1
|
Ek CJ, Alkmark M, Baburamani AA, Supramaniam VG, Sood S, Melchiotti R, de Rinaldis E, Hagberg H, Mallard C. Novel biomarkers of preterm brain injury from blood transcriptome in sheep model of intrauterine asphyxia. Pediatr Res 2024; 96:1707-1717. [PMID: 38822135 PMCID: PMC11772238 DOI: 10.1038/s41390-024-03224-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 03/06/2024] [Accepted: 04/02/2024] [Indexed: 06/02/2024]
Abstract
BACKGROUND Infants born preterm have a higher incidence of neurological deficits. A key step in finding effective treatments is to identify biomarkers that reliably predict outcome. METHODS Following umbilical cord occlusion (UCO) in pregnant sheep, whole fetal blood RNA was sequenced pre- and post-UCO, brain injury outcome was determined by battery of neuropathology scoring and the transcriptome signature correlated to the degree of brain injury. Additionally, we developed a novel analytical procedure to deduce cell blood composition over time. RESULTS Sixty-one genes were identified with significant altered expression after UCO. In pre-UCO blood, the level of three mRNAs (Trex2, Znf280b, novel miRNA) and in post-UCO, four mRNAs (Fam184a, Angptl2, novel lincRNA and an unknown protein-coding gene) were associated to brain injury (FDR < 0.01). Several of these mRNAs are related to inflammation and angiogenesis. Pathway analysis highlighted genes playing a role in perinatal death and growth failure. Results also indicate that several leukocyte populations undergo significant changes after UCO. CONCLUSION We have used a whole transcriptomic approach to uncover novel biomarkers in fetal blood that correlate to neuropathology in the preterm sheep brain. The current data forms a basis for future studies to investigate mechanisms of these mRNAs in the injury progression. IMPACT Trend analysis of genes following asphyxia reveal a group of genes associated with perinatal death and growth failure. Several pre-asphyxia transcripts were associated to brain injury severity suggesting genomic susceptibility to injury. Several post-asphyxia transcripts were correlated to brain injury severity, thus, serve as potential novel biomarkers of injury outcome. Successfully adaptation of cell profiling algorithms suggests significant changes in blood cell composition following asphyxia.
Collapse
Affiliation(s)
- C Joakim Ek
- Centre for Perinatal Medicine and Health, Institutes of Neuroscience and Physiology & Clinical Sciences, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden.
| | - Mårten Alkmark
- Centre for Perinatal Medicine and Health, Institutes of Neuroscience and Physiology & Clinical Sciences, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
| | - Ana A Baburamani
- Centre for the Developing Brain, Department of Perinatal Imaging and Health, School of Biomedical Engineering and Imaging Sciences, King's Health Partners, St Thomas' Hospital, London, SE1 7EH, UK
| | - Veena G Supramaniam
- Centre for the Developing Brain, Department of Perinatal Imaging and Health, School of Biomedical Engineering and Imaging Sciences, King's Health Partners, St Thomas' Hospital, London, SE1 7EH, UK
| | - Sanjana Sood
- Department of Cancer Epidemiology and Population Health, King's College London, London, UK
| | - Rossella Melchiotti
- Department of Cancer Epidemiology and Population Health, King's College London, London, UK
| | - Emanuele de Rinaldis
- Department of Cancer Epidemiology and Population Health, King's College London, London, UK
| | - Henrik Hagberg
- Centre for Perinatal Medicine and Health, Institutes of Neuroscience and Physiology & Clinical Sciences, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
- Centre for the Developing Brain, Department of Perinatal Imaging and Health, School of Biomedical Engineering and Imaging Sciences, King's Health Partners, St Thomas' Hospital, London, SE1 7EH, UK
| | - Carina Mallard
- Centre for Perinatal Medicine and Health, Institutes of Neuroscience and Physiology & Clinical Sciences, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
| |
Collapse
|
2
|
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
|
3
|
Fan G, Lu J, Zha J, Guo W, Zhang Y, Liu Y, Zhang L. TAK1 in Vascular Signaling: "Friend or Foe"? J Inflamm Res 2024; 17:3031-3041. [PMID: 38770174 PMCID: PMC11104388 DOI: 10.2147/jir.s458948] [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: 01/10/2024] [Accepted: 04/16/2024] [Indexed: 05/22/2024] Open
Abstract
The maintenance of normal vascular function and homeostasis is largely dependent on the signaling mechanisms that occur within and between cells of the vasculature. TGF-β-activated kinase 1 (TAK1), a multifaceted signaling molecule, has been shown to play critical roles in various tissue types. Although the precise function of TAK1 in the vasculature remains largely unknown, emerging evidence suggests its potential involvement in both physiological and pathological processes. A comprehensive search strategy was employed to identify relevant studies, PubMed, Web of Science, and other relevant databases were systematically searched using keywords related to TAK1, TABs and MAP3K7.In this review, we discussed the role of TAK1 in vascular signaling, with a focus on its function, activation, and related signaling pathways. Specifically, we highlight the TA1-TABs complex is a key factor, regulating vascular smooth muscle cells (VSMCs) and endothelial cells (ECs) involved in the processes of inflammation, vascular proliferation and angiogenesis. This mini review aims to elucidate the evidence supporting TAK1 signaling in the vasculature, in order to better comprehend its beneficial and potential harmful effects upon TAK1 activation in vascular tissue.
Collapse
Affiliation(s)
- Gang Fan
- Department of Urology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, People’s Republic of China
| | - Jingfen Lu
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, People’s Republic of China
| | - Jinhui Zha
- Shenzhen University, Shenzhen, 518000, People’s Republic of China
| | - Weiming Guo
- Department of Urology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, People’s Republic of China
| | - Yifei Zhang
- The First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, People’s Republic of China
| | - Yuxin Liu
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
| | - Liyuan Zhang
- Department of Urology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, People’s Republic of China
| |
Collapse
|
4
|
Zhong W, Hong C, Zhang Y, Li Y, Xiao C, Liu X. ASH2L-mediated H3K4me3 drives diabetic nephropathy through HIPK2 and Notch1 pathway. Transl Res 2024; 264:85-96. [PMID: 37879562 DOI: 10.1016/j.trsl.2023.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 10/22/2023] [Accepted: 10/22/2023] [Indexed: 10/27/2023]
Abstract
Diabetic nephropathy (DN) is one of the complications of diabetes. Long-term hyperglycemia in the kidney results in renal insufficiency, and eventually leads to end-stage renal disease. Epigenetic factor ASH2L has long been identified as a transcriptional activator, and we previously indicated that ASH2L aggravated fibrosis and inflammation in high glucose-induced glomerular mesangial cells, but the pathophysiological relevance and the mechanism of ASH2L-mediated H3K4me3 in DN is not well understood. Here we demonstrated that ASH2L is upregulated in glomeruli isolated from db/db mice. Loss of ASH2L protected glomerular injury caused by hyperglycemia, as evidenced by reduced albuminuria, preserved structure, decreased glomerular extracellular matrix deposition, and lowered renal glomerular expression of proinflammatory and profibrotic markers in db/db mice. Furthermore, we demonstrated that enrichment of ASH2L-mediated H3K4me3 on the promoter regions of ADAM17 and HIPK2 triggered their transcription, leading to aberrant activation of Notch1 signaling pathway, thereby contributing to fibrosis and inflammation in DN. The findings of this study provide compelling evidence for targeting ASH2L as a potential therapeutic strategy to prevent or slow down the progression of DN.
Collapse
Affiliation(s)
- Wen Zhong
- Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai 201203, China
| | - Chen Hong
- Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai 201203, China
| | - Yuyu Zhang
- Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai 201203, China
| | - Yuhui Li
- Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai 201203, China
| | - Chenxi Xiao
- Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai 201203, China
| | - Xinhua Liu
- Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai 201203, China.
| |
Collapse
|
5
|
Ariyasinghe NR, de Souza Santos R, Gross A, Aghamaleky-Sarvestany A, Kreimer S, Escopete S, Parker SJ, Sareen D. Proteomics of novel induced pluripotent stem cell-derived vascular endothelial cells reveal extensive similarity with an immortalized human endothelial cell line. Physiol Genomics 2023; 55:324-337. [PMID: 37306406 PMCID: PMC10396221 DOI: 10.1152/physiolgenomics.00166.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 05/23/2023] [Accepted: 05/27/2023] [Indexed: 06/13/2023] Open
Abstract
The vascular endothelium constitutes the inner lining of the blood vessel, and malfunction and injuries of the endothelium can cause cardiovascular diseases as well as other diseases including stroke, tumor growth, and chronic kidney failure. Generation of effective sources to replace injured endothelial cells (ECs) could have significant clinical impact, and somatic cell sources like peripheral or cord blood cannot credibly supply enough endothelial cell progenitors for multitude of treatments. Pluripotent stem cells are a promising source for a reliable EC supply, which have the potential to restore tissue function and treat vascular diseases. We have developed methods to differentiate induced pluripotent stem cells (iPSCs) efficiently and robustly across multiple iPSC lines into nontissue-specific pan vascular ECs (iECs) with high purity. These iECs present with canonical endothelial cell markers and exhibit measures of endothelial cell functionality with the uptake of Dil fluorescent dye-labeled acetylated low-density lipoprotein (Dil-Ac-LDL) and tube formation. Using proteomic analysis, we revealed that the iECs are more proteomically similar to established human umbilical vein ECs (HUVECs) than to iPSCs. Posttranslational modifications (PTMs) were most shared between HUVECs and iECs, and potential targets for increasing the proteomic similarity of iECs to HUVECs were identified. Here we demonstrate an efficient robust method to differentiate iPSCs into functional ECs, and for the first time provide a comprehensive protein expression profile of iECs, which indicates their similarities with a widely used immortalized HUVECs, allowing for further mechanistic studies of EC development, signaling, and metabolism for future regenerative applications.NEW & NOTEWORTHY We have developed methods to differentiate induced pluripotent stem cells (iPSCs) across multiple iPSC lines into nontissue-specific pan vascular ECs (iECs) and demonstrated the proteomic similarity of these cells to a widely used endothelial cell line (HUVECs). We also identified posttranslational modifications and targets for increasing the proteomic similarity of iECs to HUVECs. In the future, iECs can be used to study EC development, signaling, and metabolism for future regenerative applications.
Collapse
Affiliation(s)
- Nethika R Ariyasinghe
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
- Board of Governors Innovation Center, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Roberta de Souza Santos
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Andrew Gross
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Arwin Aghamaleky-Sarvestany
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Simion Kreimer
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
- Board of Governors Innovation Center, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Sean Escopete
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Sarah J Parker
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
- Board of Governors Innovation Center, Cedars-Sinai Medical Center, Los Angeles, California, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Dhruv Sareen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, California, United States
- iPSC Core, Cedars-Sinai Medical Center, Los Angeles, California, United States
- Board of Governors Innovation Center, Cedars-Sinai Medical Center, Los Angeles, California, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, United States
| |
Collapse
|
6
|
Bei Y, Zhu Y, Wei M, Yin M, Li L, Chen C, Huang Z, Liang X, Gao J, Yao J, van der Kraak PH, Vink A, Lei Z, Dai Y, Chen H, Liang Y, Sluijter JPG, Xiao J. HIPK1 Inhibition Protects against Pathological Cardiac Hypertrophy by Inhibiting the CREB-C/EBPβ Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300585. [PMID: 37098980 PMCID: PMC10288234 DOI: 10.1002/advs.202300585] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/04/2023] [Indexed: 06/19/2023]
Abstract
Inhibition of pathological cardiac hypertrophy is recognized as an important therapeutic strategy for heart failure, although effective targets are still lacking in clinical practice. Homeodomain interacting protein kinase 1 (HIPK1) is a conserved serine/threonine kinase that can respond to different stress signals, however, whether and how HIPK1 regulates myocardial function is not reported. Here, it is observed that HIPK1 is increased during pathological cardiac hypertrophy. Both genetic ablation and gene therapy targeting HIPK1 are protective against pathological hypertrophy and heart failure in vivo. Hypertrophic stress-induced HIPK1 is present in the nucleus of cardiomyocytes, while HIPK1 inhibition prevents phenylephrine-induced cardiomyocyte hypertrophy through inhibiting cAMP-response element binding protein (CREB) phosphorylation at Ser271 and inactivating CCAAT/enhancer-binding protein β (C/EBPβ)-mediated transcription of pathological response genes. Inhibition of HIPK1 and CREB forms a synergistic pathway in preventing pathological cardiac hypertrophy. In conclusion, HIPK1 inhibition may serve as a promising novel therapeutic strategy to attenuate pathological cardiac hypertrophy and heart failure.
