways. The correlation of RAI2 and CtBP2 in CRC was analyzed by TCGA dataset, and the interaction between RAI2 and CtBP2 was explored by co-immunoprecipitation (Co-IP) in CRC cells. The effect of RAI2 on the activity of Wnt signaling and the location of β-catenin was detected by Dual-Luciferase reporter assay and Immunofluorescence respectively. Western blotting analysis was performed to detect the expression of target genes involved in Wnt signaling. Sphere formation assay was employed to detect the effect of RAI2 on stem cell like properties. Cell viability assay was used to detect the chemosensitivity of cells before and after transfection of RAI2. Results The interaction between RAI2 and CtBP2 was confirmed by Co-IP in CRC cells. Besides, the negative correlation of RAI2 and CtBP2 in CRC was found by analyzing the TCGA dataset. Re-expression of RAI2 in human colon cancer cells (HCT116 and LoVo) suppressed the fluorescent activity of Wnt signaling, increased the phosphorylation and inhibited nuclear translocation of β-catenin, with down-regulation of target genes like c-Myc, CyclinD1, ASCL2, and LGR5. In contrast, the mutated RAI2, which can’t interact with CtBP2, has no above effects. We observed low expression of RAI2 in 33.89% (101/298) of CRC patients, which was significantly associated with reduced phosphorylation of β-catenin (r=0.8866, P<0.0001), poor 5-year relapse-free survival (RFS) (P = 0.0029) and overall survival (OS) (P = 0.0102). Restoration of RAI2 in HCT116 and LoVo cells inhibited stem cell-like properties of CRC cells and increased chemosensitivity of these cells to oxaliplatin and fluorouracil. Conclusion Low expression of RAI2 can serve as an independent poor prognostic marker. RAI2 inhibits Wnt signaling by interacting with or down-regulating CtBP2, resulting in repression of stem cell-like properties and increased chemosensitivity of CRC cells.
Collapse
Affiliation(s)
- Weitao Zhang
- Department of Oncology, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,Cancer Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Lu Kong
- Department of Oncology, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,Medical Department, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Hongbin Zhu
- Department of Gastroenterology and Hepatology, Chinese People's Liberation Army (PLA) NO.983 Hospital, Tianjin, China
| | - Decong Sun
- Department of Oncology, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Quanli Han
- Department of Oncology, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Bin Yan
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Zhi Cui
- Department of Oncology, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Weiwei Zhang
- Department of Oncology, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Shurong Zhang
- Cancer Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xindan Kang
- Department of Oncology, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Guanghai Dai
- Department of Oncology, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Niansong Qian
- Department of Oncology, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Wenji Yan
- Department of Oncology, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|
9
|
Erlandsen H, Jecrois AM, Nichols JC, Cole JL, Royer WE. NADH/NAD + binding and linked tetrameric assembly of the oncogenic transcription factors CtBP1 and CtBP2. FEBS Lett 2022; 596:479-490. [PMID: 34997967 DOI: 10.1002/1873-3468.14276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/14/2021] [Accepted: 12/22/2021] [Indexed: 11/08/2022]
Abstract
The activation of oncogenic C-terminal binding Protein (CtBP) transcriptional activity is coupled with NAD(H) binding and homo-oligomeric assembly, although the level of CtBP assembly and nucleotide binding affinity continues to be debated. Here, we apply biophysical techniques to address these fundamental issues for CtBP1 and CtBP2. Our ultracentrifugation results unambiguously demonstrate that CtBP assembles into tetramers in the presence of saturating NAD+ or NADH with tetramer to dimer dissociation constants about 100 nm. Isothermal titration calorimetry measurements of NAD(H) binding to CtBP show dissociation constants between 30 and 500 nm, depending on the nucleotide and paralog. Given cellular levels of NAD+ , CtBP is likely to be fully saturated with NAD under physiological concentrations suggesting that CtBP is unable to act as a sensor for NADH levels.
Collapse
Affiliation(s)
- Heidi Erlandsen
- Center for Open Research Resources & Equipment, University of Connecticut, Storrs, CT, USA
| | - Anne M Jecrois
- Department of Biochemistry and Molecular Biotechnology, UMass Chan Medical School, Worcester, MA, USA
| | - Jeffry C Nichols
- Department of Biochemistry and Molecular Biotechnology, UMass Chan Medical School, Worcester, MA, USA.,Chemistry Department, Worcester State University, MA, USA
| | - James L Cole
- Department of Molecular and Cell Biology, Department of Chemistry, University of Connecticut, CT, USA
| | - William E Royer
- Department of Biochemistry and Molecular Biotechnology, UMass Chan Medical School, Worcester, MA, USA
| |
Collapse
|
10
|
He J, Chu Z, Lai W, Lan Q, Zeng Y, Lu D, Jin S, Xu H, Su P, Yin D, Chu Z, Liu L. Circular RNA circHERC4 as a novel oncogenic driver to promote tumor metastasis via the miR-556-5p/CTBP2/E-cadherin axis in colorectal cancer. J Hematol Oncol 2021; 14:194. [PMID: 34781990 PMCID: PMC8591961 DOI: 10.1186/s13045-021-01210-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 11/04/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The main cause of death in colorectal cancer patients is metastasis. Accumulating evidences suggest that circRNA plays pivotal roles in cancer initiation and development. However, the underlying molecular mechanisms of circRNAs that orchestrate cancer metastasis remain vague and need further clarification. METHODS Two paired CRC and adjacent normal tissues were used to screen the upregulated circRNAs by circRNA-seq; then, cell invasion assay was applied to confirm the functional invasion-related circRNAs. According to the above methods, circHERC4 (hsa_circ_0007113) was selected for further research. Next, we investigated the clinical significance of circHERC4 in a large cohort of patients with CRC. The oncogenic activity of circHERC4 was investigated in both CRC cell lines and animal xenograft studies. Finally, we explored the molecular mechanisms underlying circHERC4 as a malignant driver. RESULTS We demonstrated that circHERC4 was aberrantly elevated in CRC tissues (P < 0.001), and was positively associated with lymph node metastasis and advanced tumor grade (P < 0.01). Notably, the expression of circHERC4 was associated with worse survival in patients with CRC. Silencing of circHERC4 significantly inhibited the proliferation and migration of two highly aggressive CRC cell lines and reduced liver and lung metastasis in vivo. Mechanistically, we revealed that circHERC4 inactivated the tumor suppressor, miR-556-5p, leading to the activation of CTBP2/E-cadherin pathway which promotes tumor metastasis in CRC. CONCLUSIONS CircHERC4 exerts critical roles in promoting tumor aggressiveness through miR-556-5p/CTBP2/E-cadherin pathway and is a prognostic biomarker of the disease, suggesting that circHERC4 may serve as an exploitable therapeutic target for patients with CRC.
