51
|
Jing Y, Kobayashi M, Shoulkamy MI, Zhou M, Thi Vu H, Arakawa H, Sabit H, Iwabuchi S, Quang Vu C, Kasahara A, Ueno M, Tadokoro Y, Kurayoshi K, Chen X, Yan Y, Arai S, Hashimoto S, Soga T, Todo T, Nakada M, Hirao A. Lysine-arginine imbalance overcomes therapeutic tolerance governed by the transcription factor E3-lysosome axis in glioblastoma. Nat Commun 2025; 16:2876. [PMID: 40169552 PMCID: PMC11962137 DOI: 10.1038/s41467-025-56946-z] [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: 12/14/2023] [Accepted: 02/06/2025] [Indexed: 04/03/2025] Open
Abstract
Recent advances in cancer therapy have underscored the importance of targeting specific metabolic pathways. In this study, we propose a precision nutrition approach aimed at lysosomal function in glioblastoma multiforme (GBM). Using patient-derived GBM cells, we identify lysosomal activity as a unique metabolic biomarker of tumorigenesis, controlling the efficacy of temozolomide (TMZ), a standard GBM therapy. Employing combined analyses of clinical patient samples and xenograft models, we further elucidate the pivotal role of Transcription Factor Binding To IGHM Enhancer 3 (TFE3), a master regulator of lysosomal biogenesis, in modulating malignant properties, particularly TMZ tolerance, by regulating peroxisome proliferator-activated receptor-gamma coactivator 1-alpha (PGC1α)-mediated mitochondrial activity. Notably, we find that lysine protects GBM cells from lysosomal stress by counteracting arginine's effects on nitric oxide production. The lysine restriction mimetic, homoarginine administration, significantly enhances the efficacy of anticancer therapies through lysosomal dysfunction. This study underscores the critical role of lysosomal function modulated by amino acid metabolism in GBM pathogenesis and treatment.
Collapse
Affiliation(s)
- Yongwei Jing
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Masahiko Kobayashi
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Mahmoud I Shoulkamy
- WPI Nano Life Science Institute (WPI-Nano LSI), Kanazawa University, Kanazawa, Ishikawa, Japan
- Department of Zoology, Faculty of Science, Minia University, Minia, Egypt
| | - Meiqi Zhou
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Ha Thi Vu
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan
- Department of Medical Biology and Genetics, Hanoi Medical University, Ha Noi, Vietnam
| | - Hiroshi Arakawa
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Hemragul Sabit
- Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Sadahiro Iwabuchi
- Department of Molecular Pathophysiology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Cong Quang Vu
- WPI Nano Life Science Institute (WPI-Nano LSI), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Atsuko Kasahara
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan
- WPI Nano Life Science Institute (WPI-Nano LSI), Kanazawa University, Kanazawa, Ishikawa, Japan
- Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Masaya Ueno
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan
- WPI Nano Life Science Institute (WPI-Nano LSI), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Yuko Tadokoro
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan
- WPI Nano Life Science Institute (WPI-Nano LSI), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Kenta Kurayoshi
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Xi Chen
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Yuhang Yan
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Satoshi Arai
- WPI Nano Life Science Institute (WPI-Nano LSI), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Shinichi Hashimoto
- Department of Molecular Pathophysiology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
- Human Biology-Microbiome-Quantum Research Center (WPI-Bio2Q), Keio University, Tokyo, Japan
| | - Tomoki Todo
- Division of Innovative Cancer Therapy, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Mitsutoshi Nakada
- Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Atsushi Hirao
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan.
- WPI Nano Life Science Institute (WPI-Nano LSI), Kanazawa University, Kanazawa, Ishikawa, Japan.
| |
Collapse
|
52
|
Jia G, He P, Dai T, Goh D, Wang J, Sun M, Wee F, Li F, Lim JCT, Hao S, Liu Y, Lim TKH, Ngo NT, Tao Q, Wang W, Umar A, Nashan B, Zhang Y, Ding C, Yeong J, Liu L, Sun C. Spatial immune scoring system predicts hepatocellular carcinoma recurrence. Nature 2025; 640:1031-1041. [PMID: 40074893 DOI: 10.1038/s41586-025-08668-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: 03/18/2023] [Accepted: 01/17/2025] [Indexed: 03/14/2025]
Abstract
Given the high recurrence rates of hepatocellular carcinoma (HCC) post-resection1-3, improved early identification of patients at high risk for post-resection recurrence would help to improve patient outcomes and prioritize healthcare resources4-6. Here we observed a spatial and HCC recurrence-associated distribution of natural killer (NK) cells in the invasive front and tumour centre from 61 patients. Using extreme gradient boosting and inverse-variance weighting, we developed the tumour immune microenvironment spatial (TIMES) score based on the spatial expression patterns of five biomarkers (SPON2, ZFP36L2, ZFP36, VIM and HLA-DRB1) to predict HCC recurrence risk. The TIMES score (hazard ratio = 88.2, P < 0.001) outperformed current standard tools for patient risk stratification including the TNM and BCLC systems. We validated the model in 231 patients from five multicentred cohorts, achieving a real-world accuracy of 82.2% and specificity of 85.7%. The predictive power of these biomarkers emerged through the integration of their spatial distributions, rather than individual marker expression levels alone. In vivo models, including NK cell-specific Spon2-knockout mice, revealed that SPON2 enhances IFNγ secretion and NK cell infiltration at the invasive front. Our study introduces TIMES, a publicly accessible tool for predicting HCC recurrence risk, offering insights into its potential to inform treatment decisions for early-stage HCC.
Collapse
MESH Headings
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/diagnosis
- Carcinoma, Hepatocellular/surgery
- Carcinoma, Hepatocellular/genetics
- Humans
- Liver Neoplasms/immunology
- Liver Neoplasms/pathology
- Liver Neoplasms/diagnosis
- Liver Neoplasms/surgery
- Liver Neoplasms/genetics
- Killer Cells, Natural/immunology
- Killer Cells, Natural/cytology
- Neoplasm Recurrence, Local/immunology
- Neoplasm Recurrence, Local/diagnosis
- Neoplasm Recurrence, Local/pathology
- Animals
- Mice
- Male
- Tumor Microenvironment/immunology
- Female
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Biomarkers, Tumor/analysis
- Interferon-gamma/metabolism
- Mice, Knockout
- Middle Aged
- Cohort Studies
- Extracellular Matrix Proteins/genetics
- Extracellular Matrix Proteins/deficiency
- Extracellular Matrix Proteins/metabolism
- Reproducibility of Results
Collapse
Affiliation(s)
- Gengjie Jia
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Peiqi He
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Tianli Dai
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Denise Goh
- Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Jiabei Wang
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Mengyuan Sun
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Felicia Wee
- Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Fuling Li
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Jeffrey Chun Tatt Lim
- Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Shuxia Hao
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Yao Liu
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Tony Kiat Hon Lim
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Duke-NUS Medical School, Singapore, Singapore
| | | | - Qingping Tao
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Wei Wang
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Ahitsham Umar
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Björn Nashan
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
| | - Yongchang Zhang
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Central South University, Changsha, China
| | - Chen Ding
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institutes of Biomedical Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Joe Yeong
- Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Singapore.
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore.
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Singapore, Singapore.
- Cancer Science Institute, National University of Singapore, Singapore, Singapore.
| | - Lianxin Liu
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China.
| | - Cheng Sun
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China.
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
53
|
Gharaee N, Wegrzyn-Woltosz J, Jiang J, Akhade VS, Bridgers J, Stubbins RJ, Hiwase D, Kutyna MM, Chan O, Komrokji R, Padron E, Deng Y, Cole G, Umlandt P, Fuller M, Kim A, Karsan A. Haploinsufficiency of miR-143 and miR-145 reveal targetable dependencies in resistant del(5q) myelodysplastic neoplasm. Leukemia 2025; 39:917-928. [PMID: 40000845 PMCID: PMC11976265 DOI: 10.1038/s41375-025-02537-2] [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: 04/28/2024] [Revised: 12/21/2024] [Accepted: 02/11/2025] [Indexed: 02/27/2025]
Abstract
Myelodysplastic neoplasms (MDS) are stem cell disorders characterized by ineffective hematopoiesis and risk of transformation to acute myeloid leukemia (AML). Chromosomal alterations are frequent in MDS, with interstitial deletion of chromosome 5q (del(5q)) being the most common. Lenalidomide is the current first-line treatment for del(5q) MDS and its efficacy relies on degradation of CK1α which is encoded by the CSNK1A1 gene located in the commonly deleted region (CDR) of chromosome 5q. However, lenalidomide-resistance is common, often secondary to loss-of-function mutations in TP53 or RUNX1. The CDR in del(5q) harbors several genes, including noncoding miRNAs, the loss of which contribute to disease phenotypes. miR-143 and miR-145 are located within the del(5q) CDR, but precise understanding of their role in human hematopoiesis and in the pathogenesis of del(5q) MDS is lacking. Here we provide evidence that deficiency of miR-143 and miR-145 plays a role in clonal expansion of del(5q) MDS. We show that insulin-like growth factor 1 receptor (IGF-1R) is a direct target of both miR-143 and miR-145. Our data demonstrate that IGF-1R inhibition reduces proliferation and viability of del(5q) cells in vitro and in vivo, and that lenalidomide-resistant del(5q) MDS cells depleted of either TP53 or RUNX1 are sensitive to IGF-1R inhibition. Resistant del(5q) MDS-L cells, as well as primary MDS marrow cells, are also sensitive to targeting of IGF-1R-related dependencies in del(5q) MDS, which include the Abl and MAPK signaling pathways. This work thus provides potential new therapeutic avenues for lenalidomide-resistant del(5q) MDS.
Collapse
Affiliation(s)
- Nadia Gharaee
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
- Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Joanna Wegrzyn-Woltosz
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
- Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Jihong Jiang
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
- Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Vijay Suresh Akhade
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Joshua Bridgers
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Ryan J Stubbins
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
- Leukemia/BMT Program of BC, BC Cancer and Vancouver Coastal Health, Vancouver, BC, Canada
- Division of Hematology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Devendra Hiwase
- Department of Haematology, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Monika M Kutyna
- Department of Haematology, Royal Adelaide Hospital, Adelaide, SA, Australia
| | | | | | | | - Yu Deng
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
- Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Gary Cole
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Patricia Umlandt
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Megan Fuller
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Ada Kim
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Aly Karsan
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada.
- Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
54
|
Blass E, Colarusso A, Aid M, Larocca RA, Reeves RK, Barouch DH. Early spatiotemporal evolution of the immune response elicited by adenovirus serotype 26 vector vaccination in mice. J Virol 2025:e0024725. [PMID: 40162786 DOI: 10.1128/jvi.00247-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 03/11/2025] [Indexed: 04/02/2025] Open
Abstract
As the first responder to immunological challenges, the innate immune system shapes and regulates the ensuing adaptive immune response. Many clinical studies evaluating the role of innate immunity in initiating vaccine-elicited adaptive immune responses have largely been confined to blood due to the inherent difficulty in acquiring tissue samples. However, the absence of vaccine-site and draining lymph node information limits the understanding of early events induced by vaccination that could potentially shape vaccine-elicited immunity. We, therefore, utilized a mouse model to investigate the spatiotemporal evolution of the immune response within the first 24 hours following intramuscular adenovirus serotype 26 (Ad26) vector vaccination in tissues. We show that the Ad26 vaccine-elicited innate immune response commences by 1 hour and rapidly evolves in tissues and blood within the first 24 hours, as reflected by the detection of cytokines, chemokines, cellular responses, and transcriptomic pathways. Furthermore, serum levels of IL-6, MIG, MIP-1α, MIP-1β, and TNF-α at 6 hours post-vaccination correlated with the frequency of vaccine-elicited memory CD8+ T cell responses evaluated at 60 days post-vaccination in blood and tissues. Taken together, our data suggest that the immune response to Ad26 vector vaccination commences quickly in tissues by 1 hour and that events by as early as 6 hours post-vaccination can shape vaccine-elicited CD8+ T cell responses at later memory time points.IMPORTANCEPrior studies have largely concentrated on innate immune activation in peripheral blood following vaccination. In this study, we report the detailed spatial and temporal innate immune activation in tissues following Ad26 vaccination in mice. We observed rapid innate activation not only in peripheral blood but also in draining lymph nodes and at the site of inoculation. Our findings provide a more detailed picture of the host response to vaccination than previously reported.
Collapse
Affiliation(s)
- Eryn Blass
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Alessandro Colarusso
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Malika Aid
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Rafael A Larocca
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - R Keith Reeves
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
55
|
Yang JQ, Wang C, Nayak RC, Kolla M, Cai M, Pujato M, Zheng Y, Lu QR, Guo F. Genetic and epigenetic regulation of Treg cell fitness by autism-related chromatin remodeler CHD8. Cell Mol Biol Lett 2025; 30:36. [PMID: 40155813 PMCID: PMC11954365 DOI: 10.1186/s11658-025-00711-z] [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: 10/01/2024] [Accepted: 02/25/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Chromatin remodeler chromodomain helicase DNA-binding protein 8 (CHD8) defines a subtype of autism that is associated with immune disorders. It remains unknown whether CHD8 plays a cell-intrinsic role in immune cells such as regulatory T cells (Tregs) that maintain immune tolerance through suppressing CD4+ and CD8+ effector T cells. METHODS Treg-specific conditional CHD8-deficient mice were generated by crossing Chd8Flox/Flox mice with Foxp3YFP-cre transgenic mice. Effects of CHD8 deficiency were investigated using hematoxylin and eosin (H&E) staining, flow cytometry, and multi-omics, including RNA-sequencing (RNA-seq), assay for transposase-accessible chromatin sequencing (ATAC-seq), and chromatin immunoprecipitation sequencing (CHIP-seq). RESULTS We found that Treg-specific CHD8 deletion led to early, fatal inflammation owing to increased CD4+ and CD8+ effector T cells. CHD8 deletion did not alter Treg homeostasis but increased their functional plasticity with elevated expression of effector T cell cytokines. CHIP-seq of Tregs uncovered that CHD8 binding genes were enriched in phosphatidylinositol-3 kinase (PI3K)-protein kinase B (Akt)-mammalian target of rapamycin (mTOR) signaling and several other pathways. RNA-seq and ATAC-seq revealed that CHD8 deletion upregulated a number of pathways, notably mammalian target of rapamycin complex 1 (mTORC1) signaling and its mediated glycolysis that have been reported to promote Treg plasticity. Integrating RNA-seq data with CHIP-seq and ATAC-seq data identified a number of CHD8 target genes whose expression depends on CHD8 direct binding-mediated chromatin remodeling. CONCLUSIONS Our findings suggest that CHD8 plays an important role in maintaining Treg fitness through genetic and epigenetic mechanisms to control autoimmunity, which may have important implications in immune changes in autism.
Collapse
Affiliation(s)
- Jun-Qi Yang
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Chen Wang
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Ramesh C Nayak
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Manohar Kolla
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Mingjun Cai
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Mario Pujato
- Life Sciences Computational Services LLC, Huntingdon Valley, PA, 19006, USA
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Q Richard Lu
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA.
| | - Fukun Guo
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA.
| |
Collapse
|
56
|
Rahimian E, Koochak M, Traikov S, Schroeder M, Brilloff S, Schäfer S, Kufrin V, Küchler S, Krüger A, Mirtschink P, Baretton G, Schröck E, Schewe DM, Ball CR, Bornhäuser M, Glimm H, Bill M, Wurm AA. A quiescence-like/TGF-β1-specific CRISPRi screen reveals drug uptake transporters as secondary targets of kinase inhibitors in AML. Drug Resist Updat 2025; 81:101242. [PMID: 40184725 DOI: 10.1016/j.drup.2025.101242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/26/2025] [Accepted: 03/23/2025] [Indexed: 04/07/2025]
Abstract
Relapse in acute myeloid leukemia (AML) is driven by resistant subclones that survive chemotherapy. It is assumed that these resilient leukemic cells can modify their proliferative behavior by entering a quiescent-like state, similar to healthy hematopoietic stem cells (HSCs). These dormant cells can evade the effects of cytostatic drugs that primarily target actively dividing cells. Although quiescence has been extensively studied in healthy hematopoiesis and various solid cancers, its role in AML has remained unexplored. In this study, we applied an HSC-derived quiescence-associated gene signature to an AML patient cohort and found it to be strongly correlated with poor prognosis and active TGF-β signaling. In vitro treatment with TGF-β1 induces a quiescence-like phenotype, resulting in a G0 shift and reduced sensitivity to cytarabine. To find potential therapeutic targets that prevent AML-associated quiescence and improve response to cytarabine, we conducted a comprehensive CRISPR interference (CRISPRi) screen combined with TGF-β1 stimulation. This approach identified TGFBR1 inhibitors, like vactosertib, as effective agents for preventing the G0 shift in AML cell models. However, pretreatment with vactosertib unexpectedly induced complete resistance to cytarabine. To elucidate the underlying mechanism, we performed a multi-faceted approach combining a second CRISPRi screen, liquid chromatography-tandem mass spectrometry (LC-MS/MS), and in silico analysis. Our findings revealed that TGFBR1 inhibitors unintentionally target the nucleoside transporter SLC29A1 (ENT1), leading to reduced intracellular cytarabine levels. Importantly, we found that this drug interaction is not unique to TGFBR1 inhibitors, but extends to other clinically significant kinase inhibitors, such as the FLT3 inhibitor midostaurin. These findings may have important implications for optimizing combination therapies in AML treatment.
