1
|
Esparza M, El Zahed SS, Karakus U, Niederstrasser H, Gao B, Batten K, Shay JW, Posner B, Hirsch FR, Girard L, Huang LJS, Minna J, García-Sastre A, Fontoura BMA. Contrasting interferon-mediated antiviral responses in human lung adenocarcinoma cells. J Virol 2025:e0046925. [PMID: 40434103 DOI: 10.1128/jvi.00469-25] [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: 03/12/2025] [Accepted: 05/02/2025] [Indexed: 05/29/2025] Open
Abstract
Lung cancers develop from lung epithelial cells after a series of genetic and epigenetic changes, and these cells are major sites of influenza virus infection. Thus, we explored how changes found in patient-derived lung cancer cell lines impacted influenza virus replication and identified two lines with opposite responses to influenza A viral infection. We show that the NCI-H820 lung adenocarcinoma (LUAD) is resistant to influenza A virus and VSV infection, while LUAD line NCI-H322 is highly susceptible to infection by both viruses. H322 cells have a homozygous deletion in a region of chromosome 9 encoding IFNαgenes, IFNβ1, IFNω1, and IFNε genes, leading to downregulation of immune response and high infection rates. In contrast, the resistant H820 cell line has three copies of these same interferon genes and shows increased expression of interferon-regulated genes. We found that the resistance of H820 cells to influenza infection is likely linked to impaired viral entry-due to high basal levels of interferon-induced proteins known to inhibit endocytosis (IFITM1/2/3, NCOA7, and CH25H)-and to increased expression of mRNAs that encode other antiviral factors. In contrast, H322 cells show the absence or low levels of interferon-regulated genes involved in the inhibition of viral entry. These results suggest that the opposite phenotypes on viral entry of H322 and H820 cells may be at least in part associated with impaired or enhanced interferon response, respectively. Since most lung cancer patients have genomic characterization of their tumors, individualized differences in interferon responses may have therapeutic and patient management implications. IMPORTANCE Lung cancers develop from genetic and epigenetic changes that can dramatically influence patients' susceptibility to viral infection and replication. This study evaluates the responses to influenza virus infection of two patient-derived lung cancer cell lines. Interestingly, the cell lines investigated are of the same cancer type, lung adenocarcinomas, yet one cell line is highly susceptible, while the other cell line is highly resistant to viral infection. This is in part due to contrasting genetic alterations that lead to changes in the interferon response pathways, which differentially impact viral entry. Thus, identifying these risk factors can inform the prognosis of patients infected with influenza virus and guide their personalized treatment plans.
Collapse
Affiliation(s)
- Matthew Esparza
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Sara S El Zahed
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Umut Karakus
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Boning Gao
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Kimberly Batten
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jerry W Shay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Bruce Posner
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Fred R Hirsch
- Center for Thoracic Oncology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Luc Girard
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Lily Jun-Shen Huang
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - John Minna
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Departments of Internal Medicine and Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- The Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Beatriz M A Fontoura
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
2
|
Salih MM, Weindel CG, Malekos E, Sudek L, Katzman S, Mabry CJ, Chapman MJ, Coleman AK, Azam S, Watson RO, Patrick KL, Carpenter S. Myeloid-specific HNRNPA2B1 deficiency disrupts macrophage function and in vivo responses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf073. [PMID: 40414614 DOI: 10.1093/jimmun/vkaf073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 03/12/2025] [Accepted: 03/25/2025] [Indexed: 05/27/2025]
Abstract
The mechanisms through which heterogeneous nuclear ribonucleoprotein A2B1 (HNRNPA2B1) contributes to innate immune gene regulation are poorly understood. To fill this gap, we generated a myeloid lineage-specific HNRNPA2B1-conditional mouse using LysMCre. In an endotoxic shock model, HNRNPA2B1-deficient mice exhibit dampened expression of inflammatory mediators despite increased infiltration of macrophages and neutrophils. Likewise, during infection with the gram-negative bacterial pathogen Salmonella enterica, HNRNPA2B1-deficient mice fail to mount protective inflammatory responses and experience higher bacterial burdens. To better understand the molecular mechanisms driving these phenotypes in vivo, we performed transcriptomics analysis of LPS-treated HNRNPA2B1-deficient macrophages ex vivo. We noted an increase in transcripts encoding nonproductive isoforms of a number of Interferon (IFN)-regulated genes, including the IFNG receptor (IFNGR). Focusing on IFNGR, we confirmed lower surface expression on HNRNPA2B1-deficient macrophages and dampened responsiveness in response to IFNG treatment. In conclusion, our data demonstrates that HNRNPA2B1 is essential for optimal macrophage function, particularly in the context of intracellular bacterial restriction in the case of Salmonella infection. This highlights a previously unappreciated role for RNA-binding proteins in mounting effective immune defenses.
Collapse
Affiliation(s)
- Mays Mohammed Salih
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA, United States
| | - Chi G Weindel
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX, United States
| | - Eric Malekos
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA, United States
| | - Lisa Sudek
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA, United States
| | - Sol Katzman
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA, United States
| | - Cory J Mabry
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Morgan J Chapman
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Aja K Coleman
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Sikandar Azam
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX, United States
| | - Robert O Watson
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Kristin L Patrick
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Susan Carpenter
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA, United States
| |
Collapse
|
3
|
Walter Z, Li M, Molho M, Berish L, Isopi A, O'Mara M, Dittmar M, Nwaezeapu C, Richards A, McCullagh M, Krogan NJ, Cherry S, Johnson JR, Ramage H. An integrated proteomics approach identifies phosphorylation sites on viral and host proteins that regulate West Nile virus infection. Cell Rep 2025; 44:115728. [PMID: 40381193 DOI: 10.1016/j.celrep.2025.115728] [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: 05/02/2024] [Revised: 03/22/2025] [Accepted: 04/30/2025] [Indexed: 05/20/2025] Open
Abstract
Upon infection, viruses alter the proteome, creating a hospitable environment for infection. Cells respond to limit viral replication, including through protein regulation by post-translational modifications. We use mass spectrometry to define proteome alterations during West Nile virus (WNV) infection. Our studies identify upregulation of HERPUD1, which restricts WNV replication through a mechanism independent of its role in endoplasmic reticulum (ER)-associated degradation (ERAD). We also identify modifications on viral proteins, including a WNV NS3 phosphorylation site that impacts viral replication. Finally, we reveal activation of two host kinases with antiviral activity. We identify phosphorylation at S108 of AMPKβ1, a non-catalytic subunit that regulates activity of the AMPK complex. We also show activation of PAK2 by phosphorylation at S141, which restricts translation of the viral genome. This work contributes to our understanding of the interplay between host and virus while providing a resource to define the changes to the proteome that regulate viral infection.
Collapse
Affiliation(s)
- Zachary Walter
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Minghua Li
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Melissa Molho
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Lauren Berish
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Andrew Isopi
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Mary O'Mara
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Mark Dittmar
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Chike Nwaezeapu
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Alicia Richards
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94143, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA 94143, USA
| | - Martin McCullagh
- Department of Chemistry, Oklahoma State University, Stillwater, OK 74078, USA
| | - Nevan J Krogan
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94143, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA 94143, USA; The J. David Gladstone Institutes, San Francisco, CA 94158, USA
| | - Sara Cherry
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Jeffrey R Johnson
- Department of Microbiology, Icahn School of Medicine at Mt. Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Holly Ramage
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
4
|
Rannikko JH, Turpin R, Boström P, Virtakoivu R, Harth C, Takeda A, Tamminen A, Koskivuo I, Hollmén M. Macrophage sensitivity to bexmarilimab-induced reprogramming is shaped by the tumor microenvironment. J Immunother Cancer 2025; 13:e011292. [PMID: 40379271 PMCID: PMC12083384 DOI: 10.1136/jitc-2024-011292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 04/26/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND Tumor-associated macrophages (TAMs) adapt to the tumor microenvironment (TME), either aiding cancer eradication or promoting tumor growth and immune evasion. To manipulate TAMs therapeutically, a deep understanding of their interaction with the TME is essential. This study explores the responsiveness of TMEs to bexmarilimab, a macrophage reprogramming therapy showing clinical benefit in various solid tumors. METHODS We exploited a breast cancer patient-derived explant culture (PDEC) model to characterize bexmarilimab responses in both tumor and adjacent cancer-free tissues by RNA sequencing and multiplex cytokine profiling. Using single-cell RNA sequencing, spatial transcriptomics, and conditioned media treatment, we further investigated the effects of Clever-1+ macrophages and TME features on bexmarilimab sensitivity. RESULTS The PDEC model captured key aspects of bexmarilimab's mode of action and validated a gene signature for determining treatment sensitivity. We identified three distinct responses to bexmarilimab in tumors and adjacent cancer-free tissues, shaped by the local microenvironment and macrophage phenotype, origin, and localization. The inflammatory state of the TME emerged as the primary determinant of response. Immune activation occurred in immunologically cold TMEs lacking late-stage activated TAMs, whereas interferon-regulated TMEs exhibited suppressed inflammation. In cancer-free breast tissue, bexmarilimab activated B cell responses independent of treatment sensitivity in the adjacent tumor. CONCLUSIONS These findings reveal the complexity of TAM targeting in cancer and emphasize the need for patient selection to maximize bexmarilimab's efficacy.
Collapse
Affiliation(s)
- Jenna H Rannikko
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Turku, Finland
| | - Rita Turpin
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Turku, Finland
| | - Pia Boström
- Department of Pathology, TYKS Turku University Hospital, Turku, Finland
| | - Reetta Virtakoivu
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Turku, Finland
| | - Chantal Harth
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Turku, Finland
| | - Akira Takeda
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Turku, Finland
| | - Anselm Tamminen
- Department of Plastic and General Surgery, TYKS Turku University Hospital, Turku, Finland
| | - Ilkka Koskivuo
- Department of Plastic and General Surgery, TYKS Turku University Hospital, Turku, Finland
| | - Maija Hollmén
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Turku, Finland
| |
Collapse
|
5
|
Bayat M, Nahid-Samiei R, Sadri Nahand J, Naghili B. Interferon and immunity: the role of microRNA in viral evasion strategies. Front Immunol 2025; 16:1567459. [PMID: 40416980 PMCID: PMC12101089 DOI: 10.3389/fimmu.2025.1567459] [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: 01/27/2025] [Accepted: 03/26/2025] [Indexed: 05/27/2025] Open
Abstract
Interferons (IFNs) are indispensable innate antiviral cytokines that orchestrate the vertebrate immune response against viral incursions. Nearly every cell possesses the remarkable ability to release IFNs upon detecting viral threats, triggering a robust signaling cascade that alerts neighboring cells and halts viral propagation via paracrine communication. The intricate influence of IFNs is mediated by an extensive network of proteins activated through the Jak-STAT pathways, facilitating the swift transcription of over 300 interferon-stimulated genes (ISGs) that fortify cellular defenses against replication. However, the cunning nature of viruses has led to the evolution of sophisticated evasion strategies, notably through the manipulation of host microRNAs (miRNAs) that disrupt vital components of the IFN signaling machinery. This review delves into the intricate interplay between viral infections and both host- and viral-derived miRNAs, exploring their potent roles in modulating RIG-I-like receptors, Toll-like receptors, IFN receptors, and the JAK/STAT pathway, ultimately shaping the landscape of antiviral immunity.
Collapse
Affiliation(s)
- Mobina Bayat
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rahil Nahid-Samiei
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Javid Sadri Nahand
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behrouz Naghili
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
6
|
Broomfield BJ, Tan CW, Qin RZ, Abberger H, Duckworth BC, Alvarado C, Dalit L, Lee CL, Shandre Mugan R, Mazrad ZA, Muramatsu H, Mackiewicz L, Williams BE, Chen J, Takanashi A, Fabb S, Pellegrini M, Rogers KL, Moon WJ, Pouton CW, Davis MJ, Nutt SL, Pardi N, Wimmer VC, Groom JR. Transient inhibition of type I interferon enhances CD8+ T cell stemness and vaccine protection. J Exp Med 2025; 222:e20241148. [PMID: 40062995 PMCID: PMC11893171 DOI: 10.1084/jem.20241148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 11/25/2024] [Accepted: 02/04/2025] [Indexed: 03/14/2025] Open
Abstract
Developing vaccines that promote CD8+ T cell memory is a challenge for infectious disease and cancer immunotherapy. TCF-1+ stem cell-like memory CD8+ T (TSCM) cells are important determinants of long-lived memory. Yet, the developmental requirements for TSCM cell formation are unclear. Here, we identify the temporal window for type I interferon receptor (IFNAR) blockade to drive TSCM cell generation following viral infection and mRNA-lipid nanoparticle vaccination. We reveal a reversible developmental trajectory where transcriptionally distinct TSCM cells emerged from a transitional precursor of exhausted T cellular state concomitant with viral clearance. TSCM cell differentiation correlated with T cell retention within the lymph node paracortex due to disrupted CXCR3 chemokine gradient formation. These effects were linked to increased antigen load and a counterintuitive increase in IFNγ, which controlled cell location. Vaccination with the IFNAR blockade promoted TSCM cell differentiation and enhanced protection against chronic infection. These findings propose an approach to vaccine design whereby modulation of inflammation promotes memory formation and function.
Collapse
Affiliation(s)
- Benjamin J. Broomfield
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Chin Wee Tan
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Raymond Z. Qin
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Hanna Abberger
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Brigette C. Duckworth
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Carolina Alvarado
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Lennard Dalit
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Chee Leng Lee
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Rekha Shandre Mugan
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Zihnil A.I. Mazrad
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Hiromi Muramatsu
- Department of Microbiology, Perelman School of Medicine, Philadelphia, PA, USA
| | - Liana Mackiewicz
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Bailey E. Williams
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Jinjin Chen
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Asuka Takanashi
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Stewart Fabb
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Marc Pellegrini
- Centenary Institute of Cancer Medicine and Cell Biology, Camperdown, Australia
| | - Kelly L. Rogers
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | | | - Colin W. Pouton
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Melissa J. Davis
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Australia
- School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia
| | - Stephen L. Nutt
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, Philadelphia, PA, USA
| | - Verena C. Wimmer
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Joanna R. Groom
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| |
Collapse
|
7
|
Zhao Y, Metzler AD, Li Y, Urlich P, Wang Y, Gilbert DM, Yao B, Tang H. Interferon-dependent R-loop induction by Zika virus contributes to growth attenuation. PNAS NEXUS 2025; 4:pgaf147. [PMID: 40386679 PMCID: PMC12082298 DOI: 10.1093/pnasnexus/pgaf147] [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: 08/20/2024] [Accepted: 03/31/2025] [Indexed: 05/20/2025]
Abstract
Zika virus (ZIKV) infection in human neural progenitors triggers DNA damage and activates DNA damage response, leading to cell cycle arrest that can retard brain development. Here, we link the ZIKV-induced S phase arrest to replication fork stalling and R-loop induction. DRIP-seq reveals that ZIKV infection induces R loops at specific loci strongly enriched in the interferon (IFN)-stimulated genes (ISGs). Bromouridine sequencing results further indicate that nascent ISGs transcripts are prone to R-loop induction upon infection. Knockout of IFN receptor eliminated the R loops on ISGs and partially rescued S-phase arrest in infected cells. And overexpression of RNaseH1 reduced ZIKV-mediated DNA damage and cell cycle arrest. We conclude that unscheduled expression of ISGs induced by ZIKV alters R-loop homeostasis and perturbs replication fork progression, leading to fork stalling and eventually DNA damage. IFN-dependent R-loop induction represents a previously unknown, nucleic acid-based mechanism for cell cycle arrest.
