1
|
Roh T, Seo W, Won M, Yang WS, Sapkota A, Park EJ, Yun SH, Jeon SM, Kim KT, Lee B, Ryu G, Lee SH, Shin JM, Shin HJ, Kim YJ, Lee Y, Chung C, Song IC, Song HK, Jo EK. The inflammasome-activating poxvirus peptide IAMP29 promotes antimicrobial and anticancer responses. Exp Mol Med 2024:10.1038/s12276-024-01339-3. [PMID: 39511430 DOI: 10.1038/s12276-024-01339-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 11/15/2024] Open
Abstract
Poxviruses are implicated in a variety of infectious diseases; however, little is known about the molecular mechanisms that underlie the immune response during poxvirus infection. We investigated the function and mechanisms of the monkeypox virus envelope protein (A30L) and its core peptide (IAMP29) during the activation of innate immune responses. The A30L protein and its core peptide, IAMP29 (a 29-amino-acid inflammasome-activating peptide encompassing His40 to Asp69 of A30L), strongly activated the nucleotide-binding oligomerization domain, leucine rich repeat and pyrin domain-containing 3 (NLRP3) inflammasome by inducing the production of mitochondrial reactive oxygen species in human monocytes. Specifically, IAMP29 triggered metabolic reprogramming toward glycolysis and interacted with pyruvate kinase M isoforms (PKM1 and PKM2), thus activating the NLRP3 inflammasome and interleukin (IL)-1β production in human monocytes and murine macrophages. In human primary monocyte-derived macrophages, IAMP29-induced inflammasome activation promoted an antimicrobial response to rapidly growing non-tuberculous mycobacteria. Furthermore, IAMP29 exhibited cytotoxic activity against leukemia cells, which was mediated by pyroptosis and apoptosis. These findings provide insights into the immunological function of the poxvirus envelope peptide and suggest its therapeutic potential.
Collapse
Affiliation(s)
- Taylor Roh
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Infection Control Convergence Research Center, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Brain Korea 21 FOUR Project for Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Wonhyoung Seo
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Infection Control Convergence Research Center, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Minho Won
- Department of Biochemistry, Chungnam National University College of Natural Sciences, Daejeon, Republic of Korea
| | - Woo Seok Yang
- Department of Life Sciences, Korea University, Seoul, Republic of Korea
| | - Asmita Sapkota
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Infection Control Convergence Research Center, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Eun-Jin Park
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Infection Control Convergence Research Center, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Sung-Ho Yun
- Bio-Chemical Analysis Team, Korea Basic Science Institute, Cheongju, Republic of Korea
| | - Sang Min Jeon
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Infection Control Convergence Research Center, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Brain Korea 21 FOUR Project for Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Kyung Tae Kim
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Infection Control Convergence Research Center, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Brain Korea 21 FOUR Project for Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Bomi Lee
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Infection Control Convergence Research Center, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Brain Korea 21 FOUR Project for Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Gyoungah Ryu
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Infection Control Convergence Research Center, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Sang-Hee Lee
- Center for Research Equipment, Korea Basic Science Institute, Cheongju, Republic of Korea
| | - Jung-Min Shin
- Department of Dermatology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Hyo Jung Shin
- Department of Anatomy and Cell Biology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Brain Research Institute, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Young Jae Kim
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Infection Control Convergence Research Center, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Brain Korea 21 FOUR Project for Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Young Lee
- Department of Dermatology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Chaeuk Chung
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Ik-Chan Song
- Division of Hematology/Oncology, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Hyun Kyu Song
- Department of Life Sciences, Korea University, Seoul, Republic of Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea.
- Infection Control Convergence Research Center, Chungnam National University College of Medicine, Daejeon, Republic of Korea.
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea.
