1
|
Schnappauf O, Wang H, Aksentijevich I, Kastner DL, Laxer RM. A 33-year diagnostic odyssey in an Ashkenazi Jewish patient with Aicardi-Goutières syndrome. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2025; 4:100400. [PMID: 39906892 PMCID: PMC11791129 DOI: 10.1016/j.jacig.2025.100400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/25/2024] [Accepted: 11/08/2024] [Indexed: 02/06/2025]
Abstract
The critical need for awareness and genetic testing of the SAMHD1 deletion in Ashkenazi Jewish patients is highlighted owing to its relatively high carrier frequency. Early detection can prevent severe disease complications through targeted therapy.
Collapse
Affiliation(s)
- Oskar Schnappauf
- Department of Natural Sciences and Institute for Functional Gene Analytics, Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach, Germany
| | - Hongying Wang
- Inflammatory Disease Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, Md
| | - Ivona Aksentijevich
- Inflammatory Disease Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, Md
| | - Daniel L. Kastner
- Inflammatory Disease Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, Md
| | - Ronald M. Laxer
- The Hospital for Sick Children and St Michael’s Hospital, Division of Rheumatology, Departments of Paediatrics and Medicine, GRIID Program, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
2
|
Li JB, Walkley CR. Leveraging genetics to understand ADAR1-mediated RNA editing in health and disease. Nat Rev Genet 2025:10.1038/s41576-025-00830-5. [PMID: 40229561 DOI: 10.1038/s41576-025-00830-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2025] [Indexed: 04/16/2025]
Abstract
Endogenous, long double-stranded RNA (dsRNA) can resemble viral dsRNA and be recognized by cytosolic dsRNA sensors, triggering autoimmunity. Genetic studies of rare, inherited human diseases and experiments using mouse models have established the importance of adenosine-to-inosine RNA editing by the enzyme adenosine deaminase acting on RNA 1 (ADAR1) as a critical safeguard against autoinflammatory responses to cellular dsRNA. More recently, human genetic studies have revealed that dsRNA editing and sensing mechanisms are involved in common inflammatory diseases, emphasizing the broader role of dsRNA in modulating immune responses and disease pathogenesis. These findings have highlighted the therapeutic potential of targeting dsRNA editing and sensing, as exemplified by the emergence of ADAR1 inhibition in cancer therapy.
Collapse
Affiliation(s)
- Jin Billy Li
- Department of Genetics, Stanford University, Stanford, CA, USA.
| | - Carl R Walkley
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia.
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia.
- Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia.
| |
Collapse
|
3
|
Aydin H, Bolat H. The Rare Syndrome Aicardi-Goutières 4: A Case Report and Literature Review. Dev Neurobiol 2025; 85:e22965. [PMID: 40263931 PMCID: PMC12015151 DOI: 10.1002/dneu.22965] [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: 03/06/2025] [Revised: 03/12/2025] [Accepted: 03/17/2025] [Indexed: 04/24/2025]
Abstract
Aicardi-Goutières syndrome (AGS) is a genetically heterogeneous type of interferonopathy resulting from defects in the processing or sensing of nucleic acids. The AGS phenotype encompasses a broad range of neurological and non-neurological findings. It presents with a congenital or subacute onset, manifesting as microcephaly, spasticity, dystonia, seizures, cortical blindness, and psychomotor retardation in the first year of life. The radiological and laboratory findings of AGS are generally accompanied by intracranial calcification, white matter abnormalities, cerebral atrophy, and cerebrospinal fluid lymphocytic pleocytosis. A case diagnosed as AGS type 4 among patients presenting to the Balikesir University Medical Faculty pediatric neurology clinic, Türkiye, between August 1, 2024, and February 1, 2025, and undergoing genetic testing was included in the study. The patient exhibited a coarse facial appearance, a low ear line, scoliosis, contractures in the upper and lower extremities, hyperactive deep tendon reflexes, an equivocal Babinski response, and upper and lower extremity muscle strength of 3/5. The patient was started on levetiracetam at 20 mg/kg in two doses for epilepsy. Whole exome sequencing revealed a homozygous pathogenic variant in RNASEH2A. Parental genetic analyses for the targeted variant were heterozygous. In conclusion, the diagnosis of AGS relies on clinical characteristics and genetic testing. Basic neurological characteristics include developmental delay, dystonia, microcephaly, brain calcification, and leukodystrophy. Although data concerning genotype-phenotype in AGS type 4 have been reported in the literature, these are still limited.
Collapse
Affiliation(s)
- Hilal Aydin
- Faculty of Medicine, Department of PediatricsBalikesir UniversityBalikesirTürkiye
| | - Hilmi Bolat
- Faculty of Medicine, Department of Medical GeneticsBalikesir UniversityBalikesirTürkiye
| |
Collapse
|
4
|
Yamazaki S, Kaneko S, Shimbo A, Irabu H, Ogino R, Miyamoto T, Izawa K, Segawa Y, Kakizaki J, Mori M, Shimizu M. Overlapping Aicardi-Goutières and Singleton-Merten syndromes with a heterozygous gain-of-function mutation in IFIH1 mimicking juvenile idiopathic arthritis. Immunol Med 2025:1-5. [PMID: 40116369 DOI: 10.1080/25785826.2025.2479148] [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: 09/03/2024] [Accepted: 03/01/2025] [Indexed: 03/23/2025] Open
Abstract
Aicardi-Goutières syndrome (AGS) and Singleton-Merten syndrome (SMS) are associated with heterozygous gain-of-function mutations in the interferon induced with helicase C domain 1 (IFIH1) gene. Recent reports describe patients exhibiting overlapping clinical features of AGS and SMS, along with marked type I interferon (IFN) overproduction. However, the clinical characteristics and optimal treatment strategies remain unclear. Herein, we present a patient with overlapping clinical features of AGS and SMS who was initially misdiagnosed with juvenile idiopathic arthritis. Surgical soft tissue release of the hip and knee joints improved joint deformities and spastic paraparesis. Baricitinib effectively treated refractory chilblains and skin ulcers while reducing IFN-stimulated gene overexpression in peripheral blood. These findings indicate that baricitinib may be a safe and effective treatment for AGS-SMS overlap, and surgical intervention may be a viable option for refractory joint deformities with spastic paraparesis.
Collapse
Affiliation(s)
- Susumu Yamazaki
- Department of Pediatrics and Adolescent Medicine, Juntendo University Nerima Hospital, Tokyo, Japan
- Department of Lifetime Clinical Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shuya Kaneko
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Asami Shimbo
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hitoshi Irabu
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ryo Ogino
- Department of Pediatrics, Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Takayuki Miyamoto
- Department of Pediatrics, Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Kazushi Izawa
- Department of Pediatrics, Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Yuko Segawa
- Department of Orthopedic Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Jun Kakizaki
- Department of Orthopedic Surgery, Chiba Children's Hospital, Chiba, Japan
| | - Masaaki Mori
- Department of Lifetime Clinical Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masaki Shimizu
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
5
|
Braidotti S, Ferraro RM, Franca R, Genova E, Giambuzzi F, Mancini A, Marinozzi V, Pugnetti L, Zudeh G, Tesser A, Tommasini A, Decorti G, Giliani SC, Stocco G. Pharmacological evaluation of drug therapies in Aicardi-Goutières syndrome: insights from patient-derived neural stem cells. Front Pharmacol 2025; 16:1549183. [PMID: 40183101 PMCID: PMC11966042 DOI: 10.3389/fphar.2025.1549183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/26/2025] [Indexed: 04/05/2025] Open
Abstract
Aicardi-Goutières syndrome (AGS) is a rare genetic disorder classified among type I interferonopathies. Current pharmacological management of AGS is symptomatic and supportive, with recent clinical applications of JAK inhibitors (JAKi) and antiretroviral therapies (RTIs). To investigate the effects of these therapies, patient-specific induced pluripotent stem cells (iPSCs) were generated by reprogramming fibroblasts from three AGS patients with distinct genetic mutations (AGS1, AGS2, AGS7) and differentiated into neural stem cells (NSCs). iPSCs and NSCs derived from commercial BJ fibroblasts of a healthy donor served as control. The cytotoxic effects of glucocorticoids, thiopurines, JAK inhibitors (ruxolitinib, baricitinib, tofacitinib, pacritinib), and RTIs (abacavir, lamivudine, zidovudine) were evaluated using the MTT assay. Results showed that glucocorticoids did not compromise NSC viability. Among thiopurines, thioguanine, but not mercaptopurine, exhibited cytotoxicity in NSCs. All tested JAK inhibitors, except pacritinib, were non-toxic to iPSCs and NSCs. Interestingly, high concentrations of certain JAK inhibitors (ruxolitinib, baricitinib, tofacitinib) led to an unexpected increase in cell viability in AGS patient-derived cells compared to control, suggesting potential alterations in cell proliferation or stress responses. RTIs demonstrated no cytotoxicity, except for zidovudine, which showed selective toxicity in AGS2-derived iPSCs compared to controls. These findings suggest that glucocorticoids, JAK inhibitors (excluding pacritinib), and RTIs are likely safe for NSCs of AGS patients, while caution is warranted with thioguanine and pacritinib. Further studies are needed to explore the mechanisms underlying increased cell viability at high JAK inhibitor concentrations and the selective sensitivity to zidovudine.
Collapse
Affiliation(s)
- Stefania Braidotti
- Department of Paediatrics, Institute for Maternal and Child Health (I.R.C.C.S) Burlo Garofolo, Trieste, Italy
| | - Rosalba Monica Ferraro
- “Angelo Nocivelli” Institute for Molecular Medicine, ASST Spedali Civili, Brescia, Italy
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Raffaella Franca
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Elena Genova
- Department of Paediatrics, Institute for Maternal and Child Health (I.R.C.C.S) Burlo Garofolo, Trieste, Italy
| | - Francesco Giambuzzi
- Department of Advanced Translational Diagnostics, Institute for Maternal & Child Health (I.R.C.C.S) Burlo Garofolo, Trieste, Italy
| | - Andrea Mancini
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Valentina Marinozzi
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Letizia Pugnetti
- Department of Advanced Translational Diagnostics, Institute for Maternal & Child Health (I.R.C.C.S) Burlo Garofolo, Trieste, Italy
| | - Giulia Zudeh
- Department of Advanced Translational Diagnostics, Institute for Maternal & Child Health (I.R.C.C.S) Burlo Garofolo, Trieste, Italy
| | - Alessandra Tesser
- Department of Paediatrics, Institute for Maternal and Child Health (I.R.C.C.S) Burlo Garofolo, Trieste, Italy
| | - Alberto Tommasini
- Department of Paediatrics, Institute for Maternal and Child Health (I.R.C.C.S) Burlo Garofolo, Trieste, Italy
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Giuliana Decorti
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
- Department of Advanced Translational Diagnostics, Institute for Maternal & Child Health (I.R.C.C.S) Burlo Garofolo, Trieste, Italy
| | - Silvia Clara Giliani
- “Angelo Nocivelli” Institute for Molecular Medicine, ASST Spedali Civili, Brescia, Italy
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Gabriele Stocco
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
- Department of Advanced Translational Diagnostics, Institute for Maternal & Child Health (I.R.C.C.S) Burlo Garofolo, Trieste, Italy
| |
Collapse
|
6
|
Han F, Fan S, Hou B, Zhou L, Yao M, Shen M, Zhu Y, Wardlaw JM, Ni J. Inflammatory disorders that affect the cerebral small vessels. Chin Med J (Engl) 2025:00029330-990000000-01479. [PMID: 40090970 DOI: 10.1097/cm9.0000000000003574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Indexed: 03/19/2025] Open
Abstract
ABSTRACT This comprehensive review synthesizes the latest advancements in understanding inflammatory disorders affecting cerebral small vessels, a distinct yet understudied category within cerebral small vessel diseases (SVD). Unlike classical SVD, these inflammatory conditions exhibit unique clinical presentations, imaging patterns, and pathophysiological mechanisms, posing significant diagnostic and therapeutic challenges. Highlighting their heterogeneity, this review spans primary angiitis of the central nervous system, cerebral amyloid angiopathy-related inflammation, systemic vasculitis, secondary vasculitis, and vasculitis in autoinflammatory diseases. Key discussions focus on emerging insights into immune-mediated processes, neuroimaging characteristics, and histopathological distinctions. Furthermore, this review underscores the importance of standardized diagnostic frameworks, individualized immunomodulation approaches, and novel targeted therapies to address unmet clinical demands.
Collapse
Affiliation(s)
- Fei Han
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Siyuan Fan
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Bo Hou
- Department of Radiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Lixin Zhou
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Ming Yao
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Min Shen
- Department of Rare Diseases, Department of Rheumatology and Clinical Immunology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Yicheng Zhu
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Joanna M Wardlaw
- Centre for Clinical Brain Sciences and UK Dementia Research Institute, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
| | - Jun Ni
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
7
|
Cao AB, Devant P, Wang C, Sun M, Kennedy SN, Ma W, Ruan J, Kagan JC. LPS binding caspase activation and recruitment domains (CARDs) are bipartite lipid binding modules. SCIENCE ADVANCES 2025; 11:eadt9027. [PMID: 40053584 PMCID: PMC11887843 DOI: 10.1126/sciadv.adt9027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025]
Abstract
Caspase-11 is an innate immune pattern recognition receptor (PRR) that detects cytosolic bacterial lipopolysaccharides (LPS) through its caspase activation and recruitment domain (CARD). Caspase-11 also detects eukaryotic (i.e., self) lipids. This observation raises the question of whether common or distinct mechanisms govern caspase interactions with self- and nonself-lipids. In this study, using biochemical, computational, and cell-based assays, we report that the caspase-11 CARD functions as a bipartite lipid-binding module. Distinct regions within the CARD bind to phosphate groups and long acyl chains of self- and nonself-lipids. Self-lipid binding capability is conserved across numerous caspase-11 homologs and orthologs. The symmetry in self- and nonself-lipid detection mechanisms enabled us to engineer an LPS-binding domain de novo, using an ancestral CARD-like domain present in the fish Amphilophus citrinellus. These findings offer insights into the molecular basis of LPS recognition by caspase-11 and highlight the fundamental and likely inseparable relationship between self and nonself discrimination.