Collapse
Affiliation(s)
- Yijing Zhao
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Anna D Metzler
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Yangping Li
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Paola Urlich
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Yilin Wang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - David M Gilbert
- San Diego Biomedical Research Institute, San Diego, CA 92121, USA
| | - Bing Yao
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hengli Tang
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
8
|
Souto EP, Gong P, Landua JD, Rajaram Srinivasan R, Ganesan A, Dobrolecki LE, Purdy SC, Pan X, Zeosky M, Chung A, Yi SS, Ford HL, Lewis MT. Lineage Tracing and Single-Cell RNA Sequencing Reveal a Common Transcriptional State in Breast Cancer Tumor-Initiating Cells Characterized by IFN/STAT1 Activity. Cancer Res 2025; 85:1390-1409. [PMID: 40230213 PMCID: PMC11997551 DOI: 10.1158/0008-5472.can-23-4022] [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: 12/19/2023] [Revised: 09/03/2024] [Accepted: 01/31/2025] [Indexed: 04/16/2025]
Abstract
A tumor cell subpopulation of tumor-initiating cells (TIC) or "cancer stem cells" is associated with therapeutic resistance, as well as both local and distant recurrences. Signal transducer and activator of transcription (STAT) activity is elevated in TICs in claudin-low models of human triple-negative breast cancer, which enables enrichment of TICs using a STAT-responsive reporter. Lineage tracing of TICs as they undergo cell state changes could enable a better understanding of the molecular phenotypes of TIC and uncover strategies to selectively target TICs. In this study, we developed a STAT-responsive lineage-tracing system and used it in conjunction with the original reporter to enrich for cells with enhanced mammosphere-forming potential. This approach was able to detect TICs in some, but not all, basal-like triple-negative breast cancer xenograft models, indicating that STAT signaling has both TIC-related and TIC-independent functions. Single-cell RNA sequencing (RNA-seq) of reporter-tagged xenografts and clinical samples identified a common IFN/STAT1-associated transcriptional state in TICs that was previously linked to inflammation and macrophage differentiation. Surprisingly, most of the identified genes were not present in previously published TIC signatures derived using bulk RNA-seq. Finally, bone marrow stromal cell antigen-2 was identified as a cell surface marker of this state that functionally regulated TIC frequency. These results suggest that TICs may exploit the IFN/STAT1 signaling axis to promote their activity and that targeting this pathway may help eliminate TICs. Significance: Coupling single-cell transcriptomics with tumor-initiating cell enrichment identified IFN response gene expression not previously reported in bulk RNA-sequencing-derived signatures and proposed IFN/STAT1 signaling as a candidate therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Eric P. Souto
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Cancer and Cell Biology Graduate Program, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Ping Gong
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - John D. Landua
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | | | - Abhinaya Ganesan
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Cancer and Cell Biology Graduate Program, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Lacey E. Dobrolecki
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Stephen C. Purdy
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (UC-AMC), Aurora, Colorado
- Pharmacology Graduate Program, UC-AMC, Aurora, Colorado
- University of Colorado Cancer Center, UC-AMC, Aurora, Colorado
| | - Xingxin Pan
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, Texas
| | - Michael Zeosky
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, Texas
| | - Anna Chung
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, Texas
| | - S. Stephen Yi
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, Texas
| | - Heide L. Ford
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (UC-AMC), Aurora, Colorado
- Pharmacology Graduate Program, UC-AMC, Aurora, Colorado
- University of Colorado Cancer Center, UC-AMC, Aurora, Colorado
| | - Michael T. Lewis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Cancer and Cell Biology Graduate Program, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Department of Radiology, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
9
|
Wang YQ, Wang S, Yi HM, Qian Y, Wang Y, Xu HM, Xu-Monette ZY, Au K, Tian S, Dong Y, Zhao J, Fu D, Mu RJ, Wang SY, Wang L, Young KH, Xu PP, Zhao WL. Practical microenvironment classification in diffuse large B cell lymphoma using digital pathology. Cell Rep Med 2025; 6:102030. [PMID: 40112808 PMCID: PMC12047489 DOI: 10.1016/j.xcrm.2025.102030] [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: 10/07/2024] [Revised: 01/15/2025] [Accepted: 02/24/2025] [Indexed: 03/22/2025]
Abstract
Diffuse large B cell lymphoma (DLBCL) is a heterogeneous B cell neoplasm with variable clinical outcomes influenced by both tumor-derived and lymphoma microenvironment (LME) alterations. A recent transcriptomic study identifies four DLBCL subtypes based on LME characteristics: germinal center (GC)-like, mesenchymal (MS), inflammatory (IN), and depleted (DP). However, integrating this classification into clinical practice remains challenging. Here, we utilize deconvolution methods to assess microenvironment component abundance, establishing an LME classification of DLBCL using immunohistochemistry markers and digital pathology based on CD3, CD8, CD68, PD-L1, and collagen. This staining-based algorithm demonstrates over 80% concordance with transcriptome-based classification. Single-cell sequencing confirms that the immune microenvironments distinguished by this algorithm align with transcriptomic profiles. Significant disparities in overall and progression-free survival are observed among LME subtypes following rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) or R-CHOP with targeted agents (R-CHOP-X) immunochemotherapy. LME subtypes differed from distinct immune escape mechanisms, highlighting specific immunotherapeutic targets and supporting application of this classification in future precision medicine trials.
Collapse
Affiliation(s)
- Yu-Qing Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shuo Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong-Mei Yi
- Department of Pathology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Qian
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hai-Min Xu
- Department of Pathology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zijun Y Xu-Monette
- Hematopathology Division and Department of Pathology, Duke University Medical Center, Durham, NC, USA; Duke Cancer Institute, Durham, NC, USA
| | - Kelly Au
- Hematopathology Division and Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Shuang Tian
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Dong
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Zhao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Di Fu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rong-Ji Mu
- Department of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shu-Ye Wang
- Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Li Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai, China
| | - Ken H Young
- Hematopathology Division and Department of Pathology, Duke University Medical Center, Durham, NC, USA; Duke Cancer Institute, Durham, NC, USA.
| | - Peng-Peng Xu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Wei-Li Zhao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai, China.
| |
Collapse
|
10
|
Goldkamp AK, Atchison RG, Falkenberg SM, Dassanayake RP, Neill JD, Casas E. Host transcriptome response to Mycoplasma bovis and bovine viral diarrhea virus in bovine tissues. BMC Genomics 2025; 26:361. [PMID: 40211134 PMCID: PMC11987210 DOI: 10.1186/s12864-025-11549-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 03/28/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND Mycoplasma bovis is a prominent pathogen associated with respiratory disease in livestock. Respiratory disease in cattle often involves co-infection, where a primary viral infection can weaken the host immune system and thus enhance subsequent bacterial infection. The objective of this study was to investigate changes in the host (cattle) transcriptome during bacterial-viral co-infection. RNA sequencing was done in whole blood cells (WBC), liver, mesenteric lymph node (MLN), tracheal-bronchial lymph node (TBLN), spleen, and thymus collected from Control animals (n = 2), animals infected with M. bovis (MB; n = 3), and animals infected with M. bovis and bovine viral diarrhea virus (BVDV) (Dual; n = 3). RESULTS Thymus and spleen had the greatest number of differentially expressed genes (DEGs) out of all tissues analyzed. In spleen, genes involved in maintenance of the extracellular matrix (ECM) including collagen type XV alpha 1 chain (COL15A1), collagen type IV alpha 2 chain (COL4A2), and heparan sulfate proteoglycan 2 (HSPG2) were the most significantly downregulated in Dual compared to Control and MB. In thymus, complement 3 (C3) was a highly significant DEG and upregulated in Dual compared to Control and MB. Interferon alpha inducible protein 6 (IFI6) and interferon-induced transmembrane proteins (IFITM1 and IFITM3), were significantly associated with infection status and upregulated in spleen and thymus of Dual compared to Control and MB. CONCLUSION Downregulation of ECM components may cause degradation of the ECM and contribute to increased viral spread due to co-infection. Hyperactivation of complement pathway genes may contribute to damage to the thymus and influence severity of co-infection. Co-expression of IFI6, IFITM1 and IFITM3 across lymphoid tissues may be connected to enhanced pathogenesis in co-infection. These findings suggest co-infection exacerbates disease severity through modulation of ECM components in spleen and complement and coagulation cascades in the thymus. These impacted pathways may underlie thymic atrophy and impaired pathogen clearance due to BVDV and M. bovis co-infection.
Collapse
Affiliation(s)
- Anna K Goldkamp
- Ruminant Diseases and Immunology Research Unit, National Animal Disease Center, Agricultural Research Service, Department of Agriculture, Ames, IA, USA
| | - Randy G Atchison
- Ruminant Diseases and Immunology Research Unit, National Animal Disease Center, Agricultural Research Service, Department of Agriculture, Ames, IA, USA
- Animal Plant Health Inspection Service, Department of Agriculture, Centers for Veterinary Biologics, Ames, IA, USA
| | - Shollie M Falkenberg
- Ruminant Diseases and Immunology Research Unit, National Animal Disease Center, Agricultural Research Service, Department of Agriculture, Ames, IA, USA
- College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Rohana P Dassanayake
- Ruminant Diseases and Immunology Research Unit, National Animal Disease Center, Agricultural Research Service, Department of Agriculture, Ames, IA, USA
| | - John D Neill
- Ruminant Diseases and Immunology Research Unit, National Animal Disease Center, Agricultural Research Service, Department of Agriculture, Ames, IA, USA
| | - Eduardo Casas
- Ruminant Diseases and Immunology Research Unit, National Animal Disease Center, Agricultural Research Service, Department of Agriculture, Ames, IA, USA.
| |
Collapse
|
11
|
Munhoz DD, Fonseca DLM, Filgueiras IS, Dias HD, Nakaya HI, Jurisica I, Ochs HD, Schimke LF, Rizzo LV, Cabral-Marques O. Integrative immunology identified interferome signatures in uveitis and systemic disease-associated uveitis. Front Immunol 2025; 16:1509805. [PMID: 40270958 PMCID: PMC12014655 DOI: 10.3389/fimmu.2025.1509805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 03/10/2025] [Indexed: 04/25/2025] Open
Abstract
Introduction Uveitis accounts for up to 25% of global legal blindness and involves intraocular inflammation, classifed as infectious or non-infectious. Its complex pathophysiology includes dysregulated cytokines, particularly interferons (IFNs). However, the global signature of type I, II, and III interferon-regulated genes (Interferome) remains largely uncharacterized in uveitis. Methods In this study, we conducted an integrative systems biology analysis of blood transcriptome data from 169 non-infectious uveitis patients (56 isolated uveitis, 113 systemic disease-associated uveitis) and 82 healthy controls. Results Modular co-expression analysis identified distinct cytokine signaling networks, emphasizing interleukin and interferon pathways. A meta-analysis revealed 110 differentially expressed genes (metaDEGs) in isolated uveitis and 91 in systemic disease-associated uveitis, predominantly linked to immune responses. The Interferome database confirmed a predominance of type I and II IFN signatures in both groups. Pathway enrichment analysis highlighted inflammatory responses, including cytokine production (IL-8, IL1-β, IFN-γ, β, and α) and toll-like receptor signaling (TLR4, TLR7, TLR8, CD180). Principal component analysis emphasized the IFN signature's discriminative power, particularly in systemic disease-associated uveitis. Machine learning identified IFN-associated genes as robust predictors, while linear discriminant analysis pinpointed CCR2, CD180, GAPT, and PTGS2 as key risk factors in isolated uveitis and CA1, SIAH2, and PGS in systemic disease-associated uveitis. Conclusion These findings highlight IFN-driven imune dysregulation and potential molecular targets for precision therapies in uveitis.
Collapse
Affiliation(s)
- Danielle Dias Munhoz
- Experimental Biology Laboratory Prof. Dr. Geraldo Medeiros-Neto, Hospital Israelita Albert Einsten, Sao Paulo, Brazil
| | - Dennyson Leandro M. Fonseca
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Igor Salerno Filgueiras
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Haroldo Dutra Dias
- Department of Neuroscience, Institute of Biomedical Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Helder I. Nakaya
- Experimental Biology Laboratory Prof. Dr. Geraldo Medeiros-Neto, Hospital Israelita Albert Einsten, Sao Paulo, Brazil
| | - Igor Jurisica
- Division of Orthopaedics, Osteoarthritis Research Program, Schroeder Arthritis Institute, and Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Departments of Medical Biophysics and Computer Science, and Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Hans D. Ochs
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States
- Seattle Children’s Research Institute, Seattle, WA, United States
| | - Lena F. Schimke
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Luiz Vicente Rizzo
- Experimental Biology Laboratory Prof. Dr. Geraldo Medeiros-Neto, Hospital Israelita Albert Einsten, Sao Paulo, Brazil
| | - Otavio Cabral-Marques
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), University of Sao Paulo (USP), Sao Paulo, SP, Brazil
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
- Network of Immunity in Infection, Malignancy, and Autoimmunity (NIIMA), Universal Scientific Education and Research Network, (USERN), Sao Paulo, Brazil
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo (USP), Sao Paulo, Brazil
- D’Or Institute for Research and Education (IDOR), São Paulo, Brazil
- Department of Medicine, Division of Molecular Medicine, Laboratory of Medical Investigation 29, University of São Paulo, Sao Paulo, Brazil
| |
Collapse
|
12
|
Chen H, Charles PD, Gu Q, Liberatori S, Robertson DL, Palmarini M, Wilson SJ, Mohammed S, Castello A. Omics Analyses Uncover Host Networks Defining Virus-Permissive and -Hostile Cellular States. Mol Cell Proteomics 2025; 24:100966. [PMID: 40204275 DOI: 10.1016/j.mcpro.2025.100966] [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: 11/15/2024] [Revised: 03/24/2025] [Accepted: 04/04/2025] [Indexed: 04/11/2025] Open
Abstract
The capacity of host cells to sustain or restrict virus infection is influenced by their proteome. Understanding the compendium of proteins defining cellular permissiveness is key to many questions in fundamental virology. Here, we apply a multi-omic approach to determine the proteins that are associated with highly permissive, intermediate, and hostile cellular states. We observed two groups of differentially regulated genes: (i) with robust changes in mRNA and protein levels and (ii) with protein/RNA discordances. While many of the latter are classified as interferon-stimulated genes (ISGs), most exhibit no antiviral effects in overexpression screens. This suggests that IFN-dependent protein changes can be better indicators of antiviral function than mRNA levels. Phosphoproteomics revealed an additional regulatory layer involving non-signaling proteins with altered phosphorylation. Indeed, we confirmed that several permissiveness-associated proteins with changes in abundance or phosphorylation regulate infection fitness. Altogether, our study provides a comprehensive and systematic map of the cellular alterations driving virus susceptibility.
Collapse
Affiliation(s)
- Honglin Chen
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK; Department of Biochemistry, University of Oxford, Oxford, UK
| | | | - Quan Gu
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | | | | | | | - Sam J Wilson
- Cambridge Institute of Therapeutic Immunol & Infect Disease, Jeffrey Cheah Biomedical Centre, Cambridge, UK
| | - Shabaz Mohammed
- Department of Biochemistry, University of Oxford, Oxford, UK; The Rosalind Franklin Institute, Oxfordshire, UK; Department of Chemistry, University of Oxford, Oxford, UK.
| | - Alfredo Castello
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK.
| |
Collapse
|
13
|
Ernst IVS, Lehtonen L, Nilsson SM, Nielsen FL, Marcher AB, Mandrup S, Madsen JGS. Single Nucleus Multiome Analysis Reveals Early Inflammatory Response to High-Fat Diet in Mouse Pancreatic Islets. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.01.646568. [PMID: 40236154 PMCID: PMC11996447 DOI: 10.1101/2025.04.01.646568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
In periods of sustained hyper-nutrition, pancreatic β-cells undergo functional compensation through transcriptional upregulation of gene programs driving insulin secretion. This adaptation is essential for maintaining systemic glucose homeostasis and metabolic health. Using single nuclei multiomics, we have mapped the early transcriptional compensation mechanisms in murine islets of Langerhans exposed to high-fat diet (HFD) for one and three weeks. We show that β-cells exhibit the largest transcriptional response to HFD, characterized by early activation of proinflammatory eRegulons and downregulation of β-cell identity genes, particularly in a distinct subset of β-cells. Our observations translate to humans, as we observe an increase in the inflammatory gene signatures in human β-cells in pre-diabetes and diabetes. Collectively, these observations point to cellular cross-talk through proinflammatory signaling as a central and early driver of β-cell dysfunction that limits the compensatory capacity of β-cells, which is closely linked to the development of diabetes.