| |
Collapse
|
2
|
Stojanovic L, Abbotts R, Tripathi K, Coon CM, Rajendran S, Farid EA, Hostetter G, Guarnieri JW, Wallace DC, Liu S, Wan J, Calendo G, Marker R, Gohari Z, Inayatullah MMA, Tiwari VK, Kader T, Santagata S, Drapkin R, Kommoss S, Pfisterer J, Konecny GE, Coopergard R, Issa JP, Winterhoff BJN, Topper MJ, Sandusky GE, Miller KD, Baylin SB, Nephew KP, Rassool FV. ZNFX1 is a Novel Master Regulator in Epigenetically-induced Pathogen Mimicry and Inflammasome Signaling in Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.18.618659. [PMID: 39484607 PMCID: PMC11526927 DOI: 10.1101/2024.10.18.618659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
DNA methyltransferase and poly(ADP-ribose) polymerase inhibitors (DNMTis, PARPis) induce a stimulator of interferon (IFN) genes (STING)-dependent pathogen mimicry response (PMR) in ovarian (OC) and other cancers. We now show that combining DNMTis and PARPis upregulates expression of a little-studied nucleic-acid sensor, NFX1-type zinc finger-containing 1 protein (ZNFX1). We demonstrate that ZNFX1 is a novel master regulator for PMR induction in mitochondria, serving as a gateway for STING-dependent PMR. In patient OC databases, high ZNFX1 expression levels correlate with advanced stage disease. ZNFX1 expression alone significantly correlates with an increase in overall survival in a phase 3 trial for therapy-resistant OC patients receiving bevacizumab in combination with chemotherapy. In correlative RNA-seq data, inflammasome signaling through ZNFX1 correlates with abnormal vasculogenesis. ZNFX1 controls PMR signaling through the mitochondria and may serve as a biomarker to facilitate offering personalized therapy in OC patients, highlighting the strong translational significance of our findings. Significance statement DNA methyltransferase and poly(ADP-ribose) polymerase inhibitors upregulate expression of a novel nucleic-acid sensor, ZNFX1 that serves as a mitochondrial gateway to STING-dependent interferon/inflammasome signaling with tumor suppressor properties in ovarian cancer.
Collapse
Affiliation(s)
- Lora Stojanovic
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Rachel Abbotts
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Kaushlendra Tripathi
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Collin M. Coon
- Medical Sciences Program, Indiana University School of Medicine-Bloomington, Bloomington, IN 47405, USA
| | - Saranya Rajendran
- Medical Sciences Program, Indiana University School of Medicine-Bloomington, Bloomington, IN 47405, USA
| | - Elnaz Abbasi Farid
- Medical Sciences Program, Indiana University School of Medicine-Bloomington, Bloomington, IN 47405, USA
| | | | - Joseph W. Guarnieri
- Center for Mitochondrial and Epigenomic Medicine, Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Douglas C. Wallace
- Center for Mitochondrial and Epigenomic Medicine, Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, Division of Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine Indianapolis, IN 46202, USA
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine Indianapolis, IN 46202, USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine Indianapolis, IN 46202, USA
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine Indianapolis, IN 46202, USA
| | | | - Rebecca Marker
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Zahra Gohari
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | - Tanjina Kader
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
- Ludwig Center at Harvard, Boston, MA, USA
| | - Sandro Santagata
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
- Ludwig Center at Harvard, Boston, MA, USA
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Ronny Drapkin
- Penn Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Basser Center for BRCA, Abramson Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Stefan Kommoss
- Diakonie-Klinikum Schwäbisch Hall, Germany, Institute for Health Informatics
- Kliniken Essen-Mitte, Gynäkologie und Gynäkologische Onkologie, Germany
| | | | | | - Ryan Coopergard
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Boris J. N. Winterhoff
- Department of Obstetrics, Gynecology and Women’s Health (OBGYN), Division of Gynecologic Oncology, University of Minnesota, MN 55812-3011, USA
| | - Michael J. Topper
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21231, USA
| | - George E. Sandusky
- Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
| | - Kathy D. Miller
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Stephen B. Baylin
- Diakonie-Klinikum Schwäbisch Hall, Germany, Institute for Health Informatics
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21231, USA
| | - Kenneth P. Nephew
- Medical Sciences Program, Indiana University School of Medicine-Bloomington, Bloomington, IN 47405, USA
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine Indianapolis, IN 46202, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
| | - Feyruz V. Rassool
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
3
|
Negara I, Tomuleasa C, Buruiana S, Efremov DG. Molecular Subtypes and the Role of TP53 in Diffuse Large B-Cell Lymphoma and Richter Syndrome. Cancers (Basel) 2024; 16:2170. [PMID: 38927876 PMCID: PMC11201917 DOI: 10.3390/cancers16122170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 05/30/2024] [Accepted: 06/01/2024] [Indexed: 06/28/2024] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common lymphoid malignancy and a heterogeneous entity comprised of several biologically distinct subtypes. Recently, novel genetic classifications of DLBCL have been resolved based on common mutational patterns indicative of distinct pathways of transformation. However, the complicated and costly nature of the novel classifiers has precluded their inclusion into routine practice. In view of this, the status of the TP53 gene, which is mutated or deleted in 20-30% of the cases, has emerged as an important prognostic factor for DLBCL patients, setting itself apart from other predictors. TP53 genetic lesions are particularly enriched in a genetic subtype of DLBCL that shares genomic features with Richter Syndrome, highlighting the possibility of a subset of DLBCL arising from the transformation of an occult chronic lymphocytic leukemia-like malignancy, such as monoclonal B-cell lymphocytosis. Patients with TP53-mutated DLBCL, including those with Richter Syndrome, have a particularly poor prognosis and display inferior responses to standard chemoimmunotherapy regimens. The data presented in this manuscript argue for the need for improved and more practical risk-stratification models for patients with DLBCL and show the potential for the use of TP53 mutational status for prognostication and, in prospect, treatment stratification in DLBCL.