Collapse
Affiliation(s)
- Anh B. Cao
- Division of Gastroenterology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Pascal Devant
- Division of Gastroenterology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Chengliang Wang
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Mengyu Sun
- Division of Gastroenterology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Stephanie N. Kennedy
- Division of Gastroenterology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Weiyi Ma
- Division of Gastroenterology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Jianbin Ruan
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Jonathan C. Kagan
- Division of Gastroenterology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
8
|
Kyawe PP, Liu P, Jiang Z, Bradley ES, Cicuto T, Trombly MI, Silverman N, Fitzgerald KA, McDougall WM, Wang JP. CRISPR editing of candidate host factors that impact influenza A virus infection. Microbiol Spectr 2025; 13:e0262724. [PMID: 39887213 DOI: 10.1128/spectrum.02627-24] [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] [Received: 10/28/2024] [Accepted: 12/23/2024] [Indexed: 02/01/2025] Open
Abstract
Influenza A virus (IAV) is a respiratory pathogen with a segmented negative-sense RNA genome that can cause epidemics and pandemics. The host factors required for the complete IAV infectious cycle have not been fully identified. Here, we examined three host factors for their contributions to IAV infectivity. We performed CRISPR-mediated knockout of cytidine monophosphate N-acetylneuraminic acid synthetase (CMAS) as well as CRISPR-mediated overexpression of beta-1,4 N-acetylgalactosaminyltransferase 2 (B4GALNT2) and adenosine deaminase acting on RNA 1 (ADAR1) in the human bronchial epithelial A549 cell line and evaluated the impact on IAV and other RNA viruses. We confirmed that knockout of CMAS or overexpression of B4GALNT2 restricts IAV infection by diminishing binding to the cell surface but has no effect on vesicular stomatitis virus infection. Although ADAR1 overexpression does not significantly inhibit IAV replication, it has a pro-viral effect with coxsackie B virus (CVB) infection. This pro-viral effect is not likely secondary to reduced type I interferon (IFN) production, as the induction of the IFN-stimulated genes ISG15 and CXCL10 is negligible in both parent and ADAR1-overexpressing A549 cells following CVB challenge. In contrast, ISG15 and CXCL10 production is robust and equal for parent and ADAR1-overexpressing A549 cells challenged with IAV. Taken together, these data provide insights into how host factors can be further explored to understand the dynamics of pro- and anti-viral factors.IMPORTANCEInfluenza A virus (IAV) remains a global threat due to its ability to cause pandemics, making the identification of host factors essential for developing new antiviral strategies. In this study, we utilized CRISPR-based techniques to investigate host factors that impact IAV infectivity. Knockout of CMAS, a key enzyme in sialic acid biosynthesis, significantly reduced IAV binding and infection by disrupting sialic acid production on the cell surface. Overexpression of B4GALNT2 had similar effects, conferring resistance to IAV infection through diminished cell-surface binding. Overexpression of ADAR1, known for its role in RNA editing and immune regulation, impacted IAV replication minimally but enhanced coxsackie B virus replication. Such findings reveal the diverse roles of host factors in viral infection, offering insights for targeted therapeutic development against IAV and other pathogens.
Collapse
Affiliation(s)
- Pyae Phyo Kyawe
- Department of Medicine, Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Interdisciplinary Graduate Program, Morningside Graduate School of Biomedical Sciences, Worcester, Massachusetts, USA
| | - Ping Liu
- Department of Medicine, Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Zhaozhao Jiang
- Department of Medicine, Division of Innate Immunity, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Evan S Bradley
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Thomas Cicuto
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Melanie I Trombly
- Department of Medicine, Division of Innate Immunity, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Neal Silverman
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Katherine A Fitzgerald
- Department of Medicine, Division of Innate Immunity, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - William M McDougall
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Jennifer P Wang
- Department of Medicine, Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
9
|
Hou L, Zhou P, Du Y, Wang X, Zhao C. Short-term efficacy of tofacitinib, a JAK inhibitor, in IFIH1-related Aicardi-Goutières syndrome. Eur J Med Genet 2025; 75:105006. [PMID: 40043752 DOI: 10.1016/j.ejmg.2025.105006] [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: 01/15/2025] [Revised: 02/23/2025] [Accepted: 03/02/2025] [Indexed: 03/10/2025]
Abstract
Aicardi-Goutières syndrome (AGS) is a genetically heterogeneous type-I interferonopathy presenting in infancy with intracranial calcifications, white matter lesions, and brain atrophy. AGS7, caused by gain-of-function (GOF) mutations in the IFIH1 gene, triggers excessive type-I interferon production, leading to autoimmune responses. We describe an 18-year-old female diagnosed with AGS7 due to a somatic GOF mutation in IFIH1. In 2014, she presented with multiple joint swelling, facial rash, and hair loss, and received a diagnosis of juvenile idiopathic arthritis and systemic lupus erythematosus. Traditional immunosuppressants were administered, but provided little benefit. Genetic testing in 2023 revealed a GOF variant (p.R720G) in IFIH1. Given the link between IFIH1 variants and the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway, we administered tofacitinib (a JAK inhibitor) and oral methylprednisolone, with tapering of the traditional immunosuppressants. After nearly one year, the patient showed no significant disease activity and normal hair growth, with no notable changes in thyroid function. Although treatment of AGS remains challenging, this case suggests tofacitinib can successfully manage AGS7 symptoms. More clinical studies are needed to verify the long-term safety and efficacy of tofacitinib.
Collapse
Affiliation(s)
- Ling Hou
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Peng Zhou
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Yue Du
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Xiuli Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Chengguang Zhao
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|
10
|
Zeng Y, Tao P, Wang J, Li T, Du Y, Wang X, Wang W, Peng S, Wang W, Ma M, Song H, Yu X, Zhou Q. Somatic gain-of-function mutation in TLR7 causes early-onset systemic lupus erythematosus. Ann Rheum Dis 2025; 84:442-450. [PMID: 39919974 DOI: 10.1016/j.ard.2025.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/23/2024] [Accepted: 12/27/2024] [Indexed: 02/09/2025]
Abstract
OBJECTIVES We identified a case of early-onset systemic lupus erythematosus (SLE) characterised by acute immune thrombocytopenia, recurrent fever, pneumonia, myocardial damage, thyroid dysfunction, lymphadenopathy, hepatosplenomegaly, and intracranial calcification. Our objective was to investigate the genetic and molecular mechanisms underlying the disease. METHODS Whole exome sequencing and targeted sequencing were performed and a somatic mutation in TLR7 was identified. RNA sequencing, quantitative polymerase chain reaction (qPCR), intracellular cytokine staining, and phospho-flow cytometry were performed to characterise inflammatory signatures. In addition, nuclear factor κB dual-luciferase reporter assays, qPCR, and RNA pull-down assays were performed to assess the functional impact of the TLR7 mutation on immune signalling. RESULTS We identified a novel somatic TLR7 mutation (p.Phe506Ser) that is likely to arise during early embryonic development. This mutation led to transcriptional upregulation of proinflammatory cytokines and interferon-stimulated genes, such as TNF and IFI27, with significant increases in intracellular cytokine expression, including TNF, following stimulation with the ligand single-stranded RNA (ssRNA) and the agonist R848 in the patient's peripheral blood mononuclear cells (PBMCs). In addition, functional analysis in HEK293T cells demonstrated that the mutant TLR7 exhibited increased binding affinity for ssRNA and enhanced responsiveness to agonists, resulting in hyperactivation of TLR7-mediated signalling. CONCLUSIONS We report the first case of early-onset SLE caused by a somatic TLR7 gain-of-function mutation. Our findings demonstrate that the TLR7 F506S mutation drives excessive proinflammatory signalling in the patient's PBMCs, contributing to disease pathogenesis.
Collapse
Affiliation(s)
- Yi Zeng
- Department of Rheumatology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine and Liangzhu Laboratory of Zhejiang University, Hangzhou, China
| | - Panfeng Tao
- Department of Rheumatology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine and Liangzhu Laboratory of Zhejiang University, Hangzhou, China; College of Life Sciences, Zhejiang University, Hangzhou, China.
| | - Jun Wang
- Department of Rheumatology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine and Liangzhu Laboratory of Zhejiang University, Hangzhou, China
| | - Ting Li
- Department of Pediatrics, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yue Du
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiuli Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wei Wang
- Department of Pediatrics, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Siming Peng
- Department of Pediatrics, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wei Wang
- Department of Pediatrics, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Mingsheng Ma
- Department of Pediatrics, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Hongmei Song
- Department of Pediatrics, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Xiaomin Yu
- Department of Rheumatology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine and Liangzhu Laboratory of Zhejiang University, Hangzhou, China; Department of Rheumatology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Qing Zhou
- Department of Rheumatology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine and Liangzhu Laboratory of Zhejiang University, Hangzhou, China.
| |
Collapse
|
11
|
Charras A, Hiraki LT, Lewandowski L, Hedrich CM. Genetic and epigenetic factors shape phenotypes and outcomes in systemic lupus erythematosus - focus on juvenile-onset systemic lupus erythematosus. Curr Opin Rheumatol 2025; 37:149-163. [PMID: 39660463 PMCID: PMC11789615 DOI: 10.1097/bor.0000000000001072] [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: 12/12/2024]
Abstract
PURPOSE OF REVIEW Systemic lupus erythematosus (SLE) is a severe autoimmune/inflammatory disease. Patients with juvenile disease-onset and those of non-European ancestry are most severely affected. While the exact pathophysiology remains unknown, common and rare gene variants in the context of environmental exposure and epigenetic alterations are involved. This manuscript summarizes the current understanding of genetic and epigenetic contributors to SLE risk, manifestations and outcomes. RECENT FINDINGS Though SLE is a mechanistically complex disease, we are beginning to understand the impact of rare and common gene variants on disease expression and associated outcomes. Recent trans -ancestral and multigenerational studies suggest that differential genetic and environmental impacts shape phenotypic variability between age-groups and ancestries. High genetic burden associates with young age at disease-onset, organ involvement, and severity. Additional epigenetic impact contributes to disease-onset and severity, including SLE-phenotypes caused by rare single gene variants. Studies aiming to identify predictors of organ involvement and disease outcomes promise future patient stratification towards individualized treatment and care. SUMMARY An improved understanding of genetic variation and epigenetic marks explain phenotypic differences between age-groups and ancestries, promising their future exploitation for diagnostic, prognostic and therapeutic considerations.
Collapse
Affiliation(s)
- Amandine Charras
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Linda T. Hiraki
- Genetics & Genome Biology, Research Institute, and Division of Rheumatology, The Hospital for Sick Children, & Division of Rheumatology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Laura Lewandowski
- National Institute of Arthritis and Musculoskeletal and Skin diseases, NIH, Bethesda, Maryland, USA
| | - Christian M. Hedrich
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- Department of Rheumatology, Alder Hey Children's NHS Foundation Trust, Liverpool, UK
| |
Collapse
|
12
|
Williams SG, Sim S, Wolin SL. RNA sensing at the crossroads of autoimmunity and autoinflammation. RNA (NEW YORK, N.Y.) 2025; 31:369-381. [PMID: 39779213 PMCID: PMC11874990 DOI: 10.1261/rna.080304.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 12/31/2024] [Indexed: 01/11/2025]
Abstract
Immune-mediated diseases are common in humans. The immune system is a complex host defense system that evolved to protect us from pathogens, but also plays an important role in homeostatic processes, removing dead or senescent cells, and participating in tumor surveillance. The human immune system has two arms: the older innate immune system and the newer adaptive immune system. Sensing of foreign RNA is critical to the innate immune system's ability to recognize pathogens, especially viral infections. However, RNA sensors are also strongly implicated in autoimmune and autoinflammatory diseases, highlighting the importance of balancing pathogen recognition with tolerance to host RNAs that can resemble their viral counterparts. We describe how RNA sensors bind their ligands, how this binding is coupled to upregulation of type I interferon-stimulated genes, and the ways in which mutations in RNA sensors and genes that play important roles in RNA homeostasis have been linked to autoimmune and autoinflammatory diseases.
Collapse
Affiliation(s)
- Sandra G Williams
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
- RNA Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, USA
| | - Soyeong Sim
- RNA Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, USA
| | - Sandra L Wolin
- RNA Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, USA
| |
Collapse
|
13
|
Al Zayer L, Al Zayer M, Alrujaib A, Buhasan A, Jadah RHSH. Delayed Diagnosis of Aicardi-Goutières Syndrome in a 10-Year-Old Female Child With TREX1 Mutation: A Case Report. Cureus 2025; 17:e79730. [PMID: 40161088 PMCID: PMC11953621 DOI: 10.7759/cureus.79730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2025] [Indexed: 04/02/2025] Open
Abstract
Aicardi-Goutières syndrome (AGS) is a rare hereditary autoinflammatory disease of subacute encephalopathy, characterized by a wide range of neurological and extra-neurological manifestations, primarily affecting the brain and skin. Key features include increased expression of interferon-stimulated genes (ISGs), acquired microcephaly, dystonia, spasticity, chilblains, and panniculitis. Radiological findings include cerebral calcifications, leukodystrophy, cerebral atrophy, and cerebrospinal fluid abnormalities such as chronic lymphocytosis and elevated interferon-alpha (INF-α) levels. Seven pathogenic genes have been identified in association with AGS. The management of AGS is primarily supportive, as there is currently no definitive cure for the condition. The primary goals are to address symptoms, improve quality of life, and prevent complications. The aim of this study was to emphasize the importance of early diagnosis of this rare genetic condition, as timely identification enables prompt intervention and management. Early diagnosis improves clinical outcomes, enhances the quality of life for affected individuals, and provides valuable guidance for family planning and genetic counseling.