Collapse
Affiliation(s)
- Jiehua He
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan-Jiang Xi Road, Guangzhou, 510120, People's Republic of China
| | - Ziqiang Chu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan-Jiang Xi Road, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Wei Lai
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan-Jiang Xi Road, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Qiusheng Lan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan-Jiang Xi Road, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Yujie Zeng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan-Jiang Xi Road, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Daning Lu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan-Jiang Xi Road, Guangzhou, 510120, People's Republic of China
| | - Shaowen Jin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan-Jiang Xi Road, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Heyang Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan-Jiang Xi Road, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Pengwei Su
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan-Jiang Xi Road, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Dong Yin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China.
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan-Jiang Xi Road, Guangzhou, 510120, People's Republic of China.
| | - Zhonghua Chu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China.
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan-Jiang Xi Road, Guangzhou, 510120, Guangdong, People's Republic of China.
| | - Lu Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China.
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan-Jiang Xi Road, Guangzhou, 510120, Guangdong, People's Republic of China.
| |
Collapse
|
11
|
The transrepression and transactivation roles of CtBPs in the pathogenesis of different diseases. J Mol Med (Berl) 2021; 99:1335-1347. [PMID: 34196767 DOI: 10.1007/s00109-021-02107-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/31/2021] [Accepted: 06/25/2021] [Indexed: 02/06/2023]
Abstract
Gene transcription is strictly controlled by transcriptional complexes, which are assemblies of transcription factors, transcriptional regulators, and co-regulators. Mammalian genomes encode two C-terminal-binding proteins (CtBPs), CtBP1 and CtBP2, which are both well-known transcriptional corepressors of oncogenic processes. Their overexpression in tumors is associated with malignant behavior, such as uncontrolled cell proliferation, migration, and invasion, as well as with an increase in the epithelial-mesenchymal transition. CtBPs coordinate with other transcriptional regulators, such as histone deacetylases (HDACs) and histone acetyltransferases (p300 and CBP [CREBP-binding protein]) that contain the PXDLS motif, and with transcription factors to assemble transcriptional complexes that dock onto the promoters of genes to initiate gene transcription. Emerging evidence suggests that CtBPs function as both corepressors and coactivators in different biological processes ranging from apoptosis to inflammation and osteogenesis. Therapeutic targeting of CtBPs or the interactions required to form transcriptional complexes has also shown promising effects in preventing disease progression. This review summarizes the most recent progress in the study of CtBP functions and therapeutic inhibitors in different biological processes. This knowledge may enable a better understanding of the complexity of the roles of CtBPs, while providing new insights into therapeutic strategies that target CtBPs.
Collapse
|
12
|
Nichols JC, Schiffer CA, Royer WE. NAD(H) phosphates mediate tetramer assembly of human C-terminal binding protein (CtBP). J Biol Chem 2021; 296:100351. [PMID: 33524397 PMCID: PMC7949142 DOI: 10.1016/j.jbc.2021.100351] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/22/2021] [Accepted: 01/27/2021] [Indexed: 12/27/2022] Open
Abstract
C-terminal binding proteins (CtBPs) are cotranscriptional factors that play key roles in cell fate. We have previously shown that NAD(H) promotes the assembly of similar tetramers from either human CtBP1 and CtBP2 and that CtBP2 tetramer destabilizing mutants are defective for oncogenic activity. To assist structure-based design efforts for compounds that disrupt CtBP tetramerization, it is essential to understand how NAD(H) triggers tetramer assembly. Here, we investigate the moieties within NAD(H) that are responsible for triggering tetramer formation. Using multiangle light scattering (MALS), we show that ADP is able to promote tetramer formation of both CtBP1 and CtBP2, whereas AMP promotes tetramer assembly of CtBP1, but not CtBP2. Other NAD(H) moieties that lack the adenosine phosphate, including adenosine and those incorporating nicotinamide, all fail to promote tetramer assembly. Our crystal structures of CtBP1 with AMP reveal participation of the adenosine phosphate in the tetrameric interface, pinpointing its central role in NAD(H)-linked assembly. CtBP1 and CtBP2 have overlapping but unique roles, suggesting that a detailed understanding of their unique structural properties might have utility in the design of paralog-specific inhibitors. We investigated the different responses to AMP through a series of site-directed mutants at 13 positions. These mutations reveal a central role for a hinge segment, which we term the 120s hinge that connects the substrate with coenzyme-binding domains and influences nucleotide binding and tetramer assembly. Our results provide insight into suitable pockets to explore in structure-based drug design to interfere with cotranscriptional activity of CtBP in cancer.
