1
|
Hong J, Wu Y, Li M, Man KF, Song D, Koh SB. cAMP response element-binding protein: A credible cancer drug target. J Pharmacol Exp Ther 2025; 392:103529. [PMID: 40157009 PMCID: PMC12060161 DOI: 10.1016/j.jpet.2025.103529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 02/22/2025] [Indexed: 04/01/2025] Open
Abstract
Despite advancements in radiotherapy, chemotherapy, endocrine therapy, targeted therapy, and immunotherapy, resistance to therapy remains a pervasive challenge in oncology, in part owing to tumor heterogeneity. Identifying new therapeutic targets is key to addressing this challenge because it can both diversify and enhance existing treatment options, particularly through combination regimens. The cAMP response element-binding protein (CREB) is a transcription factor involved in various biological processes. It is aberrantly activated in several aggressive cancer types, including breast cancer. Clinically, high CREB expression is associated with increased breast tumor aggressiveness and poor prognosis. Functionally, CREB promotes breast cancer cell proliferation, survival, invasion, metastasis, as well as therapy resistance by deregulating genes related to apoptosis, cell cycle, and metabolism. Targeting CREB with small molecule inhibitors has demonstrated promise in preclinical studies. This review summarizes the current understanding of CREB mechanisms and their potential as a therapeutic target. SIGNIFICANCE STATEMENT: cAMP response element-binding protein (CREB) is a master regulator of multiple biological processes, including neurodevelopment, metabolic regulation, and immune response. CREB is a putative proto-oncogene in breast cancer that regulates the cell cycle, apoptosis, and cellular migration. Preclinical development of CREB-targeting small molecules is underway.
Collapse
Affiliation(s)
- Jinghui Hong
- Department of Breast Surgery, General Surgery Centre, The First Hospital of Jilin University, Changchun, Jilin, China; Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Yuheng Wu
- Department of Breast Surgery, General Surgery Centre, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Mengxin Li
- Department of Breast Surgery, General Surgery Centre, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Ki-Fong Man
- Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Dong Song
- Department of Breast Surgery, General Surgery Centre, The First Hospital of Jilin University, Changchun, Jilin, China.
| | - Siang-Boon Koh
- Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom; University Hospitals Bristol and Weston, National Health Service (NHS) Foundation Trust, Bristol, United Kingdom.
| |
Collapse
|
2
|
Kuravsky M, Kelly C, Redfield C, Shammas SL. The transition state for coupled folding and binding of a disordered DNA binding domain resembles the unbound state. Nucleic Acids Res 2024; 52:11822-11837. [PMID: 39315703 PMCID: PMC11514473 DOI: 10.1093/nar/gkae794] [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: 02/12/2024] [Revised: 08/22/2024] [Accepted: 09/02/2024] [Indexed: 09/25/2024] Open
Abstract
The basic zippers (bZIPs) are one of two large eukaryotic families of transcription factors whose DNA binding domains are disordered in isolation but fold into stable α-helices upon target DNA binding. Here, we systematically disrupt pre-existing helical propensity within the DNA binding region of the homodimeric bZIP domain of cAMP-response element binding protein (CREB) using Ala-Gly scanning and examine the impact on target binding kinetics. We find that the secondary structure of the transition state strongly resembles that of the unbound state. The residue closest to the dimerization domain is largely folded within both unbound and transition states; dimerization apparently propagates additional helical propensity into the basic region. The results are consistent with electrostatically-enhanced DNA binding, followed by rapid folding from the folded zipper outwards. Fly-casting theory suggests that protein disorder can accelerate binding. Interestingly however, we did not observe higher association rate constants for mutants with lower levels of residual structure in the unbound state.
Collapse
Affiliation(s)
- Mikhail Kuravsky
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
| | - Conor Kelly
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
| | | | - Sarah L Shammas
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
| |
Collapse
|
3
|
Chandra Jena B, Flaherty DP, O'Brien VP, Watts VJ. Biochemical pharmacology of adenylyl cyclases in cancer. Biochem Pharmacol 2024; 228:116160. [PMID: 38522554 PMCID: PMC11410551 DOI: 10.1016/j.bcp.2024.116160] [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: 01/20/2024] [Revised: 03/11/2024] [Accepted: 03/21/2024] [Indexed: 03/26/2024]
Abstract
Globally, despite extensive research and pharmacological advancement, cancer remains one of the most common causes of mortality. Understanding the signaling pathways involved in cancer progression is essential for the discovery of new drug targets. The adenylyl cyclase (AC) superfamily comprises glycoproteins that regulate intracellular signaling and convert ATP into cyclic AMP, an important second messenger. The present review highlights the involvement of ACs in cancer progression and suppression, broken down for each specific mammalian AC isoform. The precise mechanisms by which ACs contribute to cancer cell proliferation and invasion are not well understood and are variable among cancer types; however, AC overactivation, along with that of downstream regulators, presents a potential target for novel anticancer therapies. The expression patterns of ACs in numerous cancers are discussed. In addition, we highlight inhibitors of AC-related signaling that are currently under investigation, with a focus on possible anti-cancer strategies. Recent discoveries with small molecules regarding more direct modulation AC activity are also discussed in detail. A more comprehensive understanding of different components in AC-related signaling could potentially lead to the development of novel therapeutic strategies for personalized oncology and might enhance the efficacy of chemoimmunotherapy in the treatment of various cancers.
Collapse
Affiliation(s)
- Bikash Chandra Jena
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, USA
| | - Daniel P Flaherty
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, USA
| | - Valerie P O'Brien
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, USA.
| | - Val J Watts
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, USA.
| |
Collapse
|
4
|
Nayar JC, Abboud M, Dixon KM. Cyclic AMP-regulatory element-binding protein: a novel UV-targeted transcription factor in skin cancer. Photochem Photobiol Sci 2024; 23:1209-1215. [PMID: 38743195 DOI: 10.1007/s43630-024-00578-7] [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: 01/22/2024] [Accepted: 04/12/2024] [Indexed: 05/16/2024]
Abstract
Common therapeutics in relation to melanoma and non-melanoma cancers include the use of kinase inhibitors. The long-term benefits of kinases, however, are limited by development of drug resistance. An alternative approach for treatment would be to focus on transcription factors. Cyclic AMP-regulatory element-binding protein (CREB) is a transcription factor that is commonly overactivated or overexpressed in many different cancers including skin cancer. Ultraviolet radiation (UVR), one of the main causes of skin cancer, can activate CREB in both melanocytes and keratinocytes. In addition, CREB has been found to be activated in skin cancers. Considering the prominent role that CREB plays in skin cancers, the studies reviewed herein raise the possibility of CREB as a potential prognostic and diagnostic marker of skin cancer and a novel target for therapeutic intervention.
Collapse
Affiliation(s)
- Julianne C Nayar
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, 2050, Camperdown, NSW, Australia
| | - Myriam Abboud
- Department of Health, Zayed University, P.O. Box 144534, Abu Dhabi, United Arab Emirates
| | - Katie M Dixon
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, 2050, Camperdown, NSW, Australia.
| |
Collapse
|
5
|
Dinevska M, Widodo SS, Cook L, Stylli SS, Ramsay RG, Mantamadiotis T. CREB: A multifaceted transcriptional regulator of neural and immune function in CNS tumors. Brain Behav Immun 2024; 116:140-149. [PMID: 38070619 DOI: 10.1016/j.bbi.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 11/16/2023] [Accepted: 12/04/2023] [Indexed: 01/21/2024] Open
Abstract
Cancers of the central nervous system (CNS) are unique with respect to their tumor microenvironment. Such a status is due to immune-privilege and the cellular behaviors within a highly networked, neural-rich milieu. During tumor development in the CNS, neural, immune and cancer cells establish complex cell-to-cell communication networks which mimic physiological functions, including paracrine signaling and synapse-like formations. This crosstalk regulates diverse pathological functions contributing to tumor progression. In the CNS, regulation of physiological and pathological functions relies on various cell signaling and transcription programs. At the core of these events lies the cyclic adenosine monophosphate (cAMP) response element binding protein (CREB), a master transcriptional regulator in the CNS. CREB is a kinase inducible transcription factor which regulates many CNS functions, including neurogenesis, neuronal survival, neuronal activation and long-term memory. Here, we discuss how CREB-regulated mechanisms operating in diverse cell types, which control development and function of the CNS, are co-opted in CNS tumors.
