1
|
Klein B, Reynolds MB, Xu B, Gharaee-Kermani M, Gao Y, Berthier CC, Henning S, Tsoi LC, Loftus SN, McNeely KE, Goudsmit CM, Victory AM, Dobry C, Hile GA, Ma F, Turnier JL, Gudjonsson JE, O'Riordan MX, Kahlenberg JM. Epidermal ZBP1 stabilizes mitochondrial Z-DNA to drive UV-induced IFN signaling in autoimmune photosensitivity. Sci Immunol 2025; 10:eado1710. [PMID: 40053607 DOI: 10.1126/sciimmunol.ado1710] [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: 01/22/2024] [Revised: 11/14/2024] [Accepted: 02/11/2025] [Indexed: 03/09/2025]
Abstract
Photosensitivity is observed in numerous autoimmune diseases and drives poor quality of life and disease flares. Elevated epidermal type I interferon (IFN) production primes for photosensitivity and enhanced inflammation, but the substrates that sustain and amplify this cycle remain undefined. We show that IFN-induced Z-DNA binding protein 1 (ZBP1) stabilizes ultraviolet (UV) B-induced cytosolic Z-DNA derived from oxidized mitochondrial DNA. ZBP1 is up-regulated in the epidermis of adult and pediatric patients with autoimmune photosensitivity. In patient-derived samples, lupus keratinocytes accumulate extensive cytosolic Z-DNA after UVB exposure, and transfection of keratinocytes with Z-DNA results in stronger IFN production through cyclic guanosine monophosphate-adenosine monophosphate synthase-stimulator of interferon genes (cGAS-STING) activation compared with the more conventional B-DNA. ZBP1 knockdown abrogates UVB-induced IFN responses, whereas overexpression results in a lupus-like phenotype with spontaneous Z-DNA accumulation and IFN production. Our results highlight Z-DNA and ZBP1 as critical mediators for UVB-induced inflammation and uncover how type I IFNs prime for cutaneous inflammation in photosensitivity.
Collapse
Affiliation(s)
- Benjamin Klein
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Mack B Reynolds
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Bin Xu
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Mehrnaz Gharaee-Kermani
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Yiqing Gao
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Celine C Berthier
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Svenja Henning
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Lam C Tsoi
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Shannon N Loftus
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Kelsey E McNeely
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Christine M Goudsmit
- Division of Pediatric Rheumatology, Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Amanda M Victory
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Craig Dobry
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Grace A Hile
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Feiyang Ma
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Jessica L Turnier
- Division of Pediatric Rheumatology, Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Johann E Gudjonsson
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Mary X O'Riordan
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| | - J Michelle Kahlenberg
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
2
|
Navarro HI, Daly AE, Rodriguez B, Wu S, Ngo KA, Fraser A, Schiffman A, Liu Y, Smale ST, Chia JJ, Hoffmann A. NF-κB RelB suppresses the inflammatory gene expression programs of dendritic cells by competing with RelA for binding to target gene promoters. Cell Discov 2025; 11:13. [PMID: 39929805 PMCID: PMC11811218 DOI: 10.1038/s41421-024-00767-9] [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: 10/19/2023] [Accepted: 12/27/2024] [Indexed: 02/13/2025] Open
Abstract
A group of autoinflammatory disorders termed relopathies arise as a consequence of NF-κB dysregulation. Genetic loss of the NF-κB subunit RelB in humans and mice leads to autoimmunity and lethal multi-organ inflammatory pathology. Our recent study showed that this inflammatory pathology is independent of type I interferon signaling, and further identified dysregulation of a set of pro-inflammatory NF-κB target genes. However, it remains unknown how the loss of RelB leads to the dysregulation of these NF-κB motif-containing pro-inflammatory genes. Here, we report epigenome profiling studies revealing that RelB is associated with pro-inflammatory genes in dendritic cells. While these genes recruit RelA binding upon exposure to a maturation stimulus, we observed substantially more RelA recruitment in the absence of RelB. For these genes, we found that elevated RelA recruitment is correlated with elevated gene expression. To test whether RelB may compete with RelA for binding to NF-κB-regulated gene promoters via competition for κB sites, we generated a new mouse strain (RelBDB/DB) that harbors targeted point mutations in the RelB DNA binding domain that eliminates high-affinity DNA binding. We found that this targeted mutation in the RelB DNA binding domain is sufficient to drive multi-organ inflammatory pathology. These results provide insights into the biological mechanism of RelB as a suppressor of pro-inflammatory gene expression and autoimmune pathology.