Collapse
Affiliation(s)
- Jeffry C Nichols
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, USA; Chemistry Department, Worcester State University, Worcester, Massachusetts, USA
| | - Celia A Schiffer
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - William E Royer
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, USA.
| |
Collapse
|
13
|
Gromisch C, Qadan M, Machado MA, Liu K, Colson Y, Grinstaff MW. Pancreatic Adenocarcinoma: Unconventional Approaches for an Unconventional Disease. Cancer Res 2020; 80:3179-3192. [PMID: 32220831 PMCID: PMC7755309 DOI: 10.1158/0008-5472.can-19-2731] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 02/08/2020] [Accepted: 03/24/2020] [Indexed: 12/16/2022]
Abstract
This review highlights current treatments, limitations, and pitfalls in the management of pancreatic cancer and discusses current research in novel targets and drug development to overcome these clinical challenges. We begin with a review of the clinical landscape of pancreatic cancer, including genetic and environmental risk factors, as well as limitations in disease diagnosis and prevention. We next discuss current treatment paradigms for pancreatic cancer and the shortcomings of targeted therapy in this disease. Targeting major driver mutations in pancreatic cancer, such as dysregulation in the KRAS and TGFβ signaling pathways, have failed to improve survival outcomes compared with nontargeted chemotherapy; thus, we describe new advances in therapy such as Ras-binding pocket inhibitors. We then review next-generation approaches in nanomedicine and drug delivery, focusing on preclinical advancements in novel optical probes, antibodies, small-molecule agents, and nucleic acids to improve surgical outcomes in resectable disease, augment current therapies, expand druggable targets, and minimize morbidity. We conclude by summarizing progress in current research, identifying areas for future exploration in drug development and nanotechnology, and discussing future prospects for management of this disease.
Collapse
Affiliation(s)
- Christopher Gromisch
- Departments of Pharmacology and Experimental Therapeutics, Biomedical Engineering, and Chemistry, Boston University, Boston, Massachusetts
| | - Motaz Qadan
- Division of Surgical Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Mariana Albuquerque Machado
- Departments of Pharmacology and Experimental Therapeutics, Biomedical Engineering, and Chemistry, Boston University, Boston, Massachusetts
| | - Kebin Liu
- Department of Biochemistry and Molecular Biology and Georgia Cancer Center, Medical College of Georgia, Augusta, Georgia
| | - Yolonda Colson
- Division of Thoracic Surgery, Massachusetts General Hospital, Boston, Massachusetts
| | - Mark W Grinstaff
- Departments of Pharmacology and Experimental Therapeutics, Biomedical Engineering, and Chemistry, Boston University, Boston, Massachusetts.
| |
Collapse
|
14
|
CtBP determines ovarian cancer cell fate through repression of death receptors. Cell Death Dis 2020; 11:286. [PMID: 32332713 PMCID: PMC7181866 DOI: 10.1038/s41419-020-2455-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/18/2020] [Accepted: 03/19/2020] [Indexed: 02/06/2023]
Abstract
C-terminal binding protein 2 (CtBP2) is elevated in epithelial ovarian cancer, especially in the aggressive and highly lethal subtype, high-grade serous ovarian cancer (HGSOC). However, whether HGSOC tumor progression is dependent on CtBP2 or its paralog CtBP1, is not well understood. Here we report that CtBP1/2 repress HGSOC cell apoptosis through silencing of death receptors (DRs) 4/5. CtBP1 or 2 knockdown upregulated DR4/5 expression, and triggered autonomous apoptosis via caspase 8 activation, but dependent on cell-type context. Activation of DR4/5 by CtBP1/2 loss also sensitized HGSOC cell susceptibility to the proapoptotic DR4/5 ligand TRAIL. Consistent with its function as transcription corepressor, CtBP1/2 bound to the promoter regions of DR4/5 and repressed DR4/5 expression, presumably through recruitment to a repressive transcription regulatory complex. We also found that CtBP1 and 2 were both required for repression of DR4/5. Collectively, this study identifies CtBP1 and 2 as potent repressors of DR4/5 expression and activity, and supports the targeting of CtBP as a promising therapeutic strategy for HGSOC.
Collapse
|
15
|
Honjo K, Won WJ, King RG, Ianov L, Crossman DK, Easlick JL, Shakhmatov MA, Khass M, Vale AM, Stephan RP, Li R, Davis RS. Fc Receptor-Like 6 (FCRL6) Discloses Progenitor B Cell Heterogeneity That Correlates With Pre-BCR Dependent and Independent Pathways of Natural Antibody Selection. Front Immunol 2020; 11:82. [PMID: 32117244 PMCID: PMC7033751 DOI: 10.3389/fimmu.2020.00082] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 01/13/2020] [Indexed: 11/24/2022] Open
Abstract
B-1a cells produce "natural" antibodies (Abs) to neutralize pathogens and clear neo self-antigens, but the fundamental selection mechanisms that shape their polyreactive repertoires are poorly understood. Here, we identified a B cell progenitor subset defined by Fc receptor-like 6 (FCRL6) expression, harboring innate-like defense, migration, and differentiation properties conducive for natural Ab generation. Compared to FCRL6- pro B cells, the repressed mitotic, DNA damage repair, and signaling activity of FCRL6+ progenitors, yielded VH repertoires with biased distal Ighv segment accessibility, constrained diversity, and hydrophobic and charged CDR-H3 sequences. Beyond nascent autoreactivity, VH11 productivity, which predominates phosphatidylcholine-specific B-1a B cell receptors (BCRs), was higher for FCRL6+ cells as was pre-BCR formation, which was required for Myc induction and VH11, but not VH12, B-1a development. Thus, FCRL6 revealed unexpected heterogeneity in the developmental origins, regulation, and selection of natural Abs at the pre-BCR checkpoint with implications for autoimmunity and lymphoproliferative disorders.
