1
|
Langille BL, Juárez M, Prieto N, Boison S, Lim PS, Swift BD, Garber AF. Candidate genes associated with fatty acid compositions in north American Atlantic salmon (Salmo salar). BMC Genomics 2024; 25:1208. [PMID: 39695999 DOI: 10.1186/s12864-024-11131-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 12/05/2024] [Indexed: 12/20/2024] Open
Abstract
Fatty acids are a requirement for normal development, however, since humans are unable to de novo produce essential fatty acids, they must be obtained from diet. Atlantic salmon is a major dietary source of nutritious and digestible fatty acids. Here, we set out to uncover the genomic basis of individual fatty acids and indices (saturated fatty acids, monounsaturated fatty acids, polyunsaturated fatty acids, n-3, and n-6) in 208 North American Atlantic salmon, to understand selection potential toward increasing relative quantities of essential fatty acids and to identify candidate genes for future research. Total n-6 (pro-inflammatory) was higher than total n-3 (anti-inflammatory) fatty acids with a ratio of 1 : 1.31 (n-3 : n-6). Heritability of fatty acids ranged from 0 to 0.99, however, most fatty acids and indices had moderate to high heritabilities (ranged from 0.20 to 0.88), implying that selection for improvement of traits could be possible. We found the same significant markers on chromosome 23 (based on false discovery rate thresholds of 2.0e-6 and suggestive significant thresholds of 2.0e-5 in Manhattan plots) in four fatty acids (γ-linoleic acid, stearidonic acid, dihimo-γ-linolenic acid, and eicosatrienoic acid), where three genes (sin3b, acbd6, and fads2) are known to be involved in lipid metabolism. These genes, fads2 in particular, would all make ideal candidates for future functional studies. In addition, there were four fatty acids with loci over the suggestive significant threshold with a variety of markers on different chromosomes (lauric acid, stearic acid, eicosatetraenoic acid (ETA), and docosadienoic acid), with associated genes that had relevant functions to fatty acids or adipose cells in general.
Collapse
Affiliation(s)
- Barbara L Langille
- The Huntsman Marine Science Centre, 1 Lower Campus Rd., St. Andrews, NB E5B 2L7, Canada.
| | - Manuel Juárez
- Agriculture and Agri-Food Canada Lacombe Research and Development Centre, 6000 C and E Trail, Lacombe, AB T4L 1W1, Canada
| | - Nuria Prieto
- Agriculture and Agri-Food Canada Lacombe Research and Development Centre, 6000 C and E Trail, Lacombe, AB T4L 1W1, Canada
| | - Solomon Boison
- Mowi Genetics AS, Sandviksbodene 77A, Bergen, 5035, Norway
| | - Panya Sae Lim
- Mowi Genetics AS, Sandviksbodene 77A, Bergen, 5035, Norway
| | - Bruce D Swift
- Tri-Gen Fish Improvement Ltd., Site 13 Comp 27 RR1, Lacombe, AB, T4L 2N1, Canada
| | - Amber F Garber
- The Huntsman Marine Science Centre, 1 Lower Campus Rd., St. Andrews, NB E5B 2L7, Canada
| |
Collapse
|
2
|
Xia C, Jiang Y, Zhao Y, Chen Z, Sun Y, Sun Z, Cui R, Tao W. Genipin 1-O-β-D-gentiobioside ameliorates CUMS-induced prefrontal cortex neuron neuronal apoptosis by modulating HIPK2 SUMOylation. Int Immunopharmacol 2024; 141:112985. [PMID: 39213873 DOI: 10.1016/j.intimp.2024.112985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/11/2024] [Accepted: 08/17/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Depression is a common mental illness with more than 280 million sufferers worldwide. Inflammation, particularly the c-Jun amino-terminal kinase (JNK) pathway, contributes to depression development and neuronal apoptosis. Gardenia is a herb with therapeutic effects on depression that has been shown to inhibit neuronal apoptosis. However, one of the components in gardenia, Genipin 1-O-β-D-gentiobioside(GG), has been less studied for its mechanism on depression. Thus, in the current study, we investigate how Genipin 1-O-β-D-gentiobioside improves depression and elucidate its possible mechanism of action. METHODS In this investigation, we utilize a chronic unpredictable mild stress (CUMS) mouse model and corticosterone-induced primary cortical neurons to examine the role of GG in ameliorating depressive symptoms and neuronal apoptosis. TUNEL staining and flow cytometry assessed the effects of GG on neuronal apoptosis. Western Blot analyses and immunofluorescence assays apoptosis-related proteins in the prefrontal cortex and primary neurons. The site of action of GG in regulating homeodomain interacting protein kinase 2 (HIPK2) SUMOylation was further explored in primary neurons. We constructed siRNA-SUMO1 vectors to transfect primary neuronal cells with intracellular SUMO1 knockdown. Proximity ligation assay (PLA) experiments were performed on primary neurons according to the instructions of the assay kit to observe the physical relationship between HIPK2 and SUMO1. We predicted the HIPK2 SUMOylation modification site by an online database and constructed vectors to target and site-directed mutagenesis, then to transfected primary neuronal cells. RESULTS The results showed that GG effectively alleviated depressive-like behaviours, down-regulated apoptosis-related proteins (p-JNK, Bax, Cleaved-Caspase-3), and inhibited neuronal apoptosis in CUMS-induced depressed mice and corticosterone-induced primary cortical neurons. We reveal a complex mechanism underlying the link between GG, SUMOylation of HIPK2, and complex pathways of neuronal apoptosis regulation. K326 and K1189 are the key SUMOylation sites regulated by GG in intricate interactions of apoptosis-related proteins. CONCLUSION Our study demonstrated that GG exerts antidepressant-like actions through neuroprotective effects by inhibiting the apoptosis of prefrontal cortex neurons, revealing the mechanism of GG inhibition of JNK phosphorylation by enhancing HIPK2 SUMOylation.