Collapse
Affiliation(s)
- Elahe Rahimian
- Mildred Scheel Early Career Center, National Center for Tumor Diseases (NCT/UCC) Dresden, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany; Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Germany
| | - Masoud Koochak
- Mildred Scheel Early Career Center, National Center for Tumor Diseases (NCT/UCC) Dresden, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany; Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Germany
| | - Sofia Traikov
- Institute for Clinical Chemistry and Laboratory Medicine, TU Dresden, Dresden, Germany
| | - Michael Schroeder
- Biotechnology Center (BIOTEC), Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Silke Brilloff
- Mildred Scheel Early Career Center, National Center for Tumor Diseases (NCT/UCC) Dresden, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany; Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Germany
| | - Silvia Schäfer
- Mildred Scheel Early Career Center, National Center for Tumor Diseases (NCT/UCC) Dresden, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany; Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Germany
| | - Vida Kufrin
- Mildred Scheel Early Career Center, National Center for Tumor Diseases (NCT/UCC) Dresden, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany; Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Germany
| | - Sandra Küchler
- Mildred Scheel Early Career Center, National Center for Tumor Diseases (NCT/UCC) Dresden, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany; Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Germany
| | - Alexander Krüger
- Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases Dresden (NCT), NCT/UCC Dresden, a partnership between German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Germany
| | - Peter Mirtschink
- Institute for Clinical Chemistry and Laboratory Medicine, TU Dresden, Dresden, Germany
| | - Gustavo Baretton
- Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases Dresden (NCT), NCT/UCC Dresden, a partnership between German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Germany; Institute for Pathology, Faculty of Medicine Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Evelin Schröck
- Institute for Clinical Genetics, University Hospital Carl Gustav Carus at TUD Dresden University of Technology and Faculty of Medicine of TUD Dresden University of Technology, Dresden, Germany; ERN GENTURIS, Hereditary Cancer Syndrome Center Dresden, Germany; National Center for Tumor Diseases (NCT), NCT/UCC Dresden, a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Germany; German Cancer Consortium (DKTK), Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Denis M Schewe
- Department of Pediatric Hematology and Oncology, University Hospital Dresden, Dresden, Germany
| | - Claudia R Ball
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Germany; Translational Medical Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany; German Cancer Consortium (DKTK), partner site Dresden, Germany; TUD Dresden University of Technology, Faculty of Biology, Dresden, Germany
| | - Martin Bornhäuser
- Mildred Scheel Early Career Center, National Center for Tumor Diseases (NCT/UCC) Dresden, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany; Department of Internal Medicine I, University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Hanno Glimm
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Germany; Translational Medical Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany; German Cancer Consortium (DKTK), partner site Dresden, Germany; Translational Functional Cancer Genomics, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marius Bill
- Mildred Scheel Early Career Center, National Center for Tumor Diseases (NCT/UCC) Dresden, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany; Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Germany; German Cancer Consortium (DKTK), partner site Dresden, Germany; Department of Internal Medicine I, University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Alexander A Wurm
- Mildred Scheel Early Career Center, National Center for Tumor Diseases (NCT/UCC) Dresden, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany; Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Germany; Department of Pediatric Hematology and Oncology, University Hospital Dresden, Dresden, Germany; Translational Medical Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany; German Cancer Consortium (DKTK), partner site Dresden, Germany.
| |
Collapse
|
57
|
Vaiasicca S, James DW, Melone G, Saeed O, Francis LW, Corradetti B. Amniotic fluid-derived mesenchymal stem cells as a therapeutic tool against cytokine storm: a comparison with umbilical cord counterparts. Stem Cell Res Ther 2025; 16:151. [PMID: 40156072 PMCID: PMC11951844 DOI: 10.1186/s13287-025-04262-0] [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] [Accepted: 03/04/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Several immunosuppressive therapies have been proposed as key treatment options for critically ill patients since the first appearance of severe acute respiratory syndrome coronavirus 2. Mesenchymal stem cells (MSCs) from different sources have been considered for their potential to attenuate the cytokine storm associated to COVID-19 and the consequent multi-organ failure, providing evidence for safe and efficacious treatments. Among them, administration of umbilical cord-derived MSCs (UC-MSCs) has demonstrated a significant increase in survival rates, largely due to their potent immunosuppressive properties. METHODS We applied next-generation sequencing (NGS) analysis to compare the transcriptomic profiles of MSCs isolated from two gestational sources: amniotic fluid (AF) obtained during prenatal diagnosis and their clinically relevant umbilical cord counterparts, for which datasets were publicly available. A full meta-analysis was performed to identify suitable GEO and NGS datasets for comparison between AF- and UC-MSC samples. RESULTS Transcriptome analysis revelaed significant differences between groups, despite both cell lines being strongly involved in the tissue development, crucial to achieve the complex task of wound healing. Significantly enriched hallmark genes suggest AF-MSC superior immunomodulatory features against signaling pathways actively involved in the cytokine storm (i.e., IL-2/STAT, TNF-a/NFkB, IL-2/STAT5, PI3K/AKT/mTOR). CONCLUSIONS The data presented here suggest that AF-MSCs hold significant promise for treating not only COVID-19-associated cytokine storms but also a variety of other inflammatory syndromes (i.e., those induced by bacterial infections, autoimmune disorders, and therapeutic interventions). Realizing the full potential of AF-MSCs as a comprehensive therapeutic approach in inflammatory disease management will require more extensive clinical trials and in-depth mechanistic studies.
Collapse
Affiliation(s)
- Salvatore Vaiasicca
- Advanced Technology Center for Aging Research, IRCCS INRCA, Ancona, Italy
- Department of Life and Environmental Life, Polytechnic University of Marche, Ancona, Italy
| | - David W James
- Centre of NanoHealth, Swansea University Medical School, Swansea, UK
| | - Gianmarco Melone
- Centre of NanoHealth, Swansea University Medical School, Swansea, UK
| | - Omar Saeed
- Centre of NanoHealth, Swansea University Medical School, Swansea, UK
| | - Lewis W Francis
- Centre of NanoHealth, Swansea University Medical School, Swansea, UK
| | - Bruna Corradetti
- Centre of NanoHealth, Swansea University Medical School, Swansea, UK.
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA.
- Department of Medicine, Section Oncology/Hematology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
58
|
Wang Q, Li S, Wei Y, Xu C, Liu X, Wang X, Chai W, Mou W, Chen X, Li C, Wang C, Gui J. An increase in IL-10-producing DNT cells is associated with the pathogenesis of pediatric SLE. Clin Immunol 2025; 276:110490. [PMID: 40158789 DOI: 10.1016/j.clim.2025.110490] [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: 01/18/2025] [Revised: 03/20/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease that causes immune system overactivity and organ damage. Among T-cell subsets involved in SLE, CD4 and CD8 double-negative αβT (DNT) cells have attracted attention in recent years, although their role in SLE remains poorly understood. Examining the minute intricacies, particularly signaling pathway modifications is crucial, as it may unveil potential therapeutic targets and lead to the development of more effective treatments. Our study found increased DNT cells in pediatric SLE patients, with elevated IL-10 signaling. These IL-10-producing DNT cells were positively related to disease activity defined by SLE Disease Activity Index (SLEDAI), and were further elevated in patients with lupus nephritis. Additionally, our results indicated that IL-10-producing DNT cells correlated positively with anti-Sm autoantibodies. Collectively, our study revealed that modulation of IL-10 production within DNT-cell subset could affect both immune regulation and autoantibody production, contributing to the immunological dysregulation in SLE.
Collapse
Affiliation(s)
- Qixin Wang
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China; Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Shipeng Li
- Department of Rheumatology, National Centre for Children's Health, Beijing Children's Hospital, Capital Medical University, No. 56 Nan Li Shi Lu, Beijing 100045, China
| | - Yannan Wei
- Inner Mongolia Xilingol League Central Hospital, Xilinhaote 026000, China
| | - Chen Xu
- Inner Mongolia Xilingol League Central Hospital, Xilinhaote 026000, China
| | - Xiangjun Liu
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China; Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Xiaolin Wang
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China; Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Wenjia Chai
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China; Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Wenjun Mou
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China; Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Xi Chen
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China; Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Caifeng Li
- Department of Rheumatology, National Centre for Children's Health, Beijing Children's Hospital, Capital Medical University, No. 56 Nan Li Shi Lu, Beijing 100045, China.
| | - Caisheng Wang
- Inner Mongolia Xilingol League Central Hospital, Xilinhaote 026000, China.
| | - Jingang Gui
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China; Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China.
| |
Collapse
|
59
|
Lee JH, Son S, Ko Y, Lim H, Lee M, Kang MG, Kim H, Lee KM, Shin I. Nidogen-1 suppresses cell proliferation, migration, and glycolysis via integrin β1-mediated HIF-1α downregulation in triple-negative breast cancer. Sci Rep 2025; 15:10633. [PMID: 40148359 PMCID: PMC11950294 DOI: 10.1038/s41598-024-84880-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 12/27/2024] [Indexed: 03/29/2025] Open
Abstract
Nidogen-1 (NID1) is a secreted glycoprotein widely distributed in basement membranes. NID1 interacts with extracellular matrix proteins such as collagen and laminin and has been implicated in the progression of various cancers. However, study on the role of NID1 in breast cancer is scarce and inconsistent. In this work, we found that the expression of NID1 is significantly lower in breast cancer tissue than in normal tissue. In addition, NID1 expression correlated negatively with a poor prognosis for breast cancer patients. Based on those findings, we speculated that NID1 might act as a cancer suppressor in breast cancer. To investigate the role of NID1 in breast cancer, we constructed NID1-overexpressing cell lines. NID1 overexpression decreased breast cancer cell proliferation, migration, and in vivo tumor growth. Moreover, glucose metabolism, which is known to enhance cancer cell proliferation and migration, was also decreased by NID1 overexpression. Mechanistically, NID1 overexpression downregulated hypoxia-inducible factor-1α (HIF-1α) expression at the transcription level. Furthermore, we found that NID1 reduced integrin β1 stability and downregulated the transcription of HIF-1α through the FAK/Src/NF-κB p65 signaling axis, which is downstream of integrin β1. Together, the results of this study demonstrate the tumor suppressive role of NID1 in triple-negative breast cancer.
Collapse
Affiliation(s)
- Joo-Hyung Lee
- Department of Life Science, Hanyang University, Seoul, 04763, Korea
| | - Seogho Son
- Department of Life Science, Hanyang University, Seoul, 04763, Korea
| | - Yunhyo Ko
- Department of Life Science, Hanyang University, Seoul, 04763, Korea
| | - Hogeun Lim
- Department of Life Science, Hanyang University, Seoul, 04763, Korea
| | - Minhyeok Lee
- Department of Life Science, Hanyang University, Seoul, 04763, Korea
| | - Min-Gyeong Kang
- Department of Life Science, Hanyang University, Seoul, 04763, Korea
| | - Hyungjoo Kim
- Department of Life Science, Hanyang University, Seoul, 04763, Korea
| | - Kyung-Min Lee
- Department of Life Science, Hanyang University, Seoul, 04763, Korea
| | - Incheol Shin
- Department of Life Science, Hanyang University, Seoul, 04763, Korea.
- Natural Science Institute, Hanyang University, Seoul, 04763, Korea.
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, 04763, Korea.
| |
Collapse
|
60
|
Kansakar SB, Sterben SP, Anamala CC, Thielen MD, Liaudanskaya V. The Silent Saboteur: How Mitochondria Shape the Long-Term Fate of the Injured Brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.19.644244. [PMID: 40166284 PMCID: PMC11957143 DOI: 10.1101/2025.03.19.644244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Traumatic brain injury (TBI) is a major risk factor for neurodegenerative diseases, including Alzheimer's disease (AD), yet the mechanistic link remains unclear. Here, we integrated human patient-derived transcriptomics with a 3D in vitro brain injury model to dissect cell-specific mitochondrial dysfunction as a driver of injury-induced neurodegeneration. Comparative transcriptomic analysis at 6 and 48 hours post-injury revealed conserved mitochondrial impairments across excitatory neurons, interneurons, astrocytes, and microglia. Using a novel cell-specific mitochondria tracking system, we demonstrate prolonged neuronal mitochondrial fragmentation, bioenergetic failure, and metabolic instability, coinciding with the emergence of AD markers, including pTau, APP, and Aβ42/40 dysregulation. Glial mitochondria exhibited delayed but distinct metabolic dysfunctions, with astrocytes impaired metabolic support and microglia sustained chronic inflammation. These findings establish neuronal mitochondrial failure as an early trigger of injury-induced neurodegeneration, reinforcing mitochondrial dysfunction as a therapeutic target for preventing TBI-driven AD pathology.
Collapse
|
61
|
Pan D, Chen P, Zhang H, Zhao Q, Fang W, Ji S, Chen T. Mitochondrial quality control: A promising target of traditional Chinese medicine in the treatment of cardiovascular disease. Pharmacol Res 2025; 215:107712. [PMID: 40154932 DOI: 10.1016/j.phrs.2025.107712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/08/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
Cardiovascular disease remains the leading cause of death globally, and drugs for new targets are urgently needed. Mitochondria are the primary sources of cellular energy, play crucial roles in regulating cellular homeostasis, and are tightly associated with pathological processes in cardiovascular disease. In response to physiological signals and external stimuli in cardiovascular disease, mitochondrial quality control, which mainly includes mitophagy, mitochondrial dynamics, and mitochondrial biogenesis, is initiated to meet cellular requirements and maintain cellular homeostasis. Traditional Chinese Medicine (TCM) has been shown to have pharmacological effects on alleviating cardiac injury in various cardiovascular diseases, including myocardial ischemia/reperfusion, myocardial infarction, and heart failure, by regulating mitochondrial quality control. Recently, several molecular mechanisms of TCM in the treatment of cardiovascular disease have been elucidated. However, mitochondrial quality control by TCM for treating cardiovascular disease has not been investigated. In this review, we aim to decipher the pharmacological effects and molecular mechanisms of TCM in regulating mitochondrial quality in various cardiovascular diseases. We also present our perspectives regarding future research in this field.
Collapse
Affiliation(s)
- Deng Pan
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang, China.
| | - Pengfei Chen
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China; National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - He Zhang
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China; National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Qian Zhao
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang, China
| | - Wei Fang
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang, China
| | - Siyan Ji
- Stomatology Department of Qiqihar Medical College School, Heilongjiang, China
| | - Tielong Chen
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang, China.
| |
Collapse
|
62
|
Zapata-Linares N, Toillon I, Wanherdrick K, Pigenet A, Duhalde F, Binvignat M, Martin-Uriz PS, Louvet L, Calleja-Cervantes ME, Ghali Mhenni O, Guibert C, Nourissat G, Nogier A, Leterme D, Broux O, Magneron P, Prosper F, Chauveau C, Landoulsi J, Berenbaum F, Rodriguez-Madoz JR, Lafage-Proust MH, Lucas S, Houard X. Implication of bone marrow adipose tissue in bone homeostasis during osteoarthritis. Osteoarthritis Cartilage 2025:S1063-4584(25)00870-2. [PMID: 40154729 DOI: 10.1016/j.joca.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 03/16/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
OBJECTIVE To explore the role of bone marrow adipocytes (BMAds) in osteoarthritis (OA). METHODS Male and female C57BL/6 mice (n=4/group) underwent meniscectomy (MNX) or SHAM surgery. OA was determined using Osteoarthritis Research Society International (OARSI) score, and the number of perilipin+ adipocytes was quantified. Mesenchymal Stromal Cells (MSCs) from MNX and SHAM mice were differentiated into osteoblasts and adipocytes. Human adipocytes and MSCs (n=8) were enzymatically isolated from epiphyseal and metaphyseal marrow, and from subcutaneous adipose tissue (SCAT) of hip OA patients. Human OA MSCs were differentiated into osteoblasts and adipocytes (OA-Diff-hAdipo). Gene expression patterns of epiphyseal and metaphyseal BMAds, SCAT adipocytes and OA-Diff-hAdipo were evaluated by RNAseq (n=4). The effect conditioned media from OA epiphyseal bone (n=5) on the alkaline phosphatase (ALP) activity and mineralization kinetics was assessed in vitro. RESULTS Increase in BMAd density was positively correlated with cartilage degradation in MNX mice. OA modified the differentiation capacity of MSCs, accelerating adipocyte differentiation and failing to produce osteoblasts in both human and mice. Human epiphyseal, metaphyseal and SCAT adipocytes from the same OA patients each displayed a specific transcriptome, suggesting different functions. Enrichment analysis defined metaphyseal OA-BMAds as cells implicated in hematopoietic stem cell differentiation. On the other hand, epiphyseal OA-BMAds were considered as osteogenic cells showing an up-regulation of genes related to bone mineralization and remodeling. Specifically, OA epiphysis-secreted molecules decreased ALP activity and altered in vitro the mineralization process. CONCLUSION All these results support the emergence of BMAds as new cell partners in OA, opening new venues for therapeutic approaches.
