1
|
Hong W, Ma H, Yang Z, Wang J, Peng B, Wang L, Du Y, Yang L, Zhang L, Li Z, Huang H, Zhu D, Yang B, He Q, Wang J, Weng Q. Optineurin restrains CCR7 degradation to guide type II collagen-stimulated dendritic cell migration in rheumatoid arthritis. Acta Pharm Sin B 2025; 15:1626-1642. [PMID: 40370566 PMCID: PMC12069250 DOI: 10.1016/j.apsb.2025.02.004] [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: 05/07/2024] [Revised: 07/28/2024] [Accepted: 12/19/2024] [Indexed: 05/16/2025] Open
Abstract
Dendritic cells (DCs) serve as the primary antigen-presenting cells in autoimmune diseases, like rheumatoid arthritis (RA), and exhibit distinct signaling profiles due to antigenic diversity. Type II collagen (CII) has been recognized as an RA-specific antigen; however, little is known about CII-stimulated DCs, limiting the development of RA-specific therapeutic interventions. In this study, we show that CII-stimulated DCs display a preferential gene expression profile associated with migration, offering a new perspective for targeting DC migration in RA treatment. Then, saikosaponin D (SSD) was identified as a compound capable of blocking CII-induced DC migration and effectively ameliorating arthritis. Optineurin (OPTN) is further revealed as a potential SSD target, with Optn deletion impairing CII-pulsed DC migration without affecting maturation. Function analyses uncover that OPTN prevents the proteasomal transport and ubiquitin-dependent degradation of C-C chemokine receptor 7 (CCR7), a pivotal chemokine receptor in DC migration. Optn-deficient DCs exhibit reduced CCR7 expression, leading to slower migration in CII-surrounded environment, thus alleviating arthritis progression. Our findings underscore the significance of antigen-specific DC activation in RA and suggest OPTN is a crucial regulator of CII-specific DC migration. OPTN emerges as a promising drug target for RA, potentially offering significant value for the therapeutic management of RA.
Collapse
Affiliation(s)
- Wenxiang Hong
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Nanhu Brain-Computer Interface Institute, Hangzhou 311100, China
| | - Hongbo Ma
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhaoxu Yang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiaying Wang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Bowen Peng
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Longling Wang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yiwen Du
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lijun Yang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Drug Safety Evaluation and Research of Zhejiang Province, Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou 310059, China
| | - Lijiang Zhang
- Key Laboratory of Drug Safety Evaluation and Research of Zhejiang Province, Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou 310059, China
| | - Zhibin Li
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310018, China
| | - Han Huang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Difeng Zhu
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Bo Yang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- School of Medicine, Hangzhou City University, Hangzhou 310015, China
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Jiajia Wang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Nanhu Brain-Computer Interface Institute, Hangzhou 311100, China
| | - Qinjie Weng
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Nanhu Brain-Computer Interface Institute, Hangzhou 311100, China
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Taizhou Institute of Zhejiang University, Taizhou 318000, China
| |
Collapse
|
2
|
Chen W, Miao J, Chen J, Chen J. Development of machine learning models for diagnostic biomarker identification and immune cell infiltration analysis in PCOS. J Ovarian Res 2025; 18:1. [PMID: 39754246 PMCID: PMC11697806 DOI: 10.1186/s13048-024-01583-1] [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: 06/15/2024] [Accepted: 12/22/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a common endocrine disorder affecting women of reproductive age. It is characterized by symptoms such as hyperandrogenemia, oligo or anovulation and polycystic ovarian, significantly impacting quality of life. However, the practical implementation of machine learning (ML) in PCOS diagnosis is hindered by the limitations related to data size and algorithmic models. To address this research gap, we have increased the sample size in our