1
|
Trelford CB, Shepherd TG. Insights into targeting LKB1 in tumorigenesis. Genes Dis 2025; 12:101402. [PMID: 39735555 PMCID: PMC11681833 DOI: 10.1016/j.gendis.2024.101402] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 06/03/2024] [Accepted: 06/22/2024] [Indexed: 12/31/2024] Open
Abstract
Genetic alterations to serine-threonine kinase 11 (STK11) have been implicated in Peutz-Jeghers syndrome and tumorigenesis. Further exploration of the context-specific roles of liver kinase B1 (LKB1; encoded by STK11) observed that it regulates AMP-activated protein kinase (AMPK) and AMPK-related kinases. Given that both migration and proliferation are enhanced with the loss of LKB1 activity combined with the prevalence of STK11 genetic alterations in cancer biopsies, LKB1 was marked as a tumor suppressor. However, the role of LKB1 in tumorigenesis is paradoxical as LKB1 activates autophagy and reactive oxygen species scavenging while dampening anoikis, which contribute to cancer cell survival. Due to the pro-tumorigenic properties of LKB1, targeting LKB1 pathways is now relevant for cancer treatment. With the recent successes of targeting LKB1 signaling in research and clinical settings, and enhanced cytotoxicity of chemical compounds in LKB1-deficient tumors, there is now a need for LKB1 inhibitors. However, validating LKB1 inhibitors is challenging as LKB1 adaptor proteins, nucleocytoplasmic shuttling, and splice variants all manipulate LKB1 activity. Furthermore, STE-20-related kinase adaptor protein (STRAD) and mouse protein 25 dictate LKB1 cellular localization and kinase activity. For these reasons, prior to assessing the efficacy and potency of pharmacological candidates, the functional status of LKB1 needs to be defined. Therefore, to improve the understanding of LKB1 in physiology and oncology, this review highlights the role of LKB1 in tumorigenesis and addresses the therapeutic relevancy of LKB1 inhibitors.
Collapse
Affiliation(s)
- Charles B. Trelford
- The Mary & John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer Program, London, ON N6A 4L6, Canada
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
| | - Trevor G. Shepherd
- The Mary & John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer Program, London, ON N6A 4L6, Canada
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
- Department of Obstetrics and Gynaecology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
| |
Collapse
|
2
|
Ma C, Lin Z, Yao J, Qin W, Wang X, Li Q, Ye Y, Liu X, Chen F, Hu J, Xu G, Tan G. Loss of USP10 promotes hepatocellular carcinoma proliferation by regulating the serine synthesis pathway through inhibition of LKB1 activity. Cancer Sci 2024; 115:3902-3914. [PMID: 39327097 PMCID: PMC11611778 DOI: 10.1111/cas.16336] [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: 04/06/2024] [Revised: 07/30/2024] [Accepted: 08/27/2024] [Indexed: 09/28/2024] Open
Abstract
Metabolic dysregulation is emerging as a critical factor in tumorigenesis, and reprogramming of serine metabolism has been identified as an essential factor in the progression of hepatocellular carcinoma (HCC). Studies have shown that LKB1 deficiency can activate mTOR to upregulate the serine synthesis pathway (SSP) and promote tumor progression. Our team discovered that ubiquitin-specific protease 10 (USP10) can inhibit HCC proliferation through mTOR, but its relationship with SSP needs further investigation. The metabolite assays revealed a significant increase in serine content in HCC tissues. Through the LKB1/mTOR/activating transcription factor 4 (ATF4) axis, loss of USP10 may increase serine biosynthesis and promote the proliferation of HCC in vitro and in vivo. Furthermore, it was found that USP10 could activate LKB1 through deubiquitination. Analyzing clinical HCC tissues revealed a positive correlation between USP10 and LKB1. Additionally, those with high expression of USP10 in HCC tissues showed a better degree of tumor differentiation and longer overall survival time. Moreover, we found increased expression of both serine and its synthase in liver tumor tissues of USP10 liver-specific KO mice. Loss of USP10 inhibits the activity of LKB1, contributing to the stimulation of the mTOR/ATF4 axis and SSP and then promoting the proliferation of HCC. This work presents a novel approach for serine-targeted treatment in HCC.
Collapse
Affiliation(s)
- Chi Ma
- Department of General SurgeryThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical Physics, Chinese Academy of SciencesDalianChina
- Liaoning Key Laboratory of Molecular Targeted Drugs in Hepatobiliary and Pancreatic CancerDalian Medical UniversityDalianChina
| | - Zhikun Lin
- Department of General SurgeryThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical Physics, Chinese Academy of SciencesDalianChina
- Liaoning Key Laboratory of Molecular Targeted Drugs in Hepatobiliary and Pancreatic CancerDalian Medical UniversityDalianChina
| | - Jiaqi Yao
- Department of AnesthesiologyThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Wangshu Qin
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical Physics, Chinese Academy of SciencesDalianChina
- Liaoning Province Key Laboratory of MetabolomicsDalianChina
| | - Xiaolin Wang
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical Physics, Chinese Academy of SciencesDalianChina
- Liaoning Province Key Laboratory of MetabolomicsDalianChina
| | - Qi Li
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical Physics, Chinese Academy of SciencesDalianChina
- Liaoning Province Key Laboratory of MetabolomicsDalianChina
| | - Yaorui Ye
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical Physics, Chinese Academy of SciencesDalianChina
- Liaoning Province Key Laboratory of MetabolomicsDalianChina
| | - Xinyu Liu
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical Physics, Chinese Academy of SciencesDalianChina
- Liaoning Province Key Laboratory of MetabolomicsDalianChina
- University of Chinese Academy of SciencesBeijingChina
| | - Fating Chen
- Department of the First Clinical CollegeDalian Medical UniversityDalianChina
| | - Jinlong Hu
- Department of the First Clinical CollegeDalian Medical UniversityDalianChina
| | - Guowang Xu
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical Physics, Chinese Academy of SciencesDalianChina
- Liaoning Province Key Laboratory of MetabolomicsDalianChina
- University of Chinese Academy of SciencesBeijingChina
| | - Guang Tan
- Department of General SurgeryThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
- Liaoning Key Laboratory of Molecular Targeted Drugs in Hepatobiliary and Pancreatic CancerDalian Medical UniversityDalianChina
| |
Collapse
|
3
|
Goleij P, Khazeei Tabari MA, Ahmed ARD, Mohamed LME, Saleh GAH, Abdu Hassan MTM, Moahmmednoor AGM, Khan H. Molecular Secrets Revealed: How Diabetes may be Paving the Way for Leukemia. Curr Treat Options Oncol 2024; 25:1563-1579. [PMID: 39585587 DOI: 10.1007/s11864-024-01281-6] [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] [Indexed: 11/26/2024]
Abstract
OPINION STATEMENT Type 2 Diabetes Mellitus (T2DM) and leukemia are two major global health concerns, both contributing significantly to morbidity and mortality. Epidemiological evidence demonstrates a strong correlation between T2DM and an increased risk of leukemia, particularly driven by insulin resistance, hyperglycemia, and the resultant metabolic dysregulation. Key shared risk factors, including obesity and chronic inflammation, create a conducive environment for leukemogenesis, intensifying cancer cell proliferation and resistance to standard therapies. Insulin resistance, in particular, triggers oncogenic pathways such as PI3K/AKT and MAPK, exacerbating the aggressive phenotype seen in leukemia patients with T2DM. Additionally, clonal hematopoiesis of indeterminate potential (CHIP) is implicated in the higher leukemia risk observed in diabetic populations, especially among the elderly. Molecular mechanisms like the insulin-like growth factor (IGF) system further highlight the intricate link between these diseases, promoting survival and proliferation of leukemia cells. The coexistence of T2DM in leukemia patients is associated with poorer prognostic outcomes, including increased susceptibility to infections, reduced survival, and greater treatment resistance. Antidiabetic agents, notably metformin and pioglitazone, show promise in enhancing chemotherapy efficacy and improving patient outcomes by targeting metabolic pathways. These results highlight the need for comprehensive treatment approaches that target both metabolic abnormalities and cancer-related mechanisms in patients suffering from both T2DM and leukemia.
