1
|
Guo C, Tang H, Ren M, Zhang Y. BHLHE40-mediated RGS16 upregulation: a driver propelling gastric cancer progression via ferroptosis suppression. Hereditas 2025; 162:87. [PMID: 40413527 PMCID: PMC12102885 DOI: 10.1186/s41065-025-00447-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Accepted: 05/05/2025] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND Gastric cancer (GC), a malignant neoplasm that arises from the epithelium of the gastric mucosa, endangers patients' lives and health severely. Regulator of G-protein signaling 16 (RGS16) has been found to be correlated with the malignant progression of various cancers, and BHLHE40 is highly expressed in GC. However, it remains unclear whether there is a regulatory mechanism between the them. METHODS The bioinformatics tools were applied to assess the differentially expressed genes in GC. Next, the expression levels of mRNA and protein were evaluated by qRT-PCR and Western blot. Cellular behaviors were assessed using CCK-8, EdU, Transwell, and flow cytometry assays. Meanwhile, the ferroptosis-related indicators were measured. Subsequently, the xenograft models were set up to estimate the role of RGS16 in vivo. Besides, the interaction between BHLHE40 and RGS16 was determined using ChIP assay and dual-luciferase reporter assay. RESULTS RGS16 exhibited an upregulated pattern in GC. In addition, silencing RGS16 impeded the proliferation, migration and invasion of GC cells while reinforcing apoptosis and ferroptosis. Moreover, RGS16 boosted the growth of tumors in vivo. Furthermore, BHLHE40 could bind to RGS16 and positively regulate its expression. Overexpression of RGS16 reversed the effects of silencing BHLHE40 on GC cells. CONCLUSION BHLHE40 curbed ferroptosis and oxidative stress of GC cells by modulating the expression of RGS16, thereby facilitating the malignant progression of GC.
Collapse
Affiliation(s)
- Caiyun Guo
- Department of Gastroenterology, Xi'an International Medical Center Hospital, Xi'an, 710117, Shaanxi, China
| | - Hua Tang
- Department of Gastroenterology, Xi'an International Medical Center Hospital, Xi'an, 710117, Shaanxi, China.
| | - Maifang Ren
- Department of Gastroenterology, Xi'an International Medical Center Hospital, Xi'an, 710117, Shaanxi, China
| | - Yongli Zhang
- Department of Gastroenterology, Tongchuan People's Hospital, Tongchuan, 727100, Shaanxi, China.
| |
Collapse
|
2
|
Boiko JR, Ensbey KS, Waltner OG, Jenkins IC, Bhise SS, MacDonald KPA, Blazar BR, Hall AM, Gooley TA, Minnie SA, Lee SJ, Furlan SN, Hill GR. Defining pathogenic IL-17 and CSF-1 gene expression signatures in chronic graft-versus-host disease. Blood 2025; 145:2214-2228. [PMID: 39977705 DOI: 10.1182/blood.2024025337] [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: 05/08/2024] [Revised: 01/23/2025] [Accepted: 01/23/2025] [Indexed: 02/22/2025] Open
Abstract
ABSTRACT Chronic graft-versus-host disease (cGVHD) remains the leading cause of nonrelapse morbidity and mortality after allogeneic hematopoietic cell transplantation (HCT). Effective therapeutic agents targeting dysregulated cytokines including interleukin-17 (IL-17) and colony-stimulating factor 1 (CSF-1) have been defined in preclinical models of cGVHD, and efficacy in subsequent clinical trials has led to their recent US Food and Drug Administration approval. Despite this, these agents are effective in only a subset of patients, expensive, difficult to access outside the United States, and used in a trial-and-error fashion. The ability to readily discern druggable, dysregulated immunity in these patients is desperately needed to facilitate the selection of appropriate treatment and to potentially identify high-risk individuals for preemptive therapy. We used single-cell sequencing-based approaches in our informative preclinical cGVHD models to "reverse engineer" temporal IL-17 and CSF-1 signatures in mouse blood that could be used to interrogate patients. We defined distinct, nonintuitive IL-17 and CSF-1 signatures in mouse blood monocytes that could be identified in relevant monocyte populations within 70% of patients at diagnosis of cGVHD and in half of patients at day +100 after HCT who subsequently developed cGVHD. These signatures can now be evaluated prospectively in clinical studies to help delineate potential responder and nonresponders to relevant therapeutics targeting these pathways.
Collapse
Affiliation(s)
- Julie R Boiko
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Pediatrics, University of Washington, Seattle, WA
| | - Kathleen S Ensbey
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Olivia G Waltner
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Isaac C Jenkins
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Shruti S Bhise
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Kelli P A MacDonald
- Department of Infection and Inflammation, Queensland Institute of Medical Research Berghofer, Brisbane, Australia
| | - Bruce R Blazar
- Division of Blood and Marrow Transplant and Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - A Marcie Hall
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Ted A Gooley
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Simone A Minnie
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Stephanie J Lee
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Scott N Furlan
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Pediatrics, University of Washington, Seattle, WA
| | - Geoffrey R Hill
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
3
|
Mohammed A, Wang W, Arreola M, Solomon BD, Slepicka PF, Hubka KM, Nguyen HD, Zheng Z, Chavez MG, Yeh CY, Kim DK, Ma MR, Martin E, Li L, Pasca AM, Winn VD, Gifford CA, Kedlian VR, Park JE, Khatri P, Hollander GA, Roncarolo MG, Sebastiano V, Teichmann SA, Gentles AJ, Weinacht KG. Distinct type I and II interferon responses direct cortical and medullary thymic epithelial cell development. Sci Immunol 2025; 10:eado4720. [PMID: 40315299 DOI: 10.1126/sciimmunol.ado4720] [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: 02/12/2024] [Revised: 12/05/2024] [Accepted: 04/10/2025] [Indexed: 05/04/2025]
Abstract
Advances in genomics have redefined our understanding of thymic epithelial heterogeneity and architecture, yet signals driving thymic epithelial differentiation remain incompletely understood. Here, we elucidated pathways instructing human thymic epithelial cell development in the context of other anterior foregut-derived organs. Activation of interferon response gene regulatory networks distinguished epithelial cells of the thymus from those of other anterior foregut-derived organs. Thymic cortex and medulla epithelia displayed distinctive interferon-responsive signatures defined by lineage-specific chromatin accessibility. We explored the effects of type I and II interferons on thymic epithelial progenitor differentiation from induced pluripotent stem cells. Type II interferon was essential for expressing proteasome and antigen-presenting molecules, whereas type I or II interferons were essential for inducing different cytokines in thymic epithelial progenitor cells. Our findings suggest that interferons are critical to cortical and medullary thymic epithelial cell differentiation.
