1
|
Zhen Y, Li D, Meng Y, Xing Z, Zheng J. Unveiling the roles of HIPK2 in atherosclerosis: Insights into the β-catenin/STAT1 signaling cascade and the involvement of SENP1. Biochem Pharmacol 2025; 237:116911. [PMID: 40164339 DOI: 10.1016/j.bcp.2025.116911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 03/03/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025]
Abstract
Atherosclerosis is a disorder of lipid metabolism, but its pathogenesis has not yet been fully elucidated. This study aimed to clarify the roles of homeodomain interacting protein kinase 2 (HIPK2) in atherosclerosis. Atherosclerotic model was constructed by feeding apolipoprotein E knockout (ApoE-/-) mice with a high-fat diet. Human THP-1 macrophages and mouse RAW 264.7 macrophages were stimulated with IFN-γ to establish an in vitro model. We showed an upregulation of HIPK2 in the aorta of atherosclerotic mice. HIPK2 knockdown reduced macrophage infiltration, M1 polarization, and attenuates atherosclerosis development. Downregulation of HIPK2 in macrophages led to a significant suppression in the expression of pro-inflammatory factors, which was accompanied by an enhancement in the phosphorylation and degradation of β-catenin, as well as the activation of the signal transducer and activator of transcription 1 (STAT1) signaling pathway. Silencing of HIPK2 alone in THP-1 macrophages resulted in anti-inflammatory effects and suppression of M1 macrophage polarization. However, simultaneous silencing of HIPK2 and β-catenin (CTNNB1) reversed these effects, counteracting the outcomes observed with HIPK2 silencing alone. We validated that small ubiquitin-like modifier (SUMO)-specific peptidase 1 (SENP1) regulated HIPK2 function by affecting the SUMOylation of HIPK2 at the K32 site. SENP1 knockdown promoted HIPK2 SUMOylation, impairing its protein stability. In the rescue experiments, IFN-γ-induced inflammation and M1 polarization were resumed upon restoration of HIPK2 expression in SENP1-silenced macrophages. Our work demonstrated that HIPK2 accelerated the progression of atherosclerosis by regulating β-catenin/STAT1 signaling cascade to promote macrophage infiltration and M1 polarization. HIPK2 was regulated by SENP1-mediated de-SUMOylation.
Collapse
Affiliation(s)
- Yanhua Zhen
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang 110022, China
| | - Dongdong Li
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang 110022, China
| | - Yulu Meng
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Zeyu Xing
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang 110022, China
| | - Jiahe Zheng
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang 110022, China.
| |
Collapse
|
2
|
Wong K, Bishop JA, Weinreb I, Motta M, Del Castillo Velasco-Herrera M, Bellacchio E, Ferreira I, van der Weyden L, Boccacino JM, Lauri A, Rotundo G, Ciolfi A, Cheema S, Olvera-León R, Offord V, Droop A, Vermes I, Allgäuer M, Hyrcza M, Anderson E, Smith K, de Saint Aubain N, Mogler C, Stenzinger A, Arends MJ, Brenn T, Tartaglia M, Adams DJ. Wnt/β-catenin activation by mutually exclusive FBXW11 and CTNNB1 hotspot mutations drives salivary basal cell adenoma. Nat Commun 2025; 16:4657. [PMID: 40389436 PMCID: PMC12089348 DOI: 10.1038/s41467-025-59871-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/07/2025] [Indexed: 05/21/2025] Open
Abstract
Basal cell adenoma (BCA) and basal cell adenocarcinoma (BCAC) of the salivary gland are rare tumours that can be difficult to distinguish from each other and other salivary gland tumour subtypes. Using next-generation sequencing, we identify a recurrent FBXW11 missense mutation (p.F517S) in BCA that is mutually exclusive with the previously reported CTNNB1 p.I35T gain-of-function (GoF) mutation with these mutations collectively accounting for 94% of BCAs. In vitro, mutant FBXW11 is characterised by defective binding to β-catenin and higher protein levels within the nucleus. This is consistent with the increased nuclear expression of β-catenin and activation of the Wnt/β-catenin pathway. The genomic profiles of BCAC are distinct from BCA, with hotspot DICER1 and HRAS mutations and putative driver mutations affecting PI3K/AKT and NF-κB signalling pathway genes. These findings have important implications for the diagnosis and treatment of BCA and BCAC, which, despite histopathologic overlap, may be unrelated entities.
Collapse
Affiliation(s)
- Kim Wong
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | - Justin A Bishop
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ilan Weinreb
- Laboratory Medicine Program, University Health Network, Toronto General Hospital, Toronto, ON, Canada
- Department of Pathobiology and Laboratory Medicine, University of Toronto, Toronto, ON, Canada
| | - Marialetizia Motta
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | | | - Emanuele Bellacchio
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Ingrid Ferreira
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | | | | | - Antonella Lauri
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Giovannina Rotundo
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Andrea Ciolfi
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Saamin Cheema
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | - Rebeca Olvera-León
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | - Victoria Offord
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | - Alastair Droop
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | - Ian Vermes
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | - Michael Allgäuer
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Martin Hyrcza
- Department of Pathology and Laboratory Medicine, University of Calgary, Arnie Charboneau Cancer Institute, Calgary, AB, Canada
| | - Elizabeth Anderson
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | - Katie Smith
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | - Nicolas de Saint Aubain
- Department of Pathology, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles, Brussels, Belgium
| | - Carolin Mogler
- School of Medicine and Health, Technical University Munich, Munich, Germany
| | | | - Mark J Arends
- Edinburgh Pathology, Cancer Research UK Scotland Centre, The University of Edinburgh, Institute of Genetics and Cancer, Edinburgh, UK
| | - Thomas Brenn
- Departments of Pathology and Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - David J Adams
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, Cambridge, UK.
| |
Collapse
|
3
|
Liu X, Zhu Y, Huang W, Chen J, Lan J, Long X, Zhou J. MYG1 interacts with HSP90 to promote breast cancer progression through Wnt/β-catenin and Notch signaling pathways. Exp Cell Res 2025; 446:114448. [PMID: 39956425 DOI: 10.1016/j.yexcr.2025.114448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/23/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025]
Abstract
BACKGROUND As an evolutionarily conserved gene involved in embryonic development, cell differentiation, and immune metabolism, MYG1 exhibits a dynamic expression pattern related to development in human and mouse embryonic tissues, especially upregulates in undifferentiated or pluripotent stem cells. However, MYG1 has been poorly studied in breast cancer and its functional mechanism still remains unclear. METHOD Immunohistochemistry and immunofluorescence were used to study MYG1 expression and localization in breast cancer. Lentivirus transfection combined with CCK8, colony formation, matrix gel experiment and breast fat pad tumor formation in nude mice were used for in vivo and in vitro functional assessment. GSEA enrichment analysis, immunofluorescence and Western blot were conducted to explore functional mechanism. RESULT MYG1 expression was upregulated in breast cancer and its higher expression correlated with a variety of clinicopathological characteristics indicating poor prognosis. In vitro and in vivo experiments showed that overexpression of MYG1 promoted breast cancer cells proliferation, migration, invasion and tumorigenesis, while downregulation of MYG1 had an opposite effect. Mechanistically, MYG1 interacted with HSP90 to significantly activate Wnt/β-catenin and Notch signaling pathways in breast cancer cells, thus promoting EMT, cell cycle process and breast cancer progression. CONCLUSION MYG1 is highly expressed in breast cancer and functions as an oncogene. Mechanistically, MYG1 interacts with HSP90 to accelerate EMT and cell cycle process by activating both Wnt/β-catenin and Notch signaling pathways.