Collapse
Affiliation(s)
- Pouya Goleij
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran.
- Department of Genetics, Faculty of Biology, Sana Institute of Higher Education, Sari, Iran.
| | | | | | | | | | | | | | - Haroon Khan
- Department of Pharmacy, Faculty of Chemical and Life Sciences, Abdul Wali Khan University Mardan, Mardan, 23200, Pakistan.
- Department of Pharmacy, Korea University, Sejong, 20019, South Korea.
| |
Collapse
|
4
|
Trelford CB, Shepherd TG. LKB1 biology: assessing the therapeutic relevancy of LKB1 inhibitors. Cell Commun Signal 2024; 22:310. [PMID: 38844908 PMCID: PMC11155146 DOI: 10.1186/s12964-024-01689-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024] Open
Abstract
Liver Kinase B1 (LKB1), encoded by Serine-Threonine Kinase 11 (STK11), is a master kinase that regulates cell migration, polarity, proliferation, and metabolism through downstream adenosine monophosphate-activated protein kinase (AMPK) and AMPK-related kinase signalling. Since genetic screens identified STK11 mutations in Peutz-Jeghers Syndrome, STK11 mutants have been implicated in tumourigenesis labelling it as a tumour suppressor. In support of this, several compounds reduce tumour burden through upregulating LKB1 signalling, and LKB1-AMPK agonists are cytotoxic to tumour cells. However, in certain contexts, its role in cancer is paradoxical as LKB1 promotes tumour cell survival by mediating resistance against metabolic and oxidative stressors. LKB1 deficiency has also enhanced the selectivity and cytotoxicity of several cancer therapies. Taken together, there is a need to develop LKB1-specific pharmacological compounds, but prior to developing LKB1 inhibitors, further work is needed to understand LKB1 activity and regulation. However, investigating LKB1 activity is strenuous as cell/tissue type, mutations to the LKB1 signalling pathway, STE-20-related kinase adaptor protein (STRAD) binding, Mouse protein 25-STRAD binding, splicing variants, nucleocytoplasmic shuttling, post-translational modifications, and kinase conformation impact the functional status of LKB1. For these reasons, guidelines to standardize experimental strategies to study LKB1 activity, associate proteins, spliced isoforms, post-translational modifications, and regulation are of upmost importance to the development of LKB1-specific therapies. Therefore, to assess the therapeutic relevancy of LKB1 inhibitors, this review summarizes the importance of LKB1 in cell physiology, highlights contributors to LKB1 activation, and outlines the benefits and risks associated with targeting LKB1.
Collapse
Affiliation(s)
- Charles B Trelford
- The Mary &, John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer Program, 790 Commissioners Road East, Room A4‑921, London, ON, N6A 4L6, Canada.
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.
| | - Trevor G Shepherd
- The Mary &, John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer Program, 790 Commissioners Road East, Room A4‑921, London, ON, N6A 4L6, Canada
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Obstetrics and Gynaecology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| |
Collapse
|
5
|
Proteau S, Krossa I, Husser C, Guéguinou M, Sella F, Bille K, Irondelle M, Dalmasso M, Barouillet T, Cheli Y, Pisibon C, Arrighi N, Nahon‐Estève S, Martel A, Gastaud L, Lassalle S, Mignen O, Brest P, Mazure NM, Bost F, Baillif S, Landreville S, Turcotte S, Hasson D, Carcamo S, Vandier C, Bernstein E, Yvan‐Charvet L, Levesque MP, Ballotti R, Bertolotto C, Strub T. LKB1-SIK2 loss drives uveal melanoma proliferation and hypersensitivity to SLC8A1 and ROS inhibition. EMBO Mol Med 2023; 15:e17719. [PMID: 37966164 PMCID: PMC10701601 DOI: 10.15252/emmm.202317719] [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: 03/17/2023] [Revised: 10/16/2023] [Accepted: 10/23/2023] [Indexed: 11/16/2023] Open
Abstract
Metastatic uveal melanomas are highly resistant to all existing treatments. To address this critical issue, we performed a kinome-wide CRISPR-Cas9 knockout screen, which revealed the LKB1-SIK2 module in restraining uveal melanoma tumorigenesis. Functionally, LKB1 loss enhances proliferation and survival through SIK2 inhibition and upregulation of the sodium/calcium (Na+ /Ca2+ ) exchanger SLC8A1. This signaling cascade promotes increased levels of intracellular calcium and mitochondrial reactive oxygen species, two hallmarks of cancer. We further demonstrate that combination of an SLC8A1 inhibitor and a mitochondria-targeted antioxidant promotes enhanced cell death efficacy in LKB1- and SIK2-negative uveal melanoma cells compared to control cells. Our study also identified an LKB1-loss gene signature for the survival prognostic of patients with uveal melanoma that may be also predictive of response to the therapy combination. Our data thus identify not only metabolic vulnerabilities but also new prognostic markers, thereby providing a therapeutic strategy for particular subtypes of metastatic uveal melanoma.
Collapse
Affiliation(s)
- Sarah Proteau
- University Côte d'AzurNiceFrance
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020, and Equipe labellisée ARC 2022, Mediterranean Centre for Molecular MedicineNiceFrance
| | - Imène Krossa
- University Côte d'AzurNiceFrance
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020, and Equipe labellisée ARC 2022, Mediterranean Centre for Molecular MedicineNiceFrance
| | - Chrystel Husser
- University Côte d'AzurNiceFrance
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020, and Equipe labellisée ARC 2022, Mediterranean Centre for Molecular MedicineNiceFrance
| | | | - Federica Sella
- Department of Dermatology, University Hospital ZurichUniversity of ZurichZurichSwitzerland
| | - Karine Bille
- University Côte d'AzurNiceFrance
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020, and Equipe labellisée ARC 2022, Mediterranean Centre for Molecular MedicineNiceFrance
| | | | - Mélanie Dalmasso
- University Côte d'AzurNiceFrance
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020, and Equipe labellisée ARC 2022, Mediterranean Centre for Molecular MedicineNiceFrance
| | - Thibault Barouillet
- Inserm, Hematometabolism and metainflammation, team 13, Mediterranean Centre for Molecular MedicineNiceFrance
| | - Yann Cheli
- University Côte d'AzurNiceFrance
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020, and Equipe labellisée ARC 2022, Mediterranean Centre for Molecular MedicineNiceFrance
| | - Céline Pisibon
- University Côte d'AzurNiceFrance
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020, and Equipe labellisée ARC 2022, Mediterranean Centre for Molecular MedicineNiceFrance
| | - Nicole Arrighi
- University Côte d'AzurNiceFrance
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020, and Equipe labellisée ARC 2022, Mediterranean Centre for Molecular MedicineNiceFrance
| | - Sacha Nahon‐Estève
- University Côte d'AzurNiceFrance
- Department of OphthalmologyCentre Hospitalier Universitaire of NiceNiceFrance
| | - Arnaud Martel
- University Côte d'AzurNiceFrance
- Department of OphthalmologyCentre Hospitalier Universitaire of NiceNiceFrance
| | | | - Sandra Lassalle
- University Côte d'AzurNiceFrance
- Laboratory of Clinical and Experimental Pathology, University Hospital of Nice, FHU OncoAge, Cote d'Azur University, Biobank BB‐0033‐00025, IRCAN team 4, OncoAge FHUNiceFrance
| | | | - Patrick Brest
- University Côte d'AzurNiceFrance
- IRCAN team 4, Inserm, CNRS, FHU‐oncoAge, IHU‐RESPIRera NiceNiceFrance
| | - Nathalie M Mazure
- University Côte d'AzurNiceFrance
- Inserm, Cancer, Metabolism and environment, team, Equipe labellisée Ligue 2022, Mediterranean Centre for Molecular MedicineNiceFrance
| | - Frédéric Bost
- University Côte d'AzurNiceFrance
- Inserm, Cancer, Metabolism and environment, team, Equipe labellisée Ligue 2022, Mediterranean Centre for Molecular MedicineNiceFrance
| | - Stéphanie Baillif
- University Côte d'AzurNiceFrance
- Department of OphthalmologyCentre Hospitalier Universitaire of NiceNiceFrance