Collapse
Affiliation(s)
- Abdulvasey Mohammed
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94304, USA
| | - Wenqing Wang
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94304, USA
| | - Martin Arreola
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94304, USA
| | - Benjamin D Solomon
- Department of Pediatrics, Division of Allergy and Immunology, Stanford School of Medicine, Stanford, CA 94304, USA
| | - Priscila F Slepicka
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94304, USA
| | - Kelsea M Hubka
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94304, USA
| | - Hanh Dan Nguyen
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94304, USA
| | - Zihao Zheng
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94304, USA
| | - Michael G Chavez
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Christine Y Yeh
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
- Department of Medicine, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Doo Kyung Kim
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Michael R Ma
- Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA
- Department of Cardiothoracic Surgery, Division of Pediatric Cardiac Surgery, Stanford University, Stanford, CA 94304, USA
| | - Elisabeth Martin
- Department of Cardiothoracic Surgery, Division of Pediatric Cardiac Surgery, Stanford University, Stanford, CA 94304, USA
| | - Li Li
- Department of Pediatrics, Division of Neonatology, Stanford School of Medicine, Stanford, CA 94304, USA
| | - Anca M Pasca
- Department of Pediatrics, Division of Neonatology, Stanford School of Medicine, Stanford, CA 94304, USA
| | - Virginia D Winn
- Department of Obstetrics and Gynecology, Stanford School of Medicine, Stanford, CA, USA
| | - Casey A Gifford
- Department of Pediatrics, Division of Cardiology, Stanford School of Medicine, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Veronika R Kedlian
- Wellcome Sanger Institute, Cambridge, UK
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | | | - Purvesh Khatri
- Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, CA 94304, USA
- Center for Biomedical Informatics Research, Department of Medicine, School of Medicine, Stanford University, CA 94305, USA
| | - Georg A Hollander
- Department of Pediatrics and Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
- Department of Biosystems and Engineering, ETH Zurich, Zurich, Switzerland
- Botnar Institute of Immune Engineering, Basel, Switzerland
| | - Maria Grazia Roncarolo
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94304, USA
| | - Vittorio Sebastiano
- Department of Obstetrics and Gynecology, Stanford School of Medicine, Stanford, CA, USA
| | - Sarah A Teichmann
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
- CIFAR Macmillan Multi-scale Human Programme, CIFAR, Toronto, Canada
| | - Andrew J Gentles
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
- Department of Pathology, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Katja G Weinacht
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94304, USA
| |
Collapse
|
4
|
Zelic M, Blazier A, Pontarelli F, LaMorte M, Huang J, Tasdemir-Yilmaz OE, Ren Y, Ryan SK, Shapiro C, Morel C, Krishnaswami P, Levit M, Sood D, Chen Y, Gans J, Tang X, Hsiao-Nakamoto J, Huang F, Zhang B, Berry JD, Bangari DS, Gaglia G, Ofengeim D, Hammond TR. Single-cell transcriptomic and functional studies identify glial state changes and a role for inflammatory RIPK1 signaling in ALS pathogenesis. Immunity 2025; 58:961-979.e8. [PMID: 40132594 DOI: 10.1016/j.immuni.2025.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 10/31/2024] [Accepted: 02/25/2025] [Indexed: 03/27/2025]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease characterized by motor neuron loss. Microglia and astrocyte-driven neuroinflammation is prominent in ALS, but the cell state dynamics and pathways driving disease remain unclear. We performed single-nucleus RNA sequencing of ALS spinal cords and identified altered glial cell states, including increased expression of inflammatory and glial activation markers. Many of these signals converged on the inflammation and cell death regulator receptor-interacting protein kinase 1 (RIPK1) and the necroptotic cell death pathway. In superoxide dismutase 1 (SOD1)G93A mice, blocking RIPK1 kinase activity delayed symptom onset and motor impairment and modulated glial responses. We used human induced pluripotent stem cell (iPSC)-derived motor neuron, astrocyte, and microglia tri-cultures to identify potential biomarkers that are secreted upon RIPK1 activation in vitro and modulated by RIPK1 inhibition in the cerebrospinal fluid (CSF) of people with ALS. These data reveal ALS-enriched glial populations associated with inflammation and suggest a deleterious role for neuroinflammatory signaling in ALS pathogenesis.
Collapse
Affiliation(s)
- Matija Zelic
- Sanofi, Rare and Neurologic Diseases, Cambridge, MA 02141, USA.
| | - Anna Blazier
- Sanofi, Rare and Neurologic Diseases, Cambridge, MA 02141, USA
| | | | - Michael LaMorte
- Sanofi, Rare and Neurologic Diseases, Cambridge, MA 02141, USA
| | - Jeremy Huang
- Sanofi, Precision Medicine and Computational Biology, Cambridge, MA 02141, USA
| | | | - Yi Ren
- Sanofi, Rare and Neurologic Diseases, Cambridge, MA 02141, USA
| | - Sean K Ryan
- Sanofi, Rare and Neurologic Diseases, Cambridge, MA 02141, USA
| | - Cynthia Shapiro
- Sanofi, Global Discovery Pathology and Multimodal Imaging, Cambridge, MA 02141, USA
| | - Caroline Morel
- Sanofi, Global Discovery Pathology and Multimodal Imaging, Cambridge, MA 02141, USA
| | | | - Mikhail Levit
- Sanofi, Precision Medicine and Computational Biology, Cambridge, MA 02141, USA
| | - Disha Sood
- Sanofi, Rare and Neurologic Diseases, Cambridge, MA 02141, USA
| | - Yao Chen
- Sanofi, Precision Medicine and Computational Biology, Cambridge, MA 02141, USA
| | - Joseph Gans
- Sanofi, Precision Medicine and Computational Biology, Cambridge, MA 02141, USA
| | - Xinyan Tang
- Denali Therapeutics, Inc., South San Francisco, CA 94080, USA
| | | | - Fen Huang
- Denali Therapeutics, Inc., South San Francisco, CA 94080, USA
| | - Bailin Zhang
- Sanofi, Precision Medicine and Computational Biology, Cambridge, MA 02141, USA
| | - James D Berry
- Healey Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Dinesh S Bangari
- Sanofi, Global Discovery Pathology and Multimodal Imaging, Cambridge, MA 02141, USA
| | - Giorgio Gaglia
- Sanofi, Precision Medicine and Computational Biology, Cambridge, MA 02141, USA
| | | | | |
Collapse
|
5
|
Willems SH, Qian S, Lång P, Overtoom BE, Alimostafazadeh S, Fuentes-Mateos R, Vasse GF, van der Veen TA, Vlasma J, de Jager MH, Guryev V, Fejer G, Andersson G, Melgert BN. TRAPping the effects of tobacco smoking: the regulation and function of Acp5 expression in lung macrophages. Am J Physiol Lung Cell Mol Physiol 2025; 328:L497-L511. [PMID: 39993028 DOI: 10.1152/ajplung.00157.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 09/02/2024] [Accepted: 02/17/2025] [Indexed: 02/26/2025] Open
Abstract