Collapse
Affiliation(s)
- Leen Al Zayer
- School of Medicine, Royal College of Surgeons in Ireland, Busaiteen, BHR
| | | | | | - Asal Buhasan
- School of Medicine, Royal College of Surgeons in Ireland, Busaiteen, BHR
| | | |
Collapse
|
14
|
Khalilian S, Fathi M, Miryounesi M, Ghafouri-Fard S. An overview of genetic mutations in Aicardi-Goutières syndrome in Iranian population. Neurol Sci 2025; 46:999-1007. [PMID: 39470906 DOI: 10.1007/s10072-024-07824-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: 05/05/2024] [Accepted: 10/15/2024] [Indexed: 11/01/2024]
Abstract
Aicardi-Goutières syndrome (AGS) is a rare neurodevelopmental disorder that can be misdiagnosed with infectious disorders. Molecular genetics tools and subsequent counseling have an important role in the estimation of recurrence risk and prevention of additional cases in the family. The current study provides an overview of genetic mutations in AGS in Iranian population. In a time period of 3 years, we assessed nine AGS cases and identified the underlying mutations using whole exome sequencing. Mutations were located in TREX1, IFIH1, RNASEH2B, RNASEH2A and SAMHD1 genes and inherited in either autosomal dominant or autosomal recessive manner. Since both modes of inheritance have been previously reported for AGS, appropriate genetic counseling is needed for estimation of recurrence risk in families.
Collapse
Affiliation(s)
- Sheyda Khalilian
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohadeseh Fathi
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Miryounesi
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Ouhenach M, Nada A, Lyahyai J, Sefiani A. The c.529G>A (p.Ala177Thr) RNASEH2B Gene Pathogenic Variant as a First-Line Genetic Test for Aicardi-Goutières Syndrome: A Case Series of Four Moroccan Families. Am J Med Genet A 2025:e63997. [PMID: 39890436 DOI: 10.1002/ajmg.a.63997] [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: 08/28/2024] [Revised: 11/26/2024] [Accepted: 01/07/2025] [Indexed: 02/03/2025]
Abstract
Aicardi-Goutières syndrome (AGS) is a hereditary encephalopathy characterized by marked clinical variability, mainly cerebral calcifications, cerebral atrophy, and leukodystrophy. The clinical diagnosis is difficult and can lead to high mortality. To date, nine genes are implicated, including RNASEH2A, RNASEH2B, RNASEH2C, TREX1, SAMHD1, ADAR1, IFIH1, LSM11, and RNU7-1. However, the p.A177T (c.529G>A) RNASEH2B gene mutation was described as the most recurrent mutation in several populations. Overall, there is a lack of research data on AGS in Morocco. Seven Moroccan patients from four families were referred for evaluation of Aicardi-Goutières syndrome (AGS). The first patient, a 1.5-year-old boy with leukodystrophy, underwent exome sequencing. The remaining six patients (a 2.5-year-old boy, three sisters aged 14, 10, and 2, and two sisters aged 5 and 3, along with another 2.5-year-old boy) were tested for the recurrent p.A177T (c.529G>A) RNASEH2B gene mutation using polymerase chain reaction and Sanger sequencing. Of the seven patients, five (two unrelated and three siblings) were homozygous for this pathogenic variant. Symptoms ranged from isolated spasticity with brain calcification to typical encephalopathy, with an average onset age of 1.5 years. Clinical variability was observed within one family. These findings demonstrate the phenotypic diversity of AGS and indicate that the first step of the diagnostic strategy should be genetic testing for the p.A177T (c.529G>A) RNASEH2B recurrent mutation.
Collapse
Affiliation(s)
- Mouna Ouhenach
- Department of Medical Genetics, National Institute of Health, Rabat, Morocco
| | - Amllal Nada
- Department of Medical Genetics, National Institute of Health, Rabat, Morocco
- Research Team in Genomics and Molecular Epidemiology of Genetic Diseases, GENOPATH Center, Faculty of Medicine and Pharmacy, University Mohammed V, Rabat, Morocco
| | - Jaber Lyahyai
- Department of Medical Genetics, National Institute of Health, Rabat, Morocco
- Research Team in Genomics and Molecular Epidemiology of Genetic Diseases, GENOPATH Center, Faculty of Medicine and Pharmacy, University Mohammed V, Rabat, Morocco
| | - Abdelaziz Sefiani
- Department of Medical Genetics, National Institute of Health, Rabat, Morocco
- Research Team in Genomics and Molecular Epidemiology of Genetic Diseases, GENOPATH Center, Faculty of Medicine and Pharmacy, University Mohammed V, Rabat, Morocco
| |
Collapse
|
16
|
Boiu S, Paschalidis N, Sentis G, Manolakou T, Nezos A, Gialitakis M, Grigoriou M, Atsali E, Giorgi M, Ntinopoulos A, Mavragani C, Makrythanasis P, Boumpas DT, Banos A. Increased interferon I signaling, DNA damage response and evidence of T-cell exhaustion in a patient with combined interferonopathy (Aicardi-Goutières Syndrome, AGS) and cohesinopathy (Cornelia de Lange Syndrome, CdLS). Pediatr Rheumatol Online J 2025; 23:11. [PMID: 39871364 PMCID: PMC11770959 DOI: 10.1186/s12969-024-01050-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 12/10/2024] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND Type I interferonopathies including Aicardi-Goutiéres Syndrome (AGS) represent a heterogeneous group of clinical phenotypes. Herein, we present a Case with combined AGS and Cornelia de Lange Syndrome (CdLS)-a cohesinopathy-with comprehensive analysis of the immune and genomic abnormalities. CASE AND METHODS A 20-year old man presented with chilblain lesions and resorption of distal phalanges of fingers and toes, somatic and psychomotor retardation, microcephaly, synophrys, hearing losing and other aberrancies consistent with the phenotype of CdLS. We used whole exome sequencing to genetically map the associated mutations and performed transcriptome profiling and enrichment analysis in CD14+ monocytes of the patient and immune phenotyping by mass cytometry (CyToF), comparing to healthy individuals and lupus patients as disease controls. DNA damage response was assayed by confocal microscopy in the peripheral blood of this patient. RESULTS Next generation exome sequencing confirmed a homozygous SAMHD1 gene mutation and a hemizygous non-synonymous mutation on SMC1A gene, responsible for the AGS and CdLS, respectively. Transcriptome profiling of CD14+ monocytes of the patient showed enrichment of type I IFN signaling and enhanced DNA damage response pathway. Broad immune phenotype of the peripheral blood of the patient revealed absence of activated T cell populations, increased frequency of NK cells and plasmablasts and enhanced granulocytic lineage. Further analysis suggested activation of the ATM branch of DNA damage response and increased apoptosis in the periphery of the patient. CONCLUSIONS A rare case of a patient bearing two genetic lesions (responsible for AGS/CdLS syndromes) exhibits distinctive features of genomic damage and interferon responses. Immune phenotype revealed granulocytic skewing and absence of activated T cells compatible with chronic antigenic stimulation and/or homing of these cells at sites of inflammation.
Collapse
Affiliation(s)
- Sorina Boiu
- Third Department of Pediatrics, Pediatric Rheumatology Unit, National and Kapodistrian University of Athens, 'Attikon' General University Hospital, Athens, Greece
- Department for Community Health, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
- Department of Pediatrics, Fribourg Hospital, Fribourg, Switzerland
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, INSERM UMR 1163, Paris, France
| | | | - George Sentis
- Laboratory of Autoimmunity and Inflammation, Center for Clinical, Biomedical Research Foundation, Experimental Surgery and Translational Research, Academy of Athens, Athens, Greece
| | - Theodora Manolakou
- Laboratory of Autoimmunity and Inflammation, Center for Clinical, Biomedical Research Foundation, Experimental Surgery and Translational Research, Academy of Athens, Athens, Greece
| | - Andrianos Nezos
- Department of Physiology, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Manolis Gialitakis
- Laboratory of Autoimmunity and Inflammation, Center for Clinical, Biomedical Research Foundation, Experimental Surgery and Translational Research, Academy of Athens, Athens, Greece
| | - Maria Grigoriou
- Laboratory of Autoimmunity and Inflammation, Center for Clinical, Biomedical Research Foundation, Experimental Surgery and Translational Research, Academy of Athens, Athens, Greece
| | - Erato Atsali
- Third Department of Pediatrics, Pediatric Rheumatology Unit, National and Kapodistrian University of Athens, 'Attikon' General University Hospital, Athens, Greece
| | - Melpomeni Giorgi
- Third Department of Pediatrics, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Argirios Ntinopoulos
- Third Department of Pediatrics, Pediatric Neurology Unit, National and Kapodistrian University of Athens, 'Attikon' General University Hospital, Athens, Greece
| | - Clio Mavragani
- Department of Physiology, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Periklis Makrythanasis
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
- Laboratory of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Department of Genetic Medicine and Development, Medical School, University of Geneva, Geneva, Switzerland
| | - Dimitrios T Boumpas
- Laboratory of Autoimmunity and Inflammation, Center for Clinical, Biomedical Research Foundation, Experimental Surgery and Translational Research, Academy of Athens, Athens, Greece
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Aggelos Banos
- Laboratory of Autoimmunity and Inflammation, Center for Clinical, Biomedical Research Foundation, Experimental Surgery and Translational Research, Academy of Athens, Athens, Greece.
| |
Collapse
|
17
|
Zhang Q, Walkley CR. Mouse models for understanding physiological functions of ADARs. Methods Enzymol 2025; 710:153-185. [PMID: 39870443 DOI: 10.1016/bs.mie.2024.11.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
Adenosine-to-inosine (A-to-I) editing, is a highly prevalent posttranscriptional modification of RNA, mediated by the adenosine deaminases acting on RNA (ADAR) proteins. Mammalian transcriptomes contain tens of thousands to millions of A-to-I editing events. Mutations in ADAR can result in rare autoinflammatory disorders such as Aicardi-Goutières syndrome (AGS) through to irreversible conditions such as motor neuron disease, amyotrophic lateral sclerosis (ALS). Mouse models have played an important role in our current understanding of the physiology of ADAR proteins. With the advancement of genetic engineering technologies, a number of new mouse models have been recently generated, each providing additional insight into ADAR function. This review highlights both past and current mouse models, exploring the methodologies used in their generation, their respective discoveries, and the significance of these findings in relation to human ADAR physiology.
Collapse
Affiliation(s)
- Qinyi Zhang
- St.Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; Department of Medicine, St. Vincent's Hospital, Melbourne Medical School, University of Melbourne, Fitzroy, Victoria, Australia; Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Carl R Walkley
- St.Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; Department of Medicine, St. Vincent's Hospital, Melbourne Medical School, University of Melbourne, Fitzroy, Victoria, Australia; Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia; Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
18
|
Goldbach-Mansky R, Alehashemi S, de Jesus AA. Emerging concepts and treatments in autoinflammatory interferonopathies and monogenic systemic lupus erythematosus. Nat Rev Rheumatol 2025; 21:22-45. [PMID: 39623155 DOI: 10.1038/s41584-024-01184-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2024] [Indexed: 12/22/2024]
Abstract
Over the past two decades, the number of genetically defined autoinflammatory interferonopathies has steadily increased. Aicardi-Goutières syndrome and proteasome-associated autoinflammatory syndromes (PRAAS, also known as CANDLE) are caused by genetic defects that impair homeostatic intracellular nucleic acid and protein processing respectively. Research into these genetic defects revealed intracellular sensors that activate type I interferon production. In SAVI and COPA syndrome, genetic defects that cause chronic activation of the dinucleotide sensor stimulator of interferon genes (STING) share features of lung inflammation and fibrosis; and selected mutations that amplify interferon-α/β receptor signalling cause central nervous system manifestations resembling Aicardi-Goutières syndrome. Research into the monogenic causes of childhood-onset systemic lupus erythematosus (SLE) demonstrates the pathogenic role of autoantibodies to particle-bound extracellular nucleic acids that distinguishes monogenic SLE from the autoinflammatory interferonopathies. This Review introduces a classification for autoinflammatory interferonopathies and discusses the divergent and shared pathomechanisms of interferon production and signalling in these diseases. Early success with drugs that block type I interferon signalling, new insights into the roles of cytoplasmic DNA or RNA sensors, pathways in type I interferon production and organ-specific pathology of the autoinflammatory interferonopathies and monogenic SLE, reveal novel drug targets that could personalize treatment approaches.