Collapse
Affiliation(s)
- Natalia Zapata-Linares
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France
| | - Indira Toillon
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France
| | - Kristell Wanherdrick
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France
| | - Audrey Pigenet
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France
| | - Fanny Duhalde
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France; Sorbonne Université, CNRS, Laboratoire de Réactivité de Surface, LRS, F-75005, Paris, France
| | - Marie Binvignat
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France
| | | | - Loïc Louvet
- Marrow Adiposity and Bone Lab (MABLab) ULR4490, Université du Littoral Côte d'Opale, F-62200 Boulogne sur Mer, Univ. Lille, F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Maria E Calleja-Cervantes
- Hemato-Oncology Program. CIMA Universidad de Navarra-IdiSNA, Pamplona, Spain; Computational Biology Program, CIMA Universidad de Navarra-IdiSNA, Pamplona, Spain
| | - Olfa Ghali Mhenni
- Marrow Adiposity and Bone Lab (MABLab) ULR4490, Université du Littoral Côte d'Opale, F-62200 Boulogne sur Mer, Univ. Lille, F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Clément Guibert
- Sorbonne Université, CNRS, Laboratoire de Réactivité de Surface, LRS, F-75005, Paris, France
| | - Geoffroy Nourissat
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France
| | | | - Damien Leterme
- Marrow Adiposity and Bone Lab (MABLab) ULR4490, Université du Littoral Côte d'Opale, F-62200 Boulogne sur Mer, Univ. Lille, F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Odile Broux
- Marrow Adiposity and Bone Lab (MABLab) ULR4490, Université du Littoral Côte d'Opale, F-62200 Boulogne sur Mer, Univ. Lille, F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Paul Magneron
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France
| | - Felipe Prosper
- Hemato-Oncology Program. CIMA Universidad de Navarra-IdiSNA, Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Hematology and Cell Therapy Department, Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain; Cancer Center Universidad de Navarra (CCUN), Pamplona, Spain
| | - Christophe Chauveau
- Marrow Adiposity and Bone Lab (MABLab) ULR4490, Université du Littoral Côte d'Opale, F-62200 Boulogne sur Mer, Univ. Lille, F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Jessem Landoulsi
- Sorbonne Université, CNRS, Laboratoire de Réactivité de Surface, LRS, F-75005, Paris, France
| | - Francis Berenbaum
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France; Rheumatology Department, AP-HP Saint-Antoine Hospital, 184, Rue du Faubourg Saint-Antoine, 75012 Paris, France
| | - Juan R Rodriguez-Madoz
- Hemato-Oncology Program. CIMA Universidad de Navarra-IdiSNA, Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Cancer Center Universidad de Navarra (CCUN), Pamplona, Spain
| | - Marie-Hélène Lafage-Proust
- Université de Lyon - Université Jean Monnet, INSERM U1059, Faculté de Médecine, F-42270 Saint-Priest en Jarez, France
| | - Stéphanie Lucas
- Marrow Adiposity and Bone Lab (MABLab) ULR4490, Université du Littoral Côte d'Opale, F-62200 Boulogne sur Mer, Univ. Lille, F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Xavier Houard
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France.
| |
Collapse
|
63
|
Xu J, Wakai M, Xiong K, Yang Y, Prabakaran A, Wu S, Ahrens D, Molina-Portela MDP, Ni M, Bai Y, Shavlakadze T, Glass DJ. The pro-inflammatory cytokine IL6 suppresses mitochondrial function via the gp130-JAK1/STAT1/3-HIF1α/ERRα axis. Cell Rep 2025; 44:115403. [PMID: 40056415 DOI: 10.1016/j.celrep.2025.115403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 11/07/2024] [Accepted: 02/14/2025] [Indexed: 03/10/2025] Open
Abstract
Chronic inflammation and a decline in mitochondrial function are hallmarks of aging. Here, we show that the two mechanisms may be linked. We found that interleukin-6 (IL6) suppresses mitochondrial function in settings where PGC1 (both PGC1α and PGC1β) expression is low. This suppression is mediated by the JAK1/STAT1/3 axis, which activates HIF1α through non-canonical mechanisms involving upregulation of HIF1A and ERRα transcription, and subsequent stabilization of the HIF1A protein by ERRα. HIF1α, in turn, inhibits ERRα, which is a master regulator of mitochondrial biogenesis, thus contributing to the inhibition of mitochondrial function. When expressed at higher levels, PGC1 rescues ERRα to boost baseline mitochondrial respiration, including under IL6-treated conditions. Our study suggests that inhibition of the IL6 signaling axis could be a potential treatment for those inflammatory settings where mitochondrial function is compromised.
Collapse
Affiliation(s)
- Jianing Xu
- Aging/Age-Related Diseases, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA.
| | - Matthew Wakai
- Aging/Age-Related Diseases, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Kun Xiong
- Molecular Profiling & Data Science, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Yanfeng Yang
- Aging/Age-Related Diseases, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Adithya Prabakaran
- Aging/Age-Related Diseases, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Sophia Wu
- Aging/Age-Related Diseases, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Diana Ahrens
- Research Flow Cytometry Core, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | | | - Min Ni
- Molecular Profiling & Data Science, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Yu Bai
- Molecular Profiling & Data Science, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Tea Shavlakadze
- Aging/Age-Related Diseases, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA.
| | - David J Glass
- Aging/Age-Related Diseases, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA.
| |
Collapse
|
64
|
Finch RH, Vitry G, Siew K, Walsh SB, Beheshti A, Hardiman G, da Silveira WA. Spaceflight causes strain-dependent gene expression changes in the kidneys of mice. NPJ Microgravity 2025; 11:11. [PMID: 40133368 PMCID: PMC11937539 DOI: 10.1038/s41526-025-00465-0] [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: 07/15/2024] [Accepted: 02/17/2025] [Indexed: 03/27/2025] Open
Abstract
Incidence of kidney stones in astronauts is a major risk factor associated with long-term missions, caused by increased blood calcium levels due to bone demineralisation triggered by microgravity and space radiation. Transcriptomic changes have been observed in tissues during spaceflight, including the kidney. We analysed kidney transcriptome patterns in two different strains of mice flown on the International Space Station, C57BL/6J and BALB/c. Here we show a link between spaceflight and transcriptome patterns associated with dysregulation of lipid and extracellular matrix metabolism and altered transforming growth factor-beta signalling. A stronger response was seen in C57BL/6J mice than BALB/c. Genetic differences in hyaluronan metabolism between strains may confer protection against extracellular matrix remodelling through the downregulation of epithelial-mesenchymal transition. We intend for our findings to contribute to the development of new countermeasures against kidney disease in astronauts and people here on Earth.
Collapse
Affiliation(s)
- Rebecca H Finch
- University of Staffordshire, Department of Sports and Science, School of Health, Education, Policing and Sciences, Science Centre, Leek Road, Stoke-on-Trent, ST4 2DF, UK
| | - Geraldine Vitry
- University of Staffordshire, Department of Sports and Science, School of Health, Education, Policing and Sciences, Science Centre, Leek Road, Stoke-on-Trent, ST4 2DF, UK
- International Space University, 1 Rue Jean-Dominique Cassini, 67400, Illkirch-Graffenstaden, France
- Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Department of Oncology, 3970 Reservoir Rd, NW, New Research Building EP11, Washington, DC, 20057, USA
| | - Keith Siew
- London Tubular Centre, Department of Renal Medicine, University College London, London, UK
| | - Stephen B Walsh
- London Tubular Centre, Department of Renal Medicine, University College London, London, UK
| | - Afshin Beheshti
- Center for Space Biomedicine, McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Gary Hardiman
- Faculty of Medicine, Health and Life Sciences, Institute for Global Food Security (IGFS), School of Biological Sciences, Queen's University Belfast, 19 Chlorine Gardens, Belfast, BT9 5DL, UK
- Department of Medicine, Medical University of South Carolina, MSC 403, 171 Ashley Ave Suite 419, Charleston, SC, 29425, USA
| | - Willian A da Silveira
- University of Staffordshire, Department of Sports and Science, School of Health, Education, Policing and Sciences, Science Centre, Leek Road, Stoke-on-Trent, ST4 2DF, UK.
- International Space University, 1 Rue Jean-Dominique Cassini, 67400, Illkirch-Graffenstaden, France.
- School of Science, Engineering and Environment. University of Salford, Manchester, M5 4WT, UK.
| |
Collapse
|
65
|
Chang J, Campbell-Hanson KR, Vanneste M, Bartschat NI, Nagel R, Arnadottir AK, Vu HN, Montgomery C, Yevdash J, Jiang J, Bhinu A, Helverson A, Henry MD, Steingrímsson E, Weigel RJ, Cornell RA, Kenny C. Antagonistic roles for MITF and TFE3 in melanoma plasticity. Cell Rep 2025; 44:115474. [PMID: 40138313 DOI: 10.1016/j.celrep.2025.115474] [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: 08/03/2024] [Revised: 01/06/2025] [Accepted: 03/05/2025] [Indexed: 03/29/2025] Open
Abstract
Melanoma cells can switch from a melanocytic and proliferative state to a mesenchymal and invasive state and back again. This plasticity drives intratumoral heterogeneity, progression, and therapeutic resistance. Microphthalmia-associated transcription factor (MITF) promotes the melanocytic/proliferative phenotype, but factors that drive the mesenchymal/invasive phenotype and the mechanisms that effect the switch between cell states are unclear. Here, we identify the MITF paralog, TFE3, and the non-canonical mTORC1 pathway as regulators of the mesenchymal state. We show that TFE3 expression drives the metastatic phenotype in melanoma cell lines and tumors. Deletion of TFE3 in MITF-low melanoma cell lines suppresses their ability to migrate and metastasize. Further, MITF suppresses the mesenchymal phenotype by directly or indirectly activating expression of FNIP1, FNIP2, and FLCN, which encode components of the non-canonical mTORC1 pathway, thereby promoting cytoplasmic retention and lysosome-mediated degradation of TFE3. These findings highlight a molecular pathway controlling melanoma plasticity and invasiveness.
Collapse
Affiliation(s)
- Jeremy Chang
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | | | - Marion Vanneste
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Nicholas I Bartschat
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Ryan Nagel
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Asdis K Arnadottir
- Department of Biochemistry and Molecular Biology, BioMedical Center, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Hong Nhung Vu
- Department of Biochemistry and Molecular Biology, BioMedical Center, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Collin Montgomery
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Julius Yevdash
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Jiarui Jiang
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Ardith Bhinu
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Annika Helverson
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA, USA
| | - Michael D Henry
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Eiríkur Steingrímsson
- Department of Biochemistry and Molecular Biology, BioMedical Center, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Ronald J Weigel
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Robert A Cornell
- Department of Oral Health Sciences, University of Washington, School of Dentistry, Seattle, WA, USA
| | - Colin Kenny
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
66
|
Hosseini TM, Jang SS, Bendik J, Guo T. Indisulam Shows an Anti-Cancer Effect on HPV+ and HPV- Head and Neck Cancer. Cancers (Basel) 2025; 17:1072. [PMID: 40227602 PMCID: PMC11987906 DOI: 10.3390/cancers17071072] [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: 02/04/2025] [Revised: 03/16/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Background/Objectives: HPV+ head and neck squamous cell carcinoma has been shown to have a unique genomic background, requiring researchers to study it as its own distinct type of cancer. HPV+ tumors have been shown to exhibit fewer genetic mutations in cancer drivers as opposed to their HPV- counterparts. In this paper, we explored how targeting post-transcriptional changes, specifically alternative splicing events, could serve as a potential mechanism to treat HPV+ cancer. Methods: Using indisulam, a drug that targets alternative splicing through the degradation of RBM39, we treated various HPV+ and HPV- cell lines and assessed tumor cell viability. We also tested indisulam in vivo to evaluate its effect on tumor volume. Additionally, we analyzed gene expression differences between indisulam-treated subjects and their non-treated counterparts. Results: Indisulam treatment led to a reduction in tumor cell viability in both HPV+ and HPV- cell lines. In vivo experiments showed a reduction in tumor volume following indisulam treatment. Gene expression analysis revealed that indisulam induces consistent differential gene expression changes and highly enriches interferon pathways in treated HPV+ cell lines. Conclusions: These findings suggest that targeting alternative splicing via indisulam may be a promising therapeutic approach for HPV+ cancers. Further research is required to establish indisulam as a viable anti-cancer treatment in clinical settings.
Collapse
Affiliation(s)
- Tara M. Hosseini
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Sophie S. Jang
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Joseph Bendik
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Theresa Guo
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
- Department of Otolaryngology-Head & Neck Surgery, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
67
|
Zhang Y, Nomura M, Nishimura K, Zang W, Koike Y, Xiao M, Ito H, Fukumoto M, Tanaka A, Aoyama Y, Saika W, Hasegawa C, Yamazaki H, Takaori-Kondo A, Inoue D. In-depth functional analysis of BRD9 in fetal hematopoiesis reveals context-dependent roles. iScience 2025; 28:112010. [PMID: 40109374 PMCID: PMC11919606 DOI: 10.1016/j.isci.2025.112010] [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: 07/18/2024] [Revised: 10/19/2024] [Accepted: 02/10/2025] [Indexed: 03/22/2025] Open
Abstract
The hierarchical organization of hematopoietic stem cells (HSCs) governing adult hematopoiesis has been extensively investigated. However, the dynamic epigenomic transition from fetal to adult hematopoiesis remains incompletely understood, particularly regarding the involvement of epigenetic factors. In this study, we investigate the roles of BRD9, an essential component of the non-canonical BAF (ncBAF) complex known to govern the fate of adult HSCs, in fetal hematopoiesis. Consistent with observations in adult hematopoiesis, BRD9 loss impairs fetal HSC stemness and disturbs erythroid maturation. Intriguingly, the impact on myeloid lineage was discrepant: BRD9 loss inhibited and promoted myeloid differentiation in fetal and adult models, respectively. Through comprehensive transcriptomic and epigenomic analysis, we elucidate the differential roles of BRD9 in a context- and lineage-dependent manner. Our data uncover how BRD9/ncBAF complex modulates transcription in a stage-specific manner, providing deeper insights into the epigenetic regulation underlying the transition from fetal to adult hematopoiesis.
Collapse
Affiliation(s)
- Yifan Zhang
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Hematology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masaki Nomura
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Facility for iPS Cell Therapy, CiRA Foundation, Kyoto, Japan
- Department of Cancer Pathology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Japan
| | - Koutarou Nishimura
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Cancer Pathology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Japan
| | - Weijia Zang
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Hematology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Cancer Pathology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Japan
| | - Yui Koike
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Cancer Pathology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Japan
| | - Muran Xiao
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Hiromi Ito
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Cancer Pathology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Japan
| | - Miki Fukumoto
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Atsushi Tanaka
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Yumi Aoyama
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Hematology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Wataru Saika
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Cancer Pathology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Japan
- Department of Hematology, Shiga University of Medical Science, Otsu, Japan
| | - Chihiro Hasegawa
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Cancer Pathology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Japan
- Department of Hematology and Oncology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Hiromi Yamazaki
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Cancer Pathology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Japan
| | - Akifumi Takaori-Kondo
- Department of Hematology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Daichi Inoue
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Hematology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Cancer Pathology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Japan
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
| |
Collapse
|
68
|
Feng X, Xu M, Liu Y, Wang X, Duan Y, Zheng X, Yin W, Cai Y, Zhang W, Jiang Q, Pang J, Li J. The sperm quality in DIO male mice is linked to the NF-κB signaling and Ppp2ca expression in the hypothalamus. iScience 2025; 28:112110. [PMID: 40160428 PMCID: PMC11951025 DOI: 10.1016/j.isci.2025.112110] [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: 07/29/2024] [Revised: 11/24/2024] [Accepted: 02/21/2025] [Indexed: 04/02/2025] Open
Abstract
Recent studies show obesity correlated with reduced sperm quality in males, but the mechanism is unclear. In this study, diet-induced obese (DIO) male mice exhibited disrupted luteinizing hormone (LH) pulse release due to altered function of the hypothalamic-pituitary-gonadal (HPG) axis. This alteration was caused by activation of nuclear factor kappa B (NF-κB) signaling in the hypothalamus, which led to decreased sperm quality. RNA sequencing (RNA-seq) analysis of the hypothalamic arcuate nucleus (ARC) revealed a signaling network involving protein phosphatase 2 catalytic subunit alpha (Ppp2ca). This network disrupted LH pulse secretion by inhibiting Akt kinase (AKT) and cAMP responsive element-binding protein 1 (CREB1) activities, thereby reducing KiSS-1 metastasis-suppressor (Kiss1) expression. Furthermore, overexpression of the Ppp2ca gene in the ARC led to disrupted LH patterns and reduced sperm quality. These findings offer new insights into the molecular mechanisms underlying sperm quality decline in DIO male mice.
Collapse
Affiliation(s)
- Xu Feng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Maoxing Xu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Ying Liu
- Clinical Center of Reproductive Medicine, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Xiaoyu Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yiman Duan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xiaoyan Zheng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Wen Yin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yafei Cai
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Wei Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Qin Jiang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Jing Pang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Juxue Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
- The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, China
| |
Collapse
|
69
|
Han C, Yuan H, Chen AK, DiPietro LA, Chen L. Differential Temperature-Induced Responses in Immortalized Oral and Skin Keratinocytes. Int J Mol Sci 2025; 26:2851. [PMID: 40243437 PMCID: PMC11988828 DOI: 10.3390/ijms26072851] [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: 02/14/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/18/2025] Open
Abstract
The epidermis of the skin and oral mucosa is constantly exposed to various environmental stimuli, including temperature changes. In particularly extreme conditions, such as excess heat or cold, significant injury may occur. Oral and skin keratinocytes exhibit tissue-specific differences in wound healing outcomes and the transcriptomic response to injury. This study investigated if skin and oral keratinocytes also have differential responses to heat- and cold-induced injury. Oral keratinocytes (TIGKs) were found to exhibit an enhanced viability following heat-induced injury compared to skin keratinocytes (HaCaTs). However, there were no discernible differences between skin and oral keratinocyte viability following cold-induced injury. To examine the transcriptomic differences between skin and oral keratinocytes in response to temperature-induced injury, we generated an mRNA-sequencing gene expression dataset. Differentially expressed genes (DEGs) including heat shock proteins (HSPs) were identified between HaCaTs and TIGKs at baseline (37 °C) and after heat- (60 °C) or cold-induced (-25 °C) injury. Our comparative analyses suggest that skin and oral keratinocytes exhibit transcriptomic differences at baseline and in their responses to heat or cold exposure. The enhanced heat tolerance of TIGKs relative to HaCaTs may be due to an advantageous expression of a subset of HSPs at baseline in TIGKs. Our work also provides a source of skin and oral keratinocyte gene expression data following heat- and cold-induced injury that can be used for future analyses.