Tartrate-resistant acid phosphatase [TRAP, gene acid phosphatase 5 (Acp5; gene name for TRAP)] is highly expressed in alveolar macrophages with proposed roles in lung inflammation and lung fibrosis development. We previously showed that its expression and activity are higher in lung macrophages of smokers and patients with chronic obstructive pulmonary disease (COPD), suggesting involvement in smoke-induced lung damage. In this study, we explored the function of TRAP and regulation of its different mRNA transcripts (Acp5 201-206) in lung tissue exposed to cigarette smoke to elucidate its function in alveolar macrophages. In mice exposed to cigarette smoke or air for 4-6 wk, higher Acp5 mRNA expression in lung tissue after smoking was mainly driven by transcript Acp5-202, which originates from macrophages. The expression of Acp5-202 correlated with transcription factors previously found to drive proliferation of macrophages. Treating fetal liver progenitor-derived alveolar-like macrophages [Max Planck Institute (MPI; macrophages derived from fetal liver progenitors) macrophages] with cigarette smoke extract resulted in more proliferation compared with nontreated cells. In contrast, Acp5-deficient MPI macrophages and MPI macrophages treated with a TRAP inhibitor proliferated significantly less than control macrophages. Mechanistically, this lack of proliferation after TRAP inhibition was associated with higher presence of phosphorylated Beta-catenin (β-catenin; a signaling protein) compared with nontreated controls. Phosphorylation of β-catenin is known to mark it for ubiquitination and degradation by the proteasome, preventing its activity in promoting cell proliferation. In conclusion, our findings provide strong evidence for TRAP stimulating alveolar macrophage proliferation by dephosphorylating β-catenin. By driving proliferation, TRAP likely helps sustain alveolar macrophage populations during smoke exposure, either compensating for their loss due to smoking or increasing their numbers to better manage smoke-induced damage.NEW & NOTEWORTHY This study has uncovered that the enzyme tartrate-resistant acid phosphatase (TRAP) is crucial for alveolar macrophage proliferation through a β-catenin-dependent pathway. Importantly, TRAP influences this important ability of alveolar macrophages through the Acp5-202 mRNA transcript. The increase in TRAP expression following smoke exposure suggests that it plays a key role in promoting cell renewal, potentially helping to mitigate smoke-induced lung damage.
Collapse
Affiliation(s)
- Suzanne H Willems
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Shilei Qian
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Pernilla Lång
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Bjarne E Overtoom
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Sina Alimostafazadeh
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Rocío Fuentes-Mateos
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Gwenda F Vasse
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
| | - T Anienke van der Veen
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
- GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jelmer Vlasma
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marina H de Jager
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Victor Guryev
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gyorgy Fejer
- School of Biomedical and Healthcare Sciences, Peninsula Schools of Medicine and Dentistry, University of Plymouth, Plymouth, United Kingdom
| | - Göran Andersson
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Barbro N Melgert
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
6
|
Ishigaki S, Suzuki K, Takeshita M, Kaneko Y. Identification of BHLHE40-Expressing CD4 + T Cells Producing GM-CSF in Rheumatoid Arthritis. Int J Rheum Dis 2025; 28:e70219. [PMID: 40223427 DOI: 10.1111/1756-185x.70219] [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: 07/02/2024] [Revised: 03/09/2025] [Accepted: 03/21/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND Granulocyte-macrophage colony-stimulating factor (GM-CSF: CSF2) plays a crucial role in the pathogenesis of autoimmune diseases. The basic helix-loop-helix family member e40 (BHLHE40) gene is important for GM-CSF production in CD4+ T cells. However, the relationship between the expression of these genes and rheumatoid arthritis (RA) remains unclear, particularly in interleukin-1 (IL-1)-enriched inflammatory sites. Therefore, we investigated the expression of BHLHE40 and CSF2 in CD4+ T cells in RA. METHODS We analyzed gene expression using previously deposited and publicly available databases containing peripheral blood (PB) and synovial fluid (SF) from patients with RA and healthy controls (HC). Comprehensive datasets, including single-cell RNA-sequencing (scRNA-seq), RNA-seq, and microarray data, were used for this analysis. RESULTS BHLHE40 expression in PB CD4+ T cells from HC was higher in central memory, effector memory, Th17, and Th1/17 cells than in naive CD4+ T cells. Furthermore, BHLHE40 and CSF2 expression in the CD45RA-CCR7+ /-CD4+ T cell subset was significantly higher in the SF of patients with RA than in those with PB. scRNA-seq revealed that BHLHE40-expressing cells showed higher CSF2 expression than those that did not. Additionally, scRNA-seq showed higher BHLHE40 expression in PB CD4+ T cells from RA patients than in those from HC. CONCLUSION We analyzed the gene expressions of BHLHE40, which is crucial for GM-CSF production, IL1R1, which regulates BHLHE40 induction, and CSF2, its resulting product, in CD4+ T cells. Their expression levels were compared across RA SF, PB, and HC. Notably, increased expression of these genes was identified in SF.
Collapse
Affiliation(s)
- Sho Ishigaki
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Katsuya Suzuki
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Masaru Takeshita
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Yuko Kaneko
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
7
|
Wu Y, Zhang C, Huang J, Chen Q, Zhang Y, Liu F, Xu D, Jiang K, Shi R, Chen M, Yuan H. Integrated analysis of scRNA-seq and bulk RNA-seq data identifies BHLHE40 as a key gene in pancreatic cancer progression and gemcitabine resistance. Semin Oncol 2025; 52:152338. [PMID: 40250076 DOI: 10.1016/j.seminoncol.2025.152338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 04/20/2025]
Abstract
OBJECTIVE Pancreatic cancer is characterized by its high mortality rate and short survival periods, and novel therapeutic targets and tailor personalized strategies are urgently needed. In this study, we aim to investigate the molecular mechanisms underlying pancreatic ductal adenocarcinoma (PDAC) progression and chemoresistance, with a focus on identifying novel therapeutic targets. METHODS Multiomics approaches were integrated to identify novel actionable targets for PDAC. Public datasets such as TCGA and GEO were utilized to investigate the relationship between gene expression and clinical outcomes. Functional enrichment, cell-cell communication, and metabolic pathway analyses were performed to reveal PDAC heterogeneity and therapeutic resistance mechanisms. RESULTS BHLHE40 was identified as a hub gene linked to high-CNV PDAC cells, Gemcitabine resistance, and poor prognosis in PDAC. High BHLHE40 expression is significantly correlated with immunosuppressive tumor microenvironment (TME) features such as reduced CD8+ T infiltration, TCR richness, and lower tumor mutational burden (TMB). ChIP-seq data analysis confirmed BHLHE40 could directly bind to the SAT1 promoter, establishing a transcriptional axis promoting chemoresistance. Single-cell RNA-seq analysis further revealed that the BHLHE40+/SAT1+ subpopulation cells are resistant to Gemcitabine in PDAC. CONCLUSIONS BHLHE40 is significantly correlated with PDAC malignancy and chemoresistance via SAT1 regulation and immune evasion. Targeting BHLHE40 may sensitize PDACs to Gemcitabine and facilitate personalized treatment for BHLHE40+ PDAC patients.