Collapse
|
14
|
Keuls RA, Ochsner SA, O'Neill MB, O'Day DR, Miyauchi A, Campbell KM, Lanners N, Goldstein JA, Yee C, McKenna NJ, Parchem RJ, Parchem JG. Single-nucleus transcriptional profiling of the placenta reveals the syncytiotrophoblast stress response to COVID-19. Am J Obstet Gynecol 2025; 232:S160-S175.e7. [PMID: 40253079 DOI: 10.1016/j.ajog.2025.01.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND COVID-19 in pregnancy is associated with placental immune activation, inflammation, and vascular malperfusion, but its impact on syncytiotrophoblast biology and function is unclear. OBJECTIVE This study aimed to determine the effects of maternal COVID-19 on placental syncytiotrophoblasts using single-nucleus transcriptional profiling and to compare placental stress responses in COVID-19 and preeclampsia. STUDY DESIGN For transcriptional characterization of syncytiotrophoblasts, we used the single-nucleus RNA sequencing platform, single-cell combinatorial indexing RNA sequencing (sci-RNA-seq3), to profile placental villi and fetal membranes from unvaccinated patients with symptomatic COVID-19 at birth (n = 4), gestational age-matched controls (n = 4), and a case of critical COVID-19 in the second trimester with delivery at term (n = 1). Clustering of nuclei and differential gene expression analysis was performed in Seurat. Gene ontology analysis was conducted using Enrichr. High-confidence transcriptional target analysis was used to identify key transcription factor nodes governing the syncytiotrophoblast response to maternal SARS-CoV-2 infection. Bioinformatic approaches were further used to compare the COVID-19 dataset to published preeclampsia gene signatures. Tissue analysis, including immunofluorescence, was conducted to validate the transcriptional data and to compare COVID-19 and preeclampsia placental histology for an expanded cohort of placentas: controls (n = 6), asymptomatic COVID-19 (n = 3), symptomatic COVID-19 (n = 5), and preeclampsia with severe features (n = 7). RESULTS The analyzed dataset comprised 15 cell clusters and 47,889 nuclei. We identified 3 clusters of syncytiotrophoblasts representing fusing and mature nuclei with overlapping but distinct transcriptional responses to COVID-19. Bioinformatic analyses indicated that COVID-19 is associated with the following alterations in syncytiotrophoblasts: (1) endoplasmic reticulum stress and activation of stress signaling pathways, including the unfolded protein response and integrated stress response; (2) regulation of gene expression by CCAAT/enhancer-binding protein beta (CEBPB), a master transcription factor of the syncytiotrophoblast lineage; and (3) upregulation of preeclampsia-associated genes. Using complementary methods, we confirmed increased levels of stress response proteins (eg, BiP, G3BP1) in syncytiotrophoblasts, unfolded protein response signaling (spliced XBP1 mRNA), and CEBPB activation (phosphorylation) in COVID-19. Increased cytotrophoblast proliferation (Ki-67) was also detected in COVID-19, consistent with a trophoblast response to injury. Markers of stress detected in preeclampsia demonstrated similarities in the placental stress phenotype of COVID-19 and preeclampsia. CONCLUSION Maternal COVID-19 is associated with syncytiotrophoblast endoplasmic reticulum stress and activation of the syncytiotrophoblast lineage transcription factor, CEBPB. Similarities between syncytiotrophoblast stress in COVID-19 and preeclampsia provide insights into their clinical association.
Collapse
Affiliation(s)
- Rachel A Keuls
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX; Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX; Department of Neuroscience, Baylor College of Medicine, Houston, TX
| | - Scott A Ochsner
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Mary B O'Neill
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA
| | - Diana R O'Day
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA
| | - Akihiko Miyauchi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX; Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX; Department of Neuroscience, Baylor College of Medicine, Houston, TX
| | - Kadeshia M Campbell
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX
| | - Natalie Lanners
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX
| | - Jeffery A Goldstein
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Connor Yee
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX; Larry C. Gilstrap MD Center for Perinatal and Women's Health Research, The University of Texas Health Science Center at Houston, Houston, TX
| | - Neil J McKenna
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Ronald J Parchem
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX; Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX; Department of Neuroscience, Baylor College of Medicine, Houston, TX.
| | - Jacqueline G Parchem
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX.
| |
Collapse
|
15
|
Dumesic PA, Wilensky SE, Bose S, Van Vranken JG, Gygi SP, Spiegelman BM. RBM43 controls PGC1α translation and a PGC1α-STING signaling axis. Cell Metab 2025; 37:742-757.e8. [PMID: 39965564 PMCID: PMC11885043 DOI: 10.1016/j.cmet.2025.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 10/17/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025]
Abstract
Obesity is associated with systemic inflammation that impairs mitochondrial function. This disruption curtails oxidative metabolism, limiting adipocyte lipid metabolism and thermogenesis, a metabolically beneficial program that dissipates chemical energy as heat. Here, we show that PGC1α, a key governor of mitochondrial biogenesis, is negatively regulated at the level of its mRNA translation by the RNA-binding protein RBM43. RBM43 is induced by inflammatory cytokines and suppresses mitochondrial biogenesis in a PGC1α-dependent manner. In mice, adipocyte-selective Rbm43 disruption elevates PGC1α translation and oxidative metabolism. In obesity, Rbm43 loss improves glucose tolerance, reduces adipose inflammation, and suppresses activation of the innate immune sensor cGAS-STING in adipocytes. We further identify a role for PGC1α in safeguarding against cytoplasmic accumulation of mitochondrial DNA, a cGAS ligand. The action of RBM43 defines a translational regulatory axis by which inflammatory signals dictate cellular energy metabolism and contribute to metabolic disease pathogenesis.
Collapse
Affiliation(s)
- Phillip A Dumesic
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Sarah E Wilensky
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Symanthika Bose
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | | | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Bruce M Spiegelman
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
16
|
Hiew JY, Lim YS, Liu H, Ng CS. Integrated transcriptomic profiling reveals a STING-mediated Type II Interferon signature in SOD1-mutant amyotrophic lateral sclerosis models. Commun Biol 2025; 8:347. [PMID: 40025162 PMCID: PMC11873215 DOI: 10.1038/s42003-025-07790-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 02/20/2025] [Indexed: 03/04/2025] Open
Abstract
Inflammation is a hallmark of amyotrophic lateral sclerosis (ALS), particularly in cases with SOD1 mutations. Using integrative transcriptomics, we analyzed gene expression changes in mouse models throughout progression, human induced-pluripotent stem cells (hiPSCs), and post-mortem spinal cord tissue from ALS patients. We identified a conserved upregulation of interferon (IFN) genes and IFN-stimulating genes (ISGs) in both mouse models and human ALS, with a predominance Type I IFNs (IFN-α/β) in mice and Type II IFNs (IFN-γ) in humans. In mouse models, we observed robust and sustained upregulation of Type I and II ISGs, including ATF3, beginning at disease onset stage and persisting throughout disease progression. Single-cell transcriptomics further pinpointed vascular endothelial cells as a major source of ISGs. Furthermore, we found that the STING-TBK1 axis is essential for the induction of Type II ISGs in ALS, as its deletion impaired their expression. Our study uncovers a conserved ISGs signature across ALS models and patients, highlighting the potential role of innate immune activation in ALS pathogenesis. These findings suggest that ISGs may serve as potential biomarkers and therapeutic targets for ALS.
Collapse
Affiliation(s)
- Jen Young Hiew
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Selangor, Malaysia
| | - Yi Shan Lim
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Selangor, Malaysia
| | - Huitao Liu
- School of Biological Engineering, College of Advanced Interdisciplinary Science and Technology, Henan University of Technology, 100 Lianhua Street, Zhongyuan District, Zhengzhou, 450001, China
| | - Chen Seng Ng
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Selangor, Malaysia.
| |
Collapse
|
17
|
Karjalainen A, Witalisz-Siepracka A, Prchal-Murphy M, Martin D, Sternberg F, Krunic M, Dolezal M, Fortelny N, Farlik M, Macho-Maschler S, Lassnig C, Meissl K, Amenitsch L, Lederer T, Pohl E, Gotthardt D, Bock C, Decker T, Strobl B, Müller M. Cell-type-specific requirement for TYK2 in murine immune cells under steady state and challenged conditions. Cell Mol Life Sci 2025; 82:98. [PMID: 40025196 PMCID: PMC11872851 DOI: 10.1007/s00018-025-05625-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/31/2025] [Accepted: 02/17/2025] [Indexed: 03/04/2025]
Abstract
Tyrosine kinase 2 (TYK2) deficiency and loss or inhibition of kinase activity in men and mice leads to similar immune compromised phenotypes, predominantly through impairment of interferon (IFN) and interleukin 12 family responses. Here we relate the transcriptome changes to phenotypical changes observed in TYK2-deficient (Tyk2-/-) and TYK2 kinase-inactive (Tyk2K923E) mice in naïve splenic immune cells and upon ex vivo IFN treatment or in vivo tumor transplant infiltration. The TYK2 activities under homeostatic and both challenged conditions are highly cell-type-specific with respect to quantity and quality of transcriptionally dependent genes. The major impact of loss of TYK2 protein or kinase activity in splenic homeostatic macrophages, NK and CD8+ T cells and tumor-derived cytolytic cells is on IFN responses. While reportedly TYK2 deficiency leads to partial impairment of IFN-I responses, we identified cell-type-specific IFN-I-repressed gene sets completely dependent on TYK2 kinase activity. Reported kinase-inactive functions of TYK2 relate to signaling crosstalk, metabolic functions and cell differentiation or maturation. None of these phenotypes relates to respective enriched gene sets in the TYK2 kinase-inactive cell types. Nonetheless, the scaffolding functions of TYK2 are capable to change transcriptional activities at single gene levels and chromatin accessibility at promoter-distal regions upon cytokine treatment most prominently in CD8+ T cells. The cell-type-specific transcriptomic and epigenetic effects of TYK2 shed new light on the biology of this JAK family member and are relevant for current and future treatment of autoimmune and inflammatory diseases with TYK2 inhibitors.
Collapse
Affiliation(s)
- Anzhelika Karjalainen
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Agnieszka Witalisz-Siepracka
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
- Division Pharmacology, Karl Landsteiner University of Health Sciences, Krems an Der Donau, Austria
| | - Michaela Prchal-Murphy
- Pharmacology and Toxicology, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - David Martin
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Felix Sternberg
- Physiology and Biophysics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
- Department of Nutritional Sciences, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Milica Krunic
- Campus Tulln, University of Applied Sciences Wiener Neustadt, Wiener Neustadt, Austria
| | - Marlies Dolezal
- Platform Biostatistics and Bioinformatics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Nikolaus Fortelny
- Department of Biosciences and Medical Biology, Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, Austria
| | - Matthias Farlik
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Sabine Macho-Maschler
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Caroline Lassnig
- Core Facility VetBiomodels, University of Veterinary Medicine, Vienna, Austria
| | - Katrin Meissl
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Lena Amenitsch
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Therese Lederer
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Elena Pohl
- Physiology and Biophysics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Dagmar Gotthardt
- Division Pharmacology, Karl Landsteiner University of Health Sciences, Krems an Der Donau, Austria
| | - Christoph Bock
- Cemm Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Institute of Artificial Intelligence, Center for Medical Data Science, Medical University of Vienna, Vienna, Austria
| | - Thomas Decker
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Center for Molecular Biology, Department of Microbiology, Immunobiology and Genetics, University of Vienna, Vienna, Austria
| | - Birgit Strobl
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Mathias Müller
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria.
| |
Collapse
|
18
|
Lu Y, Gao L, Yang Y, Shi D, Zhang Z, Wang X, Huang Y, Wu J, Meng J, Li H, Yan D. Protective role of mitophagy on microglia-mediated neuroinflammatory injury through mtDNA-STING signaling in manganese-induced parkinsonism. J Neuroinflammation 2025; 22:55. [PMID: 40022162 PMCID: PMC11869743 DOI: 10.1186/s12974-025-03396-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 02/23/2025] [Indexed: 03/03/2025] Open
Abstract
Manganese (Mn), the third most abundant transition metal in the earth's crust, has widespread applications in the emerging field of organometallic catalysis and traditional industries. Excessive Mn exposure causes neurological syndrome resembling Parkinson's disease (PD). The pathogenesis of PD is thought to involve microglia-mediated neuroinflammatory injury, with mitochondrial dysfunction playing a role in aberrant microglial activation. In the early stages of PD, PINK1/Parkin-mediated mitophagy contributes to the microglial inflammatory response via the cGAS/STING signaling pathway. Suppression of PINK1/Parkin-mediated mitophagy due to excessive Mn exposure exacerbates neuronal injury. Moreover, excessive Mn exposure leads to neuroinflammatory damage via the microglial cGAS-STING pathway. However, the precise role of microglial mitophagy in modulating neuroinflammation in Mn-induced parkinsonism and its underlying molecular mechanism remains unclear. Here, we observed that Mn-exposed mice exhibited neurobehavioral abnormalities and detrimental microglial activation, along with increased apoptosis of nerve cells, proinflammatory cytokines, and intracellular ROS. Furthermore, in vivo and in vitro experiments showed that excessive Mn exposure resulted in microglial mitochondrial dysfunction, manifested by increased mitochondrial ROS, decreased mitochondrial mass, and membrane potential. Additionally, with the escalating Mn dose, PINK1/Parkin-mediated mitophagy changed from activation to suppression. This was evidenced by decreased levels of LC3-II, PINK1, p-Parkin/Parkin, and increased levels of p62 protein expression level, as well as the colocalization between ATPB and LC3B due to excessive Mn exposure. Upregulation of mitophagy by urolithin A could mitigate Mn-induced mitochondrial dysfunction, as indicated by decreased mitochondrial ROS, increased mitochondrial mass, and membrane potential, along with improvements in neurobehavioral deficits and attenuated detrimental microglial activation. Using single-nucleus RNA-sequencing (snRNA-seq) analysis in the Mn-exposed mouse model, we identified the microglial cGAS-STING signaling pathway as a potential mechanism underlying Mn-induced neuroinflammation. This pathway is associated with an increase in cytosolic mtDNA levels, which activate STING signaling. These findings point to the induction of microglial mitophagy as a viable strategy to alleviate Mn-induced neuroinflammation through mtDNA-STING signaling.
Collapse
Affiliation(s)
- Yang Lu
- School of Public Health, Jinzhou Medical University, Section III, Linghe District, Jinzhou, China
| | - Liang Gao
- School of Public Health, Jinzhou Medical University, Section III, Linghe District, Jinzhou, China
- Collaborative Innovation Center For Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou, China
| | - Yuqing Yang
- School of Public Health, Jinzhou Medical University, Section III, Linghe District, Jinzhou, China
| | - Dihang Shi
- School of Public Health, Jinzhou Medical University, Section III, Linghe District, Jinzhou, China
| | - Zhipeng Zhang
- School of Public Health, Jinzhou Medical University, Section III, Linghe District, Jinzhou, China
| | - Xiaobai Wang
- School of Public Health, Jinzhou Medical University, Section III, Linghe District, Jinzhou, China
| | - Ying Huang
- School of Public Health, Jinzhou Medical University, Section III, Linghe District, Jinzhou, China
| | - Jie Wu
- School of Public Health, Jinzhou Medical University, Section III, Linghe District, Jinzhou, China
| | - Jia Meng
- School of Public Health, Jinzhou Medical University, Section III, Linghe District, Jinzhou, China
- Collaborative Innovation Center For Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou, China
| | - Hong Li
- School of Public Health, Jinzhou Medical University, Section III, Linghe District, Jinzhou, China
- Collaborative Innovation Center For Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou, China
| | - Dongying Yan
- School of Public Health, Jinzhou Medical University, Section III, Linghe District, Jinzhou, China.
- Collaborative Innovation Center For Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou, China.
| |
Collapse
|
19
|
Ravenhill BJ, Oliveira M, Wood G, Di Y, Kite J, Wang X, Davies CTR, Lu Y, Antrobus R, Elliott G, Irigoyen N, Hughes DJ, Lyons PA, Chung B, Borner GHH, Weekes MP. Spatial proteomics identifies a CRTC-dependent viral signaling pathway that stimulates production of interleukin-11. Cell Rep 2025; 44:115263. [PMID: 39921859 DOI: 10.1016/j.celrep.2025.115263] [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: 05/02/2024] [Revised: 12/09/2024] [Accepted: 01/12/2025] [Indexed: 02/10/2025] Open
Abstract
Appropriate cellular recognition of viruses is essential for the generation of an effective innate and adaptive immune response. Viral sensors and their downstream signaling components thus provide a crucial first line of host defense. Many of them exhibit subcellular relocalization upon activation, resulting in the expression of interferon and antiviral genes. To comprehensively identify signaling factors, we analyzed protein relocalization on a global scale during viral infection. cAMP-responsive element-binding protein (CREB)-regulated transcription coactivators 2 and 3 (CRTC2/3) exhibited early cytoplasmic-to-nuclear translocation upon infection with multiple viruses in diverse cell types. This movement was dependent on mitochondrial antiviral signaling protein (MAVS), cyclo-oxygenase proteins, and protein kinase A. A key effect of CRTC2/3 translocation is transcription of the fibro-inflammatory cytokine interleukin (IL)-11. This may be important clinically in viral infections associated with fibrosis, including SARS-CoV-2. Nuclear translocation of CRTC2/3 is, therefore, identified as an important pathway in the context of viral infection.