Collapse
Affiliation(s)
- Yihua Bei
- Institute of Geriatrics (Shanghai University)Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong)School of MedicineShanghai UniversityNantong226011China
- Cardiac Regeneration and Ageing LabInstitute of Cardiovascular SciencesShanghai Engineering Research Center of Organ RepairSchool of Life ScienceShanghai UniversityShanghai200444China
| | - Yujiao Zhu
- Institute of Geriatrics (Shanghai University)Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong)School of MedicineShanghai UniversityNantong226011China
- Cardiac Regeneration and Ageing LabInstitute of Cardiovascular SciencesShanghai Engineering Research Center of Organ RepairSchool of Life ScienceShanghai UniversityShanghai200444China
| | - Meng Wei
- Institute of Geriatrics (Shanghai University)Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong)School of MedicineShanghai UniversityNantong226011China
- Cardiac Regeneration and Ageing LabInstitute of Cardiovascular SciencesShanghai Engineering Research Center of Organ RepairSchool of Life ScienceShanghai UniversityShanghai200444China
| | - Mingming Yin
- Institute of Geriatrics (Shanghai University)Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong)School of MedicineShanghai UniversityNantong226011China
- Cardiac Regeneration and Ageing LabInstitute of Cardiovascular SciencesShanghai Engineering Research Center of Organ RepairSchool of Life ScienceShanghai UniversityShanghai200444China
| | - Lin Li
- Institute of Geriatrics (Shanghai University)Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong)School of MedicineShanghai UniversityNantong226011China
- Cardiac Regeneration and Ageing LabInstitute of Cardiovascular SciencesShanghai Engineering Research Center of Organ RepairSchool of Life ScienceShanghai UniversityShanghai200444China
| | - Chen Chen
- Institute of Geriatrics (Shanghai University)Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong)School of MedicineShanghai UniversityNantong226011China
- Cardiac Regeneration and Ageing LabInstitute of Cardiovascular SciencesShanghai Engineering Research Center of Organ RepairSchool of Life ScienceShanghai UniversityShanghai200444China
| | - Zhenzhen Huang
- Institute of Geriatrics (Shanghai University)Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong)School of MedicineShanghai UniversityNantong226011China
- Cardiac Regeneration and Ageing LabInstitute of Cardiovascular SciencesShanghai Engineering Research Center of Organ RepairSchool of Life ScienceShanghai UniversityShanghai200444China
| | - Xuchun Liang
- Institute of Geriatrics (Shanghai University)Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong)School of MedicineShanghai UniversityNantong226011China
- Cardiac Regeneration and Ageing LabInstitute of Cardiovascular SciencesShanghai Engineering Research Center of Organ RepairSchool of Life ScienceShanghai UniversityShanghai200444China
| | - Juan Gao
- Institute of Geriatrics (Shanghai University)Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong)School of MedicineShanghai UniversityNantong226011China
- Cardiac Regeneration and Ageing LabInstitute of Cardiovascular SciencesShanghai Engineering Research Center of Organ RepairSchool of Life ScienceShanghai UniversityShanghai200444China
| | - Jianhua Yao
- Department of CardiologyShanghai Tenth People's HospitalTongji University School of MedicineShanghai200072China
- Department of CardiologyShigatse People's HospitalTibet857000China
| | - Petra H. van der Kraak
- Department of PathologyUniversity Medical Center UtrechtUniversity UtrechtUtrecht3584 CXThe Netherlands
| | - Aryan Vink
- Department of PathologyUniversity Medical Center UtrechtUniversity UtrechtUtrecht3584 CXThe Netherlands
| | - Zhiyong Lei
- Department of CardiologyLaboratory of Experimental CardiologyUniversity Medical Center UtrechtUniversity UtrechtUtrecht3584 CXThe Netherlands
- Division LabCentral Diagnosis Laboratory ResearchUniversity Medical Center UtrechtUniversity UtrechtUtrecht3584 CXThe Netherlands
| | - Yuxiang Dai
- Shanghai Institute of Cardiovascular DiseasesZhongshan HospitalFudan UniversityShanghai200032China
| | - Huihua Chen
- School of Basic Medical ScienceShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Yueyang Liang
- School of Basic Medical ScienceShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Joost PG Sluijter
- Department of CardiologyLaboratory of Experimental CardiologyUniversity Medical Center UtrechtUniversity UtrechtUtrecht3584 CXThe Netherlands
- UMC Utrecht Regenerative Medicine CenterUniversity Medical Center UtrechtUtrecht3508 GAThe Netherlands
| | - Junjie Xiao
- Institute of Geriatrics (Shanghai University)Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong)School of MedicineShanghai UniversityNantong226011China
- Cardiac Regeneration and Ageing LabInstitute of Cardiovascular SciencesShanghai Engineering Research Center of Organ RepairSchool of Life ScienceShanghai UniversityShanghai200444China
| |
Collapse
|
7
|
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
|
8
|
Kozlik-Siwiec P, Buregwa-Czuma S, Zawlik I, Dziedzina S, Myszka A, Zuk-Kuwik J, Siwiec-Kozlik A, Zarychta J, Okon K, Zareba L, Soja J, Jakiela B, Kepski M, Bazan JG, Bazan-Socha S. Co-Expression Analysis of Airway Epithelial Transcriptome in Asthma Patients with Eosinophilic vs. Non-Eosinophilic Airway Infiltration. Int J Mol Sci 2023; 24:3789. [PMID: 36835202 PMCID: PMC9959255 DOI: 10.3390/ijms24043789] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/27/2023] [Accepted: 02/07/2023] [Indexed: 02/16/2023] Open
Abstract
Asthma heterogeneity complicates the search for targeted treatment against airway inflammation and remodeling. We sought to investigate relations between eosinophilic inflammation, a phenotypic feature frequent in severe asthma, bronchial epithelial transcriptome, and functional and structural measures of airway remodeling. We compared epithelial gene expression, spirometry, airway cross-sectional geometry (computed tomography), reticular basement membrane thickness (histology), and blood and bronchoalveolar lavage (BAL) cytokines of n = 40 moderate to severe eosinophilic (EA) and non-eosinophilic asthma (NEA) patients distinguished by BAL eosinophilia. EA patients showed a similar extent of airway remodeling as NEA but had an increased expression of genes involved in the immune response and inflammation (e.g., KIR3DS1), reactive oxygen species generation (GYS2, ATPIF1), cell activation and proliferation (ANK3), cargo transporting (RAB4B, CPLX2), and tissue remodeling (FBLN1, SOX14, GSN), and a lower expression of genes involved in epithelial integrity (e.g., GJB1) and histone acetylation (SIN3A). Genes co-expressed in EA were involved in antiviral responses (e.g., ATP1B1), cell migration (EPS8L1, STOML3), cell adhesion (RAPH1), epithelial-mesenchymal transition (ASB3), and airway hyperreactivity and remodeling (FBN3, RECK), and several were linked to asthma in genome- (e.g., MRPL14, ASB3) or epigenome-wide association studies (CLC, GPI, SSCRB4, STRN4). Signaling pathways inferred from the co-expression pattern were associated with airway remodeling (e.g., TGF-β/Smad2/3, E2F/Rb, and Wnt/β-catenin).
Collapse
Affiliation(s)
- Pawel Kozlik-Siwiec
- Department of Internal Medicine, Jagiellonian University Medical College, 31-066 Krakow, Poland
- Haematology Clinical Department, University Hospital, 31-501 Krakow, Poland
| | - Sylwia Buregwa-Czuma
- College of Natural Sciences, Institute of Computer Science, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland
| | - Izabela Zawlik
- Centre for Innovative Research in Medical and Natural Sciences, Institute of Medical Sciences, Medical College, University of Rzeszow, Kopisto 2a, 35-959 Rzeszow, Poland
| | - Sylwia Dziedzina
- Department of Internal Medicine, Jagiellonian University Medical College, 31-066 Krakow, Poland
| | - Aleksander Myszka
- Institute of Medical Sciences, Medical College, University of Rzeszow, Kopisto 2a, 35-959 Rzeszow, Poland
| | - Joanna Zuk-Kuwik
- Haematology Clinical Department, University Hospital, 31-501 Krakow, Poland
- Haematology Department, Jagiellonian University Medical College, 31-501 Krakow, Poland
| | | | - Jacek Zarychta
- Department of Internal Medicine, Jagiellonian University Medical College, 31-066 Krakow, Poland
- Pulmonary Hospital, 34-736 Zakopane, Poland
| | - Krzysztof Okon
- Department of Pathology, Jagiellonian University Medical College, 33-332 Krakow, Poland
| | - Lech Zareba
- College of Natural Sciences, Institute of Computer Science, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland
| | - Jerzy Soja
- Department of Internal Medicine, Jagiellonian University Medical College, 31-066 Krakow, Poland
| | - Bogdan Jakiela
- Department of Internal Medicine, Jagiellonian University Medical College, 31-066 Krakow, Poland
| | - Michał Kepski
- College of Natural Sciences, Institute of Computer Science, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland
| | - Jan G. Bazan
- College of Natural Sciences, Institute of Computer Science, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland
| | - Stanislawa Bazan-Socha
- Department of Internal Medicine, Jagiellonian University Medical College, 31-066 Krakow, Poland
| |
Collapse
|
9
|
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
|
10
|
Lee SM, Lee JW, Kim I, Woo DC, Pack CG, Sung YH, Baek IJ, Jung CH, Kim YH, Ha CH. Angiogenic adipokine C1q-TNF-related protein 9 ameliorates myocardial infarction via histone deacetylase 7-mediated MEF2 activation. SCIENCE ADVANCES 2022; 8:eabq0898. [PMID: 36459558 PMCID: PMC10936044 DOI: 10.1126/sciadv.abq0898] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 10/18/2022] [Indexed: 06/17/2023]
Abstract
C1q/tumor necrosis factor-related protein 9 (CTRP9) is an adipokine and has high potential as a therapeutic target. However, the role of CTRP9 in cardiovascular disease pathogenesis remains unclear. We found CTRP9 to induce HDAC7 and p38 MAPK phosphorylation via tight regulation of AMPK in vascular endothelial cells, leading to angiogenesis through increased MEF2 activity. The expression of CTRP9 and atheroprotective MEF2 was decreased in plaque tissue of atherosclerotic patients and the ventricle of post-infarction mice. CTRP9 treatment inhibited the formation of atherosclerotic plaques in ApoE KO and CTRP9 KO mice. In addition, CTRP9 induced significant ischemic injury prevention in the post-MI mice. Clinically, serum CTRP9 levels were reduced in patients with MI compared with healthy controls. In summary, CTRP9 induces a vasoprotective response via the AMPK/HDAC7/p38 MAPK pathway in vascular endothelial cells, whereas its absence can contribute to atherosclerosis and MI. Hence, CTRP9 may represent a valuable therapeutic target and biomarker in cardiovascular diseases.
Collapse
Affiliation(s)
- Seung Min Lee
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jin Woo Lee
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Inki Kim
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Convergence Medicine Research Center (CREDIT), Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Dong-Cheol Woo
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Convergence Medicine Research Center (CREDIT), Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Chan-Gi Pack
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Convergence Medicine Research Center (CREDIT), Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Young Hoon Sung
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Convergence Medicine Research Center (CREDIT), Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - In-Jeoung Baek
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Convergence Medicine Research Center (CREDIT), Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Chang Hee Jung
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Young-Hak Kim
- Cardiology Division, Asan Medical Center and University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chang Hoon Ha
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
11
|
Gao Y, Wu D, Chen B, Chen Y, Zhang Q, Zhao P. Rare Variants in Novel Candidate Genes Associated With Nonsyndromic Patent Ductus Arteriosus Identified With Whole-Exome Sequencing. Front Genet 2022; 13:921925. [PMID: 35734438 PMCID: PMC9207465 DOI: 10.3389/fgene.2022.921925] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 05/09/2022] [Indexed: 11/18/2022] Open
Abstract
Background: Patent ductus arteriosus (PDA) is one of the most common congenital heart defects causing pulmonary hypertension, infective endocarditis, and even death. The important role of genetics in determining spontaneous ductal closure has been well-established. However, as many of the identified variants are rare, thorough identification of the associated genetic factors is necessary to further explore the genetic etiology of PDA. Methods: We performed whole-exome sequencing (WES) on 39 isolated nonsyndromic PDA patients and 100 healthy controls. Rare variants and novel genes were identified through bioinformatic filtering strategies. The expression patterns of candidate genes were explored in human embryo heart samples. Results: Eighteen rare damaging variants of six novel PDA-associated genes (SOX8, NES, CDH2, ANK3, EIF4G1, and HIPK1) were newly identified, which were highly expressed in human embryo hearts. Conclusions: WES is an efficient diagnostic tool for exploring the genetic pathogenesis of PDA. These findings contribute new insights into the molecular basis of PDA and may inform further studies on genetic risk factors for congenital heart defects.
Collapse
Affiliation(s)
- Ying Gao
- Department of Pediatric, Shidong Hospital, Shanghai, China
| | - Dan Wu
- Department of Pediatric, Shidong Hospital, Shanghai, China
| | - Bo Chen
- Department of Cardiothoracic Surgery, School of Medicine, Heart Center, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yinghui Chen
- Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qi Zhang
- Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Pengjun Zhao
- Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Pengjun Zhao,
| |
Collapse
|
12
|
Di Segni M, Virdia I, Verdina A, Amoreo CA, Baldari S, Toietta G, Diodoro MG, Mottolese M, Sperduti I, Moretti F, Buglioni S, Soddu S, Di Rocco G. HIPK2 Cooperates with KRAS Signaling and Associates with Colorectal Cancer Progression. Mol Cancer Res 2022; 20:686-698. [PMID: 35082165 DOI: 10.1158/1541-7786.mcr-21-0628] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 11/25/2021] [Accepted: 01/21/2022] [Indexed: 11/16/2022]
Abstract
UNLABELLED Homeodomain-interacting protein kinase 2 (HIPK2) is an evolutionary conserved kinase that has gained attention as a fine tuner of multiple signaling pathways, among which those commonly altered in colorectal cancer. The aim of this study was to evaluate the relationship of HIPK2 expression with progression markers and mutational pattern and gain insights into the contribution of HIPK2 activity in colorectal cancer. We evaluated a retrospective cohort of colorectal cancer samples by IHC for HIPK2 expression and by next-generation sequencing (NGS) for the detection of mutations of cancer associated genes. We show that the percentage of HIPK2-positive cells increases with tumor progression, significantly correlates with tumor-node-metastasis (TNM) staging and associates with a worse outcome. In addition, we observed that high HIPK2 expression significantly associates with KRAS mutations but not with other cancer-related genes. Functional characterization of the link between HIPK2 and KRAS show that activation of the RAS pathway either due to KRAS mutation or via upstream receptor stimulation, increases HIPK2 expression at the protein level. Of note, HIPK2 physically participates in the active RAS complex while HIPK2 depletion impairs ERK phosphorylation and the growth of tumors derived from KRAS mutated colorectal cancer cells. Overall, this study identifies HIPK2 as a prognostic biomarker candidate in patients with colorectal cancer and underscores a previously unknown functional link between HIPK2 and the KRAS signaling pathway. IMPLICATIONS Our data indicate HIPK2 as a new player in the complex picture of the KRAS signaling network, providing rationales for future clinical studies and new treatment strategies for KRAS mutated colorectal cancer.
Collapse
Affiliation(s)
- Micol Di Segni
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS Regina Elena National Cancer Institute, Rome, Italy
- Department of Science, Roma Tre University, Rome, Italy
| | - Ilaria Virdia
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Alessandra Verdina
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Carla Azzurra Amoreo
- Pathology Division, Biostatistics and Bioinformatic Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Silvia Baldari
- Unit of Tumor Immunology and Immunotherapy, Biostatistics and Bioinformatic Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Gabriele Toietta
- Unit of Tumor Immunology and Immunotherapy, Biostatistics and Bioinformatic Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Maria Grazia Diodoro
- Pathology Division, Biostatistics and Bioinformatic Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Marcella Mottolese
- Pathology Division, Biostatistics and Bioinformatic Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Isabella Sperduti
- Clinical Trial Center, Biostatistics and Bioinformatic Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Fabiola Moretti
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (CNR), Monterotondo, Italy
| | - Simonetta Buglioni
- Pathology Division, Biostatistics and Bioinformatic Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Silvia Soddu
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giuliana Di Rocco
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
13
|
Yu Q, Liu L, Zhang X, Chang H, Ma S, Xie Z, Tang S, Ju X, Zhu H, Shen B, Zhang Q. MiR-221-3p targets HIPK2 to promote diabetic wound healing. Microvasc Res 2022; 140:104306. [PMID: 34973299 DOI: 10.1016/j.mvr.2021.104306] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/12/2021] [Accepted: 12/15/2021] [Indexed: 12/15/2022]
Abstract
Diabetic foot ulcer is a severe complication of diabetes and is prone to being a chronic non-healing wound. We previously demonstrated that endothelial progenitor cell-derived exosomes, which contain miR-221-3p, alleviate diabetic ulcers. Here, to explore the mechanisms underlying this wound healing, we investigated the potential angiogenic effects of miR-221-3p in vitro using cultured human umbilical vein endothelial cells (HUVECs) and in vivo using a streptozotocin-induced mouse model of diabetes. We found that miR-221-3p promoted HUVEC viability, migration, and capillary-like tube formation. HUVECs cultured in high glucose showed up-regulated expression of homeodomain-interacting protein kinase 2 (HIPK2), a predicted target of miR-221-3p that may decrease angiogenesis. Knockdown of HIPK2 enhanced high glucose-suppressed HUVEC viability, migration, and tube formation, counteracting the effects of high glucose. Using a dual luciferase reporter assay, we found that HIPK2 was indeed a direct target of miR-221-3p. Subcutaneous injection of miR-221-3p agomir into diabetic mice promoted wound healing and suppressed HIPK2 expression in wound margin tissue. These findings indicate that HIPK2, as a direct target of miR-221-3p, contributes to the regulatory role of miR-221-3p in diabetic wound healing and may be a novel therapeutic target for diabetic foot ulcer.