Collapse
Affiliation(s)
- Xuming Liu
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou North Road, Guangzhou, 510515, China
| | - Yurong Zhu
- Department of Pathology, The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, 78 Wandao Road, Dongguan, 523039, China; Dongguan Clinical Pathology Diagnosis Center, 14 Yuhua Road, Dongguan, 523001, China; Dongguan Key Laboratory of Clinical Pathology, 14 Yuhua Road, Dongguan, 523001, China
| | - Wenqing Huang
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou North Road, Guangzhou, 510515, China
| | - Jianxiong Chen
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou North Road, Guangzhou, 510515, China
| | - Jiawen Lan
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou North Road, Guangzhou, 510515, China
| | - Xiaoli Long
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou North Road, Guangzhou, 510515, China.
| | - Jun Zhou
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, 1838 Guangzhou North Road, Guangzhou, 510515, China; Department of Pathology, The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, 78 Wandao Road, Dongguan, 523039, China; Dongguan Clinical Pathology Diagnosis Center, 14 Yuhua Road, Dongguan, 523001, China; Dongguan Key Laboratory of Clinical Pathology, 14 Yuhua Road, Dongguan, 523001, China.
| |
Collapse
|
4
|
Verdina A, Garufi A, D’Orazi V, D’Orazi G. HIPK2 in Colon Cancer: A Potential Biomarker for Tumor Progression and Response to Therapies. Int J Mol Sci 2024; 25:7678. [PMID: 39062921 PMCID: PMC11277226 DOI: 10.3390/ijms25147678] [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: 06/24/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Colon cancer, one of the most common and fatal cancers worldwide, is characterized by stepwise accumulation of specific genetic alterations in tumor suppressor genes or oncogenes, leading to tumor growth and metastasis. HIPK2 (homeodomain-interacting protein kinase 2) is a serine/threonine protein kinase and a "bona fide" oncosuppressor protein. Its activation inhibits tumor growth mainly by promoting apoptosis, while its inactivation increases tumorigenicity and resistance to therapies of many different cancer types, including colon cancer. HIPK2 interacts with many molecular pathways by means of its kinase activity or transcriptional co-repressor function modulating cell growth and apoptosis, invasion, angiogenesis, inflammation and hypoxia. HIPK2 has been shown to participate in several molecular pathways involved in colon cancer including p53, Wnt/β-catenin and the newly identified nuclear factor erythroid 2 (NF-E2) p45-related factor 2 (NRF2). HIPK2 also plays a role in tumor-host interaction in the tumor microenvironment (TME) by inducing angiogenesis and cancer-associated fibroblast (CAF) differentiation. The aim of this review is to assess the role of HIPK2 in colon cancer and the underlying molecular pathways for a better understanding of its involvement in colon cancer carcinogenesis and response to therapies, which will likely pave the way for novel colon cancer therapies.
Collapse
Affiliation(s)
- Alessandra Verdina
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (A.V.); (A.G.)
| | - Alessia Garufi
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (A.V.); (A.G.)
| | - Valerio D’Orazi
- Department of Surgery, Sapienza University, 00185 Rome, Italy;
| | - Gabriella D’Orazi
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (A.V.); (A.G.)
- Department of Neurosciences, Imaging and Clinical Sciences, University “G. D’Annunzio”, 66013 Chieti, Italy
| |
Collapse
|
5
|
Wu Z, Wu D, Zhong Q, Zou X, Liu Z, Long H, Wei J, Li X, Dai F. The role of zyxin in signal transduction and its relationship with diseases. Front Mol Biosci 2024; 11:1371549. [PMID: 38712343 PMCID: PMC11070705 DOI: 10.3389/fmolb.2024.1371549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 04/08/2024] [Indexed: 05/08/2024] Open
Abstract
This review highlighted the pivotal role of zyxin, an essential cell focal adhesions protein, in cellular biology and various diseases. Zyxin can orchestrate the restructuring and dynamic alterations of the cellular cytoskeleton, which is involved in cell proliferation, adhesion, motility, and gene transcription. Aberrant zyxin expression is closely correlated with tumor cell activity and cardiac function in both tumorigenesis and cardiovascular diseases. Moreover, in fibrotic and inflammatory conditions, zyxin can modulate cellular functions and inflammatory responses. Therefore, a comprehensive understanding of zyxin is crucial for deciphering signal transduction networks and disease pathogenesis. Investigating its role in diseases holds promise for novel avenues in early diagnosis and therapeutic strategies. Nevertheless, targeting zyxin as a therapeutic focal point presents challenges in terms of specificity, safety, drug delivery, and resistance. Nonetheless, in-depth studies on zyxin and the application of precision medicine could offer new possibilities for personalized treatment modalities.
Collapse
Affiliation(s)
- Zelan Wu
- Department of Cardiovascular Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Daiqin Wu
- Department of Cardiovascular Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Qin Zhong
- Clinical Research Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xue Zou
- Clinical Research Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zhongjing Liu
- Clinical Research Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Hehua Long
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, China
| | - Jing Wei
- Department of Endocrinology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xia Li
- Guizhou Precision Medicine Institute, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Fangjie Dai
- Department of Cardiovascular Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
6
|
Garufi A, D’Orazi V, Pistritto G, Cirone M, D’Orazi G. HIPK2 in Angiogenesis: A Promising Biomarker in Cancer Progression and in Angiogenic Diseases. Cancers (Basel) 2023; 15:1566. [PMID: 36900356 PMCID: PMC10000595 DOI: 10.3390/cancers15051566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
Angiogenesis is the formation of new blood capillaries taking place from preexisting functional vessels, a process that allows cells to cope with shortage of nutrients and low oxygen availability. Angiogenesis may be activated in several pathological diseases, from tumor growth and metastases formation to ischemic and inflammatory diseases. New insights into the mechanisms that regulate angiogenesis have been discovered in the last years, leading to the discovery of new therapeutic opportunities. However, in the case of cancer, their success may be limited by the occurrence of drug resistance, meaning that the road to optimize such treatments is still long. Homeodomain-interacting protein kinase 2 (HIPK2), a multifaceted protein that regulates different molecular pathways, is involved in the negative regulation of cancer growth, and may be considered a "bona fide" oncosuppressor molecule. In this review, we will discuss the emerging link between HIPK2 and angiogenesis and how the control of angiogenesis by HIPK2 impinges in the pathogenesis of several diseases, including cancer.
Collapse
Affiliation(s)
- Alessia Garufi
- Unit of Cellular Networks, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Valerio D’Orazi
- Department of Surgery, Sapienza University, 00185 Rome, Italy
| | - Giuseppa Pistritto
- Centralized Procedures Office, Italian Medicines Agency (AIFA), 00187 Rome, Italy
| | - Mara Cirone
- Laboratory Affiliated to Pasteur Institute Italy Foundation Cenci Bolognetti, Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Gabriella D’Orazi
- Unit of Cellular Networks, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
- Department of Neurosciences, Imaging and Clinical Sciences, University “G. D’Annunzio”, 66013 Chieti, Italy
| |
Collapse
|
7
|
Garufi A, Pistritto G, D’Orazi G. HIPK2 as a Novel Regulator of Fibrosis. Cancers (Basel) 2023; 15:1059. [PMID: 36831402 PMCID: PMC9954661 DOI: 10.3390/cancers15041059] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/01/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Fibrosis is an unmet medical problem due to a lack of evident biomarkers to help develop efficient targeted therapies. Fibrosis can affect almost every organ and eventually induce organ failure. Homeodomain-interacting protein kinase 2 (HIPK2) is a protein kinase that controls several molecular pathways involved in cell death and development and it has been extensively studied, mainly in the cancer biology field. Recently, a role for HIPK2 has been highlighted in tissue fibrosis. Thus, HIPK2 regulates several pro-fibrotic pathways such as Wnt/β-catenin, TGF-β and Notch involved in renal, pulmonary, liver and cardiac fibrosis. These findings suggest a wider role for HIPK2 in tissue physiopathology and highlight HIPK2 as a promising target for therapeutic purposes in fibrosis. Here, we will summarize the recent studies showing the involvement of HIPK2 as a novel regulator of fibrosis.