Collapse
Affiliation(s)
| | | | | | | | - Lin Chen
- Center for Wound Healing and Tissue Regeneration, University of Illinois Chicago, Chicago, IL 60612, USA; (C.H.); (H.Y.); (A.K.C.); (L.A.D.)
| |
Collapse
|
70
|
Myers JM, Sandel C, Alvarez K, Garman L, Wiley G, Montgomery C, Gaffney P, Stavrakis S, Fairweather D, Bruno KA, Zhao YD, Cooper LT, Cunningham MW. Cardiac autoantibodies promote a fibrotic transcriptome and reduced ventricular recovery in human myocarditis. Front Immunol 2025; 16:1500909. [PMID: 40181955 PMCID: PMC11965655 DOI: 10.3389/fimmu.2025.1500909] [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: 09/24/2024] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
Myocarditis leads to dilated cardiomyopathy (DCM) with one-third failing to recover normal ejection fraction (EF 50%). Our previous studies have supported a Th17 autoimmune pathogenesis where IL17A and IL-6 are elevated in myocarditis patients who do not recover normal EF. In the non-recovered group, autoantibody mechanisms of pathogenesis in myocardial injury and systolic dysfunction are not fully understood. Furthermore, in our myocarditis cohort, cardiac myosin (CM) autoantibodies (AAbs) were elevated and cross-reactive with the β-adrenergic receptor (βAR). Here we studied cross-reactive CM/βAR serum AAbs and human myocarditis-derived monoclonal antibodies (mAbs) to define their potential pathogenic mechanisms and to identify unique human CM epitopes associated with non-recovery in a longitudinal (n=41) cohort. Elevated CM IgG AAbs in the non-recovered phenotype correlated with reduced EF and poor outcomes. Human CM epitopes unique to the non-recovered phenotype shared strong amino acid sequence homology with extracellular loops of βARs and supported molecular mimicry and cross-reactivity between CM and βAR. Myocarditis-derived IgG and human mAb 2C.4 activated protein kinase A (PKA) in an IgG, CM, and βAR-dependent manner in H9c2 heart myoblast cell line, and transcriptomic analysis revealed mAb 2C.4 induced fibrosis pathways which were highly similar pathways seen with isoproterenol, a beta receptor agonist. Our data translate into new mechanistic insights from our small longitudinal group of myocarditis/DCM patients and into potential therapeutic targets and biomarkers for future studies.
Collapse
Affiliation(s)
- Jennifer M. Myers
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Clayton Sandel
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Kathy Alvarez
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Lori Garman
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Graham Wiley
- Clinical Genomics Core, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Courtney Montgomery
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Patrick Gaffney
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Stavros Stavrakis
- Department of Cardiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - DeLisa Fairweather
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Katelyn A. Bruno
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Yan Daniel Zhao
- Biostatistics and Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Leslie T. Cooper
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Madeleine W. Cunningham
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
71
|
Ver Heul AM, Mack M, Zamidar L, Tamari M, Yang TL, Trier AM, Kim DH, Janzen-Meza H, Van Dyken SJ, Hsieh CS, Karo JM, Sun JC, Kim BS. RAG suppresses group 2 innate lymphoid cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.04.23.590767. [PMID: 38712036 PMCID: PMC11071423 DOI: 10.1101/2024.04.23.590767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Antigen specificity is the central trait distinguishing adaptive from innate immune function. Assembly of antigen-specific T cell and B cell receptors occurs through V(D)J recombination mediated by the Recombinase Activating Gene endonucleases RAG1 and RAG2 (collectively called RAG). In the absence of RAG, mature T and B cells do not develop and thus RAG is critically associated with adaptive immune function. In addition to adaptive T helper 2 (Th2) cells, group 2 innate lymphoid cells (ILC2s) contribute to type 2 immune responses by producing cytokines like Interleukin-5 (IL-5) and IL-13. Although it has been reported that RAG expression modulates the function of innate natural killer (NK) cells, whether other innate immune cells such as ILC2s are affected by RAG remains unclear. We find that in RAG-deficient mice, ILC2 populations expand and produce increased IL-5 and IL-13 at steady state and contribute to increased inflammation in atopic dermatitis (AD)-like disease. Further, we show that RAG modulates ILC2 function in a cell-intrinsic manner independent of the absence or presence of adaptive T and B lymphocytes. Lastly, employing multiomic single cell analyses of RAG1 lineage-traced cells, we identify key transcriptional and epigenomic ILC2 functional programs that are suppressed by a history of RAG expression. Collectively, our data reveal a novel role for RAG in modulating innate type 2 immunity through suppression of ILC2s.
Collapse
Affiliation(s)
- Aaron M. Ver Heul
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Madison Mack
- Immunology & Inflammation Research Therapeutic Area, Sanofi, Cambridge, MA 02141, USA
| | - Lydia Zamidar
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Masato Tamari
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ting-Lin Yang
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Anna M. Trier
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Do-Hyun Kim
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63130, USA
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 04763, Republic of Korea
| | - Hannah Janzen-Meza
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Steven J. Van Dyken
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Chyi-Song Hsieh
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Jenny M. Karo
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Immunology and Microbial Pathogenesis Program, Graduate School of Medical Sciences, Weill Cornell Medical College, New York, NY 10065, USA
| | - Joseph C. Sun
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Immunology and Microbial Pathogenesis Program, Graduate School of Medical Sciences, Weill Cornell Medical College, New York, NY 10065, USA
| | - Brian S. Kim
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Allen Discovery Center for Neuroimmune Interactions, Icahn School of Medicine at Mount Sinai 10019
| |
Collapse
|
72
|
Alhomoud M, Foley M, Sugita M, Fein JA, Yamshon S, Martinez L, Rejeski K, Astorkia M, Betel D, Brentjens R, van Besien K, Galluzzi L, Boyer O, Martinet J, Formenti S, Guzman ML. Total body irradiation primes CD19-directed CAR T cells against large B-cell lymphoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.17.643462. [PMID: 40166306 PMCID: PMC11957048 DOI: 10.1101/2025.03.17.643462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
CD19-targeting chimeric antigen receptor T cells (CART19) have demonstrated significant effectiveness in treating relapsed or refractory large B-cell lymphoma (LBCL). However, they often fail to sustain durable remissions in more than half of all treated patients. Therefore, there is an urgent need to identify approaches to enhance CART19 efficacy. Here, we studied the impact of low-dose radiation on CART19 activity in vitro and find that radiation enhances the cytotoxicity of CART19 against LBCL by upregulating death receptors. Disrupting the FAS receptor diminishes this benefit, indicating that this pathway plays an important role in enhancing the cytotoxic effects of CAR T cells. To further validate these findings, we conducted in vivo studies using a lymphoma syngeneic mouse model delivering total body irradiation (TBI). We observed that delivering TBI at a single dose of 1Gy prior to CAR T cell infusion significantly improved CART19-mediated tumor elimination and increased overall survival rates. Importantly, we characterized several important effects of TBI, including enhanced lymphodepletion, improved T cell expansion and persistence, better intra-tumoral migration, and a more favorable, anti-tumor phenotypic composition of the T cells. In summary, for the first time, we have demonstrated preclinically that administering TBI before CART19 infusion significantly accelerates tumor elimination and improves overall survival. This approach holds promise for translation into clinical practice and serves as a valuable foundation for further research to enhance outcomes for patients receiving CART19 treatment.
Collapse
|
73
|
Wang MX, Mauch BE, Williams AF, Barazande-Pour T, Araujo Hoffmann F, Harris SH, Lathrop CP, Turkal CE, Yung BS, Paw MH, Gervasio DAG, Tran T, Stuhlfire AE, Guo T, Daniels GA, Park SJ, Gutkind JS, Hangauer MJ. Antigenic cancer persister cells survive direct T cell attack. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643359. [PMID: 40166148 PMCID: PMC11956947 DOI: 10.1101/2025.03.14.643359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Drug-tolerant persister cancer cells were first reported fifteen years ago as a quiescent, reversible cell state which tolerates unattenuated cytotoxic drug stress. It remains unknown whether a similar phenomenon contributes to immune evasion. Here we report a persister state which survives weeks of direct cytotoxic T lymphocyte (CTL) attack. In contrast to previously known immune evasion mechanisms that avoid immune attack, antigenic persister cells robustly activate CTLs which deliver Granzyme B, secrete IFNγ, and induce tryptophan starvation resulting in apoptosis initiation. Instead of dying, persister cells paradoxically leverage apoptotic caspase activity to avoid inflammatory death. Furthermore, persister cells acquire mutations and epigenetic changes which enable outgrowth of CTL-resistant cells. Persister cell features are enriched in inflamed tumors which regressed during immunotherapy in vivo and in surgically resected human melanoma tissue under immune stress ex vivo. These findings reveal a persister cell state which is a barrier to immune-mediated tumor clearance.
Collapse
Affiliation(s)
- Michael X Wang
- Department of Dermatology, University of California San Diego
| | - Brandon E Mauch
- Department of Dermatology, University of California San Diego
| | | | | | | | - Sophie H Harris
- Department of Dermatology, University of California San Diego
| | | | - Claire E Turkal
- Department of Dermatology, University of California San Diego
| | - Bryan S Yung
- Department of Pharmacology, University of California San Diego
- Moores Cancer Center, University of California San Diego
| | - Michelle H Paw
- Department of Dermatology, University of California San Diego
| | | | - Tiffany Tran
- Department of Dermatology, University of California San Diego
| | | | - Theresa Guo
- Moores Cancer Center, University of California San Diego
- Department of Otolaryngology, University of California San Diego
| | - Gregory A Daniels
- Moores Cancer Center, University of California San Diego
- Division of Hematology-Oncology, Department of Medicine, University of California San Diego
| | - Soo J Park
- Moores Cancer Center, University of California San Diego
- Division of Hematology-Oncology, Department of Medicine, University of California San Diego
| | - J Silvio Gutkind
- Department of Pharmacology, University of California San Diego
- Moores Cancer Center, University of California San Diego
| | - Matthew J Hangauer
- Department of Dermatology, University of California San Diego
- Moores Cancer Center, University of California San Diego
| |
Collapse
|
74
|
Melton R, Jimenez S, Elison W, Tucciarone L, Howell A, Wang G, Berti D, Beebe E, Miller M, Zeng C, McGrail C, VanderStel K, Korgaonkar K, Elgamal R, Mummey H, Chiou J, Griffin E, Kusmartseva I, Atkinson M, Preissl S, Theis FJ, Sander M, Gaulton KJ. Single-cell multiome and spatial profiling reveals pancreas cell type-specific gene regulatory programs driving type 1 diabetes progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.13.637721. [PMID: 40027657 PMCID: PMC11870426 DOI: 10.1101/2025.02.13.637721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Cell type-specific regulatory programs that drive type 1 diabetes (T1D) in the pancreas are poorly understood. Here we performed single nucleus multiomics and spatial transcriptomics in up to 32 non-diabetic (ND), autoantibody-positive (AAB+), and T1D pancreas donors. Genomic profiles from 853,005 cells mapped to 12 pancreatic cell types, including multiple exocrine sub-types. Beta, acinar, and other cell types, and related cellular niches, had altered abundance and gene activity in T1D progression, including distinct pathways altered in AAB+ compared to T1D. We identified epigenomic drivers of gene activity in T1D and AAB+ which, combined with genetic association, revealed causal pathways of T1D risk including antigen presentation in beta cells. Finally, single cell and spatial profiles together revealed widespread changes in cell-cell signaling in T1D including signals affecting beta cell regulation. Overall, these results revealed drivers of T1D progression in the pancreas, which form the basis for therapeutic targets for disease prevention.
Collapse
Affiliation(s)
- Rebecca Melton
- Biomedical sciences program, University of California San Diego, La Jolla CA
| | - Sara Jimenez
- Department of Computational Health, Institute of Computational Biology, Helmholtz, Munich, Germany
| | - Weston Elison
- Biomedical sciences program, University of California San Diego, La Jolla CA
| | - Luca Tucciarone
- Department of Pediatrics, University of California San Diego, La Jolla CA
| | - Abigail Howell
- Department of Pediatrics, University of California San Diego, La Jolla CA
| | - Gaowei Wang
- Department of Pediatrics, University of California San Diego, La Jolla CA
| | - Denise Berti
- Department of Pediatrics, University of California San Diego, La Jolla CA
| | - Elisha Beebe
- Department of Pediatrics, University of California San Diego, La Jolla CA
| | - Michael Miller
- Center for Epigenomics, University of California San Diego, La Jolla CA
| | - Chun Zeng
- Department of Pediatrics, University of California San Diego, La Jolla CA
| | - Carolyn McGrail
- Biomedical sciences program, University of California San Diego, La Jolla CA
| | - Kennedy VanderStel
- Department of Pediatrics, University of California San Diego, La Jolla CA
| | - Katha Korgaonkar
- Department of Pediatrics, University of California San Diego, La Jolla CA
| | - Ruth Elgamal
- Biomedical sciences program, University of California San Diego, La Jolla CA
| | - Hannah Mummey
- Bioinformatics and systems biology program, University of California San Diego, La Jolla CA
| | - Joshua Chiou
- Pfizer Research and Discovery, Pfizer Inc, Cambridge, MA
| | - Emily Griffin
- Department of Pediatrics, University of California San Diego, La Jolla CA
| | - Irina Kusmartseva
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville FL
| | - Mark Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville FL
| | - Sebastian Preissl
- Center for Epigenomics, University of California San Diego, La Jolla CA
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Fabian J Theis
- Department of Computational Health, Institute of Computational Biology, Helmholtz, Munich, Germany
| | - Maike Sander
- Department of Pediatrics, University of California San Diego, La Jolla CA
- Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla CA
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Kyle J Gaulton
- Department of Pediatrics, University of California San Diego, La Jolla CA
| |
Collapse
|
75
|
Moreno-Sánchez I, Hernández-Huertas L, Nahón-Cano D, Martínez-García PM, Treichel AJ, Gómez-Marin C, Tomás-Gallardo L, da Silva Pescador G, Kushawah G, Egidy R, Perera A, Díaz-Moscoso A, Cano-Ruiz A, Walker JA, Muñoz MJ, Holden K, Galcerán J, Nieto MÁ, Bazzini AA, Moreno-Mateos MA. Enhanced RNA-targeting CRISPR-Cas technology in zebrafish. Nat Commun 2025; 16:2591. [PMID: 40091120 PMCID: PMC11911407 DOI: 10.1038/s41467-025-57792-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 02/28/2025] [Indexed: 03/19/2025] Open
Abstract
CRISPR-Cas13 RNA-targeting systems are widely used in basic and applied sciences. However, its application has recently generated controversy due to collateral activity in mammalian cells and mouse models. Moreover, its competence could be improved in vivo. Here, we optimized transient formulations as ribonucleoprotein complexes or mRNA-gRNA combinations to enhance the CRISPR-RfxCas13d system in zebrafish. We i) use chemically modified gRNAs to allow more penetrant loss-of-function phenotypes, ii) improve nuclear RNA targeting, and iii) compare different computational models and determine the most accurate to predict gRNA activity in vivo. Furthermore, we demonstrate that transient CRISPR-RfxCas13d can effectively deplete endogenous mRNAs in zebrafish embryos without inducing collateral effects, except when targeting extremely abundant and ectopic RNAs. Finally, we implement alternative RNA-targeting CRISPR-Cas systems such as CRISPR-Cas7-11 and CRISPR-DjCas13d. Altogether, these findings contribute to CRISPR-Cas technology optimization for RNA targeting in zebrafish through transient approaches and assist in the progression of in vivo applications.
Collapse
Grants
- F31 HD110268 NICHD NIH HHS
- R01 GM136849 NIGMS NIH HHS
- R21 OD034161 NIH HHS
- This work was supported by Ramon y Cajal (RyC-2017-23041), PID2021-127535NB-I00, CNS2022-135564 and CEX2020-001088-M grants funded by MICIU/AEI/ 10.13039/501100011033 by “ERDF A way of making Europe” (“ERDF/EU”), and by ESF Investing in your future from Ministerio de Ciencia, Innovación y Universidades and European Union (M.A.M.-M.). This work has also been co-financed by the Spanish Ministry of Science and Innovation with funds from the European Union NextGenerationEU (PRTR-C17.I1) and the Regional Ministry of University, Research and Innovation of the Autonomous Community of Andalusia within the framework of the Biotechnology Plan applied to Health. The Moreno-Mateos lab was also funded by European Regional Development Fund (FEDER 80% of the total funding) by the Ministry of Economy, Knowledge, Business and University, of the Government of Andalusia, within the framework of the FEDER Andalusia 2014-2020 operational program within the objective "Promotion and generation of frontier knowledge and knowledge oriented to the challenges of society, development of emerging technologies (grant UPO-1380590)” and by the Fondo Europeo de Desarrollo Regional (FEDER) and Consejería de Transformación Económica, Industria, Conocimiento y Universidades de la Junta de Andalucía, within the operative program FEDER Andalucía 2014-2020 (01 - Refuerzo de la investigación, el desarrollo tecnológico y la innovación, grant P20_00866). M.A.M.-M. was the recipient of the Genome Engineer Innovation 2019 Grant from Synthego. The CABD is an institution funded by University Pablo de Olavide, Consejo Superior de Investigaciones Científicas (CSIC), and Junta de Andalucía.