Collapse
Affiliation(s)
- Qiqi Yu
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui Province 230022, People's Republic of China
| | - Lei Liu
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui Province 230022, People's Republic of China
| | - Xin Zhang
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui Province 230022, People's Republic of China
| | - Hongfeng Chang
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui Province 230022, People's Republic of China
| | - Shaobo Ma
- School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui Province 230032, People's Republic of China
| | - Zhenhui Xie
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui Province 230022, People's Republic of China
| | - Songtao Tang
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui Province 230022, People's Republic of China
| | - Xinmin Ju
- Laboratory of Molecular Biology and Department of Biochemistry, Anhui Medical University, 81 Meishan Road, Hefei, Anhui Province 230032, People's Republic of China
| | - Huaqing Zhu
- Laboratory of Molecular Biology and Department of Biochemistry, Anhui Medical University, 81 Meishan Road, Hefei, Anhui Province 230032, People's Republic of China.
| | - Bing Shen
- School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui Province 230032, People's Republic of China.
| | - Qiu Zhang
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui Province 230022, People's Republic of China.
| |
Collapse
|
14
|
Wang X, Zhang Z, Liang H, Chen R, Huang Y. Circ_0025908 regulates cell vitality and proliferation via miR-137/HIPK2 axis of rheumatic arthritis. J Orthop Surg Res 2021; 16:472. [PMID: 34330307 PMCID: PMC8323297 DOI: 10.1186/s13018-021-02615-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/13/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Rheumatic arthritis (RA) is an autoimmune disease with bad effects. Recent researches have shown that circular RNAs (circRNAs) could affect the progress of RA, but the mechanism still indistinct. In this work, we explored the roles of circ_0025908 in RA. METHODS The levels of circ_0025908, microRNA-137 (miR-137), and mRNA of homeodomain-interacting protein kinase 2 (HIPK2) were detected by quantitative real-time reverse transcription-polymerase chain reaction (qRT-PCR) in RA tissues. Meanwhile, the level of HIPK2 was quantified by Western blot analysis. Besides, the cell functions were examined by CCK8 assay, EdU assay, flow cytometry assay, ELISA, and Western blot. Furthermore, the interplay between miR-137 and circ_0025908 or HIPK2 was detected by dual-luciferase reporter assay. RESULTS The levels of circ_0025908 and HIPK2 were upregulated, and the miR-137 level was decreased in RA tissues in contrast to that in normal tissues. For functional analysis, circ_0025908 deficiency inhibited cell vitality, cell mitotic cycle, cell proliferation, and immunoreaction in RA cells, whereas promoted cell apoptosis. Moreover, miR-137 was confirmed to repress the progression of RA cells by suppressing HIPK2. In mechanism, circ_0025908 acted as a miR-137 sponge to regulate the level of HIPK2. CONCLUSION Circ_0025908 facilitates the development of RA through increasing HIPK2 expression by regulating miR-137, which also offered an underlying targeted therapy for RA treatment.
Collapse
Affiliation(s)
- Xiaofeng Wang
- Department of Traumatic Orthopedics, Institute of Orthopedics, Huizhou Central People's Hospital, No. 41, North E'ling Road, Huizhou, 516000, Guangdong Province, China
| | - Zhiwen Zhang
- Department of Traumatic Orthopedics, Institute of Orthopedics, Huizhou Central People's Hospital, No. 41, North E'ling Road, Huizhou, 516000, Guangdong Province, China
| | - Haofeng Liang
- Department of Traumatic Orthopedics, Institute of Orthopedics, Huizhou Central People's Hospital, No. 41, North E'ling Road, Huizhou, 516000, Guangdong Province, China
| | - Ruixiong Chen
- Department of Traumatic Orthopedics, Institute of Orthopedics, Huizhou Central People's Hospital, No. 41, North E'ling Road, Huizhou, 516000, Guangdong Province, China
| | - Yuliang Huang
- Department of Traumatic Orthopedics, Institute of Orthopedics, Huizhou Central People's Hospital, No. 41, North E'ling Road, Huizhou, 516000, Guangdong Province, China.
| |
Collapse
|
15
|
Zhang F, Qi L, Feng Q, Zhang B, Li X, Liu C, Li W, Liu Q, Yang D, Yin Y, Peng C, Wu H, Tang ZH, Zhou X, Xiang Z, Zhang Z, Wang H, Wei B. HIPK2 phosphorylates HDAC3 for NF-κB acetylation to ameliorate colitis-associated colorectal carcinoma and sepsis. Proc Natl Acad Sci U S A 2021; 118:e2021798118. [PMID: 34244427 PMCID: PMC8285910 DOI: 10.1073/pnas.2021798118] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Although inflammation is critical for the clearance of pathogens, uncontrolled inflammation also contributes to the development of multiple diseases such as cancer and sepsis. Since NF-κB-mediated transactivation in the nucleus is pivotal downstream of various stimuli to induce inflammation, searching the nuclear-localized targets specifically regulating NF-κB activation will provide important therapeutic application. Here, we have identified that homeodomain-interacting protein kinase 2 (HIPK2), a nuclear serine/threonine kinase, increases its expression in inflammatory macrophages. Importantly, HIPK2 deficiency or overexpression could enhance or inhibit inflammatory responses in LPS-stimulated macrophages, respectively. HIPK2-deficient mice were more susceptible to LPS-induced endotoxemia and CLP-induced sepsis. Adoptive transfer of Hipk2+/- bone marrow cells (BMs) also aggravated AOM/DSS-induced colorectal cancer. Mechanistically, HIPK2 bound and phosphorylated histone deacetylase 3 (HDAC3) at serine 374 to inhibit its enzymatic activity, thus reducing the deacetylation of p65 at lysine 218 to suppress NF-κB activation. Notably, the HDAC3 inhibitors protected wild-type or Hipk2-/- BMs-reconstituted mice from LPS-induced endotoxemia. Our findings suggest that the HIPK2-HDAC3-p65 module in macrophages restrains excessive inflammation, which may represent a new layer of therapeutic mechanism for colitis-associated colorectal cancer and sepsis.
Collapse
Affiliation(s)
- Fang Zhang
- Institute of Geriatrics, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Shanghai 200444, China
- School of Life Sciences, Shanghai University, Shanghai 200444, China
- School of Communication & Information Engineering, Shanghai University, Shanghai 200444, China
| | - Linlin Qi
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Qiuyun Feng
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Baokai Zhang
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Xiangyue Li
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Chang Liu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of the Chinese Academy of Sciences, Shanghai 200031, China
| | - Weiyun Li
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of the Chinese Academy of Sciences, Shanghai 200031, China
| | - Qiaojie Liu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Dan Yang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Yue Yin
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai 201210, China
- Shanghai Science Research Center, Chinese Academy of Sciences, Shanghai 201204, China
| | - Chao Peng
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai 201210, China
- Shanghai Science Research Center, Chinese Academy of Sciences, Shanghai 201204, China
| | - Han Wu
- Division of Trauma Surgery, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhao-Hui Tang
- Division of Trauma Surgery, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xi Zhou
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Zou Xiang
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Zhijiang Zhang
- School of Communication & Information Engineering, Shanghai University, Shanghai 200444, China
| | - Hongyan Wang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of the Chinese Academy of Sciences, Shanghai 200031, China;
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Bin Wei
- Institute of Geriatrics, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Shanghai 200444, China;
- School of Life Sciences, Shanghai University, Shanghai 200444, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| |
Collapse
|
16
|
Xiao W, Wang T, Ye Y, Wang X, Chen B, Xing J, Yang H, Zhang X. Identification of HIPK3 as a potential biomarker and an inhibitor of clear cell renal cell carcinoma. Aging (Albany NY) 2021; 13:3536-3553. [PMID: 33495417 PMCID: PMC7906163 DOI: 10.18632/aging.202294] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 10/27/2020] [Indexed: 12/09/2022]
Abstract
Invasion and metastasis are the main causes of poor prognosis in patients with clear cell renal cell carcinoma (ccRCC). The homeodomain interacting protein kinases (HIPKs) can regulate cell proliferation and apoptosis. Little is known about the prognostic role of HIPKs in ccRCC. Here we use Kaplan-Meier survival analysis and multivariate analysis to analyze the correlation of overall survival (OS) and disease–free survival (DFS). ROC curves analyzed the relationship between clinicopathological parameters and HIPK3 expression in ccRCC. Univariate analysis and multivariate analysis confirmed that the expression of HIPK3 was associated with OS (HR, 0.701; P=0.041) and DFS (HR, 0.630; P=0.012). Low HIPK3 expression was a poor prognostic factor and HIPK3 expression was significantly down-regulated in ccRCC cancer tissues when compared with normal renal tissues. In vitro cell results also confirmed that HIPK3 over-expression could inhibit tumor growth and malignant characteristics. The results indicate that low expression of HIPK3 in ccRCC tissues is significantly associated with poor survival rates in tumor patients, and HIPK3 may be used as a valuable biomarker and inhibitor of ccRCC.
Collapse
Affiliation(s)
- Wen Xiao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Wang
- Department of Urology, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yuzhong Ye
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuegang Wang
- Department of Urology, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Bin Chen
- Department of Urology, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Jinchun Xing
- Department of Urology, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Hongmei Yang
- Department of Pathogenic Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
17
|
Abstract
Complex multicellular life in mammals relies on functional cooperation of different organs for the survival of the whole organism. The kidneys play a critical part in this process through the maintenance of fluid volume and composition homeostasis, which enables other organs to fulfil their tasks. The renal endothelium exhibits phenotypic and molecular traits that distinguish it from endothelia of other organs. Moreover, the adult kidney vasculature comprises diverse populations of mostly quiescent, but not metabolically inactive, endothelial cells (ECs) that reside within the kidney glomeruli, cortex and medulla. Each of these populations supports specific functions, for example, in the filtration of blood plasma, the reabsorption and secretion of water and solutes, and the concentration of urine. Transcriptional profiling of these diverse EC populations suggests they have adapted to local microenvironmental conditions (hypoxia, shear stress, hyperosmolarity), enabling them to support kidney functions. Exposure of ECs to microenvironment-derived angiogenic factors affects their metabolism, and sustains kidney development and homeostasis, whereas EC-derived angiocrine factors preserve distinct microenvironment niches. In the context of kidney disease, renal ECs show alteration in their metabolism and phenotype in response to pathological changes in the local microenvironment, further promoting kidney dysfunction. Understanding the diversity and specialization of kidney ECs could provide new avenues for the treatment of kidney diseases and kidney regeneration.
Collapse
|
18
|
Wu X, Zhang Y, Chen L, Han Y, Song Y, Cheng H. BTX-A Promotes Expression of Angiogenesis-Associated Genes in Human Umbilical Vein Endothelial Cells. DNA Cell Biol 2020; 39:2154-2165. [PMID: 33181024 DOI: 10.1089/dna.2020.6004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Raynaud's phenomenon (RP) is an episodic vasospasm of the peripheral arteries caused by an exaggerated reaction to cold temperature or emotional stress. Restoring the angiogenesis capability of the acral lesional skin is a critical strategy to treat RP. Local injection of botulinum toxin-A (BTX-A) has also been reported for treatment of RP. However, since the exact mechanisms of BTX-A action are still unclear, its administration for treatment of RP is not widely used. In the present study, BTX-A was found to promote angiogenesis and relieve RP in the patient. To elucidate its mechanisms against angiogenesis, BTX-A was used to treat human umbilical vein endothelial cells (HUVECs), one of the most popular in vitro models of angiogenesis, and RNA sequencing was used to investigate differentially expressed genes. A total of 413 genes were upregulated, and 1634 were downregulated, with fold-changes >2.0 in HUVECs treated with BTX-A. Gene ontology annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis showed BTX-A affected expression of angiogenesis-associated, angiogenesis-associated signaling pathway-related, metabolic pathway, and epigenetic regulation-related genes. These results demonstrate potential biomarkers of BTX-A action, thereby providing potential therapeutic mechanism(s) by which BTX-A relieves RP symptoms.
Collapse
Affiliation(s)
- Xia Wu
- Department of Dermatology and Venereology, Zhejiang University School of Medicine Sir Run Run Shaw Hospital, Hangzhou, Zhejiang, P.R. China
| | - Yu Zhang
- Department of Dermatology and Venereology, Zhejiang University School of Medicine Sir Run Run Shaw Hospital, Hangzhou, Zhejiang, P.R. China
| | - Luxia Chen
- Department of Dermatology and Venereology, Zhejiang University School of Medicine Sir Run Run Shaw Hospital, Hangzhou, Zhejiang, P.R. China
| | - Yongmei Han
- Department of Dermatology and Venereology, Zhejiang University School of Medicine Sir Run Run Shaw Hospital, Hangzhou, Zhejiang, P.R. China
| | - Yinjing Song
- Department of Dermatology and Venereology, Zhejiang University School of Medicine Sir Run Run Shaw Hospital, Hangzhou, Zhejiang, P.R. China
| | - Hao Cheng
- Department of Dermatology and Venereology, Zhejiang University School of Medicine Sir Run Run Shaw Hospital, Hangzhou, Zhejiang, P.R. China
| |
Collapse
|
19
|
Haas J, Bloesel D, Bacher S, Kracht M, Schmitz ML. Chromatin Targeting of HIPK2 Leads to Acetylation-Dependent Chromatin Decondensation. Front Cell Dev Biol 2020; 8:852. [PMID: 32984337 PMCID: PMC7490299 DOI: 10.3389/fcell.2020.00852] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 08/10/2020] [Indexed: 11/13/2022] Open
Abstract
The protein kinase homeodomain-interacting protein kinase 2 (HIPK2) plays an important role in development and in the response to external cues. The kinase associates with an exceptionally large number of different transcription factors and chromatin regulatory proteins to direct distinct gene expression programs. In order to investigate the function of HIPK2 for chromatin compaction, HIPK2 was fused to the DNA-binding domains of Gal4 or LacI, thus allowing its specific targeting to binding sites for these transcription factors that were integrated in specific chromosome loci. Tethering of HIPK2 resulted in strong decompaction of euchromatic and heterochromatic areas. HIPK2-mediated heterochromatin decondensation started already 4 h after its chromatin association and required the functionality of its SUMO-interacting motif. This process was paralleled by disappearance of the repressive H3K27me3 chromatin mark, recruitment of the acetyltransferases CBP and p300 and increased histone acetylation at H3K18 and H4K5. HIPK2-mediated chromatin decompaction was strongly inhibited in the presence of a CBP/p300 inhibitor and completely blocked by the BET inhibitor JQ1, consistent with a causative role of acetylations for this process. Chromatin tethering of HIPK2 had only a minor effect on basal transcription, while it strongly boosted estrogen-triggered gene expression by acting as a transcriptional cofactor.