Collapse
Affiliation(s)
- Changbo Xia
- Department of Pharmacy, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, Xinxiang 453000, Henan, China
| | - Yue Jiang
- Department of Pharmacy, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, Xinxiang 453000, Henan, China
| | - Yan Zhao
- Anqing First People's Hospital of Anhui Medical University, Anqing 246004, Anhui, China
| | - Zhuzi Chen
- Jiangsu Health Vocational College, Nanjing 210000, Jiangsu, China
| | - Ying Sun
- Department of Pathology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, Henan, China
| | - Zhongwen Sun
- College of Medicine, Lishui University, Lishui 323000, Fujian, China
| | - Ruijie Cui
- Department of Pharmacy, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, Xinxiang 453000, Henan, China.
| | - Weiwei Tao
- Department of Pharmacy, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, Xinxiang 453000, Henan, China; School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China.
| |
Collapse
|
3
|
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] [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
|
4
|
Feng Y, Li Z, Wang H, Liu BC, Lee K, He JC. HIPK2 C-terminal domain inhibits NF-κB signaling and renal inflammation in kidney injury. JCI Insight 2024; 9:e175153. [PMID: 38512421 PMCID: PMC11141872 DOI: 10.1172/jci.insight.175153] [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: 09/07/2023] [Accepted: 03/14/2024] [Indexed: 03/23/2024] Open
Abstract
HIPK2 is a multifunctional kinase that acts as a key pathogenic mediator of chronic kidney disease and fibrosis. It acts as a central effector of multiple signaling pathways implicated in kidney injury, such as TGF-β/Smad3-mediated extracellular matrix accumulation, NF-κB-mediated inflammation, and p53-mediated apoptosis. Thus, a better understanding of the specific HIPK2 regions necessary for distinct downstream pathway activation is critical for optimal drug development for CKD. Our study now shows that caspase-6-mediated removal of the C-terminal region of HIPK2 (HIPK2-CT) lead to hyperactive p65 NF-κB transcriptional response in kidney cells. In contrast, the expression of cleaved HIPK2-CT fragment could restrain the NF-κB transcriptional activity by cytoplasmic sequestration of p65 and the attenuation of IκBα degradation. Therefore, we examined whether HIPK2-CT expression can be exploited to restrain renal inflammation in vivo. The induction of HIPK2-CT overexpression in kidney tubular cells attenuated p65 nuclear translocation, expression of inflammatory cytokines, and macrophage infiltration in the kidneys of mice with unilateral ureteral obstruction and LPS-induced acute kidney injury. Collectively, our findings indicate that the HIPK2-CT is involved in the regulation of nuclear NF-κB transcriptional activity and that HIPK2-CT or its analogs could be further exploited as potential antiinflammatory agents to treat kidney disease.
Collapse
Affiliation(s)
- Ye Feng
- Department of Medicine/Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Zhengzhe Li
- Department of Medicine/Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Heather Wang
- Department of Medicine/Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Bi-Cheng Liu
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Kyung Lee
- Department of Medicine/Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - John Cijiang He
- Department of Medicine/Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Renal Section, James J. Peters Veterans Affairs Medical Center, New York, New York, USA
| |
Collapse
|
5
|
Apaydin T, Zonis S, Zhou C, Valencia CW, Barrett R, Strous GJ, Mol JA, Chesnokova V, Melmed S. WIP1 is a novel specific target for growth hormone action. iScience 2023; 26:108117. [PMID: 37876819 PMCID: PMC10590974 DOI: 10.1016/j.isci.2023.108117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/22/2023] [Accepted: 09/29/2023] [Indexed: 10/26/2023] Open
Abstract
DNA damage repair (DDR) is mediated by phosphorylating effectors ATM kinase, CHK2, p53, and γH2AX. We showed earlier that GH suppresses DDR by suppressing pATM, resulting in DNA damage accumulation. Here, we show GH acting through GH receptor (GHR) inducing wild-type p53-inducible phosphatase 1 (WIP1), which dephosphorylated ATM and its effectors in normal human colon cells and three-dimensional human intestinal organoids. Mice bearing GH-secreting xenografts exhibited induced colon WIP1 with suppressed pATM and γH2AX. WIP1 was also induced in buffy coats derived from patients with elevated GH from somatotroph adenomas. In contrast, decreased colon WIP1 was observed in GHR-/- mice. WIP1 inhibition restored ATM phosphorylation and reversed GH-induced DNA damage. We elucidated a novel GH signaling pathway activating Src/AMPK to trigger HIPK2 nuclear-cytoplasmic relocation and suppressing WIP1 ubiquitination. Concordantly, blocking either AMPK or Src abolished GH-induced WIP1. We identify WIP1 as a specific target for GH-mediated epithelial DNA damage accumulation.