Collapse
Affiliation(s)
- Ivan Negara
- Molecular Hematology Unit, International Centre for Genetic Engineering and Biotechnology, 34149 Trieste, Italy
- Department of Internal Medicine, Hematology, “Nicolae Testemitanu” State University of Medicine and Pharmacy, 2004 Chisinau, Moldova;
| | - Ciprian Tomuleasa
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania;
| | - Sanda Buruiana
- Department of Internal Medicine, Hematology, “Nicolae Testemitanu” State University of Medicine and Pharmacy, 2004 Chisinau, Moldova;
| | - Dimitar G. Efremov
- Molecular Hematology Unit, International Centre for Genetic Engineering and Biotechnology, 34149 Trieste, Italy
- Macedonian Academy of Sciences and Arts, 1000 Skopje, North Macedonia
| |
Collapse
|
4
|
Zhang Y, Zhao H, Deng W, Lai J, Sang K, Chen Q. Zebularine potentiates anti-tumor immunity by inducing tumor immunogenicity and improving antigen processing through cGAS-STING pathway. Commun Biol 2024; 7:587. [PMID: 38755254 PMCID: PMC11099016 DOI: 10.1038/s42003-024-06271-w] [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: 08/04/2023] [Accepted: 04/30/2024] [Indexed: 05/18/2024] Open
Abstract
DNA methylation is an important epigenetic mechanism involved in the anti-tumor immune response, and DNA methyltransferase inhibitors (DNMTi) have achieved impressive therapeutic outcomes in patients with certain cancer types. However, it is unclear how inhibition of DNA methylation bridges the innate and adaptive immune responses to inhibit tumor growth. Here, we report that DNMTi zebularine reconstructs tumor immunogenicity, in turn promote dendritic cell maturation, antigen-presenting cell activity, tumor cell phagocytosis by APCs, and efficient T cell priming. Further in vivo and in vitro analyses reveal that zebularine stimulates cGAS-STING-NF-κB/IFNβ signaling to enhance tumor cell immunogenicity and upregulate antigen processing and presentation machinery (AgPPM), which promotes effective CD4+ and CD8+ T cell-mediated killing of tumor cells. These findings support the use of combination regimens that include DNMTi and immunotherapy for cancer treatment.
Collapse
Affiliation(s)
- Yong Zhang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China
| | - Heng Zhao
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China
| | - Weili Deng
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China
| | - Junzhong Lai
- The Cancer Center, Union Hospital, Fujian Medical University, Fuzhou, Fujian Province, 350117, China
| | - Kai Sang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China
| | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China.
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China.
| |
Collapse
|
5
|
Sampaio LR, Dias RDB, Goes JVC, de Melo RPM, de Paula Borges D, de Lima Melo MM, de Oliveira RTG, Ribeiro-Júnior HL, Magalhães SMM, Pinheiro RF. Role of the STING pathway in myeloid neoplasms: a prospero-registered systematic review of principal hurdles of STING on the road to the clinical practice. Med Oncol 2024; 41:128. [PMID: 38656461 DOI: 10.1007/s12032-024-02376-8] [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: 03/10/2024] [Accepted: 03/28/2024] [Indexed: 04/26/2024]
Abstract
Myeloid neoplasms are a group of bone marrow diseases distinguished by disruptions in the molecular pathways that regulate the balance between hematopoietic stem cell (HSC) self-renewal and the generation of specialized cells. Cytokines and chemokines, two important components of the inflammatory process, also influence hematological differentiation. In this scenario, immunological dysregulation plays a pivotal role in the pathogenesis of bone marrow neoplasms. The STING pathway recognizes DNA fragments in the cell cytoplasm and triggers an immune response by type I interferons. The role of STING in cancer has not yet been established; however, both actions, as an oncogene or tumor suppressor, have been documented in other types of cancer. Therefore, we performed a systematic review (registered in PROSPERO database #CRD42023407512) to discuss the role of STING pathway in the advancement of pathogenesis and/or prognosis for different myeloid neoplasms. In brief, scientific evidence supports investigations that primarily use cell lines from myeloid neoplasms, such as leukemia. More high-quality research and clinical trials are needed to understand the role of the STING pathway in the pathology of hematological malignancies. Finally, the STING pathway suggests being a promising therapeutic molecular target, particularly when combined with current drug therapies.