Collapse
Affiliation(s)
- Héctor I Navarro
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
- Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Allison E Daly
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
- Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Benancio Rodriguez
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
- Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Sunny Wu
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Kim A Ngo
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Anna Fraser
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, CA, USA
| | - Allison Schiffman
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
- Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Yi Liu
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
- DeepKinase Biotechnologies Ltd., Beijing, China
| | - Stephen T Smale
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
- Molecular Biology Institute, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA
| | - Jennifer J Chia
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA.
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA.
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, USA.
| | - Alexander Hoffmann
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California, Los Angeles, CA, USA.
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA.
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA.
| |
Collapse
|
3
|
Maliah A, Santana-Magal N, Parikh S, Gordon S, Reshef K, Sade Y, Khateeb A, Richter A, Gutwillig A, Parikh R, Golan T, Krissi M, Na M, Binshtok G, Manich P, Elkoshi N, Grisaru-Tal S, Zemser-Werner V, Brenner R, Vaknine H, Nizri E, Moyal L, Amitay-Laish I, Rosemberg L, Munitz A, Kronfeld-Schor N, Shifrut E, Kobiler O, Madi A, Geiger T, Carmi Y, Levy C. Crosslinking of Ly6a metabolically reprograms CD8 T cells for cancer immunotherapy. Nat Commun 2024; 15:8354. [PMID: 39333093 PMCID: PMC11437002 DOI: 10.1038/s41467-024-52079-x] [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: 06/13/2023] [Accepted: 08/25/2024] [Indexed: 09/29/2024] Open
Abstract
T cell inhibitory mechanisms prevent autoimmune reactions, while cancer immunotherapy aims to remove these inhibitory signals. Chronic ultraviolet (UV) exposure attenuates autoimmunity through promotion of poorly understood immune-suppressive mechanisms. Here we show that mice with subcutaneous melanoma are not responsive to anti-PD1 immunotherapy following chronic UV irradiation, given prior to tumor injection, due to the suppression of T cell killing ability in skin-draining lymph nodes. Using mass cytometry and single-cell RNA-sequencing analyzes, we discover that skin-specific, UV-induced suppression of T-cells killing activity is mediated by upregulation of a Ly6ahigh T-cell subpopulation. Independently of the UV effect, Ly6ahigh T cells are induced by chronic type-1 interferon in the tumor microenvironment. Treatment with an anti-Ly6a antibody enhances the anti-tumoral cytotoxic activity of T cells and reprograms their mitochondrial metabolism via the Erk/cMyc axis. Treatment with an anti-Ly6a antibody inhibits tumor growth in mice resistant to anti-PD1 therapy. Applying our findings in humans could lead to an immunotherapy treatment for patients with resistance to existing treatments.
Collapse
Affiliation(s)
- Avishai Maliah
- Department of Human Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nadine Santana-Magal
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shivang Parikh
- The Ragon Institute of Mass General, MIT and Harvard 600/625 Main Street, Cambridge, MA, USA
| | - Sagi Gordon
- Department of Human Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Keren Reshef
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yuval Sade
- Department of Human Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Aseel Khateeb
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Alon Richter
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Amit Gutwillig
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Roma Parikh
- Department of Human Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tamar Golan
- Department of Human Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Matan Krissi
- Department of Human Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Manho Na
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Gal Binshtok
- Department of Human Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Paulee Manich
- Department of Human Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nadav Elkoshi
- Department of Human Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sharon Grisaru-Tal