| | - Solange Landreville
- Département d'ophtalmologie et d'ORL‐CCF, Faculté de médecineUniversité LavalQuebec CityQCCanada
- CUO‐Recherche and Axe médecine régénératriceCentre de recherche du CHU de Québec‐Université LavalQuebec CityQCCanada
- Centre de recherche sur le cancer de l'Université LavalQuebec CityQCCanada
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEXQuebec CityQCCanada
| | - Simon Turcotte
- Cancer AxisCentre de recherche du Centre Hospitalier de l'Université de Montréal/Institut du cancer de MontréalMontréalQCCanada
- Hepato‐Pancreato‐Biliary Surgery and Liver Transplantation ServiceCentre hospitalier de l'Université de MontréalMontréalQCCanada
| | - Dan Hasson
- Department of Oncological Sciences, Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Tisch Cancer Institute Bioinformatics for Next Generation Sequencing (BiNGS) FacilityIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Saul Carcamo
- Department of Oncological Sciences, Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Tisch Cancer Institute Bioinformatics for Next Generation Sequencing (BiNGS) FacilityIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | | | - Emily Bernstein
- Department of Oncological Sciences, Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Laurent Yvan‐Charvet
- University Côte d'AzurNiceFrance
- Inserm, Hematometabolism and metainflammation, team 13, Mediterranean Centre for Molecular MedicineNiceFrance
| | - Mitchell P Levesque
- Department of Dermatology, University Hospital ZurichUniversity of ZurichZurichSwitzerland
| | - Robert Ballotti
- University Côte d'AzurNiceFrance
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020, and Equipe labellisée ARC 2022, Mediterranean Centre for Molecular MedicineNiceFrance
| | - Corine Bertolotto
- University Côte d'AzurNiceFrance
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020, and Equipe labellisée ARC 2022, Mediterranean Centre for Molecular MedicineNiceFrance
| | - Thomas Strub
- University Côte d'AzurNiceFrance
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020, and Equipe labellisée ARC 2022, Mediterranean Centre for Molecular MedicineNiceFrance
| |
Collapse
|
6
|
Li H, Zou F, Zhang J, Zhu S, Chu K, Zhang X, Zhao T. YAP suppresses human T-cell leukemia virus type 1 transcription. J Med Virol 2023; 95:e29065. [PMID: 37661566 DOI: 10.1002/jmv.29065] [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: 07/17/2023] [Revised: 08/11/2023] [Accepted: 08/17/2023] [Indexed: 09/05/2023]
Abstract
Human T-cell leukemia virus type 1 (HTLV-1) is an oncogenic retrovirus that causes adult T-cell leukemia/lymphoma (ATL). HTLV-1 encodes Tax protein that activates transcription from viral long terminal repeats (LTR). Multiple cofactors are involved in the regulation of HTLV-1 transcription via association with Tax. Yes-associated protein (YAP), which is the key effector of Hippo pathway, is elevated and activated in ATL cells. In this study, we reported that YAP protein suppressed Tax activation of HTLV-1 5' LTR but not 3' LTR. The activation of the 5' LTR by Tax was potentiated when YAP was depleted. Moreover, overexpression of YAP repressed HTLV-1 plus-strand viral gene expression and virion production, whereas compromising YAP by RNA inference augmented the expression of HTLV-1 protein. As mechanisms of YAP-mediated viral transcription inhibition, we found that YAP interacted with Tax, and prevented the association between Tax and p300. It finally led to the inhibition of recruitment of Tax to the Tax-responsive element in the 5' LTR of HTLV-1. Taken together, our results demonstrate the negative regulatory function of YAP in Tax activation of HTLV-1 transcription. It may achieve sufficient transcriptional repression to maintain persistent infection and long-term latency of HTLV-1 in the host cells.
Collapse
Affiliation(s)
- Hengbo Li
- School of Medicine, Hangzhou City University, Hangzhou, China
- College of Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Feng Zou
- College of Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Jie Zhang
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Shengyu Zhu
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Kaifei Chu
- School of Medicine, Hangzhou City University, Hangzhou, China
- College of Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Xu Zhang
- School of Medicine, Hangzhou City University, Hangzhou, China
- College of Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Tiejun Zhao
- School of Medicine, Hangzhou City University, Hangzhou, China
- College of Life Sciences, Zhejiang Normal University, Jinhua, China
| |
Collapse
|
7
|
Zhang Y, Zhou F, Guan J, Zhou L, Chen B. Action Mechanism of Metformin and Its Application in Hematological Malignancy Treatments: A Review. Biomolecules 2023; 13:250. [PMID: 36830619 PMCID: PMC9953052 DOI: 10.3390/biom13020250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 02/03/2023] Open
Abstract
Hematologic malignancies (HMs) mainly include acute and chronic leukemia, lymphoma, myeloma and other heterogeneous tumors that seriously threaten human life and health. The common effective treatments are radiotherapy, chemotherapy and hematopoietic stem cell transplantation (HSCT), which have limited options and are prone to tumor recurrence and (or) drug resistance. Metformin is the first-line drug for the treatment of type 2 diabetes (T2DM). Recently, studies identified the potential anti-cancer ability of metformin in both T2DM patients and patients that are non-diabetic. The latest epidemiological and preclinical studies suggested a potential benefit of metformin in the prevention and treatment of patients with HM. The mechanism may involve the activation of the adenosine monophosphate-activated protein kinase (AMPK) signaling pathway by metformin as well as other AMPK-independent pathways to exert anti-cancer properties. In addition, combining current conventional anti-cancer drugs with metformin may improve the efficacy and reduce adverse drug reactions. Therefore, metformin can also be used as an adjuvant therapeutic agent for HM. This paper highlights the anti-hyperglycemic effects and potential anti-cancer effects of metformin, and also compiles the in vitro and clinical trials of metformin as an anti-cancer and chemosensitizing agent for the treatment of HM. The need for future research on the use of metformin in the treatment of HM is indicated.
Collapse
Affiliation(s)
| | | | | | | | - Baoan Chen
- Department of Hematology and Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| |
Collapse
|
8
|
Ganesh GV, Mohanram RK. Metabolic reprogramming and immune regulation in viral diseases. Rev Med Virol 2021; 32:e2268. [PMID: 34176174 DOI: 10.1002/rmv.2268] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 06/02/2021] [Accepted: 06/10/2021] [Indexed: 12/11/2022]
Abstract
The recent outbreak and transmission of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) worldwide and the ensuing coronavirus disease 2019 (COVID-19) pandemic has left us scrambling for ways to contain the disease and develop vaccines that are safe and effective. Equally important, understanding the impact of the virus on the host system in convalescent patients, healthy otherwise or with co-morbidities, is expected to aid in developing effective strategies in the management of patients afflicted with the disease. Viruses possess the uncanny ability to redirect host metabolism to serve their needs and also limit host immune response to ensure their survival. An ever-increasingly powerful approach uses metabolomics to uncover diverse molecular signatures that influence a wide array of host signalling networks in different viral infections. This would also help integrate experimental findings from individual studies to yield robust evidence. In addition, unravelling the molecular mechanisms harnessed by both viruses and tumours in their host metabolism will help broaden the repertoire of therapeutic tools available to combat viral disease.