Collapse
Affiliation(s)
- Raphaela Goldbach-Mansky
- Translational Autoinflammatory Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Sara Alehashemi
- Translational Autoinflammatory Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Adriana A de Jesus
- Translational Autoinflammatory Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
19
|
Elitt CM, Volpe JJ. Degenerative Disorders of the Newborn. VOLPE'S NEUROLOGY OF THE NEWBORN 2025:967-1007.e17. [DOI: 10.1016/b978-0-443-10513-5.00033-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
20
|
Tian Y, Li N, Li Q, Gao N. Structural insight into Okazaki fragment maturation mediated by PCNA-bound FEN1 and RNaseH2. EMBO J 2025; 44:484-504. [PMID: 39578540 PMCID: PMC11731006 DOI: 10.1038/s44318-024-00296-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: 04/24/2024] [Revised: 10/27/2024] [Accepted: 10/28/2024] [Indexed: 11/24/2024] Open
Abstract
PCNA is a master coordinator of many DNA-metabolic events. During DNA replication, the maturation of Okazaki fragments involves at least four DNA enzymes, all of which contain PCNA-interacting motifs. However, the temporal relationships and functional modulations between these PCNA-binding proteins are unclear. Here, we developed a strategy to purify endogenous PCNA-containing complexes from native chromatin, and characterized their structures using cryo-EM. Two structurally resolved classes (PCNA-FEN1 and PCNA-FEN1-RNaseH2 complexes) have captured a series of 3D snapshots for the primer-removal steps of Okazaki fragment maturation. These structures show that product release from FEN1 is a rate-liming step. Furthermore, both FEN1 and RNaseH2 undergo continuous conformational changes on PCNA that result in constant fluctuations in the bending angle of substrate DNA at the nick site, implying that these enzymes could regulate each other through conformational modulation of the bound DNA. The structures of the PCNA-FEN1-RNaseH2 complex confirm the toolbelt function of PCNA and suggests a potential unrecognized role of RNaseH2, as a dsDNA binding protein, in promoting the 5'-flap cleaving activity of FEN1.
Collapse
Affiliation(s)
- Yuhui Tian
- State Key Laboratory of Membrane Biology, Peking-Tsinghua Joint Center for Life Sciences, School of Life Sciences, Peking University, Beijing, China
| | - Ningning Li
- State Key Laboratory of Membrane Biology, Peking-Tsinghua Joint Center for Life Sciences, School of Life Sciences, Peking University, Beijing, China
- Changping Laboratory, Beijing, China
| | - Qing Li
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Ning Gao
- State Key Laboratory of Membrane Biology, Peking-Tsinghua Joint Center for Life Sciences, School of Life Sciences, Peking University, Beijing, China.
- Changping Laboratory, Beijing, China.
- National Biomedical Imaging Center, Peking University, Beijing, China.
- Beijing Advanced Center of RNA Biology (BEACON), Peking University, Beijing, China.
| |
Collapse
|
21
|
Stratigi K, Siametis A, Garinis GA. Looping forward: exploring R-loop processing and therapeutic potential. FEBS Lett 2025; 599:244-266. [PMID: 38844597 PMCID: PMC11771710 DOI: 10.1002/1873-3468.14947] [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: 02/07/2024] [Revised: 05/13/2024] [Accepted: 05/20/2024] [Indexed: 01/28/2025]
Abstract
Recently, there has been increasing interest in the complex relationship between transcription and genome stability, with specific attention directed toward the physiological significance of molecular structures known as R-loops. These structures arise when an RNA strand invades into the DNA duplex, and their formation is involved in a wide range of regulatory functions affecting gene expression, DNA repair processes or cell homeostasis. The persistent presence of R-loops, if not effectively removed, contributes to genome instability, underscoring the significance of the factors responsible for their resolution and modification. In this review, we provide a comprehensive overview of how R-loop processing can drive either a beneficial or a harmful outcome. Additionally, we explore the potential for manipulating such structures to devise rationalized therapeutic strategies targeting the aberrant accumulation of R-loops.
Collapse
Affiliation(s)
- Kalliopi Stratigi
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology‐HellasHeraklionCreteGreece
| | - Athanasios Siametis
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology‐HellasHeraklionCreteGreece
- Department of BiologyUniversity of CreteHeraklionCreteGreece
| | - George A. Garinis
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology‐HellasHeraklionCreteGreece
- Department of BiologyUniversity of CreteHeraklionCreteGreece
| |
Collapse
|
22
|
Cheng L, Liu Z, Shen C, Xiong Y, Shin SY, Hwang Y, Yang S, Chen Z, Zhang X. A Wonderful Journey: The Diverse Roles of Adenosine Deaminase Action on RNA 1 (ADAR1) in Central Nervous System Diseases. CNS Neurosci Ther 2025; 31:e70208. [PMID: 39753993 PMCID: PMC11702419 DOI: 10.1111/cns.70208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/03/2024] [Accepted: 12/20/2024] [Indexed: 01/14/2025] Open
Abstract
BACKGROUND Adenosine deaminase action on RNA 1 (ADAR1) can convert the adenosine in double-stranded RNA (dsRNA) molecules into inosine in a process known as A-to-I RNA editing. ADAR1 regulates gene expression output by interacting with RNA and other proteins; plays important roles in development, including growth; and is linked to innate immunity, tumors, and central nervous system (CNS) diseases. RESULTS In recent years, the role of ADAR1 in tumors has been widely discussed, but its role in CNS diseases has not been reviewed. It is worth noting that recent studies have shown ADAR1 has great potential in the treatment of neurodegenerative diseases, but the mechanisms are still unclear. Therefore, it is necessary to elaborate on the role of ADAR1 in CNS diseases. CONCLUSIONS Here, we focus on the effects and mechanisms of ADAR1 on CNS diseases such as Aicardi-AicardiGoutières syndrome, Alzheimer's disease, Parkinson's disease, glioblastoma, epilepsy, amyotrophic lateral sclerosis, and autism. We also evaluate the impact of ADAR1-based treatment strategies on these diseases, with a particular focus on the development and treatment strategies of new technologies such as microRNAs, nanotechnology, gene editing, and stem cell therapy. We hope to provide new directions and insights for the future development of ADAR1 gene editing technology in brain science and the treatment of CNS diseases.
Collapse
Affiliation(s)
- Lin Cheng
- Department of NeurologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Ziying Liu
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
- Department of PathologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| | - Chunxiao Shen
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
- Department of PathologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| | - Yinyi Xiong
- Department of RehabilitationAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| | - Sang Yol Shin
- Department of Emergency Medical TechnologyWonkwang University College of MedicineIksanJeonbuk‐doRepublic of Korea
| | - Yong Hwang
- Department of Emergency MedicineWonkwang University College of MedicineIksanJeonbuk‐doRepublic of Korea
| | - Seung‐Bum Yang
- Department of ParamedicineWonkwang Health Science UniversityIksanJeonbuk‐doRepublic of Korea
| | - Zhiying Chen
- Department of NeurologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Xiaorong Zhang
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
- Department of PathologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| |
Collapse
|
23
|
Gabaldon-Albero A, Martin-Grau C, Marti-Masanet M, Lopez-Jimenez A, Llorens R, Beseler-Soto B, Martin-Zamora S, Lopez B, Calvo I, Hernandez-Muela S, Rosello M, Orellana C, Martinez F. Aicardi-Goutières syndrome type 6: report of ADAR variant and clinical outcome after ruxolitinib treatment in the neonatal period. Pediatr Rheumatol Online J 2024; 22:110. [PMID: 39732715 PMCID: PMC11682636 DOI: 10.1186/s12969-024-01036-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 11/14/2024] [Indexed: 12/30/2024] Open
Abstract
BACKGROUND Aicardi-Goutières Syndrome is a monogenic type 1 interferonopathy with infantile onset, characterized by a variable degree of neurological damage. Approximately 7% of Aicardi-Goutières Syndrome cases are caused by pathogenic variants in the ADAR gene and are classified as Aicardi-Goutières Syndrome type 6. Here, we present a new homozygous pathogenic variant in the ADAR gene. Currently, Janus Kinase inhibitors have been proposed to treat selected interferonopathies such as Aicardi-Goutières Syndrome, although limited information is available on its use and results in the neonatal presentation of this disease. CASE PRESENTATION We present two siblings, a male neonate with congenital petechial rash, severe thrombopenia and generalized hypotonia and his deceased sister who had normal development until 5 months of age, when she suffered acute encephalopathy. We describe the clinical course, complementary examinations and follow-up with early treatment of the newborn with ruxolitinib. The homozygous variant c.2908G > A (p.Ala970Thr) in the ADAR gene was found in both siblings, parents were heterozygous carriers. CONCLUSIONS The homozygous variant c.2908G > A (p.Ala970Thr) in the ADAR gene causes Aicardi-Goutières Syndrome type 6. Intrafamilial phenotypic spectrum of the disease varies among individuals with the same pathogenic variant. Early initiation of ruxolitinib improved systemic signs but did not prevent the progression of neurological disease.
Collapse
Affiliation(s)
- Alba Gabaldon-Albero
- Pediatric Neurology Unit, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Translational Genetics Research Group, La Fe Health Research Institute (IIS La Fe), Avenida Fernando Abril Martorell nº 106 Tower A, 7th Floor, Valencia, Spain
| | - Carla Martin-Grau
- Translational Genetics Research Group, La Fe Health Research Institute (IIS La Fe), Avenida Fernando Abril Martorell nº 106 Tower A, 7th Floor, Valencia, Spain
- Genetics Unit, Hospital Universitario y Politécnico La Fe, Avenida Fernando Abril Martorell nº 106 Tower A, 4th Floor, Valencia, Spain
| | - Miguel Marti-Masanet
- Pediatric Rheumatology Unit, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | | | - Roberto Llorens
- Pediatric Radiology, Unit Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Beatriz Beseler-Soto
- Pediatric Neurology Unit, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | | | - Berta Lopez
- Pediatric Rheumatology Unit, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Inmaculada Calvo
- Pediatric Rheumatology Unit, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Sara Hernandez-Muela
- Pediatric Neurology Unit, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Monica Rosello
- Translational Genetics Research Group, La Fe Health Research Institute (IIS La Fe), Avenida Fernando Abril Martorell nº 106 Tower A, 7th Floor, Valencia, Spain
- Genetics Unit, Hospital Universitario y Politécnico La Fe, Avenida Fernando Abril Martorell nº 106 Tower A, 4th Floor, Valencia, Spain
| | - Carmen Orellana
- Translational Genetics Research Group, La Fe Health Research Institute (IIS La Fe), Avenida Fernando Abril Martorell nº 106 Tower A, 7th Floor, Valencia, Spain
- Genetics Unit, Hospital Universitario y Politécnico La Fe, Avenida Fernando Abril Martorell nº 106 Tower A, 4th Floor, Valencia, Spain
| | - Francisco Martinez
- Translational Genetics Research Group, La Fe Health Research Institute (IIS La Fe), Avenida Fernando Abril Martorell nº 106 Tower A, 7th Floor, Valencia, Spain.
- Genetics Unit, Hospital Universitario y Politécnico La Fe, Avenida Fernando Abril Martorell nº 106 Tower A, 4th Floor, Valencia, Spain.
| |
Collapse
|
24
|
Politano D, Tonduti D, Battini R, Fazzi E, Orcesi S. Exploring emerging JAK inhibitors in the treatment of Aicardi-Goutières syndrome. Expert Opin Emerg Drugs 2024:1-19. [PMID: 39704072 DOI: 10.1080/14728214.2024.2445508] [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: 07/15/2024] [Revised: 12/18/2024] [Accepted: 12/18/2024] [Indexed: 12/21/2024]
Abstract
INTRODUCTION Aicardi-Goutières syndrome (AGS) is a genetically heterogeneous monogenic autoinflammatory disorder classified as an 'interferonopathy'. Nine genes have been implicated in AGS, encoding proteins involved in nucleic acid clearance, repair, sensing, or histone pre-mRNA processing. Dysregulation in these pathways leads to excessive type I interferon production, the primary driver of the disease. AGS typically presents with early-life neurological regression, followed by stabilization with varying degrees of neurological impairment and common extra-neurological features, such as chilblains. Advances in understanding AGS pathogenesis have enabled the development of new therapies, with JAK inhibitors emerging as the most studied option for reducing interferon-mediated effects. AREAS COVERED This review discusses the clinical features, genetic basis, and molecular pathways of AGS while tracing the evolution of its therapeutic strategies. Particular emphasis is placed on JAK inhibitors, which target proteins activated by type I interferons, providing a novel direction in treatment. EXPERT OPINION Inhibitors effectively reduce extra-neurological symptoms in AGS, though their impact on neurological outcomes remains unclear. The unknown natural history of AGS limits treatment evaluation. Despite growing insights, key aspects of pathogenesis and treatment optimization - including timing, administration, and long-term effects - remain unresolved, highlighting the need for further research.
Collapse
Affiliation(s)
- Davide Politano
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Davide Tonduti
- Unit of Pediatric Neurology, COALA Center for Diagnosis and Treatment of Leukodystrophies, V. Buzzi Children's Hospital, Milan, Italy
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Roberta Battini
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Elisa Fazzi
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Unit of Child Neurology and Psychiatry, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Simona Orcesi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
25
|
Luca D, Kato H. Mouse models of type I interferonopathies. Hum Mol Genet 2024:ddae187. [PMID: 39680957 DOI: 10.1093/hmg/ddae187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 12/10/2024] [Indexed: 12/18/2024] Open
Abstract
Type I interferonopathies are severe monogenic diseases caused by mutations that result in chronically upregulated production of type I interferon. They present with a broad variety of symptoms, the mechanisms of which are being extensively studied. Mouse models of type I interferonopathies are an important resource for this purpose, and in this context, we review several key molecular and phenotypic findings that are advancing our understanding of the respective diseases. We focus on genotypes related to nucleic acid metabolism, sensing by cytosolic receptors and downstream signalling.