Collapse
Affiliation(s)
- Ismael Moreno-Sánchez
- Andalusian Center for Developmental Biology (CABD), Pablo de Olavide University/CSIC/Junta de Andalucía, Seville, Spain
- Department of Molecular Biology and Biochemical Engineering, Pablo de Olavide University, Seville, Spain
- Instituto de Neurociencias (CSIC-UMH), Alicante, Spain
| | - Luis Hernández-Huertas
- Andalusian Center for Developmental Biology (CABD), Pablo de Olavide University/CSIC/Junta de Andalucía, Seville, Spain
- Department of Molecular Biology and Biochemical Engineering, Pablo de Olavide University, Seville, Spain
| | - Daniel Nahón-Cano
- Andalusian Center for Developmental Biology (CABD), Pablo de Olavide University/CSIC/Junta de Andalucía, Seville, Spain
- Department of Molecular Biology and Biochemical Engineering, Pablo de Olavide University, Seville, Spain
| | - Pedro Manuel Martínez-García
- Andalusian Center for Developmental Biology (CABD), Pablo de Olavide University/CSIC/Junta de Andalucía, Seville, Spain
| | | | - Carlos Gómez-Marin
- Andalusian Center for Developmental Biology (CABD), Pablo de Olavide University/CSIC/Junta de Andalucía, Seville, Spain
- Department of Molecular Biology and Biochemical Engineering, Pablo de Olavide University, Seville, Spain
| | - Laura Tomás-Gallardo
- Andalusian Center for Developmental Biology (CABD), Pablo de Olavide University/CSIC/Junta de Andalucía, Seville, Spain
- Proteomics and Biochemistry Platform, Andalusian Center for Developmental Biology (CABD), Pablo de Olavide University/CSIC/Junta de Andalucía, Seville, Spain
| | | | - Gopal Kushawah
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Rhonda Egidy
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Anoja Perera
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Alejandro Díaz-Moscoso
- Andalusian Center for Developmental Biology (CABD), Pablo de Olavide University/CSIC/Junta de Andalucía, Seville, Spain
- Proteomics and Biochemistry Platform, Andalusian Center for Developmental Biology (CABD), Pablo de Olavide University/CSIC/Junta de Andalucía, Seville, Spain
- Instituto de Investigaciones Químicas (IIQ-CICIC), CSIC-US, Seville, Spain
| | - Alejandra Cano-Ruiz
- Andalusian Center for Developmental Biology (CABD), Pablo de Olavide University/CSIC/Junta de Andalucía, Seville, Spain
- Department of Molecular Biology and Biochemical Engineering, Pablo de Olavide University, Seville, Spain
| | | | - Manuel J Muñoz
- Andalusian Center for Developmental Biology (CABD), Pablo de Olavide University/CSIC/Junta de Andalucía, Seville, Spain
- Department of Molecular Biology and Biochemical Engineering, Pablo de Olavide University, Seville, Spain
| | | | - Joan Galcerán
- Instituto de Neurociencias (CSIC-UMH), Alicante, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
| | - M Ángela Nieto
- Instituto de Neurociencias (CSIC-UMH), Alicante, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
| | - Ariel A Bazzini
- Stowers Institute for Medical Research, Kansas City, MO, USA
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Miguel A Moreno-Mateos
- Andalusian Center for Developmental Biology (CABD), Pablo de Olavide University/CSIC/Junta de Andalucía, Seville, Spain.
- Department of Molecular Biology and Biochemical Engineering, Pablo de Olavide University, Seville, Spain.
| |
Collapse
|
76
|
Sviderskiy VO, Vasudevaraja V, Dubois LG, Stafford J, Liu EK, Serrano J, Possemato R, Snuderl M. Metabolic profiling of adult and pediatric gliomas reveals enriched glucose availability in pediatric gliomas and increased fatty acid oxidation in adult gliomas. Acta Neuropathol Commun 2025; 13:61. [PMID: 40087788 PMCID: PMC11909955 DOI: 10.1186/s40478-025-01961-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 02/15/2025] [Indexed: 03/17/2025] Open
Abstract
Gliomas are the most common primary brain tumors and a major source of mortality and morbidity in adults and children. Recent genomic studies have identified multiple molecular subtypes; however metabolic characterization of these tumors has thus far been limited. We performed metabolic profiling of 114 adult and pediatric primary gliomas and integrated metabolomic data with transcriptomics and DNA methylation classes. We identified that pediatric tumors have higher levels of glucose and reduced lactate compared to adult tumors regardless of underlying genetics or grade, suggesting differences in availability of glucose and/or utilization of glucose for downstream pathways. Differences in glucose utilization in pediatric gliomas may be facilitated through overexpression of SLC2A4, which encodes the insulin-stimulated glucose transporter GLUT4. Transcriptomic comparison of adult and pediatric tumors suggests that adult tumors may have limited access to glucose and experience more hypoxia, which is supported by enrichment of lactate, 2-hydroxyglutarate (2-HG), even in isocitrate dehydrogenase (IDH) wild-type tumors, and 3-hydroxybutyrate, a ketone body that is produced by oxidation of fatty acids and ketogenic amino acids during periods of glucose scarcity. Our data support adult tumors relying more on fatty acid oxidation, as they have an abundance of acyl carnitines compared to pediatric tumors and have significant enrichment of transcripts needed for oxidative phosphorylation. Our findings suggest striking differences exist in the metabolism of pediatric and adult gliomas, which can provide new insight into metabolic vulnerabilities for therapy.
Collapse
Affiliation(s)
- Vladislav O Sviderskiy
- Department of Pathology, NYU Langone Health, New York, NY, USA
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Luiz Gustavo Dubois
- Department of Pathology, NYU Langone Health, New York, NY, USA
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - James Stafford
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Elisa K Liu
- NYU Grossman School of Medicine, New York, NY, 10016, USA
| | | | - Richard Possemato
- Department of Pathology, NYU Langone Health, New York, NY, USA.
- NYU Grossman School of Medicine, New York, NY, 10016, USA.
- Laura and Isaac Perlmutter Cancer Center, New York, NY, 10016, USA.
- Department of Pathology, NYU Langone Health, 550 First Avenue, Smilow 611, New York, NY, 10016, USA.
| | - Matija Snuderl
- Department of Pathology, NYU Langone Health, New York, NY, USA.
- NYU Grossman School of Medicine, New York, NY, 10016, USA.
- Laura and Isaac Perlmutter Cancer Center, New York, NY, 10016, USA.
- Department of Pathology, NYU Langone Health, 240 E 38Th Street, 22Nd Floor, New York, NY, 10016, USA.
| |
Collapse
|
77
|
Senguttuvan RN, Cho H, Wu X, Frankel PH, Ruel N, Yost SE, Kebria M, Han E, Song M, de Leon M, Invernizzi M, Eng M, Tinsley R, Salehian B, Li A, Schmolze D, Chang S, Arias-Stella J, Dellinger TH. The Anti-Cancer Role of Pterostilbene in Endometrial Cancer: A Phase II Prospective, Randomized, Window-of-Opportunity Clinical Trial with Megestrol Acetate. Antioxidants (Basel) 2025; 14:345. [PMID: 40227411 PMCID: PMC11939256 DOI: 10.3390/antiox14030345] [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: 02/17/2025] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 04/15/2025] Open
Abstract
Pterostilbene (3,5-dimethoxy-40-hydroxystilbene) is a potent oral antioxidant with a promising role in anti-cancer treatment. In endometrial cancer (EC), in vitro studies demonstrated a synergistic antiproliferative effect of pterostilbene (PT) with megestrol acetate (MA), a common treatment for EC. This is a randomized phase II clinical trial (NCT03671811) of PT+MA vs. MA for three weeks prior to scheduled hysterectomy. The primary objective is to determine the antiproliferative effect of PT+MA vs. MA using Ki-67 index. The secondary objectives are toxicity, histological response, transcriptional changes, and lipid metabolism. A total of 44 patients were enrolled between January 2019 and November 2022 with 23 randomized to Arm 1 (PT+MA) and 21 to Arm 2 (MA). Toxicities included one G3 thromboembolic event (PT+MA) and one G3 hypertension event (MA). Histological responses were high in both arms (>90%). There was no difference in Ki-67 changes, although, when restricted to endometroid subtype, the relative decrease in Ki67 was 33.8% in PT+MA vs. 20.1% in MA alone (p = 0.14). Whole transcriptomic gene profiling of samples before and after PT+MA exposure demonstrated the activation of interferon alpha response pathway and suppression of mTORC1 signaling, hypoxia, oxidative phosphorylation, and IL2-STAT5 signaling. Lipid metabolism analyses did not reveal any significant changes between arms. PT is well-tolerated in the preoperative treatment of EC and demonstrated in vivo anti-cancer effects on the transcriptomic level.
Collapse
Affiliation(s)
- Rosemary N. Senguttuvan
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA; (R.N.S.); (M.K.); (E.H.); (M.S.); (M.d.L.); (M.I.)
| | - Hyejin Cho
- Integrative Genomics Core, City of Hope National Medical Center, Duarte, CA 91010, USA; (H.C.); (X.W.)
| | - Xiwei Wu
- Integrative Genomics Core, City of Hope National Medical Center, Duarte, CA 91010, USA; (H.C.); (X.W.)
| | - Paul H. Frankel
- Department of Computation and Quantitative Medicine, City of Hope National Medical Center, Duarte, CA 91010, USA; (P.H.F.); (N.R.)
| | - Nora Ruel
- Department of Computation and Quantitative Medicine, City of Hope National Medical Center, Duarte, CA 91010, USA; (P.H.F.); (N.R.)
| | - Susan E. Yost
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA 91010, USA;
| | - Mehdi Kebria
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA; (R.N.S.); (M.K.); (E.H.); (M.S.); (M.d.L.); (M.I.)
| | - Ernest Han
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA; (R.N.S.); (M.K.); (E.H.); (M.S.); (M.d.L.); (M.I.)
| | - Mihae Song
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA; (R.N.S.); (M.K.); (E.H.); (M.S.); (M.d.L.); (M.I.)
| | - Maria de Leon
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA; (R.N.S.); (M.K.); (E.H.); (M.S.); (M.d.L.); (M.I.)
| | - Marta Invernizzi
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA; (R.N.S.); (M.K.); (E.H.); (M.S.); (M.d.L.); (M.I.)
| | - Melissa Eng
- Clinical Trials Office, City of Hope National Medical Center, Duarte, CA 91010, USA; (M.E.); (R.T.)
| | - Raechelle Tinsley
- Clinical Trials Office, City of Hope National Medical Center, Duarte, CA 91010, USA; (M.E.); (R.T.)
| | - Behrouz Salehian
- Department of Endocrinology, City of Hope National Medical Center, Duarte, CA 91010, USA;
| | - Aimin Li
- Department of Pathology, City of Hope National Medical Center, Duarte, CA 91010, USA; (A.L.); (D.S.); (S.C.); (J.A.-S.)
| | - Daniel Schmolze
- Department of Pathology, City of Hope National Medical Center, Duarte, CA 91010, USA; (A.L.); (D.S.); (S.C.); (J.A.-S.)
| | - Sue Chang
- Department of Pathology, City of Hope National Medical Center, Duarte, CA 91010, USA; (A.L.); (D.S.); (S.C.); (J.A.-S.)
| | - Javier Arias-Stella
- Department of Pathology, City of Hope National Medical Center, Duarte, CA 91010, USA; (A.L.); (D.S.); (S.C.); (J.A.-S.)
| | - Thanh H. Dellinger
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA; (R.N.S.); (M.K.); (E.H.); (M.S.); (M.d.L.); (M.I.)
| |
Collapse
|
78
|
Jonker PB, Sadullozoda M, Cognet G, Saab JJA, Sokol KH, Wu VX, Kumari D, Sheehan C, Ozgurses ME, Agovino D, Croley G, Patel SA, Bock-Hughes A, Macleod KF, Shah H, Coloff JL, Lien EC, Muir A. Microenvironmental arginine restriction sensitizes pancreatic cancers to polyunsaturated fatty acids by suppression of lipid synthesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.10.642426. [PMID: 40161789 PMCID: PMC11952453 DOI: 10.1101/2025.03.10.642426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Nutrient limitation is a characteristic feature of poorly perfused tumors. In contrast to well-perfused tissues, nutrient deficits in tumors perturb cellular metabolic activity, which imposes metabolic constraints on cancer cells. The metabolic constraints created by the tumor microenvironment can lead to vulnerabilities in cancers. Identifying the metabolic constraints of the tumor microenvironment and the vulnerabilities that arise in cancers can provide new insight into tumor biology and identify promising antineoplastic targets. To identify how the microenvironment constrains the metabolism of pancreatic tumors, we challenged pancreatic cancer cells with microenvironmental nutrient levels and analyzed changes in cell metabolism. We found that arginine limitation in pancreatic tumors perturbs saturated and monounsaturated fatty acid synthesis by suppressing the lipogenic transcription factor SREBP1. Synthesis of these fatty acids is critical for maintaining a balance of saturated, monounsaturated, and polyunsaturated fatty acids in cellular membranes. As a consequence of microenvironmental constraints on fatty acid synthesis, pancreatic cancer cells and tumors are unable to maintain lipid homeostasis when exposed to polyunsaturated fatty acids, leading to cell death by ferroptosis. In sum, arginine restriction in the tumor microenvironment constrains lipid metabolism in pancreatic cancers, which renders these tumors vulnerable to polyunsaturatedenriched fat sources.
Collapse
Affiliation(s)
- Patrick B. Jonker
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| | - Mumina Sadullozoda
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| | - Guillaume Cognet
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| | - Juan J. Apiz Saab
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| | - Kelly H. Sokol
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, Michigan, USA, 49503
| | - Violet X. Wu
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| | - Deepa Kumari
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| | - Colin Sheehan
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| | - Mete E. Ozgurses
- Department of Physiology and Biophysics, University of Illinois College of Medicine, Chicago, IL, USA, 60612
| | - Darby Agovino
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| | - Grace Croley
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| | - Smit A. Patel
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| | - Althea Bock-Hughes
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| | - Kay F. Macleod
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| | - Hardik Shah
- Metabolomics Platform, Comprehensive Cancer Center, The University of Chicago, Chicago, IL, USA, 60637
| | - Jonathan L. Coloff
- Department of Physiology and Biophysics, University of Illinois College of Medicine, Chicago, IL, USA, 60612
| | - Evan C. Lien
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, Michigan, USA, 49503
| | - Alexander Muir
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| |
Collapse
|
79
|
Strefeler A, Baker ZN, Chollet S, Guerra RM, Ivanisevic J, Gallart-Ayala H, Pagliarini DJ, Jourdain AA. Uridine-sensitized screening identifies genes and metabolic regulators of nucleotide synthesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.11.642569. [PMID: 40161720 PMCID: PMC11952425 DOI: 10.1101/2025.03.11.642569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Nucleotides are essential for nucleic acid synthesis, signaling, and metabolism, and can be synthesized de novo or through salvage. Rapidly proliferating cells require large amounts of nucleotides, making nucleotide metabolism a widely exploited target for cancer therapy. However, resistance frequently emerges, highlighting the need for a deeper understanding of nucleotide regulation. Here, we harness uridine salvage and CRISPR-Cas9 screening to reveal regulators of de novo pyrimidine synthesis. We identify several factors and report that pyrimidine synthesis can continue in the absence of coenzyme Q (CoQ), the canonical electron acceptor in de novo synthesis. We further investigate NUDT5 and report its conserved interaction with PPAT, the rate-limiting enzyme in purine synthesis. We show that in the absence of NUDT5, hyperactive purine synthesis siphons the phosphoribosyl pyrophosphate (PRPP) pool at the expense of pyrimidine synthesis, promoting resistance to chemotherapy. Intriguingly, the interaction between NUDT5 and PPAT appears to be disrupted by PRPP, highlighting intricate allosteric regulation. Our findings reveal a fundamental mechanism for maintaining nucleotide balance and position NUDT5 as a potential biomarker for predicting resistance to chemotherapy.
Collapse
Affiliation(s)
- Abigail Strefeler
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Zakery N Baker
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, USA
| | - Sylvain Chollet
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Rachel M Guerra
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, USA
| | | | | | - David J Pagliarini
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, USA
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, USA
- Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, USA
| | - Alexis A Jourdain
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
80
|
Zhang N, Sun Q, Zhang J, Zhang R, Liu S, Zhao X, Ma J, Li X. Intrapancreatic adipocytes and beta cell dedifferentiation in human type 2 diabetes. Diabetologia 2025:10.1007/s00125-025-06392-9. [PMID: 40072535 DOI: 10.1007/s00125-025-06392-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/20/2025] [Indexed: 03/14/2025]
Abstract
AIMS/HYPOTHESIS Fat deposition in the pancreas is implicated in beta cell dysfunction and the progress of type 2 diabetes. However, there is limited evidence to confirm the correlation and explore how pancreatic fat links with beta cell dysfunction in human type 2 diabetes. This study aimed to examine the spatial relationship between pancreatic fat and islets in human pancreases. METHODS Histological analysis of pancreatic specimens from 50 organ donors (15 with type 2 diabetes, 35 without) assessed pancreatic fat content variation among individuals with diabetes and its correlation with estimated beta cell mass and cell distribution within islets. Bioinformatic analysis of single-cell RNA-seq of 11 type 2 diabetic donors (from the Human Pancreatic Analysis Project database) explored the impact of high pancreatic fat content on beta cell gene expression and cell fate. Validation of bioinformatic results was performed with the above diabetic pancreases. RESULTS Pancreatic fat content was higher in individuals with type 2 diabetes (10.24% [3.29-13.89%] vs 0.74% [0.34-5.11%], p<0.001), negatively correlated with estimated beta cell mass (r=-0.675, p=0.006) and positively with alpha-to-beta cell ratio (r=0.608, p=0.016). Enrichment analysis indicated that in diabetic donors with higher pancreatic fat content, the expression of ALDH1A3, beta cell dedifferentiation marker, in both alpha and beta cells was significantly increased, and in beta cells, the expression of NPY decreased. Pseudotime analysis revealed beta cell dedifferentiation and transdifferentiation towards alpha cells in diabetic donors with higher pancreatic fat content, with decreased expression of genes related to beta cell maturation and function, including INSM1, MafA and NPY. Concurrently, pathways related to inflammation and immune response were activated. Histologically, pancreatic fat content correlated positively with the percentage of beta cells positive for aldehyde dehydrogenase 1 family member A3 (ALDH1A3) within the islets (r=0.594, p=0.020) and the ALDH1A3 positivity rate in beta cells (r=0.615, p=0.015). And the number of T cells adjacent to adipocytes was related to the distribution pattern of adipocytes and the dedifferentiation phenotype in islets. CONCLUSIONS/INTERPRETATION Higher pancreatic fat content was accompanied by increased beta cell dedifferentiation in the individuals with diabetes. Clusters of adipocytes significantly contribute to higher pancreatic fat content and immune cell recruitment. Overall, the interactions among adipocytes, immune cells and beta cells in the pancreas microenvironment might contribute to beta cell failure and dedifferentiation in type 2 diabetes.