Collapse
Affiliation(s)
- Goutham V Ganesh
- Life Science Division, SRM Research Institute and Department of Biotechnology, School of Bioengineering, SRM Institute of Science & Technology, Kattankulathur, Tamil Nadu, India
| | - Ramkumar K Mohanram
- Life Science Division, SRM Research Institute and Department of Biotechnology, School of Bioengineering, SRM Institute of Science & Technology, Kattankulathur, Tamil Nadu, India
| |
Collapse
|
9
|
Activation of PERK-ATF4-CHOP pathway as a novel therapeutic approach for efficient elimination of HTLV-1-infected cells. Blood Adv 2021; 4:1845-1858. [PMID: 32369565 DOI: 10.1182/bloodadvances.2019001139] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 03/26/2020] [Indexed: 02/06/2023] Open
Abstract
Patients with adult T-cell leukemia (ATL) exhibit a poor prognosis and overall survival rate when treated with standard chemotherapy, highlighting the continued requirement for the development of novel safe and effective therapies for human T-cell leukemia virus type 1 (HTLV-1)-related diseases. In this study, we demonstrated that MK-2048, a second-generation HIV-1 integrase (IN) inhibitor, potently and selectively kills HTLV-1-infected cells. Differential transcriptome profiling revealed significantly elevated levels of gene expression of the unfolded protein response (UPR) PKR-like ER kinase (PERK) signaling pathway in ATL cell lines following MK-2048 treatment. We also identified a significant downregulation in glucose regulated protein 78 (GRP78), a master regulator of the UPR in the CD4+CADM1+ HTLV-1-infected cell population of primary HTLV-1 carrier peripheral blood mononuclear cells (PBMCs) (n = 9), suggesting that HTLV-1-infected cells are hypersensitive to endoplasmic reticulum (ER) stress-mediated apoptosis. MK-2048 efficiently reduced proviral loads in primary HTLV-1 carrier PBMCs (n = 4), but had no effect on the total numbers of these cells, indicating that MK-2048 does not affect the proliferation of HTLV-1-uninfected PBMCs. MK-2048 specifically activated the ER stress-related proapoptotic gene, DNA damage-inducible transcript 3 protein (DDIT3), also known as C/EBP homologous protein (CHOP), in HTLV-1-infected but not uninfected cells of HTLV-1-carrier PBMCs. Our findings demonstrated that MK-2048 selectively induces HTLV-1-infected cell apoptosis via the activation of the UPR. This novel regulatory mechanism of the HIV IN inhibitor MK-2048 in HTLV-1-infected cells provides a promising prophylactic and therapeutic target for HTLV-1-related diseases including ATL.
Collapse
|
10
|
Abstract
Human T cell leukemia virus type 1 (HTLV-1) is a horizontally transmitted virus infection of CD4+ lymphocytes which causes adult T cell leukemia-lymphoma (ATLL) and HTLV-associated myelopathy (HAM). The viral genome encodes two oncoproteins, transactivator protein (Tax) and helix basic zipper protein (HBZ), which are considered tumor initiator and maintenance factors, respectively. Tax is the primary inducer of clonal infected T cell expansion, and genetic instability. The immune response to Tax results in the selection of cells with little or no Tax expression, which have undergone genetic and epigenetic alterations that promote T cell activation, proliferation, and resistance to apoptosis. This selection of malignant cells occurs over several decades in 5% of infected individuals. Novel insights into the molecular details of each of these events has led to targeted therapies for ATLL.
Collapse
Affiliation(s)
- Lee Ratner
- Division of Oncology, Washington University School of Medicine, Box 8069, 660 S Euclid Ave, St Louis, MO 63110, United States.
| |
Collapse
|
11
|
Kuwako KI, Okano H. The LKB1-SIK Pathway Controls Dendrite Self-Avoidance in Purkinje Cells. Cell Rep 2019; 24:2808-2818.e4. [PMID: 30208308 DOI: 10.1016/j.celrep.2018.08.029] [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: 02/06/2018] [Revised: 06/12/2018] [Accepted: 08/08/2018] [Indexed: 02/08/2023] Open
Abstract
Strictly controlled dendrite patterning underlies precise neural connection. Dendrite self-avoidance is a crucial system preventing self-crossing and clumping of dendrites. Although many cell-surface molecules that regulate self-avoidance have been identified, the signaling pathway that orchestrates it remains poorly understood, particularly in mammals. Here, we demonstrate that the LKB1-SIK kinase pathway plays a pivotal role in the self-avoidance of Purkinje cell (PC) dendrites by ensuring dendritic localization of Robo2, a regulator of self-avoidance. LKB1 is activated in developing PCs, and PC-specific deletion of LKB1 severely disrupts the self-avoidance of PC dendrites without affecting gross morphology. SIK1 and SIK2, downstream kinases of LKB1, mediate LKB1-dependent dendrite self-avoidance. Furthermore, loss of LKB1 leads to significantly decreased Robo2 levels in the dendrite but not in the cell body. Finally, restoration of dendritic Robo2 level via overexpression largely rescues the self-avoidance defect in LKB1-deficient PCs. These findings reveal an LKB1-pathway-mediated developmental program that establishes dendrite self-avoidance.
Collapse
Affiliation(s)
- Ken-Ichiro Kuwako
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
12
|
Gao WW, Tang HMV, Cheng Y, Chan CP, Chan CP, Jin DY. Suppression of gluconeogenic gene transcription by SIK1-induced ubiquitination and degradation of CRTC1. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2018; 1861:211-223. [PMID: 29408765 DOI: 10.1016/j.bbagrm.2018.01.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 01/23/2018] [Accepted: 01/29/2018] [Indexed: 12/21/2022]
Abstract
CRTCs are a group of three transcriptional coactivators required for CREB-dependent transcription. CREB and CRTCs are critically involved in the regulation of various biological processes such as cell proliferation, metabolism, learning and memory. However, whether CRTC1 efficiently induces gluconeogenic gene expression and how CRTC1 is regulated by upstream kinase SIK1 remain to be understood. In this work, we demonstrated SIK1-induced phosphorylation, ubiquitination and degradation of CRTC1 in the context of the regulation of gluconeogenesis. CRTC1 protein was destabilized by SIK1 but not SIK2 or SIK3. This effect was likely mediated by phosphorylation at S155, S167, S188 and S346 residues of CRTC1 followed by K48-linked polyubiquitination and proteasomal degradation. Expression of gluconeogenic genes such as that coding for phosphoenolpyruvate carboxykinase was stimulated by CRTC1, but suppressed by SIK1. Depletion of CRTC1 protein also blocked forskolin-induced gluconeogenic gene expression, knockdown or pharmaceutical inhibition of SIK1 had the opposite effect. Finally, SIK1-induced ubiquitination of CRTC1 was mediated by RFWD2 ubiquitin ligase at a site not equivalent to K628 in CRTC2. Taken together, our work reveals a regulatory circuit in which SIK1 suppresses gluconeogenic gene transcription by inducing ubiquitination and degradation of CRTC1. Our findings have implications in the development of new antihyperglycemic agents.
Collapse
Affiliation(s)
- Wei-Wei Gao
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong; State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Hei-Man Vincent Tang
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong; State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Yun Cheng
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong; State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Ching-Ping Chan
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong; State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Chi-Ping Chan
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong; State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong.
| | - Dong-Yan Jin
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong; State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong.
| |
Collapse
|
13
|
Biondani G, Peyron JF. Metformin, an Anti-diabetic Drug to Target Leukemia. Front Endocrinol (Lausanne) 2018; 9:446. [PMID: 30147674 PMCID: PMC6095966 DOI: 10.3389/fendo.2018.00446] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 07/19/2018] [Indexed: 12/14/2022] Open
Abstract
Metformin, a widely used anti-diabetic molecule, has attracted a strong interest in the last 10 years as a possible new anti-cancer molecule. Metformin acts by interfering with mitochondrial respiration, leading to an activation of the AMPK tumor-suppressive pathway to promote catabolic-energy saving reactions and block anabolic ones that are associated with abnormal cell proliferation. Metformin also acts at the organism level. In type 2 diabetes patients, metformin reduces hyperglycemia and increases insulin sensitivity by enhancing insulin-stimulated glucose uptake in muscles, liver, and adipose tissue and by reducing glucose output by the liver. Lowering insulin and insulin-like growth factor 1 (IGF-1) levels that stimulate cancer growth could be important features of metformin's mode of action. Despite continuous progress in treatments with the use of targeted therapies and now immunotherapies, acute leukemias are still of very poor prognosis for relapse patients, demonstrating an important need for new treatments deriving from the identification of their pathological supportive mechanisms. In the last decade, it has been realized that if cancer cells modify and reprogram their metabolism to feed their intense biochemical needs associated with their runaway proliferation, they develop metabolic addictions that could represent attractive targets for new therapeutic strategies that intend to starve and kill cancer cells. This Mini Review explores the anti-leukemic potential of metformin and its mode of action on leukemia metabolism.