Collapse
Affiliation(s)
- Alessia Garufi
- Unit of Cellular Networks, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Giuseppa Pistritto
- Centralized Procedures Office, Italian Medicines Agency (AIFA), 00187 Rome, Italy
| | - Gabriella D’Orazi
- Unit of Cellular Networks, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
- Department of Neurosciences, Imaging and Clinical Sciences, University “G. D’Annunzio”, 66013 Chieti, Italy
| |
Collapse
|
8
|
Tsang B, Pritišanac I, Scherer SW, Moses AM, Forman-Kay JD. Phase Separation as a Missing Mechanism for Interpretation of Disease Mutations. Cell 2020; 183:1742-1756. [DOI: 10.1016/j.cell.2020.11.050] [Citation(s) in RCA: 170] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/04/2020] [Accepted: 11/25/2020] [Indexed: 02/08/2023]
|
9
|
Li Y, Lv Y, Cheng C, Huang Y, Yang L, He J, Tao X, Hu Y, Ma Y, Su Y, Wu L, Yu G, Jiang Q, Liu S, Liu X, Liu Z. SPEN induces miR-4652-3p to target HIPK2 in nasopharyngeal carcinoma. Cell Death Dis 2020; 11:509. [PMID: 32641685 PMCID: PMC7343777 DOI: 10.1038/s41419-020-2699-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 06/10/2020] [Accepted: 06/15/2020] [Indexed: 02/07/2023]
Abstract
SPEN family transcriptional repressor (SPEN), also known as the SMART/HDAC1-associated repressor protein (SHARP), has been reported to modulate the malignant phenotypes of breast cancer, colon cancer, and ovarian cancer. However, its role and the detail molecular basis in nasopharyngeal carcinoma (NPC) remain elusive. In this study, the SPEN mRNA and protein expression was found to be increased in NPC cells and tissues compared with nonmalignant nasopharyngeal epithelial cells and tissues. Elevated SPEN protein expression was found to promote the pathogenesis of NPC and lead to poor prognosis. Knockdown of SPEN expression resulted in inactivation ofPI3K/AKT and c-JUN signaling, thereby suppressing NPC migration and invasion. In addition, miR-4652-3p was found to be a downstream inducer of SPEN by targeting the homeodomain interacting protein kinase 2 (HIPK2) gene, a potential tumor suppressor that reduces the activation of epithelial-mesenchymal transition (EMT) signaling, thereby reducing its expression and leading to increased NPC migration, invasion, and metastasis. In addition, SPEN was found to induce miR-4652-3p expression by activating PI3K/AKT/c-JUN signaling to target HIPK2. Our data provided a new molecular mechanism for SPEN as a metastasis promoter through activation of PI3K/AKT signaling, thereby stimulating the c-JUN/miR-4652-3p axis to target HIPK2 in NPC.
Collapse
Affiliation(s)
- Yang Li
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Yumin Lv
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Chao Cheng
- Department of Pediatric Otorhinolaryngology, Shenzhen Key Laboratory of Viral Oncology, The Clinical Innovation & Research Centre, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Yan Huang
- Department of Pathology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Liu Yang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Jingjing He
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Xingyu Tao
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Yingying Hu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Yuting Ma
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Yun Su
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Liyang Wu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Guifang Yu
- Department of Oncology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Qingping Jiang
- Department of Pathology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shu Liu
- Department of Breast Surgery, Guiyang Maternal and Child Healthcare Hospital, Guiyang, 550003, Guizhou, China
| | - Xiong Liu
- E.N.T. Department of Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Zhen Liu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China.
| |
Collapse
|
10
|
Deshmukh V, O'Green AL, Bossard C, Seo T, Lamangan L, Ibanez M, Ghias A, Lai C, Do L, Cho S, Cahiwat J, Chiu K, Pedraza M, Anderson S, Harris R, Dellamary L, Kc S, Barroga C, Melchior B, Tam B, Kennedy S, Tambiah J, Hood J, Yazici Y. Modulation of the Wnt pathway through inhibition of CLK2 and DYRK1A by lorecivivint as a novel, potentially disease-modifying approach for knee osteoarthritis treatment. Osteoarthritis Cartilage 2019; 27:1347-1360. [PMID: 31132406 DOI: 10.1016/j.joca.2019.05.006] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 04/23/2019] [Accepted: 05/14/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVES Wnt pathway upregulation contributes to knee osteoarthritis (OA) through osteoblast differentiation, increased catabolic enzymes, and inflammation. The small-molecule Wnt pathway inhibitor, lorecivivint (SM04690), which previously demonstrated chondrogenesis and cartilage protection in an animal OA model, was evaluated to elucidate its mechanism of action. DESIGN Biochemical assays measured kinase activity. Western blots measured protein phosphorylation in human mesenchymal stem cells (hMSCs), chondrocytes, and synovial fibroblasts. siRNA knockdown effects in hMSCs and BEAS-2B cells on Wnt pathway, chondrogenic genes, and LPS-induced inflammatory cytokines was measured by qPCR. In vivo anti-inflammation, pain, and function were evaluated following single intra-articular (IA) lorecivivint or vehicle injection in the monosodium iodoacetate (MIA)-induced rat OA model. RESULTS Lorecivivint inhibited intranuclear kinases CDC-like kinase 2 (CLK2) and dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A). Lorecivivint inhibited CLK2-mediated phosphorylation of serine/arginine-rich (SR) splicing factors and DYRK1A-mediated phosphorylation of SIRT1 and FOXO1. siRNA knockdowns identified a role for CLK2 and DYRK1A in Wnt pathway modulation without affecting β-catenin with CLK2 inhibition inducing early chondrogenesis and DYRK1A inhibition enhancing mature chondrocyte function. NF-κB and STAT3 inhibition by lorecivivint reduced inflammation. DYRK1A knockdown was sufficient for anti-inflammatory effects, while combined DYRK1A/CLK2 knockdown enhanced this effect. In the MIA model, lorecivivint inhibited production of inflammatory cytokines and cartilage degradative enzymes, resulting in increased joint cartilage, decreased pain, and improved weight-bearing function. CONCLUSIONS Lorecivivint inhibition of CLK2 and DYRK1A suggested a novel mechanism for Wnt pathway inhibition, enhancing chondrogenesis, chondrocyte function, and anti-inflammation. Lorecivivint shows potential to modify structure and improve symptoms of knee OA.
Collapse
Affiliation(s)
| | | | | | - T Seo
- Samumed, LLC, San Diego, CA, USA.
| | | | - M Ibanez
- Samumed, LLC, San Diego, CA, USA.
| | - A Ghias
- Samumed, LLC, San Diego, CA, USA.
| | - C Lai
- Samumed, LLC, San Diego, CA, USA.
| | - L Do
- Samumed, LLC, San Diego, CA, USA.
| | - S Cho
- Samumed, LLC, San Diego, CA, USA.
| | | | - K Chiu
- Samumed, LLC, San Diego, CA, USA.
| | | | | | - R Harris
- Samumed, LLC, San Diego, CA, USA.
| | | | - S Kc
- Samumed, LLC, San Diego, CA, USA.
| | | | | | - B Tam
- Formerly Samumed, LLC, USA.
| | | | | | - J Hood
- Formerly Samumed, LLC, USA.
| | - Y Yazici
- Samumed, LLC, San Diego, CA, USA.
| |
Collapse
|
11
|
Cao L, Yang G, Gao S, Jing C, Montgomery RR, Yin Y, Wang P, Fikrig E, You F. HIPK2 is necessary for type I interferon-mediated antiviral immunity. Sci Signal 2019; 12:12/573/eaau4604. [PMID: 30890658 DOI: 10.1126/scisignal.aau4604] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Precise control of interferons (IFNs) is crucial to maintain immune homeostasis. Here, we demonstrated that homeodomain-interacting protein kinase 2 (HIPK2) was required for the production of type I IFNs in response to RNA virus infection. HIPK2 deficiency markedly impaired IFN production in macrophages after vesicular stomatitis virus (VSV) infection, and HIPK2-deficient mice were more susceptible to lethal VSV disease than were wild-type mice. After VSV infection, HIPK2 was cleaved by active caspases, which released a hyperactive, N-terminal fragment that translocated to the nucleus and further augmented antiviral responses. In part, HIPK2 interacted with ELF4 and promoted its phosphorylation at Ser369, which enabled Ifn-b transcription. In addition, HIPK2 production was stimulated by type I IFNs to further enhance antiviral immunity. These data suggest that the kinase activity and nuclear localization of HIPK2 are essential for the production of type I IFNs.