Collapse
Affiliation(s)
- Leticia Rodrigues Sampaio
- Cancer Cytogenomic Laboratory, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Post-Graduate Program in Medical Science, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Drug Research and Development Center (NPDM), Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - Ricardo Dyllan Barbosa Dias
- Cancer Cytogenomic Laboratory, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Post-Graduate Program in Medical Science, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Drug Research and Development Center (NPDM), Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - João Vitor Caetano Goes
- Cancer Cytogenomic Laboratory, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Drug Research and Development Center (NPDM), Federal University of Ceara, Fortaleza, Ceara, Brazil
- Post-Graduate Program of Pathology, Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - Renata Pinheiro Martins de Melo
- Cancer Cytogenomic Laboratory, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Drug Research and Development Center (NPDM), Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - Daniela de Paula Borges
- Cancer Cytogenomic Laboratory, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Post-Graduate Program in Medical Science, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Drug Research and Development Center (NPDM), Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - Mayara Magna de Lima Melo
- Cancer Cytogenomic Laboratory, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Post-Graduate Program in Medical Science, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Drug Research and Development Center (NPDM), Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - Roberta Taiane Germano de Oliveira
- Cancer Cytogenomic Laboratory, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Post-Graduate Program in Medical Science, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Drug Research and Development Center (NPDM), Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - Howard Lopes Ribeiro-Júnior
- Cancer Cytogenomic Laboratory, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Post-Graduate Program in Medical Science, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Drug Research and Development Center (NPDM), Federal University of Ceara, Fortaleza, Ceara, Brazil
- Post-Graduate Program of Pathology, Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - Silvia Maria Meira Magalhães
- Cancer Cytogenomic Laboratory, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Post-Graduate Program in Medical Science, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Drug Research and Development Center (NPDM), Federal University of Ceara, Fortaleza, Ceara, Brazil
- Post-Graduate Program of Pathology, Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - Ronald Feitosa Pinheiro
- Cancer Cytogenomic Laboratory, Federal University of Ceara, Fortaleza, Ceara, Brazil.
- Post-Graduate Program in Medical Science, Federal University of Ceara, Fortaleza, Ceara, Brazil.
- Drug Research and Development Center (NPDM), Federal University of Ceara, Fortaleza, Ceara, Brazil.
- Post-Graduate Program of Pathology, Federal University of Ceara, Fortaleza, Ceara, Brazil.
| |
Collapse
|
6
|
Chen Q, Hong Y, Chen W, Lin F, Zeng J, Huang Y, Zhang L, Yao J, Xu B. Prognostic implications of cGAS and STING gene expression in acute myeloid leukemia. Exp Biol Med (Maywood) 2024; 249:10108. [PMID: 38510490 PMCID: PMC10954193 DOI: 10.3389/ebm.2024.10108] [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: 07/09/2023] [Accepted: 09/25/2023] [Indexed: 03/22/2024] Open
Abstract
Acute myeloid leukemia (AML) is one of the most threatening hematological malignances. cGAS-STING pathway plays an important role in tumor immunity and development. However, the prognostic role of cGAS-STING pathway in AML remains unknown. Firstly, The expression of cGAS and STING was analyzed by bioinformatics analysis. Subsequently, Bone marrow samples were collected from 120 AML patients and 15 healthy individuals in an independent cohort. The cGAS and STING expression was significantly elevated in AML patients compared with healthy controls. Patients with high cGAS and STING expression had a higher NRAS/KRAS mutation rate and lower complete remission (CR) rate. High cGAS and STING expression was significantly associated with lower overall survival (OS) and disease-free survival (DFS). Our findings revealed that the expression levels of cGAS and STING in AML are elevated. High expression of cGAS and STING correlated with worse OS and DFS and may be a useful biomarker for inferior prognosis in AML patients.