Collapse
Affiliation(s)
- Benjamin J Ravenhill
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Marisa Oliveira
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - George Wood
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Ying Di
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Joanne Kite
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Xinyue Wang
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Colin T R Davies
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Yongxu Lu
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Robin Antrobus
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Gill Elliott
- Department of Microbial Sciences, School of Biosciences, University of Surrey, Guildford, UK
| | - Nerea Irigoyen
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - David J Hughes
- School of Biology, University of St. Andrews, St. Andrews, UK
| | - Paul A Lyons
- Department of Medicine, University of Cambridge, Cambridge, UK; Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Betty Chung
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Georg H H Borner
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, Martinsried, Germany
| | - Michael P Weekes
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
20
|
Xu Z, Kuhlmann-Hogan A, Xu S, Tseng H, Chen D, Tan S, Sun M, Tripple V, Bosenberg M, Miller-Jensen K, Kaech SM. Scavenger Receptor CD36 in Tumor-Associated Macrophages Promotes Cancer Progression by Dampening Type-I IFN Signaling. Cancer Res 2025; 85:462-476. [PMID: 39546763 PMCID: PMC11788022 DOI: 10.1158/0008-5472.can-23-4027] [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: 12/19/2023] [Revised: 06/25/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
Tumor-associated macrophages (TAM) are a heterogeneous population of myeloid cells that dictate the inflammatory tone of the tumor microenvironment. In this study, we unveiled a mechanism by which scavenger receptor cluster of differentiation 36 (CD36) suppresses TAM inflammatory states. CD36 was upregulated in TAMs and associated with immunosuppressive features, and myeloid-specific deletion of CD36 significantly reduced tumor growth. Moreover, CD36-deficient TAMs acquired inflammatory signatures including elevated type-I IFN (IFNI) production, mirroring the inverse correlation between CD36 and IFNI response observed in patients with cancer. IFNI, especially IFNβ, produced by CD36-deficient TAMs directly induced tumor cell quiescence and delayed tumor growth. Mechanistically, CD36 acted as a natural suppressor of IFNI signaling in macrophages through p38 activation downstream of oxidized lipid signaling. These findings establish CD36 as a critical regulator of TAM function and the tumor inflammatory microenvironment, providing additional rationale for pharmacologic inhibition of CD36 to rejuvenate antitumor immunity. Significance: CD36 in tumor-associated macrophages mediates immunosuppression and can be targeted as a therapeutic avenue for stimulating interferon production and increasing the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Ziyan Xu
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, California
- School of Biological Sciences, University of California San Diego, La Jolla, California
| | - Alexandra Kuhlmann-Hogan
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, California
| | - Shihao Xu
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, California
| | - Hubert Tseng
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, California
| | - Dan Chen
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, California
| | - Shirong Tan
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, California
| | - Ming Sun
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, California
| | - Victoria Tripple
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, California
| | - Marcus Bosenberg
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut
- Yale Center for Precision Cancer Modeling, Yale School of Medicine, New Haven, Connecticut
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut
| | - Kathryn Miller-Jensen
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut
| | - Susan M. Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, California
| |
Collapse
|
21
|
Vasou A, Nightingale K, Cetkovská V, Scheler J, Bamford CGG, Andrejeva J, Rowe JC, Swatek KN, Schwarz‐Linek U, Randall RE, McLauchlan J, Weekes MP, Bogunovic D, Hughes DJ. ISG15-Dependent Stabilisation of USP18 Is Necessary but Not Sufficient to Regulate Type I Interferon Signalling in Humans. Eur J Immunol 2025; 55:e202451651. [PMID: 39931755 PMCID: PMC11811815 DOI: 10.1002/eji.202451651] [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: 11/08/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 02/13/2025]
Abstract
Type I interferon (IFN) signalling induces the expression of several hundred IFN-stimulated genes (ISGs) that provide an unfavourable environment for viral replication. To prevent an overexuberant response and autoinflammatory disease, IFN signalling requires tight control. One critical regulator is the ubiquitin-like protein IFN-stimulated gene 15 (ISG15), evidenced by autoinflammatory disease in patients with inherited ISG15 deficiencies. Current models suggest that ISG15 stabilises ubiquitin-specific peptidase 18 (USP18), a well-established negative regulator of IFN signalling. USP18 also functions as an ISG15-specific peptidase that cleaves ISG15 from ISGylated proteins; however, USP18's catalytic activity is dispensable for controlling IFN signalling. Here, we show that the ISG15-dependent stabilisation of USP18 involves hydrophobic interactions reliant on tryptophan 123 (W123) in ISG15. Nonetheless, while USP18 stabilisation is necessary, it is not sufficient for the regulation of IFN signalling; ISG15 C-terminal mutants with significantly reduced affinity still stabilised USP18, yet the magnitude of signalling resembled ISG15-deficient cells. Hence, USP18 requires non-covalent interactions with the ISG15 C-terminal diGlycine motif to promote its regulatory function. It shows ISG15 is a repressor of type I IFN signalling beyond its role as a USP18 stabiliser.
Collapse
Affiliation(s)
- Andri Vasou
- Biomedical Sciences Research ComplexSchool of BiologyUniversity of St AndrewsSt AndrewsUK
| | - Katie Nightingale
- Department of MedicineCambridge Institute for Medical ResearchUniversity of CambridgeCambridgeUK
| | - Vladimíra Cetkovská
- Biomedical Sciences Research ComplexSchool of BiologyUniversity of St AndrewsSt AndrewsUK
| | - Jonathan Scheler
- Biomedical Sciences Research ComplexSchool of BiologyUniversity of St AndrewsSt AndrewsUK
| | - Connor G. G. Bamford
- Medical Research Council‐University of Glasgow Centre for Virus ResearchGlasgowUK
| | - Jelena Andrejeva
- Biomedical Sciences Research ComplexSchool of BiologyUniversity of St AndrewsSt AndrewsUK
| | - Jessica C. Rowe
- Medical Research Council Protein Phosphorylation and Ubiquitylation UnitSchool of Life SciencesUniversity of DundeeDundeeUK
| | - Kirby N. Swatek
- Medical Research Council Protein Phosphorylation and Ubiquitylation UnitSchool of Life SciencesUniversity of DundeeDundeeUK
| | - Ulrich Schwarz‐Linek
- Biomedical Sciences Research ComplexSchool of BiologyUniversity of St AndrewsSt AndrewsUK
| | - Richard E. Randall
- Biomedical Sciences Research ComplexSchool of BiologyUniversity of St AndrewsSt AndrewsUK
| | - John McLauchlan
- Medical Research Council‐University of Glasgow Centre for Virus ResearchGlasgowUK
| | - Michael P. Weekes
- Department of MedicineCambridge Institute for Medical ResearchUniversity of CambridgeCambridgeUK
| | - Dusan Bogunovic
- Department of PediatricsColumbia University Medical CenterNew YorkNew YorkUSA
| | - David J. Hughes
- Biomedical Sciences Research ComplexSchool of BiologyUniversity of St AndrewsSt AndrewsUK
| |
Collapse
|
22
|
Argoetti A, Shalev D, Polyak G, Shima N, Biran H, Lahav T, Hashimshony T, Mandel-Gutfreund Y. lncRNA NORAD modulates STAT3/STAT1 balance and innate immune responses in human cells via interaction with STAT3. Nat Commun 2025; 16:571. [PMID: 39794357 PMCID: PMC11723954 DOI: 10.1038/s41467-025-55822-0] [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: 08/01/2023] [Accepted: 12/30/2024] [Indexed: 01/13/2025] Open
Abstract
Long non-coding RNAs (lncRNAs) are pivotal regulators of cellular processes. Here we reveal an interaction between the lncRNA NORAD, noted for its role in DNA stability, and the immune related transcription factor STAT3 in embryonic and differentiated human cells. Results from NORAD knockdown experiments implicate NORAD in facilitating STAT3 nuclear localization and suppressing antiviral gene activation. In NORAD-deficient cells, STAT3 remains cytoplasmic, allowing STAT1 to enhance antiviral activity. Analysis of RNA expression data from in vitro experiments and clinical samples demonstrates reduced NORAD upon viral infection. Additionally, evolutionary conservation analysis suggests that this regulatory function of NORAD is restricted to humans, potentially owing to the introduction of an Alu element in hominoids. Our findings thus suggest that NORAD functions as a modulator of STAT3-mediated immune suppression, adding to the understanding of lncRNAs in immune regulation and evolutionary adaptation in host defense mechanisms.
Collapse
Affiliation(s)
- Amir Argoetti
- Technion-Israel Institute of Technology, Faculty of Biology, Emerson building, Haifa, Israel
| | - Dor Shalev
- Technion-Israel Institute of Technology, Faculty of Biology, Emerson building, Haifa, Israel
| | - Galia Polyak
- Technion-Israel Institute of Technology, Faculty of Biology, Emerson building, Haifa, Israel
| | - Noa Shima
- Technion-Israel Institute of Technology, Faculty of Biology, Emerson building, Haifa, Israel
| | - Hadas Biran
- Technion-Israel Institute of Technology, Faculty of Computer Science, Taub building, Haifa, Israel
| | - Tamar Lahav
- Technion-Israel Institute of Technology, Faculty of Biology, Emerson building, Haifa, Israel
| | - Tamar Hashimshony
- Technion-Israel Institute of Technology, Faculty of Biology, Emerson building, Haifa, Israel
| | - Yael Mandel-Gutfreund
- Technion-Israel Institute of Technology, Faculty of Biology, Emerson building, Haifa, Israel.
- Technion-Israel Institute of Technology, Faculty of Computer Science, Taub building, Haifa, Israel.
| |
Collapse
|
23
|
Hancks DC. An Evolutionary Framework Exploiting Virologs and Their Host Origins to Inform Poxvirus Protein Functions. Methods Mol Biol 2025; 2860:257-272. [PMID: 39621273 DOI: 10.1007/978-1-0716-4160-6_17] [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: 12/13/2024]
Abstract
Poxviruses represent evolutionary successful infectious agents. As a family, poxviruses can infect a wide variety of species including humans, fish, and insects. While many other viruses are species-specific, an individual poxvirus species is often capable of infecting diverse hosts and cell types. For example, the prototypical poxvirus, vaccinia, is well known to infect numerous human cell types but can also infect cells from divergent hosts like frog neurons. Notably, poxvirus infections result in both detrimental human and animal diseases. The most infamous disease linked to a poxvirus is smallpox caused by variola virus. Poxviruses are large double-stranded DNA viruses, which uniquely replicate in the cytoplasm of cells. The model poxvirus genome encodes ~200 nonoverlapping protein-coding open reading frames (ORFs). Poxvirus gene products impact various biological processes like the production of virus particles, the host range of infectivity, and disease pathogenesis. In addition, poxviruses and their gene products have biomedical application with several species commonly engineered for use as vaccines and oncolytic virotherapy. Nevertheless, we still have an incomplete understanding of the functions associated with many poxvirus genes. In this chapter, we outline evolutionary insights that can complement ongoing studies of poxvirus gene functions and biology, which may serve to elucidate new molecular activities linked to this biomedically relevant class of viruses.
Collapse
Affiliation(s)
- Dustin C Hancks
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
24
|
Shi Y, McKenery A, Dolan M, Mastri M, Hill JW, Dommer A, Benzekry S, Long M, Abrams SI, Puzanov I, Ebos JML. Acquired resistance to PD-L1 inhibition enhances a type I IFN-regulated secretory program in tumors. EMBO Rep 2025; 26:521-559. [PMID: 39663510 PMCID: PMC11772817 DOI: 10.1038/s44319-024-00333-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 11/10/2024] [Accepted: 11/14/2024] [Indexed: 12/13/2024] Open
Abstract
Therapeutic inhibition of programmed cell death ligand (PD-L1) is linked to alterations in interferon (IFN) signaling. Since IFN-regulated intracellular signaling can control extracellular secretory programs in tumors to modulate immunity, we examined IFN-related secretory changes in tumor cells following resistance to PD-L1 inhibition. Here we report an anti-PD-L1 treatment-induced secretome (PTIS) in tumor models of acquired resistance that is regulated by type I IFNs. These secretory changes can suppress activation of T cells ex vivo while diminishing tumor cell cytotoxicity, revealing that tumor-intrinsic treatment adaptations can exert broad tumor-extrinsic effects. When reimplanted in vivo, resistant tumor growth can slow or stop when PTIS components are disrupted individually, or when type I IFN signaling machinery is blocked. Interestingly, genetic and therapeutic disruption of PD-L1 in vitro can only partially recapitulate the PTIS phenotype highlighting the importance of developing in vivo-based resistance models to more faithfully mimic clinically-relevant treatment failure. Together, this study shows acquired resistance to immune-checkpoint inhibitors 'rewires' tumor secretory programs controlled by type I IFNs that, in turn, can protect from immune cell attack.
Collapse
Affiliation(s)
- Yuhao Shi
- Department of Experimental Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Amber McKenery
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Melissa Dolan
- Department of Experimental Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Michalis Mastri
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - James W Hill
- Jacobs School of Medicine and Biomedical Sciences, SUNY at Buffalo, Buffalo, USA
| | - Adam Dommer
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Sebastien Benzekry
- Computational Pharmacology and Clinical Oncology (COMPO), Inria Sophia Antipolis-Méditerranée, Centre de Recherches en Cancérologie de Marseille, Inserm U1068, CNRS UMR7258, Institut Paoli-Calmettes, Faculté de Pharmacie, Aix-Marseille University, Marseille, France
| | - Mark Long
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Scott I Abrams
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - John M L Ebos
- Department of Experimental Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
| |
Collapse
|
25
|
Paczkowska J, Tang M, Wright KT, Song L, Luu K, Shanmugam V, Welsh EL, Weirather JL, Besson N, Olszewski H, Porter BA, Pfaff KL, Redd RA, Cader FZ, Mandato E, Ouyang J, Calabretta E, Bai G, Lawton LN, Armand P, Rodig SJ, Liu XS, Shipp MA. Cancer-specific innate and adaptive immune rewiring drives resistance to PD-1 blockade in classic Hodgkin lymphoma. Nat Commun 2024; 15:10740. [PMID: 39737927 PMCID: PMC11686379 DOI: 10.1038/s41467-024-54512-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 11/11/2024] [Indexed: 01/01/2025] Open
Abstract
Hodgkin Reed-Sternberg (HRS) cells of classic Hodgkin lymphoma (cHL), like many solid tumors, elicit ineffective immune responses. However, patients with cHL are highly responsive to PD-1 blockade, which largely depends on HRS cell-specific retention of MHC class II and implicates CD4+ T cells and additional MHC class I-independent immune effectors. Here, we utilize single-cell RNA sequencing and spatial analysis to define shared circulating and microenvironmental features of the immune response to PD-1 blockade in cHL. Compared with non-responders, responding patients have more circulating CD4+ naïve and central memory T cells and B cells, as well as more diverse CD4+ T cell and B cell receptor repertoires. Importantly, a population of circulating and tumor-infiltrating IL1β+ monocytes/macrophages is detectable in patients with cHL but not healthy donors, and a proinflammatory, tumor-promoting signature of these circulating IL1β+ monocytes is associated with resistance to PD-1 blockade in cHL. Altogether, our findings reveal extensive immune rewiring and complementary roles of CD4+ T cells, B cells and IL1β+ monocytes in the response to PD-1 blockade and suggest that these features can be captured with a peripheral blood test.
Collapse
Affiliation(s)
- Julia Paczkowska
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ming Tang
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Astra Zeneca, Waltham, MA, USA
| | - Kyle T Wright
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Li Song
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biomedical Data Science, Dartmouth College, Hanover, NH, USA
| | - Kelsey Luu
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- PathAI, Boston, MA, USA
| | - Vignesh Shanmugam
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Emma L Welsh
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jason L Weirather
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Naomi Besson
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Harrison Olszewski
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Billie A Porter
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kathleen L Pfaff
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Robert A Redd
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Fathima Zumla Cader
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- AstraZeneca, City House, Cambridge, UK
| | - Elisa Mandato
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jing Ouyang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Mechanisms of Cancer Resistance Thematic Center, Bristol Myers Squibb, Cambridge, MA, USA
| | - Eleonora Calabretta
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Gali Bai
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Lee N Lawton
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Philippe Armand
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Scott J Rodig
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Xiaole Shirley Liu
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
- GV20 Therapeutics, LLC, Cambridge, MA, USA
| | - Margaret A Shipp
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
26
|
Horton MK, Nititham J, Taylor KE, Katz P, Ye CJ, Yazdany J, Dall'Era M, Hurabielle C, Barcellos LF, Criswell LA, Lanata CM. Changes in DNA methylation are associated with systemic lupus erythematosus flare remission and clinical subtypes. Clin Epigenetics 2024; 16:181. [PMID: 39696438 PMCID: PMC11656870 DOI: 10.1186/s13148-024-01792-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: 05/15/2024] [Accepted: 11/22/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) has numerous symptoms across organs and an unpredictable flare-remittance pattern. This has made it challenging to understand drivers of long-term SLE outcomes. Our objective was to identify whether changes in DNA methylation over time, in an actively flaring SLE cohort, were associated with remission and whether these changes meaningfully subtype SLE patients. METHODS Fifty-nine multi-ethnic SLE patients had clinical visits and DNA methylation profiles at a flare and approximately 3 months later. Methylation was measured using the Illumina EPIC array. We identified sites where methylation change between visits was associated with remission at the follow-up visit using limma package and a time x remission interaction term. Models adjusted for batch, age at diagnosis, time between visits, age at flare, sex, medications, and cell-type proportions. Separately, a paired T-test identified Bonferroni significant methylation sites with ≥ 3% change between visits (n = 546). Methylation changes at these sites were used for unsupervised consensus hierarchical clustering. Associations between clusters and patient features were assessed. RESULTS Nineteen patients fully remitted at the follow-up visit. For 1,953 CpG sites, methylation changed differently for remitters vs. non-remitters (Bonferroni p < 0.05). Nearly half were within genes regulated by interferon. The largest effect was at cg22873177; on average, remitters had 23% decreased methylation between visits while non-remitters had no change. Three SLE patient clusters were identified using methylation differences agnostic of clinical outcomes. All Cluster 1 subjects (n = 12) experienced complete flare remission, despite similar baseline disease activity scores, medications, and demographics as other clusters. Methylation changes at six CpG sites, including within immune-related CD45 and IFI genes, were particularly distinct for each cluster, suggesting these may be good candidates for stratifying patients in the future. CONCLUSIONS Changes in DNA methylation during active SLE were associated with remission status and identified subgroups of SLE patients with several distinct clinical and biological characteristics. DNA methylation patterns might help inform SLE subtypes, leading to targeted therapies based on relevant underlying biological pathways.