Collapse
Affiliation(s)
- Marija Dinevska
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Samuel S Widodo
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Laura Cook
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Stanley S Stylli
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia; Department of Neurosurgery, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Robert G Ramsay
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology and the Department of Clinical Pathology, The University of Melbourne, Melbourne, Australia
| | - Theo Mantamadiotis
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia; Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia; Centre for Stem Cell Systems, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
6
|
Zhu Y, Wang Z, Li Y, Peng H, Liu J, Zhang J, Xiao X. The Role of CREBBP/EP300 and Its Therapeutic Implications in Hematological Malignancies. Cancers (Basel) 2023; 15:cancers15041219. [PMID: 36831561 PMCID: PMC9953837 DOI: 10.3390/cancers15041219] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/17/2023] Open
Abstract
Disordered histone acetylation has emerged as a key mechanism in promoting hematological malignancies. CREB-binding protein (CREBBP) and E1A-binding protein P300 (EP300) are two key acetyltransferases and transcriptional cofactors that regulate gene expression by regulating the acetylation levels of histone proteins and non-histone proteins. CREBBP/EP300 dysregulation and CREBBP/EP300-containing complexes are critical for the initiation, progression, and chemoresistance of hematological malignancies. CREBBP/EP300 also participate in tumor immune responses by regulating the differentiation and function of multiple immune cells. Currently, CREBBP/EP300 are attractive targets for drug development and are increasingly used as favorable tools in preclinical studies of hematological malignancies. In this review, we summarize the role of CREBBP/EP300 in normal hematopoiesis and highlight the pathogenic mechanisms of CREBBP/EP300 in hematological malignancies. Moreover, the research basis and potential future therapeutic implications of related inhibitors were also discussed from several aspects. This review represents an in-depth insight into the physiological and pathological significance of CREBBP/EP300 in hematology.
Collapse
Affiliation(s)
- Yu Zhu
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China
| | - Zi Wang
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China
| | - Yanan Li
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China
| | - Hongling Peng
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China
| | - Jing Liu
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China
| | - Ji Zhang
- The Affiliated Nanhua Hospital, Department of Clinical Laboratory, Hengyang Medical School, University of South China, Hengyang 421001, China
- Correspondence: (J.Z.); (X.X.); Tel.: +86-734-8279050 (J.Z.); +86-731-84805449 (X.X.)
| | - Xiaojuan Xiao
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China
- Correspondence: (J.Z.); (X.X.); Tel.: +86-734-8279050 (J.Z.); +86-731-84805449 (X.X.)
| |
Collapse
|
7
|
Bin T, Lin C, Liu FJ, Wang Y, Xu XJ, Lin DJ, Tang J, Lu B. Establishment of a risk model correlated with metabolism based on RNA-binding proteins associated with cell pyroptosis in acute myeloid leukemia. Front Oncol 2022; 12:1059978. [DOI: 10.3389/fonc.2022.1059978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 11/01/2022] [Indexed: 11/18/2022] Open
Abstract
BackgroundRNA-binding protein (RBP) regulates acute myeloid leukemia (AML) by participating in mRNA editing and modification. Pyroptosis also plays an immunomodulatory function in AML. Therefore, this study aimed to identify pyroptosis-related RBP genes that could predict the prognosis of AML patients.MethodsAML related expression data were downloaded from the UCSC website and Gene Expression Omnibus (GEO) database. Pyroptosis-RPB-related differentially expressed genes (PRBP-DEGs) were conducted with a protein-protein interactions (PPI) network to screen out the key PRBP-DEGs, based on which a risk model was constructed by Cox analysis, and evaluated by plotting Receiver operating characteristic (ROC) curves and survival curves. Independent prognostic analysis was performed and a nomogram was constructed. Finally, enrichment analysis was performed for high and low risk groups.ReusltsA total of 71 PRBP-DEGs were obtained and a pyroptosis-RPB-related risk model was constructed based on IFIT5, MRPL14, MRPL21, MRPL39, MVP, and PUSL1 acquired from Cox analysis. RiskScore, age, and cytogenetics risk category were identified as independent prognostic factors, and the nomogram based on these independent prognostic factors could accurately predict 1-, 3- and 5-year survival of AML patients. Gene set enrichment analysis (GSEA) showed that the high-risk and low-risk groups were mainly enriched in metabolic- and immune-related processes and pathways.ConclusionIn this study, a risk score model correlated with metabolism based on RNA-binding proteins associated with cell pyroptosis in acute myeloid leukemia was established, which provided a theoretical basis and reference value for therapeutic studies and prognosis of AML.
Collapse
|
8
|
Salavaty A, Shehni SA, Ramialison M, Currie PD. Systematic molecular profiling of acute leukemia cancer stem cells allows identification of druggable targets. Heliyon 2022; 8:e11093. [PMID: 36281397 PMCID: PMC9586918 DOI: 10.1016/j.heliyon.2022.e11093] [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: 08/23/2022] [Revised: 10/04/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022] Open
Abstract
Acute myeloid leukemia (AML) is one of the most prevalent and acute blood cancers with a poor prognosis and low overall survival rate, especially in the elderly. Although several new AML markers and drug targets have been recently identified, the rate of long-term cancer eradication has not improved significantly due to the presence and drug resistance of AML cancer stem cells (CSCs). Here we develop a novel computational pipeline to analyze the transcriptomic profiles of AML cancer (stem) cells and identify novel candidate AML CSC markers and drug targets. In our novel pipeline we apply a top-down meta-analysis strategy to integrate The Cancer Genome Atlas data with CSC datasets to infer cell stemness features. As a result, a set of genes termed the "AML key CSC genes" along with all the available drugs/compounds that could target them were identified. Overall, our novel computational pipeline could retrieve known cancer drugs (Carfilzomib) and predicted novel drugs such as Zonisamide, Amitriptyline, and their targets amongst the top ranked drugs and drug targets for targeting AML. Additionally, the pipeline applied in this study could be used for the identification of CSC-specific markers, drivers and their respective targeting drugs in other cancer types.
Collapse
Affiliation(s)
- Adrian Salavaty
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
- Systems Biology Institute Australia, Monash University, Clayton, VIC 3800, Australia
| | - Sara Alaei Shehni
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Mirana Ramialison
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
- Systems Biology Institute Australia, Monash University, Clayton, VIC 3800, Australia
- Novo Nordisk Foundation Center for Stem Cell Medicine, Murdoch Children's Research Institute, Royal Children's Hospital, Flemington Road, Parkville, VIC, 3052, Australia
- Department of Pediatrics, The Royal Children's Hospital, University of Melbourne Parkville, VIC, 3052, Australia
| | - Peter D. Currie
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
- EMBL Australia, Monash University, Clayton, VIC 3800, Australia
| |
Collapse
|
9
|
Ahmed MB, Alghamdi AAA, Islam SU, Lee JS, Lee YS. cAMP Signaling in Cancer: A PKA-CREB and EPAC-Centric Approach. Cells 2022; 11:cells11132020. [PMID: 35805104 PMCID: PMC9266045 DOI: 10.3390/cells11132020] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/17/2022] [Accepted: 06/23/2022] [Indexed: 02/01/2023] Open
Abstract
Cancer is one of the most common causes of death globally. Despite extensive research and considerable advances in cancer therapy, the fundamentals of the disease remain unclear. Understanding the key signaling mechanisms that cause cancer cell malignancy may help to uncover new pharmaco-targets. Cyclic adenosine monophosphate (cAMP) regulates various biological functions, including those in malignant cells. Understanding intracellular second messenger pathways is crucial for identifying downstream proteins involved in cancer growth and development. cAMP regulates cell signaling and a variety of physiological and pathological activities. There may be an impact on gene transcription from protein kinase A (PKA) as well as its downstream effectors, such as cAMP response element-binding protein (CREB). The position of CREB downstream of numerous growth signaling pathways implies its oncogenic potential in tumor cells. Tumor growth is associated with increased CREB expression and activation. PKA can be used as both an onco-drug target and a biomarker to find, identify, and stage tumors. Exploring cAMP effectors and their downstream pathways in cancer has become easier using exchange protein directly activated by cAMP (EPAC) modulators. This signaling system may inhibit or accelerate tumor growth depending on the tumor and its environment. As cAMP and its effectors are critical for cancer development, targeting them may be a useful cancer treatment strategy. Moreover, by reviewing the material from a distinct viewpoint, this review aims to give a knowledge of the impact of the cAMP signaling pathway and the related effectors on cancer incidence and development. These innovative insights seek to encourage the development of novel treatment techniques and new approaches.
Collapse
Affiliation(s)
- Muhammad Bilal Ahmed
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea; (M.B.A.); (J.-S.L.)
| | | | - Salman Ul Islam
- Department of Pharmacy, Cecos University, Peshawar, Street 1, Sector F 5 Phase 6 Hayatabad, Peshawar 25000, Pakistan;
| | - Joon-Seok Lee
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea; (M.B.A.); (J.-S.L.)
| | - Young-Sup Lee
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea; (M.B.A.); (J.-S.L.)
- Correspondence: ; Tel.: +82-53-950-6353; Fax: +82-53-943-2762
| |
Collapse
|
10
|
Zhang K, Horikoshi N, Li S, Powers AS, Hameedi MA, Pintilie GD, Chae HD, Khan YA, Suomivuori CM, Dror RO, Sakamoto KM, Chiu W, Wakatsuki S. Cryo-EM, Protein Engineering, and Simulation Enable the Development of Peptide Therapeutics against Acute Myeloid Leukemia. ACS CENTRAL SCIENCE 2022; 8:214-222. [PMID: 35233453 PMCID: PMC8875425 DOI: 10.1021/acscentsci.1c01090] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Indexed: 06/14/2023]
Abstract
Cryogenic electron microscopy (cryo-EM) has emerged as a viable structural tool for molecular therapeutics development against human diseases. However, it remains a challenge to determine structures of proteins that are flexible and smaller than 30 kDa. The 11 kDa KIX domain of CREB-binding protein (CBP), a potential therapeutic target for acute myeloid leukemia and other cancers, is a protein which has defied structure-based inhibitor design. Here, we develop an experimental approach to overcome the size limitation by engineering a protein double-shell to sandwich the KIX domain between apoferritin as the inner shell and maltose-binding protein as the outer shell. To assist homogeneous orientations of the target, disulfide bonds are introduced at the target-apoferritin interface, resulting in a cryo-EM structure at 2.6 Å resolution. We used molecular dynamics simulations to design peptides that block the interaction of the KIX domain of CBP with the intrinsically disordered pKID domain of CREB. The double-shell design allows for fluorescence polarization assays confirming the binding between the KIX domain in the double-shell and these interacting peptides. Further cryo-EM analysis reveals a helix-helix interaction between a single KIX helix and the best peptide, providing a possible strategy for developments of next-generation inhibitors.