Collapse
Affiliation(s)
- Domnica Luca
- Institute of Cardiovascular Immunology, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, Bonn 53127, Germany
| | - Hiroki Kato
- Institute of Cardiovascular Immunology, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, Bonn 53127, Germany
| |
Collapse
|
26
|
Keshavan N, Mhaldien L, Gilmour K, Rahman S. Interferon Stimulated Gene Expression Is a Biomarker for Primary Mitochondrial Disease. Ann Neurol 2024; 96:1185-1200. [PMID: 39320038 DOI: 10.1002/ana.27081] [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: 06/24/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 09/26/2024]
Abstract
OBJECTIVE Mitochondria are implicated in regulation of the innate immune response. We hypothesized that abnormalities in interferon signaling may contribute to pathophysiology in patients with primary mitochondrial disease (PMD). METHODS Expression of interferon stimulated genes (ISGs) was measured by real-time polymerase chain reaction (PCR) in whole blood samples from a cohort of patients with PMD. RESULTS Upregulated ISG expression was observed in a high proportion (41/55, 75%) of patients with PMD on at least 1 occasion, most frequently IFI27 upregulation, seen in 50% of the samples. Some patients had extremely high IFI27 levels, similar to those seen in patients with primary interferonopathies. A statistically significant correlation was observed between elevated IFI27 gene expression and PMD, but not between IFI27 and secondary mitochondrial dysfunction, suggesting that ISG upregulation is a biomarker of PMD. In some patients with PMD, ISG abnormalities persisted on repeat measurement over several years, indicative of ongoing chronic inflammation. Subgroup analyses suggested common ISG signatures in patients with similar mitochondrial disease mechanisms and positive correlations with disease severity among patients with identical genetic diagnoses. INTERPRETATION Dysregulated interferon signaling is frequently seen in patients with PMD suggesting that interferon dysregulation is a contributor to pathophysiology. This may indicate a role for repurposing of immunomodulatory therapies for the treatment of PMDs by targeting interferon signaling. ANN NEUROL 2024;96:1185-1200.
Collapse
Affiliation(s)
- Nandaki Keshavan
- Metabolic Unit, Great Ormond Street Hospital, London, UK
- Mitochondrial Research Group, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Lana Mhaldien
- Department of Immunology, Camelia Botnar Laboratory, Great Ormond Street Hospital, London, UK
| | - Kimberly Gilmour
- Department of Immunology, Camelia Botnar Laboratory, Great Ormond Street Hospital, London, UK
| | - Shamima Rahman
- Metabolic Unit, Great Ormond Street Hospital, London, UK
- Mitochondrial Research Group, UCL Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
27
|
Rodríguez RD, Alarcón-Riquelme ME. Exploring the contribution of genetics on the clinical manifestations of systemic lupus erythematosus. Best Pract Res Clin Rheumatol 2024; 38:101971. [PMID: 39013664 DOI: 10.1016/j.berh.2024.101971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/18/2024]
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease characterized by diverse clinical manifestations affecting multiple organs and systems. The understanding of genetic factors underlying the various manifestations of SLE has evolved considerably in recent years. This review provides an overview of the genetic implications in some of the most prevalent manifestations of SLE, including renal involvement, neuropsychiatric, cutaneous, constitutional, musculoskeletal, and cardiovascular manifestations. We discuss the current state of knowledge regarding the genetic basis of these manifestations, highlighting key genetic variants and pathways implicated in their pathogenesis. Additionally, we explore the clinical implications of genetic findings, including their potential role in risk stratification, prognosis, and personalized treatment approaches for patients with SLE. Through a comprehensive examination of the genetic landscape of SLE manifestations, this review aims to provide insights into the underlying mechanisms driving disease heterogeneity and inform future research directions in this field.
Collapse
Affiliation(s)
- Ruth D Rodríguez
- Center for Genomics and Oncological Research (GENyO). Pfizer/ University of Granada/ Andalusian Government, Spain
| | - Marta E Alarcón-Riquelme
- Center for Genomics and Oncological Research (GENyO). Pfizer/ University of Granada/ Andalusian Government, Spain; Institute for Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
28
|
Guillet S, Lazarov T, Jordan N, Boisson B, Tello M, Craddock B, Zhou T, Nishi C, Bareja R, Yang H, Rieux-Laucat F, Fregel Lorenzo RI, Dyall SD, Isenberg D, D'Cruz D, Lachmann N, Elemento O, Viale A, Socci ND, Abel L, Nagata S, Huse M, Miller WT, Casanova JL, Geissmann F. ACK1 and BRK non-receptor tyrosine kinase deficiencies are associated with familial systemic lupus and involved in efferocytosis. eLife 2024; 13:RP96085. [PMID: 39570652 PMCID: PMC11581429 DOI: 10.7554/elife.96085] [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: 11/22/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease, the pathophysiology and genetic basis of which are incompletely understood. Using a forward genetic screen in multiplex families with SLE, we identified an association between SLE and compound heterozygous deleterious variants in the non-receptor tyrosine kinases (NRTKs) ACK1 and BRK. Experimental blockade of ACK1 or BRK increased circulating autoantibodies in vivo in mice and exacerbated glomerular IgG deposits in an SLE mouse model. Mechanistically, NRTKs regulate activation, migration, and proliferation of immune cells. We found that the patients' ACK1 and BRK variants impair efferocytosis, the MERTK-mediated anti-inflammatory response to apoptotic cells, in human induced pluripotent stem cell (hiPSC)-derived macrophages, which may contribute to SLE pathogenesis. Overall, our data suggest that ACK1 and BRK deficiencies are associated with human SLE and impair efferocytosis in macrophages.
Collapse
Affiliation(s)
- Stephanie Guillet
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
- Ecole doctorale Bio Sorbonne Paris Cité, Université Paris Descartes-Sorbonne Paris CitéParisFrance
| | - Tomi Lazarov
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical SciencesNew YorkUnited States
| | - Natasha Jordan
- Centre for Molecular and Cellular Biology of Inflammation (CMCBI), King’s College London and Louise Coote Lupus Unit, Guy’s and Thomas’ HospitalsLondonUnited Kingdom
| | - Bertrand Boisson
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller UniversityNew YorkUnited States
- University of Paris Cité, Imagine InstituteParisFrance
| | - Maria Tello
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Barbara Craddock
- SKI Stem Cell Research Core, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Ting Zhou
- SKI Stem Cell Research Core, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Chihiro Nishi
- Laboratory of Biochemistry & Immunology, World Premier International Immunology Frontier Research Center, Osaka UniversityOsakaJapan
| | - Rohan Bareja
- Cary and Israel Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Meyer Cancer Center Weill Cornell Medical CollegeNew YorkUnited States
| | - Hairu Yang
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | | | | | - Sabrina D Dyall
- Department of Biosciences and Ocean Studies, Faculty of Science, University of MauritiusReduitMauritius
| | - David Isenberg
- Bioinformatics Core, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - David D'Cruz
- Centre for Molecular and Cellular Biology of Inflammation (CMCBI), King’s College London and Louise Coote Lupus Unit, Guy’s and Thomas’ HospitalsLondonUnited Kingdom
| | - Nico Lachmann
- Centre for Rheumatology, Division of Medicine, University College London, The Rayne BuildingLondonUnited Kingdom
| | - Olivier Elemento
- Cary and Israel Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Meyer Cancer Center Weill Cornell Medical CollegeNew YorkUnited States
| | - Agnes Viale
- Institute of Experimental Hematology, REBIRTH Cluster of Excellence, Hannover Medical SchoolHannoverGermany
| | - Nicholas D Socci
- Institute of Experimental Hematology, REBIRTH Cluster of Excellence, Hannover Medical SchoolHannoverGermany
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Laurent Abel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller UniversityNew YorkUnited States
- University of Paris Cité, Imagine InstituteParisFrance
| | - Shigekazu Nagata
- Laboratory of Biochemistry & Immunology, World Premier International Immunology Frontier Research Center, Osaka UniversityOsakaJapan
| | - Morgan Huse
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - W Todd Miller
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
- Department of Physiology and Biophysics, Stony Brook University School of MedicineStony BrookUnited States
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller UniversityNew YorkUnited States
- University of Paris Cité, Imagine InstituteParisFrance
- Howard Hughes Medical InstituteNew YorkUnited States
- Lab of Human Genetics of Infectious Diseases, INSERM, Necker Hospital for Sick ChildrenParisFrance
- Department of Pediatrics, Necker Hospital for Sick ChildrenParisFrance
| | - Frédéric Geissmann
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical SciencesNew YorkUnited States
- Centre for Molecular and Cellular Biology of Inflammation (CMCBI), King’s College London and Louise Coote Lupus Unit, Guy’s and Thomas’ HospitalsLondonUnited Kingdom
| |
Collapse
|
29
|
Crow YJ. CNS disease associated with enhanced type I interferon signalling. Lancet Neurol 2024; 23:1158-1168. [PMID: 39424561 PMCID: PMC7616788 DOI: 10.1016/s1474-4422(24)00263-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 10/21/2024]
Abstract
The ability to mount an interferon-mediated innate immune response is essential in protection against neurotropic viruses, but antiviral type I interferons also have neurotoxic potential. The production of type I interferons can be triggered by self-derived nucleic acids, and the brain can be susceptible to inappropriate upregulation of type I interferon signalling. Homoeostatic dysregulation of type I interferons has been implicated in rare inborn errors of immunity (referred to as type I interferonopathies) and more common neurodegenerative disorders (eg, Parkinson's disease, Alzheimer's disease, and amyotrophic lateral sclerosis). Recent developments include new insights into the pathogenesis of these disorders that involve dysregulated type I interferon signalling, as well as advances in their diagnosis and management. The role of type I interferons in brain cellular health suggests the future therapeutic potential of approaches that target these interferons and their signalling.
Collapse
Affiliation(s)
- Yanick J Crow
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK; Laboratory of Neurogenetics and Neuroinflammation, Imagine Institute, INSERM UMR1163, Paris, France.
| |
Collapse
|
30
|
Maxwell G, Souzeau E. Childhood glaucoma: Implications for genetic counselling. Clin Genet 2024; 106:545-563. [PMID: 39206700 DOI: 10.1111/cge.14603] [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: 05/09/2024] [Revised: 07/17/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024]
Abstract
Childhood glaucoma is a heterogeneous group of ocular disorders defined by an age of onset from birth to 18 years. These vision-threatening disorders require early diagnosis, timely treatment, and lifelong management to maintain vision and minimise irreversible blindness. The genetics of childhood glaucoma is complex with both phenotypic and genetic heterogeneity. The purpose of this review is to summarise the different types of childhood glaucoma and their genetic architecture to aid in the genetic counselling process with patients and their families. We provide an overview of associated syndromes and discuss implications for genetic counselling, including genetic testing strategies, cascade genetic testing, and reproductive options.
Collapse
Affiliation(s)
- Giorgina Maxwell
- Department of Ophthalmology, Flinders University, Adelaide, South Australia, Australia
| | - Emmanuelle Souzeau
- Department of Ophthalmology, Flinders University, Adelaide, South Australia, Australia
| |
Collapse
|
31
|
Arroyo-Andrés J, Arteche-López AR, González-Granado I, Postigo Llorente C. Pseudo-catatonia and Acral Degos-like Lesions: An Atypical Form of the Aicardi-Goutières Syndrome. ACTAS DERMO-SIFILIOGRAFICAS 2024; 115:1045-1046. [PMID: 38552783 DOI: 10.1016/j.ad.2023.04.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/27/2023] [Accepted: 04/19/2023] [Indexed: 05/27/2024] Open
Affiliation(s)
- J Arroyo-Andrés
- Department of Dermatology, 12 de Octubre Hospital, Institute i+12, CIBERONC, Medical School, University Complutense, Madrid, Spain.
| | - A R Arteche-López
- Department of Genetics, University 12 de Octubre Hospital, Madrid, Spain
| | - I González-Granado
- Pediatrics Unit, 12 de Octubre Hospital, Av Cordoba S/N 28041, Madrid, Spain
| | - C Postigo Llorente
- Department of Dermatology, 12 de Octubre Hospital, Institute i+12, CIBERONC, Medical School, University Complutense, Madrid, Spain
| |
Collapse
|
32
|
Arroyo-Andrés J, Arteche-López AR, González-Granado I, Postigo Llorente C. Pseudo-catatonia and Acral Degos-like Lesions: An Atypical Form of the Aicardi-Goutières Syndrome. ACTAS DERMO-SIFILIOGRAFICAS 2024; 115:T1045-T1046. [PMID: 39413913 DOI: 10.1016/j.ad.2024.10.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/27/2023] [Accepted: 04/19/2023] [Indexed: 10/18/2024] Open
Affiliation(s)
- J Arroyo-Andrés
- Department of Dermatology, 12 de Octubre Hospital, Institute i+12, CIBERONC, Medical School, University Complutense, Madrid, Spain.
| | - A R Arteche-López
- Department of Genetics, University 12 de Octubre Hospital, Madrid, Spain
| | - I González-Granado
- Pediatrics Unit, 12 de Octubre Hospital, Av Cordoba S/N 28041, Madrid, Spain
| | - C Postigo Llorente
- Department of Dermatology, 12 de Octubre Hospital, Institute i+12, CIBERONC, Medical School, University Complutense, Madrid, Spain
| |
Collapse
|
33
|
Cao AB, Devant P, Wang C, Sun M, Kennedy SN, Ruan J, Kagan JC. LPS binding caspase activation and recruitment domains (CARDs) are bipartite lipid binding modules. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.07.617105. [PMID: 39416091 PMCID: PMC11482759 DOI: 10.1101/2024.10.07.617105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Caspase-11 is an innate immune pattern recognition receptor (PRR) that detects cytosolic bacterial lipopolysaccharides (LPS) through its caspase activation and recruitment domain (CARD), triggering inflammatory cell death known as pyroptosis. Caspase-11 also detects eukaryotic (i.e. self) lipids. This observation raises the question of whether common or distinct mechanisms govern the interactions with self and nonself lipids. In this study, using biochemical, computational, and cell-based assays, we report that the caspase-11 CARD functions as a bipartite lipid-binding module. Distinct regions within the CARD bind to phosphate groups and long acyl chains of self and nonself lipids. Self-lipid binding capability is conserved across numerous caspase-11 homologs and orthologs. The symmetry in self and nonself lipid detection mechanisms enabled us to engineer an LPS-binding domain de novo, using an ancestral CARD-like domain present in the fish Amphilophus citrinellus. These findings offer critical insights into the molecular basis of LPS recognition by caspase-11 and highlight the fundamental and likely inseparable relationship between self and nonself discrimination.