Collapse
Affiliation(s)
- Lili Cao
- Institute of Systems Biomedicine, Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| | - Guang Yang
- Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT 208022, USA.,Department of Parasitology, Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, No. 601, Huangpu Avenue West, Guangzhou, Guangdong 510632, China
| | - Shandian Gao
- Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT 208022, USA
| | - Chunxia Jing
- Department of Epidemiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Ruth R Montgomery
- Section of Rheumatology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Yuxin Yin
- Institute of Systems Biomedicine, Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| | - Penghua Wang
- Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT 208022, USA.,Department of Immunology, University of Connecticut Health Center, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT 208022, USA. .,Howard Hughes Medical Institute, Chevy Chase, MA 20815, USA
| | - Fuping You
- Institute of Systems Biomedicine, Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China.
| |
Collapse
|
12
|
Dar MS, Singh P, Mir RA, Dar MJ. Βeta-catenin N-terminal domain: An enigmatic region prone to cancer causing mutations. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2017; 773:122-133. [PMID: 28927523 DOI: 10.1016/j.mrrev.2017.06.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 05/29/2017] [Accepted: 06/05/2017] [Indexed: 12/21/2022]
Abstract
The Wnt/β-catenin is a highly conserved signaling pathway involved in cell fate decisions during various stages of development. Dysregulation of canonical Wnt/β-catenin signaling has been associated with various diseases including cancer. β-Catenin, the central component of canonical Wnt signaling pathway, is a multi-functional protein playing both structural and signaling roles. β-Catenin is composed of three distinct domains: N-terminal domain, C-terminal domain and a central armadillo repeat domain. N-terminal domain of β-catenin harbours almost all of the cancer causing mutations, thus deciphering its critical structural and functional roles offers great potential in cancer detection and therapy. Here, in this review, we have collected information from pharmacological analysis, bio-physical and structural studies, molecular modeling, in-vivo and in-vitro assays, and transgenic animal experiments employing various N-terminal domain variants of β-catenin to discuss the interaction of β-catenin with its binding partners that specifically interact with this domain and the implications of these interactions on signaling, cell fate determination, and in tumorigenesis. A thorough understanding of interactions between β-catenin and its binding partners will enable us to more effectively understand how β-catenin switches between its multiple roles, and will lead to the development of specific assays for the identification of small molecules as chemotherapeutic agents to treat diseases, including cancer and neurological disorders, where Wnt/β-catenin signaling is dysregulated.
Collapse
Affiliation(s)
- Mohd Saleem Dar
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, India; Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, J&K, India
| | - Paramjeet Singh
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, India; Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, J&K, India
| | - Riyaz A Mir
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Ansari Nagar, New Delhi, India
| | - Mohd Jamal Dar
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, India; Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, J&K, India.
| |
Collapse
|
13
|
Anzilotti S, Tornincasa M, Gerlini R, Conte A, Brancaccio P, Cuomo O, Bianco G, Fusco A, Annunziato L, Pignataro G, Pierantoni GM. Genetic ablation of homeodomain-interacting protein kinase 2 selectively induces apoptosis of cerebellar Purkinje cells during adulthood and generates an ataxic-like phenotype. Cell Death Dis 2015; 6:e2004. [PMID: 26633710 PMCID: PMC4720876 DOI: 10.1038/cddis.2015.298] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 08/27/2015] [Accepted: 08/28/2015] [Indexed: 12/12/2022]
Abstract
Homeodomain-interacting protein kinase 2 (HIPK2) is a multitalented coregulator of an increasing number of transcription factors and cofactors involved in cell death and proliferation in several organs and systems. As Hipk2−/− mice show behavioral abnormalities consistent with cerebellar dysfunction, we investigated whether Hipk2 is involved in these neurological symptoms. To this aim, we characterized the postnatal developmental expression profile of Hipk2 in the brain cortex, hippocampus, striatum, and cerebellum of mice by real-time PCR, western blot analysis, and immunohistochemistry. Notably, we found that whereas in the brain cortex, hippocampus, and striatum, HIPK2 expression progressively decreased with age, that is, from postnatal day 1 to adulthood, it increased in the cerebellum. Interestingly, mice lacking Hipk2 displayed atrophic lobules and a visibly smaller cerebellum than did wild-type mice. More important, the cerebellum of Hipk2−/− mice showed a strong reduction in cerebellar Purkinje neurons during adulthood. Such reduction is due to the activation of an apoptotic process associated with a compromised proteasomal function followed by an unpredicted accumulation of ubiquitinated proteins. In particular, Purkinje cell dysfunction was characterized by a strong accumulation of ubiquitinated β-catenin. Moreover, our behavioral tests showed that Hipk2−/− mice displayed muscle and balance impairment, indicative of Hipk2 involvement in cerebellar function. Taken together, these results indicate that Hipk2 exerts a relevant role in the survival of cerebellar Purkinje cells and that Hipk2 genetic ablation generates cerebellar dysfunction compatible with an ataxic-like phenotype.
Collapse
Affiliation(s)
| | - M Tornincasa
- Institute of Endocrinology and Experimental Oncology of National Research Council and Department of Molecular Medicine and Medical Biotechnology, School of Medicine, 'Federico II' University of Naples, Naples, Italy
| | - R Gerlini
- Institute of Endocrinology and Experimental Oncology of National Research Council and Department of Molecular Medicine and Medical Biotechnology, School of Medicine, 'Federico II' University of Naples, Naples, Italy
| | - A Conte
- Institute of Endocrinology and Experimental Oncology of National Research Council and Department of Molecular Medicine and Medical Biotechnology, School of Medicine, 'Federico II' University of Naples, Naples, Italy
| | - P Brancaccio
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, 'Federico II' University of Naples, Naples, Italy
| | - O Cuomo
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, 'Federico II' University of Naples, Naples, Italy
| | - G Bianco
- Institute of Endocrinology and Experimental Oncology of National Research Council and Department of Molecular Medicine and Medical Biotechnology, School of Medicine, 'Federico II' University of Naples, Naples, Italy
| | - A Fusco
- Institute of Endocrinology and Experimental Oncology of National Research Council and Department of Molecular Medicine and Medical Biotechnology, School of Medicine, 'Federico II' University of Naples, Naples, Italy
| | - L Annunziato
- SDN IRCCS, Naples, Italy.,Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, 'Federico II' University of Naples, Naples, Italy
| | - G Pignataro
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, 'Federico II' University of Naples, Naples, Italy
| | - G M Pierantoni
- Institute of Endocrinology and Experimental Oncology of National Research Council and Department of Molecular Medicine and Medical Biotechnology, School of Medicine, 'Federico II' University of Naples, Naples, Italy
| |
Collapse
|
14
|
Tan M, Gong H, Zeng Y, Tao L, Wang J, Jiang J, Xu D, Bao E, Qiu J, Liu Z. Downregulation of homeodomain-interacting protein kinase-2 contributes to bladder cancer metastasis by regulating Wnt signaling. J Cell Biochem 2015; 115:1762-7. [PMID: 24824041 DOI: 10.1002/jcb.24842] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 04/25/2014] [Accepted: 05/09/2014] [Indexed: 11/07/2022]
Abstract
Homeodomain-interacting protein kinase-2 (Hipk2) has been shown to have important regulatory roles in cancer biology, such as cancer cell proliferation, cell cycle, and cell invasion. However, the contributions of Hipk2 to bladder cancer metastasis remain largely unknown. In the current study, we assayed the expression level of Hipk2 in bladder cancer tissues by real-time PCR, and defined its biological functions. We found that Hipk2 levels were downregulated in most bladder cancer tissues compared with adjacent normal tissues, and Hipk2 levels were remarkably decreased in metastasized tumor tissues when compared with primary tumors. SiRNA-mediated Hipk2 silencing increased bladder cancer cell invasion. Hipk2 knockdown resulted in decrease of E-cadherin expression and increase of N-cadherin and fibronectin expression, indicated that epithelial-mesenchymal transition (EMT) was induced. We further demonstrated that Hipk2 knockdown induced Wnt signaling activation and β-catenin nuclear localization. Finally, we confirmed that Hipk2 inhibition promoted EMT and subsequent cell invasion, at least in part by activating Wnt signaling. These data suggest an important role of Hipk2 in regulating metastasis of bladder cancer and implicate the potential application of Hipk2 in bladder cancer therapy.
Collapse
Affiliation(s)
- Mingyue Tan
- Department of Urology, Shanghai First People's Hospital, Medical College of Shanghai Jiao Tong University, Shanghai, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Dewald DN, Steinmetz EL, Walldorf U. Homeodomain-interacting protein kinase (Hipk) phosphorylates the small SPOC family protein Spenito. INSECT MOLECULAR BIOLOGY 2014; 23:706-719. [PMID: 25040100 DOI: 10.1111/imb.12117] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The Drosophila homeodomain-interacting protein kinase (Hipk) is a versatile regulator involved in a variety of pathways, such as Notch and Wingless signalling, thereby acting in processes including the promotion of eye development or control of cell numbers in the nervous system. In vertebrates, extensive studies have related its homologue HIPK2 to important roles in the control of p53-mediated apoptosis and tumour suppression. Spenito (Nito) belongs to the group of small SPOC family proteins and has a role, amongst others, as a regulator of Wingless signalling downstream of Armadillo. In the present study, we show that both proteins have an enzyme-substrate relationship, adding a new interesting component to the broad range of Hipk interactions, and we map several phosphorylation sites of Nito. Furthermore, we were able to define a preliminary consensus motif for Hipk target sites, which will simplify the identification of new substrates of this kinase.