Collapse
Affiliation(s)
- Kaiming Zhang
- MOE
Key Laboratory for Cellular Dynamics and Division of Life Sciences
and Medicine, University of Science and
Technology of China, Hefei 230027, China
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
| | - Naoki Horikoshi
- Life
Science Center for Survival Dynamics, University
of Tsukuba, Tsukuba, Ibaraki 305-8577, Japan
- Department
of Structural Biology, Stanford University, Stanford, California 94305, United States
| | - Shanshan Li
- MOE
Key Laboratory for Cellular Dynamics and Division of Life Sciences
and Medicine, University of Science and
Technology of China, Hefei 230027, China
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
| | - Alexander S. Powers
- Department
of Chemistry, Stanford University, Stanford, California 94305, United States
- Department
of Computer Science, Stanford University, Stanford, California 94305, United States
| | - Mikhail A. Hameedi
- Department
of Structural Biology, Stanford University, Stanford, California 94305, United States
- Biosciences
Division, SLAC National Accelerator Laboratory, Stanford University, Menlo
Park, California 94025, United States
| | - Grigore D. Pintilie
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
| | - Hee-Don Chae
- Department
of Pediatrics, Stanford University School
of Medicine, Stanford, California 94305, United States
| | - Yousuf A. Khan
- Department
of Computer Science, Stanford University, Stanford, California 94305, United States
- Department
of Molecular and Cellular Physiology, Stanford
University School of Medicine, Stanford, California 94305, United States
| | - Carl-Mikael Suomivuori
- Department
of Computer Science, Stanford University, Stanford, California 94305, United States
| | - Ron O. Dror
- Department
of Computer Science, Stanford University, Stanford, California 94305, United States
| | - Kathleen M. Sakamoto
- Department
of Pediatrics, Stanford University School
of Medicine, Stanford, California 94305, United States
| | - Wah Chiu
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
- CryoEM
and Bioimaging Division, Stanford Synchrotron Radiation Lightsource,
SLAC National Accelerator Laboratory, Stanford
University, Menlo
Park, California 94025, United States
| | - Soichi Wakatsuki
- Department
of Structural Biology, Stanford University, Stanford, California 94305, United States
- Biosciences
Division, SLAC National Accelerator Laboratory, Stanford University, Menlo
Park, California 94025, United States
| |
Collapse
|
11
|
Gutierrez DA, Contreras L, Villanueva PJ, Borrego EA, Morán-Santibañez K, Hess JD, DeJesus R, Larragoity M, Betancourt AP, Mohl JE, Robles-Escajeda E, Begum K, Roy S, Kirken RA, Varela-Ramirez A, Aguilera RJ. Identification of a Potent Cytotoxic Pyrazole with Anti-Breast Cancer Activity That Alters Multiple Pathways. Cells 2022; 11:254. [PMID: 35053370 PMCID: PMC8773755 DOI: 10.3390/cells11020254] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/14/2021] [Accepted: 01/07/2022] [Indexed: 11/26/2022] Open
Abstract
In this study, we identified a novel pyrazole-based derivative (P3C) that displayed potent cytotoxicity against 27 human cancer cell lines derived from different tissue origins with 50% cytotoxic concentrations (CC50) in the low micromolar and nanomolar range, particularly in two triple-negative breast cancer (TNBC) cell lines (from 0.25 to 0.49 µM). In vitro assays revealed that P3C induces reactive oxygen species (ROS) accumulation leading to mitochondrial depolarization and caspase-3/7 and -8 activation, suggesting the participation of both the intrinsic and extrinsic apoptotic pathways. P3C caused microtubule disruption, phosphatidylserine externalization, PARP cleavage, DNA fragmentation, and cell cycle arrest on TNBC cells. In addition, P3C triggered dephosphorylation of CREB, p38, ERK, STAT3, and Fyn, and hyperphosphorylation of JNK and NF-kB in TNBC cells, indicating the inactivation of both p38MAPK/STAT3 and ERK1/2/CREB signaling pathways. In support of our in vitro assays, transcriptome analyses of two distinct TNBC cell lines (MDA-MB-231 and MDA-MB-468 cells) treated with P3C revealed 28 genes similarly affected by the treatment implicated in apoptosis, oxidative stress, protein kinase modulation, and microtubule stability.
Collapse
Affiliation(s)
- Denisse A. Gutierrez
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Lisett Contreras
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Paulina J. Villanueva
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Edgar A. Borrego
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Karla Morán-Santibañez
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Jessica D. Hess
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Rebecca DeJesus
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Manuel Larragoity
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Ana P. Betancourt
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Jonathon E. Mohl
- Department of Bioinformatics, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA;
| | - Elisa Robles-Escajeda
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Khodeza Begum
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Sourav Roy
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Robert A. Kirken
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Armando Varela-Ramirez
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Renato J. Aguilera
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| |
Collapse
|
12
|
Bolandi SM, Pakjoo M, Beigi P, Kiani M, Allahgholipour A, Goudarzi N, Khorashad JS, Eiring AM. A Role for the Bone Marrow Microenvironment in Drug Resistance of Acute Myeloid Leukemia. Cells 2021; 10:2833. [PMID: 34831055 PMCID: PMC8616250 DOI: 10.3390/cells10112833] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 02/08/2023] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease with a poor prognosis and remarkable resistance to chemotherapeutic agents. Understanding resistance mechanisms against currently available drugs helps to recognize the therapeutic obstacles. Various mechanisms of resistance to chemotherapy or targeted inhibitors have been described for AML cells, including a role for the bone marrow niche in both the initiation and persistence of the disease, and in drug resistance of the leukemic stem cell (LSC) population. The BM niche supports LSC survival through direct and indirect interactions among the stromal cells, hematopoietic stem/progenitor cells, and leukemic cells. Additionally, the BM niche mediates changes in metabolic and signal pathway activation due to the acquisition of new mutations or selection and expansion of a minor clone. This review briefly discusses the role of the BM microenvironment and metabolic pathways in resistance to therapy, as discovered through AML clinical studies or cell line and animal models.
Collapse
Affiliation(s)
- Seyed Mohammadreza Bolandi
- Department of Immunology, Razi Vaccine and Sera Research Institute, Karaj, Iran; (S.M.B.); (N.G.)
- Department of Pharmacology, Karaj Branch, Islamic Azad University, Karaj, Iran; (M.K.); (A.A.)
| | - Mahdi Pakjoo
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; (M.P.); (P.B.)
| | - Peyman Beigi
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; (M.P.); (P.B.)
| | - Mohammad Kiani
- Department of Pharmacology, Karaj Branch, Islamic Azad University, Karaj, Iran; (M.K.); (A.A.)
| | - Ali Allahgholipour
- Department of Pharmacology, Karaj Branch, Islamic Azad University, Karaj, Iran; (M.K.); (A.A.)
| | - Negar Goudarzi
- Department of Immunology, Razi Vaccine and Sera Research Institute, Karaj, Iran; (S.M.B.); (N.G.)
| | - Jamshid S. Khorashad
- Centre for Haematology, Hammersmith Hospital, Imperial College London, London W12 0HS, UK;
| | - Anna M. Eiring
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center at El Paso, El Paso, TX 79905, USA
| |
Collapse
|
13
|
Wang Z, Zhang X, Tian X, Yang Y, Ma L, Wang J, Yu Y. CREB stimulates GPX4 transcription to inhibit ferroptosis in lung adenocarcinoma. Oncol Rep 2021; 45:88. [PMID: 33846793 PMCID: PMC8042667 DOI: 10.3892/or.2021.8039] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 02/15/2021] [Indexed: 12/11/2022] Open
Abstract
Ferroptosis is a new form of regulated cell death and closely related to cancer. However, the mechanism underlying the regulation of ferroptosis in lung adenocarcinoma (LUAD) remains unclear. IB, IHC and ELISA were performed to analyze protein expression. RT-qPCR was used to analyze mRNA expression. Cell viability, 3D cell growth, MDA, the generation of lipid ROS and the Fe2+ concentration were measured to evaluate the responses to the induction of ferroptosis. Measurement of luciferase activity and ChIP were used to analyze the promoter activity regulated by the transcriptional regulator. Co-IP assays were performed to identify protein-protein interactions. In the present study, it was revealed that cAMP response element-binding protein (CREB) was highly expressed in LUAD, and knockdown of CREB inhibited cell viability and growth by promoting apoptosis- and ferroptosis-like cell death, concurrently. It was observed that CREB suppressed lipid peroxidation by binding the promoter region of glutathione peroxidase 4 (GPX4), and this binding could be enhanced by E1A binding protein P300 (EP300). The bZIP domain in CREB and the CBP/p300-HAT domain in EP300 were essential for CREB-EP300 binding in LUAD cells. Finally, it was revealed that CREB, GPX4, EP300 and 4-HNE were closely related to tumor size and stage, and tumors with a higher degree of malignancy were more likely to have a low degree of lipid peroxidation. Therefore, targeting this CREB/EP300/GPX4 axis may provide new strategies for treating LUAD.