Collapse
Affiliation(s)
- Yang Wu
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chun Zhang
- Department of Gastroenterology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiacheng Huang
- Department of Urology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qun Chen
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yufeng Zhang
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fengyuan Liu
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Dong Xu
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Kuirong Jiang
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Run Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mengxing Chen
- Department of Nephrology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hao Yuan
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
8
|
Mestrallet G, Brown M, Vaninov N, Cho NW, Velazquez L, Ananthanarayanan A, Spitzer M, Vabret N, Cimen Bozkus C, Samstein RM, Bhardwaj N. Coordinated macrophage and T cell interactions mediate response to checkpoint blockade in colorectal cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.12.637954. [PMID: 40027748 PMCID: PMC11870396 DOI: 10.1101/2025.02.12.637954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Mismatch repair deficiency (MMRd), either due to inherited or somatic mutation, is prevalent in colorectal cancer (CRC) and other cancers. While anti-PD-1 therapy is utilized in both local and advanced disease, up to 50% of MMRd CRC fail to respond. Using animal and human models of MMRd, we determined that interactions between MHC+ C1Q+ CXCL9+ macrophages and TCF+ BHLHE40+ PRF1+ T cell subsets are associated with control of MMRd tumor growth, during anti-PD-1 treatment. In contrast, resistance is associated with upregulation of TIM3, LAG3, TIGIT, and PD-1 expression on T cells, and infiltration of the tumor with immunosuppressive TREM2+ macrophages and monocytes. By combining anti-PD-1 with anti-LAG3/CTLA4/TREM2, up to 100% tumor eradication was achieved in MMRd CRC and remarkably, in >70% in MMRp CRC. This study identifies key T cell and macrophage subsets mediating the efficacy of immunotherapy in overcoming immune escape in both MMRd and MMRp CRC settings. Abstract Figure Highlights Anti-PD-1 therapy leads to the accumulation and colocalization of MHCI/II+ C1Q+ CXCL9+ macrophages and DCs with TCF+ CCL5+ T cells that have high TCR diversity.Resistance to anti-PD-1 therapy involves multiple T cell checkpoints, TREM2+ macrophages, IL1B+ TREM1+ monocytes and neutrophils, and IFITM+ tumor cells.Simultaneous blockade of PD-1, LAG3, CTLA-4 and TREM2 dramatically prevents progression of both MMRd and MMRp tumors.Combination therapy completely eliminates tumors by leveraging MHC+ macrophage, CD4+ and CD8+ T cell interactions, facilitating durable anti-tumor effects.
Collapse
|
9
|
Chen J, Zou P, Quan L, Gong C, Fang Z, Lin B, Lang J, Chen M. Huaxian formula prevents the progression of radiation-induced pulmonary fibrosis by inhibiting the pro-fibrotic effects of macrophages. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119026. [PMID: 39515679 DOI: 10.1016/j.jep.2024.119026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Huaxian formula (HXF), a traditional Chinese medicine (TCM) remedy, specifically targets the pathological factors of "heat toxicity" and "phlegm stasis" induced by radiation in radiation-induced pulmonary fibrosis (RIPF). It works by clearing heat and invigorating the blood, addressing these key factors in the development of RIPF. AIM OF THE STUDY The HXF has demonstrated potential in preventing RIPF, although its underlying mechanisms remain unclear. This study aims to investigate the efficacy, molecular targets, and mechanisms of action of HXF. MATERIALS AND METHODS The major constituents of the HXF were identified by ultra performance liquid chromatography and tandem mass spectrometry (UPLC-MS). C57BL/6j mice were divided into four groups: control (Ctrl), HXF alone (HXF), 17Gy-irradiation alone (IR), and irradiation plus HXF (IR + HXF). Lung damage and fibrosis were assessed by histopathological staining, and the flow cytometry and immunohistochemistry (IHC) were used to detect the macrophages phenotype of lung tissues in vivo at 16 weeks post-irradiation. Transcriptomic sequencing and bioinformatics analyses identified key genes modulated by HXF. In vitro assays included flow cytometry, western bolt, and quantitative PCR (qPCR) explored the impact of HXF on macrophage polarization and fibrotic activity, while co-culture experiments of the macrophage conditional medium and mouse embryo fibroblast NIH/3T3 investigated macrophage-fibroblast interactions. RESULTS 20 major constituents of HXF were identified. And the in vivo results revealed significant lung damage and fibrosis in the IR group, which were notably mitigated in the IR + HXF group. And HXF has been shown to significantly inhibit the infiltration of M2-type macrophages in lung tissues. Transcriptomic analysis identified differentially expressed genes (DEGs) such as Arg1, Mmp10, and Fgf23. Bioinformatics enrichment analysis indicated that these DEGs are involved in pathways related to the inhibition of extracellular matrix formation and inflammation. In vitro, HXF-containing serum reduced M2-type macrophage polarization and decreased the secretion of Arginase1 and TGFβ1. Conditioned medium from HXF-treated macrophages suppressed fibroblast activation. CONCLUSION HXF's preventive effects on RIPF involve multiple targets and mechanisms, including the modulation of Arg1, Mmp10, and Fgf23 expression. By inhibiting the pro-fibrotic capacity of macrophages, HXF suppresses fibroblast activation and collagen production, thereby alleviating lung fibrosis. These findings underscore the potential of HXF as a preventive strategy in managing RIPF.