Collapse
Affiliation(s)
- D N Dewald
- Developmental Biology, Saarland University, Homburg, Germany
| | | | | |
Collapse
|
16
|
Zhou L, Feng Y, Jin Y, Liu X, Sui H, Chai N, Chen X, Liu N, Ji Q, Wang Y, Li Q. Verbascoside promotes apoptosis by regulating HIPK2-p53 signaling in human colorectal cancer. BMC Cancer 2014; 14:747. [PMID: 25282590 PMCID: PMC4197337 DOI: 10.1186/1471-2407-14-747] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 09/26/2014] [Indexed: 01/18/2023] Open
Abstract
Background We investigated the role of the HIPK2–p53 signaling pathway in tumorigenesis and resistance to the drug Verbascoside (VB) in colorectal cancer (CRC), using in vivo and in vitro experiments. Methods Primary human CRC samples and normal intestinal tissues from patients were analyzed for HIPK2 expression by immunohistochemistry (IHC) and its expression was correlated against patients’ clinicopathological characteristics. Human CRC HCT-116 cells were implanted in BALB/c nude mice; mice with xenografted tumors were randomly administrated vehicle (control), 20, 40, or 80 mg/mL VB, or 1 mg/mL fluorouracil (5-FU). HIPK2, p53, Bax, and Bcl-2 expression in these tumors were determined by IHC. In vitro effects of VB on CRC cell proliferation and apoptosis were measured by CCK-8 assay and flow cytometry; HIPK2, p53, p-p53, Bax, and Bcl-2 were measured by western blot. Results IHC analysis for 100 human CRC tumor samples and 20 normal intestinal tissues, showed HIPK2 expression to inversely correlate with Dukes stage and depth of invasion in CRC (P < 0.05). In vivo, the inhibition rates of 20, 40, and 80 mg/mL VB on CRC xenograft tumor weight were 42.79%, 53.90%, and 60.99%, respectively, and were accompanied by increased expression of HIPK2, p53, and Bax, and decreased Bcl-2 expression in treated tumors. In vitro, VB significantly inhibited proliferation of CRC cell lines HCT-116, HT-29, LoVo, and SW620, in a time- and dose-dependent manner. The apoptosis rates of 25, 50, and 100 μM VB on HCT-116 cells were 10.83 ± 1.28, 11.25 ± 1.54, and 20.19 ± 2.87%, and on HT-29 cells were 18.92 ± 6.12, 21.57 ± 4.05, and 25.14 ± 6.73%, respectively. In summary, VB treatment significantly enhanced the protein expression of pro-apoptotic HIPK2, p53, p-p53, Bax, and decreased anti-apoptotic Bcl-2 expression in CRC cells. Conclusions HIPK2 protein modulates the phosphorylation status of p53, and levels of Bax and Bcl-2 in CRC. We also found that VB effectively activated the HIPK2–p53 signaling pathway, resulting in increased CRC cell apoptosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Qi Li
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
17
|
Shimizu N, Ishitani S, Sato A, Shibuya H, Ishitani T. Hipk2 and PP1c Cooperate to Maintain Dvl Protein Levels Required for Wnt Signal Transduction. Cell Rep 2014; 8:1391-404. [DOI: 10.1016/j.celrep.2014.07.040] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 06/04/2014] [Accepted: 07/23/2014] [Indexed: 11/30/2022] Open
|
18
|
Mbom BC, Siemers KA, Ostrowski MA, Nelson WJ, Barth AIM. Nek2 phosphorylates and stabilizes β-catenin at mitotic centrosomes downstream of Plk1. Mol Biol Cell 2014; 25:977-91. [PMID: 24501426 PMCID: PMC3967981 DOI: 10.1091/mbc.e13-06-0349] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 01/13/2014] [Accepted: 01/27/2014] [Indexed: 12/27/2022] Open
Abstract
β-Catenin is a multifunctional protein with critical roles in cell-cell adhesion, Wnt signaling, and the centrosome cycle. Whereas the regulation of β-catenin in cell-cell adhesion and Wnt signaling are well understood, how β-catenin is regulated at the centrosome is not. NIMA-related protein kinase 2 (Nek2), which regulates centrosome disjunction/splitting, binds to and phosphorylates β-catenin. Using in vitro and cell-based assays, we show that Nek2 phosphorylates the same regulatory sites in the N-terminus of β-catenin as glycogen synthase kinase 3β (GSK3β), which are recognized by a specific phospho-S33/S37/T41 antibody, as well as additional sites. Nek2 binding to β-catenin appears to inhibit binding of the E3 ligase β-TrCP and prevents β-catenin ubiquitination and degradation. Thus β-catenin phosphorylated by Nek2 is stabilized and accumulates at centrosomes in mitosis. We further show that polo-like kinase 1 (Plk1) regulates Nek2 phosphorylation and stabilization of β-catenin. Taken together, these results identify a novel mechanism for regulating β-catenin stability that is independent of GSK3β and provide new insight into a pathway involving Plk1, Nek2, and β-catenin that regulates the centrosome cycle.
Collapse
Affiliation(s)
| | | | | | - W. James Nelson
- Department of Biology, Stanford University, Stanford, CA 94305
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305
| | | |
Collapse
|
19
|
Zhou LH, Hu Q, Chen XZ, Liu X, Sui H, Fu XL, Ren JL, Li Q. Verbascoside exerts therapeutic effects against colorectal cancer xenografts in nude mice via HIPK2-P53 pathway. Shijie Huaren Xiaohua Zazhi 2014; 22:171-178. [DOI: 10.11569/wcjd.v22.i2.171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AIM: To investigate whether verbascoside exerts therapeutic effects against colorectal cancer xenografts in nude mice and the possible role of the HIPK2-P53 pathway in this process.
METHODS: An ectopic nude mice model of colorectal cancer was established by subcutaneously inoculating human colorectal carcinoma HCT-116 cells into the armpit of nude mice. The mice were then randomly divided into 5 groups: a model control group, low- [20 mg/(kg·d)], medium- [40 mg/(kg·d)], high-dose verbascoside groups [80 mg/(kg·d)], and a 5-fluorouracil (5-FU) group [1 mg/(kg·d)]. Drugs were injected intraperitoneally for two weeks. Two weeks later, the mice were sacrificed and the tumors were peeled off to measure tumor size and weight. The expression of apoptosis-related proteins HIPK2, P53, Bax, and Bcl-2 in tumor tissues was tested by immunohistochemistry.
RESULTS: Compared with the model control group, tumor size decreased by 48.41%, 61.04%, 63.75%, and 75.14% in the low-, medium-, high-dose verbascoside groups and the 5-FU group, respectively, and tumor weight decreased by 42.79%, 53.90%, 60.99%, and 66.19%. The relative expression levels of HIPK2 in tumor tissues were 4.83 ± 0.62, 8.46 ± 0.99, 11.90 ± 1.21 and 13.50 ± 0.94, in the low-, medium-, high-dose verbascoside groups and the 5-FU group, respectively. The corresponding values were 14.59 ± 0.90, 17.60 ± 1.40, 23.10 ± 2.10 and 22.44 ± 2.05 for P53, 14.41 ± 0.38, 15.84 ± 0.54, 26.28 ± 0.55 and 26.34 ± 2.33 for Bax, and 14.08 ± 1.04, 11.93 ± 0.93, 7.48 ± 0.86 and 5.46 ± 0.67 for Bcl-2.
CONCLUSION: Verbascoside can inhibit the growth of human colorectal cancer xenografts in mice, up-regulate the expression of HIPK2, P53 and Bax, and down-regulate Bcl-2 expression. The inhibition of colorectal cancer by verbascoside was closely with the promotion of tumor cell apoptosis.
Collapse
|
20
|
Tomimaru Y, Koga H, Shin TH, Xu CQ, Wands JR, Kim M. The SxxSS motif of T-cell factor-4 isoforms modulates Wnt/β-catenin signal activation in hepatocellular carcinoma cells. Cancer Lett 2013; 336:359-69. [PMID: 23562475 PMCID: PMC3700609 DOI: 10.1016/j.canlet.2013.03.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 03/21/2013] [Accepted: 03/24/2013] [Indexed: 12/25/2022]
Abstract
T-cell factor (TCF) proteins represent key transcription factors in Wnt signaling. We show that the SxxSS motif in TCF-4 regulates transcriptional activity in HCC cells. TCF-4K mutants increased transcriptional activity compared to TCF-4K (bearing the SxxSS); the binding pattern of co-factors in TCF-4K mutants was similar to that in TCF-4J (lacking the SxxSS). TCF activity in TCF-4K cells was suppressed by homeodomain-interacting protein kinase 2 (HIPK2), but not in TCF-4J cells. Together, our data indicates that the SxxSS motif in TCF-4K regulates transcriptional activity by modifying co-factors in the β-catenin/TCF-4 transcriptional complex and these events may be mediated through HIPK2.