Collapse
Affiliation(s)
- Tugce Apaydin
- Department of Medicine, Pituitary Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Svetlana Zonis
- Department of Medicine, Pituitary Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Cuiqi Zhou
- Department of Medicine, Pituitary Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Christian Wong Valencia
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Robert Barrett
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ger J. Strous
- Center for Molecular Medicine, University Medical Center Utrecht, Institute of Biomembranes, Utrecht University, Utrecht, the Netherlands
| | - Jan A. Mol
- Department of Clinical Sciences of Companion Animals, Utrecht University, Utrecht, the Netherlands
| | - Vera Chesnokova
- Department of Medicine, Pituitary Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Shlomo Melmed
- Department of Medicine, Pituitary Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
6
|
He J, Zhu Q, Han P, Zhou T, Li J, Wang X, Cheng J. Transcriptomic Networks Reveal the Tissue-Specific Cold Shock Responses in Japanese Flounder ( Paralichthys olivaceus). BIOLOGY 2023; 12:784. [PMID: 37372069 DOI: 10.3390/biology12060784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 05/16/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023]
Abstract
Low temperature is among the important factors affecting the distribution, survival, growth, and physiology of aquatic animals. In this study, coordinated transcriptomic responses to 10 °C acute cold stress were investigated in the gills, hearts, livers, and spleens of Japanese flounder (Paralichthys olivaceus), an important aquaculture species in east Asia. Histological examination suggested different levels of injury among P. olivaceus tissues after cold shock, mainly in the gills and livers. Based on transcriptome and weighted gene coexpression network analysis, 10 tissue-specific cold responsive modules (CRMs) were identified, revealing a cascade of cellular responses to cold stress. Specifically, five upregulated CRMs were enriched with induced differentially expressed genes (DEGs), mainly corresponding to the functions of "extracellular matrix", "cytoskeleton", and "oxidoreductase activity", indicating the induced cellular response to cold shock. The "cell cycle/division" and "DNA complex" functions were enriched in the downregulated CRMs for all four tissues, which comprised inhibited DEGs, suggesting that even with tissue-specific responses, cold shock may induce severely disrupted cellular functions in all tissues, reducing aquaculture productivity. Therefore, our results revealed the tissue-specific regulation of the cellular response to low-temperature stress, which warrants further investigation and provides more comprehensive insights for the conservation and cultivation of P. olivaceus in cold water.
Collapse
Affiliation(s)
- Jiayi He
- Key Laboratory of Marine Genetics and Breeding (Ocean University of China), Ministry of Education, 5 Yushan Road, Qingdao 266003, China
| | - Qing Zhu
- Key Laboratory of Marine Genetics and Breeding (Ocean University of China), Ministry of Education, 5 Yushan Road, Qingdao 266003, China
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya 572024, China
| | - Ping Han
- Key Laboratory of Marine Genetics and Breeding (Ocean University of China), Ministry of Education, 5 Yushan Road, Qingdao 266003, China
| | - Tianyu Zhou
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya 572024, China
| | - Juyan Li
- Key Laboratory of Marine Genetics and Breeding (Ocean University of China), Ministry of Education, 5 Yushan Road, Qingdao 266003, China
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya 572024, China
| | - Xubo Wang
- Key Laboratory of Aquacultural Biotechnology (Ningbo University), Ministry of Education, 169 Qixingnan Road, Ningbo 315832, China
| | - Jie Cheng
- Key Laboratory of Marine Genetics and Breeding (Ocean University of China), Ministry of Education, 5 Yushan Road, Qingdao 266003, China
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya 572024, China
- Laboratory for Marine Fisheries Science and Food Production Processes, National Laboratory for Marine Science and Technology (Qingdao), 1 Wenhai Road, Qingdao 266237, China
| |
Collapse
|
7
|
Garufi A, D'Orazi V, Pistritto G, Cirone M, D'Orazi G. The Sweet Side of HIPK2. Cancers (Basel) 2023; 15:2678. [PMID: 37345014 DOI: 10.3390/cancers15102678] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/04/2023] [Accepted: 05/08/2023] [Indexed: 06/23/2023] Open
Abstract
HIPK2 is an evolutionary conserved protein kinase which modulates many molecular pathways involved in cellular functions such as apoptosis, DNA damage response, protein stability, and protein transcription. HIPK2 plays a key role in the cancer cell response to cytotoxic drugs as its deregulation impairs drug-induced cancer cell death. HIPK2 has also been involved in regulating fibrosis, angiogenesis, and neurological diseases. Recently, hyperglycemia was found to positively and/or negatively regulate HIPK2 activity, affecting not only cancer cell response to chemotherapy but also the progression of some diabetes complications. The present review will discuss how HIPK2 may be influenced by the high glucose (HG) metabolic condition and the consequences of such regulation in medical conditions.
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
|
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
|
9
|
NRF2 in Cancer: Cross-Talk with Oncogenic Pathways and Involvement in Gammaherpesvirus-Driven Carcinogenesis. Int J Mol Sci 2022; 24:ijms24010595. [PMID: 36614036 PMCID: PMC9820659 DOI: 10.3390/ijms24010595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/26/2022] [Accepted: 12/27/2022] [Indexed: 12/31/2022] Open
Abstract
Expanding knowledge of the molecular mechanisms at the basis of tumor development, especially the cross-talk between oncogenic pathways, will possibly lead to better tailoring of anticancer therapies. Nuclear factor erythroid 2-related factor 2 (NRF2) plays a central role in cancer progression, not only because of its antioxidant activity but also because it establishes cross-talk with several oncogenic pathways, including Heat Shock Factor1 (HSF1), mammalian target of rapamycin (mTOR), and mutant (mut) p53. Moreover, the involvement of NRF2 in gammaherpesvirus-driven carcinogenesis is particularly interesting. These viruses indeed hijack the NRF2 pathway to sustain the survival of tumor cells in which they establish a latent infection and to avoid a too-high increase of reactive oxygen species (ROS) when these cancer cells undergo treatments that induce viral replication. Interestingly, NRF2 activation may prevent gammaherpesvirus-driven oncogenic transformation, highlighting how manipulating the NRF2 pathway in the different phases of gammaherpesvirus-mediated carcinogenesis may lead to different outcomes. This review will highlight the mechanistic interplay between NRF2 and some oncogenic pathways and its involvement in gammaherpesviruses biology to recapitulate published evidence useful for potential application in cancer therapy.
