1
|
Pacella GN, Kuprasertkul N, Bao L, Huang S, D’souza C, Prouty SM, Anderson A, Maldonado López AM, Sinkfield M, Olingou C, Seykora JT, Capell BC. UTX (KDM6A) promotes differentiation noncatalytically in somatic self-renewing epithelia. Proc Natl Acad Sci U S A 2025; 122:e2422971122. [PMID: 40372430 PMCID: PMC12107135 DOI: 10.1073/pnas.2422971122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/24/2025] [Indexed: 05/16/2025] Open
Abstract
The X-linked histone demethylase, UTX (KDM6A), is a master regulator of gene enhancers, though its role in self-renewing epithelia like the skin is not well understood. Here, we find that UTX is a key regulator of skin differentiation via the regulation of retinoic acid (RA) signaling, an essential metabolic pathway in both skin homeostasis, as well as in the treatment of an array of skin conditions ranging from cancer and acne to aging. Through deletion of Utx in the skin, we demonstrate direct regulation of both retinoid metabolic genes such as Crabp2, as well as key genes involved in epidermal stem cell fate and differentiation (i.e., Cdh1, Grhl3, Ctnnb1). Spatial analyses show that UTX loss dysregulates epidermal, sebaceous, and hair follicle differentiation programs. Strikingly, this only occurs in homozygous females, demonstrating that UTX's Y-linked paralog, UTY (Kdm6c), can compensate in males. Further, we observe genome-wide losses of H3K27 acetylation (H3K27ac) with minimal changes in H3K27 trimethylation (H3K27me3), revealing that UTX functions primarily noncatalytically to promote skin homeostasis. Together, the elucidation of these links between epigenetics, metabolic signaling, and epithelial differentiation offers new insights into how epigenetic modulation may allow for fine-tuning of key signaling pathways to treat disease.
Collapse
Affiliation(s)
- Gina N. Pacella
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Penn Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
| | - Nina Kuprasertkul
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Penn Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
| | - Lydia Bao
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Penn Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
| | - Sijia Huang
- Penn Institute of Biomedical Informatics, University of Pennsylvania, Perelman School of Medicine, PhiladelphiaPA19104
| | - Carina D’souza
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Penn Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
| | - Stephen M. Prouty
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
| | - Amy Anderson
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Penn Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
| | - Alexandra M. Maldonado López
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Penn Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
| | - Morgan Sinkfield
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Penn Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
| | - Cyria Olingou
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Penn Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
| | - John T. Seykora
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
| | - Brian C. Capell
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Penn Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Penn Institute for Regenerative Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
| |
Collapse
|
2
|
Lee S, Ho YY, Hao S, Ouyang Y, Liew UL, Goyal A, Li S, Barbour JA, He M, Huang Y, Wong JWH. A tumour necrosis factor-α responsive cryptic promoter drives overexpression of the human endogenous retrovirus ERVK-7. J Biol Chem 2025:108568. [PMID: 40316021 DOI: 10.1016/j.jbc.2025.108568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 04/09/2025] [Accepted: 04/28/2025] [Indexed: 05/04/2025] Open
Abstract
Endogenous retroviruses (ERVs) shape human genome functionality and influence disease pathogenesis, including cancer. ERVK-7, a significant ERV, acts as an immune modulator and prognostic marker in lung adenocarcinoma (LUAD). Although ERVK-7 overexpression has been linked to the amplification of the 1q22 locus in approximately 10% of LUAD cases, it predominantly arises from alternative regulatory mechanisms. Our findings indicate that the canonical 5' long terminal repeat (LTR) of ERVK-7 is methylated and inactive, necessitating the use of alternative upstream promoters. We identified two novel transcripts, ERVK-7.long and ERVK-7.short, arising from distinct promoters located 2.8 kb and 13.8 kb upstream of the 5'LTR of ERVK-7, respectively. ERVK-7.long is predominantly overexpressed in LUAD. Through comprehensive epigenetic mapping and single-cell transcriptomics, we demonstrate that ERVK-7.long activation is predetermined by cell lineage, specifically in small airway epithelial cells (SAECs), where its promoter displays tumor-specific H3K4me3 modifications. Single-cell RNA sequencing further reveals a distinct enrichment of ERVK-7.long in LUAD tumor cells and alveolar type 2 epithelial cells, underscoring a cell-type-specific origin. Additionally, inflammatory signaling significantly influences ERVK-7 expression; TNF-α enhances ERVK-7.long, while interferon signaling preferentially augments ERVK-7.short by differential recruitment of NF-κB/RELA and IRF to their respective promoters. This differential regulation clarifies the elevated ERVK-7 expression in LUAD compared to lung squamous cell carcinoma (LUSC). Our study elucidates the complex regulatory mechanisms governing ERVK-7 in LUAD and proposes these transcripts as potential biomarkers and therapeutic targets, offering new avenues to improve patient outcomes.
Collapse
Affiliation(s)
- Sojung Lee
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China; Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong SAR, China
| | - Yin Yee Ho
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Suyu Hao
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Yingqi Ouyang
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - U Ling Liew
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Ashish Goyal
- Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephen Li
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Jayne A Barbour
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Mu He
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Yuanhua Huang
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China; Center for Translational Stem Cell Biology, Hong Kong Science and Technology Park, Hong Kong SAR, China; Department of Statistics and Actuarial Science, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Jason W H Wong
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China; Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong SAR, China.
| |
Collapse
|
3
|
Naffaa MM, Al-Ewaidat OA, Gogia S, Begiashvili V. Neoantigen-based immunotherapy: advancing precision medicine in cancer and glioblastoma treatment through discovery and innovation. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002313. [PMID: 40309350 PMCID: PMC12040680 DOI: 10.37349/etat.2025.1002313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025] Open
Abstract
Neoantigen-based immunotherapy has emerged as a transformative approach in cancer treatment, offering precision medicine strategies that target tumor-specific antigens derived from genetic, transcriptomic, and proteomic alterations unique to cancer cells. These neoantigens serve as highly specific targets for personalized therapies, promising more effective and tailored treatments. The aim of this article is to explore the advances in neoantigen-based therapies, highlighting successful treatments such as vaccines, tumor-infiltrating lymphocyte (TIL) therapy, T-cell receptor-engineered T cells therapy (TCR-T), and chimeric antigen receptor T cells therapy (CAR-T), particularly in cancer types like glioblastoma (GBM). Advances in technologies such as next-generation sequencing, RNA-based platforms, and CRISPR gene editing have accelerated the identification and validation of neoantigens, moving them closer to clinical application. Despite promising results, challenges such as tumor heterogeneity, immune evasion, and resistance mechanisms persist. The integration of AI-driven tools and multi-omic data has refined neoantigen discovery, while combination therapies are being developed to address issues like immune suppression and scalability. Additionally, the article discusses the ongoing development of personalized immunotherapies targeting tumor mutations, emphasizing the need for continued collaboration between computational and experimental approaches. Ultimately, the integration of cutting-edge technologies in neoantigen research holds the potential to revolutionize cancer care, offering hope for more effective and targeted treatments.
Collapse
Affiliation(s)
- Moawiah M Naffaa
- Department of Psychology and Neuroscience, Duke University, Durham, NC 27708, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ola A Al-Ewaidat
- Department of Internal Medicine, Ascension Saint Francis Hospital, Evanston, IL 60202, USA
| | - Sopiko Gogia
- Department of Internal Medicine, Ascension Saint Francis Hospital, Evanston, IL 60202, USA
| | - Valiko Begiashvili
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66103, USA
| |
Collapse
|
4
|
Simula ER, Jasemi S, Cossu D, Fais M, Cossu I, Chessa V, Canu M, Sechi LA. Human Endogenous Retroviruses as Novel Therapeutic Targets in Neurodegenerative Disorders. Vaccines (Basel) 2025; 13:415. [PMID: 40333317 PMCID: PMC12031449 DOI: 10.3390/vaccines13040415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/11/2025] [Accepted: 04/14/2025] [Indexed: 05/09/2025] Open
Abstract
Human Endogenous Retroviruses comprise approximately 8% of the human genome, serving as fragments of ancient retroviral infections. Although they are generally maintained in a silenced state by robust epigenetic mechanisms, specific HERV groups, particularly HERV-W and HERV-K, can become derepressed under specific pathological conditions, thereby contributing to the initiation and progression of neuroinflammatory and neurodegenerative processes. Preclinical studies and clinical trials, such as those investigating monoclonal antibodies, indicate that directly targeting these elements may offer a novel therapeutic strategy. In this review, we provide an overview of HERVs' biology, examine their role in neurodegenerative diseases such as amyotrophic lateral sclerosis, multiple sclerosis, Alzheimer's disease, and Parkinson's disease, and explore their therapeutic prospects, highlighting both the challenges and the potential future research directions needed to translate these approaches into clinical interventions.
Collapse
Affiliation(s)
- Elena Rita Simula
- Department of Biomedical Sciences, Division of Microbiology and Virology, University of Sassari, 07100 Sassari, Italy; (S.J.); (D.C.); (M.F.); (I.C.)
| | - Seyedesomaye Jasemi
- Department of Biomedical Sciences, Division of Microbiology and Virology, University of Sassari, 07100 Sassari, Italy; (S.J.); (D.C.); (M.F.); (I.C.)
| | - Davide Cossu
- Department of Biomedical Sciences, Division of Microbiology and Virology, University of Sassari, 07100 Sassari, Italy; (S.J.); (D.C.); (M.F.); (I.C.)
| | - Milena Fais
- Department of Biomedical Sciences, Division of Microbiology and Virology, University of Sassari, 07100 Sassari, Italy; (S.J.); (D.C.); (M.F.); (I.C.)
| | - Ilaria Cossu
- Department of Biomedical Sciences, Division of Microbiology and Virology, University of Sassari, 07100 Sassari, Italy; (S.J.); (D.C.); (M.F.); (I.C.)
| | - Vanna Chessa
- ASL Sassari, SC Anestesia Territoriale Cure Palliatiave, 07100 Sassari, Italy; (V.C.); (M.C.)
| | - Mattia Canu
- ASL Sassari, SC Anestesia Territoriale Cure Palliatiave, 07100 Sassari, Italy; (V.C.); (M.C.)
| | - Leonardo Antonio Sechi
- Department of Biomedical Sciences, Division of Microbiology and Virology, University of Sassari, 07100 Sassari, Italy; (S.J.); (D.C.); (M.F.); (I.C.)
- Struttura Complessa Microbiologia e Virologia, Azienda Ospedaliera Universitaria Sassari, 07100 Sassari, Italy
| |
Collapse
|
5
|
Li M, Sun Y, Shan X, Tong Y, Fu Y, Ma X, Sun Z, Xiang Y, Zhu Y, Wang T, Wang X, Zhang J, Niu D. Roles of Immunity and Endogenous Retroelements in the Pathogenesis of Rheumatoid Arthritis and Treatment Strategies. Funct Integr Genomics 2025; 25:85. [PMID: 40205241 DOI: 10.1007/s10142-025-01583-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/24/2025] [Accepted: 03/17/2025] [Indexed: 04/11/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic, inflammatory, autoimmune disease that primarily affects the joints. RA usually results in synovial hyperplasia, expansion of "pannus" and destruction of cartilage. The etiology and pathogenesis of RA are not fully understood, but immunity has been shown to play an important role in the development of autoimmune diseases such as RA. In addition, endogenous retroelements, the remnants of ancient retroviruses in the human genome, are involved in cancer and/or immune disorders. As evidenced by increasing evidences that the aberrant expression of retroelements induces innate immunity, despite the fact that the expression of most retroelements has been epigenetically suppressed over a long period of evolution. With the continuous development of disease-modifying anti-rheumatic drugs (DMARDs), RA disease activity has been alleviated and improved. Unfortunately, some patients have a limited response to DMARDs, and the drugs also have the disadvantages of some side effects and high economic costs. This review summarizes the pathogenic mechanisms of RA and endogenous retroelements in autoimmunity, and concludes with a summary of treatments for RA, along with new therapeutic recommendations.