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Ronen Brenner
- Institute of Oncology, E. Wolfson Medical Center, Holon, Israel
| | - Hananya Vaknine
- Institute of Pathology, E. Wolfson Medical Center, Holon, Israel
| | - Eran Nizri
- Peritoneal Surface Malignancies and Melanoma Unit, Department of Surgery A, Tel-Aviv Sourasky Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Lilach Moyal
- Felsenstein Medical Research Center, Tel-Aviv University and the Division of Dermatology, Rabin Medical Center, Petach Tikva, Israel
| | - Iris Amitay-Laish
- Felsenstein Medical Research Center, Tel-Aviv University and the Division of Dermatology, Rabin Medical Center, Petach Tikva, Israel
| | - Luiza Rosemberg
- School of Zoology, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Ariel Munitz
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Eric Shifrut
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Faculty of Life Sciences, School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
- Dotan Center for Advanced Therapies, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Oren Kobiler
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Asaf Madi
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tamar Geiger
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Yaron Carmi
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Carmit Levy
- Department of Human Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
4
|
Li TM, Zyulina V, Seltzer ES, Dacic M, Chinenov Y, Daamen AR, Veiga KR, Schwartz N, Oliver DJ, Cabahug-Zuckerman P, Lora J, Liu Y, Shipman WD, Ambler WG, Taber SF, Onel KB, Zippin JH, Rashighi M, Krueger JG, Anandasabapathy N, Rogatsky I, Jabbari A, Blobel CP, Lipsky PE, Lu TT. The interferon-rich skin environment regulates Langerhans cell ADAM17 to promote photosensitivity in lupus. eLife 2024; 13:e85914. [PMID: 38860651 PMCID: PMC11213570 DOI: 10.7554/elife.85914] [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: 01/02/2023] [Accepted: 06/10/2024] [Indexed: 06/12/2024] Open
Abstract
The autoimmune disease lupus erythematosus (lupus) is characterized by photosensitivity, where even ambient ultraviolet radiation (UVR) exposure can lead to development of inflammatory skin lesions. We have previously shown that Langerhans cells (LCs) limit keratinocyte apoptosis and photosensitivity via a disintegrin and metalloprotease 17 (ADAM17)-mediated release of epidermal growth factor receptor (EGFR) ligands and that LC ADAM17 sheddase activity is reduced in lupus. Here, we sought to understand how the lupus skin environment contributes to LC ADAM17 dysfunction and, in the process, differentiate between effects on LC ADAM17 sheddase function, LC ADAM17 expression, and LC numbers. We show through transcriptomic analysis a shared IFN-rich environment in non-lesional skin across human lupus and three murine models: MRL/lpr, B6.Sle1yaa, and imiquimod (IMQ) mice. IFN-I inhibits LC ADAM17 sheddase activity in murine and human LCs, and IFNAR blockade in lupus model mice restores LC ADAM17 sheddase activity, all without consistent effects on LC ADAM17 protein expression or LC numbers. Anti-IFNAR-mediated LC ADAM17 sheddase function restoration is associated with reduced photosensitive responses that are dependent on EGFR signaling and LC ADAM17. Reactive oxygen species (ROS) is a known mediator of ADAM17 activity; we show that UVR-induced LC ROS production is reduced in lupus model mice, restored by anti-IFNAR, and is cytoplasmic in origin. Our findings suggest that IFN-I promotes photosensitivity at least in part by inhibiting UVR-induced LC ADAM17 sheddase function and raise the possibility that anifrolumab ameliorates lupus skin disease in part by restoring this function. This work provides insight into IFN-I-mediated disease mechanisms, LC regulation, and a potential mechanism of action for anifrolumab in lupus.
Collapse
Affiliation(s)
- Thomas Morgan Li
- Autoimmunity and Inflammation Program, Hospital for Special Surgery Research InstituteNew YorkUnited States
| | - Victoria Zyulina
- Autoimmunity and Inflammation Program, Hospital for Special Surgery Research InstituteNew YorkUnited States
- Department of Microbiology and Immunology, Weill Cornell Medical CollegeNew YorkUnited States
| | - Ethan S Seltzer
- Autoimmunity and Inflammation Program, Hospital for Special Surgery Research InstituteNew YorkUnited States
| | - Marija Dacic
- David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery Research InstituteNew YorkUnited States
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery Research InstituteNew YorkUnited States