Collapse
Affiliation(s)
- Zhixian Wang
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Xiao Zhang
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Xiaoting Tian
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Yueyue Yang
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, P.R. China
| | - Lifang Ma
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Jiayi Wang
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Yongchun Yu
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| |
Collapse
|
14
|
Zhang H, Kong Q, Wang J, Jiang Y, Hua H. Complex roles of cAMP-PKA-CREB signaling in cancer. Exp Hematol Oncol 2020; 9:32. [PMID: 33292604 PMCID: PMC7684908 DOI: 10.1186/s40164-020-00191-1] [Citation(s) in RCA: 276] [Impact Index Per Article: 55.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 11/19/2020] [Indexed: 02/08/2023] Open
Abstract
Cyclic adenosine monophosphate (cAMP) is the first discovered second messenger, which plays pivotal roles in cell signaling, and regulates many physiological and pathological processes. cAMP can regulate the transcription of various target genes, mainly through protein kinase A (PKA) and its downstream effectors such as cAMP-responsive element binding protein (CREB). In addition, PKA can phosphorylate many kinases such as Raf, GSK3 and FAK. Aberrant cAMP-PKA signaling is involved in various types of human tumors. Especially, cAMP signaling may have both tumor-suppressive and tumor-promoting roles depending on the tumor types and context. cAMP-PKA signaling can regulate cancer cell growth, migration, invasion and metabolism. This review highlights the important roles of cAMP-PKA-CREB signaling in tumorigenesis. The potential strategies to target this pathway for cancer therapy are also discussed.
Collapse
Affiliation(s)
- Hongying Zhang
- Laboratory of Oncogene, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Qingbin Kong
- Laboratory of Oncogene, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jiao Wang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yangfu Jiang
- Laboratory of Oncogene, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Hui Hua
- Laboratory of Stem Cell Biology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
15
|
Steven A, Friedrich M, Jank P, Heimer N, Budczies J, Denkert C, Seliger B. What turns CREB on? And off? And why does it matter? Cell Mol Life Sci 2020; 77:4049-4067. [PMID: 32347317 PMCID: PMC7532970 DOI: 10.1007/s00018-020-03525-8] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 03/21/2020] [Accepted: 04/06/2020] [Indexed: 12/16/2022]
Abstract
Altered expression and function of the transcription factor cyclic AMP response-binding protein (CREB) has been identified to play an important role in cancer and is associated with the overall survival and therapy response of tumor patients. This review focuses on the expression and activation of CREB under physiologic conditions and in tumors of distinct origin as well as the underlying mechanisms of CREB regulation by diverse stimuli and inhibitors. In addition, the clinical relevance of CREB is summarized, including its use as a prognostic and/or predictive marker as well as a therapeutic target.
Collapse
Affiliation(s)
- André Steven
- Institute for Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Michael Friedrich
- Institute for Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Paul Jank
- Institute of Pathology, Philipps University Marburg, 35043, Marburg, Germany
| | - Nadine Heimer
- Institute for Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Jan Budczies
- Institute of Pathology, University Clinic Heidelberg, 69120, Heidelberg, Germany
| | - Carsten Denkert
- Institute of Pathology, Philipps University Marburg, 35043, Marburg, Germany
| | - Barbara Seliger
- Institute for Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany.
| |
Collapse
|
16
|
Illiano M, Conte M, Salzillo A, Ragone A, Spina A, Nebbioso A, Altucci L, Sapio L, Naviglio S. The KDM Inhibitor GSKJ4 Triggers CREB Downregulation via a Protein Kinase A and Proteasome-Dependent Mechanism in Human Acute Myeloid Leukemia Cells. Front Oncol 2020; 10:799. [PMID: 32582541 PMCID: PMC7289982 DOI: 10.3389/fonc.2020.00799] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 04/23/2020] [Indexed: 01/02/2023] Open
Abstract
Acute myeloid leukemia (AML) is a progressive hematopoietic-derived cancer arising from stepwise genetic mutations of the myeloid lineage. cAMP response element-binding protein (CREB) is a nuclear transcription factor, which plays a key role in the multistep process of leukemogenesis, thus emerging as an attractive potential drug target for AML treatment. Since epigenetic dysregulations, such as DNA methylation, histone modifications, as well as chromatin remodeling, are a frequent occurrence in AML, an increasing and selective number of epi-drugs are emerging as encouraging therapeutic agents. Here, we demonstrate that the histone lysine demethylases (KDMs) JMJD3/UTX inhibitor GSKJ4 results in both proliferation decrease and CREB protein downregulation in AML cells. We found that GSKJ4 clearly decreases CREB protein, but not CREB mRNA levels. By cycloheximide assay, we provide evidence that GSKJ4 reduces CREB protein stability; moreover, proteasome inhibition largely counteracts the GSKJ4-induced CREB downregulation. Very interestingly, a rapid CREB phosphorylation at the Ser133 residue precedes CREB protein decrease in response to GSKJ4 treatment. In addition, protein kinase A (PKA) inhibition, but not extracellular signal-regulated kinase (ERK)1/2 inhibition, almost completely prevents both GSKJ4-induced p-Ser133-CREB phosphorylation and CREB protein downregulation. Overall, our study enforces the evidence regarding CREB as a potential druggable target, identifies the small epigenetic molecule GSKJ4 as an "inhibitor" of CREB, and encourages the design of future GSKJ4-based studies for the development of innovative approaches for AML therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Luigi Sapio
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Silvio Naviglio
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
17
|
He B, Yang N, Man CH, Ng NK, Cher C, Leung H, Kan LL, Cheng BY, Lam SS, Wang ML, Zhang C, Kwok H, Cheng G, Sharma R, Ma AC, So CE, Kwong Y, Leung AY. Follistatin is a novel therapeutic target and biomarker in FLT3/ITD acute myeloid leukemia. EMBO Mol Med 2020; 12:e10895. [PMID: 32134197 PMCID: PMC7136967 DOI: 10.15252/emmm.201910895] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 02/07/2020] [Accepted: 02/14/2020] [Indexed: 12/12/2022] Open
Abstract
Internal tandem duplication of Fms-like tyrosine kinase 3 (FLT3/ITD) occurs in about 30% of acute myeloid leukemia (AML) and is associated with poor response to conventional treatment and adverse outcome. Here, we reported that human FLT3/ITD expression led to axis duplication and dorsalization in about 50% of zebrafish embryos. The morphologic phenotype was accompanied by ectopic expression of a morphogen follistatin (fst) during early embryonic development. Increase in fst expression also occurred in adult FLT3/ITD-transgenic zebrafish, Flt3/ITD knock-in mice, and human FLT3/ITD AML cells. Overexpression of human FST317 and FST344 isoforms enhanced clonogenicity and leukemia engraftment in xenotransplantation model via RET, IL2RA, and CCL5 upregulation. Specific targeting of FST by shRNA, CRISPR/Cas9, or antisense oligo inhibited leukemic growth in vitro and in vivo. Importantly, serum FST positively correlated with leukemia engraftment in FLT3/ITD AML patient-derived xenograft mice and leukemia blast percentage in primary AML patients. In FLT3/ITD AML patients treated with FLT3 inhibitor quizartinib, serum FST levels correlated with clinical response. These observations supported FST as a novel therapeutic target and biomarker in FLT3/ITD AML.