Collapse
Affiliation(s)
- Mingyang Li
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, 666 Wusu Street, Hangzhou, 311300, Zhejiang, China
| | - Yuanyuan Sun
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, 666 Wusu Street, Hangzhou, 311300, Zhejiang, China
| | - Xueting Shan
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, 666 Wusu Street, Hangzhou, 311300, Zhejiang, China
| | - Yuhong Tong
- Fourth School of Clinical Medicine, Zhejiang University of Traditional Chinese Medicine, Hangzhou, 310053, China
| | - Yite Fu
- Nanjing Outstanding Gene Technology Co, Nanjing, 210018, Jiangsu, China
| | - Xiang Ma
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, 666 Wusu Street, Hangzhou, 311300, Zhejiang, China
| | - Zhongxin Sun
- Department of Plastic, Reconstructive & Hand Microsurgery, Ningbo NO.6 Hospital, Ningbo, 315000, Zhejiang, China
| | - Yun Xiang
- Jinhua Academy of Agricultural Sciences, Jinhua, 321000, Zhejiang, China
| | - Yidan Zhu
- Jinhua Academy of Agricultural Sciences, Jinhua, 321000, Zhejiang, China
| | - Tao Wang
- Nanjing Outstanding Gene Technology Co, Nanjing, 210018, Jiangsu, China
| | - Xin Wang
- Department of Plastic, Reconstructive & Hand Microsurgery, Ningbo NO.6 Hospital, Ningbo, 315000, Zhejiang, China.
| | - Jufang Zhang
- Department of Plastic and Aesthetic Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, Zhejiang, China.
| | - Dong Niu
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, 666 Wusu Street, Hangzhou, 311300, Zhejiang, China.
| |
Collapse
|
6
|
Della Valle F, Reddy P, Aguirre Vazquez A, Izpisua Belmonte JC. Reactivation of retrotransposable elements is associated with environmental stress and ageing. Nat Rev Genet 2025:10.1038/s41576-025-00829-y. [PMID: 40175591 DOI: 10.1038/s41576-025-00829-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2025] [Indexed: 04/04/2025]
Abstract
Retrotransposable elements (RTEs) are interspersed repetitive sequences that represent a large portion of eukaryotic genomes. Ancestral expansions of RTEs directly contributed to the shaping of these genomes and to the evolution of different species, particularly mammals. RTE activity is tightly regulated by different epigenetic mechanisms but this control becomes compromised as cells age and RTEs are reactivated. This dysregulation of RTEs leads to perturbation of cell function and organ and organismal homeostasis, which drives ageing and age-related disease. Environmental stress is associated with both ageing-related characteristics and the epigenetic mechanisms that control RTE activity, with accumulating evidence indicating that RTE reactivation mediates the effects of environmental stressors on ageing onset and progression. A better understanding of how RTEs are reactivated and their subsequent biological roles may help the development of therapies against ageing-related phenotypes and diseases.
Collapse
Affiliation(s)
| | - Pradeep Reddy
- Altos Labs, San Diego Institute of Science, San Diego, CA, USA
| | | | | |
Collapse
|
7
|
Lei X, Mao S, Li Y, Huang S, Li J, Du W, Kuang C, Yuan K. ERVcancer: a web resource designed for querying activation of human endogenous retroviruses across major cancer types. J Genet Genomics 2025; 52:583-591. [PMID: 39265822 DOI: 10.1016/j.jgg.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 09/14/2024]
Abstract
Human endogenous retroviruses (HERVs) comprise approximately 8% of the human genome, integrated into the dynamic regulatory network of cellular potency during early embryonic development. In recent studies, resurgent the transcriptional activity of HERVs has been frequently observed in many types of human cancers, suggesting their potential functions in the occurrence and progression of malignancy. However, a dedicated web resource for querying the relationship between the activation of HERVs and cancer development is lacking. Here, we construct a database to explore the sequence information, expression profiles, survival prognosis, and genetic interactions of HERVs in diverse cancer types. Our database currently contains RNA sequencing data of 580 HERVs across 16,246 samples, including that of 6478 tumoral and 634 normal tissues, 932 cancer cell lines, as well as 151 early embryonic and 8051 human adult tissues. The primary goal is to provide an easily accessible and user-friendly database for professionals in the fields of bioinformatics, pathology, pharmacology, and related areas, enabling them to efficiently screen the activity of HERVs of interest in normal and cancerous tissues and evaluate the clinical relevance. The ERVcancer database is available at http://kyuanlab.com/ervcancer/.
Collapse
Affiliation(s)
- Xiaoyun Lei
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Guangxi Health Commission Key Laboratory of Medical Genetics and Genomics, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, China
| | - Song Mao
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yinshuang Li
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Shi Huang
- Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410000, China
| | - Jinchen Li
- Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410000, China
| | - Wei Du
- Department of Pathology, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan 415000, China
| | - Chunmei Kuang
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Kai Yuan
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410000, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Furong Laboratory, Changsha, Hunan 410000, China; The Biobank of Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| |
Collapse
|
8
|
Dickter JK, Zhao Y, Parekh V, Ma H, Modi BG, Li WY, Armenian SH, Wu X, Abdulla FR. Mining metagenomes and metatranscriptomes unveils viruses associated with cutaneous squamous cell carcinoma in hematopoietic stem cell transplant recipients. Microbiol Spectr 2025; 13:e0005325. [PMID: 40162769 PMCID: PMC12054082 DOI: 10.1128/spectrum.00053-25] [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: 01/07/2025] [Accepted: 03/07/2025] [Indexed: 04/02/2025] Open
Abstract
We investigated the presence of viral DNA and RNA in cutaneous squamous cell carcinoma (cSCC) tumor and normal tissues from nine individuals with a history of hematopoietic stem cell transplantation (HCT). Microbiome quantification through DNA and RNA sequencing (RNA-seq) revealed the presence of 18 viruses in both tumor and normal tissues. DNA sequencing (DNA-seq) identified Torque teno virus, Saimiriine herpesvirus 1, Merkel cell polyomavirus, Human parvovirus B19, Human gammaherpesvirus-4, Human herpesvirus-6, and others. RNA-seq revealed additional viruses such as Tobamovirus, Pinus nigra virus, Orthohepadnavirus, Human papillomavirus-5, Human herpesvirus-7, Human gammaherpesvirus-4, Gammaretrovirus, and others. Notably, DNA-seq indicated that tumor samples exhibited low levels of Escherichia virus in three out of nine subjects and elevated levels of Human gammaherpesvirus-4 in one subject, while normal samples frequently contained Gammaretrovirus and occasionally Escherichia virus. A comparative analysis using both DNA- and RNA-seq captured three common viruses: Abelson murine leukemia virus, Murine type C retrovirus, and Human gammaherpesvirus-4. These findings were corroborated by an independent data set, supporting the reliability of the viral detection methods utilized. The study provides insights into the viral landscape in post-HCT patients, emphasizing the need for comprehensive viral monitoring in this vulnerable population. IMPORTANCE This study is important because it explores the potential role of viruses in the development of cSCC in individuals who have undergone allogeneic HCT. cSCC is common in this population, particularly in those with chronic graft-versus-host disease on long-term immunosuppression. By using advanced metagenomic and metatranscriptomic next-generation sequencing, we aimed to identify viral pathogens present in tumor and adjacent normal tissue. The results could lead to targeted preventive or therapeutic interventions for these high-risk people, potentially improving their outcomes and management of cSCC.
Collapse
Affiliation(s)
- Jana K. Dickter
- Department of Medicine, Division of Infectious Diseases, City of Hope National Medical Center, Duarte, California, USA
| | - Yuqi Zhao
- Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Vishwas Parekh
- Department of Pathology, City of Hope National Medical Center, Duarte, California, USA
| | - Huiyan Ma
- Department of Computational and Quantitative Medicine, City of Hope National Medical Center, Duarte, California, USA
| | - Badri G. Modi
- Department of Surgery, Division of Dermatology, City of Hope National Medical Center, Duarte, California, USA
| | - Wai-Yee Li
- Department of Surgery, Division of Plastic Surgery, City of Hope National Medical Center, Duarte, California, USA
| | - Saro H. Armenian
- Department of Population Sciences, Department of Pediatrics, City of Hope National Medical Center, Duarte, California, USA
| | - Xiwei Wu
- Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Farah R. Abdulla
- Department of Surgery, Division of Dermatology, City of Hope National Medical Center, Duarte, California, USA
| |
Collapse
|
9
|
Ferreira CS, Da Silva AT, Brustolini OJB, Soares BRP, Manuli ER, Ramundo MS, Paranhos-Baccala G, Sabino EC, Vasconcelos ATR. Immune and vascular modulation by HERVs: the role of CXCR1 and IL18RAP in dengue severity progression. Front Immunol 2025; 16:1557588. [PMID: 40124360 PMCID: PMC11925782 DOI: 10.3389/fimmu.2025.1557588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 02/17/2025] [Indexed: 03/25/2025] Open
Abstract
Introduction Human Endogenous Retroviruses (HERVs), which can be activated by viral infections, have complex roles in gene regulation and immune modulation. However, their contribution to disease progression is not yet fully understood. Dengue fever ranges from mild symptoms to severe cases characterized by plasma leakage and immune dysregulation, providing a relevant context to investigate these interactions. Methods This study comes up with a comprehensive analysis of differentially expressed HERVs (DE-HERVs), protein-coding genes (DEGs), and regulatory elements such as microRNAs (DE-miRNA) and non-LTR retroviruses (DE-LINEs and DE-SINEs) derived from the transcriptomes of Brazilian dengue patients across different disease stages. Results The results show that DE-HERVs are associated with key genes identified in severe dengue cases, including ARG1, SLC15A2, COL3A1, SVEP1, CH25H, CST7, CXCR1, IL18RAP, SORL1, and TACR1, suggesting their role in immune modulation and endothelial permeability. Specifically, the upregulation of CXCR1 and IL18RAP genes in patients who progressed to severe dengue correlates with a complex regulatory network involving down-regulated microRNAs (miRNAs) and non-LTR retroviruses, emphasizing their relevance to inflammation and vascular permeability. MicroRNAs and non-LTR retroviruses were found to regulate these genes differently across dengue stages, with non-LTR elements appearing predominantly in non-severe cases and miRNA expression profiles varying across the comparison groups. Discussion These findings improve our understanding of the molecular mechanisms underlying dengue progression and suggest that HERV-related regulatory networks may influence viral infections. Further research is required to clarify the specific roles of HERVs in dengue pathogenesis.
Collapse
Affiliation(s)
- Cristina Santos Ferreira
- Laboratório de Bioinformática, Laboratório Nacional de Computação Científica (LNCC/MCTIC), Rio de Janeiro, Brazil
| | - Alan Tardin Da Silva
- Laboratório de Bioinformática, Laboratório Nacional de Computação Científica (LNCC/MCTIC), Rio de Janeiro, Brazil
| | | | - Beatriz Rodrigues Pellegrina Soares
- Departamento de Moléstias Infecciosas e Parasitárias, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Instituto de Medicina Tropical, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Erika Regina Manuli
- Departamento de Moléstias Infecciosas e Parasitárias, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Instituto de Medicina Tropical, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Universidade Municipal de São Caetano do Sul, São Caetano do Sul, Brazil
| | - Mariana Severo Ramundo
- Departamento de Clínica Médica, Disciplina de Imunologia Clínica e Alergia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | | | - Ester Cerdeira Sabino
- Universidade Municipal de São Caetano do Sul, São Caetano do Sul, Brazil
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | | |
Collapse
|
10
|
Jia M, Han S, Li L, Fu Y, Zhou D. Interferon-Stimulated Genes: Novel Targets in Renal Pathogenesis. KIDNEY DISEASES (BASEL, SWITZERLAND) 2025; 11:390-401. [PMID: 40519215 PMCID: PMC12165645 DOI: 10.1159/000546141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 04/18/2025] [Indexed: 06/18/2025]
Abstract
Background Kidney diseases are a prevalent global health concern, and despite ongoing research, there remains a lack of fully effective clinical treatments to prevent or halt their progression. Consequently, it is encouraged to identify novel biomarkers, establish early diagnostic methods, pinpoint key molecular pathways, and develop innovative therapeutic targets for more effective management of renal disorders. Summary Interferons (IFNs), a group of cytokines, play pivotal roles in immune responses, particularly in antiviral and antiproliferative activities. IFNs trigger a cascade of signaling events that lead to the induction of interferon-stimulated genes (ISGs), which are essential for controlling viral infections and regulating immune responses. This review explores the impact of interferon-related genes on renal disorders, focusing on the mechanisms, therapeutic approaches, and consequences of enhanced interferon signaling in the kidney. Key Messages Most diagnostic and therapeutic strategies targeting ISGs are still far from clinical implementation. The better understanding of ISG-regulated pathophysiology and the progress of new intervention approaches are expected to facilitate the clinical translation of ISGs-based diagnosis and therapy of kidney diseases.