- Physiology, Biophysics, and Systems Biology Program, Weill Cornell Graduate School of Medical SciencesNew YorkUnited States
| | - Yurii Chinenov
- David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery Research InstituteNew YorkUnited States
| | - Andrea R Daamen
- Department of Medicine, AMPEL BioSolutionsCharlottesvilleUnited States
| | - Keila R Veiga
- Autoimmunity and Inflammation Program, Hospital for Special Surgery Research InstituteNew YorkUnited States
- Pediatric Rheumatology, Department of Medicine, Hospital for Special SurgeryNew YorkUnited States
- Department of Pediatrics, Weill Cornell Medical CollegeNew YorkUnited States
| | - Noa Schwartz
- Autoimmunity and Inflammation Program, Hospital for Special Surgery Research InstituteNew YorkUnited States
- Rheumatology, Department of Medicine, Hospital for Special SurgeryNew YorkUnited States
| | - David J Oliver
- David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery Research InstituteNew YorkUnited States
| | - Pamela Cabahug-Zuckerman
- Autoimmunity and Inflammation Program, Hospital for Special Surgery Research InstituteNew YorkUnited States
| | - Jose Lora
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery Research InstituteNew YorkUnited States
- Physiology, Biophysics, and Systems Biology Program, Weill Cornell Graduate School of Medical SciencesNew YorkUnited States
| | - Yong Liu
- Department of Dermatology, Weill Cornell Medical CollegeNew YorkUnited States
| | - William D Shipman
- Autoimmunity and Inflammation Program, Hospital for Special Surgery Research InstituteNew YorkUnited States
- Weill Cornell/Rockefeller/Sloan-Kettering Tri-Institutional MD-PhD Program, Weill Cornell Medical CollegeNew YorkUnited States
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical SciencesNew YorkUnited States
| | - William G Ambler
- Autoimmunity and Inflammation Program, Hospital for Special Surgery Research InstituteNew YorkUnited States
- Pediatric Rheumatology, Department of Medicine, Hospital for Special SurgeryNew YorkUnited States
- Department of Pediatrics, Weill Cornell Medical CollegeNew YorkUnited States
| | - Sarah F Taber
- Pediatric Rheumatology, Department of Medicine, Hospital for Special SurgeryNew YorkUnited States
- Department of Pediatrics, Weill Cornell Medical CollegeNew YorkUnited States
| | - Karen B Onel
- Pediatric Rheumatology, Department of Medicine, Hospital for Special SurgeryNew YorkUnited States
- Department of Pediatrics, Weill Cornell Medical CollegeNew YorkUnited States
| | - Jonathan H Zippin
- Department of Dermatology, Weill Cornell Medical CollegeNew YorkUnited States
| | - Mehdi Rashighi
- Department of Dermatology, University of Massachusetts Medical SchoolWorcesterUnited States
| | - James G Krueger
- Laboratory of Investigative Dermatology, Rockefeller UniversityNew YorkUnited States
| | - Niroshana Anandasabapathy
- Department of Dermatology, Weill Cornell Medical CollegeNew YorkUnited States
- Weill Cornell/Rockefeller/Sloan-Kettering Tri-Institutional MD-PhD Program, Weill Cornell Medical CollegeNew YorkUnited States
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical SciencesNew YorkUnited States
| | - Inez Rogatsky
- Department of Microbiology and Immunology, Weill Cornell Medical CollegeNew YorkUnited States
- David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery Research InstituteNew YorkUnited States
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery Research InstituteNew YorkUnited States
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical SciencesNew YorkUnited States
| | - Ali Jabbari
- Laboratory of Investigative Dermatology, Rockefeller UniversityNew YorkUnited States
| | - Carl P Blobel
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery Research InstituteNew YorkUnited States
- Physiology, Biophysics, and Systems Biology Program, Weill Cornell Graduate School of Medical SciencesNew YorkUnited States
- Department of Physiology, Biophysics, and Systems Biology, Weill Cornell Medical CollegeNew YorkUnited States
| | - Peter E Lipsky
- Department of Medicine, AMPEL BioSolutionsCharlottesvilleUnited States
| | - Theresa T Lu
- Autoimmunity and Inflammation Program, Hospital for Special Surgery Research InstituteNew YorkUnited States
- Department of Microbiology and Immunology, Weill Cornell Medical CollegeNew YorkUnited States
- Pediatric Rheumatology, Department of Medicine, Hospital for Special SurgeryNew YorkUnited States
- Department of Pediatrics, Weill Cornell Medical CollegeNew YorkUnited States
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical SciencesNew YorkUnited States
| |
Collapse
|
5
|