study and aim to utilize two ML algorithms to analyze and validate diagnostic biomarkers, as well as explore immune cell infiltration patterns in PCOS. METHODS We performed RNA-seq analysis on granulosa cell, including 13 samples from normal controls and 25 samples from women with PCOS. The data from our study were combined with publicly available databases. Batch effects were corrected using the 'sva' package in R software. Differential expression analysis was performed to identify genes that exhibited significant differences between the two groups. These differentially expressed genes (DEGs) were further analyzed for Gene Ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Hub genes were selected by intersecting the results of both methods after using LASSO and SVM-RFE for central gene selection for DEGs. Receiver Operating Characteristic (ROC) curves were employed to verify the accuracy of models by SVM and XGBoost. CIBERSORT analysis was performed to determine the relative abundances of immune cell populations. GSEA was analyzed to illustrate the expression patterns of genes within highly enriched functional pathways. RT-qPCR was used to validate the reliability of hub genes. RESULTS 824 DEGs were found between the normal control and PCOS groups, including 376 upregulated and 448 downregulated genes. These DEGs were associated with endocytosis, salmonella infection and focal adhesion based on the KEGG enrichment analysis. Through overlapping LASSO and SVM-RFE algorithms, we identified four hub genes (CNTN2, CASR, CACNB3, MFAP2) that are significantly associated with the PCOS group. The diagnostic efficacy validation set using SVM and XGBoost yielded AUC values of 0.795 and 0.875, respectively, indicating their potential as diagnostic biomarkers. Consistent with the data analysis, the upregulation of CNTN2, CASR, CACNB3, and MFAP2 in PCOS was confirmed by RT-qPCR analysis on human granulosa cells. Furthermore, according to CIBERSORT analysis, a significant reduction in CD4 memory resting T cells was revealed in the PCOS group compared to the normal control group (P < 0.05). CONCLUSIONS This study identified CNTN2, CASR, CACNB3, and MFAP2 as potential diagnostic biomarkers for PCOS, which provides strong evidence for existing research on hub genes. Furthermore, the analysis of immune cell infiltration revealed the significant involvement of CD4 memory resting T cells in the onset and progression of PCOS. These findings shed light on potential mechanisms underlying PCOS pathogenesis and provide valuable insights for future research and therapeutic interventions.
Collapse
Affiliation(s)
- Wenxiu Chen
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jianliang Miao
- First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Jingfei Chen
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Jianlin Chen
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
3
|
Gonzales GA, Huang S, Wilkinson L, Nguyen JA, Sikdar S, Piot C, Naumenko V, Rajwani J, Wood CM, Dinh I, Moore M, Cedeño E, McKenna N, Polyak MJ, Amidian S, Ebacher V, Rosin NL, Carneiro MB, Surewaard B, Peters NC, Mody CH, Biernaskie J, Yates RM, Mahoney DJ, Canton J. The pore-forming apolipoprotein APOL7C drives phagosomal rupture and antigen cross-presentation by dendritic cells. Sci Immunol 2024; 9:eadn2168. [PMID: 39485861 DOI: 10.1126/sciimmunol.adn2168] [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: 11/28/2023] [Revised: 07/23/2024] [Accepted: 10/07/2024] [Indexed: 11/03/2024]
Abstract
Conventional dendritic cells (cDCs) generate protective cytotoxic T lymphocyte (CTL) responses against extracellular pathogens and tumors. This is achieved through a process known as cross-presentation (XP), and, despite its biological importance, the mechanism(s) driving XP remains unclear. Here, we show that a cDC-specific pore-forming protein called apolipoprotein L 7C (APOL7C) is up-regulated in response to innate immune stimuli and is recruited to phagosomes. Association of APOL7C with phagosomes led to phagosomal rupture and escape of engulfed antigens to the cytosol, where they could be processed via the endogenous MHC class I antigen processing pathway. Accordingly, mice deficient in APOL7C did not efficiently prime CD8+ T cells in response to immunization with bead-bound and cell-associated antigens. Together, our data indicate the presence of dedicated apolipoproteins that mediate the delivery of phagocytosed proteins to the cytosol of activated cDCs to facilitate XP.