Collapse
Affiliation(s)
- Meng Jia
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shuangxu Han
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Liang Li
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yi Fu
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Di Zhou
- Division of Life Sciences and Medicine, Department of Phase I Clinical Trials Laboratory, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| |
Collapse
|
11
|
Turčić M, Kraljević Pavelić S, Trivanović D, Pavelić K. Interaction of HERVs with PAMPs in Dysregulation of Immune Response Cascade Upon SARS-CoV-2 Infections. Int J Mol Sci 2024; 25:13360. [PMID: 39769125 PMCID: PMC11677760 DOI: 10.3390/ijms252413360] [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: 10/27/2024] [Revised: 11/20/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Human endogenous retroviruses (HERVs) are genomic fragments integrated into human DNA from germline infections by exogenous retroviruses that threatened primates early in their evolution and are inherited vertically in the germline. So far, HERVs have been studied in the context of extensive immunopathogenic, neuropathogenic and even oncogenic effects within their host. In particular, in our paper, we elaborate on the aspects related to the possible correlation of transposable HERV elements' activation and SARS-CoV-2 spike protein's presence in cells of COVID-19 patients or upon COVID-19 vaccination with implications for natural and adaptive immunity. In particular, the release of cytokines TNF-α, IL-1β and IL-6 occurs in such cases and plays a notable role in sustaining chronic inflammation. Moreover, well-known interindividual variations of HERVs might partially account for the interpersonal variability of COVID-19 symptoms or unwanted events post-vaccination. Accordingly, further studies are required to clarify the SARS-CoV-2 spike protein's role in triggering HERVs.
Collapse
Affiliation(s)
- Marijana Turčić
- Teaching Institute of Public Health of Primorsko-Goranska County, Krešimirova 52a, 51000 Rijeka, Croatia;
| | - Sandra Kraljević Pavelić
- Faculty of Health Studies, University of Rijeka, Ulica Viktora Cara Emina 5, 51000 Rijeka, Croatia
| | - Dragan Trivanović
- Faculty of Medicine, Juraj Dobrila University of Pula, Zagrebačka 30, 52100 Pula, Croatia;
- Opća Bolnica Pula, Santoriova Ul. 24a, 52100 Pula, Croatia
| | - Krešimir Pavelić
- Faculty of Medicine, Juraj Dobrila University of Pula, Zagrebačka 30, 52100 Pula, Croatia;
- International Academy of Science, Arts and Religion, Radnička Cesta, 71000 Sarajevo, Bosnia and Herzegovina
| |
Collapse
|
12
|
Strum S, Evdokimova V, Radvanyi L, Spreafico A. Extracellular Vesicles and Their Applications in Tumor Diagnostics and Immunotherapy. Cells 2024; 13:2031. [PMID: 39682778 PMCID: PMC11639792 DOI: 10.3390/cells13232031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024] Open
Abstract
Extracellular vesicles (EVs) are cell-derived nanoparticles that have attracted significant attention in the investigation of human health and disease, including cancer biology and its clinical management. Concerning cancer, EVs have been shown to influence numerous aspects of oncogenesis, including tumor proliferation and metastasis. EVs can augment the immune system and have been implicated in virtually all aspects of innate and adaptive immunity. With immunotherapy changing the landscape of cancer treatment across multiple disease sites, it is paramount to understand their mechanisms of action and to further improve upon their efficacy. Despite a rapidly growing body of evidence supporting of the utility of EVs in cancer diagnostics and therapeutics, their application in clinical trials involving solid tumors and immunotherapy remains limited. To date, relatively few trials are known to incorporate EVs in this context, mainly employing them as biomarkers. To help address this gap, this review summarizes known applications of EVs in clinical trials and provides a brief overview of the roles that EVs play in cancer biology, immunology, and their proposed implications in immunotherapy. The impetus to leverage EVs in future clinical trials and correlative studies is crucial, as they are ideally positioned to synergize with advancements in multi-omics research to further therapeutic discovery and our understanding of cancer biology.
Collapse
Affiliation(s)
- Scott Strum
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
- Division of Medical Oncology and Hematology, Department of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada
| | | | - Laszlo Radvanyi
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Anna Spreafico
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
- Division of Medical Oncology and Hematology, Department of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada
| |
Collapse
|
13
|
Fiordoro S, Rosano C, Pechkova E, Barocci S, Izzotti A. Epigenetic modulation of immune cells: Mechanisms and implications. Adv Biol Regul 2024; 94:101043. [PMID: 39305736 DOI: 10.1016/j.jbior.2024.101043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/25/2024] [Accepted: 08/02/2024] [Indexed: 12/12/2024]
Abstract
Epigenetic modulation of the immune response entails modifiable and inheritable modifications that do not modify the DNA sequence. While there have been many studies on epigenetic changes in tumor cells, there is now a growing focus on epigenetically mediated changes in immune cells of both the innate and adaptive systems. These changes have significant implications for both the body's response to tumors and the development of potential therapeutic vaccines. This study primarily discusses the key epigenetic alterations, with a specific emphasis on pseudouridination, as well as non-coding RNAs and their transportation, which can lead to the development of cancer and the acquisition of new phenotypic traits by immune cells. Furthermore, the advancement of therapeutic vaccinations targeting the tumor will be outlined.
Collapse
Affiliation(s)
- S Fiordoro
- Department of Health Sciences, University of Genova, Via Pastore 1, 16132 Genova, Italy
| | - C Rosano
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy.
| | - E Pechkova
- Department of Experimental Medicine, University of Genoa, 16132 Genova, Italy
| | - S Barocci
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - A Izzotti
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy; Department of Experimental Medicine, University of Genoa, 16132 Genova, Italy
| |
Collapse
|
14
|
Belizário J, Garay-Malpartida M. Key Epigenetic Players in Etiology and Novel Combinatorial Therapies for Treatment of Hepatocellular Carcinoma. LIVERS 2024; 4:638-655. [DOI: 10.3390/livers4040044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of death in which the molecular tumorigenesis and cellular heterogeneity are poorly understood. The genetic principle that specific driver mutations in oncogenes, DNA repair genes, and tumor-suppressor genes can independently drive cancer development has been widely explored. Additionally, a repertory of harmful epigenetic modifications in DNA and chromatin—impacting the expression of genes involved in cellular proliferation, differentiation, genome stability, cell-cycle control, and DNA repair—are now acknowledged across various biological contexts that contribute to cancer etiology. Notably, the dynamic hypermethylation and hypomethylation in enhancer and promoter regions that promote activation or silencing of the master regulatory genes of the epigenetic programs is often altered in tumor cells due to mutation. Genome instability is one of the cancer hallmarks that contribute to transdifferentiation and intratumoral heterogeneity. Thus, it is broadly accepted that tumor tissue is dominated by genetically and epigenetically distinct sub-clones which display a set of genetic and epigenetic mutations. Here we summarize some functions of key genetic and epigenetic players and biochemical pathways leading to liver cell transformation. We discuss the role of the potential epigenetic marks in target genes thought to be involved in sequential events following liver lipid metabolism dysregulation, inflammation, fibrosis, cirrhosis, and finally hepatocellular carcinoma. We also briefly describe new findings showing how epigenetic drugs together with chemotherapy and immunotherapy can improve overall responses in patients with hepatic tumors.
Collapse
Affiliation(s)
- José Belizário
- School of Arts, Sciences and Humanities of the University of Sao Paulo, Rua Arlindo Bettio, 1000, São Paulo 03828-000, Brazil
| | - Miguel Garay-Malpartida
- School of Arts, Sciences and Humanities of the University of Sao Paulo, Rua Arlindo Bettio, 1000, São Paulo 03828-000, Brazil
| |
Collapse
|
15
|
Camp SY, He MX, Cuoco MS, Saad E, Pimenta E, Meli K, Bakouny Z, Labaki C, Titchen BM, Kang YJ, Horst J, Trowbridge R, Shannon E, Helvie K, Thorner AR, Vigneau S, Mayorga A, Kodali J, Lachmayr H, Bemus M, Park J, Choueiri T, Bi K, Van Allen EM. Single-cell epigenetic profiling reveals an interferon response-high program associated with BAP1 deficiency in kidney cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.15.623837. [PMID: 39605676 PMCID: PMC11601305 DOI: 10.1101/2024.11.15.623837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Renal cell carcinoma (RCC) is characterized by recurrent somatic mutations in epigenetic regulators, which stratify patients into clinically significant subgroups with distinct prognoses and treatment responses. However, the cell type-specific epigenetic landscape of RCC-broadly and in the context of these mutations-is incompletely understood. To investigate these open questions, we integrated single nucleus ATAC sequencing data from RCC tumors across four independent cohorts. In clear cell RCC tumors, we identified four shared malignant epigenetic programs related to angiogenesis, proximal tubule-like features, interferon (IFN) signaling, and one that lacked distinct genomic regions with increased accessibility. Among the mutated epigenetic regulators, BAP1 mutation exhibited the most significant impact on chromatin accessibility in tumor cells, and the associated epigenetic changes were linked to IFN response. We identify multiple potential sources of elevated IFN signaling in these lesions, such as increased immune infiltration and increased accessibility and expression of an IFN-associated ERV, ERV3-16A3_LTR. We find that the expression of ERV3-16A3_LTR may itself be a negative prognostic biomarker in ccRCC. Our findings highlight the convergence of malignant epigenetic programs across ccRCC tumors and suggest that BAP1 loss, potentially through ERV3-16A3_LTR dysregulation, is associated with an IFN response-high epigenetic program.
Collapse
Affiliation(s)
- Sabrina Y. Camp
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Meng Xiao He
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Harvard Graduate Program in Biophysics, Boston, MA 02115, USA
| | - Michael S. Cuoco
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, CA 92093, USA
| | - Eddy Saad
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Erica Pimenta
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Kevin Meli
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Harvard Division of Medical Sciences PhD Program in Biological and Biomedical Sciences, Boston, MA 02115, USA
| | - Ziad Bakouny
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Chris Labaki
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Breanna M. Titchen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Harvard Division of Medical Sciences PhD Program in Biological and Biomedical Sciences, Boston, MA 02115, USA
| | - Yun Jee Kang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Department of Surgery, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Jack Horst
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Rachel Trowbridge
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Erin Shannon
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Karla Helvie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Aaron R. Thorner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Sébastien Vigneau
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Angie Mayorga
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Jahnavi Kodali
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Hannah Lachmayr
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Meredith Bemus
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Jihye Park
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Toni Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Kevin Bi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Eliezer M. Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Parker Institute for Cancer Immunotherapy, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| |
Collapse
|
16
|
Studstill C, Huang N, Sundstrom S, Moscoso S, Zhang H, Damania B, Moody C. Apoptotic Caspases Suppress Expression of Endogenous Retroviruses in HPV31+ Cells That Are Associated with Activation of an Innate Immune Response. Viruses 2024; 16:1695. [PMID: 39599810 PMCID: PMC11598866 DOI: 10.3390/v16111695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/25/2024] [Accepted: 10/26/2024] [Indexed: 11/29/2024] Open
Abstract
Avoidance of an immune response is critical to completion of the human papillomavirus (HPV) life cycle, which occurs in the stratified epithelium and is linked to epithelial differentiation. We previously demonstrated that high-risk HPVs use apoptotic caspases to suppress an antiviral innate immune response during the productive phase of the life cycle. We found that caspase-8 and caspase-3 suppress a type I IFN-β and type III IFN-λ response by disabling the MDA5/MAVS double-stranded RNA (dsRNA) sensing pathway, indicating that immunogenic RNAs increase upon differentiation in HPV+ cells. In this study, we demonstrate that caspase inhibition results in aberrant transcription of a subset of endogenous retroviruses (ERVs) that have been shown to activate an IFN response through dsRNA-sensing pathways. We show that the increase in ERV transcription is accompanied by an enrichment in dsRNA formation. Additionally, we demonstrate that the robust increase in ERV expression requires activation of the JAK/STAT-signaling pathway, indicating that this subset of ERVs is IFN-inducible. Overall, these results suggest a model by which caspase activity blocks the reactivation of ERVs through the JAK/STAT pathway, protecting HPV+ cells from an increase in immunogenic dsRNAs that otherwise would trigger an IFN response that inhibits productive viral replication.