Collapse
Affiliation(s)
- Na Zhang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qiman Sun
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiaxin Zhang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ruonan Zhang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Siyi Liu
- Fudan University, Shanghai, China
| | - Xuelian Zhao
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing Ma
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaomu Li
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
81
|
White JB, Sanchez KL, Currais A, Soriano-Castell D, Maher P, Soriano S. Ferroptosis and Charcot-Marie-Tooth Disease 1A: Emerging Evidence for a Pathogenic Association. Antioxidants (Basel) 2025; 14:331. [PMID: 40227266 PMCID: PMC11939244 DOI: 10.3390/antiox14030331] [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: 02/12/2025] [Revised: 03/08/2025] [Accepted: 03/08/2025] [Indexed: 04/15/2025] Open
Abstract
Charcot-Marie-Tooth disease (CMT) is the most common hereditary peripheral neuropathy worldwide, presenting clinically as muscle weakness that progresses to impaired ambulation or quadriplegia with age. CMT1A, the most common subtype, is caused by a duplication in PMP22, encoding an essential membrane protein for Schwann cell myelin integrity. While the mechanisms of peripheral neurodegeneration in CMT1A are poorly understood, excessive oxidative stress, particularly lipid peroxidation, is a known pathological feature, and antioxidant therapy has reversed the CMT1A phenotype in a mouse model. For the first time, we define the pathogenic link between CMT1A and ferroptosis, a form of regulated cell death caused by excessive lipid peroxidation and hindered antioxidant defenses. Human-derived CMT1A fibroblasts showed greater susceptibility to RSL3, a pro-ferroptosis agent, compared with controls, alongside several ferroptosis markers, including elevated lipid peroxides and depleted GPX4, a critical anti-ferroptosis repressor. Similarly, transcriptomic analysis of human iPSC-derived Schwann cells revealed elevated ferroptosis activation and cellular stress markers in CMT1A. We propose that chronic, sublethal ferroptotic stress, mediated by lipid peroxide accumulation, depletes antioxidant defenses in CMT1A Schwann cells, leading to decompensation with age, manifesting as symptomatic disease. These results emphasize ferroptosis as a driver of CMT1A pathology, potentially revealing a new therapeutic path.
Collapse
Affiliation(s)
- Jacob B. White
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (J.B.W.); (K.L.S.)
| | - Kayla L. Sanchez
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (J.B.W.); (K.L.S.)
| | - Antonio Currais
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA; (A.C.); (D.S.-C.)
| | - David Soriano-Castell
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA; (A.C.); (D.S.-C.)
| | - Pamela Maher
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA; (A.C.); (D.S.-C.)
| | - Salvador Soriano
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (J.B.W.); (K.L.S.)
| |
Collapse
|
82
|
Ko CC, Yang PM. Hypoxia-induced MIR31HG expression promotes partial EMT and basal-like phenotype in pancreatic ductal adenocarcinoma based on data mining and experimental analyses. J Transl Med 2025; 23:305. [PMID: 40065368 PMCID: PMC11895263 DOI: 10.1186/s12967-025-06292-x] [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: 10/28/2024] [Accepted: 02/23/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is the most common and aggressive type of pancreatic cancer, with a five-year survival rate below 8%. Its high mortality is largely due to late diagnosis, metastatic potential, and resistance to therapy. Epithelial-mesenchymal transition (EMT) plays a key role in metastasis, enabling cancer cells to become mobile. Partial EMT, where cells maintain both epithelial and mesenchymal traits, is more frequent in tumors than complete EMT and contributes to cancer progression. The long non-coding RNA MIR31 host gene (MIR31HG) has recently emerged as a critical factor in PDAC oncogenesis. This study aimed to investigate MIR31HG's role in partial EMT and its association with the basal-like PDAC subtype. METHODS We analyzed the relationship between MIR31HG expression, partial EMT, and the basal-like subtype of PDAC by integrating data from public databases. We reanalyzed public data from PDAC patient-derived organoids to assess MIR31HG expression and gene signatures under hypoxic and normoxic conditions. RNA sequencing and bioinformatics analyses, including gene set enrichment analysis (GSEA), were used to investigate differentially expressed genes and pathway enrichments. EMT, partial EMT, and hypoxia scores were calculated based on the expression levels of specific gene sets. RESULTS We observed that MIR31HG overexpression strongly correlates with higher partial EMT scores and the stabilization of the epithelial phenotype in PDAC. MIR31HG is highly expressed in the basal-like subtype of PDAC, which exhibits partial EMT traits. Hypoxia, a hallmark of basal-like PDAC, was shown to significantly induce MIR31HG expression, thereby promoting the basal-like phenotype and partial EMT. In patient-derived organoids, hypoxic conditions increased MIR31HG expression and enhanced basal-like and partial EMT gene signatures, while normoxia reduced these expressions. These findings suggest that hypoxia-induced MIR31HG expression plays a crucial role in driving the aggressive basal-like subtype of PDAC. CONCLUSIONS Our results indicate that MIR31HG is crucial in regulating PDAC progression, particularly in the aggressive basal-like subtype associated with hypoxia and partial EMT. Targeting the MIR31HG-mediated network may offer a novel therapeutic approach to combat hypoxia-driven PDAC.
Collapse
Affiliation(s)
- Ching-Chung Ko
- Department of Medical Imaging, Chi Mei Medical Center, Tainan, 71004, Taiwan
- Department of Health and Nutrition, Chia Nan University of Pharmacy and Science, Tainan, 71710, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
| | - Pei-Ming Yang
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, No. 301, Yuantong Rd., Zhonghe Dist., New Taipei City, 235603, Taiwan.
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, 11031, Taiwan.
- TMU Research Center of Cancer Translational Medicine, Taipei, 11031, Taiwan.
- Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei, 11696, Taiwan.
- Taipei Cancer Center, Taipei Medical University (TMU) and Affiliated Hospitals Pancreatic Cancer Groups, Taipei Medical University, Taipei, 11031, Taiwan.
| |
Collapse
|
83
|
Jaiswal A, Negi M, Choi EH, Kaushik NK, Kaushik N. Upstream-binding protein-1 promotes breast tumorigenesis by inducing NRG2-mediated metastasis, plasticity, and macrophage polarization. Int J Biol Macromol 2025; 307:141915. [PMID: 40064277 DOI: 10.1016/j.ijbiomac.2025.141915] [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: 10/21/2024] [Revised: 03/07/2025] [Accepted: 03/07/2025] [Indexed: 03/18/2025]
Abstract
Upstream binding protein 1 (UBP1) is a transcription factor (TF) of the CP2/grainyhead family involved in various biological processes, including cancer cell proliferation, differentiation, and embryonic development. While other mammalian grainyhead-like TFs have been linked to different cancers, including breast cancer (BC), the role of UBP1 in BC remains unexplored. In this study, we provide a preliminary investigation into the novel functions of UBP1 in BC. Using online database screening, we first demonstrated that elevated UBP1 levels in breast carcinoma are associated with poor prognosis and adverse clinical outcomes. We further showed that UBP1 promotes epithelial-mesenchymal transition (EMT) and stemness in BC cells while regulating key signaling pathways, including the PI3K-Akt. Additionally, UBP1 modulates tumor metastasis by influencing tumor-associated macrophage (TAM) polarization, promoting an immunosuppressive macrophage phenotype, and driving tumor progression. Our findings highlight UBP1's pivotal role in BC progression through multiple mechanisms, including EMT induction, stemness maintenance, and macrophage polarization via activation of the NRG2/Akt axis. Moreover, higher UBP1 expression correlates with lower overall and recurrence-free survival, underscoring its potential as a prognostic marker and therapeutic target for aggressive BC.
Collapse
Affiliation(s)
- Apurva Jaiswal
- Plasma Bioscience Research Center/Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Republic of Korea
| | - Manorma Negi
- Plasma Bioscience Research Center/Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Republic of Korea
| | - Eun Ha Choi
- Plasma Bioscience Research Center/Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Republic of Korea.
| | - Nagendra Kumar Kaushik
- Plasma Bioscience Research Center/Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Republic of Korea.
| | - Neha Kaushik
- Department of Biotechnology, College of Engineering, The University of Suwon, Hwaseong 18323, Republic of Korea.
| |
Collapse
|
84
|
Ishchenko Y, Jeng AT, Feng S, Nottoli T, Manriquez-Rodriguez C, Nguyen KK, Carrizales MG, Vitarelli MJ, Corcoran EE, Greer CA, Myers SA, Koleske AJ. Heterozygosity for neurodevelopmental disorder-associated TRIO variants yields distinct deficits in behavior, neuronal development, and synaptic transmission in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.01.05.574442. [PMID: 39131289 PMCID: PMC11312463 DOI: 10.1101/2024.01.05.574442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Genetic variants in TRIO are associated with neurodevelopmental disorders (NDDs) including schizophrenia (SCZ), autism spectrum disorder (ASD) and intellectual disability. TRIO uses its two guanine nucleotide exchange factor (GEF) domains to activate GTPases (GEF1: Rac1 and RhoG; GEF2: RhoA) that control neuronal development and connectivity. It remains unclear how discrete TRIO variants differentially impact these neurodevelopmental events. Here, we investigate how heterozygosity for NDD-associated Trio variants - +/K1431M (ASD), +/K1918X (SCZ), and +/M2145T (bipolar disorder, BPD) - impact mouse behavior, brain development, and synapse structure and function. Heterozygosity for different Trio variants impacts motor, social, and cognitive behaviors in distinct ways that model clinical phenotypes in humans. Trio variants differentially impact head and brain size, with corresponding changes in dendritic arbors of motor cortex layer 5 pyramidal neurons (M1 L5 PNs). Although neuronal structure was only modestly altered in the Trio variant heterozygotes, we observe significant changes in synaptic function and plasticity. We also identified distinct changes in glutamate synaptic release in +/K1431M and +/M2145T cortico-cortical synapses. The TRIO K1431M GEF1 domain has impaired ability to promote GTP exchange on Rac1, but +/K1431M mice exhibit increased Rac1 activity, associated with increased levels of the Rac1 GEF Tiam1. Acute Rac1 inhibition with NSC23766 rescued glutamate release deficits in +/K1431M variant cortex. Our work reveals that discrete NDD-associated Trio variants yield overlapping but distinct phenotypes in mice, demonstrates an essential role for Trio in presynaptic glutamate release, and underscores the importance of studying the impact of variant heterozygosity in vivo.
Collapse
Affiliation(s)
- Yevheniia Ishchenko
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Amanda T Jeng
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT, USA
| | - Shufang Feng
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Department of Gerontology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Timothy Nottoli
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA
| | | | - Khanh K Nguyen
- Laboratory for Immunochemical Circuits, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Melissa G Carrizales
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Matthew J Vitarelli
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Ellen E Corcoran
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Charles A Greer
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Samuel A Myers
- Laboratory for Immunochemical Circuits, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Anthony J Koleske
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT, USA
| |
Collapse
|
85
|
Matthews S, Nikoonejad Fard V, Tollis M, Seoighe C. Variable Gene Copy Number in Cancer-Related Pathways Is Associated With Cancer Prevalence Across Mammals. Mol Biol Evol 2025; 42:msaf056. [PMID: 40112176 PMCID: PMC11954591 DOI: 10.1093/molbev/msaf056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/06/2025] [Accepted: 02/10/2025] [Indexed: 03/22/2025] Open
Abstract
Cancer is a disease of multicellularity, observed across the tree of life. In principle, animals with larger body sizes and longer lifespans should be at increased risk of developing cancer. However, there is no strong relationship between these traits and cancer across mammals. Previous studies have proposed that increased copy number of cancer-related genes may enhance the robustness of cancer suppression pathways in long-lived mammals, but these studies have not extended beyond known cancer-related genes. In this study, we conducted a phylogenetic generalized least squares analysis to test for associations between copy number of all protein-coding genes and longevity, body size, and cancer prevalence across 94 species of mammals. In addition to investigating the copy number of individual genes, we tested sets of related genes for a relationship between the aggregated gene copy number of the set and these traits. We did not find strong evidence to support the hypothesis that adaptive changes in gene copy number contribute to the lack of correlation between cancer prevalence and body size or lifespan. However, we found several biological processes where aggregate copy number was associated with malignancy rate. The strongest association was for the gene set relating to transforming growth factor beta, a cytokine that plays a role in cancer progression. Overall, this study provides a comprehensive evaluation of the role of gene copy number in adaptation to body size and lifespan and sheds light on the contribution of gene copy number to variation in cancer prevalence across mammals.
Collapse
Affiliation(s)
- Sophie Matthews
- School of Mathematical and Statistical Science, University of Galway, Galway, Ireland
- The SFI Centre for Research Training in Genomics Data Science, University of Galway, Galway, Ireland
| | - Vahid Nikoonejad Fard
- School of Informatics, Computing and Cyber Systems, Northern Arizona University, Flagstaff, AZ, USA
| | - Marc Tollis
- School of Informatics, Computing and Cyber Systems, Northern Arizona University, Flagstaff, AZ, USA
- Arizona Cancer Evolution Center, Arizona State University, Tempe, AZ, USA
| | - Cathal Seoighe
- School of Mathematical and Statistical Science, University of Galway, Galway, Ireland
- The SFI Centre for Research Training in Genomics Data Science, University of Galway, Galway, Ireland
| |
Collapse
|
86
|
Dill-McFarland KA, Peterson G, Lim PN, Skerrett S, Hawn TR, Rothchild AC, Campo M. Shared and distinct responses of human and murine alveolar macrophages and monocyte-derived macrophages to Mycobacterium tuberculosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.28.640814. [PMID: 40093075 PMCID: PMC11908159 DOI: 10.1101/2025.02.28.640814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Macrophages serve as important sites of bacterial replication and host immune response during Mycobacterium tuberculosis (Mtb) infection with distinct roles for alveolar macrophages (AMs) early in infection and monocyte-derived (MDMs) during later stages of disease. Here, we leverage data from human and mouse models to perform a cross-species analysis of macrophage responses to Mtb infection. Overall, we find that both subsets of human and murine macrophages mount a strong interferon response to Mtb infection. However, AM across both species do not generate as strong a pro-inflammatory response as human MDMs or murine bone marrow-derived macrophages (BMDMs), as characterized by TNFA signaling and inflammatory response pathways. Interestingly, AMs from mice that were previously vaccinated with BCG (scBCG) or from a model of contained TB (coMtb) had Mtb responses that were more similar to human AMs than control mice. We also identify species-specific pathways altered by infection differently in mouse and human macrophages, specifically in pathways related to cholesterol in AMs as well as MYC targets and Hedgehog signaling in MDMs/BMDMs. Lastly, to investigate downstream effects of the macrophage interferon responses, we examine macrophage expression of IL-10, an immunosuppressive cytokine induced by Type I Interferons, and c-Maf, a transcription factor required for IL-10 expression in myeloid cells. We find that c-Maf and IL-10 have significantly lower expression in AMs compared to MDMs in both humans and mice, suggesting one possible mechanism by which AMs mount a stronger interferon response following Mtb infection. Overall, these results highlight the dynamics of innate myeloid responses over the course of Mtb infection and the benefit of a combined analysis across species to reveal conserved and unique responses.
Collapse
|
87
|
Kumari P, Sullivan LM, Li Z, Parker Conquest E, Cornforth E, Jayakumar R, Hu N, Alexander Sizemore J, McKee BB, Kitchen RR, González-Pérez P, Linville C, Castro K, Gutierrez H, Samaan S, Townsend EL, Darras BT, Rutkove SB, Iannaccone ST, Clemens PR, Puwanant A, Das S, Wheeler TM. Analysis of human urinary extracellular vesicles reveals disordered renal metabolism in myotonic dystrophy type 1. Nat Commun 2025; 16:2158. [PMID: 40044661 PMCID: PMC11882899 DOI: 10.1038/s41467-025-56479-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 01/17/2025] [Indexed: 03/09/2025] Open
Abstract
Chronic kidney disease (CKD) and the genetic disorder myotonic dystrophy type 1 (DM1) each are associated with progressive muscle wasting, whole-body insulin resistance, and impaired systemic metabolism. However, CKD is undocumented in DM1 and the molecular pathogenesis driving DM1 is unknown to involve the kidney. Here we use urinary extracellular vesicles (EVs), RNA sequencing, droplet digital PCR, and predictive modeling to identify downregulation of metabolism transcripts Phosphoenolpyruvate carboxykinase-1, 4-Hydroxyphenylpyruvate dioxygenase, Dihydropyrimidinase, Glutathione S-transferase alpha-1, Aminoacylase-1, and Electron transfer flavoprotein B in DM1. Expression of these genes localizes to the kidney, especially the proximal tubule, and correlates with muscle strength and function. In DM1 autopsy kidney tissue, characteristic ribonuclear inclusions are evident throughout the nephron. We show that urinary organic acids and acylglycines are elevated in DM1, and correspond to enzyme deficits of downregulated genes. Our study identifies a previously unrecognized site of DM1 molecular pathogenesis and highlights the potential of urinary EVs as biomarkers of renal and metabolic disturbance in these individuals.