Lupu VV, Lupu A, Jechel E, Starcea IM, Stoleriu G, Ioniuc I, Azoicai A, Danielescu C, Knieling A, Borka-Balas R, Salaru DL, Revenco N, Fotea S. The role of vitamin D in pediatric systemic lupus erythematosus - a double pawn in the immune and microbial balance. Front Immunol 2024; 15:1373904. [PMID: 38715605 PMCID: PMC11074404 DOI: 10.3389/fimmu.2024.1373904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 04/08/2024] [Indexed: 05/23/2024] Open
Abstract
Having increased popularity during the Covid-19 pandemic, vitamin D3 is currently impressing thanks to the numerous researches aimed at its interactions with the body's homeostasis. At the same time, there is a peak in terms of recommendations for supplementation with it. Some of the studies focus on the link between autoimmune diseases and nutritional deficiencies, especially vitamin D3. Since the specialized literature aimed at children (patients between 0-18 years old) is far from equal to the informational diversity of the adult-centered branch, this review aims to bring up to date the relationship between the microbial and nutritional balance and the activity of pediatric systemic lupus erythematosus (pSLE). The desired practical purpose resides in a better understanding and an adequate, individualized management of the affected persons to reduce morbidity. The center of the summary is to establish the impact of hypovitaminosis D in the development and evolution of pediatric lupus erythematosus. We will address aspects related to the two entities of the impact played by vitamin D3 in the pathophysiological cascade of lupus, but also the risk of toxicity and its effects when the deficiency is over supplemented (hypervitaminosis D). We will debate the relationship of hypovitaminosis D with the modulation of immune function, the potentiation of inflammatory processes, the increase of oxidative stress, the perfusion of cognitive brain areas, the seasonal incidence of SLE and its severity. Finally, we review current knowledge, post-pandemic, regarding the hypovitaminosis D - pSLE relationship.
Collapse
Affiliation(s)
- Vasile Valeriu Lupu
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Ancuta Lupu
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Elena Jechel
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | | | - Gabriela Stoleriu
- Clinical Medical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, Galati, Romania
| | - Ileana Ioniuc
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Alice Azoicai
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Ciprian Danielescu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Anton Knieling
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Reka Borka-Balas
- Pediatrics, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, Targu Mures, Romania
| | - Delia Lidia Salaru
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Ninel Revenco
- Pediatrics, “Nicolae Testemitanu” State University of Medicine and Pharmacy, Chisinau, Moldova
| | - Silvia Fotea
- Clinical Medical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, Galati, Romania
| |
Collapse
|
6
|
Klein B, Reynolds MB, Xu B, Gharaee-Kermani M, Gao Y, Berthier CC, Henning S, Loftus SN, McNeely KE, Victory AM, Dobry C, Hile GA, Ma F, Turnier JL, Gudjonsson JE, O’Riordan MX, Kahlenberg JM. Epidermal ZBP1 stabilizes mitochondrial Z-DNA to drive UV-induced IFN signaling in autoimmune photosensitivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.23.576771. [PMID: 38328232 PMCID: PMC10849619 DOI: 10.1101/2024.01.23.576771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Photosensitivity is observed in numerous autoimmune diseases and drives poor quality of life and disease flares. Elevated epidermal type I interferon (IFN) production primes for photosensitivity and enhanced inflammation, but the substrates that sustain and amplify this cycle remain undefined. Here, we show that IFN-induced Z-DNA binding protein 1 (ZBP1) stabilizes ultraviolet (UV)B-induced cytosolic Z-DNA derived from oxidized mitochondrial DNA. ZBP1 is significantly upregulated in the epidermis of adult and pediatric patients with autoimmune photosensitivity. Strikingly, lupus keratinocytes accumulate extensive cytosolic Z-DNA after UVB, and transfection of keratinocytes with Z-DNA results in stronger IFN production through cGAS-STING activation compared to B-DNA. ZBP1 knockdown abrogates UV-induced IFN responses, whereas overexpression results in a lupus-like phenotype with spontaneous Z-DNA accumulation and IFN production. Our results highlight Z-DNA and ZBP1 as critical mediators for UVB-induced inflammation and uncover how type I IFNs prime for cutaneous inflammation in photosensitivity.