Collapse
Affiliation(s)
- Mary K Horton
- Genomics of Autoimmune Rheumatic Disease Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Joanne Nititham
- Genomics of Autoimmune Rheumatic Disease Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kimberly E Taylor
- Division of Rheumatology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Patricia Katz
- Division of Rheumatology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Chun Jimmie Ye
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA, USA
| | - Jinoos Yazdany
- Division of Rheumatology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Maria Dall'Era
- Division of Rheumatology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Charlotte Hurabielle
- Division of Rheumatology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Lisa F Barcellos
- Division of Epidemiology, University of California, Berkeley, CA, USA
| | - Lindsey A Criswell
- Genomics of Autoimmune Rheumatic Disease Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Cristina M Lanata
- Genomics of Autoimmune Rheumatic Disease Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
27
|
Vukić D, Cherian A, Keskitalo S, Bong YT, Marônek M, Yadav L, Keegan LP, Varjosalo M, O'Connell MA. Distinct interactomes of ADAR1 nuclear and cytoplasmic protein isoforms and their responses to interferon induction. Nucleic Acids Res 2024; 52:14184-14204. [PMID: 39673305 DOI: 10.1093/nar/gkae1106] [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: 03/04/2024] [Revised: 09/29/2024] [Accepted: 11/04/2024] [Indexed: 12/16/2024] Open
Abstract
The RNA editing enzyme adenosine deaminase acting on RNA 1 (ADAR1) is essential for correct functioning of innate immune responses. The ADAR1p110 isoform is mainly nuclear and ADAR1p150, which is interferon (IFN) inducible, is predominately cytoplasmic. Using three different methods - co-immunoprecipitation (co-IP) of endogenous ADAR1, Strep-tag co-IP and BioID with individual ADAR1 isoforms - a comprehensive interactome was generated during both homeostasis and the IFN response. Both known and novel interactors as well as editing regulators were identified. Nuclear proteins were detected as stable interactors with both ADAR1 isoforms. In contrast, BioID identified distinct protein networks for each ADAR1 isoform, with nuclear components observed with ADAR1p110 and components of cytoplasmic cellular condensates with ADAR1p150. RNase A digestion distinguished between distal and proximal interactors, as did a double-stranded RNA (dsRNA)-binding mutant of ADAR1 which demonstrated the importance of dsRNA binding for ADAR1 interactions. IFN treatment did not affect the core ADAR1 interactomes but resulted in novel interactions, the majority of which are proximal interactions retained after RNase A treatment. Short treatment with high molecular weight poly(I:C) during the IFN response resulted in dsRNA-binding-dependent changes in the proximal protein network of ADAR1p110 and association of the ADAR1p150 proximal protein network with some components of antiviral stress granules.
Collapse
Affiliation(s)
- Dragana Vukić
- Central European Institute of Technology (CEITEC), Masaryk University, Kamenice 5, Brno 62500, Czechia
- NationalCentre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, Brno 62500, Czechia
| | - Anna Cherian
- Central European Institute of Technology (CEITEC), Masaryk University, Kamenice 5, Brno 62500, Czechia
- NationalCentre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, Brno 62500, Czechia
| | - Salla Keskitalo
- Institute of Biotechnology, HelsinkiInstitute of Life Science (HiLIFE), University of Helsinki, Helsinki 00014, Finland
| | - Yih Tyng Bong
- Institute of Biotechnology, HelsinkiInstitute of Life Science (HiLIFE), University of Helsinki, Helsinki 00014, Finland
| | - Martin Marônek
- Central European Institute of Technology (CEITEC), Masaryk University, Kamenice 5, Brno 62500, Czechia
| | - Leena Yadav
- Institute of Biotechnology, HelsinkiInstitute of Life Science (HiLIFE), University of Helsinki, Helsinki 00014, Finland
| | - Liam P Keegan
- Central European Institute of Technology (CEITEC), Masaryk University, Kamenice 5, Brno 62500, Czechia
| | - Markku Varjosalo
- Institute of Biotechnology, HelsinkiInstitute of Life Science (HiLIFE), University of Helsinki, Helsinki 00014, Finland
| | - Mary A O'Connell
- Central European Institute of Technology (CEITEC), Masaryk University, Kamenice 5, Brno 62500, Czechia
| |
Collapse
|
28
|
Guanizo AC, Luong Q, Jayasekara WSN, de Geus ED, Inampudi C, Xue VS, Chen J, de Weerd NA, Matthews AY, Gantier MP, Balic JJ, Arulananda S, Garama DJ, Hertzog PJ, Ganju V, Watkins DN, Cain JE, Gough DJ. A STAT3-STING-IFN axis controls the metastatic spread of small cell lung cancer. Nat Immunol 2024; 25:2259-2269. [PMID: 39572642 DOI: 10.1038/s41590-024-02014-5] [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: 07/14/2022] [Accepted: 10/10/2024] [Indexed: 11/27/2024]
Abstract
Small cell lung cancer (SCLC) is an aggressive neuroendocrine tumor characterized by a high metastatic potential with an overall survival rate of ~5%. The transcription factor signal transducer and activator of transcription 3 (STAT3) is overexpressed by >50% of tumors, including SCLC, but its role in SCLC development and metastasis is unclear. Here, we show that, while STAT3 deletion restricts primary tumor growth, it paradoxically enhances metastatic spread by promoting immune evasion. This occurs because STAT3 is crucial for maintaining the immune sensor stimulator of interferon (IFN) genes (STING). Without STAT3, the cyclic adenosine monophosphate-guanosine monophosphate synthase-STING pathway is inactive, resulting in decreased type I IFN secretion and an IFN gene signature. Importantly, restoration of IFN signaling through re-expression of endogenous STING, enforced expression of IFN response factor 7 or administration of recombinant type I IFN re-established antitumor immunity, inhibiting metastatic SCLC in vivo. These data show the potential of augmenting the innate immune response to block metastatic SCLC.
Collapse
Affiliation(s)
- Aleks C Guanizo
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - Quinton Luong
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - W Samantha N Jayasekara
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - Eveline D de Geus
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Chaitanya Inampudi
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - Vincent Senyang Xue
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - Jasmine Chen
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - Nicole A de Weerd
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Antony Y Matthews
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Michael P Gantier
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Jesse J Balic
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Surein Arulananda
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Medical Oncology, Monash Health, Clayton, Victoria, Australia
- School of Clinical Sciences, Faculty of Medicine, Monash University, Clayton, Victoria, Australia
| | - Daniel J Garama
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - Paul J Hertzog
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Vinod Ganju
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - D Neil Watkins
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
- Research Institute in Oncology and Hematology, Cancer Care Manitoba, Winnipeg, Manitoba, Canada
- Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jason E Cain
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - Daniel J Gough
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia.
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
29
|
Zerad L, Gacem N, Gayda F, Day L, Sinigaglia K, Richard L, Parisot M, Cagnard N, Mathis S, Bole-Feysot C, O’Connell MA, Pingault V, Dambroise E, Keegan LP, Vallat JM, Bondurand N. Overexpression of Egr1 Transcription Regulator Contributes to Schwann Cell Differentiation Defects in Neural Crest-Specific Adar1 Knockout Mice. Cells 2024; 13:1952. [PMID: 39682701 PMCID: PMC11639873 DOI: 10.3390/cells13231952] [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: 10/11/2024] [Revised: 11/05/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024] Open
Abstract
Adenosine deaminase acting on RNA 1 (ADAR1) is the principal enzyme for the adenosine-to-inosine RNA editing that prevents the aberrant activation of cytosolic nucleic acid sensors by endogenous double stranded RNAs and the activation of interferon-stimulated genes. In mice, the conditional neural crest deletion of Adar1 reduces the survival of melanocytes and alters the differentiation of Schwann cells that fail to myelinate nerve fibers in the peripheral nervous system. These myelination defects are partially rescued upon the concomitant removal of the Mda5 antiviral dsRNA sensor in vitro, suggesting implication of the Mda5/Mavs pathway and downstream effectors in the genesis of Adar1 mutant phenotypes. By analyzing RNA-Seq data from the sciatic nerves of mouse pups after conditional neural crest deletion of Adar1 (Adar1cKO), we here identified the transcription factors deregulated in Adar1cKO mutants compared to the controls. Through Adar1;Mavs and Adar1cKO;Egr1 double-mutant mouse rescue analyses, we then highlighted that the aberrant activation of the Mavs adapter protein and overexpression of the early growth response 1 (EGR1) transcription factor contribute to the Adar1 deletion associated defects in Schwann cell development in vivo. In silico and in vitro gene regulation studies additionally suggested that EGR1 might mediate this inhibitory effect through the aberrant regulation of EGR2-regulated myelin genes. We thus demonstrate the role of the Mda5/Mavs pathway, but also that of the Schwann cell transcription factors in Adar1-associated peripheral myelination defects.
Collapse
Affiliation(s)
- Lisa Zerad
- Laboratory of Embryology and Genetics of Human Malformations, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 24 Boulevard du Montparnasse, 75015 Paris, France; (L.Z.); (N.G.); (F.G.); (L.D.); (V.P.)
| | - Nadjet Gacem
- Laboratory of Embryology and Genetics of Human Malformations, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 24 Boulevard du Montparnasse, 75015 Paris, France; (L.Z.); (N.G.); (F.G.); (L.D.); (V.P.)
| | - Fanny Gayda
- Laboratory of Embryology and Genetics of Human Malformations, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 24 Boulevard du Montparnasse, 75015 Paris, France; (L.Z.); (N.G.); (F.G.); (L.D.); (V.P.)
| | - Lucie Day
- Laboratory of Embryology and Genetics of Human Malformations, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 24 Boulevard du Montparnasse, 75015 Paris, France; (L.Z.); (N.G.); (F.G.); (L.D.); (V.P.)
| | - Ketty Sinigaglia
- Central European Institute for Technology, Masaryk University (CEITEC MU), Kamenice 735/5, 625 00 Brno, Czech Republic; (K.S.); (M.A.O.); (L.P.K.)
| | - Laurence Richard
- Department of Neurology, Centre de Reference “Neuropathies Périphériques Rares”, CHU Limoges, 87000 Limoges, France; (L.R.); (J.M.V.)
| | - Melanie Parisot
- Genomics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163 et INSERM US24/CNRS UAR3633, Paris Descartes Sorbonne Paris Cite University, 75015 Paris, France; (M.P.); (C.B.-F.)
| | - Nicolas Cagnard
- Bioinformatics Platform, Imagine Institute, INSERM UMR 1163, 75015 Paris, France;
| | - Stephane Mathis
- Department of Neurology (Nerve-Muscle Unit) and ‘Grand Sud-Ouest’ National Reference Center for Neuromuscular Disorders, CHU Bordeaux, Pellegrin Hospital, 33000 Bordeaux, France;
| | - Christine Bole-Feysot
- Genomics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163 et INSERM US24/CNRS UAR3633, Paris Descartes Sorbonne Paris Cite University, 75015 Paris, France; (M.P.); (C.B.-F.)
| | - Mary A. O’Connell
- Central European Institute for Technology, Masaryk University (CEITEC MU), Kamenice 735/5, 625 00 Brno, Czech Republic; (K.S.); (M.A.O.); (L.P.K.)
| | - Veronique Pingault
- Laboratory of Embryology and Genetics of Human Malformations, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 24 Boulevard du Montparnasse, 75015 Paris, France; (L.Z.); (N.G.); (F.G.); (L.D.); (V.P.)
| | - Emilie Dambroise
- Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 24 Boulevard du Montparnasse, 75015 Paris, France;
| | - Liam P. Keegan
- Central European Institute for Technology, Masaryk University (CEITEC MU), Kamenice 735/5, 625 00 Brno, Czech Republic; (K.S.); (M.A.O.); (L.P.K.)
| | - Jean Michel Vallat
- Department of Neurology, Centre de Reference “Neuropathies Périphériques Rares”, CHU Limoges, 87000 Limoges, France; (L.R.); (J.M.V.)
| | - Nadege Bondurand
- Laboratory of Embryology and Genetics of Human Malformations, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 24 Boulevard du Montparnasse, 75015 Paris, France; (L.Z.); (N.G.); (F.G.); (L.D.); (V.P.)
| |
Collapse
|
30
|
Griffiths CD, Shah M, Shao W, Borgman CA, Janes KA. Three modes of viral adaption by the heart. SCIENCE ADVANCES 2024; 10:eadp6303. [PMID: 39536108 PMCID: PMC11559625 DOI: 10.1126/sciadv.adp6303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024]
Abstract
Viruses elicit long-term adaptive responses in the tissues they infect. Understanding viral adaptions in humans is difficult in organs such as the heart, where primary infected material is not routinely collected. In search of asymptomatic infections with accompanying host adaptions, we mined for cardio-pathogenic viruses in the unaligned reads of nearly 1000 human hearts profiled by RNA sequencing. Among virus-positive cases (~20%), we identified three robust adaptions in the host transcriptome related to inflammatory nuclear factor κB (NF-κB) signaling and posttranscriptional regulation by the p38-MK2 pathway. The adaptions are not determined by the infecting virus, and they recur in infections of human or animal hearts and cultured cardiomyocytes. Adaptions switch states when NF-κB or p38-MK2 is perturbed in cells engineered for chronic infection by the cardio-pathogenic virus, coxsackievirus B3. Stratifying viral responses into reversible adaptions adds a targetable systems-level simplification for infections of the heart and perhaps other organs.
Collapse
Affiliation(s)
- Cameron D. Griffiths
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Millie Shah
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - William Shao
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Cheryl A. Borgman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Kevin A. Janes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
31
|
Sparling AC, Ward JM, Sarkar K, Schiffenbauer A, Farhadi PN, Smith MA, Rahman S, Zerrouki K, Miller FW, Li JL, Casey KA, Rider LG. Neutrophil and mononuclear leukocyte pathways and upstream regulators revealed by serum proteomics of adult and juvenile dermatomyositis. Arthritis Res Ther 2024; 26:196. [PMID: 39529136 PMCID: PMC11552237 DOI: 10.1186/s13075-024-03421-7] [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: 05/09/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
OBJECTIVES Serum protein abundance was assessed in adult and juvenile dermatomyositis (DM and JDM) patients to determine differentially regulated proteins, altered pathways, and candidate disease activity biomarkers. METHODS Serum protein expression from 17 active adult DM and JDM patients each was compared to matched, healthy control subjects by a multiplex immunoassay. Pathway analysis and protein clustering of the differentially regulated proteins were examined to assess underlying mechanisms. Candidate disease activity biomarkers were identified by correlating protein expression with disease activity measures. RESULTS Seventy-eight of 172 proteins were differentially expressed in the sera of DM and JDM patients compared to healthy controls. Forty-eight proteins were differentially expressed in DM, 32 proteins in JDM, and 14 proteins in both DM and JDM. Twelve additional differentially expressed proteins were identified after combining the DM and JDM cohorts. C-X-C motif chemokine ligand 10 (CXCL10) was the most strongly upregulated protein in both DM and JDM sera. Other highly upregulated proteins in DM included S100 calcium binding protein A12 (S100A12), CXCL9, and nicotinamide phosphoribosyltransferase (NAMPT), while highly upregulated proteins in JDM included matrix metallopeptidase 3 (MMP3), growth differentiation factor 15 (GDF15), and von Willebrand factor (vWF). Pathway analysis indicated that phosphoinositide 3-kinase (PI3K), p38 mitogen-activated protein kinase (MAPK), and toll-like receptor 7 (TLR7) signaling were activated in DM and JDM. Additional pathways specific to DM or JDM were identified. A protein cluster associated with neutrophils and mononuclear leukocytes and a cluster of interferon-associated proteins were observed in both DM and JDM. Twenty-two proteins in DM and 24 proteins in JDM sera correlated with global, muscle, and/or skin disease activity. Seven proteins correlated with disease activity measures in both DM and JDM sera. IL-1 receptor like 1 (IL1RL1) emerged as a candidate global disease activity biomarker in DM and JDM. CONCLUSION Coordinate analysis of protein expression in DM and JDM patient sera by a multiplex immunoassay validated previous gene expression studies and identified novel dysregulated proteins, altered signaling pathways, and candidate disease activity biomarkers. These findings may further inform the assessment of DM and JDM patients and aid in the identification of potential therapeutic targets.