Collapse
Affiliation(s)
- Gerone A Gonzales
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Song Huang
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Liam Wilkinson
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jenny A Nguyen
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Saif Sikdar
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Research Institute, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, Alberta, Canada
| | - Cécile Piot
- Immunobiology Laboratory, Francis Crick Institute, London, UK
| | - Victor Naumenko
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, Alberta, Canada
- Riddell Centre for Cancer Immunotherapy, Calgary, Alberta, Canada
| | - Jahanara Rajwani
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Research Institute, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, Alberta, Canada
| | - Cassandra M Wood
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Irene Dinh
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Melanie Moore
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Eymi Cedeño
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Neil McKenna
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Maria J Polyak
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Calvin, Joan and Phoebe Snyder Institute for Chronic Disease, Calgary, Alberta, Canada
| | - Sara Amidian
- Cell Imaging Core, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | | | - Nicole L Rosin
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Matheus B Carneiro
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Calvin, Joan and Phoebe Snyder Institute for Chronic Disease, Calgary, Alberta, Canada
| | - Bas Surewaard
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Calvin, Joan and Phoebe Snyder Institute for Chronic Disease, Calgary, Alberta, Canada
| | - Nathan C Peters
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Calvin, Joan and Phoebe Snyder Institute for Chronic Disease, Calgary, Alberta, Canada
| | - Christopher H Mody
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Calvin, Joan and Phoebe Snyder Institute for Chronic Disease, Calgary, Alberta, Canada
| | - Jeff Biernaskie
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Robin M Yates
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
- Calvin, Joan and Phoebe Snyder Institute for Chronic Disease, Calgary, Alberta, Canada
| | - Douglas J Mahoney
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Research Institute, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, Alberta, Canada
- Riddell Centre for Cancer Immunotherapy, Calgary, Alberta, Canada
| | - Johnathan Canton
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Riddell Centre for Cancer Immunotherapy, Calgary, Alberta, Canada
- Calvin, Joan and Phoebe Snyder Institute for Chronic Disease, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, Calgary, Alberta, Canada
| |
Collapse
|
4
|
Okada N, Oshima K, Maruko A, Sekine M, Ito N, Wakasugi A, Mori E, Odaguchi H, Kobayashi Y. Intron retention as an excellent marker for diagnosing depression and for discovering new potential pathways for drug intervention. Front Psychiatry 2024; 15:1450708. [PMID: 39364384 PMCID: PMC11446786 DOI: 10.3389/fpsyt.2024.1450708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/20/2024] [Indexed: 10/05/2024] Open
Abstract
Background Peripheral inflammation is often associated with depressive disorders, and immunological biomarkers of depression remain a focus of investigation. Methods We performed RNA-seq analysis of RNA transcripts of human peripheral blood mononuclear cells from a case-control study including subjects with self-reported depression in the pre-symptomatic state of major depressive disorder and analyzed differentially expressed genes (DEGs) and the frequency of intron retention (IR) using rMATS. Results Among the statistically significant DEGs identified, the 651 upregulated DEGs were particularly enriched in the term "bacterial infection and phagocytosis", whereas the 820 downregulated DEGs were enriched in the terms "antigen presentation" and "T-cell proliferation and maturation". We also analyzed 158 genes for which the IR was increased (IncIR) and 211 genes for which the IR was decreased (DecIR) in the depressed subjects. Although the Gene Ontology terms associated with IncIR and DecIR were very similar to those of the up- and downregulated genes, respectively, IR genes appeared to be particularly enriched in genes with sensor functions, with a preponderance of the term "ciliary assembly and function". The observation that IR genes specifically interact with innate immunity genes suggests that immune-related genes, as well as cilia-related genes, may be excellent markers of depression. Re-analysis of previously published RNA-seq data from patients with MDD showed that common IR genes, particularly our predicted immune- and cilia-related genes, are commonly detected in populations with different levels of depression, providing validity for using IR to detect depression. Conclusion Depression was found to be associated with activation of the innate immune response and relative inactivation of T-cell signaling. The DEGs we identified reflect physiological demands that are controlled at the transcriptional level, whereas the IR results reflect a more direct mechanism for monitoring protein homeostasis. Accordingly, an alteration in IR, namely IncIR or DecIR, is a stress response, and intron-retained transcripts are sensors of the physiological state of the cytoplasm. The results demonstrate the potential of relative IR as a biomarker for the immunological stratification of depressed patients and the utility of IR for the discovery of novel pathways involved in recovery from depression.