Collapse
Affiliation(s)
- Preeti Kumari
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Lauren M Sullivan
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Zhaozhi Li
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - E Parker Conquest
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Elizabeth Cornforth
- School of Health and Rehabilitation Sciences, Massachusetts General Hospital Institute of Health Professions, Boston, MA, USA
| | - Rojashree Jayakumar
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ningyan Hu
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - J Alexander Sizemore
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Brigham B McKee
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Robert R Kitchen
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Paloma González-Pérez
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Constance Linville
- Department of Neurology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Karla Castro
- Department of Pediatrics, University of Texas Southwestern, Dallas, TX, USA
| | - Hilda Gutierrez
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Soleil Samaan
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Elise L Townsend
- School of Health and Rehabilitation Sciences, Massachusetts General Hospital Institute of Health Professions, Boston, MA, USA
| | - Basil T Darras
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Seward B Rutkove
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Susan T Iannaccone
- Department of Pediatrics, University of Texas Southwestern, Dallas, TX, USA
| | - Paula R Clemens
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
- Neurology Section, Veteran's Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA
| | - Araya Puwanant
- Department of Neurology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Sudeshna Das
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Thurman M Wheeler
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
88
|
Zeballos L, García-Peral C, Ledesma MM, Auzmendi J, Lazarowski A, López DE. Changes in the Proteomic Profile After Audiogenic Kindling in the Inferior Colliculus of the GASH/Sal Model of Epilepsy. Int J Mol Sci 2025; 26:2331. [PMID: 40076950 PMCID: PMC11900993 DOI: 10.3390/ijms26052331] [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: 01/22/2025] [Revised: 02/27/2025] [Accepted: 03/02/2025] [Indexed: 03/14/2025] Open
Abstract
Epilepsy is a multifaceted neurological disorder characterized by recurrent seizures and associated with molecular and immune alterations in key brain regions. The GASH/Sal (Genetic Audiogenic Seizure Hamster, Salamanca), a genetic model for audiogenic epilepsy, provides a powerful tool to study seizure mechanisms and resistance in predisposed individuals. This study investigates the proteomic and immune responses triggered by audiogenic kindling in the inferior colliculus, comparing non-responder animals exhibiting reduced seizure severity following repeated stimulation versus GASH/Sal naïve hamsters. To assess auditory pathway functionality, Auditory Brainstem Responses (ABRs) were recorded, revealing reduced neuronal activity in the auditory nerve of non-responders, while central auditory processing remained unaffected. Cytokine profiling demonstrated increased levels of proinflammatory markers, including IL-1 alpha (Interleukin-1 alpha), IL-10 (Interleukin-10), and TGF-beta (Transforming Growth Factor beta), alongside decreased IGF-1 (Insulin-like Growth Factor 1) levels, highlighting systemic inflammation and its interplay with neuroprotection. Building on these findings, a proteomic analysis identified 159 differentially expressed proteins (DEPs). Additionally, bioinformatic approaches, including Gene Set Enrichment Analysis (GSEA) and Weighted Gene Co-expression Network Analysis (WGCNA), revealed disrupted pathways related to metabolic and inflammatory epileptic processes and a module potentially linked to a rise in the threshold of seizures, respectively. Differentially expressed genes, identified through bioinformatic and statistical analyses, were validated by RT-qPCR. This confirmed the upregulation of six genes (Gpc1-Glypican-1; Sdc3-Syndecan-3; Vgf-Nerve Growth Factor Inducible; Cpne5-Copine 5; Agap2-Arf-GAP with GTPase domain, ANK repeat, and PH domain-containing protein 2; and Dpp8-Dipeptidyl Peptidase 8) and the downregulation of two (Ralb-RAS-like proto-oncogene B-and S100b-S100 calcium-binding protein B), aligning with reduced seizure severity. This study may uncover key proteomic and immune mechanisms underlying seizure susceptibility, providing possible novel therapeutic targets for refractory epilepsy.
Collapse
Affiliation(s)
- Laura Zeballos
- Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, 37007 Salamanca, Spain; (L.Z.); (C.G.-P.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
- Departamento de Biología Celular y Patología, Facultad de Medicina, Universidad de Salamanca, 37007 Salamanca, Spain
| | - Carlos García-Peral
- Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, 37007 Salamanca, Spain; (L.Z.); (C.G.-P.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
- Departamento de Biología Celular y Patología, Facultad de Medicina, Universidad de Salamanca, 37007 Salamanca, Spain
| | - Martín M. Ledesma
- Unidad de Conocimiento Traslacional, Hospital de Alta Complejidad del Bicentenario Esteban Echeverría, Monte Grande B1842, Argentina;
- Hospital de Alta Complejidad en Red El Cruce Dr. N. C. Kirchner SAMIC, Florencio Varela B1888, Argentina
| | - Jerónimo Auzmendi
- Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires C1417, Argentina; (J.A.); (A.L.)
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Godoy Cruz M2290, Argentina
| | - Alberto Lazarowski
- Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires C1417, Argentina; (J.A.); (A.L.)
| | - Dolores E. López
- Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, 37007 Salamanca, Spain; (L.Z.); (C.G.-P.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
- Departamento de Biología Celular y Patología, Facultad de Medicina, Universidad de Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
89
|
Kogai H, Tsukamoto S, Koga M, Miyano M, Akagi T, Yamaguchi A, Mori K, Gotoh K, Nakazawa Y. Broad-Spectrum Efficacy of CEACAM6-Targeted Antibody-Drug Conjugate with BET Protein Degrader in Colorectal, Lung, and Breast Cancer Mouse Models. Mol Cancer Ther 2025; 24:392-405. [PMID: 39812376 DOI: 10.1158/1535-7163.mct-24-0444] [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: 06/08/2024] [Revised: 10/24/2024] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
Despite remarkable advances in cancer treatment, most solid cancers remain difficult to cure. We recently developed an antibody-drug conjugate (ADC; 84-EBET) for pancreatic cancer by using the carcinoembryonic antigen-related cell adhesion molecule 6 (CEACAM6) antibody #84.7 and the bromodomain and extra-terminal (BET) protein degrader EBET. In this study, we showed the overexpression of CEACAM6 in colorectal, lung, and breast cancers and the broad-spectrum efficacy of 84-EBET in mouse models of these cancers. In vitro assays using cancer organoids and cell lines of colorectal, lung, and breast cancers revealed that 84-EBET was more potent than ADCs with known approved payloads-DXd, SN38, and monomethyl auristatin E-or standard chemotherapies. In mouse studies, a single injection of 84-EBET induced marked regression of colorectal-, lung-, and breast cancer patient-derived xenograft tumors and cell line-derived xenograft tumors. Moreover, in mouse syngeneic colorectal cancer, lung cancer, and breast cancer models resistant to PD-1 antibody, the combination of 84-EBET and PD-1 antibody induced complete regression of most tumors. Mechanistically, 84-EBET degraded bromodomain-containing protein 4 in both cancer and stromal cells via bystander efficacy. It decreased stromal inflammatory phenotypes and increased activated T-cell numbers in tumors. These results demonstrate that delivering BET protein degraders to tumors and their microenvironments via a CEACAM6-targeted ADC may be effective against a wide range of solid cancers.
Collapse
Affiliation(s)
- Hiroyuki Kogai
- Tsukuba Research Laboratory, Eisai Co., Ltd., Ibaraki, Japan
| | | | - Minaho Koga
- Tsukuba Research Laboratory, Eisai Co., Ltd., Ibaraki, Japan
| | - Masayuki Miyano
- Tsukuba Research Laboratory, Eisai Co., Ltd., Ibaraki, Japan
| | - Tsuyoshi Akagi
- Tsukuba Research Laboratory, Eisai Co., Ltd., Ibaraki, Japan
| | | | - Kiyoshi Mori
- Department of Central Laboratory and Surgical Pathology, NHO Osaka National Hospital, Osaka, Japan
| | - Kunihito Gotoh
- Department of Surgery, NHO Osaka National Hospital, Osaka, Japan
| | - Youya Nakazawa
- Tsukuba Research Laboratory, Eisai Co., Ltd., Ibaraki, Japan
| |
Collapse
|
90
|
Finatto AN, Yang C, de Oliveira Costa M. Porcine β-defensin 5 (pBD-5) modulates the inflammatory and metabolic host intestinal response to infection. Sci Rep 2025; 15:7568. [PMID: 40038370 DOI: 10.1038/s41598-025-90688-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 02/14/2025] [Indexed: 03/06/2025] Open
Abstract
Swine dysentery (SD) presents considerable challenges to both animal welfare and pork industry sustainability. Control and prevention of SD rely on antibiotics and non-vaccine biosecurity practices. Host defense peptides (HDPs) have emerged as promising alternatives to treat and prevent such health concern. This study investigated the effects of porcine β-defensin 5 (pBD-5) and its potential host cytotoxicity, metabolic influence, gene expression modulation and direct antimicrobial activity on Brachyspira hyodysenteriae growth in vitro. pBD-5 does not directly inhibit B. hyodysenteriae growth or significantly alters the metabolic activity or membrane integrity of host cells, indicating no significant cytotoxicity at the tested concentrations. Host transcriptome sequencing revealed a reduction in the number of differentially expressed genes in cells exposed to B. hyodysenteriae following pBD-5 treatment, when compared to the pathogen alone, suggesting an immunomodulatory effect. Pathway analysis revealed the downregulation of immune-related pathways, including IL-17, toll-like receptor (TLR), and NOD-like receptor signalling pathways, upon pBD-5 exposure. Conversely, metabolic pathways such as ribosome, protein digestion and absorption, and renin-angiotensin system were upregulated by pBD-5 treatment, hinting at a role in producing and conserving energy during the challenge. While this study offers insights into the immunomodulatory effects of pBD-5, further research is necessary to elucidate its precise mechanisms and potential applications as an alternative treatment for infectious diseases.
Collapse
Affiliation(s)
- Arthur Nery Finatto
- Department of Large Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5E2, Canada
| | - Christine Yang
- Department of Large Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5E2, Canada
| | - Matheus de Oliveira Costa
- Department of Large Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5E2, Canada.
- Department of Population Health, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
91
|
Taylor A, Macaulay VM, Miossec MJ, Maurya AK, Buffa FM. GeneFEAST: the pivotal, gene-centric step in functional enrichment analysis interpretation. BIOINFORMATICS (OXFORD, ENGLAND) 2025; 41:btaf100. [PMID: 40037522 PMCID: PMC11919446 DOI: 10.1093/bioinformatics/btaf100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 02/05/2025] [Accepted: 02/28/2025] [Indexed: 03/06/2025]
Abstract
SUMMARY GeneFEAST, implemented in Python, is a gene-centric functional enrichment analysis summarization and visualization tool that can be applied to large functional enrichment analysis (FEA) results arising from upstream FEA pipelines. It produces a systematic, navigable HTML report, making it easy to identify sets of genes putatively driving multiple enrichments and to explore gene-level quantitative data first used to identify input genes. Further, GeneFEAST can juxtapose FEA results from multiple studies, making it possible to highlight patterns of gene expression amongst genes that are differentially expressed in at least one of multiple conditions, and which give rise to shared enrichments under those conditions. Thus, GeneFEAST offers a novel, effective way to address the complexities of linking up many overlapping FEA results to their underlying genes and data, advancing gene-centric hypotheses, and providing pivotal information for downstream validation experiments. AVAILABILITY AND IMPLEMENTATION GeneFEAST GitHub repository: https://github.com/avigailtaylor/GeneFEAST; Zenodo record: 10.5281/zenodo.14753734; Python Package Index: https://pypi.org/project/genefeast; Docker container: ghcr.io/avigailtaylor/genefeast.
Collapse
Affiliation(s)
- Avigail Taylor
- Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, United Kingdom
- Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, United Kingdom
- Nuffield Department of Surgical Sciences, University of Oxford, ORCRB, Oxford OX3 7DQ, United Kingdom
| | - Valentine M Macaulay
- Nuffield Department of Surgical Sciences, University of Oxford, ORCRB, Oxford OX3 7DQ, United Kingdom
| | - Matthieu J Miossec
- Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Anand K Maurya
- Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Francesca M Buffa
- Computational Biology & Integrative Genomics Lab, Department of Oncology, University of Oxford, ORCRB, Oxford OX3 7DQ, United Kingdom
| |
Collapse
|
92
|
Dumesic PA, Wilensky SE, Bose S, Van Vranken JG, Gygi SP, Spiegelman BM. RBM43 controls PGC1α translation and a PGC1α-STING signaling axis. Cell Metab 2025; 37:742-757.e8. [PMID: 39965564 PMCID: PMC11885043 DOI: 10.1016/j.cmet.2025.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 10/17/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025]
Abstract
Obesity is associated with systemic inflammation that impairs mitochondrial function. This disruption curtails oxidative metabolism, limiting adipocyte lipid metabolism and thermogenesis, a metabolically beneficial program that dissipates chemical energy as heat. Here, we show that PGC1α, a key governor of mitochondrial biogenesis, is negatively regulated at the level of its mRNA translation by the RNA-binding protein RBM43. RBM43 is induced by inflammatory cytokines and suppresses mitochondrial biogenesis in a PGC1α-dependent manner. In mice, adipocyte-selective Rbm43 disruption elevates PGC1α translation and oxidative metabolism. In obesity, Rbm43 loss improves glucose tolerance, reduces adipose inflammation, and suppresses activation of the innate immune sensor cGAS-STING in adipocytes. We further identify a role for PGC1α in safeguarding against cytoplasmic accumulation of mitochondrial DNA, a cGAS ligand. The action of RBM43 defines a translational regulatory axis by which inflammatory signals dictate cellular energy metabolism and contribute to metabolic disease pathogenesis.
Collapse
Affiliation(s)
- Phillip A Dumesic
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Sarah E Wilensky
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Symanthika Bose
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | | | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Bruce M Spiegelman
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
93
|
Rickelton K, Ely JJ, Hopkins WD, Hof PR, Sherwood CC, Bauernfeind AL, Babbitt CC. Transcriptomic changes across subregions of the primate cerebellum support the evolution of uniquely human behaviors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.03.641249. [PMID: 40093170 PMCID: PMC11908169 DOI: 10.1101/2025.03.03.641249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Background Compared to other primates, humans display unique behaviors including language and complex tool use. These abilities are made possible in part by the cerebellum. This region of the hindbrain, comprising the flocculus, vermis, and lateral hemispheres, has expanded throughout primate evolution, particularly in great apes. Given the cerebellum's architecture-differing in connectivity, neuron content, and functions across subregions-examining subregional differences is crucial to understanding its evolutionary trajectory. Results We performed bulk RNA-seq across samples from six primate species, representing 40-50 million years of evolutionary history, across four subregions of the cerebellum (vermis, flocculus, right lateral hemisphere, left lateral hemisphere). We analyzed changes in gene expression with respect to evolutionary relationships via the Ornstein-Uhlenbeck model and found that, on average, 8.5% of orthologous genes are differentially expressed in humans relative to other non-human primates. Subregion-specific gene expression patterns reveal that the primate lateral hemispheres exhibit significant differences in synaptic activity and glucose metabolism, which in turn are highly implicated in neural processing. Conclusions This study provides a novel perspective on gene expression divergences across cerebellar subregions in multiple primate species, offering valuable insights into the evolution of this brain structure. Our findings reveal distinct subregional transcriptomic patterns, with the lateral hemispheres emerging as key sites of divergence across the six primate species. The enrichment of genes related to synaptic activity, glucose metabolism, locomotion, and vocalization highlights the cerebellum's crucial role in supporting the neural complexity underlying uniquely human and other species-specific primate behaviors.
Collapse
Affiliation(s)
- Katherine Rickelton
- Department of Biology, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - John J Ely
- Alamogordo Primate Facility, Holloman Air Force Base, NM 88330, USA
| | - William D Hopkins
- Department of Comparative Medicine, Michale E. Keeling Center for Comparative Medicine, The University of Texas M D Anderson Cancer Center, Bastrop, TX 78602, USA
| | - Patrick R Hof
- Nash Family Department of Neuroscience and Friedman Brain Institute, Center for Discovery and Innovation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
- New York Consortium in Evolutionary Primatology, New York, NY 10124, USA
| | - Chet C Sherwood
- Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, DC 20052, USA
| | - Amy L Bauernfeind
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Anthropology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Courtney C Babbitt
- Department of Biology, University of Massachusetts Amherst, Amherst, MA 01003, USA
| |
Collapse
|
94
|
Li Y, Zeng R, Huang Y, Zhuo Y, Huang J. Integrating machine learning and single-cell sequencing to identify shared biomarkers in type 1 diabetes mellitus and clear cell renal cell carcinoma. Front Oncol 2025; 15:1543806. [PMID: 40098701 PMCID: PMC11911197 DOI: 10.3389/fonc.2025.1543806] [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: 12/11/2024] [Accepted: 02/07/2025] [Indexed: 03/19/2025] Open
Abstract
Purpose Type 1 diabetes mellitus (T1DM), as an autoimmune disease, can increase susceptibility to clear cell renal cell carcinoma (ccRCC) due to its proinflammatory effects. ccRCC is characterized by its subtle onset and unfavorable prognosis. Thus, the aim of this study was to highlight prevention and early detection opportunities in high-risk populations by identifying common biomarkers for T1DM and ccRCC. Methods Based on multiple publicly available datasets, WGCNA was applied to identify gene modules closely associated with T1DM, which were then integrated with prognostic DEGs in ccRCC. Subsequently, the LASSO and SVM algorithms were employed to identify shared hub genes between the two diseases. Additionally, clinical samples were used to validate the expression patterns of these hub genes, and scRNA-seq data were utilized to analyze the cell types expressing these genes and to explore potential mechanisms of cell communication. Results Overall, three hub genes (KIF21A, PIGH, and RPS6KA2) were identified as shared biomarkers for TIDM and ccRCC. Analysis of clinical samples and multiple datasets revealed that KIF21A and PIGH were significantly downregulated and that PIG was upregulated in the disease group. KIF21A and PIGH are mainly expressed in NK and T cells, PRS6KA2 is mainly expressed in endothelial and epithelial cells, and the MIF signaling pathway may be related to hub genes. Conclusion Our results demonstrated the pivotal roles of hub genes in T1DM and ccRCC. These genes hold promise as novel biomarkers, offering potential avenues for preventive strategies and the development of new precision treatment modalities.