Collapse
Affiliation(s)
- Bai‐Liang He
- Division of HematologyDepartment of MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
- Guangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated HospitalSun Yat‐sen UniversityZhuhaiGuangdong ProvinceChina
| | - Ning Yang
- Division of HematologyDepartment of MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
| | - Cheuk Him Man
- Division of HematologyDepartment of MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
| | - Nelson Ka‐Lam Ng
- Division of HematologyDepartment of MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
| | - Chae‐Yin Cher
- Division of HematologyDepartment of MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
| | - Ho‐Ching Leung
- Division of HematologyDepartment of MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
| | - Leo Lai‐Hok Kan
- Division of HematologyDepartment of MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
| | - Bowie Yik‐Ling Cheng
- Division of HematologyDepartment of MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
| | - Stephen Sze‐Yuen Lam
- Division of HematologyDepartment of MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
| | - Michelle Lu‐Lu Wang
- Division of HematologyDepartment of MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
| | - Chun‐Xiao Zhang
- Division of HematologyDepartment of MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
| | - Hin Kwok
- Centre for Genomic SciencesThe University of Hong KongHong Kong SARChina
| | - Grace Cheng
- Centre for Genomic SciencesThe University of Hong KongHong Kong SARChina
| | - Rakesh Sharma
- Centre for Genomic SciencesThe University of Hong KongHong Kong SARChina
| | - Alvin Chun‐Hang Ma
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHong Kong SARChina
| | - Chi‐Wai Eric So
- Leukemia and Stem Cell Biology GroupDivision of Cancer StudiesDepartment of Hematological MedicineKing's College LondonLondonUK
| | - Yok‐Lam Kwong
- Division of HematologyDepartment of MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
| | - Anskar Yu‐Hung Leung
- Division of HematologyDepartment of MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
| |
Collapse
|
18
|
Perez DR, Sklar LA, Chigaev A, Matlawska-Wasowska K. Drug repurposing for targeting cyclic nucleotide transporters in acute leukemias - A missed opportunity. Semin Cancer Biol 2020; 68:199-208. [PMID: 32044470 DOI: 10.1016/j.semcancer.2020.02.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/01/2019] [Accepted: 02/03/2020] [Indexed: 02/08/2023]
Abstract
While current treatment regimens for acute leukemia can dramatically improve patient survival, there remains room for improvement. Due to its roles in cell differentiation, cell survival, and apoptotic signaling, modulation of the cyclic AMP (cAMP) pathway has provided a meaningful target in hematological malignancies. Several studies have demonstrated that gene expression profiles associated with increased pro-survival cAMP activity or downregulation of various pro-apoptotic factors associated with the cAMP pathway are apparent in acute leukemia patients. Previous work to increase leukemia cell intracellular cAMP focused on the use of cAMP analogs, stimulating cAMP production via transmembrane-associated adenylyl cyclases, or decreasing cAMP degradation by inhibiting phosphodiesterase activity. However, targeting cyclic nucleotide efflux by ATP-binding cassette (ABC) transporters represents an unexplored approach for modulation of intracellular cyclic nucleotide levels. Preliminary studies have shown that inhibition of cAMP efflux can stimulate leukemia cell differentiation, cell growth arrest, and apoptosis, indicating that targeting cAMP efflux may show promise for future therapeutic development. Furthermore, inhibition of cyclic nucleotide transporter activity may also contribute multiple anticancer benefits by reducing extracellular pro-survival signaling in malignant cells. Hence, several opportunities for drug repurposing may exist for targeting cyclic nucleotide transporters.
Collapse
Affiliation(s)
- Dominique R Perez
- Department of Pathology, Health Sciences Center, University of New Mexico, Albuquerque, NM, USA; Center for Molecular Discovery, Health Sciences Center, University of New Mexico, Albuquerque, NM, USA
| | - Larry A Sklar
- Department of Pathology, Health Sciences Center, University of New Mexico, Albuquerque, NM, USA; Center for Molecular Discovery, Health Sciences Center, University of New Mexico, Albuquerque, NM, USA; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
| | - Alexandre Chigaev
- Department of Pathology, Health Sciences Center, University of New Mexico, Albuquerque, NM, USA; Center for Molecular Discovery, Health Sciences Center, University of New Mexico, Albuquerque, NM, USA; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA.
| | - Ksenia Matlawska-Wasowska
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA; Department of Pediatrics, Division of Hematology-Oncology, Health Sciences Center, University of New Mexico, Albuquerque, NM, USA.
| |
Collapse
|
19
|
Chae HD, Cox N, Capolicchio S, Lee JW, Horikoshi N, Kam S, Ng AA, Edwards J, Butler TL, Chan J, Lee Y, Potter G, Capece MC, Liu CW, Wakatsuki S, Smith M, Sakamoto KM. SAR optimization studies on modified salicylamides as a potential treatment for acute myeloid leukemia through inhibition of the CREB pathway. Bioorg Med Chem Lett 2019; 29:2307-2315. [PMID: 31253529 DOI: 10.1016/j.bmcl.2019.06.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/12/2019] [Accepted: 06/17/2019] [Indexed: 10/26/2022]
Abstract
Disruption of cyclic adenosine monophosphate response element binding protein (CREB) provides a potential new strategy to address acute leukemia, a disease associated with poor prognosis, and for which conventional treatment options often carry a significant risk of morbidity and mortality. We describe the structure-activity relationships (SAR) for a series of XX-650-23 derived from naphthol AS-E phosphate that disrupts binding and activation of CREB by the CREB-binding protein (CBP). Through the development of this series, we identified several salicylamides that are potent inhibitors of acute leukemia cell viability through inhibition of CREB-CBP interaction. Among them, a biphenyl salicylamide, compound 71, was identified as a potent inhibitor of CREB-CBP interaction with improved physicochemical properties relative to previously described derivatives of naphthol AS-E phosphate.
Collapse
Affiliation(s)
- Hee-Don Chae
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Nick Cox
- Medicinal Chemistry Knowledge Center, Stanford ChEM-H, Stanford, CA, USA; Presently at Novo Nordisk Research Center Seattle, Inc., USA
| | | | - Jae Wook Lee
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Naoki Horikoshi
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA; Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Sharon Kam
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Andrew A Ng
- Medicinal Chemistry Knowledge Center, Stanford ChEM-H, Stanford, CA, USA
| | - Jeffrey Edwards
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Tae-León Butler
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Justin Chan
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Yvonne Lee
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Garrett Potter
- Medicinal Chemistry Knowledge Center, Stanford ChEM-H, Stanford, CA, USA
| | - Mark C Capece
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Corey W Liu
- Macromolecular Structure Knowledge Center, Stanford ChEM-H, Stanford, CA, USA
| | - Soichi Wakatsuki
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA; BioSciences Division, SLAC National Accelerator Laboratory, Menlo Park, CA, USA
| | - Mark Smith
- Medicinal Chemistry Knowledge Center, Stanford ChEM-H, Stanford, CA, USA.
| | - Kathleen M Sakamoto
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
20
|
Understanding and exploiting cell signalling convergence nodes and pathway cross-talk in malignant brain cancer. Cell Signal 2019; 57:2-9. [PMID: 30710631 DOI: 10.1016/j.cellsig.2019.01.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 01/29/2019] [Accepted: 01/29/2019] [Indexed: 01/03/2023]
Abstract
In cancer, complex intracellular and intercellular signals constantly evolve for the advantage of the tumour cells but to the disadvantage of the whole organism. Decades of intensive research have revealed the critical roles of cellular signalling pathways in regulating complex cell behaviours which influence tumour development, growth and therapeutic response, and ultimately patient outcome. Most studies have focussed on specific pathways and the resulting tumour cell function in a rather linear fashion, partly due to the available methodologies and partly due to the traditionally reductionist approach to research. Advances in cancer research, including genomic technologies have led to a deep appreciation of the complex signals and pathway interactions operating in tumour cells. In this review we examine the role and interaction of three major cell signalling pathways, PI3K, MAPK and cAMP, in regulating tumour cell functions and discuss the prospects for exploiting this knowledge to better treat difficult to treat cancers, using glioblastoma, the most common and deadly malignant brain cancer, as the example disease.
Collapse
|
21
|
Duque-Afonso J, Lin CH, Han K, Morgens DW, Jeng EE, Weng Z, Jeong J, Wong SHK, Zhu L, Wei MC, Chae HD, Schrappe M, Cario G, Duyster J, Xiao X, Sakamoto KM, Bassik MC, Cleary ML. CBP Modulates Sensitivity to Dasatinib in Pre-BCR + Acute Lymphoblastic Leukemia. Cancer Res 2018; 78:6497-6508. [PMID: 30262461 PMCID: PMC6283070 DOI: 10.1158/0008-5472.can-18-1703] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 08/18/2018] [Accepted: 09/24/2018] [Indexed: 01/18/2023]
Abstract
Dasatinib is a multi-tyrosine kinase inhibitor approved for treatment of Ph+ acute lymphoblastic leukemia (ALL), but its efficacy is limited by resistance. Recent preclinical studies suggest that dasatinib may be a candidate therapy in additional ALL subtypes including pre-BCR+ ALL. Here we utilized shRNA library screening and global transcriptomic analysis to identify several novel genes and pathways that may enhance dasatinib efficacy or mitigate potential resistance in human pre-BCR+ ALL. Depletion of the transcriptional coactivator CBP increased dasatinib sensitivity by downregulating transcription of the pre-BCR signaling pathway previously associated with dasatinib sensitivity. Acquired resistance was due, in part, to upregulation of alternative pathways including WNT through a mechanism, suggesting transcriptional plasticity. Small molecules that disrupt CBP interactions with the CREB KID domain or β-catenin showed promising preclinical efficacy in combination with dasatinib. These findings highlight novel modulators of sensitivity to targeted therapies in human pre-BCR+ ALL, which can be reversed by small-molecule inhibitors. They also identify promising therapeutic approaches to ameliorate dasatinib sensitivity and prevent resistance in ALL.Significance: These findings reveal mechanisms that modulate sensitivity to dasatinib and suggest therapeutic strategies to improve the outcome of patients with acute lymphoblastic leukemia.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/22/6497/F1.large.jpg Cancer Res; 78(22); 6497-508. ©2018 AACR.