Collapse
Affiliation(s)
- Norihiro Okada
- School of Pharmacy, Kitasato University, Minato-ku, Tokyo, Japan
| | - Kenshiro Oshima
- School of Pharmacy, Kitasato University, Minato-ku, Tokyo, Japan
| | - Akiko Maruko
- School of Pharmacy, Kitasato University, Minato-ku, Tokyo, Japan
| | - Mariko Sekine
- Kitasato University Kitasato Institute Hospital, Minato-ku, Tokyo, Japan
- Oriental Medicine Research Center, School of Pharmacy, Kitasato University, Minato-ku, Tokyo, Japan
| | - Naoki Ito
- Oriental Medicine Research Center, School of Pharmacy, Kitasato University, Minato-ku, Tokyo, Japan
| | - Akino Wakasugi
- Kitasato University Kitasato Institute Hospital, Minato-ku, Tokyo, Japan
- Oriental Medicine Research Center, School of Pharmacy, Kitasato University, Minato-ku, Tokyo, Japan
| | - Eiko Mori
- Oriental Medicine Research Center, School of Pharmacy, Kitasato University, Minato-ku, Tokyo, Japan
| | - Hiroshi Odaguchi
- Oriental Medicine Research Center, School of Pharmacy, Kitasato University, Minato-ku, Tokyo, Japan
| | - Yoshinori Kobayashi
- School of Pharmacy, Kitasato University, Minato-ku, Tokyo, Japan
- Oriental Medicine Research Center, School of Pharmacy, Kitasato University, Minato-ku, Tokyo, Japan
| |
Collapse
|
5
|
Martus D, Williams SK, Pichi K, Mannebach-Götz S, Kaiser N, Wardas B, Fecher-Trost C, Meyer MR, Schmitz F, Beck A, Fairless R, Diem R, Flockerzi V, Belkacemi A. Cavβ3 Contributes to the Maintenance of the Blood-Brain Barrier and Alleviates Symptoms of Experimental Autoimmune Encephalomyelitis. Arterioscler Thromb Vasc Biol 2024; 44:1833-1851. [PMID: 38957986 DOI: 10.1161/atvbaha.124.321141] [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: 04/23/2024] [Accepted: 06/21/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND Tight control of cytoplasmic Ca2+ concentration in endothelial cells is essential for the regulation of endothelial barrier function. Here, we investigated the role of Cavβ3, a subunit of voltage-gated Ca2+ (Cav) channels, in modulating Ca2+ signaling in brain microvascular endothelial cells (BMECs) and how this contributes to the integrity of the blood-brain barrier. METHODS We investigated the function of Cavβ3 in BMECs by Ca2+ imaging and Western blot, examined the endothelial barrier function in vitro and the integrity of the blood-brain barrier in vivo, and evaluated disease course after induction of experimental autoimmune encephalomyelitis in mice using Cavβ3-/- (Cavβ3-deficient) mice as controls. RESULTS We identified Cavβ3 protein in BMECs, but electrophysiological recordings did not reveal significant Cav channel activity. In vivo, blood-brain barrier integrity was reduced in the absence of Cavβ3. After induction of experimental autoimmune encephalomyelitis, Cavβ3-/- mice showed earlier disease onset with exacerbated clinical disability and increased T-cell infiltration. In vitro, the transendothelial resistance of Cavβ3-/- BMEC monolayers was lower than that of wild-type BMEC monolayers, and the organization of the junctional protein ZO-1 (zona occludens-1) was impaired. Thrombin stimulates inositol 1,4,5-trisphosphate-dependent Ca2+ release, which facilitates cell contraction and enhances endothelial barrier permeability via Ca2+-dependent phosphorylation of MLC (myosin light chain). These effects were more pronounced in Cavβ3-/- than in wild-type BMECs, whereas the differences were abolished in the presence of the MLCK (MLC kinase) inhibitor ML-7. Expression of Cacnb3 cDNA in Cavβ3-/- BMECs restored the wild-type phenotype. Coimmunoprecipitation and mass spectrometry demonstrated the association of Cavβ3 with inositol 1,4,5-trisphosphate receptor proteins. CONCLUSIONS Independent of its function as a subunit of Cav channels, Cavβ3 interacts with the inositol 1,4,5-trisphosphate receptor and is involved in the tight control of cytoplasmic Ca2+ concentration and Ca2+-dependent MLC phosphorylation in BMECs, and this role of Cavβ3 in BMECs contributes to blood-brain barrier integrity and attenuates the severity of experimental autoimmune encephalomyelitis disease.