Collapse
MESH Headings
- Animals
- Antibodies/immunology
- Antibodies/metabolism
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Female
- Humans
- Immunoglobulin Heavy Chains/genetics
- Immunoglobulin Heavy Chains/immunology
- Immunoglobulin Variable Region/genetics
- Immunoglobulin Variable Region/immunology
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Phosphatidylcholines/immunology
- Phosphatidylcholines/metabolism
- Precursor Cells, B-Lymphoid/immunology
- Precursor Cells, B-Lymphoid/metabolism
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/immunology
- Receptors, Antigen, B-Cell/metabolism
- Receptors, Fc/genetics
- Receptors, Fc/immunology
- Receptors, Fc/metabolism
- Signal Transduction/genetics
- Signal Transduction/immunology
Collapse
Affiliation(s)
- Kazuhito Honjo
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Woong-Jai Won
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Rodney G. King
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Lara Ianov
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - David K. Crossman
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Juliet L. Easlick
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mikhail A. Shakhmatov
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mohamed Khass
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Genetic Engineering and Biotechnology Division, National Research Center, Cairo, Egypt
| | - Andre M. Vale
- Program in Immunobiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Robert P. Stephan
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ran Li
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Randall S. Davis
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, United States
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
16
|
Ding B, Haidurov A, Chawla A, Parmigiani A, van de Kamp G, Dalina A, Yuan F, Lee JH, Chumakov PM, Grossman SR, Budanov AV. p53-inducible SESTRINs might play opposite roles in the regulation of early and late stages of lung carcinogenesis. Oncotarget 2019; 10:6997-7009. [PMID: 31857853 PMCID: PMC6916756 DOI: 10.18632/oncotarget.27367] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 11/17/2019] [Indexed: 01/13/2023] Open
Abstract
SESTRINs (SESN1-3) are proteins encoded by an evolutionarily conserved gene family that plays an important role in the regulation of cell viability and metabolism in response to stress. Many of the effects of SESTRINs are mediated by negative and positive regulation of mechanistic target of rapamycin kinase complexes 1 and 2 (mTORC1 and mTORC2), respectively, that are often deregulated in human cancers where they support cell growth, proliferation, and cell viability. Besides their effects on regulation of mTORC1/2, SESTRINs also control the accumulation of reactive oxygen species, cell death, and mitophagy. SESN1 and SESN2 are transcriptional targets of tumor suppressor protein p53 and may mediate tumor suppressor activities of p53. Therefore, we conducted studies based on a mouse lung cancer model and human lung adenocarcinoma A549 cells to evaluate the potential impact of SESN1 and SESN2 on lung carcinogenesis. While we observed that expression of SESN1 and SESN2 is often decreased in human tumors, inactivation of Sesn2 in mice positively regulates tumor growth through a mechanism associated with activation of AKT, while knockout of Sesn1 has no additional impact on carcinogenesis in Sesn2-deficient mice. However, inactivation of SESN1 and/or SESN2 in A549 cells accelerates cell proliferation and imparts resistance to cell death in response to glucose starvation. We propose that despite their contribution to early tumor growth, SESTRINs might suppress late stages of carcinogenesis through inhibition of cell proliferation or activation of cell death in conditions of nutrient deficiency.
Collapse
Affiliation(s)
- Boxiao Ding
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA.,These authors contributed equally to this work
| | - Alexander Haidurov
- Engelhardt Institute of Molecular Biology, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Moscow, Russia.,School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.,These authors contributed equally to this work
| | - Ayesha Chawla
- Wright Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, VA, USA
| | - Anita Parmigiani
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Gerarda van de Kamp
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Alexandra Dalina
- Engelhardt Institute of Molecular Biology, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Moscow, Russia
| | - Fang Yuan
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Jun Hee Lee
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Peter M Chumakov
- Engelhardt Institute of Molecular Biology, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Moscow, Russia
| | - Steven R Grossman
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA.,Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA.,VCU Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Andrei V Budanov
- Engelhardt Institute of Molecular Biology, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Moscow, Russia.,School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.,Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
17
|
CtBP-a targetable dependency for tumor-initiating cell activity and metastasis in pancreatic adenocarcinoma. Oncogenesis 2019; 8:55. [PMID: 31586042 PMCID: PMC6778071 DOI: 10.1038/s41389-019-0163-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 07/04/2019] [Accepted: 08/21/2019] [Indexed: 02/07/2023] Open
Abstract
Ctbp2 is a uniquely targetable oncogenic transcriptional coregulator, exhibiting overexpression in most common solid tumors, and critical to the tumor-initiating cell (TIC) transcriptional program. In the “CKP” mouse pancreatic ductal adenocarcinoma (PDAC) model driven by mutant K-Ras, Ctbp2 haploinsufficiency prolonged survival, abrogated peritoneal metastasis, and caused dramatic downregulation of c-Myc, a known critical dependency for TIC activity and tumor progression in PDAC. A small-molecule inhibitor of CtBP2, 4-chloro-hydroxyimino phenylpyruvate (4-Cl-HIPP) phenocopied Ctbp2 deletion, decreasing tumor burden similarly to gemcitabine, and the combination of 4-Cl-HIPP and gemcitabine further synergistically suppressed tumor growth. Pharmacodynamic monitoring revealed that the 4-Cl-HIPP/gemcitabine combination induced robust and synergistic tumor apoptosis and marked downregulation of the TIC marker CD133 in CKP PDAC tumors. Collectively, our data demonstrate that targeting CtBP represents a fruitful avenue for development of highly active agents in PDAC that cooperate with standard therapy to limit both primary and metastatic tumor burden.