Collapse
|
10
|
Gao Y, Xiao X, Luo J, Wang J, Peng Q, Zhao J, Jiang N, Zhao Y. E3 Ubiquitin Ligase FBXO3 Drives Neuroinflammation to Aggravate Cerebral Ischemia/Reperfusion Injury. Int J Mol Sci 2022; 23:13648. [PMID: 36362432 PMCID: PMC9658360 DOI: 10.3390/ijms232113648] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 10/27/2022] [Accepted: 11/02/2022] [Indexed: 08/04/2023] Open
Abstract
Ischemic stroke, one of the most universal causes of human mortality and morbidity, is pathologically characterized by inflammatory cascade, especially during the progression of ischemia/reperfusion (I/R) injury. F-Box Protein 3 (FBXO3), a substrate-recognition subunit of SKP1-cullin 1-F-box protein (SCF) E3 ligase complexes, has recently been proven to be severed as an underlying pro-inflammatory factor in pathological processes of diverse diseases. Given these considerations, the current study aims at investigating whether FBXO3 exerts impacts on inflammation in cerebral I/R injury. In this study, first, it was verified that FBXO3 protein expression increased after a middle cerebral artery occlusion/reperfusion (MCAO/R) model in Sprague-Dawley (SD) rats and was specifically expressed in neurons other than microglia or astrocytes. Meanwhile, in mouse hippocampal neuronal cell line HT22 cells, the elevation of FBXO3 protein was observed after oxygen and glucose deprivation/reoxygenation (OGD/R) treatment. It was also found that interference of FBXO3 with siRNA significantly alleviated neuronal damage via inhibiting the inflammatory response in I/R injury both in vivo and in vitro. The FBXO3 inhibitor BC-1215 was used to confirm the pro-inflammatory effect of FBXO3 in the OGD/R model as well. Furthermore, by administration of FBXO3 siRNA and BC-1215, FBXO3 was verified to reduce the protein level of Homeodomain-Interacting Protein Kinase 2 (HIPK2), likely through the ubiquitin-proteasome system (UPS), to aggravate cerebral I/R injury. Collectively, our results underline the detrimental effect FBXO3 has on cerebral I/R injury by accelerating inflammatory response, possibly through ubiquitylating and degrading HIPK2. Despite the specific interaction between FBXO3 and HIPK2 requiring further study, we believe that our data suggest the therapeutic relevance of FBXO3 to ischemic stroke and provide a new perspective on the mechanism of I/R injury.
Collapse
Affiliation(s)
- Yu Gao
- Department of Pathology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing 400016, China
| | - Xinyu Xiao
- School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, China
| | - Jing Luo
- Department of Pathology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing 400016, China
| | - Jianwei Wang
- School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, China
| | - Qiling Peng
- School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, China
| | - Jing Zhao
- Institute of Neuroscience, Chongqing Medical University, Chongqing 400016, China
- Department of Pathophysiology, Chongqing Medical University, Chongqing 400016, China
| | - Ning Jiang
- Department of Pathology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing 400016, China
| | - Yong Zhao
- Department of Pathology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
11
|
Li X, Li W, Zhang Z, Wang W, Huang H. SIRT6 overexpression retards renal interstitial fibrosis through targeting HIPK2 in chronic kidney disease. Front Pharmacol 2022; 13:1007168. [PMID: 36172184 PMCID: PMC9510922 DOI: 10.3389/fphar.2022.1007168] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 08/22/2022] [Indexed: 11/27/2022] Open
Abstract
Introduction: Renal interstitial fibrosis is a common pathophysiological change in the chronic kidney disease (CKD). Nicotinamide adenine dinucleotide (NAD)-dependent deacetylase sirtuin 6 (SIRT6) is demonstrated to protect against kidney injury. Vitamin B3 is the mostly used form of NAD precursors. However, the role of SIRT6 overexpression in renal interstitial fibrosis of CKD and the association between dietary vitamin B3 intake and renal function remain to be elucidated. Methods: Wild-type (WT) and SIRT6-transgene (SIRT6-Tg) mice were given with high-adenine diets to establish CKD model. HK2 cells were exposed to transforming growth factor β1 (TGF-β1) in vitro to explore related mechanism. Population data from Multi-Ethnic Study of Atherosclerosis (MESA) was used to examine the association between dietary vitamin B3 intake and renal function decline. Results: Compared to WT mice, SIRT6-Tg mice exhibited alleviated renal interstitial fibrosis as evidenced by reduced collagen deposit, collagen I and α-smooth muscle actin expression. Renal function was also improved in SIRT6-Tg mice. Homeodomain interacting protein kinase 2 (HIPK2) was induced during the fibrogenesis in CKD, while HIPK2 was downregulated after SIRT6 overexpression. Further assay in vitro confirmed that SIRT6 depletion exacerbated epithelial-to-mesenchymal transition of HK2 cells, which might be linked with HIPK2 upregulation. HIPK2 was inhibited by SIRT6 in the post-transcriptional level. Population study indicated that higher dietary vitamin B3 intake was independently correlated with a lower risk of estimate glomerular filtration rate decline in those ≥65 years old during follow-up. Conclusion: SIRT6/HIPK2 axis serves as a promising target of renal interstitial fibrosis in CKD. Dietary vitamin B3 intake is beneficial for renal function in the old people.