Collapse
Affiliation(s)
- Caleb Studstill
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ning Huang
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Shelby Sundstrom
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Samantha Moscoso
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Huirong Zhang
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Blossom Damania
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Cary Moody
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
17
|
de Azevedo SSD, Ribeiro-Alves M, Côrtes FH, Delatorre E, Hoagland B, Villela LM, Grinsztejn B, Veloso VG, Morgado MG, Souza TML, Bello G. HIV-1 controllers exhibit an enhanced antiretroviral innate state characterised by overexpression of p21 and MCPIP1 and silencing of ERVK-6 RNA expression. Mem Inst Oswaldo Cruz 2024; 119:e240071. [PMID: 39292108 PMCID: PMC11404982 DOI: 10.1590/0074-02760240071] [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: 04/01/2024] [Accepted: 07/12/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Human immunodeficiency virus (HIV)-1 infection can activate the expression of human endogenous retroviruses (HERVs), particularly HERV-K (HML-2). HIV controllers (HICs) are rare people living with HIV (PLWHs) who naturally control HIV-1 replication and overexpress some cellular restriction factors that negatively regulate the LTR-driven transcription of HIV-1 proviruses. OBJECTIVES To understand the ability of HICs to control the expression of endogenous retroviruses. METHODS We measured endogenous retrovirus type K6 (ERVK-6) RNA expression in peripheral blood mononuclear cells (PBMCs) of HICs (n = 23), antiretroviral (ART)-suppressed subjects (n = 8), and HIV-1-negative (NEG) individuals (n = 10) and correlated the transcript expression of ERVK-6 with multiple HIV-1 cellular restriction factors. FINDINGS Our study revealed that ERVK-6 RNA expression in PBMCs from HICs was significantly downregulated compared with that in both the ART and NEG control groups. Moreover, we detected that ERVK-6 RNA levels in PBMCs across all groups were negatively correlated with the expression levels of p21 and MCPIP1, two cellular restriction factors that limit the activation of macrophages and T cells by downregulating the activity of NF-kB. MAIN CONCLUSIONS These findings support the hypothesis that HICs activate innate antiviral mechanisms that may simultaneously downregulate the transcription of both exogenous (HIV-1) and endogenous (ERVK-6) retroviruses. Future studies with larger cohorts should be performed to confirm this hypothesis and to explore the role of p21 and MCPIP1 in regulating HERV-K expression in physiological and pathological conditions.
Collapse
Affiliation(s)
| | - Marcelo Ribeiro-Alves
- Fundação Oswaldo Cruz-Fiocruz, Instituto Nacional de Infectologia Evandro Chagas, Laboratório de Pesquisa Clínica em DST/AIDS, Rio de Janeiro, RJ, Brasil
| | - Fernanda Heloise Côrtes
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de AIDS & Imunologia Molecular, Rio de Janeiro, RJ, Brasil
| | - Edson Delatorre
- Universidade Federal do Espírito Santo, Centro de Ciências da Saúde, Departamento de Patologia, Laboratório de Genômica e Ecologia Viral, Vitória, ES, Brasil
| | - Brenda Hoagland
- Fundação Oswaldo Cruz-Fiocruz, Instituto Nacional de Infectologia Evandro Chagas, Laboratório de Pesquisa Clínica em DST/AIDS, Rio de Janeiro, RJ, Brasil
| | - Larissa M Villela
- Fundação Oswaldo Cruz-Fiocruz, Instituto Nacional de Infectologia Evandro Chagas, Laboratório de Pesquisa Clínica em DST/AIDS, Rio de Janeiro, RJ, Brasil
| | - Beatriz Grinsztejn
- Fundação Oswaldo Cruz-Fiocruz, Instituto Nacional de Infectologia Evandro Chagas, Laboratório de Pesquisa Clínica em DST/AIDS, Rio de Janeiro, RJ, Brasil
| | - Valdilea Gonçalvez Veloso
- Fundação Oswaldo Cruz-Fiocruz, Instituto Nacional de Infectologia Evandro Chagas, Laboratório de Pesquisa Clínica em DST/AIDS, Rio de Janeiro, RJ, Brasil
| | - Mariza G Morgado
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de AIDS & Imunologia Molecular, Rio de Janeiro, RJ, Brasil
| | - Thiago Moreno L Souza
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Imunofarmacologia, Rio de Janeiro, RJ, Brasil
- Fundação Oswaldo Cruz-Fiocruz, Instituto Nacional de Ciência e Tecnologia de Inovação em Doenças de Populações Negligenciadas, Rio de Janeiro, RJ, Brasil
| | - Gonzalo Bello
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de AIDS & Imunologia Molecular, Rio de Janeiro, RJ, Brasil
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Arbovírus e Vírus Hemorrágicos, Rio de Janeiro, RJ, Brasil
| |
Collapse
|
18
|
Rodrigues CP, Collins JM, Yang S, Martinez C, Kim JW, Lama C, Nam AS, Alt C, Lin C, Zon LI. Transcripts of repetitive DNA elements signal to block phagocytosis of hematopoietic stem cells. Science 2024; 385:eadn1629. [PMID: 39264994 PMCID: PMC12012832 DOI: 10.1126/science.adn1629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 04/09/2024] [Accepted: 07/04/2024] [Indexed: 09/14/2024]
Abstract
Macrophages maintain hematopoietic stem cell (HSC) quality by assessing cell surface Calreticulin (Calr), an "eat-me" signal induced by reactive oxygen species (ROS). Using zebrafish genetics, we identified Beta-2-microglobulin (B2m) as a crucial "don't eat-me" signal on blood stem cells. A chemical screen revealed inducers of surface Calr that promoted HSC proliferation without triggering ROS or macrophage clearance. Whole-genome CRISPR-Cas9 screening showed that Toll-like receptor 3 (Tlr3) signaling regulated b2m expression. Targeting b2m or tlr3 reduced the HSC clonality. Elevated B2m levels correlated with high expression of repetitive element (RE) transcripts. Overall, our data suggest that RE-associated double-stranded RNA could interact with TLR3 to stimulate surface expression of B2m on hematopoietic stem and progenitor cells. These findings suggest that the balance of Calr and B2m regulates macrophage-HSC interactions and defines hematopoietic clonality.
Collapse
Affiliation(s)
- Cecilia Pessoa Rodrigues
- Howard Hughes Medical Institute, Boston Children’s Hospital Boston, MA, USA
- Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA, USA
| | - Joseph M. Collins
- Howard Hughes Medical Institute, Boston Children’s Hospital Boston, MA, USA
- Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA, USA
| | - Song Yang
- Howard Hughes Medical Institute, Boston Children’s Hospital Boston, MA, USA
| | - Catherine Martinez
- Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA, USA
| | - Ji Wook Kim
- Howard Hughes Medical Institute, Boston Children’s Hospital Boston, MA, USA
- Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA, USA
| | - Chhiring Lama
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Anna S. Nam
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Clemens Alt
- Wellman Center for Photomedicine, Mass General Research Institute, Boston, MA, USA
| | - Charles Lin
- Wellman Center for Photomedicine, Mass General Research Institute, Boston, MA, USA
- Center for Systems Biology, Massachusetts General Hospital, MA, USA
| | - Leonard I. Zon
- Howard Hughes Medical Institute, Boston Children’s Hospital Boston, MA, USA
- Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA, USA
| |
Collapse
|
19
|
Shi G, Pang Q, Lin Z, Zhang X, Huang K. Repetitive Sequence Stability in Embryonic Stem Cells. Int J Mol Sci 2024; 25:8819. [PMID: 39201503 PMCID: PMC11354519 DOI: 10.3390/ijms25168819] [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: 07/10/2024] [Revised: 08/07/2024] [Accepted: 08/12/2024] [Indexed: 09/02/2024] Open
Abstract
Repetitive sequences play an indispensable role in gene expression, transcriptional regulation, and chromosome arrangements through trans and cis regulation. In this review, focusing on recent advances, we summarize the epigenetic regulatory mechanisms of repetitive sequences in embryonic stem cells. We aim to bridge the knowledge gap by discussing DNA damage repair pathway choices on repetitive sequences and summarizing the significance of chromatin organization on repetitive sequences in response to DNA damage. By consolidating these insights, we underscore the critical relationship between the stability of repetitive sequences and early embryonic development, seeking to provide a deeper understanding of repetitive sequence stability and setting the stage for further research and potential therapeutic strategies in developmental biology and regenerative medicine.
Collapse
Affiliation(s)
- Guang Shi
- MOE Key Laboratory of Gene Function and Regulation, Guangzhou Key Laboratory of Healthy Aging Research and SYSU-BCM Joint Research Center, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (Q.P.); (Z.L.); (X.Z.)
| | - Qianwen Pang
- MOE Key Laboratory of Gene Function and Regulation, Guangzhou Key Laboratory of Healthy Aging Research and SYSU-BCM Joint Research Center, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (Q.P.); (Z.L.); (X.Z.)
| | - Zhancheng Lin
- MOE Key Laboratory of Gene Function and Regulation, Guangzhou Key Laboratory of Healthy Aging Research and SYSU-BCM Joint Research Center, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (Q.P.); (Z.L.); (X.Z.)
| | - Xinyi Zhang
- MOE Key Laboratory of Gene Function and Regulation, Guangzhou Key Laboratory of Healthy Aging Research and SYSU-BCM Joint Research Center, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (Q.P.); (Z.L.); (X.Z.)
| | - Kaimeng Huang
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA;
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| |
Collapse
|
20
|
Nevalainen T, Autio-Kimura A, Hurme M. Human endogenous retrovirus W in multiple sclerosis: transcriptional activity is associated with decline in oligodendrocyte proportions in the white matter of the brain. J Neurovirol 2024; 30:393-405. [PMID: 38717678 PMCID: PMC11512866 DOI: 10.1007/s13365-024-01208-9] [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: 08/18/2023] [Revised: 01/22/2024] [Accepted: 04/03/2024] [Indexed: 10/28/2024]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease. One of the basic mechanisms in this disease is the autoimmune response against the myelin sheet leading to axonal damage. There is strong evidence showing that this response is regulated by both genetic and environmental factors. In addition, the role of viruses has been extensively studied, especially in the case of human endogenous retroviruses (HERVs). However, although several associations with MS susceptibility, especially in the case of HERV-W family have been observed, the pathogenic mechanisms have remained enigmatic. To clarify these HERV-mediated mechanisms as well as the responsible HERV-W loci, we utilized RNA sequencing data obtained from the white matter of the brain of individuals with and without MS. CIBERSORTx tool was applied to estimate the proportions of neuronal, glial, and endothelial cells in the brain. In addition, the transcriptional activity of 215 HERV-W loci were analyzed. The results indicated that 65 HERV-W loci had detectable expression, of which 14 were differentially expressed between MS and control samples. Of these, 12 HERV-W loci were upregulated in MS. Expression levels of the 8 upregulated HERV-W loci had significant negative correlation with estimated oligodendrocyte proportions, suggesting that they are associated with the dynamics of oligodendrocyte generation and/or maintenance. Furthermore, Gene Set Enrichment Analysis (GSEA) results indicated that expression levels of three upregulated HERV-W loci: 2p16.2, 2q13, and Xq13.3, are associated with suppression of oligodendrocyte development and myelination. Taken together, these data suggest new HERV-W loci candidates that might take part in MS pathogenesis.
Collapse
Affiliation(s)
- Tapio Nevalainen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland.
- Gerontology Research Center (GEREC), Tampere, Finland.