Collapse
Affiliation(s)
- Benjamin Klein
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Mack B. Reynolds
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor
| | - Bin Xu
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Mehrnaz Gharaee-Kermani
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan
| | - Yiqing Gao
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Celine C. Berthier
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Svenja Henning
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Shannon N. Loftus
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Kelsey E. McNeely
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Amanda M. Victory
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Craig Dobry
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan
| | - Grace A. Hile
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan
| | - Feiyang Ma
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan
| | - Jessica L. Turnier
- Division of Pediatric Rheumatology, Department of Pediatrics, University of Michigan, Ann Arbor
| | | | - Mary X. O’Riordan
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor
| | - J. Michelle Kahlenberg
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
7
|
Lu D, Zhu X, Hong T, Yao X, Xie Z, Chen L, Wang Y, Zhang K, Ren Y, Cao Y, Wang X. Serum Metabolomics Analysis of Skin-Involved Systemic Lupus Erythematosus: Association of Anti-SSA Antibodies with Photosensitivity. J Inflamm Res 2023; 16:3811-3822. [PMID: 37667802 PMCID: PMC10475307 DOI: 10.2147/jir.s426337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/23/2023] [Indexed: 09/06/2023] Open
Abstract
Purpose Systemic lupus erythematosus is a heterogeneous autoimmune disease in which skin involvement is a common manifestation. It is currently thought that the photosensitivity of SLE skin involvement is associated with anti-SSA antibodies. This study aimed to expand the current state of knowledge surrounding the molecular pathophysiology of SLE skin photosensitivity through Serum metabolomics analysis. Patients and Methods The serum metabolites of 23 cases of skin-involved SLE (SI) group, 14 cases of no SI (NSI) group, and 30 cases of healthy controls (HC) were analyzed by using UPLC-MS/MS technology, and subgroup analysis was performed according to the expression of anti-SSA antibodies in SI. MetaboAnalyst 5.0 was used for enrichment analysis and ROC curve construction, identifying serum metabolic markers of skin-involved SLE associated with anti-SSA antibodies. Results We identified several metabolites and metabolic pathways associated with SLE photosensitivity. Two metabolites, SM (d18:1/24:0) and gamma-CEHC can distinguish between anti-SSA antibody-positive and negative SI, with AUC of 0.829 and 0.806. These two photosensitization-related substances may be potential markers of skin involvement in SLE associated with anti-SSA antibody. Conclusion This study provides new insights into the pathogenesis of SI patients, and provides a new molecular biological basis for the association between anti-SSA antibodies and skin photoallergic manifestations of SLE.
Collapse
Affiliation(s)
- Dingqi Lu
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People’s Republic of China
| | - Xinchao Zhu
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People’s Republic of China
| | - Tao Hong
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People’s Republic of China
| | - Xinyi Yao
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People’s Republic of China
| | - Zhiming Xie
- The Second Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People’s Republic of China
| | - Liying Chen
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People’s Republic of China
| | - Yihan Wang
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People’s Republic of China
| | - Kaiyuan Zhang
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People’s Republic of China
| | - Yating Ren
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People’s Republic of China
| | - Yi Cao
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People’s Republic of China
| | - Xinchang Wang
- The Second Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People’s Republic of China
| |
Collapse
|
8
|
Gamal-Eldeen AM, Fahmy CA, Raafat BM, Althobaiti F, Bassyouni IH, Talaat RM. Association of Circulating Levels of Hypoxia-Inducible Factor-1α and miR-210 with Photosensitivity in Systemic Lupus Erythematosus Patients. Curr Mol Med 2023; 23:185-192. [PMID: 35034594 DOI: 10.2174/1566524022666220114145220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/11/2021] [Accepted: 11/25/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND miR-210, a key hypoxamiR, regulates hypoxia and inflammation-linked hypoxia. Systemic lupus erythematosus (SLE), a chronic autoimmune disease, is responsible for many pathological disorders, including photosensitivity. OBJECTIVE This study aimed to find the correlation between circulating miR-210/HIF-1α levels and photosensitivity in SLE patients and other SLE-associated pathological complications in a single-center case-control study. METHODS The study population comprised 104 SLE Egyptian patients with photosensitivity, 32 SLE patients without photosensitivity, and 32 healthy subjects. SLE activity was assessed for all patients using the SLE Disease Activity Index (SLEDAI). Clinical complications/manifestations and hematological/serological analyses were recorded. HIF-α concentration was investigated by ELISA, and miR-210 expression was analyzed by qRT-PCR. RESULTS The results revealed that circulating miR-210 was significantly increased in the SLE/photosensitivity group versus the SLE and control groups. The additional occurrence of malar rash, oral ulcers, renal disorders, or hypertension resulted in a higher expression of miR-210. SLEDAI activity status showed no effect on miR-210. Erythrocyte sedimentation rate, white blood cells, hemoglobin, platelets, patient age, and disease duration were positively correlated with circulatory miR-210. HIF-α concentration was significantly induced in the SLE/photosensitivity group versus the SLE and control groups. In SLE/photosensitivity, the presence of renal disorders and hypertension resulted in the highest HIF-α concentrations. A strong positive correlation was recorded between HIF-α concentration and circulatory miR-210 in SLE/photosensitivity patients (r = 0.886). CONCLUSION The dysregulation of circulating miR-210/HIF-1α levels in SLE/ photosensitivity patients is controlled by the presence of additional pathological complications, and results suggest that the hypoxia pathway might interact positively with the pathogenesis and disease progression of SLE.