Collapse
Affiliation(s)
- Xiaoxue Li
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Wenxin Li
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Zhengzhipeng Zhang
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Weidong Wang
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Hui Huang
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
- *Correspondence: Hui Huang,
| |
Collapse
|
12
|
Atoum MF, Alajlouni MM, Alzoughool F. A Case-Control Study of the Luteinizing Hormone Level in Luteinizing Hormone Receptor Gene (rs2293275) Polymorphism in Polycystic Ovarian Syndrome Females. Public Health Genomics 2022; 25:1-9. [PMID: 35350019 DOI: 10.1159/000521971] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 01/11/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a common heterogeneous disorder characterized by chronic anovulation, infertility, polycystic ovaries, and hyperandrogenic signs. OBJECTIVE The aim of this study was to determine the association of luteinizing hormone/chorionic gonadotropin hormone receptor LHCGR polymorphism (rs2293275) with oligomenorrhea, amenorrhea, hirsutism, acne, infertility, LH, LH/FSH ratio, and body mass index (BMI) among PCOS females. METHODS This genetic case-control study recruited 55 PCOS and 55 control females, diagnosed based on the Rotterdam criteria. LH and FSH were measured by the Roche cobas c 502 automated analyzer. Genotypic analysis was carried out using the polymerase chain reaction-restriction fragment length polymorphism and restriction endonuclease digestion. RESULTS BMI was higher for PCOS patients (28.5 ± 6.59) compared to controls (25.1 ± 5.77), and ovulatory dysfunction was seen among 90% of PCOS females. Oligomenorrhea was common in PCOS (73%), and hirsutism and acne were detected in PCOS (80% and 40%; respectively). LH ≥10 were recoded among 51%, while LH/FSH ≥1.5 was recorded among 33% PCOS females. There is a statistical difference between rs2293275 polymorphism in the AG genotype between PCOS patients and controls. PCOS patients have a significantly higher mean LH level compared to controls (8.36 ± 4.86 and 5.67 ± 2.51, respectively) and showed higher LH/FSH value (1.46 ± 0.81) compared to (0.87 ± 0.30) controls. GG and AG genotypes of LHCGR showed statistically significant higher LH (8.22 ± 4.11; 9.02 ± 3.87) and LH/FSH values (1.57 ± 0.56; 1.64 ± 0.89) compared to controls. CONCLUSION LHCGR (rs2293275) GA and GG genetic variants could modulate the hormonal levels of PCOS LH levels and the LH/FSH ratio and associated with hirsutism, oligomenorrhea, BMI, and LH/FSH ratio as risk factors.
Collapse
Affiliation(s)
- Manar Fayiz Atoum
- Department of Medical Laboratory Sciences, Applied Medical Sciences, The Hashemite University, Zarqa, Jordan
| | | | - Foad Alzoughool
- Applied Medical Sciences, The Hashemite University, Zarqa, Jordan
- Faculty of Health Sciences, Fujairah Women's College, Higher Colleges of Technology, Fujairah, United Arab Emirates
| |
Collapse
|
13
|
Zhang H, Wang J, Ge Y, Ye M, Jin X. Siah1 in cancer and nervous system diseases (Review). Oncol Rep 2021; 47:35. [PMID: 34958110 DOI: 10.3892/or.2021.8246] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 09/10/2021] [Indexed: 11/06/2022] Open
Abstract
The dysregulation of the ubiquitin‑proteasome system will result in the abnormal accumulation and dysfunction of proteins, thus leading to severe diseases. Seven in absentia homolog 1 (Siah1), an E3 ubiquitin ligase, has attracted wide attention due to its varied functions in physiological and pathological conditions, and the numerous newly discovered Siah1 substrates. In cancer and nervous system diseases, the functions of Siah1 as a promoter or a suppressor of diseases are related to the change in cellular microenvironment and subcellular localization. At the same time, complex upstream regulations make Siah1 different from other E3 ubiquitin ligases. Understanding the molecular mechanism of Siah1 will help the study of various signaling pathways and benefit the therapeutic strategy of human diseases (e.g., cancer and nervous system diseases). In the present review, the functions and regulations of Siah1 are described. Moreover, novel substrates of Siah1 discovered in recent studies will be highlighted in cancer and nervous system diseases, providing ideas for future research and clinical targeted therapies using Siah1.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Oncology, The Affiliated Hospital of School of Medicine, Ningbo University, Ningbo, Zhejiang 315020, P.R. China
| | - Jie Wang
- Department of Oncology, The Affiliated Hospital of School of Medicine, Ningbo University, Ningbo, Zhejiang 315020, P.R. China
| | - Yidong Ge
- Department of Oncology, The Affiliated Hospital of School of Medicine, Ningbo University, Ningbo, Zhejiang 315020, P.R. China
| | - Meng Ye
- Department of Oncology, The Affiliated Hospital of School of Medicine, Ningbo University, Ningbo, Zhejiang 315020, P.R. China
| | - Xiaofeng Jin
- Department of Oncology, The Affiliated Hospital of School of Medicine, Ningbo University, Ningbo, Zhejiang 315020, P.R. China
| |
Collapse
|
14
|
Giuliani V, Miller MA, Liu CY, Hartono SR, Class CA, Bristow CA, Suzuki E, Sanz LA, Gao G, Gay JP, Feng N, Rose JL, Tomihara H, Daniele JR, Peoples MD, Bardenhagen JP, Geck Do MK, Chang QE, Vangamudi B, Vellano C, Ying H, Deem AK, Do KA, Genovese G, Marszalek JR, Kovacs JJ, Kim M, Fleming JB, Guccione E, Viale A, Maitra A, Emilia Di Francesco M, Yap TA, Jones P, Draetta G, Carugo A, Chedin F, Heffernan TP. PRMT1-dependent regulation of RNA metabolism and DNA damage response sustains pancreatic ductal adenocarcinoma. Nat Commun 2021; 12:4626. [PMID: 34330913 PMCID: PMC8324870 DOI: 10.1038/s41467-021-24798-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 07/05/2021] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer that has remained clinically challenging to manage. Here we employ an RNAi-based in vivo functional genomics platform to determine epigenetic vulnerabilities across a panel of patient-derived PDAC models. Through this, we identify protein arginine methyltransferase 1 (PRMT1) as a critical dependency required for PDAC maintenance. Genetic and pharmacological studies validate the role of PRMT1 in maintaining PDAC growth. Mechanistically, using proteomic and transcriptomic analyses, we demonstrate that global inhibition of asymmetric arginine methylation impairs RNA metabolism, which includes RNA splicing, alternative polyadenylation, and transcription termination. This triggers a robust downregulation of multiple pathways involved in the DNA damage response, thereby promoting genomic instability and inhibiting tumor growth. Taken together, our data support PRMT1 as a compelling target in PDAC and informs a mechanism-based translational strategy for future therapeutic development.Statement of significancePDAC is a highly lethal cancer with limited therapeutic options. This study identified and characterized PRMT1-dependent regulation of RNA metabolism and coordination of key cellular processes required for PDAC tumor growth, defining a mechanism-based translational hypothesis for PRMT1 inhibitors.