Collapse
|
18
|
Dcona MM, Damle PK, Zarate-Perez F, Morris BL, Nawaz Z, Dennis MJ, Deng X, Korwar S, Singh SJ, Ellis KC, Royer WE, Bandyopadhyay D, Escalante C, Grossman SR. Active-Site Tryptophan, the Target of Antineoplastic C-Terminal Binding Protein Inhibitors, Mediates Inhibitor Disruption of CtBP Oligomerization and Transcription Coregulatory Activities. Mol Pharmacol 2019; 96:99-108. [PMID: 31036695 PMCID: PMC6560337 DOI: 10.1124/mol.118.114363] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 04/21/2019] [Indexed: 01/20/2023] Open
Abstract
C-terminal binding proteins (CtBP1/2) are oncogenic transcriptional coregulators and dehydrogenases often overexpressed in multiple solid tumors, including breast, colon, and ovarian cancer, and associated with poor survival. CtBPs act by repressing expression of genes responsible for apoptosis (e.g., PUMA, BIK) and metastasis-associated epithelial-mesenchymal transition (e.g., CDH1), and by activating expression of genes that promote migratory and invasive properties of cancer cells (e.g., TIAM1) and genes responsible for enhanced drug resistance (e.g., MDR1). CtBP's transcriptional functions are also critically dependent on oligomerization and nucleation of transcriptional complexes. Recently, we have developed a family of CtBP dehydrogenase inhibitors, based on the parent 2-hydroxyimino-3-phenylpropanoic acid (HIPP), that specifically disrupt cancer cell viability, abrogate CtBP's transcriptional function, and block polyp formation in a mouse model of intestinal polyposis that depends on CtBP's oncogenic functions. Crystallographic analysis revealed that HIPP interacts with CtBP1/2 at a conserved active site tryptophan (W318/324; CtBP1/2) that is unique among eukaryotic D2-dehydrogenases. To better understand the mechanism of action of HIPP-class inhibitors, we investigated the contribution of W324 to CtBP2's biochemical and physiologic activities utilizing mutational analysis. Indeed, W324 was necessary for CtBP2 self-association, as shown by analytical ultracentrifugation and in vivo cross-linking. Additionally, W324 supported CtBP's association with the transcriptional corepressor CoREST, and was critical for CtBP2 induction of cell motility. Notably, the HIPP derivative 4-chloro-HIPP biochemically and biologically phenocopied mutational inactivation of CtBP2 W324. Our data support further optimization of W318/W324-interacting CtBP dehydrogenase inhibitors that are emerging as a novel class of cancer cell-specific therapeutic.
Collapse
Affiliation(s)
- M Michael Dcona
- Departments of Internal Medicine (M.M.D., P.K.D., Z.N., M.J.D., S.J.S., S.R.G.), Human and Molecular Genetics (B.L.M., S.R.G.), Physiology and Biophysics (F.Z.-P., C.E.), Medicinal Chemistry (S.K., K.C.E.), and Biostatistics (X.D., D.B.) and Massey Cancer Center (K.C.E., D.B., C.E., S.R.G.), Virginia Commonwealth University, Richmond, Virginia; and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (W.E.R.)
| | - Priyadarshan K Damle
- Departments of Internal Medicine (M.M.D., P.K.D., Z.N., M.J.D., S.J.S., S.R.G.), Human and Molecular Genetics (B.L.M., S.R.G.), Physiology and Biophysics (F.Z.-P., C.E.), Medicinal Chemistry (S.K., K.C.E.), and Biostatistics (X.D., D.B.) and Massey Cancer Center (K.C.E., D.B., C.E., S.R.G.), Virginia Commonwealth University, Richmond, Virginia; and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (W.E.R.)
| | - Francisco Zarate-Perez
- Departments of Internal Medicine (M.M.D., P.K.D., Z.N., M.J.D., S.J.S., S.R.G.), Human and Molecular Genetics (B.L.M., S.R.G.), Physiology and Biophysics (F.Z.-P., C.E.), Medicinal Chemistry (S.K., K.C.E.), and Biostatistics (X.D., D.B.) and Massey Cancer Center (K.C.E., D.B., C.E., S.R.G.), Virginia Commonwealth University, Richmond, Virginia; and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (W.E.R.)
| | - Benjamin L Morris
- Departments of Internal Medicine (M.M.D., P.K.D., Z.N., M.J.D., S.J.S., S.R.G.), Human and Molecular Genetics (B.L.M., S.R.G.), Physiology and Biophysics (F.Z.-P., C.E.), Medicinal Chemistry (S.K., K.C.E.), and Biostatistics (X.D., D.B.) and Massey Cancer Center (K.C.E., D.B., C.E., S.R.G.), Virginia Commonwealth University, Richmond, Virginia; and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (W.E.R.)