Collapse
Affiliation(s)
- Anh B. Cao
- Division of Gastroenterology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Pascal Devant
- Division of Gastroenterology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Chengliang Wang
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Mengyu Sun
- Division of Gastroenterology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Stephanie N. Kennedy
- Division of Gastroenterology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Jianbin Ruan
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Jonathan C. Kagan
- Division of Gastroenterology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
34
|
Železnik M, Vesnaver TV, Neubauer D, Soltirovska-Šalamon A. Neurological Findings and a Brief Review of the Current Literature in a Severe Case of Aicardi-Goutières Syndrome Due to an IFIH1 Mutation. Neuropediatrics 2024; 55:337-340. [PMID: 38714209 DOI: 10.1055/a-2321-0597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/09/2024]
Abstract
Aicardi-Goutières syndrome (AGS) is a rare genetic early-onset progressive encephalopathy with variable clinical manifestations. The IFIH1 mutation has been confirmed to be responsible for type I interferon production and activation of the Janus kinase signaling pathway. We herein stress neurological observations and neuroimaging findings in a severe case report of an infant with AGS type 7 due to an IFIH1 mutation who was diagnosed in the first month of life. We also review neurological characteristics of IFIH1 mutations through recent literature.
Collapse
Affiliation(s)
- Mojca Železnik
- Division of Pediatrics, Department of Neonatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | | | - David Neubauer
- Division of Pediatrics, Adolescent and Developmental Neurology, Department of Child, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Aneta Soltirovska-Šalamon
- Division of Pediatrics, Department of Neonatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Department of Pediatrics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
35
|
Hofer MJ, Modesti N, Coufal NG, Wang Q, Sase S, Miner J, Vanderver A, Bennett ML. The prototypical interferonopathy: Aicardi-Goutières syndrome from bedside to bench. Immunol Rev 2024; 327:83-99. [PMID: 39473130 PMCID: PMC11672868 DOI: 10.1111/imr.13413] [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: 11/11/2024]
Abstract
Aicardi-Goutières syndrome (AGS) is a progressive genetic encephalopathy caused by pathogenic mutations in genes controlling cellular anti-viral responses and nucleic acid metabolism. The mutations initiate autoinflammatory processes in the brain and systemically that are triggered by chronic overproduction of type I interferon (IFN), including IFN-alpha. Emerging disease-directed therapies aim to dampen autoinflammation and block cellular responses to IFN production, creating an urgent and unmet need to understand better which cells, compartments, and mechanisms underlying disease pathogenesis. In this review, we highlight existing pre-clinical models of AGS and our current understanding of how causative genetic mutations promote disease in AGS, to promote new model development and a continued focus on improving and directing future therapies.
Collapse
Affiliation(s)
- Markus J. Hofer
- Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, NSW 2006, Australia; NHMRC Ideas Grant to MJH APP2001543
| | - Nicholson Modesti
- Division of Neurology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, 19104
| | - Nicole G. Coufal
- Department of Pediatrics, University of California, San Diego CA 92093, Rady Children’s Hospital, San Diego CA 92123. Sanford Consortium for Regenerative Medicine, San Diego CA 92037
| | - Qingde Wang
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213
| | - Sunetra Sase
- Division of Neurology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, 19104
| | - Jonathan Miner
- Departments of Medicine and Microbiology, RVCL Research Center, and Colton Center for Autoimmunity, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104
| | - Adeline Vanderver
- Division of Neurology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, 19104
| | - Mariko L Bennett
- Division of Neurology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, 19104
| |
Collapse
|
36
|
Ghodke-Puranik Y, Olferiev M, Crow MK. Systemic lupus erythematosus genetics: insights into pathogenesis and implications for therapy. Nat Rev Rheumatol 2024; 20:635-648. [PMID: 39232240 DOI: 10.1038/s41584-024-01152-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2024] [Indexed: 09/06/2024]
Abstract
Systemic lupus erythematosus (SLE) is a prime example of how the interplay between genetic and environmental factors can trigger systemic autoimmunity, particularly in young women. Although clinical disease can take years to manifest, risk is established by the unique genetic makeup of an individual. Genome-wide association studies have identified almost 200 SLE-associated risk loci, yet unravelling the functional effect of these loci remains a challenge. New analytic tools have enabled researchers to delve deeper, leveraging DNA sequencing and cell-specific and immune pathway analysis to elucidate the immunopathogenic mechanisms. Both common genetic variants and rare non-synonymous mutations can interact to increase SLE risk. Notably, variants strongly associated with SLE are often located in genome super-enhancers that regulate MHC class II gene expression. Additionally, the 3D conformations of DNA and RNA contribute to genome regulation and innate immune system activation. Improved therapies for SLE are urgently needed and current and future knowledge from genetic and genomic research should provide new tools to facilitate patient diagnosis, enhance the identification of therapeutic targets and optimize testing of agents.
Collapse
Affiliation(s)
- Yogita Ghodke-Puranik
- Mary Kirkland Center for Lupus Research, Hospital for Special Surgery and Weill Cornell Medicine, New York, NY, USA
| | - Mikhail Olferiev
- Mary Kirkland Center for Lupus Research, Hospital for Special Surgery and Weill Cornell Medicine, New York, NY, USA
| | - Mary K Crow
- Mary Kirkland Center for Lupus Research, Hospital for Special Surgery and Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
37
|
van Gemert F, Drakaki A, Lozano IM, de Groot D, Uiterkamp M, Proost N, Lieftink C, van de Ven M, Beijersbergen R, Jacobs H, te Riele H. ADARp150 counteracts whole genome duplication. Nucleic Acids Res 2024; 52:10370-10384. [PMID: 39189458 PMCID: PMC11417406 DOI: 10.1093/nar/gkae700] [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: 03/08/2024] [Revised: 07/10/2024] [Accepted: 08/07/2024] [Indexed: 08/28/2024] Open
Abstract
Impaired control of the G1/S checkpoint allows initiation of DNA replication under non-permissive conditions. Unscheduled S-phase entry is associated with DNA replication stress, demanding for other checkpoints or cellular pathways to maintain proliferation. Here, we uncovered a requirement for ADARp150 to sustain proliferation of G1/S-checkpoint-defective cells under growth-restricting conditions. Besides its well-established mRNA editing function in inversely oriented short interspersed nuclear elements (SINEs), we found ADARp150 to exert a critical function in mitosis. ADARp150 depletion resulted in tetraploidization, impeding cell proliferation in mitogen-deprived conditions. Mechanistically we show that ADAR1 depletion induced aberrant expression of Cyclin B3, which was causative for mitotic failure and whole-genome duplication. Finally, we find that also in vivo ADAR1-depletion-provoked tetraploidization hampers tumor outgrowth.
Collapse
Affiliation(s)
- Frank van Gemert
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Alexandra Drakaki
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Isabel Morales Lozano
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Daniël de Groot
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Maud Schoot Uiterkamp
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Natalie Proost
- Mouse Clinic for Cancer and Aging Research, Preclinical Intervention Unit, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Cor Lieftink
- Division of Molecular Carcinogenesis, NKI Robotics and Screening Center, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marieke van de Ven
- Mouse Clinic for Cancer and Aging Research, Preclinical Intervention Unit, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Roderick L Beijersbergen
- Division of Molecular Carcinogenesis, NKI Robotics and Screening Center, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Heinz Jacobs
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Hein te Riele
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
38
|
Shim A, Luan X, Zhou W, Crow YJ, Maciejowski J. Mutations in the non-catalytic polyproline motif destabilize TREX1 and amplify cGAS-STING signaling. Hum Mol Genet 2024; 33:1555-1566. [PMID: 38796715 PMCID: PMC11373327 DOI: 10.1093/hmg/ddae089] [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: 02/16/2024] [Revised: 04/25/2024] [Indexed: 05/28/2024] Open
Abstract
The cGAS-STING pathway detects cytosolic DNA and activates a signaling cascade that results in a type I interferon (IFN) response. The endoplasmic reticulum (ER)-associated exonuclease TREX1 suppresses cGAS-STING by eliminating DNA from the cytosol. Mutations that compromise TREX1 function are linked to autoinflammatory disorders, including systemic lupus erythematosus (SLE) and Aicardi-Goutières syndrome (AGS). Despite key roles in regulating cGAS-STING and suppressing excessive inflammation, the impact of many disease-associated TREX1 mutations-particularly those outside of the core catalytic domains-remains poorly understood. Here, we characterize a recessive AGS-linked TREX1 P61Q mutation occurring within the poorly characterized polyproline helix (PPII) motif. In keeping with its position outside of the catalytic core or ER targeting motifs, neither the P61Q mutation, nor aggregate proline-to-alanine PPII mutation, disrupts TREX1 exonuclease activity, subcellular localization, or cGAS-STING regulation in overexpression systems. Introducing targeted mutations into the endogenous TREX1 locus revealed that PPII mutations destabilize the protein, resulting in impaired exonuclease activity and unrestrained cGAS-STING activation. Overall, these results demonstrate that TREX1 PPII mutations, including P61Q, impair proper immune regulation and lead to autoimmune disease through TREX1 destabilization.
Collapse
Affiliation(s)
- Abraham Shim
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY10065, USA
| | - Xiaohan Luan
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong518055, China
| | - Wen Zhou
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong518055, China
| | - Yanick J Crow
- MRC Human Genetics Unit, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
| | - John Maciejowski
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY10065, USA
| |
Collapse
|
39
|
Zhang S, Zhang W, Ding C, Ren X, Fang F, Wu Y. Neurophenotype and genetic analysis of children with Aicardi-Goutières syndrome in China. Pediatr Investig 2024; 8:193-200. [PMID: 39347527 PMCID: PMC11427897 DOI: 10.1002/ped4.12428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 03/27/2024] [Indexed: 10/01/2024] Open
Abstract
Importance Aicardi-Goutières syndrome (AGS) is a rare genetic disorder mainly affecting the central nervous system and autoimmunity. However, research on AGS among Chinese patients is limited. Objective To summarize the neurologic phenotypes and genetic causes in pediatric AGS patients, providing insights for early recognition and diagnosis in the Chinese population. Methods Clinical features and neuroimaging results of the patients diagnosed with AGS from Beijing Children's Hospital between January 2018 and January 2022 were collected. Whole exome sequencing was used for genetic analysis. Results A total of 15 patients was included, all presenting with various neurological symptoms, including developmental delay (100%), motor skill impairment (100%), language disability (78.6%), dystonia (93.3%), microcephaly (73.3%), sleep disorders (26.7%), regression (20.0%), vessel disease (6.7%), and epilepsy (6.7%). Neuroimaging revealed intracranial calcification (86.7%), cerebral atrophy (73.3%), and leukodystrophy (73.3%). Seven genes were identified, with TREX1 being the most common (40.0%, 6/15), followed by IFIH1 (20.0%, 3/15). Variant c.294dupA (p.C99Mfs*3) was detected in four unrelated patients, accounting for 66.7% (4/6) patients with the TREX1 variant. A literature review showed that TREX1 gene mutations in 35.6% (21/59) of AGS patients among the Chinese population. Interpretation Neurological symptoms are the most prevalent and severe presentation of AGS. Diagnosis may be considered when symptoms such as developmental delay, dystonia, microcephaly, brain calcification, and leukodystrophy emerge. TREX1 mutations are predominant in the Chinese population.
Collapse
Affiliation(s)
- Shen Zhang
- Department of Neurology Beijing Children's Hospital Capital Medical University National Center for Children's Health Beijing China
| | - Weihua Zhang
- Department of Neurology Beijing Children's Hospital Capital Medical University National Center for Children's Health Beijing China
| | - Changhong Ding
- Department of Neurology Beijing Children's Hospital Capital Medical University National Center for Children's Health Beijing China
| | - Xiaotun Ren
- Department of Neurology Beijing Children's Hospital Capital Medical University National Center for Children's Health Beijing China
| | - Fang Fang
- Department of Neurology Beijing Children's Hospital Capital Medical University National Center for Children's Health Beijing China
| | - Yun Wu
- Department of Neurology Beijing Children's Hospital Capital Medical University National Center for Children's Health Beijing China
| |
Collapse
|
40
|
Thuner J, Cognard J, Belot A. How to treat monogenic SLE? Best Pract Res Clin Rheumatol 2024; 38:101962. [PMID: 38876818 DOI: 10.1016/j.berh.2024.101962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/12/2024] [Accepted: 05/29/2024] [Indexed: 06/16/2024]
Abstract
Systemic lupus erythematosus is a rare and life-threatening autoimmune disease characterized by autoantibodies against double-stranded DNA, with an immunopathology that remains partially unclear. New insights into the disease have been provided by the discovery of key mutations leading to the development of monogenic SLE, occurring in the context of early-onset disease, syndromic lupus, or familial clustering. The increased frequency of discovering these mutations in recent years, thanks to the advent of genetic screening, has greatly enhanced our understanding of the immunopathogenesis of SLE. These monogenic defects include defective clearance of apoptotic bodies, abnormalities in nucleic acid sensing, activation of the type-I interferon pathway, and the breakdown of tolerance through B or T cell activation or lymphocyte proliferation due to anomalies in TLR signalling and/or NFκB pathway overactivation. The translation of genetic discoveries into therapeutic strategies is presented here, within the framework of personalized therapy.