Collapse
|
14
|
Chan CP, Kok KH, Jin DY. Human T-Cell Leukemia Virus Type 1 Infection and Adult T-Cell Leukemia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1018:147-166. [PMID: 29052136 DOI: 10.1007/978-981-10-5765-6_9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Human T-cell leukemia virus type 1 (HTLV-1) is the first retrovirus discovered to cause adult T-cell leukemia (ATL), a highly aggressive blood cancer. HTLV-1 research in the past 35 years has been most revealing in the mechanisms of viral oncogenesis. HTLV-1 establishes a lifelong persistent infection in CD4+ T lymphocytes. The infection outcome is governed by host immunity. ATL develops in 2-5% of infected individuals 30-50 years after initial exposure. HTLV-1 encodes two oncoproteins Tax and HBZ, which are required for initiation of cellular transformation and maintenance of cell proliferation, respectively. HTLV-1 oncogenesis is driven by a clonal selection and expansion process during which both host and viral factors cooperate to impair genome stability, immune surveillance, and other mechanisms of tumor suppression. A better understanding of HTLV-1 biology and leukemogenesis will reveal new strategies and modalities for ATL prevention and treatment.
Collapse
Affiliation(s)
- Chi-Ping Chan
- School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
| | - Kin-Hang Kok
- Department of Microbiology, The University of Hong Kong, 145 Pokfulam Road, Pokfulam, Hong Kong
| | - Dong-Yan Jin
- School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong.
| |
Collapse
|
15
|
Cheng Y, Gao WW, Tang HMV, Deng JJ, Wong CM, Chan CP, Jin DY. β-TrCP-mediated ubiquitination and degradation of liver-enriched transcription factor CREB-H. Sci Rep 2016; 6:23938. [PMID: 27029215 PMCID: PMC4814919 DOI: 10.1038/srep23938] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 03/16/2016] [Indexed: 12/13/2022] Open
Abstract
CREB-H is an endoplasmic reticulum-resident bZIP transcription factor which critically regulates lipid homeostasis and gluconeogenesis in the liver. CREB-H is proteolytically activated by regulated intramembrane proteolysis to generate a C-terminally truncated form known as CREB-H-ΔTC, which translocates to the nucleus to activate target gene expression. CREB-H-ΔTC is a fast turnover protein but the mechanism governing its destruction was not well understood. In this study, we report on β-TrCP-dependent ubiquitination and proteasomal degradation of CREB-H-ΔTC. The degradation of CREB-H-ΔTC was mediated by lysine 48-linked polyubiquitination and could be inhibited by proteasome inhibitor. CREB-H-ΔTC physically interacted with β-TrCP, a substrate recognition subunit of the SCFβ-TrCP E3 ubiquitin ligase. Forced expression of β-TrCP increased the polyubiquitination and decreased the stability of CREB-H-ΔTC, whereas knockdown of β-TrCP had the opposite effect. An evolutionarily conserved sequence, SDSGIS, was identified in CREB-H-ΔTC, which functioned as the β-TrCP-binding motif. CREB-H-ΔTC lacking this motif was stabilized and resistant to β-TrCP-induced polyubiquitination. This motif was a phosphodegron and its phosphorylation was required for β-TrCP recognition. Furthermore, two inhibitory phosphorylation sites close to the phosphodegron were identified. Taken together, our work revealed a new intracellular signaling pathway that controls ubiquitination and degradation of the active form of CREB-H transcription factor.
Collapse
Affiliation(s)
- Yun Cheng
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong.,State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Wei-Wei Gao
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong.,State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Hei-Man Vincent Tang
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong.,State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong.,Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen, China
| | - Jian-Jun Deng
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong.,State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong.,Department of Food Science and Engineering, College of Chemical Engineering, Northwestern University, Xi'an 710069, China
| | - Chi-Ming Wong
- Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen, China.,Department of Medicine and State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Pokfulam, Hong Kong
| | - Chi-Ping Chan
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong.,State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong.,Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen, China
| | - Dong-Yan Jin
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong.,State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong.,Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen, China
| |
Collapse
|
16
|
Suppression of Type I Interferon Production by Human T-Cell Leukemia Virus Type 1 Oncoprotein Tax through Inhibition of IRF3 Phosphorylation. J Virol 2016; 90:3902-3912. [PMID: 26819312 DOI: 10.1128/jvi.00129-16] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 01/24/2016] [Indexed: 12/21/2022] Open
Abstract
UNLABELLED Infection with human T-cell leukemia virus type 1 (HTLV-1) is associated with adult T-cell leukemia (ATL) and tropical spastic paraparesis. Type I interferons (IFNs) are key effectors of the innate antiviral response, and IFN-α combined with the nucleoside reverse transcriptase inhibitor zidovudine is considered the standard first-line therapy for ATL. HTLV-1 oncoprotein Tax is known to suppress innate IFN production and response but the underlying mechanisms remain to be fully established. In this study, we report on the suppression of type I IFN production by HTLV-1 Tax through interaction with and inhibition of TBK1 kinase that phosphorylates IRF3. Induced transcription of IFN-β was severely impaired in HTLV-1-transformed ATL cells and freshly infected T lymphocytes. The ability to suppress IRF3 activation was ascribed to Tax. The expression of Tax alone sufficiently repressed the induction of IFN production by RIG-I plus PACT, cGAMP synthase plus STING, TBK1, IKKε, IRF3, and IRF7, but not by IRF3-5D, a dominant-active phosphomimetic mutant. This suggests that Tax perturbs IFN production at the step of IRF3 phosphorylation. Tax mutants deficient for CREB or NF-κB activation were fully competent in the suppression of IFN production. Coimmunoprecipitation experiments confirmed the association of Tax with TBK1, IKKε, STING, and IRF3.In vitrokinase assay indicated an inhibitory effect of Tax on TBK1-mediated phosphorylation of IRF3. Taken together, our findings suggested a new mechanism by which HTLV-1 oncoprotein Tax circumvents the production of type I IFNs in infected cells. Our findings have implications in therapeutic intervention of ATL. IMPORTANCE Human T-cell leukemia virus type 1 (HTLV-1) is the cause of adult T-cell leukemia (ATL), an aggressive and fatal blood cancer, as well as another chronic disabling disease of the spinal cord. Treatments are unsatisfactory, and options are limited. A combination of antiviral cellular protein alpha interferon and zidovudine, which is an inhibitor of a viral enzyme called reverse transcriptase, has been recommended as the standard first-line therapy for ATL. Exactly how HTLV-1 interacts with the cellular machinery for interferon production and action is not well understood. Our work sheds light on the mechanism of action for the inhibition of interferon production by an HTLV-1 oncogenic protein called Tax. Our findings might help to improve interferon-based anti-HTLV-1 and anti-ATL therapy.
Collapse
|
17
|
Chaudhary V, Zhang S, Yuen KS, Li C, Lui PY, Fung SY, Wang PH, Chan CP, Li D, Kok KH, Liang M, Jin DY. Suppression of type I and type III IFN signalling by NSs protein of severe fever with thrombocytopenia syndrome virus through inhibition of STAT1 phosphorylation and activation. J Gen Virol 2015; 96:3204-3211. [PMID: 26353965 DOI: 10.1099/jgv.0.000280] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Severe fever with thrombocytopenia syndrome virus (SFTSV) is an emerging tick-borne pathogen causing significant morbidity and mortality in Asia. NSs protein of SFTSV is known to perturb type I IFN induction and signalling, but the mechanism remains to be fully understood. Here, we showed the suppression of both type I and type III IFN signalling by SFTSV NSs protein is mediated through inhibition of STAT1 phosphorylation and activation. Infection with live SFTSV or expression of NSs potently suppressed IFN-stimulated genes but not NFkB activation. NSs was capable of counteracting the activity of IFN-α1, IFN-β, IFN-λ1 and IFN-λ2. Mechanistically, NSs associated with STAT1 and STAT2, mitigated IFN-β-induced phosphorylation of STAT1 at S727, and reduced the expression and activity of STAT1 protein in IFN-β-treated cells, resulting in the inhibition of STAT1 and STAT2 recruitment to IFNstimulated promoters. Taken together, SFTSV NSs protein is an IFN antagonist that suppresses phosphorylation and activation of STAT1.