Collapse
Affiliation(s)
- Junyang Chen
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China
| | - Pingjin Zou
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China
| | - Li Quan
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610032, China
| | - Cuicui Gong
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610032, China
| | - Zengyi Fang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Bing Lin
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610032, China.
| | - Jinyi Lang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China; Institute of Integrated Traditional Chinese and Western Medicine Cancer Research, Chengdu University of Traditional Chinese Medicine, Chengdu, 610032, China.
| | - Meihua Chen
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China.
| |
Collapse
|
10
|
Ye Y, Huang Y, Pan J. Exploration of the diagnostic and prognostic roles of decreased autoantibodies in lung cancer. Front Immunol 2025; 16:1538071. [PMID: 39949782 PMCID: PMC11821978 DOI: 10.3389/fimmu.2025.1538071] [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: 12/02/2024] [Accepted: 01/13/2025] [Indexed: 02/16/2025] Open
Abstract
Introduction Tumor-associated antigens (TAA) are proteins expressed during the growth and development of tumor cells, and TAA autoantibodies (TAAbs) can be detected in the serum of lung cancer patients, which can be utilized in the early screening of lung cancer. Almost all the TAAbs applied for diagnosis are those elevated, however, there are still large numbers of autoantibodies detected to decrease in tumor serums, and their functions were rarely known. Diagnosing malignant small lung nodules (≤3cm) in CT scans remains a challenge in clinical practice. Methods In this study, we applied the HuProt array and the bioinformatics analysis to assess the diagnostic values of the decreased autoantibodies in lung cancers. Results In total, 15 types of decreased autoantibodies were identified, and 6 of them were constructed into a predictive model for early lung cancer, reaching a sensitivity of 76.19% and a specificity of 55.74%. We combined with 4 elevated TAAbs, the sensitivity and the specificity of the 10-marker model can attain 80.0% and 87.0%, respectively, which is higher than that of the commonly used 7-TAAbs model in diagnosis for early-stage lung cancer. Moreover, 5 of the decreased autoantibodies can also be applied for supervising bone metastasis in lung adenocarcinoma. A follow-up process for 13 patients diagnosed with early-stage lung cancer revealed that 10 of the 15 decreased autoantibodies would recover to a higher level after the tumor was resected. Bioinformatic analysis indicated that the 15 biomarkers were strongly correlated with the prognosis of lung cancer patients. Conclusion We confirmed the importance of the decreased autoantibodies in lung cancer, providing new diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Ying Ye
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yi Huang
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Jianbo Pan
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Precision Medicine Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
11
|
Buzzao D, Steininger L, Guala D, Sonnhammer ELL. The FunCoup Cytoscape App: multi-species network analysis and visualization. Bioinformatics 2024; 41:btae739. [PMID: 39700425 PMCID: PMC11684335 DOI: 10.1093/bioinformatics/btae739] [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: 10/10/2024] [Revised: 12/09/2024] [Accepted: 12/17/2024] [Indexed: 12/21/2024] Open
Abstract
MOTIVATION Functional association networks, such as FunCoup, are crucial for analyzing complex gene interactions. To facilitate the analysis and visualization of such genome-wide networks, there is a need for seamless integration with powerful network analysis tools like Cytoscape. RESULTS The FunCoup Cytoscape App integrates the FunCoup web service API with Cytoscape, allowing users to visualize and analyze gene interaction networks for 640 species. Users can input gene identifiers and customize search parameters, using various network expansion algorithms like group or independent gene search, MaxLink, and TOPAS. The app maintains consistent visualizations with the FunCoup website, providing detailed node and link information, including tissue and pathway gene annotations. The integration with Cytoscape plugins, such as ClusterMaker2, enhances the analytical capabilities of FunCoup, as exemplified by the identification of the Myasthenia gravis disease module along with potential new therapeutic targets. AVAILABILITY AND IMPLEMENTATION The FunCoup Cytoscape App is developed using the Java OSGi framework, with UI components implemented in Java Swing and build support from Maven. The App is available as a JAR file at https://bitbucket.org/sonnhammergroup/funcoup_cytoscape/ repository, and can be downloaded from the Cytoscape App store https://apps.cytoscape.org/apps/funcoup.
Collapse
Affiliation(s)
- Davide Buzzao
- Department of Biochemistry and Biophysics, Stockholm University, Science for Life Laboratory, 171 21 Solna, Sweden
| | - Lukas Steininger
- Department of Biochemistry and Biophysics, Stockholm University, Science for Life Laboratory, 171 21 Solna, Sweden
| | - Dimitri Guala
- Department of Biochemistry and Biophysics, Stockholm University, Science for Life Laboratory, 171 21 Solna, Sweden
| | - Erik L L Sonnhammer
- Department of Biochemistry and Biophysics, Stockholm University, Science for Life Laboratory, 171 21 Solna, Sweden
| |
Collapse
|
12
|
Li YT, Takaki E, Ouchi Y, Tamai K. Guided monocyte fate to FRβ/CD163 + S1 macrophage antagonises atopic dermatitis via fibroblastic matrices in mouse hypodermis. Cell Mol Life Sci 2024; 82:14. [PMID: 39720957 DOI: 10.1007/s00018-024-05543-2] [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: 05/08/2024] [Revised: 10/21/2024] [Accepted: 12/06/2024] [Indexed: 12/26/2024]
Abstract
Macrophages are versatile myeloid leukocytes with flexible cellular states to perform diverse tissue functions beyond immunity. This plasticity is however often hijacked by diseases to promote pathology. Scanning kinetics of macrophage states by single-cell transcriptomics and flow cytometry, we observed atopic dermatitis drastically exhausted a resident subtype S1. Characterized by FRβ/CD163 expression, S1 exhibited strong efferocytosis and chemoattracted monocytes and eosinophils. Here we have delineated mechanisms regulating monocyte decision to acquire S1 identity in skin. During M-CSF driven macrophage differentiation in healthy skin, FRβ was expressed via intrinsic control of STAT6 and ALK5 activities, and did not require heterotypic cellular crosstalk. In contrast, CD163 expression required exposure to fibroblastic secretion. This process depended on SHP1 activity and involved STAT5 inactivation. Suppressed STAT5 activity caused CD163 expression and rendered macrophage insensitive to further induction by fibroblasts. Parsing coculture experiments with in silico ligand expression, we identified laminin-α2 and type-V collagen secreted by hypodermal fibroblasts as CD163-driving factors. S1 identity loss in AD followed a stepwise cascade: reduced laminins availability first dampened CD163 expression, IL4 and TGFβ subsequently acted on CD163lo/- cells to downregulate FRβ. In AD skin, we showed that imitating this fibroblast-macrophage crosstalk with exogenous laminin-211 encouraged monocyte differentiation to S1 macrophages, fostered homeostatic commitment of extravasated eosinophils, and alleviated dermatitis. Hence, we demonstrated that reinforcing a steady-state cue from hypodermal fibroblasts could override maladaptive pressure on macrophage and restored tissue homeostasis.