Collapse
Affiliation(s)
- A Clare Sparling
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Building 10, CRC Rm 6-5700, MSC 1301 10 Center Drive, Bethesda, MD, 20892-1301, USA
| | - James M Ward
- Integrative Bioinformatics Support Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, USA
| | - Kakali Sarkar
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Building 10, CRC Rm 6-5700, MSC 1301 10 Center Drive, Bethesda, MD, 20892-1301, USA
| | - Adam Schiffenbauer
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Building 10, CRC Rm 6-5700, MSC 1301 10 Center Drive, Bethesda, MD, 20892-1301, USA
| | - Payam Noroozi Farhadi
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Building 10, CRC Rm 6-5700, MSC 1301 10 Center Drive, Bethesda, MD, 20892-1301, USA
| | | | - Saifur Rahman
- BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | | | - Frederick W Miller
- Integrative Bioinformatics Support Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, USA
| | - Jian-Liang Li
- Integrative Bioinformatics Support Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, USA
| | - Kerry A Casey
- BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Lisa G Rider
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Building 10, CRC Rm 6-5700, MSC 1301 10 Center Drive, Bethesda, MD, 20892-1301, USA.
| |
Collapse
|
32
|
Phan J, Chen B, Zhao Z, Allies G, Iannaccone A, Paul A, Cansiz F, Spina A, Leven AS, Gellhaus A, Schadendorf D, Kimmig R, Mettlen M, Tasdogan A, Morrison SJ. Retrotransposons are co-opted to activate hematopoietic stem cells and erythropoiesis. Science 2024; 386:eado6836. [PMID: 39446896 PMCID: PMC11709122 DOI: 10.1126/science.ado6836] [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: 02/15/2024] [Revised: 06/21/2024] [Accepted: 08/30/2024] [Indexed: 10/26/2024]
Abstract
Hematopoietic stem cells (HSCs) and erythropoiesis are activated during pregnancy and after bleeding by the derepression of retrotransposons, including endogenous retroviruses and long interspersed nuclear elements. Retrotransposon transcription activates the innate immune sensors cyclic guanosine 3',5'-monophosphate-adenosine 5'-monophosphate synthase (cGAS) and stimulator of interferon (IFN) genes (STING), which induce IFN and IFN-regulated genes in HSCs, increasing HSC division and erythropoiesis. Inhibition of reverse transcriptase or deficiency for cGAS or STING had little or no effect on hematopoiesis in nonpregnant mice but depleted HSCs and erythroid progenitors in pregnant mice, reducing red blood cell counts. Retrotransposons and IFN-regulated genes were also induced in mouse HSCs after serial bleeding and, in human HSCs, during pregnancy. Reverse transcriptase inhibitor use was associated with anemia in pregnant but not in nonpregnant people, suggesting conservation of these mechanisms from mice to humans.
Collapse
Affiliation(s)
- Julia Phan
- Children’s Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center; Dallas, TX 75390, USA
| | - Brandon Chen
- Children’s Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center; Dallas, TX 75390, USA
| | - Zhiyu Zhao
- Children’s Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center; Dallas, TX 75390, USA
| | - Gabriele Allies
- Department of Dermatology, University Hospital Essen & German Cancer Consortium; Essen, & National Center for Tumor Diseases (NCT-West), Campus Essen & Research Alliance Ruhr, Research Center One Health, University Duisburg-Essen, Campus Essen, Essen, Germany
| | - Antonella Iannaccone
- Department of Gynecology and Obstetrics, University Hospital Essen, 45147 Essen, Germany
| | - Animesh Paul
- Children’s Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center; Dallas, TX 75390, USA
| | - Feyza Cansiz
- Department of Dermatology, University Hospital Essen & German Cancer Consortium; Essen, & National Center for Tumor Diseases (NCT-West), Campus Essen & Research Alliance Ruhr, Research Center One Health, University Duisburg-Essen, Campus Essen, Essen, Germany
| | - Alberto Spina
- Department of Gynecology and Obstetrics, University Hospital Essen, 45147 Essen, Germany
| | - Anna-Sophia Leven
- Department of Dermatology, University Hospital Essen & German Cancer Consortium; Essen, & National Center for Tumor Diseases (NCT-West), Campus Essen & Research Alliance Ruhr, Research Center One Health, University Duisburg-Essen, Campus Essen, Essen, Germany
| | - Alexandra Gellhaus
- Department of Gynecology and Obstetrics, University Hospital Essen, 45147 Essen, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen & German Cancer Consortium; Essen, & National Center for Tumor Diseases (NCT-West), Campus Essen & Research Alliance Ruhr, Research Center One Health, University Duisburg-Essen, Campus Essen, Essen, Germany
| | - Rainer Kimmig
- Department of Gynecology and Obstetrics, University Hospital Essen, 45147 Essen, Germany
| | - Marcel Mettlen
- Department of Cell Biology, University of Texas Southwestern Medical Center; Dallas, Texas 75235-9039
| | - Alpaslan Tasdogan
- Department of Dermatology, University Hospital Essen & German Cancer Consortium; Essen, & National Center for Tumor Diseases (NCT-West), Campus Essen & Research Alliance Ruhr, Research Center One Health, University Duisburg-Essen, Campus Essen, Essen, Germany
| | - Sean J. Morrison
- Children’s Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center; Dallas, TX 75390, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center; Dallas, TX 75390, USA
| |
Collapse
|
33
|
Vízkeleti L, Papp O, Doma V, Gil J, Markó-Varga G, Kovács SA, Győrffy B, Kárpáti S, Tímár J. Identification of genetic fingerprint of type I interferon therapy in visceral metastases of melanoma. Sci Rep 2024; 14:26540. [PMID: 39489756 PMCID: PMC11532416 DOI: 10.1038/s41598-024-77285-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: 04/17/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024] Open
Abstract
Malignant melanoma is a difficult-to-treat skin cancer with increasing incidence worldwide. Although type-I interferon (IFN) is no longer part of guidelines, several melanoma patients are treated with type-I interferon (IFN) at some point of the disease, potentially affecting its genetic progression. We run genome-wide copy number variation (CNV) analysis on previously type-I IFN-treated (n = 17) and control (n = 11) visceral metastases of melanoma patients. Results were completed with data from the TCGA and MM500 databases. We identified metastasis- and brain metastasis-specific gene signatures mostly affected by CN gains. Some cases were genetically resistant to IFN showing characteristic gene alterations (e.g. ABCA4 or ZEB2 gain and alterations of DNA repair genes). Analysis of a previously identified type-I IFN resistance gene set indicates that only a proportion of these genes was exclusive for the IFN-treated metastases reflecting a possible selective genomic pressure of endogenous IFNs during progression. Our data suggest that previous type-I IFN treatment and/or endogenous IFN production by immune response affect genomic progression of melanoma which may have clinical relevance, potentially influence immune checkpoint regulation in the tumor microenvironment.
Collapse
Affiliation(s)
- Laura Vízkeleti
- Department of Bioinformatics, Faculty of Medicine, Semmelweis University, 1094, Budapest, Hungary
- Department of Pathology, Forensic and Insurance Medicine, Faculty of Medicine, Semmelweis University, Üllői Str. 93., 1091, Budapest, Hungary
| | - Orsolya Papp
- Department of Pathology, Forensic and Insurance Medicine, Faculty of Medicine, Semmelweis University, Üllői Str. 93., 1091, Budapest, Hungary
- Turbine Simulated Cell Technologies, Budapest, 1027, Hungary
| | - Viktória Doma
- Department of Pathology, Forensic and Insurance Medicine, Faculty of Medicine, Semmelweis University, Üllői Str. 93., 1091, Budapest, Hungary
- Department of Dermatology, Venerology and Dermato-Oncology, Faculty of Medicine, Semmelweis University, 1085, Budapest, Hungary
| | - Jeovanis Gil
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, 223 63, Lund, Sweden
| | - György Markó-Varga
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, 223 63, Lund, Sweden
- Chemical Genomics Global Research Lab, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, South Korea
- 1St Department of Surgery, Tokyo Medical University, Tokyo, 160-8582, Japan
| | - Szonja A Kovács
- Department of Bioinformatics, Faculty of Medicine, Semmelweis University, 1094, Budapest, Hungary
- Doctoral School of Pathological Sciences, Semmelweis University, 1085, Budapest, Hungary
- National Laboratory for Drug Research and Development, 1117, Budapest, Hungary
| | - Balázs Győrffy
- Department of Bioinformatics, Faculty of Medicine, Semmelweis University, 1094, Budapest, Hungary
| | - Sarolta Kárpáti
- Department of Dermatology, Venerology and Dermato-Oncology, Faculty of Medicine, Semmelweis University, 1085, Budapest, Hungary
| | - József Tímár
- Department of Pathology, Forensic and Insurance Medicine, Faculty of Medicine, Semmelweis University, Üllői Str. 93., 1091, Budapest, Hungary.
| |
Collapse
|
34
|
He X, Zhang S, Zou Z, Gao P, Yang L, Xiang B. Antiviral Effects of Avian Interferon-Stimulated Genes. Animals (Basel) 2024; 14:3062. [PMID: 39518785 PMCID: PMC11545081 DOI: 10.3390/ani14213062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/20/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Interferons (IFNs) stimulate the expression of numerous IFN-stimulating genes via the Janus kinase-signal transducers and activators of the transcription (JAK-STAT) signaling pathway, which plays an important role in the host defense against viral infections. In mammals, including humans and mice, a substantial number of IFN-stimulated genes (ISGs) have been identified, and their molecular mechanisms have been elucidated. It is important to note that avian species are phylogenetically distant from mammals, resulting in distinct IFN-induced ISGs that may have different functions. At present, only a limited number of avian ISGs have been identified. In this review, we summarized the identified avian ISGs and their antiviral activities. As gene-editing technology is widely used in avian breeding, the identification of avian ISGs and the elucidation of their molecular mechanism may provide important support for the breeding of avians for disease resistance.
Collapse
Affiliation(s)
- Xingchen He
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (X.H.); (S.Z.); (Z.Z.); (L.Y.)
- Center for Poultry Disease Control and Prevention, Yunnan Agricultural University, Kunming 650201, China
| | - Shiyuan Zhang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (X.H.); (S.Z.); (Z.Z.); (L.Y.)
- Center for Poultry Disease Control and Prevention, Yunnan Agricultural University, Kunming 650201, China
| | - Ziheng Zou
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (X.H.); (S.Z.); (Z.Z.); (L.Y.)
| | - Pei Gao
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang 453000, China;
| | - Liangyu Yang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (X.H.); (S.Z.); (Z.Z.); (L.Y.)
- Center for Poultry Disease Control and Prevention, Yunnan Agricultural University, Kunming 650201, China
| | - Bin Xiang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (X.H.); (S.Z.); (Z.Z.); (L.Y.)
- Center for Poultry Disease Control and Prevention, Yunnan Agricultural University, Kunming 650201, China
| |
Collapse
|
35
|
de Los Rios Kobara I, Jayewickreme R, Lee MJ, Wilk AJ, Blomkalns AL, Nadeau KC, Yang S, Rogers AJ, Blish CA. Interferon-mediated NK cell activation is associated with limited neutralization breadth during SARS-CoV-2 infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619639. [PMID: 39484382 PMCID: PMC11527016 DOI: 10.1101/2024.10.22.619639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Best known for their ability to kill infected or malignant cells, natural killer (NK) cells are also underappreciated regulators of the antibody response to viral infection. In mice, NK cells can kill T follicular helper (Tfh) cells, decreasing somatic hypermutation and vaccine responses. Although human NK cell activation correlates with poor vaccine response, the mechanisms of human NK cell regulation of adaptive immunity are poorly understood. We found that in human ancestral SARS-CoV-2 infection, broad neutralizers, who were capable of neutralizing Alpha, Beta, and Delta, had fewer NK cells that expressed inhibitory and immaturity markers whereas NK cells from narrow neutralizers were highly activated and expressed interferon-stimulated genes (ISGs). ISG-mediated activation in NK cells from healthy donors increased cytotoxicity and functional responses to induced Tfh-like cells. This work reveals that NK cell activation and dysregulated inflammation may play a role in poor antibody response to SARS-CoV-2 and opens exciting avenues for designing improved vaccines and adjuvants to target emerging pathogens.
Collapse
|
36
|
Collins C, Chaumont L, Peruzzi M, Jamak N, Boudinot P, Béjar J, Moreno P, Álvarez Torres D, Collet B. Effect of a loss of the mda5/ifih1 gene on the antiviral resistance in a Chinook salmon Oncorhynchus tshawytscha cell line. PLoS One 2024; 19:e0311283. [PMID: 39401233 PMCID: PMC11472919 DOI: 10.1371/journal.pone.0311283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 09/17/2024] [Indexed: 10/17/2024] Open
Abstract
Cells are equipped with intracellular RIG-like Receptors (RLRs) detecting double stranded (ds)RNA, a molecule with Pathogen-Associated Molecular Pattern (PAMPs) generated during the life cycle of many viruses. Melanoma Differentiation-Associated protein 5 (MDA5), a helicase enzyme member of the RLRs encoded by the ifih1 gene, binds to long dsRNA molecules during a viral infection and initiates production of type I interferon (IFN1) which orchestrates the antiviral response. In order to understand the contribution of MDA5 to viral resistance in fish cells, we have isolated a clonal Chinook salmon Oncorhynchus tshawytscha epithelial-like cell line invalidated for the ifih1 gene by CRISPR/Cas9 genome editing. We demonstrated that IFN1 induction is impaired in this cell line after infection with the Snakehead Rhabdovirus (SHRV), the Salmon Alphavirus (SAV) or Nervous Necrosis Virus (NNV). The cell line, however, did not show any increase in cytopathic effect when infected with SHRV or SAV. Similarly, no cytopathic effect was observed in the ifih1-/- cell line when infected with Infectious Pancreatic Necrosis Virus (IPNV), Infectious Haemorrhagic Necrotic Virus (IHNV). These results indicate the redundancy of the antiviral innate defence system in CHSE-derived cells, which helps with circumventing viral evasion strategies.
Collapse
Affiliation(s)
- Catherine Collins
- INRAE, UVSQ, VIM, Université Paris-Saclay, Jouy-en-Josas, France
- School of Biological, Earth and Environmental Sciences, University College Cork, Cork, Ireland
| | - Lise Chaumont
- INRAE, UVSQ, VIM, Université Paris-Saclay, Jouy-en-Josas, France
| | - Mathilde Peruzzi
- INRAE, UVSQ, VIM, Université Paris-Saclay, Jouy-en-Josas, France
| | - Nedim Jamak
- INRAE, UVSQ, VIM, Université Paris-Saclay, Jouy-en-Josas, France
| | - Pierre Boudinot
- INRAE, UVSQ, VIM, Université Paris-Saclay, Jouy-en-Josas, France
| | | | | | | | - Bertrand Collet
- INRAE, UVSQ, VIM, Université Paris-Saclay, Jouy-en-Josas, France
| |
Collapse
|
37
|
Fee BE, Fee LR, Menechella M, Affeldt B, Sprouse AR, Bounini A, Alwarawrah Y, Molloy CT, Ilkayeva OR, Prinz JA, Lenz DS, MacIver NJ, Rai P, Fessler MB, Coers J, Taylor GA. Type I interferon signaling and peroxisomal dysfunction contribute to enhanced inflammatory cytokine production in IRGM1-deficient macrophages. J Biol Chem 2024; 300:107883. [PMID: 39395806 DOI: 10.1016/j.jbc.2024.107883] [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: 04/01/2024] [Revised: 09/30/2024] [Accepted: 10/05/2024] [Indexed: 10/14/2024] Open
Abstract
The human IRGM gene has been linked to inflammatory diseases including sepsis and Crohn's disease. Decreased expression of human IRGM, or the mouse orthologues Irgm1 and Irgm2, leads to increased production of a number of inflammatory chemokines and cytokines in vivo and/or in cultured macrophages. Prior work has indicated that increased cytokine production is instigated by metabolic alterations and changes in mitochondrial homeostasis; however, a comprehensive mechanism has not been elucidated. In the studies presented here, RNA deep sequencing and quantitative PCR were used to show that increases in cytokine production, as well as most changes in the transcriptional profile of Irgm1-/- bone marrow-derived macrophages (BMM), are dependent on increased type I IFN production seen in those cells. Metabolic alterations that drive increased cytokines in Irgm1-/- BMM - specifically increases in glycolysis and increased accumulation of acyl-carnitines - were unaffected by quenching type I IFN signaling. Dysregulation of peroxisomal homeostasis was identified as a novel upstream pathway that governs type I IFN production and inflammatory cytokine production. Collectively, these results enhance our understanding of the complex biochemical changes that are triggered by lack of Irgm1 and contribute to inflammatory disease seen with Irgm1-deficiency.