Collapse
Affiliation(s)
- Jesús Duque-Afonso
- Department of Pathology, Stanford University School of Medicine, Stanford, California
- Department of Hematology and Oncology, University Medical Center Freiburg, Freiburg, Germany
| | - Chiou-Hong Lin
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Kyuho Han
- Department of Genetics, Stanford University School of Medicine, Stanford, California
| | - David W Morgens
- Department of Genetics, Stanford University School of Medicine, Stanford, California
| | - Edwin E Jeng
- Department of Genetics, Stanford University School of Medicine, Stanford, California
| | - Ziming Weng
- Department of Pathology, Stanford University School of Medicine, Stanford, California
- Stanford Center for Genomics and Personalized Medicine, Stanford, California
| | - Johan Jeong
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Stephen Hon Kit Wong
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Li Zhu
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Michael C Wei
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Hee-Don Chae
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Martin Schrappe
- Department of Pediatrics, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Gunnar Cario
- Department of Pediatrics, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Justus Duyster
- Department of Hematology and Oncology, University Medical Center Freiburg, Freiburg, Germany
| | - Xiangshu Xiao
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon
| | - Kathleen M Sakamoto
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Michael C Bassik
- Department of Genetics, Stanford University School of Medicine, Stanford, California
| | - Michael L Cleary
- Department of Pathology, Stanford University School of Medicine, Stanford, California.
| |
Collapse
|
22
|
Lodge JM, Majmudar CY, Clayton J, Mapp AK. Covalent Chemical Cochaperones of the p300/CBP GACKIX Domain. Chembiochem 2018; 19:1907-1912. [PMID: 29939485 PMCID: PMC10900128 DOI: 10.1002/cbic.201800173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Indexed: 12/28/2022]
Abstract
The GACKIX activator binding domain has been a compelling target for small-molecule probe discovery because of the central role of activator-GACKIX complexes in diseases ranging from leukemia to memory disorders. Additionally, GACKIX is an ideal model to dissect the context-dependent function of activator-coactivator complexes. However, the dynamic and transient protein-protein interactions (PPIs) formed by GACKIX are difficult targets for small molecules. An additional complication is that activator-binding motifs, such as GACKIX, are found in multiple coactivators, making specificity difficult to attain. In this study, we demonstrate that the strategy of tethering can be used to rapidly discover highly specific covalent modulators of the dynamic PPIs between activators and coactivators. These serve as both ortho- and allosteric modulators, enabling the tunable assembly or disassembly of the activator-coactivator complexes formed between the KIX domain and its cognate activator binding partners MLL and CREB. The molecules maintain their function and selectivity, even in human cell lysates and in bacterial cells, and thus, will ultimately be highly useful probes for cellular studies.
Collapse
Affiliation(s)
- Jean M Lodge
- University of Michigan, Life Sciences Institute, 210 Washentaw Avenue, Ann Arbor, MI, 48109, USA
| | - Chinmay Y Majmudar
- University of Michigan, Life Sciences Institute, 210 Washentaw Avenue, Ann Arbor, MI, 48109, USA
| | - James Clayton
- University of Michigan, Life Sciences Institute, 210 Washentaw Avenue, Ann Arbor, MI, 48109, USA
| | - Anna K Mapp
- University of Michigan, Life Sciences Institute, 210 Washentaw Avenue, Ann Arbor, MI, 48109, USA
| |
Collapse
|
23
|
Tolerance to sustained activation of the cAMP/Creb pathway activity in osteoblastic cells is enabled by loss of p53. Cell Death Dis 2018; 9:844. [PMID: 30154459 PMCID: PMC6113249 DOI: 10.1038/s41419-018-0944-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 07/26/2018] [Accepted: 08/02/2018] [Indexed: 12/15/2022]
Abstract
The loss of p53 function is a central event in the genesis of osteosarcoma (OS). How mutation of p53 enables OS development from osteoblastic lineage cells is poorly understood. We and others have reported a key role for elevated and persistent activation of the cAMP/PKA/Creb1 pathway in maintenance of OS. In view of the osteoblast lineage being the cell of origin of OS, we sought to determine how these pathways interact within the context of the normal osteoblast. Normal osteoblasts (p53 WT) rapidly underwent apoptosis in response to acute elevation of cAMP levels or activity, whereas p53-deficient osteoblasts tolerated this aberrant cAMP/Creb level and activity. Using the p53 activating small-molecule Nutlin-3a and cAMP/Creb1 activator forskolin, we addressed the question of how p53 responds to the activation of cAMP. We observed that p53 acts dominantly to protect cells from excessive cAMP accumulation. We identify a Creb1-Cbp complex that functions together with and interacts with p53. Finally, translating these results we find that a selective small-molecule inhibitor of the Creb1-Cbp interaction demonstrates selective toxicity to OS cells where this pathway is constitutively active. This highlights the cAMP/Creb axis as a potentially actionable therapeutic vulnerability in p53-deficient tumors such as OS. These results define a mechanism through which p53 protects normal osteoblasts from excessive or abnormal cAMP accumulation, which becomes fundamentally compromised in OS.
Collapse
|
24
|
Wiedemann B, Weisner J, Rauh D. Chemical modulation of transcription factors. MEDCHEMCOMM 2018; 9:1249-1272. [PMID: 30151079 PMCID: PMC6097187 DOI: 10.1039/c8md00273h] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 07/10/2018] [Indexed: 12/12/2022]
Abstract
Transcription factors (TFs) constitute a diverse class of sequence-specific DNA-binding proteins, which are key to the modulation of gene expression. TFs have been associated with human diseases, including cancer, Alzheimer's and other neurodegenerative diseases, which makes this class of proteins attractive targets for chemical biology and medicinal chemistry research. Since TFs lack a common binding site or structural similarity, the development of small molecules to efficiently modulate TF biology in cells and in vivo is a challenging task. This review highlights various strategies that are currently being explored for the identification and development of modulators of Myc, p53, Stat, Nrf2, CREB, ER, AR, HIF, NF-κB, and BET proteins.
Collapse
Affiliation(s)
- Bianca Wiedemann
- Technische Universität Dortmund , Fakultät für Chemie und Chemische Biologie , Otto-Hahn-Strasse 4a , D-44227 Dortmund , Germany . ; ; Tel: +49 (0)231 755 7080
| | - Jörn Weisner
- Technische Universität Dortmund , Fakultät für Chemie und Chemische Biologie , Otto-Hahn-Strasse 4a , D-44227 Dortmund , Germany . ; ; Tel: +49 (0)231 755 7080
| | - Daniel Rauh
- Technische Universität Dortmund , Fakultät für Chemie und Chemische Biologie , Otto-Hahn-Strasse 4a , D-44227 Dortmund , Germany . ; ; Tel: +49 (0)231 755 7080
| |
Collapse
|
25
|
Illiano M, Conte M, Sapio L, Nebbioso A, Spina A, Altucci L, Naviglio S. Forskolin Sensitizes Human Acute Myeloid Leukemia Cells to H3K27me2/3 Demethylases GSKJ4 Inhibitor via Protein Kinase A. Front Pharmacol 2018; 9:792. [PMID: 30079022 PMCID: PMC6063003 DOI: 10.3389/fphar.2018.00792] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/28/2018] [Indexed: 01/26/2023] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive hematological malignancy occurring very often in older adults, with poor prognosis depending on both rapid disease progression and drug resistance occurrence. Therefore, new therapeutic approaches are demanded. Epigenetic marks play a relevant role in AML. GSKJ4 is a novel inhibitor of the histone demethylases JMJD3 and UTX. To note GSKJ4 has been recently shown to act as a potent small molecule inhibitor of the proliferation in many cancer cell types. On the other hand, forskolin, a natural cAMP raising compound, used for a long time in traditional medicine and considered safe also in recent studies, is emerging as a very interesting molecule for possible use in cancer therapy. Here, we investigate the effects of forskolin on the sensitivity of human leukemia U937 cells to GSKJ4 through flow cytometry-based assays (cell-cycle progression and cell death), cell number counting, and immunoblotting experiments. We provide evidence that forskolin markedly potentiates GSKJ4-induced antiproliferative effects by apoptotic cell death induction, accompanied by a dramatic BCL2 protein down-regulation as well as caspase 3 activation and PARP protein cleavage. Comparable effects are observed with the phosphodiesterase inhibitor IBMX and 8-Br-cAMP analogous, but not by using 8-pCPT-2'-O-Me-cAMP Epac activator. Moreover, the forskolin-induced enhancement of sensitivity to GSKJ4 is counteracted by pre-treatment with Protein Kinase A (PKA) inhibitors. Altogether, our data strongly suggest that forskolin sensitizes U937 cells to GSKJ4 inhibitor via a cAMP/PKA-mediated mechanism. Our findings provide initial evidence of anticancer activity induced by forskolin/GSKJ4 combination in leukemia cells and underline the potential for use of forskolin and GSKJ4 in the development of innovative and effective therapeutic approaches for AML treatment.