| | - Zaid Nawaz
- Departments of Internal Medicine (M.M.D., P.K.D., Z.N., M.J.D., S.J.S., S.R.G.), Human and Molecular Genetics (B.L.M., S.R.G.), Physiology and Biophysics (F.Z.-P., C.E.), Medicinal Chemistry (S.K., K.C.E.), and Biostatistics (X.D., D.B.) and Massey Cancer Center (K.C.E., D.B., C.E., S.R.G.), Virginia Commonwealth University, Richmond, Virginia; and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (W.E.R.)
| | - Michael J Dennis
- Departments of Internal Medicine (M.M.D., P.K.D., Z.N., M.J.D., S.J.S., S.R.G.), Human and Molecular Genetics (B.L.M., S.R.G.), Physiology and Biophysics (F.Z.-P., C.E.), Medicinal Chemistry (S.K., K.C.E.), and Biostatistics (X.D., D.B.) and Massey Cancer Center (K.C.E., D.B., C.E., S.R.G.), Virginia Commonwealth University, Richmond, Virginia; and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (W.E.R.)
| | - Xiaoyan Deng
- Departments of Internal Medicine (M.M.D., P.K.D., Z.N., M.J.D., S.J.S., S.R.G.), Human and Molecular Genetics (B.L.M., S.R.G.), Physiology and Biophysics (F.Z.-P., C.E.), Medicinal Chemistry (S.K., K.C.E.), and Biostatistics (X.D., D.B.) and Massey Cancer Center (K.C.E., D.B., C.E., S.R.G.), Virginia Commonwealth University, Richmond, Virginia; and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (W.E.R.)
| | - Sudha Korwar
- Departments of Internal Medicine (M.M.D., P.K.D., Z.N., M.J.D., S.J.S., S.R.G.), Human and Molecular Genetics (B.L.M., S.R.G.), Physiology and Biophysics (F.Z.-P., C.E.), Medicinal Chemistry (S.K., K.C.E.), and Biostatistics (X.D., D.B.) and Massey Cancer Center (K.C.E., D.B., C.E., S.R.G.), Virginia Commonwealth University, Richmond, Virginia; and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (W.E.R.)
| | - Sahib J Singh
- Departments of Internal Medicine (M.M.D., P.K.D., Z.N., M.J.D., S.J.S., S.R.G.), Human and Molecular Genetics (B.L.M., S.R.G.), Physiology and Biophysics (F.Z.-P., C.E.), Medicinal Chemistry (S.K., K.C.E.), and Biostatistics (X.D., D.B.) and Massey Cancer Center (K.C.E., D.B., C.E., S.R.G.), Virginia Commonwealth University, Richmond, Virginia; and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (W.E.R.)
| | - Keith C Ellis
- Departments of Internal Medicine (M.M.D., P.K.D., Z.N., M.J.D., S.J.S., S.R.G.), Human and Molecular Genetics (B.L.M., S.R.G.), Physiology and Biophysics (F.Z.-P., C.E.), Medicinal Chemistry (S.K., K.C.E.), and Biostatistics (X.D., D.B.) and Massey Cancer Center (K.C.E., D.B., C.E., S.R.G.), Virginia Commonwealth University, Richmond, Virginia; and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (W.E.R.)
| | - William E Royer
- Departments of Internal Medicine (M.M.D., P.K.D., Z.N., M.J.D., S.J.S., S.R.G.), Human and Molecular Genetics (B.L.M., S.R.G.), Physiology and Biophysics (F.Z.-P., C.E.), Medicinal Chemistry (S.K., K.C.E.), and Biostatistics (X.D., D.B.) and Massey Cancer Center (K.C.E., D.B., C.E., S.R.G.), Virginia Commonwealth University, Richmond, Virginia; and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (W.E.R.)
| | - Dipankar Bandyopadhyay
- Departments of Internal Medicine (M.M.D., P.K.D., Z.N., M.J.D., S.J.S., S.R.G.), Human and Molecular Genetics (B.L.M., S.R.G.), Physiology and Biophysics (F.Z.-P., C.E.), Medicinal Chemistry (S.K., K.C.E.), and Biostatistics (X.D., D.B.) and Massey Cancer Center (K.C.E., D.B., C.E., S.R.G.), Virginia Commonwealth University, Richmond, Virginia; and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (W.E.R.)
| | - Carlos Escalante
- Departments of Internal Medicine (M.M.D., P.K.D., Z.N., M.J.D., S.J.S., S.R.G.), Human and Molecular Genetics (B.L.M., S.R.G.), Physiology and Biophysics (F.Z.-P., C.E.), Medicinal Chemistry (S.K., K.C.E.), and Biostatistics (X.D., D.B.) and Massey Cancer Center (K.C.E., D.B., C.E., S.R.G.), Virginia Commonwealth University, Richmond, Virginia; and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (W.E.R.)
| | - Steven R Grossman
- Departments of Internal Medicine (M.M.D., P.K.D., Z.N., M.J.D., S.J.S., S.R.G.), Human and Molecular Genetics (B.L.M., S.R.G.), Physiology and Biophysics (F.Z.-P., C.E.), Medicinal Chemistry (S.K., K.C.E.), and Biostatistics (X.D., D.B.) and Massey Cancer Center (K.C.E., D.B., C.E., S.R.G.), Virginia Commonwealth University, Richmond, Virginia; and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts (W.E.R.)