Collapse
Affiliation(s)
- Qiuling Chen
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian, China
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China
| | - Yan Hong
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China
| | - WeiFeng Chen
- Department of Hematology, Shantou Central Hospital, Shantou, Guangdong, China
| | - Feng Lin
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China
- Department of Hematology, Shantou Central Hospital, Shantou, Guangdong, China
| | - Jiawei Zeng
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian, China
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China
| | - Yueting Huang
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China
| | - Li Zhang
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China
| | - Jingwei Yao
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China
| | - Bing Xu
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
7
|
Liu J, Zhou J, Luan Y, Li X, Meng X, Liao W, Tang J, Wang Z. cGAS-STING, inflammasomes and pyroptosis: an overview of crosstalk mechanism of activation and regulation. Cell Commun Signal 2024; 22:22. [PMID: 38195584 PMCID: PMC10775518 DOI: 10.1186/s12964-023-01466-w] [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: 08/23/2023] [Accepted: 12/28/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND Intracellular DNA-sensing pathway cGAS-STING, inflammasomes and pyroptosis act as critical natural immune signaling axes for microbial infection, chronic inflammation, cancer progression and organ degeneration, but the mechanism and regulation of the crosstalk network remain unclear. Cellular stress disrupts mitochondrial homeostasis, facilitates the opening of mitochondrial permeability transition pore and the leakage of mitochondrial DNA to cell membrane, triggers inflammatory responses by activating cGAS-STING signaling, and subsequently induces inflammasomes activation and the onset of pyroptosis. Meanwhile, the inflammasome-associated protein caspase-1, Gasdermin D, the CARD domain of ASC and the potassium channel are involved in regulating cGAS-STING pathway. Importantly, this crosstalk network has a cascade amplification effect that exacerbates the immuno-inflammatory response, worsening the pathological process of inflammatory and autoimmune diseases. Given the importance of this crosstalk network of cGAS-STING, inflammasomes and pyroptosis in the regulation of innate immunity, it is emerging as a new avenue to explore the mechanisms of multiple disease pathogenesis. Therefore, efforts to define strategies to selectively modulate cGAS-STING, inflammasomes and pyroptosis in different disease settings have been or are ongoing. In this review, we will describe how this mechanistic understanding is driving possible therapeutics targeting this crosstalk network, focusing on the interacting or regulatory proteins, pathways, and a regulatory mitochondrial hub between cGAS-STING, inflammasomes, and pyroptosis. SHORT CONCLUSION This review aims to provide insight into the critical roles and regulatory mechanisms of the crosstalk network of cGAS-STING, inflammasomes and pyroptosis, and to highlight some promising directions for future research and intervention.
Collapse
Affiliation(s)
- Jingwen Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Jing Zhou
- The Second Hospital of Ningbo, Ningbo, 315099, China
| | - Yuling Luan
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiaoying Li
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200080, China
| | - Xiangrui Meng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Wenhao Liao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Jianyuan Tang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
| | - Zheilei Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
| |
Collapse
|
8
|
Zhao M, Wang T, Gleber-Netto FO, Chen Z, McGrail DJ, Gomez JA, Ju W, Gadhikar MA, Ma W, Shen L, Wang Q, Tang X, Pathak S, Raso MG, Burks JK, Lin SY, Wang J, Multani AS, Pickering CR, Chen J, Myers JN, Zhou G. Mutant p53 gains oncogenic functions through a chromosomal instability-induced cytosolic DNA response. Nat Commun 2024; 15:180. [PMID: 38167338 PMCID: PMC10761733 DOI: 10.1038/s41467-023-44239-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024] Open
Abstract
Inactivating TP53 mutations leads to a loss of function of p53, but can also often result in oncogenic gain-of-function (GOF) of mutant p53 (mutp53) proteins which promotes tumor development and progression. The GOF activities of TP53 mutations are well documented, but the mechanisms involved remain poorly understood. Here, we study the mutp53 interactome and find that by targeting minichromosome maintenance complex components (MCMs), GOF mutp53 predisposes cells to replication stress and chromosomal instability (CIN), leading to a tumor cell-autonomous and cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING)-dependent cytosolic DNA response that activates downstream non-canonical nuclear factor kappa light chain enhancer of activated B cell (NC-NF-κB) signaling. Consequently, GOF mutp53-MCMs-CIN-cytosolic DNA-cGAS-STING-NC-NF-κB signaling promotes tumor cell metastasis and an immunosuppressive tumor microenvironment through antagonizing interferon signaling and regulating genes associated with pro-tumorigenic inflammation. Our findings have important implications for understanding not only the GOF activities of TP53 mutations but also the genome-guardian role of p53 and its inactivation during tumor development and progression.