Collapse
Affiliation(s)
- Vidyanath Chaudhary
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Shuo Zhang
- Key Laboratory for Medical Virology and National Institute for Viral Disease Control and Prevention, Chinese Centre for Disease Control and Prevention, Beijing 102206, PR China
| | - Kit-San Yuen
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Chuan Li
- Key Laboratory for Medical Virology and National Institute for Viral Disease Control and Prevention, Chinese Centre for Disease Control and Prevention, Beijing 102206, PR China
| | - Pak-Yin Lui
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Sin-Yee Fung
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Pei-Hui Wang
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Chi-Ping Chan
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Dexin Li
- Key Laboratory for Medical Virology and National Institute for Viral Disease Control and Prevention, Chinese Centre for Disease Control and Prevention, Beijing 102206, PR China
| | - Kin-Hang Kok
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong
| | - Mifang Liang
- Key Laboratory for Medical Virology and National Institute for Viral Disease Control and Prevention, Chinese Centre for Disease Control and Prevention, Beijing 102206, PR China
| | - Dong-Yan Jin
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
18
|
SIRT1 Suppresses Human T-Cell Leukemia Virus Type 1 Transcription. J Virol 2015; 89:8623-31. [PMID: 26063426 DOI: 10.1128/jvi.01229-15] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 06/02/2015] [Indexed: 12/11/2022] Open
Abstract
UNLABELLED Human T-cell leukemia virus type 1 (HTLV-1)-associated diseases are poorly treatable, and HTLV-1 vaccines are not available. High proviral load is one major risk factor for disease development. HTLV-1 encodes Tax oncoprotein, which activates transcription from viral long terminal repeats (LTR) and various types of cellular promoters. Counteracting Tax function might have prophylactic and therapeutic benefits. In this work, we report on the suppression of Tax activation of HTLV-1 LTR by SIRT1 deacetylase. The transcriptional activity of Tax on the LTR was largely ablated when SIRT1 was overexpressed, but Tax activation of NF-κB was unaffected. On the contrary, the activation of the LTR by Tax was boosted when SIRT1 was depleted. Treatment of cells with resveratrol shunted Tax activity in a SIRT1-dependent manner. The activation of SIRT1 in HTLV-1-transformed T cells by resveratrol potently inhibited HTLV-1 proviral transcription and Tax expression, whereas compromising SIRT1 by specific inhibitors augmented HTLV-1 mRNA expression. The administration of resveratrol also decreased the production of cell-free HTLV-1 virions from MT2 cells and the transmission of HTLV-1 from MT2 cells to uninfected Jurkat cells in coculture. SIRT1 associated with Tax in HTLV-1-transformed T cells. Treatment with resveratrol prevented the interaction of Tax with CREB and the recruitment of CREB, CRTC1, and p300 to Tax-responsive elements in the LTR. Our work demonstrates the negative regulatory function of SIRT1 in Tax activation of HTLV-1 transcription. Small-molecule activators of SIRT1 such as resveratrol might be considered new prophylactic and therapeutic agents in HTLV-1-associated diseases. IMPORTANCE Human T-cell leukemia virus type 1 (HTLV-1) causes a highly lethal blood cancer or a chronic debilitating disease of the spinal cord. Treatments are unsatisfactory, and vaccines are not available. Disease progression is associated with robust expression of HTLV-1 genes. Suppressing HTLV-1 gene expression might have preventive and therapeutic benefits. It is therefore critical that host factors controlling HTLV-1 gene expression be identified and characterized. This work reveals a new host factor that suppresses HTLV-1 gene expression and a natural compound that activates this suppression. Our findings not only provide new knowledge of the host control of HTLV-1 gene expression but also suggest a new strategy of using natural compounds for prevention and treatment of HTLV-1-associated diseases.
Collapse
|
19
|
Yuen KS, Chan CP, Wong NHM, Ho CH, Ho TH, Lei T, Deng W, Tsao SW, Chen H, Kok KH, Jin DY. CRISPR/Cas9-mediated genome editing of Epstein-Barr virus in human cells. J Gen Virol 2014; 96:626-636. [PMID: 25502645 DOI: 10.1099/jgv.0.000012] [Citation(s) in RCA: 129] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9 (CRISPR-associated 9) system is a highly efficient and powerful tool for RNA-guided editing of the cellular genome. Whether CRISPR/Cas9 can also cleave the genome of DNA viruses such as Epstein-Barr virus (EBV), which undergo episomal replication in human cells, remains to be established. Here, we reported on CRISPR/Cas9-mediated editing of the EBV genome in human cells. Two guide RNAs (gRNAs) were used to direct a targeted deletion of 558 bp in the promoter region of BART (BamHI A rightward transcript) which encodes viral microRNAs (miRNAs). Targeted editing was achieved in several human epithelial cell lines latently infected with EBV, including nasopharyngeal carcinoma C666-1 cells. CRISPR/Cas9-mediated editing of the EBV genome was efficient. A recombinant virus with the desired deletion was obtained after puromycin selection of cells expressing Cas9 and gRNAs. No off-target cleavage was found by deep sequencing. The loss of BART miRNA expression and activity was verified, supporting the BART promoter as the major promoter of BART RNA. Although CRISPR/Cas9-mediated editing of the multicopy episome of EBV in infected HEK293 cells was mostly incomplete, viruses could be recovered and introduced into other cells at low m.o.i. Recombinant viruses with an edited genome could be further isolated through single-cell sorting. Finally, a DsRed selectable marker was successfully introduced into the EBV genome during the course of CRISPR/Cas9-mediated editing. Taken together, our work provided not only the first genetic evidence that the BART promoter drives the expression of the BART transcript, but also a new and efficient method for targeted editing of EBV genome in human cells.
Collapse
Affiliation(s)
- Kit-San Yuen
- Department of Biochemistry, University of Hong Kong, Pokfulam, Hong Kong
| | - Chi-Ping Chan
- Department of Biochemistry, University of Hong Kong, Pokfulam, Hong Kong
| | | | - Chau-Ha Ho
- Department of Biochemistry, University of Hong Kong, Pokfulam, Hong Kong
| | - Ting-Hin Ho
- Department of Biochemistry, University of Hong Kong, Pokfulam, Hong Kong
| | - Ting Lei
- Department of Pathology, School of Medicine, Xi'an Jiaotong University, Xi'an, PR China
| | - Wen Deng
- Department of Anatomy, University of Hong Kong, Pokfulam, Hong Kong
| | - Sai Wah Tsao
- Department of Anatomy, University of Hong Kong, Pokfulam, Hong Kong
| | - Honglin Chen
- Department of Microbiology, University of Hong Kong, Pokfulam, Hong Kong
| | - Kin-Hang Kok
- Department of Microbiology, University of Hong Kong, Pokfulam, Hong Kong.,Department of Biochemistry, University of Hong Kong, Pokfulam, Hong Kong
| | - Dong-Yan Jin
- Department of Biochemistry, University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
20
|
Tang HMV, Gao WW, Chan CP, Cheng Y, Chaudhary V, Deng JJ, Yuen KS, Wong CM, Ng IOL, Kok KH, Zhou J, Jin DY. Requirement of CRTC1 coactivator for hepatitis B virus transcription. Nucleic Acids Res 2014; 42:12455-68. [PMID: 25300488 PMCID: PMC4227773 DOI: 10.1093/nar/gku925] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Transcription of hepatitis B virus (HBV) from the covalently closed circular DNA (cccDNA) template is essential for its replication. Suppressing the level and transcriptional activity of cccDNA might have anti-HBV effect. Although cellular transcription factors, such as CREB, which mediate HBV transcription, have been well described, transcriptional coactivators that facilitate this process are incompletely understood. In this study we showed that CREB-regulated transcriptional coactivator 1 (CRTC1) is required for HBV transcription and replication. The steady-state levels of CRTC1 protein were elevated in HBV-positive hepatoma cells and liver tissues. Ectopic expression of CRTC1 or its homolog CRTC2 or CRTC3 in hepatoma cells stimulated the activity of the preS2/S promoter of HBV, whereas overexpression of a dominant inactive form of CRTC1 inhibited HBV transcription. CRTC1 interacts with CREB and they are mutually required for the recruitment to the preS2/S promoter on cccDNA and for the activation of HBV transcription. Accumulation of pregenomic RNA (pgRNA) and cccDNA was observed when CRTC1 or its homologs were overexpressed, whereas the levels of pgRNA, cccDNA and secreted HBsAg were diminished when CRTC1 was compromised. In addition, HBV transactivator protein HBx stabilized CRTC1 and promoted its activity on HBV transcription. Our work reveals an essential role of CRTC1 coactivator in facilitating and supporting HBV transcription and replication.