Collapse
MESH Headings
- Animals
- Female
- Male
- Mice
- Blood-Brain Barrier/metabolism
- Calcium/metabolism
- Calcium Channels/metabolism
- Calcium Channels/genetics
- Calcium Signaling
- Capillary Permeability
- Cells, Cultured
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Endothelial Cells/metabolism
- Inositol 1,4,5-Trisphosphate Receptors/metabolism
- Inositol 1,4,5-Trisphosphate Receptors/genetics
- Mice, Inbred C57BL
- Mice, Knockout
- Myosin Light Chains/metabolism
- Myosin-Light-Chain Kinase/metabolism
- Myosin-Light-Chain Kinase/genetics
- Phosphorylation
Collapse
Affiliation(s)
- Damian Martus
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Sarah K Williams
- Neurologische Klinik, Universitätsklinikum Heidelberg, Germany (S.K.W., K.P., R.F., R.D.)
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany (R.F., S.K.W.)
| | - Kira Pichi
- Neurologische Klinik, Universitätsklinikum Heidelberg, Germany (S.K.W., K.P., R.F., R.D.)
| | - Stefanie Mannebach-Götz
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Nicolas Kaiser
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Barbara Wardas
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Claudia Fecher-Trost
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Markus R Meyer
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Frank Schmitz
- Institut für Anatomie und Zellbiologie (F.S.), Universität des Saarlandes, Homburg, Germany
| | - Andreas Beck
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Richard Fairless
- Neurologische Klinik, Universitätsklinikum Heidelberg, Germany (S.K.W., K.P., R.F., R.D.)
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany (R.F., S.K.W.)
| | - Ricarda Diem
- Neurologische Klinik, Universitätsklinikum Heidelberg, Germany (S.K.W., K.P., R.F., R.D.)
| | - Veit Flockerzi
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Anouar Belkacemi
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
- Now with Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, Germany (A. Belkacemi)
| |
Collapse
|
6
|
Woo MS, Mayer C, Binkle-Ladisch L, Sonner JK, Rosenkranz SC, Shaposhnykov A, Rothammer N, Tsvilovskyy V, Lorenz SM, Raich L, Bal LC, Vieira V, Wagner I, Bauer S, Glatzel M, Conrad M, Merkler D, Freichel M, Friese MA. STING orchestrates the neuronal inflammatory stress response in multiple sclerosis. Cell 2024; 187:4043-4060.e30. [PMID: 38878778 DOI: 10.1016/j.cell.2024.05.031] [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: 01/16/2024] [Revised: 04/08/2024] [Accepted: 05/16/2024] [Indexed: 07/28/2024]
Abstract
Inflammation-induced neurodegeneration is a defining feature of multiple sclerosis (MS), yet the underlying mechanisms remain unclear. By dissecting the neuronal inflammatory stress response, we discovered that neurons in MS and its mouse model induce the stimulator of interferon genes (STING). However, activation of neuronal STING requires its detachment from the stromal interaction molecule 1 (STIM1), a process triggered by glutamate excitotoxicity. This detachment initiates non-canonical STING signaling, which leads to autophagic degradation of glutathione peroxidase 4 (GPX4), essential for neuronal redox homeostasis and thereby inducing ferroptosis. Both genetic and pharmacological interventions that target STING in neurons protect against inflammation-induced neurodegeneration. Our findings position STING as a central regulator of the detrimental neuronal inflammatory stress response, integrating inflammation with glutamate signaling to cause neuronal cell death, and present it as a tractable target for treating neurodegeneration in MS.