Collapse
Affiliation(s)
- Virginia Giuliani
- Traction, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Meredith A Miller
- Traction, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chiu-Yi Liu
- Traction, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stella R Hartono
- Department of Molecular and Cellular Biology and Genome Center, University of California, Davis, CA, USA
| | - Caleb A Class
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Pharmaceutical Sciences, Butler University, Indianapolis, IN, USA
| | | | - Erika Suzuki
- Traction, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lionel A Sanz
- Department of Molecular and Cellular Biology and Genome Center, University of California, Davis, CA, USA
| | - Guang Gao
- Traction, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jason P Gay
- Traction, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ningping Feng
- Traction, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Johnathon L Rose
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hideo Tomihara
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Surgery, Kindai University Nara Hospital, Nara, JP, USA
| | - Joseph R Daniele
- Traction, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael D Peoples
- Traction, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer P Bardenhagen
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mary K Geck Do
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qing E Chang
- ORBIT, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bhavatarini Vangamudi
- Traction, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Exo Therapeutics, Cambridge, MA, USA
| | - Christopher Vellano
- Traction, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Haoqiang Ying
- Department of Cellular and Molecular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Angela K Deem
- Traction, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kim-Anh Do
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Giannicola Genovese
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joseph R Marszalek
- Traction, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey J Kovacs
- Traction, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael Kim
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jason B Fleming
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Division of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Ernesto Guccione
- Department of Oncological Sciences and Pharmacological Sciences at Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrea Viale
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anirban Maitra
- Sheikh Ahmed Bin Zayed Al Nahyan Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - M Emilia Di Francesco
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Timothy A Yap
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Philip Jones
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Giulio Draetta
- Traction, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alessandro Carugo
- Traction, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Frederic Chedin
- Department of Molecular and Cellular Biology and Genome Center, University of California, Davis, CA, USA
| | - Timothy P Heffernan
- Traction, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
15
|
Zheng X, Pan Y, Chen X, Xia S, Hu Y, Zhou Y, Zhang J. Inactivation of homeodomain-interacting protein kinase 2 promotes oral squamous cell carcinoma metastasis through inhibition of P53-dependent E-cadherin expression. Cancer Sci 2020; 112:117-132. [PMID: 33063904 PMCID: PMC7780018 DOI: 10.1111/cas.14691] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 12/26/2022] Open
Abstract
Homeodomain-interacting protein kinase 2 (HIPK2), a well-known tumor suppressor, shows contradictory expression patterns in different cancers. This study was undertaken to clarify HIPK2 expression in oral squamous cell carcinoma (OSCC) and to reveal the potential mechanism of HIPK2 involvement in OSCC metastasis. Two hundred and four OSCC tissues, together with paired adjacent normal epithelia, dysplastic epithelia, and lymph node metastasis specimens, were collected to profile HIPK2 expression by immunohistochemical staining. High throughput RNA-sequencing was used to detect the dysregulated signaling pathways in HIPK2-deficient OSCC cells. Transwell assay and lymphatic metastatic orthotopic mouse model assay were undertaken to identify the effect of HIPK2 on tumor invasion. Western blotting and luciferase reporter assay were used to examine the HIPK2/P53/E-cadherin axis in OSCC. Nuclear delocalization of HIPK2 was observed during oral epithelial cancerization progression and was associated with cervical lymph node metastasis and poor outcome. Depletion of HIPK2 promoted tumor cell invasion in vitro and facilitated cervical lymph node metastasis in vivo. According to mRNA-sequencing, pathways closely related to tumor invasion were notably activated. Homeodomain-interacting protein kinase 2 was found to trigger E-cadherin expression by mediating P53, which directly targets the CDH1 (coding E-cadherin) promoter. Restoring P53 expression rescued the E-cadherin suppression induced by HIPK2 deficiency, whereas rescued cytoplasmic HIPK2 expression had no influence on the expression of E-cadherin and cell mobility. Together, nuclear delocalization of HIPK2 might serve as a valuable negative biomarker for poor prognosis of OSCC and lymph node metastasis. The depletion of HIPK2 expression promoted OSCC metastasis by suppressing the P53/E-cadherin axis, which might be a promising target for anticancer therapies.
Collapse
Affiliation(s)
- Xueqing Zheng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei_MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Oral Histopathology Department, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yuemei Pan
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei_MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Oral Histopathology Department, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Xinming Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei_MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Oral Histopathology Department, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Shu Xia
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei_MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Oral Histopathology Department, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yaying Hu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei_MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Oral Histopathology Department, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yi Zhou
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei_MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jiali Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei_MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Oral Histopathology Department, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
16
|
The Regulatory Properties of the Ccr4-Not Complex. Cells 2020; 9:cells9112379. [PMID: 33138308 PMCID: PMC7692201 DOI: 10.3390/cells9112379] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/21/2020] [Accepted: 10/26/2020] [Indexed: 12/12/2022] Open
Abstract
The mammalian Ccr4–Not complex, carbon catabolite repression 4 (Ccr4)-negative on TATA-less (Not), is a large, highly conserved, multifunctional assembly of proteins that acts at different cellular levels to regulate gene expression. In the nucleus, it is involved in the regulation of the cell cycle, chromatin modification, activation and inhibition of transcription initiation, control of transcription elongation, RNA export, nuclear RNA surveillance, and DNA damage repair. In the cytoplasm, the Ccr4–Not complex plays a central role in mRNA decay and affects protein quality control. Most of our original knowledge of the Ccr4–Not complex is derived, primarily, from studies in yeast. More recent studies have shown that the mammalian complex has a comparable structure and similar properties. In this review, we summarize the evidence for the multiple roles of both the yeast and mammalian Ccr4–Not complexes, highlighting their similarities.