Collapse
Affiliation(s)
- Michela Illiano
- Department of Precision Medicine, School of Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | | | - Luigi Sapio
- Department of Precision Medicine, School of Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Angela Nebbioso
- Department of Precision Medicine, School of Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Annamaria Spina
- Department of Precision Medicine, School of Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Lucia Altucci
- Department of Precision Medicine, School of Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Silvio Naviglio
- Department of Precision Medicine, School of Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| |
Collapse
|
26
|
Takei H, Kobayashi SS. Targeting transcription factors in acute myeloid leukemia. Int J Hematol 2018; 109:28-34. [PMID: 29956082 DOI: 10.1007/s12185-018-2488-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 06/12/2018] [Accepted: 06/25/2018] [Indexed: 12/28/2022]
Abstract
Transcription factors recognize and bind to consensus sequence elements that are specific for each transcription factor, and the transcription factors then regulate downstream gene expression. In the bone marrow, transcription factors, such as C/EBPα, PU.1, and RUNX1, control essential genes to maintain the normal hematopoietic system. Dysregulation of transcription factors caused by gene mutations, chromosomal aberrations, or aberrant expression can lead to cancer, including acute myeloid leukemia. In the past, transcription factors were not considered "druggable" targets. However, a better understanding of the pathology of malignant tumors and mechanisms of transcriptional regulation has enabled us to develop novel therapeutic strategies that target transcription factors. In this review, we focus on transcription factors that play important roles in leukemogenesis and current efforts and prospects in the development of transcriptional therapy. We believe that such a therapeutic approach will benefit patients with cancers that involve acute myeloid leukemia in the near future.
Collapse
Affiliation(s)
- Hisashi Takei
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, USA
- Department of Hematology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Susumu S Kobayashi
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, USA.
- Division of Translational Genomics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan.
- Harvard Stem Cell Institute, Harvard Medical School, Boston, USA.
| |
Collapse
|
27
|
Ramaswamy K, Forbes L, Minuesa G, Gindin T, Brown F, Kharas MG, Krivtsov AV, Armstrong SA, Still E, de Stanchina E, Knoechel B, Koche R, Kentsis A. Peptidomimetic blockade of MYB in acute myeloid leukemia. Nat Commun 2018; 9:110. [PMID: 29317678 PMCID: PMC5760651 DOI: 10.1038/s41467-017-02618-6] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 12/13/2017] [Indexed: 02/08/2023] Open
Abstract
Aberrant gene expression is a hallmark of acute leukemias. MYB-driven transcriptional coactivation with CREB-binding protein (CBP)/P300 is required for acute lymphoblastic and myeloid leukemias, including refractory MLL-rearranged leukemias. Using structure-guided molecular design, we developed a peptidomimetic inhibitor MYBMIM that interferes with the assembly of the molecular MYB:CBP/P300 complex and rapidly accumulates in the nuclei of AML cells. Treatment of AML cells with MYBMIM led to the dissociation of the MYB:CBP/P300 complex in cells, its displacement from oncogenic enhancers enriched for MYB binding sites, and downregulation of MYB-dependent gene expression, including of MYC and BCL2 oncogenes. AML cells underwent mitochondrial apoptosis in response to MYBMIM, which was partially rescued by ectopic expression of BCL2. MYBMIM impeded leukemia growth and extended survival of immunodeficient mice engrafted with primary patient-derived MLL-rearranged leukemia cells. These findings elucidate the dependence of human AML on aberrant transcriptional coactivation, and establish a pharmacologic approach for its therapeutic blockade.
Collapse
Affiliation(s)
- Kavitha Ramaswamy
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, 10065, USA
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, 10065, NY, USA
| | - Lauren Forbes
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, 10065, USA
- Departments of Pediatrics, Pharmacology, and Physiology & Biophysics, Weill Cornell Medical College, Cornell University, New York, NY, 10065, USA
| | - Gerard Minuesa
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, 10065, USA
| | - Tatyana Gindin
- Department of Pathology and Cell Biology, Columbia University Medical Center and New York Presbyterian Hospital, New York, NY, 10065, USA
| | - Fiona Brown
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, 10065, USA
| | - Michael G Kharas
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, 10065, USA
| | - Andrei V Krivtsov
- Center for Epigenetics Research, Sloan Kettering Institute, New York, NY, 10065, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Scott A Armstrong
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, 10065, NY, USA
- Center for Epigenetics Research, Sloan Kettering Institute, New York, NY, 10065, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Eric Still
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, 10065, USA
| | - Elisa de Stanchina
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Birgit Knoechel
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Richard Koche
- Center for Epigenetics Research, Sloan Kettering Institute, New York, NY, 10065, USA
| | - Alex Kentsis
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, 10065, USA.
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, 10065, NY, USA.
- Departments of Pediatrics, Pharmacology, and Physiology & Biophysics, Weill Cornell Medical College, Cornell University, New York, NY, 10065, USA.
| |
Collapse
|
28
|
Chae HD, Cox N, Dahl GV, Lacayo NJ, Davis KL, Capolicchio S, Smith M, Sakamoto KM. Niclosamide suppresses acute myeloid leukemia cell proliferation through inhibition of CREB-dependent signaling pathways. Oncotarget 2017; 9:4301-4317. [PMID: 29435104 PMCID: PMC5796975 DOI: 10.18632/oncotarget.23794] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 12/22/2017] [Indexed: 12/26/2022] Open
Abstract
CREB (cAMP Response Element Binding protein) is a transcription factor that is overexpressed in primary acute myeloid leukemia (AML) cells and associated with a decreased event-free survival and increased risk of relapse. We recently reported a small molecule inhibitor of CREB, XX-650-23, which inhibits CREB activity in AML cells. Structure-activity relationship analysis for chemical compounds with structures similar to XX-650-23 led to the identification of the anthelminthic drug niclosamide as a potent anti-leukemic agent that suppresses cell viability of AML cell lines and primary AML cells without a significant decrease in colony forming activity of normal bone marrow cells. Niclosamide significantly inhibited CREB function and CREB-mediated gene expression in cells, leading to apoptosis and G1/S cell cycle arrest with reduced phosphorylated CREB levels. CREB knockdown protected cells from niclosamide treatment-mediated cytotoxic effects. Furthermore, treatment with a combination of niclosamide and CREB inhibitor XX-650-23 showed an additive anti-proliferative effect, consistent with the hypothesis that niclosamide and XX-650-23 regulate the same targets or pathways to inhibit proliferation and survival of AML cells. Niclosamide significantly inhibited the progression of disease in AML patient-derived xenograft (PDX) mice, and prolonged survival of PDX mice. Niclosamide also showed synergistic effects with chemotherapy drugs to inhibit AML cell proliferation. While chemotherapy antagonized the cytotoxic potential of niclosamide, pretreatment with niclosamide sensitized cells to chemotherapeutic drugs, cytarabine, daunorubicin, and vincristine. Therefore, our results demonstrate niclosamide as a potential drug to treat AML by inducing apoptosis and cell cycle arrest through inhibition of CREB-dependent pathways in AML cells.
Collapse
Affiliation(s)
- Hee-Don Chae
- Division of Hematology/Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Nick Cox
- Medicinal Chemistry Knowledge Center, Stanford ChEM-H, Stanford, CA, USA
| | - Gary V Dahl
- Medicinal Chemistry Knowledge Center, Stanford ChEM-H, Stanford, CA, USA
| | - Norman J Lacayo
- Division of Hematology/Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Kara L Davis
- Division of Hematology/Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Samanta Capolicchio
- Division of Hematology/Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA.,Division of Hematology/Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Mark Smith
- Division of Hematology/Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Kathleen M Sakamoto
- Division of Hematology/Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
29
|
Mantamadiotis T. Towards Targeting PI3K-Dependent Regulation of Gene Expression in Brain Cancer. Cancers (Basel) 2017; 9:cancers9060060. [PMID: 28556811 PMCID: PMC5483879 DOI: 10.3390/cancers9060060] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 05/22/2017] [Accepted: 05/23/2017] [Indexed: 12/31/2022] Open
Abstract
The PI3K pathway is one of the most highly perturbed cell signaling pathways in human cancer, including the most common malignant brain tumors, gliomas, where either activating mutations of positive pathway effectors or loss/inactivation of pathway inhibitors occurs. Knowledge of the precise transcription factors modulated by PI3K in tumor cells remains elusive but there are numerous PI3K-responsive signaling factors, including kinases, which can activate many transcription factors. In the context of cancer, these transcription factors participate in the regulation of target genes expression networks to support cancer cell characteristics such as survival, proliferation, migration and differentiation. This review focuses on the role of PI3K signaling-regulated transcription in brain cancer cells from a series of recent investigations. A deeper understanding of this regulation is beginning to provide the hope of developing more sophisticated anti-cancer targeting approaches, where both upstream and downstream components of the PI3K pathway may be targeted by existing and novel drugs.