Collapse
MESH Headings
- Dermatitis, Atopic/metabolism
- Dermatitis, Atopic/pathology
- Receptors, Cell Surface/metabolism
- Antigens, Differentiation, Myelomonocytic/metabolism
- Animals
- Antigens, CD/metabolism
- Antigens, CD/genetics
- Fibroblasts/metabolism
- Fibroblasts/pathology
- Macrophages/metabolism
- Monocytes/metabolism
- Mice
- Cell Differentiation
- STAT6 Transcription Factor/metabolism
- Skin/metabolism
- Skin/pathology
- Receptor, Transforming Growth Factor-beta Type I/metabolism
- Receptor, Transforming Growth Factor-beta Type I/genetics
- Mice, Inbred C57BL
- Macrophage Colony-Stimulating Factor/metabolism
- Eosinophils/metabolism
- Laminin/metabolism
Collapse
Affiliation(s)
- Yu-Tung Li
- Department of Stem Cell Therapy Science, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan.
| | | | - Yuya Ouchi
- StemRIM Inc., Ibaraki, Osaka, 567-0085, Japan
| | - Katsuto Tamai
- Department of Stem Cell Therapy Science, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
13
|
Akula S, Alvarado-Vazquez A, Haide Mendez Enriquez E, Bal G, Franke K, Wernersson S, Hallgren J, Pejler G, Babina M, Hellman L. Characterization of Freshly Isolated Human Peripheral Blood B Cells, Monocytes, CD4+ and CD8+ T Cells, and Skin Mast Cells by Quantitative Transcriptomics. Int J Mol Sci 2024; 25:13050. [PMID: 39684762 DOI: 10.3390/ijms252313050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 11/25/2024] [Accepted: 12/01/2024] [Indexed: 12/18/2024] Open
Abstract
Quantitative transcriptomics offers a new way to obtain a detailed picture of freshly isolated cells. By direct isolation, the cells are unaffected by in vitro culture, and the isolation at cold temperatures maintains the cells relatively unaltered in phenotype by avoiding activation through receptor cross-linking or plastic adherence. Simultaneous analysis of several cell types provides the opportunity to obtain detailed pictures of transcriptomic differences between them. Here, we present such an analysis focusing on four human blood cell populations and compare those to isolated human skin mast cells. Pure CD19+ peripheral blood B cells, CD14+ monocytes, and CD4+ and CD8+ T cells were obtained by fluorescence-activated cell sorting, and KIT+ human connective tissue mast cells (MCs) were purified by MACS sorting from healthy skin. Detailed information concerning expression levels of the different granule proteases, protease inhibitors, Fc receptors, other receptors, transcription factors, cell signaling components, cytoskeletal proteins, and many other protein families relevant to the functions of these cells were obtained and comprehensively discussed. The MC granule proteases were found exclusively in the MC samples, and the T-cell granzymes in the T cells, of which several were present in both CD4+ and CD8+ T cells. High levels of CD4 were also observed in MCs and monocytes. We found a large variation between the different cell populations in the expression of Fc receptors, as well as for lipid mediators, proteoglycan synthesis enzymes, cytokines, cytokine receptors, and transcription factors. This detailed quantitative comparative analysis of more than 780 proteins of importance for the function of these populations can now serve as a good reference material for research into how these entities shape the role of these cells in immunity and tissue homeostasis.
Collapse
Affiliation(s)
- Srinivas Akula
- Department of Cell and Molecular Biology, Uppsala University, The Biomedical Center, Box 596, SE-751 24 Uppsala, Sweden
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, Box 7023, SE-75007 Uppsala, Sweden
| | - Abigail Alvarado-Vazquez
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Box 582, SE-75123 Uppsala, Sweden
| | - Erika Haide Mendez Enriquez
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Box 582, SE-75123 Uppsala, Sweden
| | - Gürkan Bal
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Kristin Franke
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Sara Wernersson
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, Box 7023, SE-75007 Uppsala, Sweden
| | - Jenny Hallgren
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Box 582, SE-75123 Uppsala, Sweden
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Box 582, SE-75123 Uppsala, Sweden
| | - Magda Babina
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Lars Hellman
- Department of Cell and Molecular Biology, Uppsala University, The Biomedical Center, Box 596, SE-751 24 Uppsala, Sweden
| |
Collapse
|
14
|
Keshari S, Shavkunov AS, Miao Q, Saha A, Minowa T, Molgora M, Williams CD, Chaib M, Highsmith AM, Pineda JE, Alekseev S, Alspach E, Hu KH, Colonna M, Pauken KE, Chen K, Gubin MM. Comparing neoantigen cancer vaccines and immune checkpoint therapy unveils an effective vaccine and anti-TREM2 macrophage-targeting dual therapy. Cell Rep 2024; 43:114875. [PMID: 39446585 PMCID: PMC11785356 DOI: 10.1016/j.celrep.2024.114875] [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: 03/05/2024] [Revised: 08/12/2024] [Accepted: 09/30/2024] [Indexed: 10/26/2024] Open
Abstract
The goal of therapeutic cancer vaccines and immune checkpoint therapy (ICT) is to promote T cells with anti-tumor capabilities. Here, we compared mutant neoantigen (neoAg) peptide-based vaccines with ICT in preclinical models. NeoAg vaccines induce the most robust expansion of proliferating and stem-like PD-1+TCF-1+ neoAg-specific CD8 T cells in tumors. Anti-CTLA-4 and/or anti-PD-1 ICT promotes intratumoral TCF-1- neoAg-specific CD8 T cells, although their phenotype depends in part on the specific ICT used. Anti-CTLA-4 also prompts substantial changes to CD4 T cells, including induction of ICOS+Bhlhe40+ T helper 1 (Th1)-like cells. Although neoAg vaccines or ICTs expand iNOS+ macrophages, neoAg vaccines maintain CX3CR1+CD206+ macrophages expressing the TREM2 receptor, unlike ICT, which suppresses them. TREM2 blockade enhances neoAg vaccine efficacy and is associated with fewer CX3CR1+CD206+ macrophages and induction of neoAg-specific CD8 T cells. Our findings highlight different mechanisms underlying neoAg vaccines and different forms of ICT and identify combinatorial therapies to enhance neoAg vaccine efficacy.