Collapse
Affiliation(s)
- Marcel S Woo
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christina Mayer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lars Binkle-Ladisch
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jana K Sonner
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sina C Rosenkranz
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Artem Shaposhnykov
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola Rothammer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Volodymyr Tsvilovskyy
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Svenja M Lorenz
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | - Lukas Raich
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lukas C Bal
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Vanessa Vieira
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ingrid Wagner
- Department of Pathology and Immunology, Division of Clinical Pathology, Faculty of Medicine, University and University Hospital of Geneva, Geneva, Switzerland
| | - Simone Bauer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | - Doron Merkler
- Department of Pathology and Immunology, Division of Clinical Pathology, Faculty of Medicine, University and University Hospital of Geneva, Geneva, Switzerland
| | - Marc Freichel
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
7
|
Su C, Kent CL, Pierpoint M, Floyd W, Luo L, Williams NT, Ma Y, Peng B, Lazarides AL, Subramanian A, Himes JE, Perez VM, Hernansaiz-Ballesteros RD, Roche KE, Modliszewski JL, Selitsky SR, Shinohara ML, Wisdom AJ, Moding EJ, Mowery YM, Kirsch DG. Enhancing radiotherapy response via intratumoral injection of a TLR9 agonist in autochthonous murine sarcomas. JCI Insight 2024; 9:e178767. [PMID: 39133651 PMCID: PMC11383182 DOI: 10.1172/jci.insight.178767] [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: 01/03/2024] [Accepted: 06/11/2024] [Indexed: 08/21/2024] Open
Abstract
Radiation therapy (RT) is frequently used to treat cancers, including soft-tissue sarcomas. Prior studies established that the toll-like receptor 9 (TLR9) agonist cytosine-phosphate-guanine oligodeoxynucleotide (CpG) enhances the response to RT in transplanted tumors, but the mechanisms of this enhancement remain unclear. Here, we used CRISPR/Cas9 and the chemical carcinogen 3-methylcholanthrene (MCA) to generate autochthonous soft-tissue sarcomas with high tumor mutation burden. Treatment with a single fraction of 20 Gy RT and 2 doses of CpG significantly enhanced tumor response, which was abrogated by genetic or immunodepletion of CD8+ T cells. To characterize the immune response to CpG+RT, we performed bulk RNA-Seq, single-cell RNA-Seq, and mass cytometry. Sarcomas treated with 20 Gy and CpG demonstrated increased CD8 T cells expressing markers associated with activation and proliferation, such as Granzyme B, Ki-67, and IFN-γ. CpG+RT also upregulated antigen presentation pathways on myeloid cells. Furthermore, in sarcomas treated with CpG+RT, TCR clonality analysis suggests an increase in clonal T cell dominance. Collectively, these findings demonstrate that CpG+RT significantly delays tumor growth in a CD8 T cell-dependent manner. These results provide a strong rationale for clinical trials evaluating CpG or other TLR9 agonists with RT in patients with soft-tissue sarcoma.
Collapse
Affiliation(s)
- Chang Su
- Department of Pharmacology and Cancer Biology and
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Collin L Kent
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Matthew Pierpoint
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Warren Floyd
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Lixia Luo
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Nerissa T Williams
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Yan Ma
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Brian Peng
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Alexander L Lazarides
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Ajay Subramanian
- Department of Radiation Oncology, Stanford University, Stanford, California, USA
| | - Jonathon E Himes
- Department of Pharmacology and Cancer Biology and
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | | | | | - Kimberly E Roche
- Tempus AI Inc., Durham, North Carolina, USA
- QuantBio LLC, Durham, North Carolina, USA
| | - Jennifer L Modliszewski
- QuantBio LLC, Durham, North Carolina, USA
- Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA
| | - Sara R Selitsky
- Tempus AI Inc., Durham, North Carolina, USA
- QuantBio LLC, Durham, North Carolina, USA
| | - Mari L Shinohara
- Department of Integrative Immunology
- Department of Molecular Genetics and Microbiology, and
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Amy J Wisdom
- Harvard Radiation Oncology Program, Boston, Massachusetts, USA
| | - Everett J Moding
- Department of Radiation Oncology, Stanford University, Stanford, California, USA
- Stanford Cancer Institute, Stanford University, Stanford, California, USA
| | - Yvonne M Mowery
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - David G Kirsch
- Department of Pharmacology and Cancer Biology and
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Radiation Oncology and
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
8
|
Tang Y, Dou S, Wei C, Sun Z, Sun D, Zhou Q, Xie L. Single-Nuclei Characterization of Lacrimal Gland in Scopolamine-Induced Dry Eye Disease. Invest Ophthalmol Vis Sci 2024; 65:46. [PMID: 38687491 PMCID: PMC11067549 DOI: 10.1167/iovs.65.4.46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/26/2024] [Indexed: 05/02/2024] Open
Abstract
Purpose The lacrimal gland (LG) is the main organ responsible for tear secretion and an important pathogenic site for dry eye disease (DED). This study aimed to comprehensively characterize LG cellular heterogeneity under normal and DED conditions using single-nucleus RNA sequencing (snRNA-seq). Methods Single LG nuclei isolated from mice with or without DED induced by scopolamine (SCOP)/desiccating stress (DS) were subjected to snRNA-seq using the 10x Genomics platform. These cells were clustered and annotated using the t-distributed stochastic neighbor embedding (t-SNE) method and unbiased computational informatic analysis. Cluster identification and functional analysis were performed based on marker gene expression and bioinformatic data mining. Results The snRNA-seq analysis of 30,351 nuclei identified eight major cell types, with acinar cells (∼72.6%) being the most abundant cell type in the LG. Subclustering analysis revealed that the LG mainly contained two acinar cell subtypes, two ductal cell subclusters, three myoepithelial cell (MECs) subtypes, and four immunocyte subclusters. In the SCOP-induced DED model, three major LG parenchymal cell types were significantly altered, characterized by a reduced proportion of acinar cells with a lowered secretion potential and an augmented proportion of ductal cells and MECs. LG immunocytes in DED scenarios showed an intensified inflammatory response and dysregulated intercellular communication with three major LG parenchymal cells. Conclusions Overall, this study offers a systemic single-nucleus transcriptomic profile of LGs in both normal and DED conditions and an atlas of the complicated interactions of immunocytes with major LG parenchymal cells. The findings also facilitate understanding the pathogenesis of DED.