Collapse
Affiliation(s)
- Mei Zhao
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Tianxiao Wang
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Head and Neck Surgery, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Frederico O Gleber-Netto
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Zhen Chen
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Daniel J McGrail
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, 44195, USA
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Javier A Gomez
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Wutong Ju
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Mayur A Gadhikar
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Wencai Ma
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Li Shen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Qi Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ximing Tang
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Sen Pathak
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Maria Gabriela Raso
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jared K Burks
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Shiaw-Yih Lin
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Asha S Multani
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Curtis R Pickering
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Surgery-Otolaryngology, Yale School of Medicine, New Haven, CT, 06250, USA
| | - Junjie Chen
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jeffrey N Myers
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Ge Zhou
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
9
|
Zhang MC, Tian S, Fu D, Wang L, Cheng S, Yi HM, Jiang XF, Song Q, Zhao Y, He Y, Li JF, Mu RJ, Fang H, Yu H, Xiong H, Li B, Chen SJ, Xu PP, Zhao WL. Genetic subtype-guided immunochemotherapy in diffuse large B cell lymphoma: The randomized GUIDANCE-01 trial. Cancer Cell 2023; 41:1705-1716.e5. [PMID: 37774697 DOI: 10.1016/j.ccell.2023.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/25/2023] [Accepted: 09/05/2023] [Indexed: 10/01/2023]
Abstract
We report the results of GUIDANCE-01 (NCT04025593), a randomized, phase II trial of R-CHOP alone or combined with targeted agents (R-CHOP-X) guided by genetic subtyping of newly diagnosed, intermediate-risk, or high-risk diffuse large B cell lymphoma (DLBCL). A total of 128 patients were randomized 1:1 to receive R-CHOP-X or R-CHOP. The study achieved the primary endpoint, showing significantly higher complete response rate with R-CHOP-X than R-CHOP (88% vs. 66%, p = 0.003), with overall response rate of 92% vs. 73% (p = 0.005). Two-year progression-free survival rates were 88% vs. 63% (p < 0.001), and 2-year overall survival rates were 94% vs. 77% (p = 0.001). Meanwhile, post hoc RNA-sequencing validated our simplified genetic subtyping algorithm and previously established lymphoma microenvironment subtypes. Our findings highlight the efficacy and safety of R-CHOP-X, a mechanism-based tailored therapy, which dually targeted genetic and microenvironmental alterations in patients with newly diagnosed DLBCL.
Collapse
Affiliation(s)
- Mu-Chen Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuang Tian
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Di Fu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai, China
| | - Shu Cheng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong-Mei Yi
- Department of Pathology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xu-Feng Jiang
- Department of Nuclear Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Song
- Department of Radiology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Zhao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yang He
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian-Feng Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rong-Ji Mu
- Clinical Research Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hai Fang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Yu
- Department of Research and Development, Shanghai Righton Biotechnology Co. Ltd, Shanghai, China
| | - Hui Xiong
- Department of Research and Development, Shanghai Righton Biotechnology Co. Ltd, Shanghai, China
| | - Biao Li
- Department of Nuclear Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sai-Juan Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peng-Peng Xu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Wei-Li Zhao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai, China.
| |
Collapse
|
10
|
Fang Y, Zhang MC, He Y, Li C, Fang H, Xu PP, Cheng S, Zhao Y, Feng Y, Liu Q, Wang L, Zhao WL. Human endogenous retroviruses as epigenetic therapeutic targets in TP53-mutated diffuse large B-cell lymphoma. Signal Transduct Target Ther 2023; 8:381. [PMID: 37798292 PMCID: PMC10556001 DOI: 10.1038/s41392-023-01626-x] [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: 03/09/2023] [Revised: 08/13/2023] [Accepted: 08/21/2023] [Indexed: 10/07/2023] Open
Abstract
TP53 mutation (TP53mut) occurs in 10-20% of diffuse large B-cell lymphoma (DLBCL) cases and serves as an unfavorable biomarker of DLBCL progression. It confers resistance to immunochemotherapy, high-dose chemotherapy, autologous stem cell transplantation, and anti-CD19 chimeric antigen receptor T-cell therapy. Therapeutic targeting of TP53mut remains a significant challenge in DLBCL treatment. Here we assessed TP53mut in 667 patients with newly diagnosed DLBCL, including 576 patients treated with immunochemotherapy rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) and 91 patients with decitabine plus R-CHOP (DR-CHOP, NCT02951728 and NCT04025593). TP53mut independently predicted an inferior prognosis in R-CHOP-treated DLBCL, although this could be mitigated by DR-CHOP treatment. In TP53mut patients, multiple viral regulation pathways were repressed, resulting in the inhibition of immune modulation, as revealed by gene set enrichment analysis. TP53mut DLBCL exhibited increased methyltransferase SUV39H1 expression and H3K9 trimethylation (H3K9me3), contributing to repression of endogenous retroviruses (ERVs) and immunosuppressive tumor microenvironment. In TP53mut DLBCL cell lines, decitabine down-regulated SUV39H1, inhibited H3K9me3 occupancy on ERVs, and triggered ERV expression, thereby unleashing interferons program and CD4+T/CD8+T cell activation. Molecular silencing of SUV39H1 significantly abrogated decitabine-induced H3K9me3 inhibition and ERV expression. In TP53mut patient-derived xenograft models and TP53mut patients, the anti-tumor effect was improved upon the use of combined treatment of decitabine and doxorubicin via SUV39H1-H3K9me3-ERVs axis. Collectively, our findings highlight an ERV regulatory circuitry in TP53mut DLBCL and the crucial roles ERVs for epigenetically reprogramming tumor microenvironment for treating TP53mut-driven cancers.