Collapse
Affiliation(s)
- Brian E Fee
- Department of Medicine, Division of Geriatrics, and Center for the Study of Aging and Human Development; Duke University Medical Center, Durham, North Carolina, USA
| | - Lanette R Fee
- Department of Medicine, Division of Geriatrics, and Center for the Study of Aging and Human Development; Duke University Medical Center, Durham, North Carolina, USA
| | - Mark Menechella
- Department of Medicine, Division of Geriatrics, and Center for the Study of Aging and Human Development; Duke University Medical Center, Durham, North Carolina, USA
| | - Bethann Affeldt
- Department of Medicine, Division of Geriatrics, and Center for the Study of Aging and Human Development; Duke University Medical Center, Durham, North Carolina, USA
| | - Aemilia R Sprouse
- Department of Medicine, Division of Geriatrics, and Center for the Study of Aging and Human Development; Duke University Medical Center, Durham, North Carolina, USA
| | - Amina Bounini
- Department of Medicine, Division of Geriatrics, and Center for the Study of Aging and Human Development; Duke University Medical Center, Durham, North Carolina, USA
| | - Yazan Alwarawrah
- Department of Pediatrics, Division of Pediatric Endocrinology and Diabetes, and Department of Nutrition, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Caitlyn T Molloy
- Department of Pediatrics, Division of Pediatric Endocrinology and Diabetes, and Department of Nutrition, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Olga R Ilkayeva
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina, USA; Division of Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Joseph A Prinz
- Duke University School of Medicine, Sequencing and Genomic Technologies, Durham, North Carolina, USA
| | - Devi Swain Lenz
- Duke University School of Medicine, Sequencing and Genomic Technologies, Durham, North Carolina, USA; Departments of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Nancie J MacIver
- Department of Pediatrics, Division of Pediatric Endocrinology and Diabetes, and Department of Nutrition, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Prashant Rai
- Immunity, Inflammation and Disease Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Michael B Fessler
- Immunity, Inflammation and Disease Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Jörn Coers
- Departments of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA; Department of Immunobiology; Duke University Medical Center, Durham, North Carolina, USA
| | - Gregory A Taylor
- Department of Medicine, Division of Geriatrics, and Center for the Study of Aging and Human Development; Duke University Medical Center, Durham, North Carolina, USA; Departments of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA; Department of Immunobiology; Duke University Medical Center, Durham, North Carolina, USA; Geriatric Research, Education, and Clinical Center, Durham VA Health Care System, Durham, North Carolina, USA.
| |
Collapse
|
38
|
Sari G, Dhatchinamoorthy K, Orellano-Ariza L, Ferreira LM, Brehm MA, Rock K. IRF2 loss is associated with reduced MHC I pathway transcripts in subsets of most human cancers and causes resistance to checkpoint immunotherapy in human and mouse melanomas. J Exp Clin Cancer Res 2024; 43:276. [PMID: 39354629 PMCID: PMC11446056 DOI: 10.1186/s13046-024-03187-5] [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: 06/25/2024] [Accepted: 09/07/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND In order for cancers to progress, they must evade elimination by CD8 T cells or other immune mechanisms. CD8 T cells recognize and kill tumor cells that display immunogenic tumor peptides bound to MHC I molecules. One of the ways that cancers can escape such killing is by reducing expression of MHC I molecules, and loss of MHC I is frequently observed in tumors. There are multiple different mechanisms that can underly the loss of MHC I complexes on tumor and it is currently unclear whether there are particular mechanisms that occur frequently and, if so, in what types of cancers. Also of importance to know is whether the loss of MHC I is reversible and how such loss and/or its restoration would impact responses to immunotherapy. Here, we investigate these issues for loss of IRF1 and IRF2, which are transcription factors that drive expression of MHC I pathway genes and some killing mechanisms. METHODS Bioinformatics analyses of IRF2 and IRF2-dependent gene transcripts were performed for all human cancers in the TCGA RNAseq database. IRF2 protein-DNA-binding was analyzed in ChIPseq databases. CRISRPcas9 was used to knock out IRF1 and IRF2 genes in human and mouse melanoma cells and the resulting phenotypes were analyzed in vitro and in vivo. RESULTS Transcriptomic analysis revealed that IRF2 expression was reduced in a substantial subset of cases in almost all types of human cancers. When this occurred there was a corresponding reduction in the expression of IRF2-regulated genes that were needed for CD8 T cell recognition. To test cause and effect for these IRF2 correlations and the consequences of IRF2 loss, we gene-edited IRF2 in a patient-derived melanoma and a mouse melanoma. The IRF2 gene-edited melanomas had reduced expression of transcripts for genes in the MHC I pathway and decreased levels of MHC I complexes on the cell surface. Levels of Caspase 7, an IRF2 target gene involved in CD8 T cell killing of tumors, were also reduced. This loss of IRF2 caused both human and mouse melanomas to become resistant to immunotherapy with a checkpoint inhibitor. Importantly, these effects were reversible. Stimulation of the IRF2-deficient melanomas with interferon induced the expression of a functionally homologous transcription factor, IRF1, which then restored the MHC I pathway and responsiveness to CPI. CONCLUSIONS Our study shows that a subset of cases within most types of cancers downregulates IRF2 and that this can allow cancers to escape immune control. This can cause resistance to checkpoint blockade immunotherapy and is reversible with currently available biologics.
Collapse
Affiliation(s)
- G Sari
- Department of Pathology, UMass Chan Medical School, 55 Lake Avenue North, Worcester, MA, 01655, USA
| | - K Dhatchinamoorthy
- Department of Pathology, UMass Chan Medical School, 55 Lake Avenue North, Worcester, MA, 01655, USA
| | - L Orellano-Ariza
- Department of Pathology, UMass Chan Medical School, 55 Lake Avenue North, Worcester, MA, 01655, USA
| | - L M Ferreira
- Program in Molecular Medicine, Diabetes Center of Excellence, UMass Chan Medical School, Worcester, MA, USA
| | - M A Brehm
- Program in Molecular Medicine, Diabetes Center of Excellence, UMass Chan Medical School, Worcester, MA, USA
| | - K Rock
- Department of Pathology, UMass Chan Medical School, 55 Lake Avenue North, Worcester, MA, 01655, USA.
| |
Collapse
|
39
|
Ortega-Prieto AM, Jimenez-Guardeño JM. Interferon-stimulated genes and their antiviral activity against SARS-CoV-2. mBio 2024; 15:e0210024. [PMID: 39171921 PMCID: PMC11389394 DOI: 10.1128/mbio.02100-24] [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: 08/23/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic remains an international health problem caused by the recent emergence of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). As of May 2024, SARS-CoV-2 has caused more than 775 million cases and over 7 million deaths globally. Despite current vaccination programs, infections are still rapidly increasing, mainly due to the appearance and spread of new variants, variations in immunization rates, and limitations of current vaccines in preventing transmission. This underscores the need for pan-variant antivirals and treatments. The interferon (IFN) system is a critical element of the innate immune response and serves as a frontline defense against viruses. It induces a generalized antiviral state by transiently upregulating hundreds of IFN-stimulated genes (ISGs). To gain a deeper comprehension of the innate immune response to SARS-CoV-2, its connection to COVID-19 pathogenesis, and the potential therapeutic implications, this review provides a detailed overview of fundamental aspects of the diverse ISGs identified for their antiviral properties against SARS-CoV-2. It emphasizes the importance of these proteins in controlling viral replication and spread. Furthermore, we explore methodological approaches for the identification of ISGs and conduct a comparative analysis with other viruses. Deciphering the roles of ISGs and their interactions with viral pathogens can help identify novel targets for antiviral therapies and enhance our preparedness to confront current and future viral threats.
Collapse
Affiliation(s)
- Ana Maria Ortega-Prieto
- Departamento de Microbiología, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Málaga, Spain
| | - Jose M Jimenez-Guardeño
- Departamento de Microbiología, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Málaga, Spain
| |
Collapse
|
40
|
Bertlin JAC, Pauzaite T, Liang Q, Wit N, Williamson JC, Sia JJ, Matheson NJ, Ortmann BM, Mitchell TJ, Speak AO, Zhang Q, Nathan JA. VHL synthetic lethality screens uncover CBF-β as a negative regulator of STING. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.03.610968. [PMID: 39282259 PMCID: PMC11398426 DOI: 10.1101/2024.09.03.610968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Clear cell renal cell carcinoma (ccRCC) represents the most common form of kidney cancer and is typified by biallelic inactivation of the von Hippel-Lindau (VHL) tumour suppressor gene. Here, we undertake genome-wide CRISPR/Cas9 screening to reveal synthetic lethal interactors of VHL, and uncover that loss of Core Binding Factor β (CBF-β) causes cell death in VHL-null ccRCC cell lines and impairs tumour establishment and growth in vivo. This synthetic relationship is independent of the elevated activity of hypoxia inducible factors (HIFs) in VHL-null cells, but does involve the RUNX transcription factors that are known binding partners of CBF-β. Mechanistically, CBF-β loss leads to upregulation of type I interferon signalling, and we uncover a direct inhibitory role for CBF-β at the STING locus controlling Interferon Stimulated Gene expression. Targeting CBF-β in kidney cancer both selectively induces tumour cell lethality and promotes activation of type I interferon signalling.
Collapse
Affiliation(s)
- James A C Bertlin
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Department of Medicine, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Tekle Pauzaite
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Department of Medicine, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Qian Liang
- Simmons Comprehensive Cancer Center, Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Niek Wit
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Department of Medicine, University of Cambridge, Cambridge, CB2 0AW, UK
| | - James C Williamson
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Department of Medicine, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Jia Jhing Sia
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Department of Medicine, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Nicholas J Matheson
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Department of Medicine, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Brian M Ortmann
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Department of Medicine, University of Cambridge, Cambridge, CB2 0AW, UK
- Wolfson Childhood Cancer Research Centre, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| | - Thomas J Mitchell
- Early Cancer Institute and Department of Surgery, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Anneliese O Speak
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Department of Medicine, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Qing Zhang
- Simmons Comprehensive Cancer Center, Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - James A Nathan
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Department of Medicine, University of Cambridge, Cambridge, CB2 0AW, UK
| |
Collapse
|
41
|
Serrero MC, Paludan SR. Restriction factors regulating human herpesvirus infections. Trends Immunol 2024; 45:662-677. [PMID: 39198098 DOI: 10.1016/j.it.2024.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 09/01/2024]
Abstract
Herpesviruses are DNA viruses and the cause of diseases ranging from mild skin conditions to severe brain diseases. Mammalian antiviral host defense comprises an array of mechanisms, including restriction factors (RFs), which block specific steps in viral replication cycles. In recent years, knowledge of RFs that contribute to controlling herpesvirus infections has expanded significantly, along with a new understanding of viral evasion mechanisms and disease pathogenesis. By integrating findings from human genetics, murine models, and cellular studies, this review provides a current view of RF control of herpesvirus infections. We also explore the regulation of RF expression, discuss the roles of RFs in diseases, and point towards their growing potential as candidate therapeutic targets.
Collapse
Affiliation(s)
- Manutea C Serrero
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus, Denmark
| | - Søren R Paludan
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus, Denmark.
| |
Collapse
|
42
|
Guduric‐Fuchs J, Pedrini E, Bertelli PM, McDonnell S, Pathak V, McLoughlin K, O'Neill CL, Stitt AW, Medina RJ. A new gene signature for endothelial senescence identifies self-RNA sensing by retinoic acid-inducible gene I as a molecular facilitator of vascular aging. Aging Cell 2024; 23:e14240. [PMID: 39422883 PMCID: PMC11488300 DOI: 10.1111/acel.14240] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/30/2024] [Accepted: 05/20/2024] [Indexed: 10/19/2024] Open
Abstract
The number of senescent vascular endothelial cells increases during aging and their dysfunctional phenotype contributes to age-related cardiovascular disease. Identification of senescent cells is challenging as molecular changes are often tissue specific and occur amongst clusters of normal cells. Here, we established, benchmarked, and validated a new gene signature called EndoSEN that pinpoints senescent endothelial cells. The EndoSEN signature was enriched for interferon-stimulated genes (ISG) and correlated with the senescence-associated secretory phenotype (SASP). SASP establishment is classically attributed to DNA damage and cyclic GMP-AMP synthase activation, but our results revealed a pivotal role for RNA accumulation and sensing in senescent endothelial cells. Mechanistically, we showed that endothelial cell senescence hallmarks include self-RNA accumulation, RNA sensor RIG-I upregulation, and an ISG signature. Moreover, a virtual model of RIG-I knockout in endothelial cells underscored senescence as a key pathway regulated by this sensor. We tested and confirmed that RIG-I knockdown was sufficient to extend the lifespan and decrease the SASP in endothelial cells. Taken together, our evidence suggests that targeting RNA sensing is a potential strategy to delay vascular aging.
Collapse
Affiliation(s)
- Jasenka Guduric‐Fuchs
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queen's University BelfastBelfastUK
| | - Edoardo Pedrini
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queen's University BelfastBelfastUK
- Center for Omics Sciences (COSR)San Raffaele Scientific InstituteMilanItaly
| | - Pietro M. Bertelli
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queen's University BelfastBelfastUK
| | - Shannon McDonnell
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queen's University BelfastBelfastUK
| | - Varun Pathak
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queen's University BelfastBelfastUK
| | - Kiran McLoughlin
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queen's University BelfastBelfastUK
| | - Christina L. O'Neill
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queen's University BelfastBelfastUK
| | - Alan W. Stitt
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queen's University BelfastBelfastUK
| | - Reinhold J. Medina
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queen's University BelfastBelfastUK
- Department of Eye and Vision ScienceInstitute for Life Course and Medical Science, University of LiverpoolLiverpoolUK
| |
Collapse
|
43
|
Halama A, Zaghlool S, Thareja G, Kader S, Al Muftah W, Mook-Kanamori M, Sarwath H, Mohamoud YA, Stephan N, Ameling S, Pucic Baković M, Krumsiek J, Prehn C, Adamski J, Schwenk JM, Friedrich N, Völker U, Wuhrer M, Lauc G, Najafi-Shoushtari SH, Malek JA, Graumann J, Mook-Kanamori D, Schmidt F, Suhre K. A roadmap to the molecular human linking multiomics with population traits and diabetes subtypes. Nat Commun 2024; 15:7111. [PMID: 39160153 PMCID: PMC11333501 DOI: 10.1038/s41467-024-51134-x] [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: 08/01/2023] [Accepted: 07/26/2024] [Indexed: 08/21/2024] Open
Abstract
In-depth multiomic phenotyping provides molecular insights into complex physiological processes and their pathologies. Here, we report on integrating 18 diverse deep molecular phenotyping (omics-) technologies applied to urine, blood, and saliva samples from 391 participants of the multiethnic diabetes Qatar Metabolomics Study of Diabetes (QMDiab). Using 6,304 quantitative molecular traits with 1,221,345 genetic variants, methylation at 470,837 DNA CpG sites, and gene expression of 57,000 transcripts, we determine (1) within-platform partial correlations, (2) between-platform mutual best correlations, and (3) genome-, epigenome-, transcriptome-, and phenome-wide associations. Combined into a molecular network of > 34,000 statistically significant trait-trait links in biofluids, our study portrays "The Molecular Human". We describe the variances explained by each omics in the phenotypes (age, sex, BMI, and diabetes state), platform complementarity, and the inherent correlation structures of multiomics data. Further, we construct multi-molecular network of diabetes subtypes. Finally, we generated an open-access web interface to "The Molecular Human" ( http://comics.metabolomix.com ), providing interactive data exploration and hypotheses generation possibilities.
Collapse
Affiliation(s)
- Anna Halama
- Bioinformatics Core, Weill Cornell Medicine-Qatar, Education City, Doha, Qatar.