Collapse
Affiliation(s)
- Hei-Man Vincent Tang
- Department of Biochemistry, The University of Hong Kong, Pokfulam, Hong Kong State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Wei-Wei Gao
- Department of Biochemistry, The University of Hong Kong, Pokfulam, Hong Kong State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Chi-Ping Chan
- Department of Biochemistry, The University of Hong Kong, Pokfulam, Hong Kong State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Yun Cheng
- Department of Biochemistry, The University of Hong Kong, Pokfulam, Hong Kong State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Vidyanath Chaudhary
- Department of Biochemistry, The University of Hong Kong, Pokfulam, Hong Kong State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Jian-Jun Deng
- Department of Biochemistry, The University of Hong Kong, Pokfulam, Hong Kong State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Kit-San Yuen
- Department of Biochemistry, The University of Hong Kong, Pokfulam, Hong Kong State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Chun-Ming Wong
- State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong
| | - Irene Oi-Lin Ng
- State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong
| | - Kin-Hang Kok
- Department of Biochemistry, The University of Hong Kong, Pokfulam, Hong Kong State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong
| | - Jie Zhou
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong
| | - Dong-Yan Jin
- Department of Biochemistry, The University of Hong Kong, Pokfulam, Hong Kong State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
21
|
Tang HMV, Gao WW, Chan CP, Kok KH, Jin DY. CRTC1 transcriptional coactivator is required for hepatitis B virus gene expression and replication. Cancer Metab 2014. [PMCID: PMC4072930 DOI: 10.1186/2049-3002-2-s1-p31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
22
|
Siu KL, Yeung ML, Kok KH, Yuen KS, Kew C, Lui PY, Chan CP, Tse H, Woo PCY, Yuen KY, Jin DY. Middle east respiratory syndrome coronavirus 4a protein is a double-stranded RNA-binding protein that suppresses PACT-induced activation of RIG-I and MDA5 in the innate antiviral response. J Virol 2014; 88:4866-76. [PMID: 24522921 PMCID: PMC3993821 DOI: 10.1128/jvi.03649-13] [Citation(s) in RCA: 160] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 02/07/2014] [Indexed: 12/21/2022] Open
Abstract
UNLABELLED Middle East respiratory syndrome coronavirus (MERS-CoV) is an emerging pathogen that causes severe disease in human. MERS-CoV is closely related to bat coronaviruses HKU4 and HKU5. Evasion of the innate antiviral response might contribute significantly to MERS-CoV pathogenesis, but the mechanism is poorly understood. In this study, we characterized MERS-CoV 4a protein as a novel immunosuppressive factor that antagonizes type I interferon production. MERS-CoV 4a protein contains a double-stranded RNA-binding domain capable of interacting with poly(I · C). Expression of MERS-CoV 4a protein suppressed the interferon production induced by poly(I · C) or Sendai virus. RNA binding of MERS-CoV 4a protein was required for IFN antagonism, a property shared by 4a protein of bat coronavirus HKU5 but not by the counterpart in bat coronavirus HKU4. MERS-CoV 4a protein interacted with PACT in an RNA-dependent manner but not with RIG-I or MDA5. It inhibited PACT-induced activation of RIG-I and MDA5 but did not affect the activity of downstream effectors such as RIG-I, MDA5, MAVS, TBK1, and IRF3. Taken together, our findings suggest a new mechanism through which MERS-CoV employs a viral double-stranded RNA-binding protein to circumvent the innate antiviral response by perturbing the function of cellular double-stranded RNA-binding protein PACT. PACT targeting might be a common strategy used by different viruses, including Ebola virus and herpes simplex virus 1, to counteract innate immunity. IMPORTANCE Middle East respiratory syndrome coronavirus (MERS-CoV) is an emerging and highly lethal human pathogen. Why MERS-CoV causes severe disease in human is unclear, and one possibility is that MERS-CoV is particularly efficient in counteracting host immunity, including the sensing of virus invasion. It will therefore be critical to clarify how MERS-CoV cripples the host proteins that sense viruses and to compare MERS-CoV with its ancestral viruses in bats in the counteraction of virus sensing. This work not only provides a new understanding of the abilities of MERS-CoV and closely related bat viruses to subvert virus sensing but also might prove useful in revealing new strategies for the development of vaccines and antivirals.
Collapse
Affiliation(s)
- Kam-Leung Siu
- Department of Biochemistry, University of Hong Kong, Pokfulam, Hong Kong
| | - Man Lung Yeung
- Department of Microbiology and State Key Laboratory of Emerging Infectious Diseases, University of Hong Kong, Pokfulam, Hong Kong
| | - Kin-Hang Kok
- Department of Biochemistry, University of Hong Kong, Pokfulam, Hong Kong
| | - Kit-San Yuen
- Department of Biochemistry, University of Hong Kong, Pokfulam, Hong Kong
| | - Chun Kew
- Department of Biochemistry, University of Hong Kong, Pokfulam, Hong Kong
| | - Pak-Yin Lui
- Department of Biochemistry, University of Hong Kong, Pokfulam, Hong Kong
| | - Chi-Ping Chan
- Department of Biochemistry, University of Hong Kong, Pokfulam, Hong Kong
| | - Herman Tse
- Department of Microbiology and State Key Laboratory of Emerging Infectious Diseases, University of Hong Kong, Pokfulam, Hong Kong
| | - Patrick C. Y. Woo
- Department of Microbiology and State Key Laboratory of Emerging Infectious Diseases, University of Hong Kong, Pokfulam, Hong Kong
| | - Kwok-Yung Yuen
- Department of Microbiology and State Key Laboratory of Emerging Infectious Diseases, University of Hong Kong, Pokfulam, Hong Kong
| | - Dong-Yan Jin
- Department of Biochemistry, University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
23
|
Siu KL, Chan CP, Kok KH, Chiu-Yat Woo P, Jin DY. Suppression of innate antiviral response by severe acute respiratory syndrome coronavirus M protein is mediated through the first transmembrane domain. Cell Mol Immunol 2014; 11:141-9. [PMID: 24509444 PMCID: PMC4003381 DOI: 10.1038/cmi.2013.61] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Revised: 11/13/2013] [Accepted: 11/27/2013] [Indexed: 12/16/2022] Open
Abstract
Coronaviruses have developed various measures to evade innate immunity. We have previously shown that severe acute respiratory syndrome (SARS) coronavirus M protein suppresses type I interferon (IFN) production by impeding the formation of functional TRAF3-containing complex. In this study, we demonstrate that the IFN-antagonizing activity is specific to SARS coronavirus M protein and is mediated through its first transmembrane domain (TM1) located at the N terminus. M protein from human coronavirus HKU1 does not inhibit IFN production. Whereas N-linked glycosylation of SARS coronavirus M protein has no influence on IFN antagonism, TM1 is indispensable for the suppression of IFN production. TM1 targets SARS coronavirus M protein and heterologous proteins to the Golgi apparatus, yet Golgi localization is required but not sufficient for IFN antagonism. Mechanistically, TM1 is capable of binding with RIG-I, TRAF3, TBK1 and IKKε, and preventing the interaction of TRAF3 with its downstream effectors. Our work defines the molecular architecture of SARS coronavirus M protein required for suppression of innate antiviral response.