Collapse
Grants
- National Key R&D Program of China (2022YFC2502600), Chang Jiang Scholars Program, Shanghai Commission of Science and Technology (17PJ1405800), Shanghai Municipal Education Commission Gaofeng Clinical Medicine Grant Support (20152206, 20152208, and 20171902), Clinical Research Plan of Shanghai hospital development center (SHDC, 2020CR1032B), Multicenter Clinical Research Project by Shanghai Jiao Tong University School of Medicine (DLY201601), Samuel Waxman Cancer Research Foundation, and the Foundation of National Facility for Translational Medicine (Shanghai, TMSK-2020-115).
Collapse
Affiliation(s)
- Ying Fang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mu-Chen Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yang He
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Li
- Network and Information Center, Shanghai Jiao Tong University, Shanghai, China
| | - Hai Fang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peng-Peng Xu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shu Cheng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Zhao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Feng
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Qian Liu
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Li Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Wei-Li Zhao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
11
|
Chang YS, Hsu MH, Chung CC, Chen HD, Tu SJ, Lee YT, Yen JC, Liu TC, Chang JG. Comprehensive Analysis and Drug Modulation of Human Endogenous Retrovirus in Hepatocellular Carcinomas. Cancers (Basel) 2023; 15:3664. [PMID: 37509325 PMCID: PMC10377948 DOI: 10.3390/cancers15143664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/12/2023] [Accepted: 07/16/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Human endogenous retroviruses (HERVs) play an important role in the development of cancer and many diseases. Here, we comprehensively explored the impact of HERVs on hepatocellular carcinomas (HCCs). METHODS We employed Telescope to identify HERVs and quantify their expression in the total RNA sequencing data obtained from 254 HCC samples, comprising 254 tumor tissues and 34 matched normal tissues. RESULTS In total, 3357 locus-specific activations of HERVs were differentially expressed, and 180 were correlated with patient survival. Using these 180 HERVs for classification, we found four subgroups with survival correlation. Higher expression levels of the 180 HERVs were correlated with poorer survival, while age, AFP, some mutations, and copy and structural variants differed among subgroups. The differential expression of host genes in high expression of these 180 HERVs primarily involved the activation of pathways related to immunity and infection, lipid and atherosclerosis, MAPK and NF-kB signaling, and cytokine-cytokine receptor interactions. Conversely, there was a suppression of pathways associated with RNA processing, including nucleocytoplasmic transport, surveillance and ribosome biogenesis, and transcriptional misregulation in cancer pathways. Almost all genes involved in HERV activation restriction, KRAB zinc finger proteins, RNA nucleocytoplasmic transport, stemness, HLA and antigen processing and presentation, and immune checkpoints were overexpressed in cancerous tissues, and many over-expressed HERV-related nearby genes were correlated with high HERV activation and poor survival. Twenty-three immune and stromal cells showed higher expression in non-cancerous than cancerous tissues, and seven were correlated with HERV activation. Small-molecule modulation of alternative splicing (AS) altered the expression of survival-related HERVs and their activation-related genes, as well as nearby genes. CONCLUSION Comprehensive and integrated approaches for evaluating HERV expression and their correlation with specific pathways have the potential to provide new companion diagnostics and therapeutic strategies for HCC.