Collapse
|
17
|
D'Orazi G, Garufi A, Cirone M. Nuclear factor erythroid 2 (NF-E2) p45-related factor 2 interferes with homeodomain-interacting protein kinase 2/p53 activity to impair solid tumors chemosensitivity. IUBMB Life 2020; 72:1634-1639. [PMID: 32593231 DOI: 10.1002/iub.2334] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 12/20/2022]
Abstract
Resistance to chemotherapy represents a major hurdle to successful cancer treatment. A key role for efficient response to anticancer therapies is played by TP53 oncosuppressor gene that indeed is mutated in 50% of human cancers or inactivated at protein level in the remaining 50%. Homeodomain-interacting protein kinase 2 (HIPK2) is the wild-type p53 (wtp53) apoptotic activator, and its inhibition by hypoxia or hyperglycemia may contribute to tumor chemoresistance mainly by impairing p53 apoptotic activity. Another important molecule able to induce chemoresistance is nuclear factor erythroid 2 (NF-E2) p45-related factor 2 (NRF2) transcription factor, whose activation by oxidative and/or electrophilic stress regulates a transcriptional antioxidant program allowing cancer cells to adapt and survive to stresses. NRF2 may shift from cytoprotective to tumor-promoting function, according to tumor phases. NRF2 may crosstalk with both wtp53 and mutant p53 (mutp53), inhibiting the wtp53 apoptotic function and strengthening the mutp53 oncogenic function. NRF2 has also been shown to induce HIPK2 mRNA expression cooperating in inducing cytoprotection. Although HIPK2, p53, and NRF2 have been individually extensively studied, their interplay has not been clearly addressed yet. On the basis of the background and our results, we aim at hypothesizing the unexpected pro-survival activity played by the NRF2/HIPK2/p53 interplay that can be hijacked by cancer cells to bypass drugs cytotoxicity.
Collapse
Affiliation(s)
- Gabriella D'Orazi
- Department of Research, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Alessia Garufi
- Department of Research, IRCCS Regina Elena National Cancer Institute, Rome, Italy.,Department of Medical Sciences, University 'G. d'Annunzio', Chieti, Italy
| | - Mara Cirone
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.,Laboratory affiliated to Pasteur Institute, Italy-Foundation Cenci Bolognetti, Rome, Italy
| |
Collapse
|
18
|
An Alternative Splice Variant of HIPK2 with Intron Retention Contributes to Cytokinesis. Cells 2020; 9:cells9020484. [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] [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
|
19
|
Sung KS, Kim SJ, Cho SW, Park YJ, Tae K, Choi CY. Functional impairment of the HIPK2 small ubiquitin-like modifier (SUMO)-interacting motif in acute myeloid leukemia. Am J Cancer Res 2019; 9:94-107. [PMID: 30755814 PMCID: PMC6356924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 12/02/2018] [Indexed: 06/09/2023] Open
Abstract
Covalent conjugations of the SUMO-1 moiety on a target protein play important roles in the regulation of cellular protein function. SUMO-conjugation of PML is a regulatory step for PML nuclear body (PML-NB) formation, and HIPK2 is SUMO-conjugated and recruited into the PML-NBs. Although HIPK2 mutations (R861W and N951I) were found in acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) patients, little is known about the underlying mechanisms by which HIPK2 mutations are associated with the pathogenesis of leukemia. Here we show that HIPK2 mutants found in AML and MDS patients are defective in SUMO-interacting motif (SIM) function. Due to defective SIM function, the HIPK2 mutants were not modified with SUMO-1, and not recruited to the PML-NBs. However, the HIPK2 mutants can normally bind to and phosphorylate AML1b. Therefore, the HIPK2 mutants can sequestrate the AML1 complex out of the PML-NBs, resulting in the disruption of AML1-mediated activation of target genes for myeloid differentiation. In addition, the differentiation of K562 blast cells was impaired by the expression of the HIPK2 SIM-defective mutants. These results suggest that HIPK2 targeting into the PML-NBs via the SIMs is crucial for HIPK2-mediated induction of myeloid differentiation, and is associated with AML pathogenesis.