Collapse
Affiliation(s)
- Sunita Keshari
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alexander S Shavkunov
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qi Miao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Akata Saha
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tomoyuki Minowa
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Martina Molgora
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, St. Louis, MO, USA
| | - Charmelle D Williams
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mehdi Chaib
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anna M Highsmith
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Josué E Pineda
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sayan Alekseev
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Program of Biology, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Elise Alspach
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Kenneth H Hu
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The Parker Institute for Cancer Immunotherapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The James P. Allison Institute, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, St. Louis, MO, USA
| | - Kristen E Pauken
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ken Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Matthew M Gubin
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
15
|
Liu B, Sun Y, Wang W, Ren J, Wang D. BHLHE40-mediated transcriptional activation of GRIN2D in gastric cancer is involved in metabolic reprogramming. Funct Integr Genomics 2024; 24:214. [PMID: 39546079 DOI: 10.1007/s10142-024-01495-9] [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: 08/08/2024] [Revised: 10/25/2024] [Accepted: 11/01/2024] [Indexed: 11/17/2024]
Abstract
Gastric cancer (GC) is the third leading cause of death in developed countries. The reprogramming of energy metabolism represents a hallmark of cancer, particularly amplified dependence on aerobic glycolysis. Here, we aimed to illustrate the functional role of glutamate ionotropic receptor N-methyl-D-aspartate type subunit 2D (GRIN2D) in the regulation of glycolysis in GC and the mechanisms involved. Differentially expressed genes were analyzed using the GEO and GEPIA databases, followed by prognostic value prediction using the Kaplan-Meier Plotter database. The effect of GRIN2D knockdown on the malignant behavior and glycolysis of GC cells was explored. GRIN2D expression was upregulated in GC cells and promoted the malignant behavior of GC cells by activating glycolysis. Class E basic helix-loop-helix protein 40 (BHLHE40) was overexpressed in GC cells and mediated transcriptional activation of GRIN2D. The anti-tumor effects of BHLHE40 knockdown on GC cells in vitro and in vivo were reversed by GRIN2D overexpression. Knockdown of GRIN2D or BHLHE40 downregulated the expression of mRNA of electron transport chain subunits and phosphorylation of p38 MARK and inhibited calcium efflux in GC cells. Overexpression of GRIN2D promoted calcium efflux, phosphorylation of p38 MARK protein, and proliferation of GES1 cells. Altogether, the findings derived from this study suggest that BHLHE40 knockdown suppresses the growth, mobility, and glycolysis of GC cells by inhibiting GRIN2D transcription and disrupting the BHLHE40/GRIN2D axis may be an attractive therapeutic strategy for GC.
Collapse
Affiliation(s)
- Bin Liu
- Department of Gastrointestinal Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, No. 98, Nantong West Road, Guangling District, Yangzhou, Jiangsu, 225001, P.R. China
| | - Yuanlin Sun
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong, 270000, P.R. China
| | - Wei Wang
- Department of Gastrointestinal Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, No. 98, Nantong West Road, Guangling District, Yangzhou, Jiangsu, 225001, P.R. China
| | - Jun Ren
- Department of Gastrointestinal Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, No. 98, Nantong West Road, Guangling District, Yangzhou, Jiangsu, 225001, P.R. China
| | - Daorong Wang
- Department of Gastrointestinal Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, No. 98, Nantong West Road, Guangling District, Yangzhou, Jiangsu, 225001, P.R. China.
| |
Collapse
|
16
|
Pasanen A, Sliz E, Huilaja L, Reimann E, Mägi R, Laisk T, Tasanen K, Kettunen J. Identifying Atopic Dermatitis Risk Loci in 1,094,060 Individuals with Subanalysis of Disease Severity and Onset. J Invest Dermatol 2024; 144:2417-2425. [PMID: 38663478 DOI: 10.1016/j.jid.2024.02.036] [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: 08/28/2023] [Revised: 02/01/2024] [Accepted: 02/15/2024] [Indexed: 06/07/2024]
Abstract
Atopic dermatitis (AD) is a common inflammatory skin disease highly attributable to genetic factors. In this study, we report results from a genome-wide meta-analysis of AD in 37,541 cases and 1,056,519 controls with data from the FinnGen project, the Estonian Biobank, the UK Biobank, the EAGLE Consortium, and the BioBank Japan. We detected 77 independent AD-associated loci, of which 10 were, to our knowledge, previously unreported. The associated loci showed enrichment in various immune regulatory processes. We further performed subgroup analyses of mild and severe AD and of early- and late-onset AD, with data from the FinnGen project. Fifty-five of the 79 tested variants in the associated loci showed larger effect estimates for severe than for mild AD as determined through administered treatment. The age of onset, as determined by the first hospital visit with AD diagnosis, was lower in patients with particular AD-risk alleles. Our findings add to the knowledge of the genetic background of AD and may underlie the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Anu Pasanen
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland; Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland; Department of Dermatology, Oulu University Hospital, Oulu, Finland
| | - Eeva Sliz
- Center for Life-Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland; Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Laura Huilaja
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland; Department of Dermatology, Oulu University Hospital, Oulu, Finland
| | - Ene Reimann
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Reedik Mägi
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Triin Laisk
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Kaisa Tasanen
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland; Department of Dermatology, Oulu University Hospital, Oulu, Finland.
| | - Johannes Kettunen
- Center for Life-Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland; Biocenter Oulu, University of Oulu, Oulu, Finland
| |
Collapse
|
17
|
Park MD, Berichel JL, Hamon P, Wilk CM, Belabed M, Yatim N, Saffon A, Boumelha J, Falcomatà C, Tepper A, Hegde S, Mattiuz R, Soong BY, LaMarche NM, Rentzeperis F, Troncoso L, Halasz L, Hennequin C, Chin T, Chen EP, Reid AM, Su M, Cahn AR, Koekkoek LL, Venturini N, Wood-isenberg S, D’souza D, Chen R, Dawson T, Nie K, Chen Z, Kim-Schulze S, Casanova-Acebes M, Swirski FK, Downward J, Vabret N, Brown BD, Marron TU, Merad M. Hematopoietic aging promotes cancer by fueling IL-1⍺-driven emergency myelopoiesis. Science 2024; 386:eadn0327. [PMID: 39236155 PMCID: PMC7616710 DOI: 10.1126/science.adn0327] [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: 11/19/2023] [Revised: 06/18/2024] [Accepted: 08/22/2024] [Indexed: 09/07/2024]
Abstract
Age is a major risk factor for cancer, but how aging impacts tumor control remains unclear. In this study, we establish that aging of the immune system, regardless of the age of the stroma and tumor, drives lung cancer progression. Hematopoietic aging enhances emergency myelopoiesis, resulting in the local accumulation of myeloid progenitor-like cells in lung tumors. These cells are a major source of interleukin (IL)-1⍺, which drives the enhanced myeloid response. The age-associated decline of DNA methyltransferase 3A enhances IL-1⍺ production, and disrupting IL-1 receptor 1 signaling early during tumor development normalized myelopoiesis and slowed the growth of lung, colonic, and pancreatic tumors. In human tumors, we identified an enrichment for IL-1⍺-expressing monocyte-derived macrophages linked to age, poorer survival, and recurrence, unraveling how aging promotes cancer and offering actionable therapeutic strategies.