Collapse
Affiliation(s)
- Yang Tang
- Shandong First Medical University, Jinan, China
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute of Shandong First Medical University, Qingdao, China
| | - Shengqian Dou
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute of Shandong First Medical University, Qingdao, China
| | - Chao Wei
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute of Shandong First Medical University, Qingdao, China
| | - Ziwen Sun
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Di Sun
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute of Shandong First Medical University, Qingdao, China
| | - Qingjun Zhou
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute of Shandong First Medical University, Qingdao, China
| | - Lixin Xie
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute of Shandong First Medical University, Qingdao, China
| |
Collapse
|
9
|
Su C, Kent CL, Pierpoint M, Floyd W, Luo L, Wiliams NT, Ma Y, Peng B, Lazarides AL, Subramanian A, Himes JE, Perez VM, Hernansaiz-Ballesteros RD, Roche KE, Modliszewski JL, Selitsky SR, Mari Shinohara, Wisdom AJ, Moding EJ, Mowery YM, Kirsch DG. Enhancing radiotherapy response via intratumoral injection of the TLR9 agonist CpG to stimulate CD8 T cells in an autochthonous mouse model of sarcoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.03.573968. [PMID: 38260522 PMCID: PMC10802286 DOI: 10.1101/2024.01.03.573968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Radiation therapy is frequently used to treat cancers including soft tissue sarcomas. Prior studies established that the toll-like receptor 9 (TLR9) agonist cytosine-phosphate-guanine oligodeoxynucleotide (CpG) enhances the response to radiation therapy (RT) in transplanted tumors, but the mechanism(s) remain unclear. Here, we used CRISPR/Cas9 and the chemical carcinogen 3-methylcholanthrene (MCA) to generate autochthonous soft tissue sarcomas with high tumor mutation burden. Treatment with a single fraction of 20 Gy RT and two doses of CpG significantly enhanced tumor response, which was abrogated by genetic or immunodepletion of CD8+ T cells. To characterize the immune response to RT + CpG, we performed bulk RNA-seq, single-cell RNA-seq, and mass cytometry. Sarcomas treated with 20 Gy and CpG demonstrated increased CD8 T cells expressing markers associated with activation and proliferation, such as Granzyme B, Ki-67, and interferon-γ. CpG + RT also upregulated antigen presentation pathways on myeloid cells. Furthermore, in sarcomas treated with CpG + RT, TCR clonality analysis suggests an increase in clonal T-cell dominance. Collectively, these findings demonstrate that RT + CpG significantly delays tumor growth in a CD8 T cell-dependent manner. These results provide a strong rationale for clinical trials evaluating CpG or other TLR9 agonists with RT in patients with soft tissue sarcoma.
Collapse
Affiliation(s)
- Chang Su
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Collin L. Kent
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Matthew Pierpoint
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | | | - Lixia Luo
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Nerissa T. Wiliams
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Yan Ma
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Brian Peng
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | | | - Ajay Subramanian
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Jonathan E. Himes
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | | | | | | | | | | | - Mari Shinohara
- Department of Immunology, Duke University, Durham, NC, USA
| | - Amy J. Wisdom
- Department of Radiation Oncology, Harvard University, Cambridge, MA, USA
| | - Everett J. Moding
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Yvonne M. Mowery
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
- MD Anderson Cancer Center, Houston, TX, USA
- Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - David G. Kirsch
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| |
Collapse
|