Collapse
Affiliation(s)
- Jonathan Thuner
- Internal Medicine Department, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre-Bénite, France; CIRI, Centre International de Recherche en Infectiologie/International Center for Infectiology Research, Université de Lyon, INSERM, Institut National de La Santé Et de La Recherche Médicale, U1111, Université Claude Bernard Lyon 1, Lyon, France
| | - Jade Cognard
- Pediatric Rheumatology, Nephrology, Dermatology Department, CMR RAISE, Women-Mother-Child Hospital, Hospices Civils de Lyon, Bron, France; CIRI, Centre International de Recherche en Infectiologie/International Center for Infectiology Research, Université de Lyon, INSERM, Institut National de La Santé Et de La Recherche Médicale, U1111, Université Claude Bernard Lyon 1, Lyon, France
| | - Alexandre Belot
- Pediatric Rheumatology, Nephrology, Dermatology Department, CMR RAISE, Women-Mother-Child Hospital, Hospices Civils de Lyon, Bron, France; CIRI, Centre International de Recherche en Infectiologie/International Center for Infectiology Research, Université de Lyon, INSERM, Institut National de La Santé Et de La Recherche Médicale, U1111, Université Claude Bernard Lyon 1, Lyon, France; CNRS, Centre National de La Recherche Scientifique, UMR5308, Lyon, France.
| |
Collapse
|
41
|
Gavazzi F, Gonzalez CD, Arnold K, Swantkowski M, Charlton L, Modesti N, Dar AA, Vanderver A, Bennett M, Adang LA. Nucleotide metabolism, leukodystrophies, and CNS pathology. J Inherit Metab Dis 2024; 47:860-875. [PMID: 38421058 PMCID: PMC11358362 DOI: 10.1002/jimd.12721] [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: 12/22/2023] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 03/02/2024]
Abstract
The balance between a protective and a destructive immune response can be precarious, as exemplified by inborn errors in nucleotide metabolism. This class of inherited disorders, which mimics infection, can result in systemic injury and severe neurologic outcomes. The most common of these disorders is Aicardi Goutières syndrome (AGS). AGS results in a phenotype similar to "TORCH" infections (Toxoplasma gondii, Other [Zika virus (ZIKV), human immunodeficiency virus (HIV)], Rubella virus, human Cytomegalovirus [HCMV], and Herpesviruses), but with sustained inflammation and ongoing potential for complications. AGS was first described in the early 1980s as familial clusters of "TORCH" infections, with severe neurology impairment, microcephaly, and basal ganglia calcifications (Aicardi & Goutières, Ann Neurol, 1984;15:49-54) and was associated with chronic cerebrospinal fluid (CSF) lymphocytosis and elevated type I interferon levels (Goutières et al., Ann Neurol, 1998;44:900-907). Since its first description, the clinical spectrum of AGS has dramatically expanded from the initial cohorts of children with severe impairment to including individuals with average intelligence and mild spastic paraparesis. This broad spectrum of potential clinical manifestations can result in a delayed diagnosis, which families cite as a major stressor. Additionally, a timely diagnosis is increasingly critical with emerging therapies targeting the interferon signaling pathway. Despite the many gains in understanding about AGS, there are still many gaps in our understanding of the cell-type drivers of pathology and characterization of modifying variables that influence clinical outcomes and achievement of timely diagnosis.
Collapse
Affiliation(s)
- Francesco Gavazzi
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | | - Kaley Arnold
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Meghan Swantkowski
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Lauren Charlton
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Nicholson Modesti
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Asif A. Dar
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Adeline Vanderver
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mariko Bennett
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Laura A. Adang
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
42
|
Song W, Bian WJ, Li H, Guo QH, Wang J, Tang B, Zhang JY, Wei W, Liu XR, Liao WP, Li B, He N. IFIH1 variants are associated with generalised epilepsy preceded by febrile seizures. J Med Genet 2024; 61:895-903. [PMID: 38964834 PMCID: PMC11420756 DOI: 10.1136/jmg-2024-109950] [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: 02/27/2024] [Accepted: 06/17/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND IFIH1 variants have been reported to be associated with immune-related disorders with/without seizures. It is unknown whether IFIH1 variants are associated with common epilepsy without acquired causes and the mechanism underlying phenotypic variation remains elusive. METHODS Trio-based whole-exome sequencing was performed on patients with febrile seizures or epilepsy with antecedent febrile seizures. Previously reported variants were systematically reviewed to investigate genotype-phenotype associations. RESULTS Two de novo heterozygous and three biallelic missense variants were identified in five patients with generalised epilepsy with antecedent febrile seizures. The variants were predicted to be damaging by in silico tools and were associated with hydrogen bonding changes to neighbouring amino acids or decreased protein stability. Patients exhibited an early onset age and became seizure-free with favourable outcome. Further analysis revealed that de novo missense variants located in the Hel region resulted in seizures with multiple neurological abnormalities, while those in the pincer domain or C-terminal domain led to seizures with normal neurodevelopment, suggesting a sub-molecular effect. Biallelic missense variants, which were inherited from unaffected parents and presented low allele frequencies in general populations, were associated with seizures without neurological abnormalities. Truncation variants were related to refractory epilepsy and severe developmental delay, suggesting a genotype-phenotype correlation. IFIH1 is predominantly expressed in the neonatal stage and decreases dramatically in the adulthood, which is consistent with the early onset age and favourable outcome of the patients. CONCLUSIONS IFIH1 variants are potentially associated with generalised epilepsy with antecedent febrile seizures. The sub-molecular implication and genotype-phenotype association help explain phenotype variations of IFIH1 variants.
Collapse
Affiliation(s)
- Wang Song
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wen-Jun Bian
- Department of Neurology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Hua Li
- Department of Neurology, The Guangdong 999 Brain Hospital, Guangzhou, Guangdong, China
| | - Qing-Hui Guo
- Department of Pediatrics, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Jie Wang
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Bin Tang
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jia-Yuan Zhang
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wei Wei
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiao-Rong Liu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wei-Ping Liao
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Bin Li
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Na He
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
43
|
Shibata Y, Shibata A, Mizuguchi T, Matsumoto N, Osaka H. A case of severe Aicardi-Goutières syndrome with a homozygous RNASEH2B intronic variant. Hum Genome Var 2024; 11:33. [PMID: 39183359 PMCID: PMC11345432 DOI: 10.1038/s41439-024-00291-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 08/27/2024] Open
Abstract
We report a case of severe Aicardi-Goutières syndrome caused by a novel homozygous RNASEH2B intronic variant, NC_000013.10(NM_024570.4):c.65-13G > A p.Glu22Valfs*5. The patient was born with pseudo-TORCH symptoms, including intracranial calcification, cataracts, and hepatosplenomegaly. Furthermore, the patient exhibited profound intellectual impairment and died at 14 months due to aspiration pneumonia accompanied by interstitial lung abnormalities. The severity of the patient's symptoms underscores the critical role of the C-terminal region of RNase H2B.
Collapse
Affiliation(s)
- Yuri Shibata
- Department of Pediatrics, Sano Kosei General Hospital, Tochigi, Japan
- Department of Pediatrics, Jichi Medical University, Tochigi, Japan
| | - Akimichi Shibata
- Department of Pediatrics, Japanese Red Cross Ashikaga Hospital, Tochigi, Japan
| | - Takeshi Mizuguchi
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Kanagawa, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Kanagawa, Japan
| | - Hitoshi Osaka
- Department of Pediatrics, Jichi Medical University, Tochigi, Japan.
| |
Collapse
|
44
|
Arkin LM, Costa-da-Silva AC, Frere J, Ng A, Sharma R, Moon JJ, Bussan HE, Kim CH, Javaid A, Steidl OR, Yatim A, Saidoune F, Gilliet M, Nguyen JT, Nihal A, Luong G, Kenfield M, Carrau L, Tran JM, Hinshaw MA, Brooks EG, Ayuso JM, O'Connor DH, Casanova JL, Cowen EW, Drolet BA, Singh AM, tenOever B, Mays JW. Pandemic-associated pernio harbors footprints of an abortive SARS-CoV-2 infection. iScience 2024; 27:110525. [PMID: 39156641 PMCID: PMC11326933 DOI: 10.1016/j.isci.2024.110525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 05/31/2024] [Accepted: 07/12/2024] [Indexed: 08/20/2024] Open
Abstract
Elevated pernio incidence was observed during the COVID-19 pandemic. This prospective study enrolled subjects with pandemic-associated pernio in Wisconsin and Switzerland. Because pernio is a cutaneous manifestation of the interferonopathies, and type I interferon (IFN-I) immunity is critical to COVID-19 recovery, we tested the hypothesis that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-mediated IFN-I signaling might underlie some pernio cases. Tissue-level IFN-I activity and plasmacytoid dendritic cell infiltrates were demonstrated in 100% of the Wisconsin cases. Across both cohorts, sparse SARS-CoV-2 RNA was captured in 25% (6/22) of biopsies, all with high inflammation. Affected patients lacked adaptive immunity to SARS-CoV-2. A hamster model of intranasal SARS-CoV-2 infection was used as a proof-of-principle experiment: RNA was detected in lungs and toes with IFN-I activity at both the sites, while replicating virus was found only in the lung. These data support a viral trigger for some pernio cases, where sustained local IFN-I activity can be triggered in the absence of seroconversion.
Collapse
Affiliation(s)
- Lisa M. Arkin
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Dermatology, Madison, WI 53726, USA
| | - Ana C. Costa-da-Silva
- Oral Immunobiology Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Justin Frere
- Department of Microbiology, New York University, Grossman School of Medicine, New York, NY 10016, USA
| | - Ashley Ng
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Dermatology, Madison, WI 53726, USA
| | - Rubina Sharma
- Oral Immunobiology Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - John J. Moon
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Dermatology, Madison, WI 53726, USA
| | - Hailey E. Bussan
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Pathology and Laboratory Medicine, Madison, WI 53726, USA
| | - Clara H. Kim
- Oral Immunobiology Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ayesha Javaid
- Oral Immunobiology Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Olivia R. Steidl
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Pediatrics, Madison, WI 53726, USA
| | - Ahmad Yatim
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
- Department of Dermatology at the University Hospital CHUV, 1011 Lausanne, Switzerland
| | - Fanny Saidoune
- Department of Dermatology at the University Hospital CHUV, 1011 Lausanne, Switzerland
| | - Michel Gilliet
- Department of Dermatology at the University Hospital CHUV, 1011 Lausanne, Switzerland
| | - Joe T. Nguyen
- Oral Immunobiology Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Aman Nihal
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Dermatology, Madison, WI 53726, USA
| | - George Luong
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Dermatology, Madison, WI 53726, USA
| | - Meaghan Kenfield
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Dermatology, Madison, WI 53726, USA
| | - Lucia Carrau
- Department of Microbiology, New York University, Grossman School of Medicine, New York, NY 10016, USA
| | - Jennifer M. Tran
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Dermatology, Madison, WI 53726, USA
| | - Molly A. Hinshaw
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Dermatology, Madison, WI 53726, USA
| | - Erin G. Brooks
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Pathology and Laboratory Medicine, Madison, WI 53726, USA
| | - Jose M. Ayuso
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Dermatology, Madison, WI 53726, USA
| | - David H. O'Connor
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Pathology and Laboratory Medicine, Madison, WI 53726, USA
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
- University of Paris Cité, Imagine Institute, 75013 Paris, France
- Howard Hughes Medical Institute, New York, NY 10065, USA
- Lab of Human Genetics of Infectious Diseases, INSERM, Necker Hospital for Sick Children, Paris, France
- Department of Pediatrics, Necker Hospital for Sick Children, 75015 Paris, France
| | - Edward W. Cowen
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Beth A. Drolet
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Dermatology, Madison, WI 53726, USA
| | - Anne Marie Singh
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Pediatrics, Madison, WI 53726, USA
| | - Benjamin tenOever
- Department of Microbiology, New York University, Grossman School of Medicine, New York, NY 10016, USA
| | - Jacqueline W. Mays
- Oral Immunobiology Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
45
|
Santoro L, Grillo F, D'Armiento M, Buccoliero AM, Rocco M, Ferro J, Vanoli A, Cafferata B, Macciomei MC, Mescoli C, Cananzi M, Alaggio R, Fassan M, Mastracci L, Francalanci P, Parente P. Clinicopathologic Features of Primary Immunodeficiency Monogenic Disease-related Very Early Onset Inflammatory Bowel Disease: Focus on Gastrointestinal Histologic Features in IFIH1 Mutations. Adv Anat Pathol 2024:00125480-990000000-00113. [PMID: 39140676 DOI: 10.1097/pap.0000000000000457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Very early onset inflammatory bowel disease (VEO-IBD) is a clinical term referring to IBD-like symptomatology arising in children younger than 6 years. VEO-IBD may be due to polygenic etiology in "pure" IBD (Crohn disease-CD and ulcerative colitis-UC), or it may be caused by primary immunodeficiency underlined by monogenic disease. Primary immunodeficiency monogenic diseases have a Mendelian inheritance and affect the immune system with multiorgan morbidity and possible effects on the gastrointestinal system. Primary Immunodeficiency monogenic diseases differ from "pure" IBD as the latter primarily affect the gastrointestinal tract with mitigated extraintestinal symptomatology. Since their first description, primary immunodeficiency monogenic diseases, although rare, have been the subject of increasing interest due to their dramatic phenotype, difficulty in reaching a timely diagnosis, and specific therapeutic approach. In this paper, we present a brief review of primary immunodeficiency monogenic diseases, focusing on to their clinicopathologic features as well as delving, in greater detail, into monogenic diseases caused by IFIH1 mutations. The clinicopathologic features of 4 patients with IFIH1, a gene involved in interferon pathway deficiency, will be described using a histologic pattern of damage approach confirming the need to avoid the histologic diagnosis of VEO-IBD in children younger than 6 years.