Collapse
Affiliation(s)
- Ya-Sian Chang
- Center for Precision Medicine, China Medical University Hospital, Taichung 40447, Taiwan
- Epigenome Research Center, China Medical University Hospital, Taichung 40447, Taiwan
- Department of Laboratory Medicine, China Medical University Hospital, Taichung 40447, Taiwan
- School of Medicine, China Medical University, Taichung 40402, Taiwan
| | - Ming-Hon Hsu
- Center for Precision Medicine, China Medical University Hospital, Taichung 40447, Taiwan
- Epigenome Research Center, China Medical University Hospital, Taichung 40447, Taiwan
- Department of Laboratory Medicine, China Medical University Hospital, Taichung 40447, Taiwan
| | - Chin-Chun Chung
- Center for Precision Medicine, China Medical University Hospital, Taichung 40447, Taiwan
- Epigenome Research Center, China Medical University Hospital, Taichung 40447, Taiwan
| | - Hong-Da Chen
- Center for Precision Medicine, China Medical University Hospital, Taichung 40447, Taiwan
- Epigenome Research Center, China Medical University Hospital, Taichung 40447, Taiwan
- Department of Laboratory Medicine, China Medical University Hospital, Taichung 40447, Taiwan
| | - Siang-Jyun Tu
- Center for Precision Medicine, China Medical University Hospital, Taichung 40447, Taiwan
- Epigenome Research Center, China Medical University Hospital, Taichung 40447, Taiwan
- Department of Laboratory Medicine, China Medical University Hospital, Taichung 40447, Taiwan
| | - Ya-Ting Lee
- Center for Precision Medicine, China Medical University Hospital, Taichung 40447, Taiwan
- Epigenome Research Center, China Medical University Hospital, Taichung 40447, Taiwan
| | - Ju-Chen Yen
- Center for Precision Medicine, China Medical University Hospital, Taichung 40447, Taiwan
- Epigenome Research Center, China Medical University Hospital, Taichung 40447, Taiwan
| | - Ta-Chih Liu
- Department of Hematology-Oncology, Chang Bing Show Chwan Memorial Hospital, Changhua 50544, Taiwan
| | - Jan-Gowth Chang
- Center for Precision Medicine, China Medical University Hospital, Taichung 40447, Taiwan
- Epigenome Research Center, China Medical University Hospital, Taichung 40447, Taiwan
- Department of Laboratory Medicine, China Medical University Hospital, Taichung 40447, Taiwan
- School of Medicine, China Medical University, Taichung 40402, Taiwan
| |
Collapse
|
12
|
Fan TJ, Cui J. Human Endogenous Retroviruses in Diseases. Subcell Biochem 2023; 106:403-439. [PMID: 38159236 DOI: 10.1007/978-3-031-40086-5_15] [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: 01/03/2024]
Abstract
Human endogenous retroviruses (HERVs), which are conserved sequences of ancient retroviruses, are widely distributed in the human genome. Although most HERVs have been rendered inactive by evolution, some have continued to exhibit important cytological functions. HERVs in the human genome perform dual functions: on the one hand, they are involved in important physiological processes such as placental development and immune regulation; on the other hand, their aberrant expression is closely associated with the pathological processes of several diseases, such as cancers, autoimmune diseases, and viral infections. HERVs can also regulate a variety of host cellular functions, including the expression of protein-coding genes and regulatory elements that have evolved from HERVs. Here, we present recent research on the roles of HERVs in viral infections and cancers, including the dysregulation of HERVs in various viral infections, HERV-induced epigenetic modifications of histones (such as methylation and acetylation), and the potential mechanisms of HERV-mediated antiviral immunity. We also describe therapies to improve the efficacy of vaccines and medications either by directly or indirectly targeting HERVs, depending on the HERV.
Collapse
Affiliation(s)
- Tian-Jiao Fan
- CAS Key Laboratory of Molecular Virology & Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Jie Cui
- CAS Key Laboratory of Molecular Virology & Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
13
|
The stimulator of interferon genes (STING) agonists for treating acute myeloid leukemia (AML): current knowledge and future outlook. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2022; 25:1545-1553. [PMID: 36587109 DOI: 10.1007/s12094-022-03065-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/26/2022] [Indexed: 01/01/2023]
Abstract
Acute myeloid leukemia (AML) is an aggressive hematologic cancer in adults. Some patients exhibit restricted T cell infiltration and do not respond to routine treatments. This may be prevented by enhancing adaptive immunity by stimulating innate immune cells inside the tumor microenvironment (TME). To activate the adaptive immunological reaction against tumors, type I interferons (IFNs) can promote the presentation of tumor-specific cytotoxic T lymphocyte (CTL) cell recruitment. During the activation of innate immunity, cyclic di-nucleotides (CDNs) bind to and stimulate the stimulator of interferon genes (STING), a protein localized inside the endoplasmic reticulum (ER) membrane, resulting in the expression of type I IFNs. The efficacy of STING agonists as effective stimulators of the anti-tumor response in AML is being investigated in numerous clinical studies. Therefore, the purpose of this investigation was to thoroughly review existing knowledge in this field and provide perspective into the clinical potential of STING agonists in AML.
Collapse
|
14
|
Chen C, Xu P. Cellular functions of cGAS-STING signaling. Trends Cell Biol 2022:S0962-8924(22)00252-5. [DOI: 10.1016/j.tcb.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/27/2022]
|