Collapse
Affiliation(s)
- Ki Sa Sung
- Department of Biological Sciences, Sungkyunkwan UniversitySuwon 16419, Republic of Korea
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew York, New York 10029, United States
| | - Sun-Jick Kim
- Department of Biological Sciences, Sungkyunkwan UniversitySuwon 16419, Republic of Korea
| | - Sang Woo Cho
- Department of Biological Sciences, Sungkyunkwan UniversitySuwon 16419, Republic of Korea
| | - Ye-Jin Park
- Department of Biological Sciences, Sungkyunkwan UniversitySuwon 16419, Republic of Korea
| | - Kun Tae
- Department of Biological Sciences, Sungkyunkwan UniversitySuwon 16419, Republic of Korea
| | - Cheol Yong Choi
- Department of Biological Sciences, Sungkyunkwan UniversitySuwon 16419, Republic of Korea
| |
Collapse
|
20
|
Xu X, Wang J, Yang R, Dong Z, Zhang D. Genetic or pharmacologic inhibition of EGFR ameliorates sepsis-induced AKI. Oncotarget 2017; 8:91577-91592. [PMID: 29207668 PMCID: PMC5710948 DOI: 10.18632/oncotarget.21244] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 08/06/2017] [Indexed: 12/19/2022] Open
Abstract
Despite recent studies have demonstrated that the EGF receptor (EGFR) activation provided a renoprotective role during ischemic and folic acid-induced AKI, the role and regulation mechanism of EGFR in septic AKI remains unclear. Here, gefitinib, a highly selective EGFR inhibitor, abrogated LPS-induced phosphorylation of EGFR, ERK1/2, and STAT3 as well as expression of COX, eNOS, and proinflammatory cytokines in HK-2 cells. In addition, c-Src is an upstream of EGFR signaling pathway and mediates LPS-induced EGFR transactivation. In vivo, either gefitinib or genetic approaches (Wave-2 mutant mice, which have reduced EGFR tyrosine kinase activity) protected against LPS or cecal ligation and puncture (CLP) induced AKI respectively. Interestingly, the beneficial effects of gefitinib or genetic approaches were accompanied by the dephosphorylation of EGFR, ERK1/2, and STAT3, the down regulation of expression of COX, eNOS, macrophage infiltration, proinflammatory cytokines production and the renal cell apoptosis. Furthermore, mRNA array results indicated that gene families involved in cell death, inflammation, proliferation and signal transduction were down regulated in Wave-2 (Wa-2) mice. Take together, these data suggest that EGFR may mediate renal injury by promoting production of inflammatory factors and cell apoptosis. Inhibition of EGFR may have therapeutic potential for AKI during endotoxemia.
Collapse
Affiliation(s)
- Xuan Xu
- Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.,Emergency Medicine and Difficult Diseases Institute, Central South University, Changsha, Hunan, People's Republic of China.,Department of Emergency Medicine, Second Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Juan Wang
- Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.,Emergency Medicine and Difficult Diseases Institute, Central South University, Changsha, Hunan, People's Republic of China
| | - Ruhao Yang
- Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.,Emergency Medicine and Difficult Diseases Institute, Central South University, Changsha, Hunan, People's Republic of China
| | - Zheng Dong
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.,Department of Cellular Biology and Anatomy, Medical College of Georgia and Charlie Norwood VA Medical Center, Augusta, Georgia, USA
| | - Dongshan Zhang
- Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.,Emergency Medicine and Difficult Diseases Institute, Central South University, Changsha, Hunan, People's Republic of China.,Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
21
|
Verdina A, Di Rocco G, Virdia I, Monteonofrio L, Gatti V, Policicchio E, Bruselles A, Tartaglia M, Soddu S. HIPK2-T566 autophosphorylation diversely contributes to UV- and doxorubicin-induced HIPK2 activation. Oncotarget 2017; 8:16744-16754. [PMID: 28060750 PMCID: PMC5369998 DOI: 10.18632/oncotarget.14421] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 12/15/2016] [Indexed: 12/30/2022] Open
Abstract
HIPK2 is a Y-regulated S/T kinase involved in various cellular processes, including cell-fate decision during development and DNA damage response. Cis-autophosphorylation in the activation-loop and trans-autophosphorylation at several S/T sites along the protein are required for HIPK2 activation, subcellular localization, and subsequent posttranslational modifications. The specific function of a few of these autophosphorylations has been recently clarified; however, most of the sites found phosphorylated by mass spectrometry in human and/or mouse HIPK2 are still uncharacterized. In the process of studying HIPK2 in human colorectal cancers, we identified a mutation (T566P) in a site we previously found autophosphorylated in mouse Hipk2. Biochemical and functional characterization of this site showed that compared to wild type (wt) HIPK2, HIPK2-T566P maintains nuclear-speckle localization and has only a mild reduction in kinase and growth arresting activities upon overexpression. Next, we assessed cell response following UV-irradiation or treatment with doxorubicin, two well-known HIPK2 activators, by evaluating cell number and viability, p53-Ser46 phosphorylation, p21 induction, and caspase cleavage. Interestingly, cells expressing HIPK2-T566P mutant did not respond to UV-irradiation, while behaved similarly to wt HIPK2 upon doxorubicin-treatment. Evaluation of HIPK2-T566 phosphorylation status by a T566-phospho-specific antibody showed constitutive phosphorylation in unstressed cells, which was maintained after doxorubicin-treatment but inhibited by UV-irradiation. Taken together, these data show that HIPK2-T566 phosphorylation contributes to UV-induced HIPK2 activity but it is dispensable for doxorubicin response.
Collapse
Affiliation(s)
- Alessandra Verdina
- Unit of Cellular Networks and Molecular Therapeutic Targets, Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute - IRCCS, Rome, Italy
| | - Giuliana Di Rocco
- Unit of Cellular Networks and Molecular Therapeutic Targets, Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute - IRCCS, Rome, Italy
| | - Ilaria Virdia
- Unit of Cellular Networks and Molecular Therapeutic Targets, Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute - IRCCS, Rome, Italy
| | - Laura Monteonofrio
- Unit of Cellular Networks and Molecular Therapeutic Targets, Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute - IRCCS, Rome, Italy
| | - Veronica Gatti
- Unit of Cellular Networks and Molecular Therapeutic Targets, Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute - IRCCS, Rome, Italy.,Present address: Istituto di Biologia Cellulare e Neurobiologia, CNR, Monterotondo Scalo, Rome, Italy
| | - Eleonora Policicchio
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Alessandro Bruselles
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù - IRCCS, Rome, Italy
| | - Silvia Soddu
- Unit of Cellular Networks and Molecular Therapeutic Targets, Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute - IRCCS, Rome, Italy
| |
Collapse
|