Collapse
Affiliation(s)
- Jana Haas
- Institute of Biochemistry, Justus-Liebig-University, Giessen, Germany.,Member of the German Center for Lung Research, Giessen, Germany
| | - Daniel Bloesel
- Institute of Biochemistry, Justus-Liebig-University, Giessen, Germany.,Member of the German Center for Lung Research, Giessen, Germany
| | - Susanne Bacher
- Institute of Biochemistry, Justus-Liebig-University, Giessen, Germany.,Member of the German Center for Lung Research, Giessen, Germany
| | - Michael Kracht
- Member of the German Center for Lung Research, Giessen, Germany.,Rudolf-Buchheim-Institute of Pharmacology, Justus-Liebig-University, Giessen, Germany
| | - M Lienhard Schmitz
- Institute of Biochemistry, Justus-Liebig-University, Giessen, Germany.,Member of the German Center for Lung Research, Giessen, Germany
| |
Collapse
|
20
|
Su C, Johnson ME, Torres A, Thomas RM, Manduchi E, Sharma P, Mehra P, Le Coz C, Leonard ME, Lu S, Hodge KM, Chesi A, Pippin J, Romberg N, Grant SFA, Wells AD. Mapping effector genes at lupus GWAS loci using promoter Capture-C in follicular helper T cells. Nat Commun 2020; 11:3294. [PMID: 32620744 PMCID: PMC7335045 DOI: 10.1038/s41467-020-17089-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 06/02/2020] [Indexed: 01/14/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is mediated by autoreactive antibodies that damage multiple tissues. Genome-wide association studies (GWAS) link >60 loci with SLE risk, but the causal variants and effector genes are largely unknown. We generated high-resolution spatial maps of SLE variant accessibility and gene connectivity in human follicular helper T cells (TFH), a cell type required for anti-nuclear antibodies characteristic of SLE. Of the ~400 potential regulatory variants identified, 90% exhibit spatial proximity to genes distant in the 1D genome sequence, including variants that loop to regulate the canonical TFH genes BCL6 and CXCR5 as confirmed by genome editing. SLE 'variant-to-gene' maps also implicate genes with no known role in TFH/SLE disease biology, including the kinases HIPK1 and MINK1. Targeting these kinases in TFH inhibits production of IL-21, a cytokine crucial for class-switched B cell antibodies. These studies offer mechanistic insight into the SLE-associated regulatory architecture of the human genome.
Collapse
Affiliation(s)
- Chun Su
- Division of Human Genetics, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
| | - Matthew E Johnson
- Division of Human Genetics, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
| | - Annabel Torres
- Department of Pathology, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
| | - Rajan M Thomas
- Department of Pathology, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
| | - Elisabetta Manduchi
- Division of Human Genetics, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
- Institute for Biomedical Informatics, University of Pennsylvania, 3700 Hamilton Walk, Philadelphia, PA, USA
| | - Prabhat Sharma
- Department of Pathology, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
| | - Parul Mehra
- Department of Pathology, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
| | - Carole Le Coz
- Division of Allergy and Immunology, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
| | - Michelle E Leonard
- Division of Human Genetics, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
| | - Sumei Lu
- Division of Human Genetics, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
| | - Kenyaita M Hodge
- Division of Human Genetics, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
| | - Alessandra Chesi
- Division of Human Genetics, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
| | - James Pippin
- Division of Human Genetics, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
| | - Neil Romberg
- Division of Allergy and Immunology, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, 3615 Civic Center Boulevard, Philadelphia, PA, USA
| | - Struan F A Grant
- Division of Human Genetics, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, 3615 Civic Center Boulevard, Philadelphia, PA, USA
- Division of Diabetes and Endocrinology, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, 3615 Civic Center Boulevard, Philadelphia, PA, USA
| | - Andrew D Wells
- Department of Pathology, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, 3615 Civic Center Boulevard, Philadelphia, PA, USA.
| |
Collapse
|
21
|
Gatti V, Ferrara M, Virdia I, Matteoni S, Monteonofrio L, di Martino S, Diodoro MG, Di Rocco G, Rinaldo C, Soddu S. An Alternative Splice Variant of HIPK2 with Intron Retention Contributes to Cytokinesis. Cells 2020; 9:484. [PMID: 32093146 PMCID: PMC7072727 DOI: 10.3390/cells9020484] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/10/2020] [Accepted: 02/17/2020] [Indexed: 12/27/2022] Open
Abstract
HIPK2 is a DYRK-like kinase involved in cellular stress response pathways, development, and cell division. Two alternative splice variants of HIPK2, HIPK2-FL and HIPK2-Δe8, have been previously identified as having different protein stability but similar functional activity in the stress response. Here, we describe one additional HIPK2 splice variant with a distinct subcellular distribution and functional activity in cytokinesis. This novel splice variant lacks the last two exons and retains intron13 with a stop codon after 89 bp of the intron, generating a short isoform, HIPK2-S, that is detectable by 2D Western blots. RT-PCR analyses of tissue arrays and tumor samples show that HIPK2-FL and HIPK2-S are expressed in normal human tissues in a tissue-dependent manner and differentially expressed in human colorectal and pancreatic cancers. Gain- and loss-of-function experiments showed that in contrast to HIPK2-FL, HIPK2-S has a diffuse, non-speckled distribution and is not involved in the DNA damage response. Rather, we found that HIPK2-S, but not HIPK2-FL, localizes at the intercellular bridge, where it phosphorylates histone H2B and spastin, both required for faithful cell division. Altogether, these data show that distinct human HIPK2 splice variants are involved in distinct HIPK2-regulated functions like stress response and cytokinesis.
Collapse
Affiliation(s)
- Veronica Gatti
- Unit of Cellular Networks and Molecular Therapeutic Targets; IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (V.G.); (I.V.); (S.M.); (L.M.); (G.D.R.)
| | - Manuela Ferrara
- Institute of Molecular Biology and Pathology (IBPM), National Research Council (CNR), c/o Sapienza University, 00185 Rome, Italy;
| | - Ilaria Virdia
- Unit of Cellular Networks and Molecular Therapeutic Targets; IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (V.G.); (I.V.); (S.M.); (L.M.); (G.D.R.)
- Department of Sciences, University Roma Tre, 00154 Rome, Italy
| | - Silvia Matteoni
- Unit of Cellular Networks and Molecular Therapeutic Targets; IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (V.G.); (I.V.); (S.M.); (L.M.); (G.D.R.)
| | - Laura Monteonofrio
- Unit of Cellular Networks and Molecular Therapeutic Targets; IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (V.G.); (I.V.); (S.M.); (L.M.); (G.D.R.)
| | - Simona di Martino
- Pathology Unit, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (S.d.M.); (M.G.D.)
| | - Maria Grazia Diodoro
- Pathology Unit, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (S.d.M.); (M.G.D.)
| | - Giuliana Di Rocco
- Unit of Cellular Networks and Molecular Therapeutic Targets; IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (V.G.); (I.V.); (S.M.); (L.M.); (G.D.R.)
| | - Cinzia Rinaldo
- Unit of Cellular Networks and Molecular Therapeutic Targets; IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (V.G.); (I.V.); (S.M.); (L.M.); (G.D.R.)
- Institute of Molecular Biology and Pathology (IBPM), National Research Council (CNR), c/o Sapienza University, 00185 Rome, Italy;
| | - Silvia Soddu
- Unit of Cellular Networks and Molecular Therapeutic Targets; IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (V.G.); (I.V.); (S.M.); (L.M.); (G.D.R.)
| |
Collapse
|
22
|
The nutrient sensor OGT regulates Hipk stability and tumorigenic-like activities in Drosophila. Proc Natl Acad Sci U S A 2020; 117:2004-2013. [PMID: 31932432 PMCID: PMC6994980 DOI: 10.1073/pnas.1912894117] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Environmental cues such as nutrients alter cellular behaviors by acting on a wide array of molecular sensors inside cells. Of emerging interest is the link observed between effects of dietary sugars on cancer proliferation. Here, we identify the requirements of hexosamine biosynthetic pathway (HBP) and O-GlcNAc transferase (OGT) for Drosophila homeodomain-interacting protein kinase (Hipk)-induced growth abnormalities in response to a high sugar diet. On a normal diet, OGT is both necessary and sufficient for inducing Hipk-mediated tumor-like growth. We further show that OGT maintains Hipk protein stability by blocking its proteasomal degradation and that Hipk is O-GlcNAcylated by OGT. In mammalian cells, human HIPK2 proteins accumulate posttranscriptionally upon OGT overexpression. Mass spectrometry analyses reveal that HIPK2 is at least O-GlcNAc modified at S852, T1009, and S1147 residues. Mutations of these residues reduce HIPK2 O-GlcNAcylation and stability. Together, our data demonstrate a conserved role of OGT in positively regulating the protein stability of HIPKs (fly Hipk and human HIPK2), which likely permits the nutritional responsiveness of HIPKs.
Collapse
|
23
|
Bochenek ML, Leidinger C, Rosinus NS, Gogiraju R, Guth S, Hobohm L, Jurk K, Mayer E, Münzel T, Lankeit M, Bosmann M, Konstantinides S, Schäfer K. Activated Endothelial TGFβ1 Signaling Promotes Venous Thrombus Nonresolution in Mice Via Endothelin-1: Potential Role for Chronic Thromboembolic Pulmonary Hypertension. Circ Res 2019; 126:162-181. [PMID: 31747868 DOI: 10.1161/circresaha.119.315259] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
RATIONALE Chronic thromboembolic pulmonary hypertension (CTEPH) is characterized by defective thrombus resolution, pulmonary artery obstruction, and vasculopathy. TGFβ (transforming growth factor-β) signaling mutations have been implicated in pulmonary arterial hypertension, whereas the role of TGFβ in the pathophysiology of CTEPH is unknown. OBJECTIVE To determine whether defective TGFβ signaling in endothelial cells contributes to thrombus nonresolution and fibrosis. METHODS AND RESULTS Venous thrombosis was induced by inferior vena cava ligation in mice with genetic deletion of TGFβ1 in platelets (Plt.TGFβ-KO) or TGFβ type II receptors in endothelial cells (End.TGFβRII-KO). Pulmonary endarterectomy specimens from CTEPH patients were analyzed using immunohistochemistry. Primary human and mouse endothelial cells were studied using confocal microscopy, quantitative polymerase chain reaction, and Western blot. Absence of TGFβ1 in platelets did not alter platelet number or function but was associated with faster venous thrombus resolution, whereas endothelial TGFβRII deletion resulted in larger, more fibrotic and higher vascularized venous thrombi. Increased circulating active TGFβ1 levels, endothelial TGFβRI/ALK1 (activin receptor-like kinase), and TGFβRI/ALK5 expression were detected in End.TGFβRII-KO mice, and activated TGFβ signaling was present in vessel-rich areas of CTEPH specimens. CTEPH-endothelial cells and murine endothelial cells lacking TGFβRII simultaneously expressed endothelial and mesenchymal markers and transcription factors regulating endothelial-to-mesenchymal transition, similar to TGFβ1-stimulated endothelial cells. Mechanistically, increased endothelin-1 levels were detected in TGFβRII-KO endothelial cells, murine venous thrombi, or endarterectomy specimens and plasma of CTEPH patients, and endothelin-1 overexpression was prevented by inhibition of ALK5, and to a lesser extent of ALK1. ALK5 inhibition and endothelin receptor antagonization inhibited mesenchymal lineage conversion in TGFβ1-exposed human and murine endothelial cells and improved venous thrombus resolution and pulmonary vaso-occlusions in End.TGFβRII-KO mice. CONCLUSIONS Endothelial TGFβ1 signaling via type I receptors and endothelin-1 contribute to mesenchymal lineage transition and thrombofibrosis, which were prevented by blocking endothelin receptors. Our findings may have relevant implications for the prevention and management of CTEPH.
Collapse
Affiliation(s)
- Magdalena L Bochenek
- From the Center for Cardiology, Cardiology I (M.L.B., C.L., N.S.R., R.G., L.H., T.M., K.S.), University Medical Center Mainz, Germany.,Center for Thrombosis and Hemostasis (M.L.B., L.H., K.J., M.L., M.B., S.K.), University Medical Center Mainz, Germany.,German Center for Cardiovascular Research (DZHK e.V.; RheinMain) (M.L.B., N.S.R., R.G., E.M., T.M., K.S.)
| | - Christiane Leidinger
- From the Center for Cardiology, Cardiology I (M.L.B., C.L., N.S.R., R.G., L.H., T.M., K.S.), University Medical Center Mainz, Germany
| | - Nico S Rosinus
- From the Center for Cardiology, Cardiology I (M.L.B., C.L., N.S.R., R.G., L.H., T.M., K.S.), University Medical Center Mainz, Germany.,German Center for Cardiovascular Research (DZHK e.V.; RheinMain) (M.L.B., N.S.R., R.G., E.M., T.M., K.S.)
| | - Rajinikanth Gogiraju
- From the Center for Cardiology, Cardiology I (M.L.B., C.L., N.S.R., R.G., L.H., T.M., K.S.), University Medical Center Mainz, Germany.,German Center for Cardiovascular Research (DZHK e.V.; RheinMain) (M.L.B., N.S.R., R.G., E.M., T.M., K.S.)
| | - Stefan Guth
- Thoracic Surgery, Kerckhoff Clinic, Bad Nauheim, Germany (S.G., E.M.)
| | - Lukas Hobohm
- From the Center for Cardiology, Cardiology I (M.L.B., C.L., N.S.R., R.G., L.H., T.M., K.S.), University Medical Center Mainz, Germany.,Center for Thrombosis and Hemostasis (M.L.B., L.H., K.J., M.L., M.B., S.K.), University Medical Center Mainz, Germany
| | - Kerstin Jurk
- Center for Thrombosis and Hemostasis (M.L.B., L.H., K.J., M.L., M.B., S.K.), University Medical Center Mainz, Germany
| | - Eckhard Mayer
- Thoracic Surgery, Kerckhoff Clinic, Bad Nauheim, Germany (S.G., E.M.).,German Center for Cardiovascular Research (DZHK e.V.; RheinMain) (M.L.B., N.S.R., R.G., E.M., T.M., K.S.)
| | - Thomas Münzel
- From the Center for Cardiology, Cardiology I (M.L.B., C.L., N.S.R., R.G., L.H., T.M., K.S.), University Medical Center Mainz, Germany.,German Center for Cardiovascular Research (DZHK e.V.; RheinMain) (M.L.B., N.S.R., R.G., E.M., T.M., K.S.)
| | - Mareike Lankeit
- Center for Thrombosis and Hemostasis (M.L.B., L.H., K.J., M.L., M.B., S.K.), University Medical Center Mainz, Germany.,Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Charité -University Medicine, Berlin, Germany (M.L.)
| | - Markus Bosmann
- Center for Thrombosis and Hemostasis (M.L.B., L.H., K.J., M.L., M.B., S.K.), University Medical Center Mainz, Germany.,Department of Medicine, Boston University School of Medicine, MA (M.B.)
| | - Stavros Konstantinides
- Center for Thrombosis and Hemostasis (M.L.B., L.H., K.J., M.L., M.B., S.K.), University Medical Center Mainz, Germany.,Department of Cardiology, Democritus University of Thrace, Alexandroupolis, Greece (S.K.)
| | - Katrin Schäfer
- From the Center for Cardiology, Cardiology I (M.L.B., C.L., N.S.R., R.G., L.H., T.M., K.S.), University Medical Center Mainz, Germany.,German Center for Cardiovascular Research (DZHK e.V.; RheinMain) (M.L.B., N.S.R., R.G., E.M., T.M., K.S.)