- Tampere University Hospital, Tampere, Finland.
| | - Arttu Autio-Kimura
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
- Gerontology Research Center (GEREC), Tampere, Finland
| | - Mikko Hurme
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
- Gerontology Research Center (GEREC), Tampere, Finland
- Tampere University Hospital, Tampere, Finland
| |
Collapse
|
21
|
Lai TJ, Sun L, Li K, Prins TJ, Treger J, Li T, Sun MZ, Nathanson DA, Liau LM, Lai A, Prins RM, Everson RG. Epigenetic Induction of Cancer-Testis Antigens and Endogenous Retroviruses at Single-Cell Level Enhances Immune Recognition and Response in Glioma. CANCER RESEARCH COMMUNICATIONS 2024; 4:1834-1849. [PMID: 38856710 PMCID: PMC11275559 DOI: 10.1158/2767-9764.crc-23-0566] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/22/2024] [Accepted: 06/05/2024] [Indexed: 06/11/2024]
Abstract
Glioblastoma (GBM) is the most common malignant primary brain tumor and remains incurable. Previous work has shown that systemic administration of Decitabine (DAC) induces sufficient expression of cancer-testis antigens (CTA) in GBM for targeting by adoptive T-cell therapy in vivo. However, the mechanisms by which DAC enhances immunogenicity in GBM remain to be elucidated. Using New York esophageal squamous cell carcinoma 1 (NY-ESO-1) as a representative inducible CTA, we demonstrate in patient tissue, immortalized glioma cells, and primary patient-derived gliomaspheres that basal CTA expression is restricted by promoter hypermethylation in gliomas. DAC treatment of glioma cells specifically inhibits DNA methylation silencing to render NY-ESO-1 and other CTA into inducible tumor antigens at single-cell resolution. Functionally, NY-ESO-1 T-cell receptor-engineered effector cell targeting of DAC-induced antigen in primary glioma cells promotes specific and polyfunctional T-cell cytokine profiles. In addition to induction of CTA, DAC concomitantly reactivates tumor-intrinsic human endogenous retroviruses, interferon response signatures, and MHC-I. Overall, we demonstrate that DAC induces targetable tumor antigen and enhances T-cell functionality against GBM, ultimately contributing to the improvement of targeted immune therapies in glioma. SIGNIFICANCE This study dissects the tumor-intrinsic epigenetic and transcriptional mechanisms underlying enhanced T-cell functionality targeting decitabine-induced cancer-testis antigens in glioma. Our findings demonstrate concomitant induction of tumor antigens, reactivation of human endogenous retroviruses, and stimulation of interferon signaling as a mechanistic rationale to epigenetically prime human gliomas to immunotherapeutic targeting.
Collapse
Affiliation(s)
- Thomas J. Lai
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.
| | - Lu Sun
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.
| | - Kevin Li
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.
| | - Terry J. Prins
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.
| | - Janet Treger
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.
| | - Tie Li
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.
| | - Matthew Z. Sun
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.
| | - David A. Nathanson
- Department of Medical and Molecular Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California.
| | - Linda M. Liau
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.
- Department of Medical and Molecular Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California.
| | - Albert Lai
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.
- UCLA Neuro-Oncology Program, University of California, Los Angeles, Los Angeles, California.
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California.
| | - Robert M. Prins
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.
- Department of Medical and Molecular Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California.
- Parker Institute for Cancer Immunotherapy, San Francisco, California.
| | - Richard G. Everson
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
22
|
Apostolou E, Rosén A. Epigenetic reprograming in myalgic encephalomyelitis/chronic fatigue syndrome: A narrative of latent viruses. J Intern Med 2024; 296:93-115. [PMID: 38693641 DOI: 10.1111/joim.13792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a chronic disease presenting with severe fatigue, post-exertional malaise, and cognitive disturbances-among a spectrum of symptoms-that collectively render the patient housebound or bedbound. Epigenetic studies in ME/CFS collectively confirm alterations and/or malfunctions in cellular and organismal physiology associated with immune responses, cellular metabolism, cell death and proliferation, and neuronal and endothelial cell function. The sudden onset of ME/CFS follows a major stress factor that, in approximately 70% of cases, involves viral infection, and ME/CFS symptoms overlap with those of long COVID. Viruses primarily linked to ME/CFS pathology are the symbiotic herpesviruses, which follow a bivalent latent-lytic lifecycle. The complex interaction between viruses and hosts involves strategies from both sides: immune evasion and persistence by the viruses, and immune activation and viral clearance by the host. This dynamic interaction is imperative for herpesviruses that facilitate their persistence through epigenetic regulation of their own and the host genome. In the current article, we provide an overview of the epigenetic signatures demonstrated in ME/CFS and focus on the potential strategies that latent viruses-particularly Epstein-Barr virus-may employ in long-term epigenetic reprograming in ME/CFS. Epigenetic studies could aid in elucidating relevant biological pathways impacted in ME/CFS and reflect the physiological variations among the patients that stem from environmental triggers, including exogenous viruses and/or altered viral activity.
Collapse
Affiliation(s)
- Eirini Apostolou
- Division of Cell and Neurobiology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Anders Rosén
- Division of Cell and Neurobiology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
23
|
Del Prete R, Nesta D, Triggiano F, Lorusso M, Garzone S, Vitulano L, Denicolò S, Indraccolo F, Mastria M, Ronga L, Inchingolo F, Aityan SK, Nguyen KCD, Tran TC, Gargiulo Isacco C, Santacroce L. Human Papillomavirus Carcinogenicity and the Need of New Perspectives: Thoughts from a Retrospective Analysis on Human Papillomavirus Outcomes Conducted at the Hospital University of Bari, Apulia, Italy, between 2011 and 2022. Diagnostics (Basel) 2024; 14:968. [PMID: 38732382 PMCID: PMC11083870 DOI: 10.3390/diagnostics14090968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/26/2024] [Accepted: 04/28/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND The current manuscript's aim was to determine the human papillomavirus (HPV) genotype-specific prevalence and distribution among individuals, males, and females, of different ages in the region of Apulia, Italy, highlighting the possible variables involved in the carcinogenicity mechanism. In addition, we proposed two hypothetical models of HPV's molecular dynamics, intending to clarify the impact of prevention and therapeutic strategies, explicitly modeled by recent survey data. METHODS We presented clinical data from 9647 participants tested for either high-risk (HR) or low-risk (LR) HPV at the affiliated Bari Policlinic University Hospital of Bari from 2011 to 2022. HPV DNA detection was performed using nested-polymerase chain reaction (PCR) and multiplex real-time PCR assay. Statistical analysis showed significant associations for all genders and ages and both HR- and LR-HPV types. A major number of significant pairwise associations were detected for the higher-risk types and females and lower-risk types and males. RESULTS The overall prevalence of HPV was 50.5% (n-4.869) vs. 49.5% (n-4.778) of the study population, of which 74.4% (n-3621) were found to be HPV high-risk (HR-HPV) genotypes and 57.7% (n-2.807) low-risk HPV (LR-HPV) genotypes, of which males were 58% and females 49%; the three most prevalent HR-HPV genotypes were HPV 53 (n707-15%), 16 (n704-14%), and 31 (n589-12%), and for LR-HPV, they were 42 (19%), 6 (16%), and 54 (13%); 56% of patients screened for HPV were ≤ 30 years old, 53% were between 31 and 40 years old, 46% were 41-50 and 51-60 years old, and finally, 44% of subjects were >60 years old. CONCLUSIONS Our study provided comprehensive epidemiological data on HPV prevalence and genotype distribution among 9647 participants, which could serve as a significant reference for clinical practice, and it implied the necessity for more effective screening methods for HPV carcinogenesis covering the use of more specific molecular investigations. Although this is a predominantly descriptive and epidemiological study, the data obtained offer not only a fairly unique trend compared to other studies of different realities and latitudes but also lead us to focus on the HPV infection within two groups of young people and adults and hypothesize the possible involvement of dysbiosis, stem cells, and the retrotransposition mechanism.
Collapse
Affiliation(s)
- Raffaele Del Prete
- Department of Interdisciplinary Medicine (DIM), U.O.C. Microbiology and Virology, University-Hospital of Bari, 70100 Bari, Italy; (R.D.P.); (D.N.); (F.T.); (M.L.); (S.G.); (L.V.); (S.D.); (F.I.); (M.M.); (L.R.); (F.I.); (K.C.D.N.); (L.S.)
| | - Daniela Nesta
- Department of Interdisciplinary Medicine (DIM), U.O.C. Microbiology and Virology, University-Hospital of Bari, 70100 Bari, Italy; (R.D.P.); (D.N.); (F.T.); (M.L.); (S.G.); (L.V.); (S.D.); (F.I.); (M.M.); (L.R.); (F.I.); (K.C.D.N.); (L.S.)
| | - Francesco Triggiano
- Department of Interdisciplinary Medicine (DIM), U.O.C. Microbiology and Virology, University-Hospital of Bari, 70100 Bari, Italy; (R.D.P.); (D.N.); (F.T.); (M.L.); (S.G.); (L.V.); (S.D.); (F.I.); (M.M.); (L.R.); (F.I.); (K.C.D.N.); (L.S.)
| | - Mara Lorusso
- Department of Interdisciplinary Medicine (DIM), U.O.C. Microbiology and Virology, University-Hospital of Bari, 70100 Bari, Italy; (R.D.P.); (D.N.); (F.T.); (M.L.); (S.G.); (L.V.); (S.D.); (F.I.); (M.M.); (L.R.); (F.I.); (K.C.D.N.); (L.S.)
| | - Stefania Garzone
- Department of Interdisciplinary Medicine (DIM), U.O.C. Microbiology and Virology, University-Hospital of Bari, 70100 Bari, Italy; (R.D.P.); (D.N.); (F.T.); (M.L.); (S.G.); (L.V.); (S.D.); (F.I.); (M.M.); (L.R.); (F.I.); (K.C.D.N.); (L.S.)
| | - Lorenzo Vitulano
- Department of Interdisciplinary Medicine (DIM), U.O.C. Microbiology and Virology, University-Hospital of Bari, 70100 Bari, Italy; (R.D.P.); (D.N.); (F.T.); (M.L.); (S.G.); (L.V.); (S.D.); (F.I.); (M.M.); (L.R.); (F.I.); (K.C.D.N.); (L.S.)
| | - Sofia Denicolò
- Department of Interdisciplinary Medicine (DIM), U.O.C. Microbiology and Virology, University-Hospital of Bari, 70100 Bari, Italy; (R.D.P.); (D.N.); (F.T.); (M.L.); (S.G.); (L.V.); (S.D.); (F.I.); (M.M.); (L.R.); (F.I.); (K.C.D.N.); (L.S.)
| | - Francesca Indraccolo
- Department of Interdisciplinary Medicine (DIM), U.O.C. Microbiology and Virology, University-Hospital of Bari, 70100 Bari, Italy; (R.D.P.); (D.N.); (F.T.); (M.L.); (S.G.); (L.V.); (S.D.); (F.I.); (M.M.); (L.R.); (F.I.); (K.C.D.N.); (L.S.)
| | - Michele Mastria
- Department of Interdisciplinary Medicine (DIM), U.O.C. Microbiology and Virology, University-Hospital of Bari, 70100 Bari, Italy; (R.D.P.); (D.N.); (F.T.); (M.L.); (S.G.); (L.V.); (S.D.); (F.I.); (M.M.); (L.R.); (F.I.); (K.C.D.N.); (L.S.)
| | - Luigi Ronga
- Department of Interdisciplinary Medicine (DIM), U.O.C. Microbiology and Virology, University-Hospital of Bari, 70100 Bari, Italy; (R.D.P.); (D.N.); (F.T.); (M.L.); (S.G.); (L.V.); (S.D.); (F.I.); (M.M.); (L.R.); (F.I.); (K.C.D.N.); (L.S.)
| | - Francesco Inchingolo
- Department of Interdisciplinary Medicine (DIM), U.O.C. Microbiology and Virology, University-Hospital of Bari, 70100 Bari, Italy; (R.D.P.); (D.N.); (F.T.); (M.L.); (S.G.); (L.V.); (S.D.); (F.I.); (M.M.); (L.R.); (F.I.); (K.C.D.N.); (L.S.)
| | - Sergey K. Aityan
- College of Engineering, Northeastern University, 5000 MacArthur Blvd., Oakland, CA 94613, USA;
| | - Kieu C. D. Nguyen
- Department of Interdisciplinary Medicine (DIM), U.O.C. Microbiology and Virology, University-Hospital of Bari, 70100 Bari, Italy; (R.D.P.); (D.N.); (F.T.); (M.L.); (S.G.); (L.V.); (S.D.); (F.I.); (M.M.); (L.R.); (F.I.); (K.C.D.N.); (L.S.)
| | - Toai Cong Tran
- Department of Basic Medical Sciences and Biomedical Research Center, Pham Ngoc Thach University of Medicine, Ho Chi Minh City 700100, Vietnam;
| | - Ciro Gargiulo Isacco
- Department of Interdisciplinary Medicine (DIM), U.O.C. Microbiology and Virology, University-Hospital of Bari, 70100 Bari, Italy; (R.D.P.); (D.N.); (F.T.); (M.L.); (S.G.); (L.V.); (S.D.); (F.I.); (M.M.); (L.R.); (F.I.); (K.C.D.N.); (L.S.)
| | - Luigi Santacroce
- Department of Interdisciplinary Medicine (DIM), U.O.C. Microbiology and Virology, University-Hospital of Bari, 70100 Bari, Italy; (R.D.P.); (D.N.); (F.T.); (M.L.); (S.G.); (L.V.); (S.D.); (F.I.); (M.M.); (L.R.); (F.I.); (K.C.D.N.); (L.S.)
| |
Collapse
|
24
|
Terrazzan A, Vanini R, Ancona P, Bianchi N, Taccioli C, Aguiari G. State-of-the-art in transposable element modulation affected by drugs in malignant prostatic cancer cells. J Cell Biochem 2024; 125:e30557. [PMID: 38501160 DOI: 10.1002/jcb.30557] [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: 08/30/2023] [Revised: 02/14/2024] [Accepted: 03/07/2024] [Indexed: 03/20/2024]
Abstract
Over recent years, the investigation of transposable elements (TEs) has granted researchers a deeper comprehension of their characteristics and functions, particularly regarding their significance in the mechanisms contributing to cancer development. This manuscript focuses on prostate carcinoma cell lines and offers a comprehensive review intended to scrutinize the associations and interactions between TEs and genes, as well as their response to treatment using various chemical drugs, emphasizing their involvement in cancer progression. We assembled a compendium of articles retrieved from the PubMed database to construct networks demonstrating correlations with genes and pharmaceuticals. In doing so, we linked the transposition of certain TE types to the expression of specific transcripts directly implicated in carcinogenesis. Additionally, we underline that treatment employing different drugs revealed unique patterns of TE reactivation. Our hypothesis gathers the current understanding and guides research toward evidence-based investigations, emphasizing the association between antiviral drugs, chemotherapy, and the reduced expression of TEs in patients affected by prostate cancer.