Collapse
Affiliation(s)
- Yi Li
- Department of Ultrasound, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Rui Zeng
- Department of Pathology, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Pathology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangzhou, China
| | - Yuhua Huang
- Department of Ultrasound, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yumin Zhuo
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jun Huang
- Department of Ultrasound, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
95
|
Lee HJ, Park SW, Lee JH, Chang SY, Oh SM, Mun S, Kang J, Park JE, Choi JK, Kim TI, Kim JY, Kim P. Differential cellular origins of the extracellular matrix of tumor and normal tissues according to colorectal cancer subtypes. Br J Cancer 2025:10.1038/s41416-025-02964-z. [PMID: 40032993 DOI: 10.1038/s41416-025-02964-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 01/07/2025] [Accepted: 02/13/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Understanding the proteomic-level heterogeneity of the tumor microenvironment (TME) in colorectal cancer (CRC) is crucial due to its well-known heterogeneity. While heterogenous CRC has been extensively characterized at the molecular subtype level, research into the functional heterogeneity of fibroblasts, particularly their relationship with extracellular matrix (ECM) alterations, remains limited. Addressing this gap is essential for a comprehensive understanding of CRC progression and the development of targeted therapies. METHODS 24 tissue samples from 21 CRC patients, along with adjacent normal tissues (NAT), were collected and decellularized using a detergent-based method to enrich the ECM component. Proteomic analysis of ECM-enriched samples was performed using tandem mass tag (TMT) spectrometry, followed by statistical analysis including differential expression protein (DEP) analysis. Single-cell RNA sequencing (scRNA-Seq) data from public datasets were integrated and analyzed to delineate cell states within the TME. Bulk tissue RNA-Seq and bioinformatics analysis, including consensus molecular subtype (CMS) classification and single-cell level deconvolution of TCGA bulk RNA-seq data, were conducted to further explore gene expression patterns and TME composition. RESULTS Differential cellular origin of the NAT and tumorous ECM proteins were identified, revealing 110 ECM proteins enriched in NAT and 28 ECM proteins in tumor tissues. Desmoplastic and WNT5A+ inflammatory fibroblasts were indicated as the sources of tumor-enriched ECM proteins, while ADAMDEC1+ expressing fibroblasts and PI16+ expressing fibroblast were identified as the sources of NAT-enriched ECM proteins. Deconvolution of bulk RNA-seq of CRC tissues discriminated CMS-specific fibroblast state, reflecting the biological traits of each CMS subtype. Specially, seven ECM genes specific to mesenchymal subtype (CMS4), including PI16+ fibroblast-related 4 genes (SFRP2, PRELP, OGN, SRPX) and desmoplastic fibroblast-related 3 genes (THBS2, CTHRC1, BGN), showed a significant association with poorer survival in patient with CRC. CONCLUSION We conducted an extracellular matrix (ECM)-focused profiling of the TME by integrating quantitative proteomics with single-cell RNA sequencing (scRNA-seq) data from CRC patients. We identified the ECM proteins of NAT and tumor tissue, and established a cell-matrisome database. We defined mesenchymal subtype-specific molecules associated with specific fibroblast subtypes showing a significant association with poorer survival in patients with CRC. Our ECM-focused profiling of tumor stroma provides new insights as indicators for biological processes and clinical endpoints.
Collapse
Affiliation(s)
- Hyun Jin Lee
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Sang Woo Park
- Korea Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Ochang, 28119, Republic of Korea
| | - Jun Hyeong Lee
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Shin Young Chang
- Department of Internal Medicine, Institute of Gastroenterology, Brain Korea 21 Project for Medical Science, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Sang Mi Oh
- Department of Internal Medicine, Institute of Gastroenterology, Brain Korea 21 Project for Medical Science, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Siwon Mun
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Junho Kang
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Jong-Eun Park
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
- SCL-KAIST Institute of Translational Research, KAIST, Daejeon, Republic of Korea
| | - Jung Kyoon Choi
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
- SCL-KAIST Institute of Translational Research, KAIST, Daejeon, Republic of Korea
| | - Tae Il Kim
- Department of Internal Medicine, Institute of Gastroenterology, Brain Korea 21 Project for Medical Science, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| | - Jin Young Kim
- Korea Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Ochang, 28119, Republic of Korea.
| | - Pilnam Kim
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea.
- SCL-KAIST Institute of Translational Research, KAIST, Daejeon, Republic of Korea.
| |
Collapse
|
96
|
DiCillo EB, Kountikov E, Zhu M, Lanker S, Harlow DE, Piette ER, Zhang W, Hayward B, Heuler J, Korich J, Bennett JL, Pisetsky D, Tedder T. Patterns of autoantibody expression in multiple sclerosis identified through development of an autoantigen discovery technology. J Clin Invest 2025; 135:e171948. [PMID: 40026247 DOI: 10.1172/jci171948] [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: 08/14/2023] [Accepted: 01/08/2025] [Indexed: 03/05/2025] Open
Abstract
Multiple sclerosis (MS) is a debilitating autoimmune disease of the CNS, which is characterized by demyelination and axonal injury and frequently preceded by a demyelinating event called clinically isolated syndrome (CIS). Despite the importance of B cells and autoantibodies in MS pathology, their target specificities remain largely unknown. For an agnostic and comprehensive evaluation of autoantibodies in MS, we developed and employed what we believe to be a novel autoantigen discovery technology, the Antigenome Platform. This Platform is a high-throughput assay comprising large-fragment (approximately 100 amino acids) cDNA libraries, phage display, serum antibody screening technology, and robust bioinformatics analysis pipelines. For autoantibody discovery, we assayed serum samples from CIS patients who received either placebo or treatment who were enrolled in the REFLEX clinical trial, which assessed the effects of IFN-β-1a (Rebif) clinical and MRI activity in patients with CIS. Serum autoantibodies from patients with CIS were significantly and reproducibly enriched for known and previously unreported protein targets; 166 targets were selected by over 10% of patients' sera. Further, 10 autoantibody biomarkers associated with disease activity and 17 associated with patient response to IFN-β-1a therapy. These findings indicate widespread autoantibody production in MS and provide biomarkers for continued study and prediction of disease progression.
Collapse
Affiliation(s)
- Europe B DiCillo
- Department of Integrated Immunobiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Evgueni Kountikov
- Department of Integrated Immunobiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Minghua Zhu
- Department of Integrated Immunobiology, Duke University Medical Center, Durham, North Carolina, USA
| | | | | | | | - Weiguo Zhang
- Department of Integrated Immunobiology, Duke University Medical Center, Durham, North Carolina, USA
| | | | - Joshua Heuler
- Department of Integrated Immunobiology, Duke University Medical Center, Durham, North Carolina, USA
| | | | - Jeffrey L Bennett
- Departments of Neurology and Ophthalmology, Programs in Neurosciences and Immunology - University of Colorado Anschutz Medical Campus; Aurora, Colorado, USA
| | - David Pisetsky
- Department of Integrated Immunobiology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Medicine, Duke University Medical Center and Medical Research Service, Veterans Administration Medical Center, Durham, North Carolina, USA
| | - Thomas Tedder
- Department of Integrated Immunobiology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
97
|
Roussis IM, Pearton DJ, Niazi U, Tsaknakis G, Papadopoulos GL, Cook R, Saqi M, Ragoussis J, Strouboulis J. A novel role for Friend of GATA1 (FOG-1) in regulating cholesterol transport in murine erythropoiesis. PLoS Genet 2025; 21:e1011617. [PMID: 40048486 PMCID: PMC11913303 DOI: 10.1371/journal.pgen.1011617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 03/17/2025] [Accepted: 02/12/2025] [Indexed: 03/18/2025] Open
Abstract
Friend of GATA1 (FOG-1) is an essential transcriptional co-factor of the master erythroid transcription factor GATA1. The knockout of the Zfpm1 gene, coding for FOG-1, results in early embryonic lethality due to anemia in mice, similar to the embryonic lethal phenotype of the Gata1 gene knockout. However, a detailed molecular analysis of the Zfpm1 knockout phenotype in erythropoiesis is presently incomplete. To this end, we used CRISPR/Cas9 to knockout Zfpm1 in mouse erythroleukemic (MEL) cells. Phenotypic characterization of DMSO-induced terminal erythroid differentiation showed that the Zfpm1 knockout MEL cells did not progress past the proerythroblast stage of differentiation. Expression profiling of the Zfpm1 knockout MEL cells by RNAseq showed a lack of up-regulation of erythroid-related gene expression profiles. Bioinformatic analysis highlighted cholesterol transport as a pathway affected in the Zfpm1 knockout cells. Moreover, we show that the cholesterol transporters Abca1 and Ldlr fail to be repressed during erythroid differentiation in Zfpm1 knockout cells, resulting in higher intracellular lipid levels and higher membrane fluidity. We also show that in FOG-1 knockout cells, the nuclear levels of SREBP2, a key transcriptional regulator of cholesterol biosynthesis and transport, are markedly increased. On the basis of these findings we propose that FOG-1 (and, potentially, GATA1) regulate cholesterol homeostasis during erythroid differentiation directly through the down regulation of cholesterol transport genes and indirectly, through the repression of the SREBP2 transcriptional activator of cholesterol homeostasis. Taken together, our work provides a molecular basis for understanding FOG-1 functions in erythropoiesis and reveals a novel role for FOG-1 in cholesterol transport.
Collapse
Affiliation(s)
- Ioannis-Marios Roussis
- Red Cell Haematology Lab, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
- Department of Biology, University of Crete, Heraklion, Crete, Greece
| | - David J. Pearton
- Red Cell Haematology Lab, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Umar Niazi
- Translational Bioinformatics, National Institute for Health Research Biomedical Centre, Guy’s and St Thomas’ NHS Foundation Trust and King’s College London, London, United Kingdom
| | - Grigorios Tsaknakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research & Technology Hellas, Heraklion, Crete, Greece
| | - Giorgio L. Papadopoulos
- Institute of Molecular Biology and Biotechnology, Foundation for Research & Technology Hellas, Heraklion, Crete, Greece
| | - Riley Cook
- Bone Marrow Failure Group, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Mansoor Saqi
- Translational Bioinformatics, National Institute for Health Research Biomedical Centre, Guy’s and St Thomas’ NHS Foundation Trust and King’s College London, London, United Kingdom
| | - Jiannis Ragoussis
- Department of Human Genetics, McGill University and McGill Genome Centre, Montreal, Quebec, Canada
| | - John Strouboulis
- Red Cell Haematology Lab, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| |
Collapse
|
98
|
Xie Y, Wang X, Cheng S, Liu W, Yi C, You Y, Zhang W, Wang Y, Tang E, Wang J, Hu W. RNAi screening of uncharacterized genes identifies promising druggable targets in Schistosoma japonicum. PLoS Pathog 2025; 21:e1013014. [PMID: 40153463 PMCID: PMC11977999 DOI: 10.1371/journal.ppat.1013014] [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: 10/25/2024] [Revised: 04/08/2025] [Accepted: 03/03/2025] [Indexed: 03/30/2025] Open
Abstract
Schistosomiasis affects more than 250 million people worldwide and is one of the neglected tropical diseases. Currently, the treatment of schistosomiasis relies on a single drug-praziquantel-which has led to increasing pressure from drug resistance. Therefore, there is an urgent need to find new treatments. The development of genome sequencing has provided valuable information for understanding the biology of schistosomes. In the genome of Schistosoma japonicum, approximately 11% of the protein-coding sequences are uncharacterized genes (UGs) annotated as "hypothetical protein" or "protein of unknown function." These poorly understood genes have been unjustifiably neglected, although some may be essential for the survival of the parasites and serve as potential drug targets. In this study, we systematically mined the highly expressed UGs in both genders of this parasite throughout key developmental stages in their mammalian host, using our previously published S. japonicum genome and RNA-seq data. By employing in vitro RNA interference (RNAi), we screened 126 UGs that lack homologs in Homo sapiens and identified 8 that are essential for the parasite vitality. We further investigated two UGs, Sjc_0002003 and Sjc_0009272, which resulted in the most severe phenotypes. Fluorescence in situ hybridization demonstrated that both genes were expressed throughout the body without sex bias. Silencing either Sjc_0002003 or Sjc_0009272 reduced the cell proliferation in the body. Furthermore, in vivo RNAi indicated both genes are required for the growth and survival of the parasites in the mammalian host. For Sjc_0002003, we further characterize the underlying molecular cause of the observed phenotype. Through RNA-seq analysis and functional studies, we revealed that silencing Sjc_0002003 reduces the expression of a series of intestinal genes, including Sjc_0007312 (hypothetical protein), Sjc_0008276 (vha-17), Sjc_0002942 (PLA2G15), and Sjc_0003646 (SJCHGC09134 protein), leading to gut dilation. Our work highlights the importance of UGs in schistosomes as promising targets for drug development in the treatment of the schistosomiasis.
Collapse
Affiliation(s)
- Yuxiang Xie
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Xiaoling Wang
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Shaoyun Cheng
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Wanling Liu
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Cun Yi
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Yanmin You
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Wei Zhang
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Yuepeng Wang
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Enlu Tang
- College of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Jipeng Wang
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Wei Hu
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
- College of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, China
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), NHC Key Laboratory of Parasite and Vector Biology, WHO Collaborating Center for Tropical Diseases, National Center for International Research on Tropical Diseases, Shanghai, China
| |
Collapse
|
99
|
Bu A, Afghah F, Castro N, Bawa M, Kohli S, Shah K, Rios B, Butty V, Raman R. Actuating Extracellular Matrices Decouple the Mechanical and Biochemical Effects of Muscle Contraction on Motor Neurons. Adv Healthc Mater 2025; 14:e2403712. [PMID: 39523700 PMCID: PMC11874633 DOI: 10.1002/adhm.202403712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/03/2024] [Indexed: 11/16/2024]
Abstract
Emerging in vivo evidence suggests that repeated muscle contraction, or exercise, impacts peripheral nerves. However, the difficulty of isolating the muscle-specific impact on motor neurons in vivo, as well as the inability to decouple the biochemical and mechanical impacts of muscle contraction in this setting, motivates investigating this phenomenon in vitro. This study demonstrates that tuning the mechanical properties of fibrin enables longitudinal culture of highly contractile skeletal muscle monolayers, enabling functional characterization of and long-term secretome harvesting from exercised tissues. Motor neurons stimulated with exercised muscle-secreted factors significantly upregulate neurite outgrowth and migration, with an effect size dependent on muscle contraction intensity. Actuating magnetic microparticles embedded within fibrin hydrogels enable dynamically stretching motor neurons and non-invasively mimicking the mechanical effects of muscle contraction. Interestingly, axonogenesis is similarly upregulated in both mechanically and biochemically stimulated motor neurons, but RNA sequencing reveals different transcriptomic signatures between groups, with biochemical stimulation having a greater impact on cell signaling related to axonogenesis and synapse maturation. This study leverages actuating extracellular matrices to robustly validate a previously hypothesized role for muscle contraction in regulating motor neuron growth and maturation from the bottom-up through both mechanical and biochemical signaling.
Collapse
Affiliation(s)
- Angel Bu
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Ferdows Afghah
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Nicolas Castro
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Maheera Bawa
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Sonika Kohli
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Karina Shah
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Brandon Rios
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Vincent Butty
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Ritu Raman
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| |
Collapse
|
100
|
Ramezani M, Weisbart E, Bauman J, Singh A, Yong J, Lozada M, Way GP, Kavari SL, Diaz C, Leardini E, Jetley G, Pagnotta J, Haghighi M, Batista TM, Pérez-Schindler J, Claussnitzer M, Singh S, Cimini BA, Blainey PC, Carpenter AE, Jan CH, Neal JT. A genome-wide atlas of human cell morphology. Nat Methods 2025; 22:621-633. [PMID: 39870862 PMCID: PMC11903339 DOI: 10.1038/s41592-024-02537-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 10/25/2024] [Indexed: 01/29/2025]
Abstract
A key challenge of the modern genomics era is developing empirical data-driven representations of gene function. Here we present the first unbiased morphology-based genome-wide perturbation atlas in human cells, containing three genome-wide genotype-phenotype maps comprising CRISPR-Cas9-based knockouts of >20,000 genes in >30 million cells. Our optical pooled cell profiling platform (PERISCOPE) combines a destainable high-dimensional phenotyping panel (based on Cell Painting) with optical sequencing of molecular barcodes and a scalable open-source analysis pipeline to facilitate massively parallel screening of pooled perturbation libraries. This perturbation atlas comprises high-dimensional phenotypic profiles of individual cells with sufficient resolution to cluster thousands of human genes, reconstruct known pathways and protein-protein interaction networks, interrogate subcellular processes and identify culture media-specific responses. Using this atlas, we identify the poorly characterized disease-associated TMEM251/LYSET as a Golgi-resident transmembrane protein essential for mannose-6-phosphate-dependent trafficking of lysosomal enzymes. In sum, this perturbation atlas and screening platform represents a rich and accessible resource for connecting genes to cellular functions at scale.
Collapse
Affiliation(s)
- Meraj Ramezani
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Erin Weisbart
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Julia Bauman
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Stanford University, Stanford, CA, USA
| | - Avtar Singh
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Genentech Department of Cellular and Tissue Genomics, South San Francisco, CA, USA
| | - John Yong
- Calico Life Sciences LLC, South San Francisco, CA, USA
| | - Maria Lozada
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Gregory P Way
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sanam L Kavari
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- University of Pennsylvania, Philadelphia, PA, USA
| | - Celeste Diaz
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Stanford University, Stanford, CA, USA
| | - Eddy Leardini
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Gunjan Jetley
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jenlu Pagnotta
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Thiago M Batista
- Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute, Cambridge, MA, USA
| | - Joaquín Pérez-Schindler
- Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute, Cambridge, MA, USA
| | - Melina Claussnitzer
- Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute, Cambridge, MA, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | | - Beth A Cimini
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Paul C Blainey
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biological Engineering, MIT, Cambridge, MA, USA
- Koch Institute for Integrative Research, MIT, Cambridge, MA, USA
| | | | - Calvin H Jan
- Calico Life Sciences LLC, South San Francisco, CA, USA
| | - James T Neal
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute, Cambridge, MA, USA.
| |
Collapse
|