| |
Collapse
|
24
|
Guo Y, Sui JY, Kim K, Zhang Z, Qu XA, Nam YJ, Willette RN, Barnett JV, Knollmann BC, Force T, Lal H. Cardiomyocyte Homeodomain-Interacting Protein Kinase 2 Maintains Basal Cardiac Function via Extracellular Signal-Regulated Kinase Signaling. Circulation 2019; 140:1820-1833. [PMID: 31581792 DOI: 10.1161/circulationaha.119.040740] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Cardiac kinases play a critical role in the development of heart failure, and represent potential tractable therapeutic targets. However, only a very small fraction of the cardiac kinome has been investigated. To identify novel cardiac kinases involved in heart failure, we used an integrated transcriptomics and bioinformatics analysis and identified Homeodomain-Interacting Protein Kinase 2 (HIPK2) as a novel candidate kinase. The role of HIPK2 in cardiac biology is unknown. METHODS We used the Expression2Kinase algorithm for the screening of kinase targets. To determine the role of HIPK2 in the heart, we generated cardiomyocyte (CM)-specific HIPK2 knockout and heterozygous mice. Heart function was examined by echocardiography, and related cellular and molecular mechanisms were examined. Adeno-associated virus serotype 9 carrying cardiac-specific constitutively active MEK1 (TnT-MEK1-CA) was administrated to rescue cardiac dysfunction in CM-HIPK2 knockout mice. RESULTS To our knowledge, this is the first study to define the role of HIPK2 in cardiac biology. Using multiple HIPK2 loss-of-function mouse models, we demonstrated that reduction of HIPK2 in CMs leads to cardiac dysfunction, suggesting a causal role in heart failure. It is important to note that cardiac dysfunction in HIPK2 knockout mice developed with advancing age, but not during development. In addition, CM-HIPK2 knockout mice and CM-HIPK2 heterozygous mice exhibited a gene dose-response relationship of CM-HIPK2 on heart function. HIPK2 expression in the heart was significantly reduced in human end-stage ischemic cardiomyopathy in comparison to nonfailing myocardium, suggesting a clinical relevance of HIPK2 in cardiac biology. In vitro studies with neonatal rat ventricular CMscorroborated the in vivo findings. Specifically, adenovirus-mediated overexpression of HIPK2 suppressed the expression of heart failure markers, NPPA and NPPB, at basal condition and abolished phenylephrine-induced pathological gene expression. An array of mechanistic studies revealed impaired extracellular signal-regulated kinase 1/2 signaling in HIPK2-deficient hearts. An in vivo rescue experiment with adeno-associated virus serotype 9 TnT-MEK1-CA nearly abolished the detrimental phenotype of knockout mice, suggesting that impaired extracellular signal-regulated kinase signaling mediated apoptosis as the key factor driving the detrimental phenotype in CM-HIPK2 knockout mice hearts. CONCLUSIONS Taken together, these findings suggest that CM-HIPK2 is required to maintain normal cardiac function via extracellular signal-regulated kinase signaling.
Collapse
Affiliation(s)
- Yuanjun Guo
- Division of Cardiovascular Medicine (Y.G., J.Y.S., Z.Z., Y.-J.N., T.F., H.L.), Vanderbilt University Medical Center, Nashville, TN.,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN (Y. Guo, J.V.B.)
| | - Jennifer Y Sui
- Division of Cardiovascular Medicine (Y.G., J.Y.S., Z.Z., Y.-J.N., T.F., H.L.), Vanderbilt University Medical Center, Nashville, TN
| | - Kyungsoo Kim
- Division of Clinical Pharmacology (K.K., B.C.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Zhentao Zhang
- Division of Cardiovascular Medicine (Y.G., J.Y.S., Z.Z., Y.-J.N., T.F., H.L.), Vanderbilt University Medical Center, Nashville, TN.,Department of Cell and Developmental Biology (Z.Z., Y.-J.N.), Vanderbilt University, Nashville, TN.,Vanderbilt Center for Stem Cell Biology (Z.Z., Y.-J.N.), Vanderbilt University, Nashville, TN
| | - Xiaoyan A Qu
- PAREXEL International, Research Triangle Park, Durham, NC (X.A.Q.)
| | - Young-Jae Nam
- Division of Cardiovascular Medicine (Y.G., J.Y.S., Z.Z., Y.-J.N., T.F., H.L.), Vanderbilt University Medical Center, Nashville, TN.,Department of Cell and Developmental Biology (Z.Z., Y.-J.N.), Vanderbilt University, Nashville, TN.,Vanderbilt Center for Stem Cell Biology (Z.Z., Y.-J.N.), Vanderbilt University, Nashville, TN
| | - Robert N Willette
- Heart Failure Discovery Performance Unit, Metabolic Pathways and Cardiovascular Therapeutic Area GlaxoSmithKline, King of Prussia, PA (R.N.W.)
| | - Joey V Barnett
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN (Y. Guo, J.V.B.)
| | - Bjorn C Knollmann
- Division of Clinical Pharmacology (K.K., B.C.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Thomas Force
- Division of Cardiovascular Medicine (Y.G., J.Y.S., Z.Z., Y.-J.N., T.F., H.L.), Vanderbilt University Medical Center, Nashville, TN
| | - Hind Lal
- Division of Cardiovascular Medicine (Y.G., J.Y.S., Z.Z., Y.-J.N., T.F., H.L.), Vanderbilt University Medical Center, Nashville, TN.,Division of Cardiovascular Disease, University of Alabama at Birmingham, AL (H.L.)
| |
Collapse
|
25
|
Agnew C, Liu L, Liu S, Xu W, You L, Yeung W, Kannan N, Jablons D, Jura N. The crystal structure of the protein kinase HIPK2 reveals a unique architecture of its CMGC-insert region. J Biol Chem 2019; 294:13545-13559. [PMID: 31341017 PMCID: PMC6746438 DOI: 10.1074/jbc.ra119.009725] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/11/2019] [Indexed: 01/07/2023] Open
Abstract
The homeodomain-interacting protein kinase (HIPK) family is comprised of four nuclear protein kinases, HIPK1-4. HIPK proteins phosphorylate a diverse range of transcription factors involved in cell proliferation, differentiation, and apoptosis. HIPK2, thus far the best-characterized member of this largely understudied family of protein kinases, plays a role in the activation of p53 in response to DNA damage. Despite this tumor-suppressor function, HIPK2 is also found overexpressed in several cancers, and its hyperactivation causes chronic fibrosis. There are currently no structures of HIPK2 or of any other HIPK kinase. Here, we report the crystal structure of HIPK2's kinase domain bound to CX-4945, a casein kinase 2α (CK2α) inhibitor currently in clinical trials against several cancers. The structure, determined at 2.2 Å resolution, revealed that CX-4945 engages the HIPK2 active site in a hybrid binding mode between that seen in structures of CK2α and Pim1 kinases. The HIPK2 kinase domain crystallized in the active conformation, which was stabilized by phosphorylation of the activation loop. We noted that the overall kinase domain fold of HIPK2 closely resembles that of evolutionarily related dual-specificity tyrosine-regulated kinases (DYRKs). Most significant structural differences between HIPK2 and DYRKs included an absence of the regulatory N-terminal domain and a unique conformation of the CMGC-insert region and of a newly defined insert segment in the αC-β4 loop. This first crystal structure of HIPK2 paves the way for characterizing the understudied members of the HIPK family and for developing HIPK2-directed therapies for managing cancer and fibrosis.
Collapse
Affiliation(s)
- Christopher Agnew
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94158
| | - Lijun Liu
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94158
| | - Shu Liu
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94115
| | - Wei Xu
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94115
| | - Liang You
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94115
| | - Wayland Yeung
- Institute of Bioinformatics and Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia 30602
| | - Natarajan Kannan
- Institute of Bioinformatics and Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia 30602
| | - David Jablons
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94115, Supported by the Kazan McClain Partners' Foundation and the H. N. and Frances C. Berger Foundation. To whom correspondence may be addressed:
1600 Divisadero St., A745, San Francisco, CA 94115. Tel.:
415-353-7502; E-mail:
| | - Natalia Jura
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94158,Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California 94158, To whom correspondence may be addressed:
555 Mission Bay Blvd. S., Rm. 452W, San Francisco, CA 94158. Tel.:
415-514-1133; E-mail:
| |
Collapse
|
26
|
Ke CY, Mei HH, Wong FH, Lo LJ. IRF6 and TAK1 coordinately promote the activation of HIPK2 to stimulate apoptosis during palate fusion. Sci Signal 2019; 12:12/593/eaav7666. [DOI: 10.1126/scisignal.aav7666] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cleft palate is a common craniofacial defect caused by a failure in palate fusion. The palatal shelves migrate toward one another and meet at the embryonic midline, creating a seam. Transforming growth factor–β3 (TGF-β3)–induced apoptosis of the medial edge epithelium (MEE), the cells located along the seam, is required for completion of palate fusion. The transcription factor interferon regulatory factor 6 (IRF6) promotes TGF-β3–induced MEE cell apoptosis by stimulating the degradation of the transcription factor ΔNp63 and promoting the expression of the gene encoding the cyclin-dependent kinase inhibitor p21. Because homeodomain-interacting protein kinase 2 (HIPK2) functions downstream of IRF6 in human cancer cells and is required for ΔNp63 protein degradation in keratinocytes, we investigated whether HIPK2 played a role in IRF6-induced ΔNp63 degradation in palate fusion. HIPK2 was present in the MEE cells of mouse palatal shelves during seam formation in vivo, and ectopic expression of IRF6 in palatal shelves cultured ex vivo stimulated the expression of Hipk2 and the accumulation of phosphorylated HIPK2. Knockdown and ectopic expression experiments in organ culture demonstrated that p21 was required for HIPK2- and IRF6-dependent activation of caspase 3, MEE apoptosis, and palate fusion. Contact between palatal shelves enhanced the phosphorylation of TGF-β–activated kinase 1 (TAK1), which promoted the phosphorylation of HIPK2 and palate fusion. Our findings demonstrate that HIPK2 promotes seam cell apoptosis and palate fusion downstream of IRF6 and that IRF6 and TAK1 appear to coordinately enhance the abundance and activation of HIPK2 during palate fusion.
Collapse
|
27
|
Mukherjee T, Udupa VAV, Prakhar P, Chandra K, Chakravortty D, Balaji KN. Epidermal Growth Factor Receptor-Responsive Indoleamine 2,3-Dioxygenase Confers Immune Homeostasis During Shigella flexneri Infection. J Infect Dis 2019; 219:1841-1851. [PMID: 30615126 DOI: 10.1093/infdis/jiz009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 01/05/2019] [Indexed: 12/19/2022] Open
Abstract
The resolution of Shigella flexneri infection-associated hyperinflammation is crucial for host survival. Using in vitro and in vivo models of shigellosis, we found that S. flexneri induces the expression of indoleamine 2,3-dioxygenase 1 (IDO1) through the nucleotide oligomerization domain 2 (NOD2) and epidermal growth factor receptor (EGFR) signaling pathway. Congruently, abrogation of NOD2 or EGFR compromises the ability of S. flexneri to induce IDO1 expression. We observed that the loss of IDO1 function in vivo exacerbates shigellosis by skewing the inflammatory cytokine response, disrupting colon epithelial barrier integrity and consequently limiting the host life-span. Interestingly, administration of recombinant EGF rescued mice from IDO1 inhibition-driven aggravated shigellosis by restoring the cytokine balance and subsequently restricting bacterial growth. This is the first study that underscores the direct implication of the NOD2-EGFR axis in IDO1 production and its crucial homeostatic contributions during shigellosis. Together, these findings reveal EGF as a potential therapeutic intervention for infectious diseases.
Collapse
Affiliation(s)
- Tanushree Mukherjee
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Vibha A V Udupa
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Praveen Prakhar
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Kasturi Chandra
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Dipshikha Chakravortty
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | | |
Collapse
|
28
|
Ritter O, Schmitz ML. Differential intracellular localization and dynamic nucleocytoplasmic shuttling of homeodomain-interacting protein kinase family members. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:1676-1686. [PMID: 31029697 DOI: 10.1016/j.bbamcr.2019.04.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 12/20/2018] [Accepted: 01/06/2019] [Indexed: 12/14/2022]
Abstract
The three canonical members of the family of homeodomain-interacting protein (HIP) kinases fulfill overlapping and distinct roles in cellular stress response pathways. Here we systematically compared all three endogenous HIPKs for their intracellular distribution and mutual interactions. The endogenous HIPKs are contained in high molecular weight complexes of ~700 kDa but do not directly interact physically. Under basal conditions, HIPK1 was mostly cytoplasmic, while HIPK3 was found in the nucleus and HIPK2 occurred in both compartments. Inhibition of nuclear export by leptomycin B resulted in the nuclear accumulation of mainly HIPK1 and HIPK2, indicating constitutive dynamic nucleocytoplasmic shuttling. The carcinogenic chemical stressor sodium arsenite caused the induction of HIPK2-dependent cell death and also resulted in a rapid and complete nuclear translocation of HIPK2, showing that the intracellular distribution of this kinase can undergo dynamic regulation.
Collapse
Affiliation(s)
- Olesja Ritter
- Institute of Biochemistry, Medical Faculty, Justus-Liebig-University, Member of the German Center for Lung Research, Friedrichstrasse 24, D-35392 Giessen, Germany
| | - M Lienhard Schmitz
- Institute of Biochemistry, Medical Faculty, Justus-Liebig-University, Member of the German Center for Lung Research, Friedrichstrasse 24, D-35392 Giessen, Germany.
| |
Collapse
|
29
|
Hu HY, Yu CH, Zhang HH, Zhang SZ, Yu WY, Yang Y, Chen Q. Exosomal miR-1229 derived from colorectal cancer cells promotes angiogenesis by targeting HIPK2. Int J Biol Macromol 2019; 132:470-477. [PMID: 30936013 DOI: 10.1016/j.ijbiomac.2019.03.221] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 03/07/2019] [Accepted: 03/29/2019] [Indexed: 02/06/2023]
Abstract
Circulating exosomal microRNAs (exomiR) have been demonstrated to be novel diagnostic biomarkers for various cancers. In this study, we found that circulating exomiR-1229 levels were significantly increased in the serum exosomes of patients with colorectal cancer (CRC) and significantly associated with tumor size, lymphatic metastasis, TNM stage and poor survival. Treatment with siRNA-Drosha, siRNA-ALIX and GW4869 repressed the expression of exomiR-1229 secreted from CRC cells. Both CRC-derived exosomes and exomiR-1229 mimic promoted the tubulogenesis of HUVECs, but transfection with exomiR-1229 inhibitor anta-miR-1229 significantly suppressed tube formation. Subsequently, HIPK2 was identified as a target of exomiR-1229 and responsible for the effect of exomiR-1229 on angiogenesis of HUVECs. ExomiR-1229 inhibited the protein expression of HIPK2, thereby activating VEGF pathway. Finally, anta-miR-1229 effectively inhibited tumor growth and angiogenesis in the nude mouse xenograft model. These results highlighted a novel mechanism of CRC angiogenesis and the biological roles of exomiR-1229.