Collapse
Affiliation(s)
- Kam-Leung Siu
- Department of Biochemistry, The University of Hong Kong, Pokfulam, Hong Kong
| | - Chi-Ping Chan
- Department of Biochemistry, The University of Hong Kong, Pokfulam, Hong Kong
| | - Kin-Hang Kok
- Department of Biochemistry, The University of Hong Kong, Pokfulam, Hong Kong
| | | | - Dong-Yan Jin
- Department of Biochemistry, The University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
24
|
Metformin: a metabolic disruptor and anti-diabetic drug to target human leukemia. Cancer Lett 2014; 346:188-96. [PMID: 24462823 DOI: 10.1016/j.canlet.2014.01.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 01/14/2014] [Accepted: 01/15/2014] [Indexed: 12/15/2022]
Abstract
There is a global and urgent need for expanding our current therapeutical arsenal against leukemia in order to improve their actual cure rates and fight relapse. Targeting the reprogrammed, altered cancer metabolism is an emerging strategy which should profoundly affect cancer cells in their intimate and irrepressible needs and addictions for nutrients uptake and incorporation into the biomass during malignant proliferation. We present here how metformin, an anti-diabetic drug that has attracted a strong interest for its recently discovered anti-cancer properties, can be envisioned as a new adjuvant approach to treat leukemia. Metformin may have a double-edged sword effect (i) by acting on the organism to decrease hyperglycaemia and hyperinsulinemia in diabetic patients and (ii) at the cellular level, by inhibiting the mTORC1-cancer supporting pathway through AMPK-dependent and independent mechanisms.
Collapse
|
25
|
Comparative analysis of the activation of unfolded protein response by spike proteins of severe acute respiratory syndrome coronavirus and human coronavirus HKU1. Cell Biosci 2014; 4:3. [PMID: 24410900 PMCID: PMC3930072 DOI: 10.1186/2045-3701-4-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 10/02/2013] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Whereas severe acute respiratory syndrome (SARS) coronavirus (SARS-CoV) is associated with severe disease, human coronavirus HKU1 (HCoV-HKU1) commonly circulates in the human populations causing generally milder illness. Spike (S) protein of SARS-CoV activates the unfolded protein response (UPR). It is not understood whether HCoV-HKU1 S protein has similar activity. In addition, the UPR-activating domain in SARS-CoV S protein remains to be identified. RESULTS In this study we compared S proteins of SARS-CoV and HCoV-HKU1 for their ability to activate the UPR. Both S proteins were found in the endoplasmic reticulum. Transmembrane serine protease TMPRSS2 catalyzed the cleavage of SARS-CoV S protein, but not the counterpart in HCoV-HKU1. Both S proteins showed a similar pattern of UPR-activating activity. Through PERK kinase they activated the transcription of UPR effector genes such as Grp78, Grp94 and CHOP. N-linked glycosylation was not required for the activation of the UPR by S proteins. S1 subunit of SARS-CoV but not its counterpart in HCoV-HKU1 was capable of activating the UPR. A central region (amino acids 201-400) of SARS-CoV S1 was required for this activity. CONCLUSIONS SARS-CoV and HCoV-HKU1 S proteins use distinct UPR-activating domains to exert the same modulatory effects on UPR signaling.
Collapse
|
26
|
Suppression of PACT-induced type I interferon production by herpes simplex virus 1 Us11 protein. J Virol 2013; 87:13141-9. [PMID: 24067967 DOI: 10.1128/jvi.02564-13] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Herpes simplex virus 1 (HSV-1) Us11 protein is a double-stranded RNA-binding protein that suppresses type I interferon production through the inhibition of the cytoplasmic RNA sensor RIG-I. Whether additional cellular mediators are involved in this suppression remains to be determined. In this study, we report on the requirement of cellular double-stranded RNA-binding protein PACT for Us11-mediated perturbation of type I interferon production. Us11 associates with PACT tightly to prevent it from binding with and activating RIG-I. The Us11-deficient HSV-1 was indistinguishable from the Us11-proficient virus in the suppression of interferon production when PACT was compromised. More importantly, HSV-1-induced activation of interferon production was abrogated in PACT knockout murine embryonic fibroblasts. Our findings suggest a new mechanism for viral evasion of innate immunity through which a viral double-stranded RNA-binding protein interacts with PACT to circumvent type I interferon production. This mechanism might also be used by other PACT-binding viral interferon-antagonizing proteins such as Ebola virus VP35 and influenza A virus NS1.
Collapse
|
27
|
Romanelli MG, Diani E, Bergamo E, Casoli C, Ciminale V, Bex F, Bertazzoni U. Highlights on distinctive structural and functional properties of HTLV Tax proteins. Front Microbiol 2013; 4:271. [PMID: 24058363 PMCID: PMC3766827 DOI: 10.3389/fmicb.2013.00271] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 08/20/2013] [Indexed: 12/15/2022] Open
Abstract
Human T cell leukemia viruses (HTLVs) are complex human retroviruses of the Deltaretrovirus genus. Four types have been identified thus far, with HTLV-1 and HTLV-2 much more prevalent than HTLV-3 or HTLV-4. HTLV-1 and HTLV-2 possess strictly related genomic structures, but differ significantly in pathogenicity, as HTLV-1 is the causative agent of adult T cell leukemia and of HTLV-associated myelopathy/tropical spastic paraparesis, whereas HTLV-2 is not associated with neoplasia. HTLVs code for a protein named Tax that is responsible for enhancing viral expression and drives cell transformation. Much effort has been invested to dissect the impact of Tax on signal transduction pathways and to identify functional differences between the HTLV Tax proteins that may explain the distinct oncogenic potential of HTLV-1 and HTLV-2. This review summarizes our current knowledge of Tax-1 and Tax-2 with emphasis on their structure, role in activation of the NF-κB (nuclear factor kappa-B) pathway, and interactions with host factors.
Collapse
|
28
|
Chan CP, Siu YT, Kok KH, Ching YP, Tang HMV, Jin DY. Group I p21-activated kinases facilitate Tax-mediated transcriptional activation of the human T-cell leukemia virus type 1 long terminal repeats. Retrovirology 2013; 10:47. [PMID: 23622267 PMCID: PMC3651266 DOI: 10.1186/1742-4690-10-47] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 04/23/2013] [Indexed: 12/12/2022] Open
Abstract
Background Human T-cell leukemia virus type 1 (HTLV-1) is the causative agent of adult T-cell leukemia and tropical spastic paraparesis. HTLV-1 encodes transactivator protein Tax that interacts with various cellular factors to modulate transcription and other biological functions. Additional cellular mediators of Tax-mediated transcriptional activation of HTLV-1 long terminal repeats (LTR) remain to be identified and characterized. Results In this study, we investigated the regulatory role of group I p21-activated kinases (Paks) in Tax-induced LTR activation. Both wild-type and kinase-dead mutants of Pak3 were capable of potentiating the activity of Tax to activate LTR transcription. The effect of Paks on the LTR was attributed to the N-terminal regulatory domain and required the action of CREB, CREB-regulating transcriptional coactivators (CRTCs) and p300/CREB-binding protein. Paks physically associated with Tax and CRTCs. Paks were recruited to the LTR in the presence of Tax. siRNAs against either Pak1 or Pak3 prevented the interaction of Tax with CRTC1 and the recruitment of Tax to the LTR. These siRNAs also inhibited LTR-dependent transcription in HTLV-1-transformed MT4 cells and in cells transfected with an infectious clone of HTLV-1. Conclusion Group I Paks augment Tax-mediated transcriptional activation of HTLV-1 LTR in a kinase-independent manner.
Collapse
Affiliation(s)
- Ching-Ping Chan
- Department of Biochemistry, The University of Hong Kong, 3/F Laboratory Block, 21 Sassoon Road, Pokfulam, Hong Kong
| | | | | | | | | | | |
Collapse
|