Collapse
Affiliation(s)
- Amira M Gamal-Eldeen
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
- High Altitude Research Center, Prince Sultan Medical Complex, Al- Hawiyah, Taif University, Taif, Saudi Arabia
| | - Cinderella A Fahmy
- Cancer Biology and Genetics Laboratory, Centre of Excellence for Advanced Sciences, National Research Centre, 33 El Buhouth St. Dokki, Cairo, 12622, Egypt
- Biochemistry Department, National Research Centre, 33 El Buhouth St. Dokki, Cairo, 12622, Egypt
| | - Bassem M Raafat
- Radiological Sciences Department, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Fayez Althobaiti
- High Altitude Research Center, Prince Sultan Medical Complex, Al- Hawiyah, Taif University, Taif, Saudi Arabia
- Biotechnology Department, Faculty of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Iman H Bassyouni
- Department of Rheumatology and Rehabilitation, Faculty of Medicine, Cairo University, El-Kasr El-Aini Hospital, Cairo 12613, Egypt
| | - Roba M Talaat
- Molecular Biology Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), Sadat City University, Egypt
| |
Collapse
|
9
|
Munir H, Lu TT. T2B or not to B: Calming neutrophils offshore. J Exp Med 2021; 218:e20211407. [PMID: 34374712 PMCID: PMC8357534 DOI: 10.1084/jem.20211407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
In this issue of JEM, Podstawka et al. (2021. J. Exp. Med.https://doi.org/10.1084/jem.20210409) show that B cells can limit neutrophil responses within the lung microvasculature by marginating and acting on marginated neutrophils. This study provides a new view of B cells and reveals a novel mechanism of cell-mediated intravascular regulation.
Collapse
Affiliation(s)
- Hafsa Munir
- Hospital for Special Surgery Research Institute, New York, NY
- Weill Cornell Medicine, New York, NY
| | - Theresa T. Lu
- Hospital for Special Surgery Research Institute, New York, NY
- Weill Cornell Medicine, New York, NY
| |
Collapse
|
10
|
Honan AM, Chen Z. Stromal Cells Underlining the Paths From Autoimmunity, Inflammation to Cancer With Roles Beyond Structural and Nutritional Support. Front Cell Dev Biol 2021; 9:658984. [PMID: 34113615 PMCID: PMC8185233 DOI: 10.3389/fcell.2021.658984] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/29/2021] [Indexed: 12/14/2022] Open
Abstract
Stromal cells provide structural support and nutrients in secondary lymphoid organs and non-lymphoid tissues. However, accumulating evidence suggests that a complex relationship exists between stromal cells and immune cells. Interactions between immune cells and stromal cells have been shown to influence the pathology of both autoimmunity and cancer. This review examines the heterogeneity of stromal cells within the lymph node and non-lymphoid tissues during both homeostatic and inflammatory conditions, in particular autoimmunity and cancer, with the goal of better understanding the complex and apparently paradoxical relationship between these two classes of diseases. The review surveys potential novel mechanisms involving the interactions between stromal cells and immune cells which may contribute to the development, pathology and underlying connection between autoimmunity and cancer, including potential pathways from autoimmune inflammation to either “hot” or “cold” tumors. These interactions may provide some insights to explain the rising incidence of both autoimmunity and cancer in young women in industrialized countries and have the potential to be exploited in the development of new interventions for preventions and treatments of both autoimmune diseases and cancer.
Collapse
Affiliation(s)
- Amanda M Honan
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Zhibin Chen
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States.,Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
11
|
Oltz EM, Schwab S. Stromal Immunology: Frameworks for Development and Response. THE JOURNAL OF IMMUNOLOGY 2021; 206:241-242. [PMID: 33397735 DOI: 10.4049/jimmunol.2090022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|