Collapse
Affiliation(s)
- Hui-Ying Hu
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou 310013, China
| | - Chen-Huan Yu
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou 310013, China
| | - Huan-Huan Zhang
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou 310013, China
| | - Song-Zhao Zhang
- Department of Clinical Laboratory Medicine, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Wen-Ying Yu
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou 310013, China
| | - Yang Yang
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou 310013, China
| | - Qin Chen
- Department of Clinical Laboratory Medicine, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China.
| |
Collapse
|
30
|
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
|
31
|
HIPK2-Mediated Transcriptional Control of NMDA Receptor Subunit Expression Regulates Neuronal Survival and Cell Death. J Neurosci 2018; 38:4006-4019. [PMID: 29581378 DOI: 10.1523/jneurosci.3577-17.2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/01/2018] [Accepted: 02/20/2018] [Indexed: 11/21/2022] Open
Abstract
NMDA receptors are critical for neuronal communication. Dysfunction in NMDA receptors has been implicated in neuropsychiatric diseases. While it is well recognized that the composition of NMDA receptors undergoes a GluN2B-to-GluN2A switch in early postnatal life, the mechanism regulating this switch remains unclear. Using transcriptomic and functional analyses in brain tissues from male and female Hipk2+/+ and Hipk2-/- mice, we showed that the HIPK2-JNK-c-Jun pathway is important in suppressing the transcription of Grin2a and Grin2c, which encodes the GluN2A and GluN2C subunits of the NMDA receptors, respectively. Loss of HIPK2 leads to a significant decrease in JNK-c-Jun signaling, which in turn derepresses the transcription of Grin2a and Grin2c mRNA and upregulates GluN2A and GluN2C protein levels. These changes result in a significant increase of GluN2A/GluN2B ratio in synapse and mitochondria, a persistent activation of the ERK-CREB pathway and the upregulation of synaptic activity-regulated genes, which collectively contribute to the resistance of Hipk2-/- neurons to cell death induced by mitochondrial toxins.SIGNIFICANCE STATEMENT We identify HIPK2-JNK-c-Jun signaling as a key mechanism that regulates the transcription of NMDA receptor subunits GluN2A and GluN2C in vivo Our results provide insights into a previously unrecognized molecular mechanism that control the switch of NMDA receptor subunits in early postnatal brain development. Furthermore, we provide evidence that changes in the ratio of NMDA subunits GluN2A/GluN2B can also be detected in the synapse and mitochondria, which contributes to a persistent activation of the prosurvival ERK-CREB pathway and its downstream target genes. Collectively, these changes protect HIPK2 deficient neurons from mitochondrial toxins.
Collapse
|
32
|
Scaglione A, Monteonofrio L, Parisi G, Cecchetti C, Siepi F, Rinaldo C, Giorgi A, Verzili D, Zamparelli C, Savino C, Soddu S, Vallone B, Montemiglio LC. Effects of Y361-auto-phosphorylation on structural plasticity of the HIPK2 kinase domain. Protein Sci 2017; 27:725-737. [PMID: 29277937 DOI: 10.1002/pro.3367] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 11/21/2017] [Accepted: 12/13/2017] [Indexed: 12/30/2022]
Abstract
The dual-specificity activity of the homeodomain interacting protein kinase 2 (HIPK2) is regulated by cis-auto-phosphorylation of tyrosine 361 (Y361) on the activation loop. Inhibition of this process or substitution of Y361 with nonphosphorylatable amino acid residues result in aberrant HIPK2 forms that show altered functionalities, pathological-like cellular relocalization, and accumulation into cytoplasmic aggresomes. Here, we report an in vitro characterization of wild type HIPK2 kinase domain and of two mutants, one at the regulating Y361 (Y361F, mimicking a form of HIPK2 lacking Y361 phosphorylation) and another at the catalytic lysine 228 (K228A, inactivating the enzyme). Gel filtration and thermal denaturation analyzes along with equilibrium binding experiments and kinase assays performed in the presence or absence of ATP-competitors were performed. The effects induced by mutations on overall stability, oligomerization and activity support the existence of different conformations of the kinase domain linked to Y361 phosphorylation. In addition, our in vitro data are consistent with both the cross-talk between the catalytic site and the activation loop of HIPK2 and the aberrant activities and accumulation previously reported for the Y361 nonphosphorylated HIPK2 in mammalian cells.
Collapse
Affiliation(s)
- Antonella Scaglione
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Sapienza Università di Roma, P.le A. Moro 5, 00185, Italy, Rome.,Dipartimento di Scienze Biochimiche, "A. Rossi Fanelli", Sapienza Università di Roma, P.le A. Moro 5, 00185, Italy, Rome
| | - Laura Monteonofrio
- Unit of Cellular Networks and Molecular Therapeutic Targets, Department of Research, Advanced Diagnostic, and Technological Innovation, Regina Elena National Cancer Institute - IRCCS, Via Elio Chianesi, 53, Rome, 00144, Italy
| | - Giacomo Parisi
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Sapienza Università di Roma, P.le A. Moro 5, 00185, Italy, Rome.,Dipartimento di Scienze Biochimiche, "A. Rossi Fanelli", Sapienza Università di Roma, P.le A. Moro 5, 00185, Italy, Rome
| | - Cristina Cecchetti
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Sapienza Università di Roma, P.le A. Moro 5, 00185, Italy, Rome.,Dipartimento di Scienze Biochimiche, "A. Rossi Fanelli", Sapienza Università di Roma, P.le A. Moro 5, 00185, Italy, Rome
| | - Francesca Siepi
- Unit of Cellular Networks and Molecular Therapeutic Targets, Department of Research, Advanced Diagnostic, and Technological Innovation, Regina Elena National Cancer Institute - IRCCS, Via Elio Chianesi, 53, Rome, 00144, Italy
| | - Cinzia Rinaldo
- Unit of Cellular Networks and Molecular Therapeutic Targets, Department of Research, Advanced Diagnostic, and Technological Innovation, Regina Elena National Cancer Institute - IRCCS, Via Elio Chianesi, 53, Rome, 00144, Italy.,CNR Institute of Molecular Biology and Pathology, P.le A. Moro 5, Rome, 00185, Italy
| | - Alessandra Giorgi
- Dipartimento di Scienze Biochimiche, "A. Rossi Fanelli", Sapienza Università di Roma, P.le A. Moro 5, 00185, Italy, Rome
| | - Daniela Verzili
- CNR Institute of Molecular Biology and Pathology, P.le A. Moro 5, Rome, 00185, Italy
| | - Carlotta Zamparelli
- Dipartimento di Scienze Biochimiche, "A. Rossi Fanelli", Sapienza Università di Roma, P.le A. Moro 5, 00185, Italy, Rome
| | - Carmelinda Savino
- CNR Institute of Molecular Biology and Pathology, P.le A. Moro 5, Rome, 00185, Italy
| | - Silvia Soddu
- Unit of Cellular Networks and Molecular Therapeutic Targets, Department of Research, Advanced Diagnostic, and Technological Innovation, Regina Elena National Cancer Institute - IRCCS, Via Elio Chianesi, 53, Rome, 00144, Italy
| | - Beatrice Vallone
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Sapienza Università di Roma, P.le A. Moro 5, 00185, Italy, Rome.,Dipartimento di Scienze Biochimiche, "A. Rossi Fanelli", Sapienza Università di Roma, P.le A. Moro 5, 00185, Italy, Rome
| | - Linda Celeste Montemiglio
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Sapienza Università di Roma, P.le A. Moro 5, 00185, Italy, Rome.,Dipartimento di Scienze Biochimiche, "A. Rossi Fanelli", Sapienza Università di Roma, P.le A. Moro 5, 00185, Italy, Rome
| |
Collapse
|
33
|
Suh JH, Lai L, Nam D, Kim J, Jo J, Taffet GE, Kim E, Kaelber JT, Lee HK, Entman ML, Cooke JP, Reineke EL. Steroid receptor coactivator-2 (SRC-2) coordinates cardiomyocyte paracrine signaling to promote pressure overload-induced angiogenesis. J Biol Chem 2017; 292:21643-21652. [PMID: 29127200 DOI: 10.1074/jbc.m117.804740] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 11/09/2017] [Indexed: 12/23/2022] Open
Abstract
Pressure overload-induced cardiac stress induces left ventricular hypertrophy driven by increased cardiomyocyte mass. The increased energetic demand and cardiomyocyte size during hypertrophy necessitate increased fuel and oxygen delivery and stimulate angiogenesis in the left ventricular wall. We have previously shown that the transcriptional regulator steroid receptor coactivator-2 (SRC-2) controls activation of several key cardiac transcription factors and that SRC-2 loss results in extensive cardiac transcriptional remodeling. Pressure overload in mice lacking SRC-2 induces an abrogated hypertrophic response and decreases sustained cardiac function, but the cardiomyocyte-specific effects of SRC-2 in these changes are unknown. Here, we report that cardiomyocyte-specific loss of SRC-2 (SRC-2 CKO) results in a blunted hypertrophy accompanied by a rapid, progressive decrease in cardiac function. We found that SRC-2 CKO mice exhibit markedly decreased left ventricular vasculature in response to transverse aortic constriction, corresponding to decreased expression of the angiogenic factor VEGF. Of note, SRC-2 knockdown in cardiomyocytes decreased VEGF expression and secretion to levels sufficient to blunt in vitro tube formation and proliferation of endothelial cells. During pressure overload, both hypertrophic and hypoxic signals can stimulate angiogenesis, both of which stimulated SRC-2 expression in vitro Furthermore, SRC-2 coactivated the transcription factors GATA-binding protein 4 (GATA-4) and hypoxia-inducible factor (HIF)-1α and -2α in response to angiotensin II and hypoxia, respectively, which drive VEGF expression. These results suggest that SRC-2 coordinates cardiomyocyte secretion of VEGF downstream of the two major angiogenic stimuli occurring during pressure overload bridging both hypertrophic and hypoxia-stimulated paracrine signaling.
Collapse
Affiliation(s)
- Ji Ho Suh
- From the Center for Bioenergetics and
| | - Li Lai
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas 77030
| | | | - Jong Kim
- the University of Houston 77004, Houston, Texas
| | - Juyeon Jo
- the Department of Pediatrics and Neuroscience, Baylor College of Medicine and Neurological Research Institute at Texas Children's Hospital, Houston, Texas 77030, and
| | - George E Taffet
- the Division of Cardiovascular Sciences, Department of Medicine, and
| | - Eunah Kim
- From the Center for Bioenergetics and
| | - Jason T Kaelber
- the National Center for Macromolecular Imaging and Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas 77030
| | - Hyun-Kyoung Lee
- the Department of Pediatrics and Neuroscience, Baylor College of Medicine and Neurological Research Institute at Texas Children's Hospital, Houston, Texas 77030, and
| | - Mark L Entman
- the Division of Cardiovascular Sciences, Department of Medicine, and
| | - John P Cooke
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas 77030
| | | |
Collapse
|
34
|
Torrente L, Sanchez C, Moreno R, Chowdhry S, Cabello P, Isono K, Koseki H, Honda T, Hayes JD, Dinkova-Kostova AT, de la Vega L. Crosstalk between NRF2 and HIPK2 shapes cytoprotective responses. Oncogene 2017; 36:6204-6212. [PMID: 28692050 PMCID: PMC5641449 DOI: 10.1038/onc.2017.221] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 04/28/2017] [Accepted: 05/30/2017] [Indexed: 12/29/2022]
Abstract
Homeodomain interacting protein kinase-2 (HIPK2) is a member of the HIPK family of stress-responsive kinases that modulates cell growth, apoptosis, proliferation and development. HIPK2 has several well-characterised tumour suppressor roles, but recent studies suggest it can also contribute to tumour progression, although the underlying mechanisms are unknown. Herein, we have identified novel crosstalk between HIPK2 and the cytoprotective transcription factor NRF2. We show that HIPK2 is a direct transcriptional target of NRF2, identifying a functional NRF2 binding site in the HIPK2 gene locus and demonstrating for the first time a transcriptional mode of regulation for this kinase. In addition, HIPK2 is required for robust NRF2 responsiveness in cells and in vivo. By using both gain-of-function and loss-of-function approaches, we demonstrate that HIPK2 can elicit a cytoprotective response in cancer cells via NRF2. Our results have uncovered a new downstream effector of HIPK2, NRF2, which is frequently activated in human tumours correlating with chemoresistance and poor prognosis. Furthermore, our results suggest that modulation of either HIPK2 levels or activity could be exploited to impair NRF2-mediated signalling in cancer cells, and thus sensitise them to chemotherapeutic drugs.
Collapse
Affiliation(s)
- L Torrente
- Division of Cancer Research, School of Medicine, Jacqui Wood Cancer Centre, James Arrott Drive, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland
| | - C Sanchez
- Division of Cancer Research, School of Medicine, Jacqui Wood Cancer Centre, James Arrott Drive, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland
| | - R Moreno
- Division of Cancer Research, School of Medicine, Jacqui Wood Cancer Centre, James Arrott Drive, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland
| | - S Chowdhry
- Division of Cancer Research, School of Medicine, Jacqui Wood Cancer Centre, James Arrott Drive, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland
| | - P Cabello
- Division of Cancer Research, School of Medicine, Jacqui Wood Cancer Centre, James Arrott Drive, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland
| | - K Isono
- Developmental Genetics, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - H Koseki
- Developmental Genetics, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - T Honda
- Department of Chemistry and Institute of Chemical Biology & Drug Discovery, Stony Brook University, Stony Brook, NY, USA
| | - J D Hayes
- Division of Cancer Research, School of Medicine, Jacqui Wood Cancer Centre, James Arrott Drive, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland
| | - A T Dinkova-Kostova
- Division of Cancer Research, School of Medicine, Jacqui Wood Cancer Centre, James Arrott Drive, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland
| | - L de la Vega
- Division of Cancer Research, School of Medicine, Jacqui Wood Cancer Centre, James Arrott Drive, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland
| |
Collapse
|
35
|
Blaquiere JA, Verheyen EM. Homeodomain-Interacting Protein Kinases: Diverse and Complex Roles in Development and Disease. Curr Top Dev Biol 2016; 123:73-103. [PMID: 28236976 DOI: 10.1016/bs.ctdb.2016.10.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The Homeodomain-interacting protein kinase (Hipk) family of proteins plays diverse, and at times conflicting, biological roles in normal development and disease. In this review we will highlight developmental and cellular roles for Hipk proteins, with an emphasis on the pleiotropic and essential physiological roles revealed through genetic studies. We discuss the myriad ways of regulating Hipk protein function, and how these may contribute to the diverse cellular roles. Furthermore we will describe the context-specific activities of Hipk family members in diseases such as cancer and fibrosis, including seemingly contradictory tumor-suppressive and oncogenic activities. Given the diverse signaling pathways regulated by Hipk proteins, it is likely that Hipks act to fine-tune signaling and may mediate cross talk in certain contexts. Such regulation is emerging as vital for development and in disease.
Collapse
Affiliation(s)
- Jessica A Blaquiere
- Department of Molecular Biology and Biochemistry, Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC, Canada
| | - Esther M Verheyen
- Department of Molecular Biology and Biochemistry, Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC, Canada.
| |
Collapse
|
36
|
Kuwano Y, Nishida K, Akaike Y, Kurokawa K, Nishikawa T, Masuda K, Rokutan K. Homeodomain-Interacting Protein Kinase-2: A Critical Regulator of the DNA Damage Response and the Epigenome. Int J Mol Sci 2016; 17:ijms17101638. [PMID: 27689990 PMCID: PMC5085671 DOI: 10.3390/ijms17101638] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Revised: 09/16/2016] [Accepted: 09/20/2016] [Indexed: 12/29/2022] Open
Abstract
Homeodomain-interacting protein kinase 2 (HIPK2) is a serine/threonine kinase that phosphorylates and activates the apoptotic program through interaction with diverse downstream targets including tumor suppressor p53. HIPK2 is activated by genotoxic stimuli and modulates cell fate following DNA damage. The DNA damage response (DDR) is triggered by DNA lesions or chromatin alterations. The DDR regulates DNA repair, cell cycle checkpoint activation, and apoptosis to restore genome integrity and cellular homeostasis. Maintenance of the DDR is essential to prevent development of diseases caused by genomic instability, including cancer, defects of development, and neurodegenerative disorders. Recent studies reveal a novel HIPK2-mediated pathway for DDR through interaction with chromatin remodeling factor homeodomain protein 1γ. In this review, we will highlight the molecular mechanisms of HIPK2 and show its functions as a crucial DDR regulator.