Collapse
Affiliation(s)
- Anna Terrazzan
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
- Laboratory for Advanced Therapy Technologies (LTTA), University of Ferrara, Ferrara, Italy
| | - Riccardo Vanini
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Pietro Ancona
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Nicoletta Bianchi
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Cristian Taccioli
- Department of Animal Medicine, Production and Health (MAPS), University of Padua, Padua, Italy
| | - Gianluca Aguiari
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| |
Collapse
|
25
|
Ruzanov P, Evdokimova V, Pachva MC, Minkovich A, Zhang Z, Langman S, Gassmann H, Thiel U, Orlic-Milacic M, Zaidi SH, Peltekova V, Heisler LE, Sharma M, Cox ME, McKee TD, Zaidi M, Lapouble E, McPherson JD, Delattre O, Radvanyi L, Burdach SE, Stein LD, Sorensen PH. Oncogenic ETS fusions promote DNA damage and proinflammatory responses via pericentromeric RNAs in extracellular vesicles. J Clin Invest 2024; 134:e169470. [PMID: 38530366 PMCID: PMC11060741 DOI: 10.1172/jci169470] [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: 02/09/2023] [Accepted: 03/12/2024] [Indexed: 03/28/2024] Open
Abstract
Aberrant expression of the E26 transformation-specific (ETS) transcription factors characterizes numerous human malignancies. Many of these proteins, including EWS:FLI1 and EWS:ERG fusions in Ewing sarcoma (EwS) and TMPRSS2:ERG in prostate cancer (PCa), drive oncogenic programs via binding to GGAA repeats. We report here that both EWS:FLI1 and ERG bind and transcriptionally activate GGAA-rich pericentromeric heterochromatin. The respective pathogen-like HSAT2 and HSAT3 RNAs, together with LINE, SINE, ERV, and other repeat transcripts, are expressed in EwS and PCa tumors, secreted in extracellular vesicles (EVs), and are highly elevated in plasma of patients with EwS with metastatic disease. High human satellite 2 and 3 (HSAT2,3) levels in EWS:FLI1- or ERG-expressing cells and tumors were associated with induction of G2/M checkpoint, mitotic spindle, and DNA damage programs. These programs were also activated in EwS EV-treated fibroblasts, coincident with accumulation of HSAT2,3 RNAs, proinflammatory responses, mitotic defects, and senescence. Mechanistically, HSAT2,3-enriched cancer EVs induced cGAS-TBK1 innate immune signaling and formation of cytosolic granules positive for double-strand RNAs, RNA-DNA, and cGAS. Hence, aberrantly expressed ETS proteins derepress pericentromeric heterochromatin, yielding pathogenic RNAs that transmit genotoxic stress and inflammation to local and distant sites. Monitoring HSAT2,3 plasma levels and preventing their dissemination may thus improve therapeutic strategies and blood-based diagnostics.
Collapse
Affiliation(s)
- Peter Ruzanov
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | | | - Manideep C. Pachva
- Department of Molecular Oncology, British Columbia Cancer Research Centre and
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Alon Minkovich
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Zhenbo Zhang
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Sofya Langman
- Department of Molecular Oncology, British Columbia Cancer Research Centre and
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hendrik Gassmann
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Uwe Thiel
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | | | - Syed H. Zaidi
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Vanya Peltekova
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | | | - Manju Sharma
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Michael E. Cox
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Trevor D. McKee
- STTARR Innovation Centre, Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Pathomics Inc., Toronto, Ontario, Canada
| | - Mark Zaidi
- Pathomics Inc., Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Eve Lapouble
- Unité Génétique Somatique (UGS), Institut Curie, Centre Hospitalier Paris, France
| | - John D. McPherson
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Department of Biochemistry and Molecular Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, California, USA
| | - Olivier Delattre
- Unité Génétique Somatique (UGS), Institut Curie, Centre Hospitalier Paris, France
- Diversity and Plasticity of Childhood tumors, INSERM U830, Institut Curie Research Center, PSL Research University, Paris, France
| | - Laszlo Radvanyi
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Stefan E.G. Burdach
- Department of Molecular Oncology, British Columbia Cancer Research Centre and
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- CCC München Comprehensive Cancer Center, DKTK German Cancer Consortium, Munich, Germany
- Institute of Pathology, Translation Pediatric Cancer Research Action, School of Medicine, Technical University of Munich, Munich, Germany
| | - Lincoln D. Stein
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Poul H. Sorensen
- Department of Molecular Oncology, British Columbia Cancer Research Centre and
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
26
|
Takahashi Ueda M. Retrotransposon-derived transcripts and their functions in immunity and disease. Genes Genet Syst 2024; 98:305-319. [PMID: 38199240 DOI: 10.1266/ggs.23-00187] [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: 01/12/2024] Open
Abstract
Retrotransposons, which account for approximately 42% of the human genome, have been increasingly recognized as "non-self" pathogen-associated molecular patterns (PAMPs) due to their virus-like sequences. In abnormal conditions such as cancer and viral infections, retrotransposons that are aberrantly expressed due to impaired epigenetic suppression display PAMPs, leading to their recognition by pattern recognition receptors (PRRs) of the innate immune system and triggering inflammation. This viral mimicry mechanism has been observed in various human diseases, including aging and autoimmune disorders. However, recent evidence suggests that retrotransposons possess highly regulated immune reactivity and play important roles in the development and function of the immune system. In this review, I discuss a wide range of retrotransposon-derived transcripts, their role as targets in immune recognition, and the diseases associated with retrotransposon activity. Furthermore, I explore the implications of chimeric transcripts formed between retrotransposons and known gene mRNAs, which have been previously underestimated, for the increase of immune-related gene isoforms and their influence on immune function. Retrotransposon-derived transcripts have profound and multifaceted effects on immune system function. The aim of this comprehensive review is to provide a better understanding of the complex relationship between retrotransposon transcripts and immune defense.
Collapse
Affiliation(s)
- Mahoko Takahashi Ueda
- Department of Genomic Function and Diversity, Medical Research Institute, Tokyo Medical and Dental University
| |
Collapse
|
27
|
Shen J, Wen X, Xing X, Fozza C, Sechi LA. Endogenous retroviruses Suppressyn and Syncytin-2 as innovative prognostic biomarkers in Acute Myeloid Leukemia. Front Cell Infect Microbiol 2024; 13:1339673. [PMID: 38274728 PMCID: PMC10808309 DOI: 10.3389/fcimb.2023.1339673] [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: 11/16/2023] [Accepted: 12/19/2023] [Indexed: 01/27/2024] Open
Abstract
Introduction Emerging evidence has proven that human endogenous retroviruses (HERVs) play a critical role in the pathogenesis of Acute Myeloid Leukemia (AML), whereas the specific HERVs influencing the prognosis of AML patients have yet to be fully understood. Methods In this study, a systematic exploration was achieved to identify potential prognostic HERVs for AML, sourced from TCGA and GTEx database. Differential analysis and functional enrichment studies were conducted using GO, KEGG, GSEA, and GSVA. The ESTIMATE algorithm was applied to explore the immune infiltration of HERVs in AML. A prognostic risk-score model was evaluated with predicted yearly accuracy using ROC analysis. Results Two HERVs Suppressyn and Syncytin-2, were identified as promising prognostic biomarkers, with high discrimination ability based on ROC analysis between AML and healthy cohorts from TCGA. Their expression was notably higher in AML patients compared to those in healthy individuals but correlates with favorable clinical outcomes in sub-groups such as white race, lower WBC counts, favorable and intermediate risks, and NPM1 or IDH1 mutation. Suppressyn and Syncytin-2 participated in immune-related pathways and exhibited correlations with multiple immune infiltration cells, such as T cells, mast cells, and tumor-associated macrophages. Finally, we developed a prognostic risk-scoring model combining Suppressyn and Syncytin-2, where a high risk-score is associated with better prognosis. Discussion Collectively, our findings revealed that Suppressyn and Syncytin-2 may act as valuable diagnostic and prognostic biomarkers for individuals with AML, while highlighting links between HERV activation, immunogenicity, and future therapeutic targets.
Collapse
Affiliation(s)
- Jiaxin Shen
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- Department of Hematology, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Xiaofen Wen
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- Department of Medical Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Xueyang Xing
- Department of Hematology, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Claudio Fozza
- Department of Medicine and Pharmacy, University of Sassari, Sassari, Italy
| | - Leonardo Antonio Sechi
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- SC of Microbiology and Virology, Azienda Ospedaliera Universitaria (AOU) of Sassari, Sassari, Italy
| |
Collapse
|
28
|
Milovic A, Duong JV, Barbour AG. The infection-tolerant white-footed deermouse tempers interferon responses to endotoxin in comparison to the mouse and rat. eLife 2024; 12:RP90135. [PMID: 38193896 PMCID: PMC10945503 DOI: 10.7554/elife.90135] [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] [Indexed: 01/10/2024] Open
Abstract
The white-footed deermouse Peromyscus leucopus, a long-lived rodent, is a key reservoir in North America for agents of several zoonoses, including Lyme disease, babesiosis, anaplasmosis, and a viral encephalitis. While persistently infected, this deermouse is without apparent disability or diminished fitness. For a model for inflammation elicited by various pathogens, the endotoxin lipopolysaccharide (LPS) was used to compare genome-wide transcription in blood by P. leucopus, Mus musculus, and Rattus norvegicus and adjusted for white cell concentrations. Deermice were distinguished from the mice and rats by LPS response profiles consistent with non-classical monocytes and alternatively-activated macrophages. LPS-treated P. leucopus, in contrast to mice and rats, also displayed little transcription of interferon-gamma and lower magnitude fold-changes in type 1 interferon-stimulated genes. These characteristics of P. leucopus were also noted in a Borrelia hermsii infection model. The phenomenon was associated with comparatively reduced transcription of endogenous retrovirus sequences and cytoplasmic pattern recognition receptors in the deermice. The results reveal a mechanism for infection tolerance in this species and perhaps other animal reservoirs for agents of human disease.