Collapse
Affiliation(s)
- Yoshito Tomimaru
- Liver Research Center, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | | | | | | | | | | |
Collapse
|
21
|
Siepi F, Gatti V, Camerini S, Crescenzi M, Soddu S. HIPK2 catalytic activity and subcellular localization are regulated by activation-loop Y354 autophosphorylation. BIOCHIMICA ET BIOPHYSICA ACTA 2013; 1833:1443-1453. [PMID: 23485397 PMCID: PMC3787740 DOI: 10.1016/j.bbamcr.2013.02.018] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Revised: 01/21/2013] [Accepted: 02/18/2013] [Indexed: 12/20/2022]
Abstract
HIPK2 (homeodomain-interacting protein kinase-2) binds to and phosphorylates, at Ser and Thr residues, a large number of targets involved in cell division and cell fate decision in response to different physiological or stress stimuli. Inactivation of HIPK2 has been observed in human and mouse cancers supporting its role as a tumor suppressor. Despite the biological relevance of this kinase, very little is known on how HIPK2 becomes catalytically active. Based on sequence homologies, HIPK2 has been taxonomically classified as a subfamily member of the dual-specificity tyrosine-regulated kinases (DYRKs) and the activation-loop Y354 of HIPK2 has been found phosphorylated in different cells; however, the relevance of this Y phosphorylation is presently unknown. Here, we show that HIPK2, which is extensively phosphorylated at S/T sites throughout its functional domains, becomes catalytically active by autophosphorylation at the activation-loop Y354. In particular, we found that, in analogy to DYRKs, HIPK2-Y354 phosphorylation is an autocatalytic event and its prevention, through Y354 substitution with non-phosphorylatable amino acids or by using the kinase inhibitor purvalanol A, induces a strong reduction of the HIPK2 S/T-kinase activity on different substrates. Interestingly, at variance from DYRKs, inhibition of HIPK2-Y354 phosphorylation induces a strong out-of-target Y-kinase activity in cis and a strong cytoplasmic relocalization of the kinase. Together, these results demonstrate that the catalytic activity, substrate specificity, and subcellular localization of HIPK2 are regulated by autophosphorylation of its activation-loop Y354.
Collapse
Affiliation(s)
- Francesca Siepi
- Dipartimento di Oncologia Sperimentale, Istituto Nazionale Tumori Regina Elena, 00158, Roma, Italy
| | - Veronica Gatti
- Dipartimento di Oncologia Sperimentale, Istituto Nazionale Tumori Regina Elena, 00158, Roma, Italy
| | - Serena Camerini
- Dipartimento di Ematologia, Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, 00161, Roma, Italy
| | - Marco Crescenzi
- Dipartimento di Biologia Cellulare e Neuroscienze, Istituto Superiore di Sanità, 00161, Roma, Italy
| | - Silvia Soddu
- Dipartimento di Oncologia Sperimentale, Istituto Nazionale Tumori Regina Elena, 00158, Roma, Italy
| |
Collapse
|
22
|
Abstract
The Wnt pathway is a major embryonic signaling pathway that controls cell proliferation, cell fate, and body-axis determination in vertebrate embryos. Soon after egg fertilization, Wnt pathway components play a role in microtubule-dependent dorsoventral axis specification. Later in embryogenesis, another conserved function of the pathway is to specify the anteroposterior axis. The dual role of Wnt signaling in Xenopus and zebrafish embryos is regulated at different developmental stages by distinct sets of Wnt target genes. This review highlights recent progress in the discrimination of different signaling branches and the identification of specific pathway targets during vertebrate axial development.
Collapse
Affiliation(s)
- Hiroki Hikasa
- Division of Cancer Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | | |
Collapse
|
23
|
Shojima N, Hara K, Fujita H, Horikoshi M, Takahashi N, Takamoto I, Ohsugi M, Aburatani H, Noda M, Kubota N, Yamauchi T, Ueki K, Kadowaki T. Depletion of homeodomain-interacting protein kinase 3 impairs insulin secretion and glucose tolerance in mice. Diabetologia 2012; 55:3318-30. [PMID: 22983607 DOI: 10.1007/s00125-012-2711-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Accepted: 08/07/2012] [Indexed: 01/13/2023]
Abstract
AIMS/HYPOTHESIS Insufficient insulin secretion and reduced pancreatic beta cell mass are hallmarks of type 2 diabetes. Here, we focused on a family of serine-threonine kinases known as homeodomain-interacting protein kinases (HIPKs). HIPKs are implicated in the modulation of Wnt signalling, which plays a crucial role in transcriptional activity, and in pancreas development and maintenance. The aim of the present study was to characterise the role of HIPKs in glucose metabolism. METHODS We used RNA interference to characterise the role of HIPKs in regulating insulin secretion and transcription activity. We conducted RT-PCR and western blot analyses to analyse the expression and abundance of HIPK genes and proteins in the islets of high-fat diet-fed mice. Glucose-induced insulin secretion and beta cell proliferation were measured in islets from Hipk3 ( -/- ) mice, which have impaired glucose tolerance owing to an insulin secretion deficiency. The abundance of pancreatic duodenal homeobox (PDX)-1 and glycogen synthase kinase (GSK)-3β phosphorylation in Hipk3 ( -/- ) islets was determined by immunohistology and western blot analyses. RESULTS We found that HIPKs regulate insulin secretion and transcription activity. Hipk3 expression was most significantly increased in the islets of high-fat diet-fed mice. Furthermore, glucose-induced insulin secretion and beta cell proliferation were decreased in the islets of Hipk3 ( -/- ) mice. Levels of PDX1 and GSK-3β phosphorylation were significantly decreased in Hipk3 ( -/- ) islets. CONCLUSIONS/INTERPRETATION Depletion of HIPK3 impairs insulin secretion and glucose tolerance. Decreased levels of HIPK3 may play a substantial role in the pathogenesis of type 2 diabetes.
Collapse
Affiliation(s)
- N Shojima
- Department of Diabetes and Metabolic Disease, Graduate School of Medicine, University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Garufi A, Pistritto G, Ceci C, Di Renzo L, Santarelli R, Faggioni A, Cirone M, D’Orazi G. Targeting COX-2/PGE(2) pathway in HIPK2 knockdown cancer cells: impact on dendritic cell maturation. PLoS One 2012; 7:e48342. [PMID: 23144866 PMCID: PMC3492329 DOI: 10.1371/journal.pone.0048342] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 09/24/2012] [Indexed: 02/04/2023] Open
Abstract
Background Homeodomain-interacting protein kinase 2 (HIPK2) is a multifunctional protein that exploits its kinase activity to modulate key molecular pathways in cancer to restrain tumor growth and induce response to therapies. For instance, HIPK2 knockdown induces upregulation of oncogenic hypoxia-inducible factor-1 (HIF-1) activity leading to a constitutive hypoxic and angiogenic phenotype with increased tumor growth in vivo. HIPK2 inhibition, therefore, releases pathways leading to production of pro-inflammatory molecules such as vascular endothelial growth factor (VEGF) or prostaglandin E2 (PGE2). Tumor-produced inflammatory mediators other than promote tumour growth and vascular development may permit evasion of anti-tumour immune responses. Thus, dendritic cells (DCs) dysfunction induced by tumor-produced molecules, may allow tumor cells to escape immunosurveillance. Here we evaluated the molecular mechanism of PGE2 production after HIPK2 depletion and how to modulate it. Methodology/Principal findings We show that HIPK2 knockdown in colon cancer cells resulted in cyclooxygenase-2 (COX-2) upregulation and COX-2-derived PGE2 generation. At molecular level, COX-2 upregulation depended on HIF-1 activity. We previously reported that zinc treatment inhibits HIF-1 activity. Here, zinc supplementation to HIPK2 depleted cells inhibited HIF-1-induced COX-2 expression and PGE2/VEGF production. At translational level, while conditioned media of both siRNA control and HIPK2 depleted cells inhibited DCs maturation, conditioned media of only zinc-treated HIPK2 depleted cells efficiently restored DCs maturation, seen as the expression of co-stimulatory molecules CD80 and CD86, cytokine IL-10 release, and STAT3 phosphorylation. Conclusion/Significance These findings show that: 1) HIPK2 knockdown induced COX-2 upregulation, mostly depending on HIF-1 activity; 2) zinc treatment downregulated HIF-1-induced COX-2 and inhibited PGE2/VEGF production; and 3) zinc treatment of HIPK2 depleted cells restored DCs maturation.