Collapse
Affiliation(s)
- Yuki Kuwano
- Department of Pathophysiology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan.
| | - Kensei Nishida
- Department of Pathophysiology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan.
| | - Yoko Akaike
- Department of Pathophysiology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan.
| | - Ken Kurokawa
- Department of Pathophysiology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan.
| | - Tatsuya Nishikawa
- Department of Pathophysiology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan.
| | - Kiyoshi Masuda
- Department of Pathophysiology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan.
| | - Kazuhito Rokutan
- Department of Pathophysiology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan.
| |
Collapse
|
37
|
Lee S, Shang Y, Redmond SA, Urisman A, Tang AA, Li KH, Burlingame AL, Pak RA, Jovičić A, Gitler AD, Wang J, Gray NS, Seeley WW, Siddique T, Bigio EH, Lee VMY, Trojanowski JQ, Chan JR, Huang EJ. Activation of HIPK2 Promotes ER Stress-Mediated Neurodegeneration in Amyotrophic Lateral Sclerosis. Neuron 2016; 91:41-55. [PMID: 27321923 DOI: 10.1016/j.neuron.2016.05.021] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 02/16/2016] [Accepted: 05/04/2016] [Indexed: 12/13/2022]
Abstract
Persistent accumulation of misfolded proteins causes endoplasmic reticulum (ER) stress, a prominent feature in many neurodegenerative diseases including amyotrophic lateral sclerosis (ALS). Here we report the identification of homeodomain interacting protein kinase 2 (HIPK2) as the essential link that promotes ER-stress-induced cell death via the IRE1α-ASK1-JNK pathway. ER stress, induced by tunicamycin or SOD1(G93A), activates HIPK2 by phosphorylating highly conserved serine and threonine residues (S359/T360) within the activation loop of the HIPK2 kinase domain. In SOD1(G93A) mice, loss of HIPK2 delays disease onset, reduces cell death in spinal motor neurons, mitigates glial pathology, and improves survival. Remarkably, HIPK2 activation positively correlates with TDP-43 proteinopathy in NEFH-tTA/tetO-hTDP-43ΔNLS mice, sporadic ALS and C9ORF72 ALS, and blocking HIPK2 kinase activity protects motor neurons from TDP-43 cytotoxicity. These results reveal a previously unrecognized role of HIPK2 activation in ER-stress-mediated neurodegeneration and its potential role as a biomarker and therapeutic target for ALS. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Sebum Lee
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yulei Shang
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Stephanie A Redmond
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Anatoly Urisman
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Amy A Tang
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Kathy H Li
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Alma L Burlingame
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ryan A Pak
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ana Jovičić
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Aaron D Gitler
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jinhua Wang
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School & Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Nathanael S Gray
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School & Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - William W Seeley
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Teepu Siddique
- Division of Neuromuscular Medicine and Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Eileen H Bigio
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Virginia M-Y Lee
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jonah R Chan
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Eric J Huang
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA; Pathology Service 113B, VA Medical Center, San Francisco, CA 94121, USA.
| |
Collapse
|
38
|
Jiang Y, Liu H, Liu WJ, Tong HB, Chen CJ, Lin FG, Zhuo YH, Qian XZ, Wang ZB, Wang Y, Zhang P, Jia HL. Endothelial Aquaporin-1 (AQP1) Expression Is Regulated by Transcription Factor Mef2c. Mol Cells 2016; 39:292-298. [PMID: 26923194 PMCID: PMC4844935 DOI: 10.14348/molcells.2016.2223] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 11/25/2015] [Accepted: 11/26/2015] [Indexed: 11/30/2022] Open
Abstract
Aquaporin 1 (AQP1) is expressed in most microvasculature endothelial cells and forms water channels that play major roles in a variety of physiologic processes. This study aimed to delineate the transcriptional regulation of AQP1 by Mef2c in endothelial cells. Mef2c cooperated with Sp1 to activate human AQP1 transcription by binding to its proximal promoter in human umbilical cord vein endothelial cells (HUVEC). Over-expression of Mef2c, Sp1, or Mef2c/Sp1 increased HUVEC migration and tube-forming ability, which can be abolished AQP1 knockdown. These data indicate that AQP1 is a direct target of Mef2c in regulating angiogenesis and vasculogenesis of endothelial cells.
Collapse
Affiliation(s)
- Yong Jiang
- Medical Examination College, Jilin Medical University, People’s
Republic of China
| | - He Liu
- Medical Examination College, Jilin Medical University, People’s
Republic of China
| | - Wen-jing Liu
- Medical Examination College, Jilin Medical University, People’s
Republic of China
| | - Hai-bin Tong
- Life Science Research Center, Beihua University, Jilin, People’s
Republic of China
| | - Chang-jun Chen
- Medical Examination College, Jilin Medical University, People’s
Republic of China
| | - Fu-gui Lin
- Medical Examination College, Jilin Medical University, People’s
Republic of China
| | - Yan-hang Zhuo
- Medical Examination College, Jilin Medical University, People’s
Republic of China
| | - Xiao-zhen Qian
- Medical Examination College, Jilin Medical University, People’s
Republic of China
| | - Zeng-bin Wang
- Medical Examination College, Jilin Medical University, People’s
Republic of China
| | - Yu Wang
- Medical Examination College, Jilin Medical University, People’s
Republic of China
| | - Peng Zhang
- Medical Examination College, Jilin Medical University, People’s
Republic of China
| | - Hong-liang Jia
- Medical Examination College, Jilin Medical University, People’s
Republic of China
| |
Collapse
|
39
|
Rodriguez-Gil A, Ritter O, Hornung J, Stekman H, Krüger M, Braun T, Kremmer E, Kracht M, Schmitz ML. HIPK family kinases bind and regulate the function of the CCR4-NOT complex. Mol Biol Cell 2016; 27:1969-80. [PMID: 27122605 PMCID: PMC4907730 DOI: 10.1091/mbc.e15-09-0629] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 04/18/2016] [Indexed: 12/11/2022] Open
Abstract
Down-regulation of the HIPK interactor CNOT2 leads to reduced HIPK2 protein levels, identifying the CCR4-NOT complex as a new regulator of HIPK2 abundance. Functional assays reveal that HIPK2 and HIPK1 restrict CNOT2-dependent mRNA decay, thus extending the regulatory potential of these kinases to the level of posttranscriptional gene regulation. The serine/threonine kinase HIPK2 functions as a regulator of developmental processes and as a signal integrator of a wide variety of stress signals, such as DNA damage, hypoxia, and reactive oxygen intermediates. Because the kinase is generated in a constitutively active form, its expression levels are restricted by a variety of different mechanisms. Here we identify the CCR4-NOT complex as a new regulator of HIPK2 abundance. Down-regulation or knockout of the CCR4-NOT complex member CNOT2 leads to reduced HIPK2 protein levels without affecting the expression level of HIPK1 or HIPK3. A fraction of all HIPK family members associates with the CCR4-NOT components CNOT2 and CNOT3. HIPKs also phosphorylate the CCR4-NOT complex, a feature that is shared with their yeast progenitor kinase, YAK1. Functional assays reveal that HIPK2 and HIPK1 restrict CNOT2-dependent mRNA decay. HIPKs are well known regulators of transcription, but the mutual regulation between CCR4-NOT and HIPKs extends the regulatory potential of these kinases by enabling posttranscriptional gene regulation.
Collapse
Affiliation(s)
- Alfonso Rodriguez-Gil
- Institute of Biochemistry, Medical Faculty, Justus-Liebig-University, Member of the German Center for Lung Research, D-35392 Giessen, Germany
| | - Olesja Ritter
- Institute of Biochemistry, Medical Faculty, Justus-Liebig-University, Member of the German Center for Lung Research, D-35392 Giessen, Germany
| | - Juliane Hornung
- Institute of Biochemistry, Medical Faculty, Justus-Liebig-University, Member of the German Center for Lung Research, D-35392 Giessen, Germany
| | - Hilda Stekman
- Institute of Biochemistry, Medical Faculty, Justus-Liebig-University, Member of the German Center for Lung Research, D-35392 Giessen, Germany
| | - Marcus Krüger
- Max Planck Institute for Heart and Lung Research, D-61231 Bad Nauheim, Germany
| | - Thomas Braun
- Max Planck Institute for Heart and Lung Research, D-61231 Bad Nauheim, Germany
| | - Elisabeth Kremmer
- Institute of Molecular Immunology, Helmholtz Center Munich, German Research Center for Environmental Health, D-81377 Munich; Germany
| | - Michael Kracht
- Rudolf-Buchheim-Institute of Pharmacology, Justus-Liebig-University, Member of the German Center for Lung Research, D-35392 Giessen, Germany
| | - M Lienhard Schmitz
- Institute of Biochemistry, Medical Faculty, Justus-Liebig-University, Member of the German Center for Lung Research, D-35392 Giessen, Germany
| |
Collapse
|
40
|
Luo SX, Huang EJ. Dopaminergic Neurons and Brain Reward Pathways: From Neurogenesis to Circuit Assembly. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 186:478-88. [PMID: 26724386 DOI: 10.1016/j.ajpath.2015.09.023] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 09/01/2015] [Accepted: 09/23/2015] [Indexed: 11/26/2022]
Abstract
Midbrain dopaminergic (DA) neurons in the substantia nigra pars compacta and ventral tegmental area regulate extrapyramidal movement and important cognitive functions, including motivation, reward associations, and habit learning. Dysfunctions in DA neuron circuitry have been implicated in several neuropsychiatric disorders, including addiction and schizophrenia, whereas selective degeneration of DA neurons in substantia nigra pars compacta is a key neuropathological feature in Parkinson disease. Efforts to understand these disorders have focused on dissecting the underlying causes, as well as developing therapeutic strategies to replenish dopamine deficiency. In particular, the promise of cell replacement therapies for clinical intervention has led to extensive research in the identification of mechanisms involved in DA neuron development. It is hoped that a comprehensive understanding of these mechanisms will lead to therapeutic strategies that improve the efficiency of DA neuron production, engraftment, and function. This review provides a comprehensive discussion on how Wnt/β-catenin and sonic hedgehog-Smoothened signaling mechanisms control the specification and expansion of DA progenitors and the differentiation of DA neurons. We also discuss how mechanisms involving transforming growth factor-β and transcriptional cofactor homeodomain interacting protein kinase 2 regulate the survival and maturation of DA neurons in early postnatal life. These results not only reveal fundamental mechanisms regulating DA neuron development, but also provide important insights to their potential contributions to neuropsychiatric and neurodegenerative diseases.
Collapse
Affiliation(s)
- Sarah X Luo
- Neuroscience Graduate Program, University of California San Francisco, San Francisco, California; Department of Pathology, University of California San Francisco, San Francisco, California
| | - Eric J Huang
- Neuroscience Graduate Program, University of California San Francisco, San Francisco, California; Department of Pathology, University of California San Francisco, San Francisco, California; Pathology Service 113B, San Francisco Veterans Affairs Medical Center, San Francisco, California.
| |
Collapse
|
41
|
RNA Seq profiling reveals a novel expression pattern of TGF-β target genes in human blood eosinophils. Immunol Lett 2015; 167:1-10. [PMID: 26112417 DOI: 10.1016/j.imlet.2015.06.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 06/05/2015] [Accepted: 06/15/2015] [Indexed: 12/16/2022]
Abstract
Despite major advances in our understanding of TGF-β signaling in multiple cell types, little is known about the direct target genes of this pathway in human eosinophils. These cells constitute the major inflammatory component present in the sputum and lung of active asthmatics and their numbers correlate well with disease severity. During the transition from acute to chronic asthma, TGF-β levels rise several fold in the lung which drives fibroblasts to produce extracellular matrix (ECM) and participate in airway and parenchymal remodeling. In this report, we use purified blood eosinophils from healthy donors and analyze baseline and TGF-β responsive genes by RNA Seq, and demonstrate that eosinophils (PBE) express 7981 protein-coding genes of which 178 genes are up-regulated and 199 genes are down-regulated by TGF-β. While 18 target genes have been previously associated with asthma and eosinophilic disorders, the vast majority have been implicated in cell death and survival, differentiation, and cellular function. Ingenuity pathway analysis revealed that 126 canonical pathways are activated by TGF-β including iNOS, TREM1, p53, IL-8 and IL-10 signaling. As TGF-β is an important cytokine for eosinophil function and survival, and pulmonary inflammation and fibrosis, our results represent a significant step toward the identification of novel TGF-β responsive genes and provide a potential therapeutic opportunity by selectively targeting relevant genes and pathways.
Collapse
|
42
|
Griseri P, Pagès G. Control of pro-angiogenic cytokine mRNA half-life in cancer: the role of AU-rich elements and associated proteins. J Interferon Cytokine Res 2015; 34:242-54. [PMID: 24697202 DOI: 10.1089/jir.2013.0140] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Control of mRNA half-life plays a central role in normal development and disease. Several pathological conditions, such as inflammation and cancer, tightly correlate with deregulation in mRNA stability of pro-inflammatory genes. Among these, pro-angiogenesis cytokines, which play a crucial role in the formation of new blood vessels, normally show rapid mRNA decay patterns. The mRNA half-life of these genes appears to be regulated by mRNA-binding proteins that interact with AU-rich elements (AREs) in the 3'-untranslated region of mRNAs. Some of these RNA-binding proteins, such as tristetraprolin (TTP), ARE RNA-binding protein 1, and KH-type splicing regulatory protein, normally promote mRNA degradation. Conversely, other proteins, such as embryonic lethal abnormal vision-like protein 1 (HuR) and polyadenylate-binding protein-interacting protein 2, act as antagonists, stabilizing the mRNA. The steady state levels of mRNA-binding proteins and their relative ratio is often perturbed in human cancers and associated with invasion and aggressiveness. Compelling evidence also suggests that underexpression of TTP and overexpression of HuR may be a useful prognostic and predictive marker in breast, colon, prostate, and brain cancers, indicating a potential therapeutic approach for these tumors. In this review, we summarize the main mechanisms involved in the regulation of mRNA decay of pro-angiogenesis cytokines in different cancers and discuss the interactions between the AU-rich-binding proteins and their mRNA targets.