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA.
| | - Shaza Zaghlool
- Bioinformatics Core, Weill Cornell Medicine-Qatar, Education City, Doha, Qatar
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Gaurav Thareja
- Bioinformatics Core, Weill Cornell Medicine-Qatar, Education City, Doha, Qatar
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Sara Kader
- Bioinformatics Core, Weill Cornell Medicine-Qatar, Education City, Doha, Qatar
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Wadha Al Muftah
- Qatar Genome Program, Qatar Foundation, Qatar Science and Technology Park, Innovation Center, Doha, Qatar
- Department of Genetic Medicine, Weill Cornell Medicine, Doha, Qatar
| | | | - Hina Sarwath
- Proteomics Core, Weill Cornell Medicine-Qatar, Education City, Doha, Qatar
| | | | - Nisha Stephan
- Bioinformatics Core, Weill Cornell Medicine-Qatar, Education City, Doha, Qatar
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Sabine Ameling
- German Centre for Cardiovascular Research, Partner Site Greifswald, University Medicine Greifswald, Greifswald, Germany
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | | | - Jan Krumsiek
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Cornelia Prehn
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, Neuherberg, Germany
| | - Jerzy Adamski
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Jochen M Schwenk
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Solna, Sweden
| | - Nele Friedrich
- German Centre for Cardiovascular Research, Partner Site Greifswald, University Medicine Greifswald, Greifswald, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Uwe Völker
- German Centre for Cardiovascular Research, Partner Site Greifswald, University Medicine Greifswald, Greifswald, Germany
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Gordan Lauc
- Genos Glycoscience Research Laboratory, Zagreb, Croatia
- Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| | - S Hani Najafi-Shoushtari
- MicroRNA Core Laboratory, Division of Research, Weill Cornell Medicine-Qatar, Education City, Doha, Qatar
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA
| | - Joel A Malek
- Department of Genetic Medicine, Weill Cornell Medicine, Doha, Qatar
- Genomics Core, Weill Cornell Medicine-Qatar, Education City, Doha, Qatar
| | - Johannes Graumann
- Institute of Translational Proteomics, Department of Medicine, Philipps-Universität Marburg, Marburg, Germany
| | - Dennis Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, the Netherlands
| | - Frank Schmidt
- Proteomics Core, Weill Cornell Medicine-Qatar, Education City, Doha, Qatar
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Karsten Suhre
- Bioinformatics Core, Weill Cornell Medicine-Qatar, Education City, Doha, Qatar.
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA.
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
44
|
Swart M, Redpath AN, Ogbechi J, Cardenas R, Topping L, Compeer EB, Goddard M, Chanalaris A, Williams R, Brewer DS, Smart N, Monaco C, Troeberg L. The extracellular heparan sulfatase SULF2 limits myeloid IFNβ signaling and Th17 responses in inflammatory arthritis. Cell Mol Life Sci 2024; 81:350. [PMID: 39141086 PMCID: PMC11335274 DOI: 10.1007/s00018-024-05333-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 06/18/2024] [Accepted: 06/25/2024] [Indexed: 08/15/2024]
Abstract
Heparan sulfate (HS) proteoglycans are important regulators of cellular responses to soluble mediators such as chemokines, cytokines and growth factors. We profiled changes in expression of genes encoding HS core proteins, biosynthesis enzymes and modifiers during macrophage polarisation, and found that the most highly regulated gene was Sulf2, an extracellular HS 6-O-sulfatase that was markedly downregulated in response to pro-inflammatory stimuli. We then generated Sulf2+/- bone marrow chimeric mice and examined inflammatory responses in antigen-induced arthritis, as a model of rheumatoid arthritis. Resolution of inflammation was impaired in myeloid Sulf2+/- chimeras, with elevated joint swelling and increased abundance of pro-arthritic Th17 cells in synovial tissue. Transcriptomic and in vitro analyses indicated that Sulf2 deficiency increased type I interferon signaling in bone marrow-derived macrophages, leading to elevated expression of the Th17-inducing cytokine IL6. This establishes that dynamic remodeling of HS by Sulf2 limits type I interferon signaling in macrophages, and so protects against Th17-driven pathology.
Collapse
Affiliation(s)
- Maarten Swart
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7FY, UK
| | - Andia N Redpath
- Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Oxford, OX1 3PT, UK
| | - Joy Ogbechi
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7FY, UK
| | - Ryan Cardenas
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Bob Champion Research and Education Building, Rosalind Franklin Road, Norwich, NR4 7UQ, UK
| | - Louise Topping
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7FY, UK
| | - Ewoud B Compeer
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7FY, UK
| | - Michael Goddard
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7FY, UK
| | - Anastasios Chanalaris
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7FY, UK
| | - Richard Williams
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7FY, UK
| | - Daniel S Brewer
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Bob Champion Research and Education Building, Rosalind Franklin Road, Norwich, NR4 7UQ, UK
| | - Nicola Smart
- Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Oxford, OX1 3PT, UK
| | - Claudia Monaco
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7FY, UK
| | - Linda Troeberg
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7FY, UK.
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Bob Champion Research and Education Building, Rosalind Franklin Road, Norwich, NR4 7UQ, UK.
| |
Collapse
|
45
|
Huang L, Tang W, He L, Li M, Lin X, Hu A, Huang X, Wu Z, Wu Z, Chen S, Hu Y. Engineered probiotic Escherichia coli elicits immediate and long-term protection against influenza A virus in mice. Nat Commun 2024; 15:6802. [PMID: 39122688 PMCID: PMC11315933 DOI: 10.1038/s41467-024-51182-3] [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: 02/09/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
Influenza virus infection remains a major global health problem and requires a universal vaccine with broad protection against different subtypes as well as a rapid-response vaccine to provide immediate protection in the event of an epidemic outbreak. Here, we show that intranasal administration of probiotic Escherichia coli Nissle 1917 activates innate immunity in the respiratory tract and provides immediate protection against influenza virus infection within 1 day. Based on this vehicle, a recombinant strain is engineered to express and secret five tandem repeats of the extracellular domain of matrix protein 2 from different influenza virus subtypes. Intranasal vaccination with this strain induces durable humoral and mucosal responses in the respiratory tract, and provides broad protection against the lethal challenge of divergent influenza viruses in female BALB/c mice. Our findings highlight a promising delivery platform for developing mucosal vaccines that provide immediate and sustained protection against respiratory pathogens.
Collapse
Affiliation(s)
- Ling Huang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wei Tang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Lina He
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Mengke Li
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xian Lin
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- Hubei JiangXia Laboratory, Wuhan, 430071, China
| | - Ao Hu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xindi Huang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Zhouyu Wu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhiyong Wu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shiyun Chen
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
| | - Yangbo Hu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
- Hubei JiangXia Laboratory, Wuhan, 430071, China.
| |
Collapse
|
46
|
Danac JMC, Matthews RE, Gungi A, Qin C, Parsons H, Antrobus R, Timms RT, Tchasovnikarova IA. Competition between two HUSH complexes orchestrates the immune response to retroelement invasion. Mol Cell 2024; 84:2870-2881.e5. [PMID: 39013473 DOI: 10.1016/j.molcel.2024.06.020] [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: 02/26/2024] [Revised: 05/31/2024] [Accepted: 06/20/2024] [Indexed: 07/18/2024]
Abstract
The human silencing hub (HUSH) preserves genome integrity through the epigenetic repression of invasive genetic elements. However, despite our understanding of HUSH as an obligate complex of three subunits, only loss of MPP8 or Periphilin, but not TASOR, triggers interferon signaling following derepression of endogenous retroelements. Here, we resolve this paradox by characterizing a second HUSH complex that shares MPP8 and Periphilin but assembles around TASOR2, an uncharacterized paralog of TASOR. Whereas HUSH represses LINE-1 retroelements marked by the repressive histone modification H3K9me3, HUSH2 is recruited by the transcription factor IRF2 to repress interferon-stimulated genes. Mechanistically, HUSH-mediated retroelement silencing sequesters the limited pool of the shared subunits MPP8 and Periphilin, preventing TASOR2 from forming HUSH2 complexes and hence relieving the HUSH2-mediated repression of interferon-stimulated genes. Thus, competition between two HUSH complexes intertwines retroelement silencing with the induction of an immune response, coupling epigenetic and immune aspects of genome defense.
Collapse
Affiliation(s)
- Joshua Miguel C Danac
- The Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK; Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Rachael E Matthews
- The Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK; Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Akhila Gungi
- The Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK; Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Chuyan Qin
- The Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK; Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Harriet Parsons
- Department of Medicine, Cambridge Institute for Medical Research, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0XY, UK
| | - Robin Antrobus
- Department of Medicine, Cambridge Institute for Medical Research, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0XY, UK
| | - Richard T Timms
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge CB2 0AW, UK
| | - Iva A Tchasovnikarova
- The Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK; Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK.
| |
Collapse
|
47
|
Ohnezeit D, Huang J, Westerkamp U, Brinschwitz V, Schmidt C, Günther T, Czech-Sioli M, Weißelberg S, Schlemeyer T, Nakel J, Mai J, Schreiner S, Schneider C, Friedel CC, Schwanke H, Brinkmann MM, Grundhoff A, Fischer N. Merkel cell polyomavirus small tumor antigen contributes to immune evasion by interfering with type I interferon signaling. PLoS Pathog 2024; 20:e1012426. [PMID: 39110744 PMCID: PMC11333005 DOI: 10.1371/journal.ppat.1012426] [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: 08/09/2023] [Revised: 08/19/2024] [Accepted: 07/15/2024] [Indexed: 08/21/2024] Open
Abstract
Merkel cell polyomavirus (MCPyV) is the causative agent of the majority of Merkel cell carcinomas (MCC). The virus has limited coding capacity, with its early viral proteins, large T (LT) and small T (sT), being multifunctional and contributing to infection and transformation. A fundamental difference in early viral gene expression between infection and MCPyV-driven tumorigenesis is the expression of a truncated LT (LTtr) in the tumor. In contrast, sT is expressed in both conditions and contributes significantly to oncogenesis. Here, we identified novel functions of early viral proteins by performing genome-wide transcriptome and chromatin studies in primary human fibroblasts. Due to current limitations in infection and tumorigenesis models, we mimic these conditions by ectopically expressing sT, LT or LTtr, individually or in combination, at different time points. In addition to its known function in cell cycle and inflammation modulation, we reveal a fundamentally new function of sT. We show that sT regulates the type I interferon (IFN) response downstream of the type I interferon receptor (IFNAR) by interfering with the interferon-stimulated gene factor 3 (ISGF3)-induced interferon-stimulated gene (ISG) response. Expression of sT leads to a reduction in the expression of interferon regulatory factor 9 (IRF9) which is a central component of the ISGF3 complex. We further show that this function of sT is conserved in BKPyV. We provide a first mechanistic understanding of which early viral proteins trigger and control the type I IFN response, which may influence MCPyV infection, persistence and, during MCC progression, regulation of the tumor microenvironment.
Collapse
Affiliation(s)
- Denise Ohnezeit
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jiabin Huang
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ute Westerkamp
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Veronika Brinschwitz
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Claudia Schmidt
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Manja Czech-Sioli
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Samira Weißelberg
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tabea Schlemeyer
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Julia Mai
- Institute of Virology, Hannover Medical School, Hannover, Germany
- Institute of Virology, Freiburg, Germany
| | - Sabrina Schreiner
- Institute of Virology, Hannover Medical School, Hannover, Germany
- Institute of Virology, Freiburg, Germany
| | | | - Caroline C. Friedel
- Institute of Informatics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Hella Schwanke
- Institute of Genetics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Melanie M. Brinkmann
- Institute of Genetics, Technische Universität Braunschweig, Braunschweig, Germany
- Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | - Nicole Fischer
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
48
|
Bassan VL, de Freitas Martins Felício R, Ribeiro Malmegrim KC, Attié de Castro F. Myeloproliferative Neoplasms Transcriptome Reveals Pro-Inflammatory Signature and Enrichment in Peripheral Blood Monocyte-Related Genes. Cancer Invest 2024; 42:605-618. [PMID: 38958254 DOI: 10.1080/07357907.2024.2371371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 12/15/2023] [Accepted: 06/19/2024] [Indexed: 07/04/2024]
Abstract
Myeloproliferative neoplasms (MPN) are hematological diseases associated with genetic driver mutations in the JAK2, CALR, and MPL genes and exacerbated oncoinflammatory status. Analyzing public microarray data from polycythemia vera (n = 41), essential thrombocythemia (n = 21), and primary myelofibrosis (n = 9) patients' peripheral blood by in silico approaches, we found that pro-inflammatory and monocyte-related genes were differentially expressed in MPN patients' transcriptome. Genes related to cell activation, secretion of pro-inflammatory and pro-angiogenic mediators, activation of neutrophils and platelets, coagulation, and interferon pathway were upregulated in monocytes compared to controls. Together, our results suggest that molecular alterations in monocytes may contribute to oncoinflammation in MPN.
Collapse
Affiliation(s)
- Vitor Leonardo Bassan
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Rafaela de Freitas Martins Felício
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Kelen Cristina Ribeiro Malmegrim
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Fabíola Attié de Castro
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
49
|
Röszer T. MicroRNA Profile of Mouse Adipocyte-Derived Extracellular Vesicles. Cells 2024; 13:1298. [PMID: 39120327 PMCID: PMC11311276 DOI: 10.3390/cells13151298] [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: 07/08/2024] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
The post-transcriptional control of gene expression is a complex and evolving field in adipocyte biology, with the premise that the delivery of microRNA (miRNA) species to the obese adipose tissue may facilitate weight loss. Cells shed extracellular vesicles (EVs) that may deliver miRNAs as intercellular messengers. However, we know little about the miRNA profile of EVs secreted by adipocytes during postnatal development. Here, we defined the miRNA cargo of EVs secreted by mouse adipocytes in two distinct phases of development: on postnatal day 6, when adipocytes are lipolytic and thermogenic, and on postnatal day 56, when adipocytes have active lipogenesis. EVs were collected from cell culture supernatants, and their miRNA profile was defined by small RNA sequencing. The most abundant miRNA of mouse adipocyte-derived EVs was mmu-miR-148a-3p. Adipocyte EVs on postnatal day 6 were hallmarked with mmu-miR-98-5p, and some miRNAs were specific to this developmental stage, such as mmu-miR-466i-5p and 12 novel miRNAs. Adipocytes on postnatal day 56 secreted mmu-miR-365-3p, and 16 miRNAs were specific to this developmental stage. The miRNA cargo of adipocyte EVs targeted gene networks of cell proliferation, insulin signaling, interferon response, thermogenesis, and lipogenesis. We provided here a database of miRNAs secreted by developing mouse adipocytes, which may be a tool for further studies on the regulation of gene networks that control mouse adipocyte development.
Collapse
Affiliation(s)
- Tamás Röszer
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
50
|
Wright AP, Harris S, Madden S, Reyes BR, Mulamula E, Gibson A, Rauch I, Constant DA, Nice TJ. Interferon regulatory factor 6 (IRF6) determines intestinal epithelial cell development and immunity. Mucosal Immunol 2024; 17:633-650. [PMID: 38604478 PMCID: PMC11323225 DOI: 10.1016/j.mucimm.2024.03.013] [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: 01/12/2024] [Revised: 03/21/2024] [Accepted: 03/31/2024] [Indexed: 04/13/2024]
Abstract
Intestinal epithelial cell (IEC) responses to interferon (IFN) favor antiviral defense with minimal cytotoxicity, but IEC-specific factors that regulate these responses remain poorly understood. Interferon regulatory factors (IRFs) are a family of nine related transcription factors, and IRF6 is preferentially expressed by epithelial cells, but its roles in IEC immunity are unknown. In this study, Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) screens found that Irf6 deficiency enhanced IFN-stimulated antiviral responses in transformed mouse IECs but not macrophages. Furthermore, knockout (KO) of Irf6 in IEC organoids resulted in profound changes to homeostasis and immunity gene expression. Irf6 KO organoids grew more slowly, and single-cell ribonucleic acid sequencing indicated reduced expression of genes in epithelial differentiation and immunity pathways. IFN-stimulated gene expression was also significantly different in Irf6 KO organoids, with increased expression of stress and apoptosis-associated genes. Functionally, the transcriptional changes in Irf6 KO organoids were associated with increased cytotoxicity upon IFN treatment or inflammasome activation. These data indicate a previously unappreciated role for IRF6 in IEC biology, including regulation of epithelial development and moderation of innate immune responses to minimize cytotoxicity and maintain barrier function.
Collapse
Affiliation(s)
- Austin P Wright
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | - Sydney Harris
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | - Shelby Madden
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | - Bryan Ramirez Reyes
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | - Ethan Mulamula
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | - Alexis Gibson
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | - Isabella Rauch
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | - David A Constant
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | - Timothy J Nice
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA.
| |
Collapse
|