Collapse
Affiliation(s)
- Luisa Santoro
- Pathology Unit, Azienda Ospedaliera Padova, Via Ospedale Vecchio
| | - Federica Grillo
- Anatomic Pathology Unit, IRCCS Ospedale Policlinico San Martino, Largo Rosanna
- Anatomic Pathology, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova
| | - Maria D'Armiento
- Pathology Unit, Department of Public Health, University of Naples Federico II, Via Sergio Pansini, Napoli
| | | | - Michele Rocco
- Department of Pathology, Santobono-Pausilipon, Children's Hospital, Naples
| | - Jacopo Ferro
- Pathology Unit, IRCCS Istituto Giannina Gaslini, Via Gerolamo, Genova
| | - Alessandro Vanoli
- Department of Molecular Medicine, University of Pavia
- Unit of Anatomic Pathology, IRCCS San Matteo Hospital Foundation, Viale Camillo, Pavia
| | - Barbara Cafferata
- Pathology Unit, IRCCS Istituto Giannina Gaslini, Via Gerolamo, Genova
| | | | - Claudia Mescoli
- Pathology Unit, Azienda Ospedaliera Padova, Via Ospedale Vecchio
| | - Mara Cananzi
- Unit of Pediatric Gastroenterology, Digestive Endoscopy, Hepatology and Care of the Child with Liver Transplantation, Department of Women's and Children's Health, University Hospital of Padova, Via Ospedale Vecchio, Padova, Italy
| | - Rita Alaggio
- Pathology Unit, Department of Laboratories, IRCCS Bambino Gesù Children's Hospital, Roma
| | - Matteo Fassan
- Surgical Pathology Unit, Department of Medicine (DIMED), University Hospital of Padua, Via Aristide Gabelli
- Veneto Institute of Oncology IOV-IRCCS, Viale Gattamelata, Padua
| | - Luca Mastracci
- Anatomic Pathology Unit, IRCCS Ospedale Policlinico San Martino, Largo Rosanna
- Anatomic Pathology, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova
| | - Paola Francalanci
- Pathology Unit, Department of Laboratories, IRCCS Bambino Gesù Children's Hospital, Roma
| | - Paola Parente
- Pathology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Cappuccini, San Giovanni Rotondo (FG), Italy
| |
Collapse
|
46
|
Small AM, Yutzey KE, Binstadt BA, Voigts Key K, Bouatia-Naji N, Milan D, Aikawa E, Otto CM, St Hilaire C. Unraveling the Mechanisms of Valvular Heart Disease to Identify Medical Therapy Targets: A Scientific Statement From the American Heart Association. Circulation 2024; 150:e109-e128. [PMID: 38881493 PMCID: PMC11542557 DOI: 10.1161/cir.0000000000001254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Valvular heart disease is a common cause of morbidity and mortality worldwide and has no effective medical therapy. Severe disease is managed with valve replacement procedures, which entail high health care-related costs and postprocedural morbidity and mortality. Robust ongoing research programs have elucidated many important molecular pathways contributing to primary valvular heart disease. However, there remain several key challenges inherent in translating research on valvular heart disease to viable molecular targets that can progress through the clinical trials pathway and effectively prevent or modify the course of these common conditions. In this scientific statement, we review the basic cellular structures of the human heart valves and discuss how these structures change in primary valvular heart disease. We focus on the most common primary valvular heart diseases, including calcific aortic stenosis, bicuspid aortic valves, mitral valve prolapse, and rheumatic heart disease, and outline the fundamental molecular discoveries contributing to each. We further outline potential therapeutic molecular targets for primary valvular heart disease and discuss key knowledge gaps that might serve as future research priorities.
Collapse
|
47
|
Tusseau M, Belot A. [Rare Autoimmune Diseases Role of Genetics - Example of Systemic Lupus Erythematosus]. Biol Aujourdhui 2024; 218:9-18. [PMID: 39007772 DOI: 10.1051/jbio/2024005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Indexed: 07/16/2024]
Abstract
Systemic lupus erythematosus (SLE) presents a complex clinical landscape with diverse manifestations, suggesting a multifactorial etiology. However, the identification of rare monogenic forms of the disease has shed light on specific genetic defects underlying SLE pathogenesis, offering valuable insights into its underlying mechanisms and clinical heterogeneity. By categorizing these monogenic forms based on the implicated signaling pathways, such as apoptotic body clearance, type I interferon signaling, JAK-STAT pathway dysregulation, innate immune receptor dysfunction and lymphocytic abnormalities, a more nuanced understanding of SLE's molecular basis emerges. Particularly in pediatric populations, where monogenic forms are more prevalent, routine genetic testing becomes increasingly important, with a diagnostic yield of approximately 10% depending on the demographic and methodological factors involved. This approach not only enhances diagnostic accuracy but also informs personalized treatment strategies tailored to the specific molecular defects driving the disease phenotype.
Collapse
Affiliation(s)
- Maud Tusseau
- Laboratoire de génétique des cancers et maladies multifactorielles, Service de génétique médicale, Hospices Civils de Lyon, Bron, France - Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard, Lyon 1, CNRS UMR5308, ENS de Lyon, Lyon, France
| | - Alexandre Belot
- Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard, Lyon 1, CNRS UMR5308, ENS de Lyon, Lyon, France - Centre de référence des maladies rhumatologiques inflammatoires, des maladies auto-immunes et interféronopathies systémiques de l'enfant, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Bron, France - Service de néphrologie, rhumatologie, dermatologie pédiatrique, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
48
|
Viengkhou B, Hayashida E, McGlasson S, Emelianova K, Forbes D, Wiseman S, Wardlaw J, Verdillo R, Irani SR, Duffy D, Piehl F, Loo L, Pagenstecher A, Neely GG, Crow YJ, Campbell IL, Hunt DPJ, Hofer MJ. The brain microvasculature is a primary mediator of interferon-α neurotoxicity in human cerebral interferonopathies. Immunity 2024; 57:1696-1709.e10. [PMID: 38878770 PMCID: PMC11250091 DOI: 10.1016/j.immuni.2024.05.017] [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: 02/22/2022] [Revised: 01/10/2024] [Accepted: 05/17/2024] [Indexed: 07/12/2024]
Abstract
Aicardi-Goutières syndrome (AGS) is an autoinflammatory disease characterized by aberrant interferon (IFN)-α production. The major cause of morbidity in AGS is brain disease, yet the primary source and target of neurotoxic IFN-α remain unclear. Here, we demonstrated that the brain was the primary source of neurotoxic IFN-α in AGS and confirmed the neurotoxicity of intracerebral IFN-α using astrocyte-driven Ifna1 misexpression in mice. Using single-cell RNA sequencing, we demonstrated that intracerebral IFN-α-activated receptor (IFNAR) signaling within cerebral endothelial cells caused a distinctive cerebral small vessel disease similar to that observed in individuals with AGS. Magnetic resonance imaging (MRI) and single-molecule ELISA revealed that central and not peripheral IFN-α was the primary determinant of microvascular disease in humans. Ablation of endothelial Ifnar1 in mice rescued microvascular disease, stopped the development of diffuse brain disease, and prolonged lifespan. These results identify the cerebral microvasculature as a primary mediator of IFN-α neurotoxicity in AGS, representing an accessible target for therapeutic intervention.
Collapse
Affiliation(s)
- Barney Viengkhou
- School of Life and Environmental Sciences and the Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Emina Hayashida
- School of Life and Environmental Sciences and the Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Sarah McGlasson
- UK Dementia Research Institute at University of Edinburgh, Edinburgh EH16 4SB, UK; Centre for Clinical Brain Sciences at University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Katie Emelianova
- UK Dementia Research Institute at University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Deborah Forbes
- UK Dementia Research Institute at University of Edinburgh, Edinburgh EH16 4SB, UK; Centre for Clinical Brain Sciences at University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Stewart Wiseman
- Centre for Clinical Brain Sciences at University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Joanna Wardlaw
- UK Dementia Research Institute at University of Edinburgh, Edinburgh EH16 4SB, UK; Centre for Clinical Brain Sciences at University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Rovin Verdillo
- School of Life and Environmental Sciences and the Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Sarosh R Irani
- Oxford Autoimmune Neurology Group, University of Oxford, Oxford, UK
| | - Darragh Duffy
- Translational Immunology Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | - Fredrik Piehl
- Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Lipin Loo
- School of Life and Environmental Sciences and the Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Axel Pagenstecher
- Department of Neuropathology, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany
| | - G Greg Neely
- School of Life and Environmental Sciences and the Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Yanick J Crow
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK; Laboratory of Neurogenetics and Neuroinflammation, Institut Imagine, Université de Paris, Paris, France
| | - Iain L Campbell
- School of Life and Environmental Sciences and the Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - David P J Hunt
- UK Dementia Research Institute at University of Edinburgh, Edinburgh EH16 4SB, UK; Centre for Clinical Brain Sciences at University of Edinburgh, Edinburgh EH16 4SB, UK.
| | - Markus J Hofer
- School of Life and Environmental Sciences and the Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
49
|
Crow MK, Olferiev M, Kirou KA. Standing on Shoulders: Interferon Research From Viral Interference to Lupus Pathogenesis and Treatment. Arthritis Rheumatol 2024; 76:1002-1012. [PMID: 38500017 DOI: 10.1002/art.42849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/24/2024] [Accepted: 02/29/2024] [Indexed: 03/20/2024]
Abstract
The discovery of interferon in the 1950s represents much more than the identification of the first cytokine and the key mediator of antiviral host defense. Defining the molecular nature and complexity of the type I interferon family, as well as its inducers and molecular mechanisms of action, was the work of investigators working at the highest level and producing insights of great consequence. Current knowledge of receptor-ligand interactions, cell signaling, and transcriptional regulation derives from studies of type I interferon. It is on the shoulders of the giants who produced that knowledge that others stand and have revealed critical mechanisms of the pathogenesis of systemic lupus erythematosus and other autoimmune diseases. The design of novel therapeutics is informed by the advances in investigation of type I interferon, with the potential for important impact on patient management.
Collapse
Affiliation(s)
- Mary K Crow
- Mary Kirkland Center for Lupus Research, Hospital for Special Surgery and Weill Cornell Medicine, New York City, New York
| | - Mikhail Olferiev
- Mary Kirkland Center for Lupus Research, Hospital for Special Surgery and Weill Cornell Medicine, New York City, New York
| | - Kyriakos A Kirou
- Mary Kirkland Center for Lupus Research, Hospital for Special Surgery and Weill Cornell Medicine, New York City, New York
| |
Collapse
|
50
|
Zhou Y, Song HM. Type I interferon pathway in pediatric systemic lupus erythematosus. World J Pediatr 2024; 20:653-668. [PMID: 38914753 PMCID: PMC11269505 DOI: 10.1007/s12519-024-00811-4] [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: 11/27/2023] [Accepted: 03/27/2024] [Indexed: 06/26/2024]
Abstract
BACKGROUND The role of type I interferon (IFN-I) signaling in systemic lupus erythematosus (SLE) has been well established. However, unanswered questions remain regarding the applicability of these findings to pediatric-onset SLE. The aim of this review is to provide an overview of the novel discoveries on IFN-I signaling in pediatric-onset SLE. DATA SOURCES A literature search was conducted in the PubMed database using the following keywords: "pediatric systemic lupus erythematosus" and "type I interferon". RESULTS IFN-I signaling is increased in pediatric SLE, largely due to the presence of plasmacytoid dendritic cells and pathways such as cyclic GMP-AMP synthase-stimulator of interferon genes-TANK-binding kinase 1 and Toll-like receptor (TLR)4/TLR9. Neutrophil extracellular traps and oxidative DNA damage further stimulate IFN-I production. Genetic variants in IFN-I-related genes, such as IFN-regulatory factor 5 and tyrosine kinase 2, are linked to SLE susceptibility in pediatric patients. In addition, type I interferonopathies, characterized by sustained IFN-I activation, can mimic SLE symptoms and are thus important to distinguish. Studies on interferonopathies also contribute to exploring the pathogenesis of SLE. Measuring IFN-I activation is crucial for SLE diagnosis and stratification. Both IFN-stimulated gene expression and serum IFN-α2 levels are common indicators. Flow cytometry markers such as CD169 and galectin-9 are promising alternatives. Anti-IFN therapies, such as sifalimumab and anifrolumab, show promise in adult patients with SLE, but their efficacy in pediatric patients requires further investigation. Janus kinase inhibitors are another treatment option for severe pediatric SLE patients. CONCLUSIONS This review presents an overview of the IFN-I pathway in pediatric SLE. Understanding the intricate relationship between IFN-I and pediatric SLE may help to identify potential diagnostic markers and targeted therapies, paving the way for improved patient care and outcomes.
Collapse
Affiliation(s)
- Yu Zhou
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
| | - Hong-Mei Song
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China.
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China.
| |
Collapse
|