Collapse
Affiliation(s)
- Paola Griseri
- 1 U.O.C Medical Genetics, Institute Giannina Gaslini , Genoa, Italy
| | | |
Collapse
|
43
|
Schmitz ML, Rodriguez-Gil A, Hornung J. Integration of stress signals by homeodomain interacting protein kinases. Biol Chem 2015; 395:375-86. [PMID: 24225127 DOI: 10.1515/hsz-2013-0264] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 11/08/2013] [Indexed: 11/15/2022]
Abstract
The family of homeodomain interacting protein kinases (HIPKs) consists of four related kinases, HIPK1 to HIPK4. These serine/threonine kinases are evolutionary conserved and derive from the yeast kinase Yak1. The largest group of HIPK phosphorylation substrates is represented by transcription factors and chromatin-associated regulators of gene expression, thus transferring HIPK-derived signals into changes of gene expression programs. The HIPKs mainly function as regulators of developmental processes and as integrators of a wide variety of stress signals. A number of conditions representing precarious situations, such as DNA damage, hypoxia, reactive oxygen intermediates and metabolic stress affect the function of HIPKs. The kinases function as integrators for these stress signals and feed them into many different downstream effector pathways that serve to cope with these precarious situations. HIPKs do not function as essential core components in the different stress signaling pathways, but rather serve as modulators of signal output and as connectors of different stress signaling pathways. Their central role as signaling hubs with the ability to shape many downstream effector pathways frequently implies them in proliferative diseases such as cancer or fibrosis.
Collapse
|
44
|
van der Laden J, Soppa U, Becker W. Effect of tyrosine autophosphorylation on catalytic activity and subcellular localisation of homeodomain-interacting protein kinases (HIPK). Cell Commun Signal 2015; 13:3. [PMID: 25630557 PMCID: PMC4353451 DOI: 10.1186/s12964-014-0082-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 12/30/2014] [Indexed: 12/18/2022] Open
Abstract
Background Homeodomain interacting protein kinases (HIPKs) function as modulators of cellular stress responses and regulate cell differentiation, proliferation and apoptosis. The HIPK family includes HIPK1, HIPK2 and HIPK3, which share a similar domain structure, and the more distantly related HIPK4. Although HIPKs phosphorylate their substrates on serine or threonine residues, it was recently reported that HIPK2 depends on the autophosphorylation of a conserved tyrosine in the activation loop to acquire full catalytic activity and correct subcellular localization. In this study we addressed the question whether tyrosine autophosphorylation in the activation loop has a similar function in the other members of the HIPK family. Results All HIPKs contained phosphotyrosine when expressed in HeLa cells. Catalytically inactive point mutants were not tyrosine-phosphorylated, indicating that HIPKs are dual-specificity protein kinases that autophosphorylate on tyrosine residues. HIPK point mutants lacking the conserved tyrosine residue in the activation loop showed reduced catalytic activity towards peptide and protein substrates. Analysis of these mutants revealed that HIPK1, HIPK2 and HIPK3 but not HIPK4 are capable of autophosphorylating on other tyrosines. Inhibition of tyrosine phosphatase activity by treatment with vanadate enhanced global phosphotyrosine content of HIPK1, HIPK2 and HIPK3 but did not affect tyrosine phosphorylation in the activation loop. Mutation of the activation-loop tyrosines resulted in a redistribution of HIPK1 and HIPK2 from a speckle-like subnuclear compartment to the cytoplasm, whereas catalytically inactive point mutants showed the same pattern of cellular distribution as the wild type proteins. In contrast, mutation of the activating tyrosine did not increase the low percentage of cells with extranuclear HIPK3. HIPK4 was excluded from the nucleus with no difference between the wild type kinase and the point mutants. Conclusions These results show that HIPKs share the mechanism of activation by tyrosine autophosphorylation with the closely related DYRK family (dual-specificity tyrosine phosphorylation regulated kinase). However, members of the HIPK family differ regarding the subcellular localization and its dependence on tyrosine autophosphorylation. Electronic supplementary material The online version of this article (doi:10.1186/s12964-014-0082-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jan van der Laden
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Wendlingweg 2, 52057, Aachen, Germany.
| | - Ulf Soppa
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Wendlingweg 2, 52057, Aachen, Germany.
| | - Walter Becker
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Wendlingweg 2, 52057, Aachen, Germany.
| |
Collapse
|
45
|
Wook Choi D, Yong Choi C. HIPK2 modification code for cell death and survival. Mol Cell Oncol 2014; 1:e955999. [PMID: 27308327 PMCID: PMC4905192 DOI: 10.1080/23723548.2014.955999] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Revised: 07/07/2014] [Accepted: 07/08/2014] [Indexed: 11/03/2022]
Abstract
Homeodomain-interacting protein kinase 2 (HIPK2) is a serine/threonine protein kinase that participates in the regulation of diverse cellular activities as a transcriptional cofactor and signal transducer. HIPK2 senses various signaling cues that in turn phosphorylate downstream substrates to coordinate developmental processes, cell cycle regulation, cell proliferation, differentiation, and the DNA damage response. HIPK2 functions are affected by its catalytic activity, stability, and subcellular localization, which in turn are dynamically regulated by diverse post-translational modifications such as polyubiquitination, SUMOylation, phosphorylation, and acetylation. HIPK2 is not modified with small molecules and/or peptides individually or independently, but in a combinatorial manner that is referred to as the “HIPK2 modification code.” HIPK2 integrates various signaling cues and senses different doses of DNA damage and ROS stimuli, which are reflected by unique patterns of HIPK2 modification. Hence, the HIPK2 modification code differentially contributes to cellular homeostasis and determination of cell fate depending on cellular context.
Collapse
Affiliation(s)
- Dong Wook Choi
- Department of Biological Sciences; Sungkyunkwan University ; Suwon, Republic of Korea
| | - Cheol Yong Choi
- Department of Biological Sciences; Sungkyunkwan University ; Suwon, Republic of Korea
| |
Collapse
|
46
|
Bai XL, Zhang Q, Ye LY, Liang F, Sun X, Chen Y, Hu QD, Fu QH, Su W, Chen Z, Zhuang ZP, Liang TB. Myocyte enhancer factor 2C regulation of hepatocellular carcinoma via vascular endothelial growth factor and Wnt/β-catenin signaling. Oncogene 2014; 34:4089-97. [PMID: 25328135 DOI: 10.1038/onc.2014.337] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 08/15/2014] [Accepted: 08/29/2014] [Indexed: 01/11/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the leading malignancies worldwide. Myocyte enhancer factor 2C (MEF2C) was traditionally regarded as a development-associated factor and was recently reported to be an oncogene candidate. We have previously reported overexpression of MEF2C in HCC; however, the roles of MEF2C in HCC remain to be clarified. In this study, HCC cell lines and a xenograft mouse model were used to determine the functions of MEF2C in vitro and in vivo, respectively. Specific plasmids and small interfering RNA were used to upregulate and downregulate MEF2C expression, respectively. Functional assays were performed to assess the influence of MEF2C on cell proliferation, and VEGF-induced vasculogenic mimicry, migration/invasion as well as angiogenesis. Co-immunoprecipitation was conducted to identify the interaction of MEF2C and β-catenin. Human HCC tissue microarrays were used to investigate correlations among MEF2C, β-catenin and involved biomarkers. MEF2C was found to mediate VEGF-induced vasculogenic mimicry, angiogenesis and migration/invasion, with involvement of the p38 MAPK and PKC signaling pathways. However, MEF2C itself inhibited tumor growth in vitro and in vivo. MEF2C was upregulated by and directly interacted with β-catenin. The nuclear translocation of β-catenin blocked by MEF2C was responsible for MEF2C-mediated growth inhibition. The nuclear translocation of MEF2C was associated with intracellular calcium signaling induced by β-catenin. HCC microarrays showed correlations of nuclear MEF2C with the angiogenesis-associated biomarker, CD31, and cytosolic MEF2C with the proliferation-associated biomarker, Ki-67. MEF2C showed double-edged activities in HCC, namely mediating VEGF-induced malignancy enhancement while inhibiting cancer proliferation via blockade of Wnt/β-catenin signaling. The overall effect of MEF2C in HCC progression regulation was dictated by its subcellular distribution. This should be determined prior to any MEF2C-associated intervention in HCC.
Collapse
Affiliation(s)
- X L Bai
- 1] Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China [2] Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Q Zhang
- 1] Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China [2] Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - L Y Ye
- 1] Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China [2] Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - F Liang
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - X Sun
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Y Chen
- Department of General Surgery, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Q D Hu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Q H Fu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - W Su
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Z Chen
- Zhejiang Key Laboratory of Gastro-Intestinal Pathophysiology, Zhejiang Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Z P Zhuang
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - T B Liang
- 1] Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China [2] Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
47
|
Hu Q, Zhou LH, Liu X, Sui H, Fu XL, Yan LL, Ren JL, Li Q. HIPK2 reduces VEGF expression by regulating expression of COX-2 and β-catenin in human colorectal cancer cells. Shijie Huaren Xiaohua Zazhi 2014; 22:1266-1274. [DOI: 10.11569/wcjd.v22.i9.1266] [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: 02/06/2023] Open
Abstract
AIM: To study the effects of overexpression of homeodomain-interacting protein kinase 2 (HIPK2) on the expression of cyclooxygenase 2 (COX-2) and β-catenin in HCT-116 cells, and to explore the regulatory effect of the HIPK2 gene on angiogenic factor expression in human colorectal cancer cells.
METHODS: The pEGFP-N3 vector or pEGFP-N3-HIPK2 plasmid was transfected into human colorectal cancer HCT-116 cells using Lipofectamine 2000, and untransfected cells were used as normal controls. The expression of VEGF (vascular endothelial growth factor) in cell culture medium was detected by ELISA assay, and the protein expression of β-catenin was tested by Western blot. The COX-2 promoter containing plasmid pGL3-basic-COX-2 was co-transfected with pRL-SV40 in the three groups above. COX-2 promoter activity was detected by dual luciferase activity assay, and COX-2 mRNA level was determined by real-time PCR.
RESULTS: The pEGFP-N3-HIPK2 group and pEGFP-N3 group had the same fluorescent level, and there was no statistically significant difference in HCT-116 cells. The VEGF expression was significantly inhibited (857.54 pg/mL ± 65.04 pg/mL vs 368.32 pg/mL ± 98.82 pg/mL, P < 0.05) after pEGFP-N3-HIPK2 plasmid transfection. COX-2 promoter activity and mRNA expression were significantly lower in the pEGFP-N3-HIPK2 group than in the pEGFP-N3 group (75.467×10-5 ± 18.666×10-5vs 266.407×10-5 ± 40.902×10-5, 1.07×10-4 ± 0.32×10-4vs 3.48×10-4 ± 0.64×10-4P < 0.05 for both). HIPK2 could inhibit the protein expression of β-catenin but up-regulate p-β-catenin expression.
CONCLUSION: pEGFP-N3-HIPK2 transfection significantly increased the expression of HIPK2, and reduced the transcription of the COX2 gene and VEGF level in human colorectal cancer HCT-116 cells. The mechanisms may be related to the inhibition of COX-2 and β-catenin expression in cells, and regulation of β-catenin-COX-2 signal transduction.
Collapse
|
48
|
Zippel N, Malik RA, Frömel T, Popp R, Bess E, Strilic B, Wettschureck N, Fleming I, Fisslthaler B. Transforming Growth Factor-β–Activated Kinase 1 Regulates Angiogenesis via AMP-Activated Protein Kinase-α1 and Redox Balance in Endothelial Cells. Arterioscler Thromb Vasc Biol 2013; 33:2792-9. [DOI: 10.1161/atvbaha.113.301848] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Objective—
Transforming growth factor-β–activated kinase 1 (TAK1) is a mitogen-activated protein 3-kinase and an AMP-activated protein kinase (AMPK) kinase in some cell types. Although TAK1
−/−
mice display defects in developmental vasculogenesis, the role of TAK1 in endothelial cells has not been investigated in detail.
Approach and Results—
TAK1 downregulation (small interfering RNA) in human endothelial cells attenuated proliferation without inducing apoptosis and diminished endothelial cell migration, as well as tube formation. Cytokine- and vascular endothelial growth factor (VEGF)–induced endothelial cell sprouting in a modified spheroid assay were abrogated by TAK1 downregulation. Moreover, VEGF–induced endothelial sprouting was impaired in aortic rings from mice lacking TAK1 in endothelial cells (TAK
ΔEC
). TAK1 inhibition and downregulation also inhibited VEGF–stimulated phosphorylation of several kinases, including AMPK. Proteomic analyses revealed that superoxide dismutase 2 (SOD2) expression was reduced in TAK1-deficient endothelial cells, resulting in attenuated hydrogen peroxide production but increased mitochondrial superoxide production. Endothelial cell SOD2 expression was also attenuated by AMPK inhibition and in endothelial cells from AMPKα1
−/−
mice but was unaffected by inhibitors of c-Jun N-terminal kinase, p38, extracellular signal–regulated kinase 1/2, or phosphatidylinositol 3-kinase/Akt. Moreover, the impaired endothelial sprouting from TAK
ΔEC
aortic rings was abrogated in the presence of polyethylene glycol-SOD, and tube formation was normalized by the overexpression of SOD2. A similar rescue of angiogenesis was observed in polyethylene glycol-SOD–treated aortic rings from mice with endothelial cell–specific deletion of the AMPKα1.
Conclusions—
These results establish TAK1 as an AMPKα1 kinase that regulates vascular endothelial growth factor–induced and cytokine-induced angiogenesis by modulating SOD2 expression and the superoxide anion:hydrogen peroxide balance.
Collapse
Affiliation(s)
- Nina Zippel
- From the Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt, Germany and DZHK (German Centre for Cardiovascular Research) partner site Rhine-Main (N.Z., R.A.M., T.F., R.P., E.B., I.F., B.F.); and Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (B.S., N.W.)
| | - Randa Abdel Malik
- From the Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt, Germany and DZHK (German Centre for Cardiovascular Research) partner site Rhine-Main (N.Z., R.A.M., T.F., R.P., E.B., I.F., B.F.); and Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (B.S., N.W.)
| | - Timo Frömel
- From the Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt, Germany and DZHK (German Centre for Cardiovascular Research) partner site Rhine-Main (N.Z., R.A.M., T.F., R.P., E.B., I.F., B.F.); and Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (B.S., N.W.)
| | - Rüdiger Popp
- From the Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt, Germany and DZHK (German Centre for Cardiovascular Research) partner site Rhine-Main (N.Z., R.A.M., T.F., R.P., E.B., I.F., B.F.); and Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (B.S., N.W.)
| | - Elke Bess
- From the Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt, Germany and DZHK (German Centre for Cardiovascular Research) partner site Rhine-Main (N.Z., R.A.M., T.F., R.P., E.B., I.F., B.F.); and Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (B.S., N.W.)
| | - Boris Strilic
- From the Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt, Germany and DZHK (German Centre for Cardiovascular Research) partner site Rhine-Main (N.Z., R.A.M., T.F., R.P., E.B., I.F., B.F.); and Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (B.S., N.W.)
| | - Nina Wettschureck
- From the Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt, Germany and DZHK (German Centre for Cardiovascular Research) partner site Rhine-Main (N.Z., R.A.M., T.F., R.P., E.B., I.F., B.F.); and Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (B.S., N.W.)
| | - Ingrid Fleming
- From the Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt, Germany and DZHK (German Centre for Cardiovascular Research) partner site Rhine-Main (N.Z., R.A.M., T.F., R.P., E.B., I.F., B.F.); and Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (B.S., N.W.)
| | - Beate Fisslthaler
- From the Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt, Germany and DZHK (German Centre for Cardiovascular Research) partner site Rhine-Main (N.Z., R.A.M., T.F., R.P., E.B., I.F., B.F.); and Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (B.S., N.W.)
| |
Collapse
|