Collapse
Affiliation(s)
- Alessia Garufi
- Department of Experimental Oncology, Molecular Oncogenesis Laboratory, Regina Elena National Cancer Institute, Rome, Italy
| | - Giuseppa Pistritto
- Department of Neuroscience, Section of Pharmacology, University “Tor Vergata”, Rome, Italy
| | - Claudia Ceci
- Department of Neuroscience, Section of Pharmacology, University “Tor Vergata”, Rome, Italy
| | - Livia Di Renzo
- Department of Experimental Medicine, Institute Pasteur-Foundation Cenci Bolognetti, “Sapienza” University, Rome, Italy
| | - Roberta Santarelli
- Department of Experimental Medicine, Institute Pasteur-Foundation Cenci Bolognetti, “Sapienza” University, Rome, Italy
| | - Alberto Faggioni
- Department of Experimental Medicine, Institute Pasteur-Foundation Cenci Bolognetti, “Sapienza” University, Rome, Italy
| | - Mara Cirone
- Department of Experimental Medicine, Institute Pasteur-Foundation Cenci Bolognetti, “Sapienza” University, Rome, Italy
- * E-mail: (GD); (MC)
| | - Gabriella D’Orazi
- Department of Experimental Oncology, Molecular Oncogenesis Laboratory, Regina Elena National Cancer Institute, Rome, Italy
- Department of Medical, Oral and Biotechnological Sciences, University “G. d’Annunzio”, Chieti, Italy
- * E-mail: (GD); (MC)
| |
Collapse
|
25
|
Becker W. Emerging role of DYRK family protein kinases as regulators of protein stability in cell cycle control. Cell Cycle 2012; 11:3389-94. [PMID: 22918246 PMCID: PMC3466549 DOI: 10.4161/cc.21404] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Dual-specificity tyrosine phosphorylation-regulated kinases (DYRKs) constitute an evolutionarily conserved family of protein kinases with key roles in the control of cell proliferation and differentiation. Members of the DYRK family phosphorylate many substrates, including critical regulators of the cell cycle. A recent report revealed that human DYRK2 acts as a negative regulator of G1/S transition by phosphorylating c-Jun and c-Myc, thereby inducing ubiquitination-mediated degradation. Other DYRKs also function as cell cycle regulators by modulating the turnover of their target proteins. DYRK1B can induce reversible cell arrest in a quiescent G0 state by targeting cyclin D1 for proteasomal degradation and stabilizing p27 (Kip1). The DYRK2 ortholog of C. elegans, MBK-2, triggers the proteasomal destruction of oocyte proteins after meiosis to allow the mitotic divisions in embryo development. This review summarizes the accumulating results that provide evidence for a general role of DYRKs in the regulation of protein stability.
Collapse
Affiliation(s)
- Walter Becker
- Institute of Pharmacology and Toxicology, Medical Faculty of the RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
26
|
Hong JY, Park JI, Lee M, Muñoz WA, Miller RK, Ji H, Gu D, Ezan J, Sokol SY, McCrea PD. Down's-syndrome-related kinase Dyrk1A modulates the p120-catenin-Kaiso trajectory of the Wnt signaling pathway. J Cell Sci 2012; 125:561-9. [PMID: 22389395 DOI: 10.1242/jcs.086173] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The Wnt pathways contribute to many processes in cancer and development, with β-catenin being a key canonical component. p120-catenin, which is structurally similar to β-catenin, regulates the expression of certain Wnt target genes, relieving repression conferred by the POZ- and zinc-finger-domain-containing transcription factor Kaiso. We have identified the kinase Dyrk1A as a component of the p120-catenin-Kaiso trajectory of the Wnt pathway. Using rescue and other approaches in Xenopus laevis embryos and mammalian cells, we found that Dyrk1A positively and selectively modulates p120-catenin protein levels, thus having an impact on p120-catenin and Kaiso (and canonical Wnt) gene targets such as siamois and wnt11. The Dyrk1A gene resides within the Down's syndrome critical region, which is amplified in Down's syndrome. A consensus Dyrk phosphorylation site in p120-catenin was identified, with a mutant mimicking phosphorylation exhibiting the predicted enhanced capacity to promote endogenous Wnt-11 and Siamois expression, and gastrulation defects. In summary, we report the biochemical and functional relationship of Dyrk1A with the p120-catenin-Kaiso signaling trajectory, with a linkage to canonical Wnt target genes. Conceivably, this work might also prove relevant to understanding the contribution of Dyrk1A dosage imbalance in Down's syndrome.
Collapse
Affiliation(s)
- Ji Yeon Hong
- Department of Biochemistry and Molecular Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, TX 77030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
D'Orazi G, Rinaldo C, Soddu S. Updates on HIPK2: a resourceful oncosuppressor for clearing cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2012; 31:63. [PMID: 22889244 PMCID: PMC3432601 DOI: 10.1186/1756-9966-31-63] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 07/27/2012] [Indexed: 02/04/2023]
Abstract
Homeodomain-interacting protein kinase 2 (HIPK2) is a multitalented protein that exploits its kinase activity to modulate key molecular pathways in cancer to restrain tumor growth and induce response to therapies. HIPK2 phosphorylates oncosuppressor p53 for apoptotic activation. In addition, also p53-independent apoptotic pathways are regulated by HIPK2 and can be exploited for anticancer purpose too. Therefore, HIPK2 activity is considered a central switch in targeting tumor cells toward apoptosis upon genotoxic damage and the preservation and/or restoration of HIPK2 function is crucial for an efficient tumor response to therapies. As a proof of principle, HIPK2 knockdown impairs p53 function, induces chemoresistance, angiogenesis, and tumor growth in vivo, on the contrary, HIPK2 overexpression activates apoptotic pathways, counteracts hypoxia, inhibits angiogenesis, and induces chemosensitivity both in p53-dependent and -independent ways. The role of HIPK2 in restraining tumor development was also confirmed by studies with HIPK2 knockout mice. Recent findings demonstrated that HIPK2 inhibitions do exist in tumors and depend by several mechanisms including HIPK2 cytoplasmic localization, protein degradation, and loss of heterozygosity (LOH), recapitulating the biological outcome obtained by RNA interference studies in tumor cells, such as p53 inactivation, resistance to therapies, apoptosis inhibition, and tumor progression. These findings may lead to new diagnostic and therapeutic approaches for treating cancer patients. This review will focus on the last updates about HIPK2 contribution in tumorigenesis and cancer treatment.
Collapse
Affiliation(s)
- Gabriella D'Orazi
- Department of Medical, Oral, and Biotechnological Sciences, University "G, d'Annunzio", Chieti 66013, Italy.
| | | | | |
Collapse
|
28
|
Kirby LA, Schott JT, Noble BL, Mendez DC, Caseley PS, Peterson SC, Routledge TJ, Patel NV. Glycogen synthase kinase 3 (GSK3) inhibitor, SB-216763, promotes pluripotency in mouse embryonic stem cells. PLoS One 2012; 7:e39329. [PMID: 22745733 PMCID: PMC3383737 DOI: 10.1371/journal.pone.0039329] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Accepted: 05/18/2012] [Indexed: 11/18/2022] Open
Abstract
Canonical Wnt/β-catenin signaling has been suggested to promote self-renewal of pluripotent mouse and human embryonic stem cells. Here, we show that SB-216763, a glycogen synthase kinase-3 (GSK3) inhibitor, can maintain mouse embryonic stem cells (mESCs) in a pluripotent state in the absence of exogenous leukemia inhibitory factor (LIF) when cultured on mouse embryonic fibroblasts (MEFs). MESCs maintained with SB-216763 for one month were morphologically indistinguishable from LIF-treated mESCs and expressed pluripotent-specific genes Oct4, Sox2, and Nanog. Furthermore, Nanog immunostaining was more homogenous in SB-216763-treated colonies compared to LIF. Embryoid bodies (EBs) prepared from these mESCs expressed early-stage markers for all three germ layers, and could efficiently differentiate into cardiac-like cells and MAP2-immunoreactive neurons. To our knowledge, SB-216763 is the first GSK3 inhibitor that can promote self-renewal of mESC co-cultured with MEFs for more than two months.