Collapse
Affiliation(s)
- Ana Milovic
- Department of Microbiology & Molecular Genetics, University of California, IrvineIrvineUnited States
| | - Jonathan V Duong
- Department of Microbiology & Molecular Genetics, University of California, IrvineIrvineUnited States
| | - Alan G Barbour
- Departments of Medicine, Microbiology & Molecular Genetics, and Ecology & Evolutionary Biology, University of California, IrvineIrvineUnited States
| |
Collapse
|
29
|
Deng S, Tian X, Belshaw R, Zhou J, Zhang S, Yang Y, Huang C, Chen W, Qiu H, Choo SW. An RNA-Seq analysis of coronavirus in the skin of the Pangolin. Sci Rep 2024; 14:910. [PMID: 38195813 PMCID: PMC10776870 DOI: 10.1038/s41598-024-51261-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: 10/26/2023] [Accepted: 01/02/2024] [Indexed: 01/11/2024] Open
Abstract
Protection of the Critically Endangered East Asian Pangolin species is hampered by the vulnerability of captive individuals to infection. Studies have previously shown the pangolin to have a unique pseudogenisation of many immunity genes (including IFNE, IFIH1, cGAS, STING, TLR5, and TLR11), and we suspected that these losses could account for this vulnerability. Here we used RNA-Seq data to show the effect of these gene losses on the transcriptional response to a viral skin infection in a deceased pangolin. This virus is very closely related to the one causing the current COVID-19 pandemic in the human population (SARS-CoV2), and we found the most upregulated pathway was the same one previously identified in the lungs of SARS-CoV2-infected humans. As predicted, we found that the pathways downstream of the lost genes were not upregulated. For example, the pseudogenised interferon epsilon (IFNE) is known to be particularly important in epithelial immunity, and we show that interferon-related responses were not upregulated in the infected pangolin skin. We suggest that the pangolin's innate gene pseudogenisation is indeed likely to be responsible for the animal's vulnerability to infection.
Collapse
Affiliation(s)
- Siwei Deng
- Department of Biology, College of Science, Mathematics and Technology, Wenzhou-Kean University, 88 Daxue Road, Ouhai, Wenzhou, Zhejiang, 325060, China
| | - Xuechen Tian
- Department of Biology, College of Science, Mathematics and Technology, Wenzhou-Kean University, 88 Daxue Road, Ouhai, Wenzhou, Zhejiang, 325060, China
- Zhejiang Bioinformatics International Science and Technology Cooperation Centre, Wenzhou-Kean University, 88 Daxue Road, Ouhai, Wenzhou, Zhejiang, 325060, China
- Wenzhou Municipal Key Laboratory for Applied Biomedical and Biopharmaceutical Informatics, 88 Daxue Road, Ouhai, Wenzhou, Zhejiang, 325060, China
| | - Robert Belshaw
- Department of Biology, College of Science, Mathematics and Technology, Wenzhou-Kean University, 88 Daxue Road, Ouhai, Wenzhou, Zhejiang, 325060, China
| | - Jinfeng Zhou
- China Biodiversity Conservation and Green Development Foundation (CBCGDF), Empark International Apartment, No. 69, Banding Road, Haidian District, Beijing, China
| | - Siyuan Zhang
- China Biodiversity Conservation and Green Development Foundation (CBCGDF), Empark International Apartment, No. 69, Banding Road, Haidian District, Beijing, China
| | - Yixin Yang
- Department of Biology, College of Science, Mathematics and Technology, Wenzhou-Kean University, 88 Daxue Road, Ouhai, Wenzhou, Zhejiang, 325060, China
- Zhejiang Bioinformatics International Science and Technology Cooperation Centre, Wenzhou-Kean University, 88 Daxue Road, Ouhai, Wenzhou, Zhejiang, 325060, China
- Wenzhou Municipal Key Laboratory for Applied Biomedical and Biopharmaceutical Informatics, 88 Daxue Road, Ouhai, Wenzhou, Zhejiang, 325060, China
- Dorothy and George Hennings College of Science, Mathematics and Technology, Kean University, 1000 Morris Ave, Union, NJ, 07083, USA
| | - Chang Huang
- Department of Biology, College of Science, Mathematics and Technology, Wenzhou-Kean University, 88 Daxue Road, Ouhai, Wenzhou, Zhejiang, 325060, China
| | - Weikang Chen
- Department of Biology, College of Science, Mathematics and Technology, Wenzhou-Kean University, 88 Daxue Road, Ouhai, Wenzhou, Zhejiang, 325060, China
| | - Hailu Qiu
- Department of Biology, College of Science, Mathematics and Technology, Wenzhou-Kean University, 88 Daxue Road, Ouhai, Wenzhou, Zhejiang, 325060, China
| | - Siew Woh Choo
- Department of Biology, College of Science, Mathematics and Technology, Wenzhou-Kean University, 88 Daxue Road, Ouhai, Wenzhou, Zhejiang, 325060, China.
- Zhejiang Bioinformatics International Science and Technology Cooperation Centre, Wenzhou-Kean University, 88 Daxue Road, Ouhai, Wenzhou, Zhejiang, 325060, China.
- Wenzhou Municipal Key Laboratory for Applied Biomedical and Biopharmaceutical Informatics, 88 Daxue Road, Ouhai, Wenzhou, Zhejiang, 325060, China.
- Dorothy and George Hennings College of Science, Mathematics and Technology, Kean University, 1000 Morris Ave, Union, NJ, 07083, USA.
| |
Collapse
|
30
|
Katoh H, Honda T. Roles of Human Endogenous Retroviruses and Endogenous Virus-Like Elements in Cancer Development and Innate Immunity. Biomolecules 2023; 13:1706. [PMID: 38136578 PMCID: PMC10741599 DOI: 10.3390/biom13121706] [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: 10/28/2023] [Revised: 11/18/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
Human endogenous retroviruses (HERVs) are remnants of ancient retroviral infections in the host genome. Although mutations and silencing mechanisms impair their original role in viral replication, HERVs are believed to play roles in various biological processes. Long interspersed nuclear elements (LINEs) are non-LTR retrotransposons that have a lifecycle resembling that of retroviruses. Although LINE expression is typically silenced in somatic cells, it also contributes to various biological processes. The aberrant expression of HERVs and LINEs is closely associated with the development of cancer and/or immunological diseases, suggesting that they are integrated into various pathways related to the diseases. HERVs/LINEs control gene expression depending on the context as promoter/enhancer elements. Some RNAs and proteins derived from HERVs/LINEs have oncogenic potential, whereas others stimulate innate immunity. Non-retroviral endogenous viral elements (nrEVEs) are a novel type of virus-like element in the genome. nrEVEs may also be involved in host immunity. This article provides a current understanding of how these elements impact cellular physiology in cancer development and innate immunity, and provides perspectives for future studies.
Collapse
Affiliation(s)
- Hirokazu Katoh
- Department of Virology, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan;
| | - Tomoyuki Honda
- Department of Virology, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan;
- Department of Virology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| |
Collapse
|
31
|
Ko EJ, Kim MH, Kim DY, An H, Leem SH, Choi YH, Kim HS, Cha HJ. The Role of Human Endogenous Retrovirus (HERV)-K119 env in THP-1 Monocytic Cell Differentiation. Int J Mol Sci 2023; 24:15566. [PMID: 37958549 PMCID: PMC10648273 DOI: 10.3390/ijms242115566] [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: 08/25/2023] [Revised: 10/16/2023] [Accepted: 10/16/2023] [Indexed: 11/15/2023] Open
Abstract
Human endogenous retrovirus (HERV)-K was reportedly inserted into the human genome millions of years ago and is closely related to various diseases, including cancer and immune regulation. In our previous studies, CRISPR-Cas9-enabled knockout (KO) of the HERV-K env gene was found to potentially reduce cell proliferation, cell migration, and invasion in colorectal and ovarian cancer cell lines. The immune response involves the migration and invasion of cells and is similar to cancer; however, in certain ways, it is completely unlike cancer. Therefore, we induced HERV-K119 env gene KO in THP-1, a monocytic cell that can be differentiated into a macrophage, to investigate the role of HERV-K119 env in immune regulation. Cell migration and invasion were noted to be significantly increased in HERV-K119 env KO THP-1 cells than in MOCK, and these results were contrary to those of cancer cells. To identify the underlying mechanism of HERV-K119 env KO in THP-1 cells, transcriptome analysis and cytokine array analysis were conducted. Semaphorin7A (SEMA7A), which induces the production of cytokines in macrophages and monocytic cells and plays an important role in immune effector cell activation during an inflammatory immune response, was significantly increased in HERV-K119 env KO THP-1 cells. We also found that HERV-K119 env KO THP-1 cells expressed various macrophage-specific surface markers, suggesting that KO of HERV-K119 env triggers the differentiation of THP-1 cells from monocytic cells into macrophages. In addition, analysis of the expression of M1 and M2 macrophage markers showed that M1 macrophage marker cluster of differentiation 32 (CD32) was significantly increased in HERV-K119 env KO cells. These results suggest that HERV-K119 env is implicated in the differentiation of monocytic cells into M1 macrophages and plays important roles in the immune response.
Collapse
Affiliation(s)
- Eun-Ji Ko
- Departments of Parasitology and Genetics, Kosin University College of Medicine, Busan 49267, Republic of Korea; (E.-J.K.); (M.-H.K.); (D.-Y.K.); (H.A.)
| | - Min-Hye Kim
- Departments of Parasitology and Genetics, Kosin University College of Medicine, Busan 49267, Republic of Korea; (E.-J.K.); (M.-H.K.); (D.-Y.K.); (H.A.)
- Department of Biomedical Sciences, Dong-A University, Busan 49315, Republic of Korea;
| | - Do-Ye Kim
- Departments of Parasitology and Genetics, Kosin University College of Medicine, Busan 49267, Republic of Korea; (E.-J.K.); (M.-H.K.); (D.-Y.K.); (H.A.)
| | - Hyojin An
- Departments of Parasitology and Genetics, Kosin University College of Medicine, Busan 49267, Republic of Korea; (E.-J.K.); (M.-H.K.); (D.-Y.K.); (H.A.)
| | - Sun-Hee Leem
- Department of Biomedical Sciences, Dong-A University, Busan 49315, Republic of Korea;
| | - Yung Hyun Choi
- Department of Biochemistry, College of Oriental Medicine, Dongeui University, Busan 47227, Republic of Korea;
| | - Heui-Soo Kim
- Department of Biological Sciences, College of Natural Sciences, Pusan National University, Busan 49241, Republic of Korea;
| | - Hee-Jae Cha
- Departments of Parasitology and Genetics, Kosin University College of Medicine, Busan 49267, Republic of Korea; (E.-J.K.); (M.-H.K.); (D.-Y.K.); (H.A.)
- Institute for Medical Science, Kosin University College of Medicine, Busan 49267, Republic of Korea
| |
Collapse
|
32
|
Milovic A, Duong JV, Barbour AG. The white-footed deermouse, an infection-tolerant reservoir for several zoonotic agents, tempers interferon responses to endotoxin in comparison to the mouse and rat. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.06.543964. [PMID: 37745581 PMCID: PMC10515768 DOI: 10.1101/2023.06.06.543964] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
The white-footed deermouse Peromyscus leucopus, a long-lived rodent, is a key reservoir for agents of several zoonoses, including Lyme disease. While persistently infected, this deermouse is without apparent disability or diminished fitness. For a model for inflammation elicited by various pathogens, the endotoxin lipopolysaccharide (LPS) was used to compare genome-wide transcription in blood by P. leucopus, Mus musculus and Rattus norvegicus and adjusted for white cell concentrations. Deermice were distinguished from the mice and rats by LPS response profiles consistent with non-classical monocytes and alternatively-activated macrophages. LPS-treated P. leucopus, in contrast to mice and rats, also displayed little transcription of interferon-gamma and lower magnitude fold-changes in type 1 interferon-stimulated genes. This was associated with comparatively reduced transcription of endogenous retrovirus sequences and cytoplasmic pattern recognition receptors in the deermice. The results reveal a mechanism for infection tolerance in this species and perhaps other animal reservoirs for agents of human disease.