| |
Collapse
|
19
|
An intestinal stem cell niche in Apc mutated neoplasia targetable by CtBP inhibition. Oncotarget 2018; 9:32408-32418. [PMID: 30197752 PMCID: PMC6126694 DOI: 10.18632/oncotarget.25784] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 06/19/2018] [Indexed: 12/18/2022] Open
Abstract
C-terminal binding protein 2 (CtBP2) drives intestinal polyposis in the Apcmin mouse model of human Familial Adenomatous Polyposis. As CtBP2 is targetable by an inhibitor of its dehydrogenase domain, understanding CtBP2’s role in adenoma formation is necessary to optimize CtBP-targeted therapies in Apc mutated human neoplasia. Tumor initiating cell (TIC) populations were substantially decreased in ApcminCtbp2+/- intestinal epithelia. Moreover, normally nuclear Ctbp2 was mislocalized to the cytoplasm of intestinal crypt stem cells in Ctbp2+/- mice, both Apcmin and wildtype, correlating with low/absent CD133 expression in those cells, and possibly explaining the lower burden of polyps in Apcmin Ctbp2+/- mice. The CtBP inhibitor 4-chloro-hydroxyimino phenylpyruvate (4-Cl-HIPP) also robustly downregulated TIC populations and significantly decreased intestinal polyposis in Apcmin mice. We have therefore demonstrated a critical link between polyposis, intestinal TIC’s and Ctbp2 gene dosage or activity, supporting continued efforts targeting CtBP in the treatment or prevention of Apc mutated neoplasia.
Collapse
|
20
|
Blevins MA, Zhang C, Zhang L, Li H, Li X, Norris DA, Huang M, Zhao R. CPP-E1A fusion peptides inhibit CtBP-mediated transcriptional repression. Mol Oncol 2018; 12:1358-1373. [PMID: 29879296 PMCID: PMC6068344 DOI: 10.1002/1878-0261.12330] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 05/17/2018] [Accepted: 05/18/2018] [Indexed: 12/29/2022] Open
Abstract
The carboxyl‐terminal binding proteins (CtBP) are transcriptional corepressors that regulate the expression of multiple epithelial‐specific and pro‐apoptotic genes. Overexpression of CtBP occurs in many human cancers where they promote the epithelial‐to‐mesenchymal transition, stem cell‐like features, and cell survival, while knockdown of CtBP in tumor cells results in p53‐independent apoptosis. CtBPs are recruited to their target genes by binding to a conserved PXDLS peptide motif present in multiple DNA‐binding transcription factors. Disrupting the interaction between CtBP and its transcription factor partners may be a means of altering CtBP‐mediated transcriptional repression and a potential approach for cancer therapies. However, small molecules targeting protein–protein interactions have traditionally been difficult to identify. In this study, we took advantage of the fact that CtBP binds to a conserved peptide motif to explore the feasibility of using peptides containing the PXDLS motif fused to cell‐penetrating peptides (CPP) to inhibit CtBP function. We demonstrate that these peptides disrupt the ability of CtBP to interact with its protein partner, E1A, in an AlphaScreen assay. Moreover, these peptides can enter both lung carcinoma and melanoma cells, disrupt the interaction between CtBP and a transcription factor partner, and inhibit CtBP‐mediated transcriptional repression. Finally, the constitutive expression of one such peptide, Pep1‐E1A‐WT, in a melanoma cell line reverses CtBP‐mediated oncogenic phenotypes including proliferation, migration, and sphere formation and limits tumor growth in vivo. Together, our results suggest that CPP‐fused PXDLS‐containing peptides can potentially be developed into a research tool or therapeutic agent targeting CtBP‐mediated transcriptional events in various biological pathways.
Collapse
Affiliation(s)
- Melanie A Blevins
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Caiguo Zhang
- Department of Dermatology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Lingdi Zhang
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Hong Li
- Department of Dermatology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Xueni Li
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - David A Norris
- Department of Dermatology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Mingxia Huang
- Department of Dermatology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Rui Zhao
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
21
|
Bellesis AG, Jecrois AM, Hayes JA, Schiffer CA, Royer WE. Assembly of human C-terminal binding protein (CtBP) into tetramers. J Biol Chem 2018; 293:9101-9112. [PMID: 29700119 DOI: 10.1074/jbc.ra118.002514] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 04/24/2018] [Indexed: 11/06/2022] Open
Abstract
C-terminal binding protein 1 (CtBP1) and CtBP2 are transcriptional coregulators that repress numerous cellular processes, such as apoptosis, by binding transcription factors and recruiting chromatin-remodeling enzymes to gene promoters. The NAD(H)-linked oligomerization of human CtBP is coupled to its co-transcriptional activity, which is implicated in cancer progression. However, the biologically relevant level of CtBP assembly has not been firmly established; nor has the stereochemical arrangement of the subunits above that of a dimer. Here, multi-angle light scattering (MALS) data established the NAD+- and NADH-dependent assembly of CtBP1 and CtBP2 into tetramers. An examination of subunit interactions within CtBP1 and CtBP2 crystal lattices revealed that both share a very similar tetrameric arrangement resulting from assembly of two dimeric pairs, with specific interactions probably being sensitive to NAD(H) binding. Creating a series of mutants of both CtBP1 and CtBP2, we tested the hypothesis that the crystallographically observed interdimer pairing stabilizes the solution tetramer. MALS data confirmed that these mutants disrupt both CtBP1 and CtBP2 tetramers, with the dimer generally remaining intact, providing the first stereochemical models for tetrameric assemblies of CtBP1 and CtBP2. The crystal structure of a subtle destabilizing mutant suggested that small structural perturbations of the hinge region linking the substrate- and NAD-binding domains are sufficient to weaken the CtBP1 tetramer. These results strongly suggest that the tetramer is important in CtBP function, and the series of CtBP mutants reported here can be used to investigate the physiological role of the tetramer.