Collapse
Affiliation(s)
- Leslie A. Kirby
- Department of Biological Science, California State University – Fullerton, Fullerton, California, United States of America
| | - Jason T. Schott
- Department of Biological Science, California State University – Fullerton, Fullerton, California, United States of America
| | - Brenda L. Noble
- Department of Biological Science, California State University – Fullerton, Fullerton, California, United States of America
| | - Daniel C. Mendez
- Department of Biological Science, California State University – Fullerton, Fullerton, California, United States of America
| | - Paul S. Caseley
- Department of Biological Science, California State University – Fullerton, Fullerton, California, United States of America
| | - Sarah C. Peterson
- Department of Biological Science, California State University – Fullerton, Fullerton, California, United States of America
| | - Tyler J. Routledge
- Department of Biological Science, California State University – Fullerton, Fullerton, California, United States of America
| | - Nilay V. Patel
- Department of Biological Science, California State University – Fullerton, Fullerton, California, United States of America
- Center for Applied Biotechnology Studies, California State University – Fullerton, Fullerton, California, United States of America
- * E-mail:
| |
Collapse
|
29
|
Rinaldo C, Moncada A, Gradi A, Ciuffini L, D'Eliseo D, Siepi F, Prodosmo A, Giorgi A, Pierantoni GM, Trapasso F, Guarguaglini G, Bartolazzi A, Cundari E, Schininà ME, Fusco A, Soddu S. HIPK2 controls cytokinesis and prevents tetraploidization by phosphorylating histone H2B at the midbody. Mol Cell 2012; 47:87-98. [PMID: 22658722 DOI: 10.1016/j.molcel.2012.04.029] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 03/29/2012] [Accepted: 04/26/2012] [Indexed: 11/25/2022]
Abstract
Failure in cytokinesis, the final step in cell division, by generating tetra- and polyploidization promotes chromosomal instability, a hallmark of cancer. Here we show that HIPK2, a kinase involved in cell fate decisions in development and response to stress, controls cytokinesis and prevents tetraploidization through its effects on histone H2B. HIPK2 binds and phosphorylates histone H2B at S14 (H2B-S14(P)), and the two proteins colocalize at the midbody. HIPK2 depletion by targeted gene disruption or RNA interference results in loss of H2B-S14(P) at the midbody, prevention of cell cleavage, and tetra- and polyploidization. In HIPK2 null cells, restoration of wild-type HIPK2 activity or expression of a phosphomimetic H2B-S14D derivative abolishes cytokinesis defects and rescues cell proliferation, showing that H2B-S14(P) is required for a faithful cytokinesis. Overall, our data uncover mechanisms of a critical HIPK2 function in cytokinesis and in the prevention of tetraploidization.
Collapse
Affiliation(s)
- Cinzia Rinaldo
- Laboratorio di Oncogenesi Molecolare, Dipartimento di Oncologia Sperimentale, Istituto Nazionale dei Tumori Regina Elena, 00158 Rome, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
The many faces and functions of β-catenin. EMBO J 2012; 31:2714-36. [PMID: 22617422 DOI: 10.1038/emboj.2012.150] [Citation(s) in RCA: 1265] [Impact Index Per Article: 97.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 04/30/2012] [Indexed: 02/07/2023] Open
Abstract
β-Catenin (Armadillo in Drosophila) is a multitasking and evolutionary conserved molecule that in metazoans exerts a crucial role in a multitude of developmental and homeostatic processes. More specifically, β-catenin is an integral structural component of cadherin-based adherens junctions, and the key nuclear effector of canonical Wnt signalling in the nucleus. Imbalance in the structural and signalling properties of β-catenin often results in disease and deregulated growth connected to cancer and metastasis. Intense research into the life of β-catenin has revealed a complex picture. Here, we try to capture the state of the art: we try to summarize and make some sense of the processes that regulate β-catenin, as well as the plethora of β-catenin binding partners. One focus will be the interaction of β-catenin with different transcription factors and the potential implications of these interactions for direct cross-talk between β-catenin and non-Wnt signalling pathways.
Collapse
|
31
|
Abstract
Embryonic signaling pathways often lead to a switch from default repression to transcriptional activation of target genes. A major consequence of Wnt signaling is stabilization of β-catenin, which associates with T-cell factors (TCFs) and 'converts' them from repressors into transcriptional activators. The molecular mechanisms responsible for this conversion remain poorly understood. Several studies have reported on the regulation of TCF by phosphorylation, yet its physiological significance has been unclear: in some cases it appears to promote target gene activation, in others Wnt-dependent transcription is inhibited. This review focuses on recent progress in the understanding of context-dependent post-translational regulation of TCF function by Wnt signaling.
Collapse
|
32
|
[Research progress and application of the homeodomain-interacting protein kinase-2]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2011; 14:373-7. [PMID: 21496439 PMCID: PMC5999714 DOI: 10.3779/j.issn.1009-3419.2011.04.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
33
|
Hikasa H, Sokol SY. Phosphorylation of TCF proteins by homeodomain-interacting protein kinase 2. J Biol Chem 2011; 286:12093-100. [PMID: 21285352 DOI: 10.1074/jbc.m110.185280] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Wnt pathways play essential roles in cell proliferation, morphogenesis, and cell fate specification during embryonic development. According to the consensus view, the Wnt pathway prevents the degradation of the key signaling component β-catenin by the protein complex containing the negative regulators Axin and glycogen synthase kinase 3 (GSK3). Stabilized β-catenin associates with TCF proteins and enters the nucleus to promote target gene expression. This study examines the involvement of HIPK2 (homeodomain-interacting protein kinase 2) in the regulation of different TCF proteins in Xenopus embryos in vivo. We show that the TCF family members LEF1, TCF4, and TCF3 are phosphorylated in embryonic ectoderm after Wnt8 stimulation and HIPK2 overexpression. We also find that TCF3 phosphorylation is triggered by canonical Wnt ligands, LRP6, and dominant negative mutants for Axin and GSK3, indicating that this process shares the same upstream regulators with β-catenin stabilization. HIPK2-dependent phosphorylation caused the dissociation of LEF1, TCF4, and TCF3 from a target promoter in vivo. This result provides a mechanistic explanation for the context-dependent function of HIPK2 in Wnt signaling; HIPK2 up-regulates transcription by phosphorylating TCF3, a transcriptional repressor, but inhibits transcription by phosphorylating LEF1, a transcriptional activator. Finally, we show that upon HIPK2-mediated phosphorylation, TCF3 is replaced with positively acting TCF1 at a target promoter. These observations emphasize a critical role for Wnt/HIPK2-dependent TCF phosphorylation and suggest that TCF switching is an important mechanism of Wnt target gene activation in vertebrate embryos.
Collapse
Affiliation(s)
- Hiroki Hikasa
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | |
Collapse
|
34
|
Regulation of TCF3 by Wnt-dependent phosphorylation during vertebrate axis specification. Dev Cell 2010; 19:521-32. [PMID: 20951344 DOI: 10.1016/j.devcel.2010.09.005] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2009] [Revised: 07/23/2010] [Accepted: 09/07/2010] [Indexed: 11/24/2022]
Abstract
A commonly accepted model of Wnt/β-catenin signaling involves target gene activation by a complex of β-catenin with a T-cell factor (TCF) family member. TCF3 is a transcriptional repressor that has been implicated in Wnt signaling and plays key roles in embryonic axis specification and stem cell differentiation. Here we demonstrate that Wnt proteins stimulate TCF3 phosphorylation in gastrulating Xenopus embryos and mammalian cells. This phosphorylation event involves β-catenin-mediated recruitment of homeodomain-interacting protein kinase 2 (HIPK2) to TCF3 and culminates in the dissociation of TCF3 from a target gene promoter. Mutated TCF3 proteins resistant to Wnt-dependent phosphorylation function as constitutive inhibitors of Wnt-mediated activation of Vent2 and Cdx4 during anteroposterior axis specification. These findings reveal an alternative in vivo mechanism of Wnt signaling that involves TCF3 phosphorylation and subsequent derepression of target genes and link this molecular event to a specific developmental process.
Collapse
|