Collapse
Affiliation(s)
- Ana Milovic
- Department of Microbiology & Molecular Genetics, University of California Irvine
| | - Jonathan V. Duong
- Department of Microbiology & Molecular Genetics, University of California Irvine
| | - Alan G. Barbour
- Departments of Medicine, Microbiology & Molecular Genetics, and Ecology & Evolutionary Biology, University of California Irvine
| |
Collapse
|
33
|
Balestrieri E, Corinaldesi E, Fabi M, Cipriani C, Giudice M, Conti A, Minutolo A, Petrone V, Fanelli M, Miele MT, Andreozzi L, Guida F, Filice E, Meli M, Grelli S, Rasi G, Toschi N, Torcetta F, Matteucci C, Lanari M, Sinibaldi-Vallebona P. Preliminary Evidence of the Differential Expression of Human Endogenous Retroviruses in Kawasaki Disease and SARS-CoV-2-Associated Multisystem Inflammatory Syndrome in Children. Int J Mol Sci 2023; 24:15086. [PMID: 37894766 PMCID: PMC10606856 DOI: 10.3390/ijms242015086] [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: 08/23/2023] [Revised: 10/03/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
Multisystem inflammatory syndrome in children (MIS-C) is a postinfectious sequela of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), with some clinical features overlapping with Kawasaki disease (KD). Our research group and others have highlighted that the spike protein of SARS-CoV-2 can trigger the activation of human endogenous retroviruses (HERVs), which in turn induces inflammatory and immune reactions, suggesting HERVs as contributing factors in COVID-19 immunopathology. With the aim to identify new factors involved in the processes underlying KD and MIS-C, we analysed the transcriptional levels of HERVs, HERV-related genes, and immune mediators in children during the acute and subacute phases compared with COVID-19 paediatric patients and healthy controls. The results showed higher levels of HERV-W, HERV-K, Syn-1, and ASCT-1/2 in KD, MIS-C, and COV patients, while higher levels of Syn-2 and MFSD2A were found only in MIS-C patients. Moreover, KD and MIS-C shared the dysregulation of several inflammatory and regulatory cytokines. Interestingly, in MIS-C patients, negative correlations have been found between HERV-W and IL-10 and between Syn-2 and IL-10, while positive correlations have been found between HERV-K and IL-10. In addition, HERV-W expression positively correlated with the C-reactive protein. This pilot study supports the role of HERVs in inflammatory diseases, suggesting their interplay with the immune system in this setting. The elevated expression of Syn-2 and MFSD2A seems to be a distinctive trait of MIS-C patients, allowing to distinguish them from KD ones. The understanding of pathological mechanisms can lead to the best available treatment for these two diseases, limiting complications and serious outcomes.
Collapse
Affiliation(s)
- Emanuela Balestrieri
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (M.G.); (A.M.); (V.P.); (M.F.); (M.T.M.); (S.G.); (G.R.); (C.M.); (P.S.-V.)
| | - Elena Corinaldesi
- Pediatric Unit, Ramazzini Hospital, 41012 Carpi, Italy; (E.C.); (F.T.)
| | - Marianna Fabi
- Pediatric Emergency Unit, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40126 Bologna, Italy; (L.A.); (F.G.); (E.F.); (M.M.); (M.L.)
| | - Chiara Cipriani
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (M.G.); (A.M.); (V.P.); (M.F.); (M.T.M.); (S.G.); (G.R.); (C.M.); (P.S.-V.)
| | - Martina Giudice
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (M.G.); (A.M.); (V.P.); (M.F.); (M.T.M.); (S.G.); (G.R.); (C.M.); (P.S.-V.)
| | - Allegra Conti
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (A.C.); (N.T.)
| | - Antonella Minutolo
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (M.G.); (A.M.); (V.P.); (M.F.); (M.T.M.); (S.G.); (G.R.); (C.M.); (P.S.-V.)
| | - Vita Petrone
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (M.G.); (A.M.); (V.P.); (M.F.); (M.T.M.); (S.G.); (G.R.); (C.M.); (P.S.-V.)
| | - Marialaura Fanelli
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (M.G.); (A.M.); (V.P.); (M.F.); (M.T.M.); (S.G.); (G.R.); (C.M.); (P.S.-V.)
| | - Martino Tony Miele
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (M.G.); (A.M.); (V.P.); (M.F.); (M.T.M.); (S.G.); (G.R.); (C.M.); (P.S.-V.)
| | - Laura Andreozzi
- Pediatric Emergency Unit, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40126 Bologna, Italy; (L.A.); (F.G.); (E.F.); (M.M.); (M.L.)
| | - Fiorentina Guida
- Pediatric Emergency Unit, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40126 Bologna, Italy; (L.A.); (F.G.); (E.F.); (M.M.); (M.L.)
| | - Emanuele Filice
- Pediatric Emergency Unit, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40126 Bologna, Italy; (L.A.); (F.G.); (E.F.); (M.M.); (M.L.)
| | - Matteo Meli
- Pediatric Emergency Unit, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40126 Bologna, Italy; (L.A.); (F.G.); (E.F.); (M.M.); (M.L.)
| | - Sandro Grelli
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (M.G.); (A.M.); (V.P.); (M.F.); (M.T.M.); (S.G.); (G.R.); (C.M.); (P.S.-V.)
| | - Guido Rasi
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (M.G.); (A.M.); (V.P.); (M.F.); (M.T.M.); (S.G.); (G.R.); (C.M.); (P.S.-V.)
| | - Nicola Toschi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (A.C.); (N.T.)
- Martinos Center for Biomedical Imaging and Harvard Medical School, Boston, MA 02129, USA
| | | | - Claudia Matteucci
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (M.G.); (A.M.); (V.P.); (M.F.); (M.T.M.); (S.G.); (G.R.); (C.M.); (P.S.-V.)
| | - Marcello Lanari
- Pediatric Emergency Unit, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40126 Bologna, Italy; (L.A.); (F.G.); (E.F.); (M.M.); (M.L.)
| | - Paola Sinibaldi-Vallebona
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (M.G.); (A.M.); (V.P.); (M.F.); (M.T.M.); (S.G.); (G.R.); (C.M.); (P.S.-V.)
- National Research Council, Institute of Translational Pharmacology, 00133 Rome, Italy
| |
Collapse
|
34
|
Costa B, Vale N. Exploring HERV-K (HML-2) Influence in Cancer and Prospects for Therapeutic Interventions. Int J Mol Sci 2023; 24:14631. [PMID: 37834078 PMCID: PMC10572383 DOI: 10.3390/ijms241914631] [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: 08/29/2023] [Revised: 09/19/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
This review investigates the intricate role of human endogenous retroviruses (HERVs) in cancer development and progression, explicitly focusing on HERV-K (HML-2). This paper sheds light on the latest research advancements and potential treatment strategies by examining the historical context of HERVs and their involvement in critical biological processes such as embryonic development, immune response, and disease progression. This review covers computational modeling for drug-target binding assessment, systems biology modeling for simulating HERV-K viral cargo dynamics, and using antiviral drugs to combat HERV-induced diseases. The findings presented in this review contribute to our understanding of HERV-mediated disease mechanisms and provide insights into future therapeutic approaches. They emphasize why HERV-K holds significant promise as a biomarker and a target.
Collapse
Affiliation(s)
- Bárbara Costa
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, s/n, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, s/n, 4200-450 Porto, Portugal
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, s/n, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, s/n, 4200-450 Porto, Portugal
| |
Collapse
|
35
|
Keighley LM, Lynch-Sutherland CF, Almomani SN, Eccles MR, Macaulay EC. Unveiling the hidden players: The crucial role of transposable elements in the placenta and their potential contribution to pre-eclampsia. Placenta 2023; 141:57-64. [PMID: 37301654 DOI: 10.1016/j.placenta.2023.05.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 05/21/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023]
Abstract
The human placenta is a vital connection between maternal and fetal tissues, allowing for the exchange of molecules and modulation of immune interactions during pregnancy. Interestingly, some of the placenta's unique functions can be attributed to transposable elements (TEs), which are DNA sequences that have mobilised into the genome. Co-option throughout mammalian evolution has led to the generation of TE-derived regulators and TE-derived genes, some of which are expressed in the placenta but silenced in somatic tissues. TE genes encompass both TE-derived genes with a repeat element in the coding region and TE-derived regulatory regions such as alternative promoters and enhancers. Placental-specific TE genes are known to contribute to the placenta's unique functions, and interestingly, they are also expressed in some cancers and share similar functions. There is evidence to support that aberrant activity of TE genes may contribute to placental pathologies, cancer and autoimmunity. In this review, we highlight the crucial roles of TE genes in placental function, and how their dysregulation may lead to pre-eclampsia, a common and dangerous placental condition. We provide a summary of the functional TE genes in the placenta to offer insight into their significance in normal and abnormal human development. Ultimately, this review highlights an opportunity for future research to investigate the potential dysregulation of TE genes in the development of placental pathologies such as pre-eclampsia. Further understanding of TE genes and their role in the placenta could lead to significant improvements in maternal and fetal health.
Collapse
Affiliation(s)
- Laura M Keighley
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, 9054, New Zealand
| | - Chiemi F Lynch-Sutherland
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, 9054, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Level 2, 3A Symonds Street, Auckland, New Zealand
| | - Suzan N Almomani
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, 9054, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Level 2, 3A Symonds Street, Auckland, New Zealand
| | - Michael R Eccles
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, 9054, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Level 2, 3A Symonds Street, Auckland, New Zealand
| | - Erin C Macaulay
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, 9054, New Zealand.
| |
Collapse
|
36
|
Charvet B, Brunel J, Pierquin J, Iampietro M, Decimo D, Queruel N, Lucas A, Encabo-Berzosa MDM, Arenaz I, Marmolejo TP, Gonzalez AI, Maldonado AC, Mathieu C, Küry P, Flores-Rivera J, Torres-Ruiz F, Avila-Rios S, Salgado Montes de Oca G, Schoorlemmer J, Perron H, Horvat B. SARS-CoV-2 awakens ancient retroviral genes and the expression of proinflammatory HERV-W envelope protein in COVID-19 patients. iScience 2023; 26:106604. [PMID: 37091988 PMCID: PMC10079620 DOI: 10.1016/j.isci.2023.106604] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 01/29/2023] [Accepted: 03/30/2023] [Indexed: 04/25/2023] Open
Abstract
Patients with COVID-19 may develop abnormal inflammatory response, followed in some cases by severe disease and long-lasting syndromes. We show here that in vitro exposure to SARS-CoV-2 activates the expression of the human endogenous retrovirus (HERV) HERV-W proinflammatory envelope protein (ENV) in peripheral blood mononuclear cells from a subset of healthy donors, in ACE2 receptor and infection-independent manner. Plasma and/or sera of 221 COVID-19 patients from different cohorts, infected with successive SARS-CoV-2 variants including the Omicron, had detectable HERV-W ENV, which correlated with ENV expression in T lymphocytes and peaked with the disease severity. HERV-W ENV was also found in postmortem tissues of lungs, heart, gastrointestinal tract, brain olfactory bulb, and nasal mucosa from COVID-19 patients. Altogether, these results demonstrate that SARS-CoV-2 could induce HERV-W envelope protein expression and suggest its involvement in the immunopathogenesis of certain COVID-19-associated syndromes and thereby its relevance in the development of personalized treatment of patients.
Collapse
Affiliation(s)
| | | | | | - Mathieu Iampietro
- CIRI, International Center for Infectiology Research, INSERM U1111, CNRS UMR5308, Université de Lyon, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Didier Decimo
- CIRI, International Center for Infectiology Research, INSERM U1111, CNRS UMR5308, Université de Lyon, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| | | | - Alexandre Lucas
- We-Met platform, I2MC/Inserm/Université Paul Sabatier UMR1297, Toulouse, France
| | | | - Izaskun Arenaz
- Biobanco del Sistema de Salud de Aragón, Instituto Aragonés de Ciencias de la Salud (IACS), Zaragoza, Spain
| | - Tania Perez Marmolejo
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, México Ciudad, México
| | - Arturo Ivan Gonzalez
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, México Ciudad, México
| | | | - Cyrille Mathieu
- CIRI, International Center for Infectiology Research, INSERM U1111, CNRS UMR5308, Université de Lyon, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Dusseldorf, Germany
| | - Jose Flores-Rivera
- Department of Neurology, National Institute of Neurology and Neurosurgery, Mexico City, Mexico
| | - Fernanda Torres-Ruiz
- Centro de investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, México Ciudad, México
| | - Santiago Avila-Rios
- Centro de investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, México Ciudad, México
| | - Gonzalo Salgado Montes de Oca
- Centro de investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, México Ciudad, México
| | - Jon Schoorlemmer
- ARAID Fundación; Instituto Aragonés de Ciencias de la Salud (IACS); Grupo B46_20R de la DGA and GIIS-028 del IISA; all Zaragoza, Spain
| | - Hervé Perron
- GeNeuro Innovation, Lyon, France
- GeNeuro, Plan les Ouates, Geneva, Switzerland
| | - Branka Horvat
- CIRI, International Center for Infectiology Research, INSERM U1111, CNRS UMR5308, Université de Lyon, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| |
Collapse
|