Collapse
Affiliation(s)
- Andrew G Bellesis
- From the Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605 and.,the Carlson School of Chemistry and Biochemistry, Clark University, Worcester, Massachusetts 01610
| | - Anne M Jecrois
- From the Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605 and
| | - Janelle A Hayes
- From the Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605 and
| | - Celia A Schiffer
- From the Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605 and
| | - William E Royer
- From the Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605 and
| |
Collapse
|
22
|
Shi H, Mao Y, Ju Q, Wu Y, Bai W, Wang P, Zhang Y, Jiang M. C-terminal binding protein‑2 mediates cisplatin chemoresistance in esophageal cancer cells via the inhibition of apoptosis. Int J Oncol 2018; 53:167-176. [PMID: 29658564 DOI: 10.3892/ijo.2018.4367] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 03/09/2018] [Indexed: 11/05/2022] Open
Abstract
C-terminal binding protein‑2 (CtBP2) is a transcriptional co-repressor that is associated with tumorigenesis and tumor progression. It has been reported to predict a poor prognosis in several human cancers, including esophageal squamous cell carcinoma (ESCC). The present study aimed to investigate the involvement of CtBP2 in the cisplatin (DDP) resistance of the ECA109 ESCC cell line and its effect on the expression of apoptosis-associated proteins. Constructed recombinant lentiviruses were used for the knockdown or overexpression of CtBP2 in ECA109 cells, and the expression of CtBP2 was measured using reverse transcription-quantitative polymerase chain reaction and western blotting following transfection. MTT assays, Hoechst 33342 staining and flow cytometry (FCM) were applied to detect the influence of CtBP2 on the DDP-induced viability and apoptosis of the transfected ECA109 cells. In addition, the levels of apoptosis-associated proteins, including p53, B‑cell lymphoma 2 (Bcl‑2), Bcl‑2‑associated X protein (Bax) and activated caspase-3 were investigated in the transfected ECA109 cells. Stable ECA109 cells with CtBP2 overexpression or knockdown were successfully established. The results of the MTT, Hoechst 33342 and FCM assays demonstrated that overexpression of CtBP2 attenuated the reduction of cell viability and inhibited the cell apoptosis induced by DDP. Furthermore, the western blotting results indicated that CtBP2 overexpression inhibited the DDP-induced apoptosis of ECA109 cells via the reduction of p53, activated caspase-3 and Bax expression, and promotion of Bcl‑2 expression. Therefore, the present study indicated that CtBP2 reduced the susceptibility of ECA109 cells to DDP by regulating the expression of apoptosis-related proteins, suggesting that it may be a promising therapeutic target in ESCC in the future.
Collapse
Affiliation(s)
- Hui Shi
- Laboratory Animals Center, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yinting Mao
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Qianqian Ju
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yingcheng Wu
- Medical College of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Wen Bai
- Medical College of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Peiwen Wang
- Medical College of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yudong Zhang
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Maorong Jiang
- Laboratory Animals Center, Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
23
|
Dcona MM, Morris BL, Ellis KC, Grossman SR. CtBP- an emerging oncogene and novel small molecule drug target: Advances in the understanding of its oncogenic action and identification of therapeutic inhibitors. Cancer Biol Ther 2017; 18:379-391. [PMID: 28532298 PMCID: PMC5536941 DOI: 10.1080/15384047.2017.1323586] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
C-terminal Binding Proteins (CtBP) 1 and 2 are oncogenic transcriptional co-regulators overexpressed in many cancer types, with their expression level correlating to worse prognostic outcomes and aggressive tumor features. CtBP negatively regulates the expression of many tumor suppressor genes, while coactivating genes that promote proliferation, epithelial-mesenchymal transition, and cancer stem cell self-renewal activity. In light of this evidence, the development of novel inhibitors that mitigate CtBP function may provide clinically actionable therapeutic tools. This review article focuses on the progress made in understanding CtBP structure, role in tumor progression, and discovery and development of CtBP inhibitors that target CtBP's dehydrogenase activity and other functions, with a focus on the theory and rationale behind the designs of current inhibitors. We provide insight into the future development and use of rational combination therapy that may further augment the efficacy of CtBP inhibitors, specifically addressing metastasis and cancer stem cell populations within tumors.
Collapse
Affiliation(s)
- M Michael Dcona
- a Department of Internal Medicine , Virginia Commonwealth University , Richmond , VA , USA
| | - Benjamin L Morris
- b Department of Human and Molecular Genetics , Virginia Commonwealth University , Richmond , VA , USA
| | - Keith C Ellis
- c Department of Medicinal Chemistry , Virginia Commonwealth University , Richmond , VA , USA.,d Institute for Structural Biology , Drug Discovery and Development, Virginia Commonwealth University , Richmond , VA , USA.,e VCU Massey Cancer Center , Virginia Commonwealth University , Richmond , VA , USA
| | - Steven R Grossman
- a Department of Internal Medicine , Virginia Commonwealth University , Richmond , VA , USA.,b Department of Human and Molecular Genetics , Virginia Commonwealth University , Richmond , VA , USA.,d Institute for Structural Biology , Drug Discovery and Development, Virginia Commonwealth University , Richmond , VA , USA.,e VCU Massey Cancer Center , Virginia Commonwealth University , Richmond , VA , USA
| |
Collapse
|