Collapse
Affiliation(s)
- Theo Mantamadiotis
- Department of Pathology, School of Biomedical Sciences, University of Melbourne, Parkville 3010, VIC, Australia.
| |
Collapse
|
30
|
Xie F, Li BX, Xiao X. Design, synthesis and biological evaluation of regioisomers of 666-15 as inhibitors of CREB-mediated gene transcription. Bioorg Med Chem Lett 2017; 27:994-998. [PMID: 28073675 PMCID: PMC5296214 DOI: 10.1016/j.bmcl.2016.12.078] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/28/2016] [Accepted: 12/28/2016] [Indexed: 11/19/2022]
Abstract
cAMP-response element binding protein (CREB) is a nuclear transcription factor that has been implicated in the pathogenesis and maintenance of various types of human cancers. Identification of small molecule inhibitors of CREB-mediated gene transcription has been pursued as a novel strategy for developing cancer therapeutics. We recently discovered a potent and cell-permeable CREB inhibitor called 666-15. 666-15 is a bisnaphthamide and has been shown to possess efficacious anti-breast cancer activity without toxicity in vivo. In this study, we designed and synthesized a series of analogs of 666-15 to probe the importance of regiochemistry in naphthalene ring B. Biological evaluations of these analogs demonstrated that the substitution pattern of the alkoxy and carboxamide in naphthalene ring B is very critical for maintaining potent CREB inhibition activity, suggesting that the unique bioactive conformation accessible in 666-15 is critically important.
Collapse
Affiliation(s)
- Fuchun Xie
- Program in Chemical Biology, Department of Physiology and Pharmacology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
| | - Bingbing X Li
- Program in Chemical Biology, Department of Physiology and Pharmacology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
| | - Xiangshu Xiao
- Program in Chemical Biology, Department of Physiology and Pharmacology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA; Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA; Knight Cardiovascular Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA.
| |
Collapse
|
31
|
Bisio V, Zampini M, Tregnago C, Manara E, Salsi V, Di Meglio A, Masetti R, Togni M, Di Giacomo D, Minuzzo S, Leszl A, Zappavigna V, Rondelli R, Mecucci C, Pession A, Locatelli F, Basso G, Pigazzi M. NUP98-fusion transcripts characterize different biological entities within acute myeloid leukemia: a report from the AIEOP-AML group. Leukemia 2016; 31:974-977. [PMID: 27890935 DOI: 10.1038/leu.2016.361] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- V Bisio
- Dipartimento di Salute della Donna e del Bambino, Clinica di Oncoematologia Pediatrica, Università di Padova, Padova, Italy
| | - M Zampini
- Istituto di Ricerca Pediatrica-Città della Speranza, Padova, Italy
| | - C Tregnago
- Dipartimento di Salute della Donna e del Bambino, Clinica di Oncoematologia Pediatrica, Università di Padova, Padova, Italy
| | - E Manara
- Istituto di Ricerca Pediatrica-Città della Speranza, Padova, Italy
| | - V Salsi
- Dipartimento di Scienze Mediche e Chirurgiche Materno-Infantili e dell'Adulto, University of Modena and Reggio Emilia, Modena, Italy
| | - A Di Meglio
- Istituto di Ricerca Pediatrica-Città della Speranza, Padova, Italy
| | - R Masetti
- Clinica Pediatrica, Università di Bologna, Ospedale 'S. Orsola', Bologna, Italy
| | - M Togni
- Clinica Pediatrica, Università di Bologna, Ospedale 'S. Orsola', Bologna, Italy
| | - D Di Giacomo
- Department of Medicine, Laboratory of Molecular Medicine, CREO, University of Perugia, Perugia, Italy
| | - S Minuzzo
- Dipartimento di Scienze Chirurgiche, Oncologiche e Gastroenterologiche, Università di Padova, Padova, Italy
| | - A Leszl
- UOC Oncoematologia Pediatrica, Azienda Ospedaliera-Università degli Studi di Padova, Padova, Italy
| | - V Zappavigna
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - R Rondelli
- Clinica Pediatrica, Università di Bologna, Ospedale 'S. Orsola', Bologna, Italy
| | - C Mecucci
- Department of Medicine, Laboratory of Molecular Medicine, CREO, University of Perugia, Perugia, Italy
| | - A Pession
- Clinica Pediatrica, Università di Bologna, Ospedale 'S. Orsola', Bologna, Italy
| | - F Locatelli
- Department of Pediatric Hematology/Oncology, IRCCS Bambino Gesù Children's Hospital, Rome, Università di Pavia, Italy
| | - G Basso
- Dipartimento di Salute della Donna e del Bambino, Clinica di Oncoematologia Pediatrica, Università di Padova, Padova, Italy
| | - M Pigazzi
- Dipartimento di Salute della Donna e del Bambino, Clinica di Oncoematologia Pediatrica, Università di Padova, Padova, Italy
| |
Collapse
|
32
|
Li BX, Gardner R, Xue C, Qian DZ, Xie F, Thomas G, Kazmierczak SC, Habecker BA, Xiao X. Systemic Inhibition of CREB is Well-tolerated in vivo. Sci Rep 2016; 6:34513. [PMID: 27694829 PMCID: PMC5046085 DOI: 10.1038/srep34513] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 09/15/2016] [Indexed: 11/30/2022] Open
Abstract
cAMP-response element binding protein (CREB) is a nuclear transcription factor activated by multiple extracellular signals including growth factors and hormones. These extracellular cues activate CREB through phosphorylation at Ser133 by various protein serine/threonine kinases. Once phosphorylated, it promotes its association with transcription coactivators CREB-binding protein (CBP) and its paralog p300 to activate CREB-dependent gene transcription. Tumor tissues of different origins have been shown to present overexpression and/or overactivation of CREB, indicating CREB as a potential cancer drug target. We previously identified 666-15 as a potent inhibitor of CREB with efficacious anti-cancer activity both in vitro and in vivo. Herein, we investigated the specificity of 666-15 and evaluated its potential in vivo toxicity. We found that 666-15 was fairly selective in inhibiting CREB. 666-15 was also found to be readily bioavailable to achieve pharmacologically relevant concentrations for CREB inhibition. Furthermore, the mice treated with 666-15 showed no evidence of changes in body weight, complete blood count, blood chemistry profile, cardiac contractility and tissue histologies from liver, kidney and heart. For the first time, these results demonstrate that pharmacological inhibition of CREB is well-tolerated in vivo and indicate that such inhibitors should be promising cancer therapeutics.
Collapse
Affiliation(s)
- Bingbing X Li
- Program in Chemical Biology, Department of Physiology and Pharmacology, Oregon Health &Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
| | - Ryan Gardner
- Program in Chemical Biology, Department of Physiology and Pharmacology, Oregon Health &Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
| | - Changhui Xue
- Knight Cancer Institute, Oregon Health &Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
| | - David Z Qian
- Knight Cancer Institute, Oregon Health &Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
| | - Fuchun Xie
- Program in Chemical Biology, Department of Physiology and Pharmacology, Oregon Health &Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
| | - George Thomas
- Knight Cancer Institute, Oregon Health &Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
| | - Steven C Kazmierczak
- Department of Pathology, Oregon Health &Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
| | - Beth A Habecker
- Program in Chemical Biology, Department of Physiology and Pharmacology, Oregon Health &Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA.,Knight Cardiovascular Institute, Department of Medicine, Oregon Health &Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
| | - Xiangshu Xiao
- Program in Chemical Biology, Department of Physiology and Pharmacology, Oregon Health &Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA.,Knight Cancer Institute, Oregon Health &Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA.,Knight Cardiovascular Institute, Department of Medicine, Oregon Health &Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
| |
Collapse
|
33
|
Varicella-Zoster Virus Activates CREB, and Inhibition of the pCREB-p300/CBP Interaction Inhibits Viral Replication In Vitro and Skin Pathogenesis In Vivo. J Virol 2016; 90:8686-97. [PMID: 27440893 PMCID: PMC5021407 DOI: 10.1128/jvi.00920-16] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 07/13/2016] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED Varicella-zoster virus (VZV) is an alphaherpesvirus that causes varicella upon primary infection and zoster upon reactivation from latency in sensory ganglion neurons. The replication of herpesviruses requires manipulation of cell signaling pathways. Notably, CREB, a factor involved in the regulation of several cellular processes, is activated upon infection of T cells with VZV. Here, we report that VZV infection also induced CREB phosphorylation in fibroblasts and that XX-650-23, a newly identified inhibitor of the phosphorylated-CREB (pCREB) interaction with p300/CBP, restricted cell-cell spread of VZV in vitro CREB phosphorylation did not require the viral open reading frame 47 (ORF47) and ORF66 kinases encoded by VZV. Evaluating the biological relevance of these observations during VZV infection of human skin xenografts in the SCID mouse model of VZV pathogenesis showed both that pCREB was upregulated in infected skin and that treatment with XX-650-23 reduced infectious-virus production and limited lesion formation compared to treatment with a vehicle control. Thus, processes of CREB activation and p300/CBP binding are important for VZV skin infection and may be targeted for antiviral drug development. IMPORTANCE Varicella-zoster virus (VZV) is a common pathogen that causes chicken pox and shingles. As with all herpesviruses, the infection is acquired for life, and the virus can periodically reactivate from latency. Although VZV infection is usually benign with few or no deleterious consequences, infection can be life threatening in immunocompromised patients. Otherwise healthy elderly individuals who develop zoster as a consequence of viral reactivation are at risk for postherpetic neuralgia (PHN), a painful and long-lasting complication. Current vaccines use a live attenuated virus that is usually safe but cannot be given to many immunodeficient patients and retains the capacity to establish latency and reactivate, causing zoster. Antiviral drugs are effective against severe VZV infections but have little impact on PHN. A better understanding of virus-host cell interactions is relevant for developing improved therapies to safely interfere with cellular processes that are crucial for VZV pathogenesis.
Collapse
|