Collapse
Affiliation(s)
- Matthew D. Park
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Jessica Le Berichel
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Pauline Hamon
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - C. Matthias Wilk
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Meriem Belabed
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Nader Yatim
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Alexis Saffon
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- INSERM U932, Immunity and Cancer, Institut Curie, Paris-Cité University; Paris, France
| | - Jesse Boumelha
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Chiara Falcomatà
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Alexander Tepper
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Samarth Hegde
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Raphaël Mattiuz
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Brian Y. Soong
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Nelson M. LaMarche
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Frederika Rentzeperis
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Leanna Troncoso
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Laszlo Halasz
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Clotilde Hennequin
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Theodore Chin
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Earnest P. Chen
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Amanda M. Reid
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Matthew Su
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Ashley Reid Cahn
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Laura L. Koekkoek
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Brain and Body Research Center, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Nicholas Venturini
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Shira Wood-isenberg
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Darwin D’souza
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Rachel Chen
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Travis Dawson
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Kai Nie
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Zhihong Chen
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Seunghee Kim-Schulze
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Maria Casanova-Acebes
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Filip K. Swirski
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Brain and Body Research Center, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Julian Downward
- Oncogene Biology Laboratory, Francis Crick Institute; London, UK
- Lung Cancer Group, Division of Molecular Pathology, Institute of Cancer Research; London, UK
| | - Nicolas Vabret
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Brian D. Brown
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Thomas U. Marron
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Center for Thoracic Oncology, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Miriam Merad
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| |
Collapse
|
18
|
Watkins TA. Differential enrichment of retinal ganglion cells underlies proposed core neurodegenerative transcription programs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.21.618927. [PMID: 39484484 PMCID: PMC11527038 DOI: 10.1101/2024.10.21.618927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
In a published Correction 1 , a revised analysis updated two "core transcription programs" proposed to underlie axon injury-induced retinal ganglion cell (RGC) neurodegeneration. Though extensive, the Correction purported to leave the two principal conclusions of its parent study 2 unaltered. The first of those findings was that a core program mediated by the Activating Transcription Factor-4 (ATF4) and its likely heterodimeric partner does not include numerous canonical ATF4 target genes stimulated by RGC axon injury. The second was that the Activating Transcription Factor-3 (ATF3) and C/EBP Homologous Protein (CHOP) function with unprecedented coordination in a parallel program regulating innate immunity pathways. Here those unexpected findings are revealed to instead reflect insufficient knockout coupled with differences in RGC enrichment across conditions. This analysis expands on the published Correction's redefinition of the purported transcription programs to raise foundational questions about the proposed functions and relationships of these transcription factors in neurodegeneration.
Collapse
|
19
|
Chen X, Wang M, Wang H, Yang J, Li X, Zhang R, Ding X, Hou H, Zhou J, Wu M. METTL3 inhibitor suppresses the progression of prostate cancer via IGFBP3/AKT pathway and synergizes with PARP inhibitor. Biomed Pharmacother 2024; 179:117366. [PMID: 39232384 DOI: 10.1016/j.biopha.2024.117366] [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: 06/10/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/06/2024] Open
Abstract
The RNA N6-methyladenosine (m6A) regulator METTL3 is an important regulatory gene in various progressive processes of prostate cancer (PCa). METTL3 inhibitors have been reported to possess potent tumor suppression capacity in some cancer types. Nevertheless, the detailed influence and mechanism of METTL3 inhibitors on PCa progression and their potential synergy with other drugs are poorly understood. In this study, we demonstrated that METTL3 was overexpressed and associated with poor survival in most PCa patients. METTL3 inhibitor STM2457 reduced m6A levels of PCa cells, thus inhibiting their proliferation, colony formation, migration, invasion, and stemness in vitro. Furthermore, STM2457 suppressed PCa progression in both the CDX and PDX models in vivo. MeRIP-seq analysis coupled with biological validation revealed that STM2457 influenced multiple biological processes in PCa cells, mainly through the IGFBP3/AKT pathway. We also proved that STM2457 induced DNA damage and showed synergistic anti-PCa effects with the PARP inhibitor olaparib both in vitro and in vivo. All in all, this work provides a novel therapeutic strategy for targeting RNA m6A modifications for the treatment of PCa and provides a meaningful reference for further clinical trials.
Collapse
Affiliation(s)
- Xin Chen
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, PR China
| | - Miaomiao Wang
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, PR China
| | - Haoran Wang
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, PR China
| | - Jingxin Yang
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, PR China
| | - Xiaoxin Li
- Center for Drug Research and Evaluation, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, PR China
| | - Rongyu Zhang
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Department of Chemistry, Zhejiang Normal University, 688 Yingbin Road, Jinhua 321004, PR China
| | - Xin Ding
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, PR China
| | - Huimin Hou
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, PR China.
| | - Jinming Zhou
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Department of Chemistry, Zhejiang Normal University, 688 Yingbin Road, Jinhua 321004, PR China.
| | - Meng Wu
- Center for Drug Research and Evaluation, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, PR China.
| |
Collapse
|
20
|
Pahl MC, Sharma P, Thomas RM, Thompson Z, Mount Z, Pippin JA, Morawski PA, Sun P, Su C, Campbell D, Grant SFA, Wells AD. Dynamic chromatin architecture identifies new autoimmune-associated enhancers for IL2 and novel genes regulating CD4+ T cell activation. eLife 2024; 13:RP96852. [PMID: 39302339 PMCID: PMC11418197 DOI: 10.7554/elife.96852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024] Open
Abstract
Genome-wide association studies (GWAS) have identified hundreds of genetic signals associated with autoimmune disease. The majority of these signals are located in non-coding regions and likely impact cis-regulatory elements (cRE). Because cRE function is dynamic across cell types and states, profiling the epigenetic status of cRE across physiological processes is necessary to characterize the molecular mechanisms by which autoimmune variants contribute to disease risk. We localized risk variants from 15 autoimmune GWAS to cRE active during TCR-CD28 co-stimulation of naïve human CD4+ T cells. To characterize how dynamic changes in gene expression correlate with cRE activity, we measured transcript levels, chromatin accessibility, and promoter-cRE contacts across three phases of naive CD4+ T cell activation using RNA-seq, ATAC-seq, and HiC. We identified ~1200 protein-coding genes physically connected to accessible disease-associated variants at 423 GWAS signals, at least one-third of which are dynamically regulated by activation. From these maps, we functionally validated a novel stretch of evolutionarily conserved intergenic enhancers whose activity is required for activation-induced IL2 gene expression in human and mouse, and is influenced by autoimmune-associated genetic variation. The set of genes implicated by this approach are enriched for genes controlling CD4+ T cell function and genes involved in human inborn errors of immunity, and we pharmacologically validated eight implicated genes as novel regulators of T cell activation. These studies directly show how autoimmune variants and the genes they regulate influence processes involved in CD4+ T cell proliferation and activation.
Collapse
Affiliation(s)
- Matthew C Pahl
- Center for Spatial and Functional Genomics, Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Division of Human Genetics, The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Prabhat Sharma
- Center for Spatial and Functional Genomics, Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Department of Pathology, The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Rajan M Thomas
- Center for Spatial and Functional Genomics, Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Department of Pathology, The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Zachary Thompson
- Center for Spatial and Functional Genomics, Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Department of Pathology, The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Zachary Mount
- Center for Spatial and Functional Genomics, Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Department of Pathology, The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - James A Pippin
- Center for Spatial and Functional Genomics, Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Division of Human Genetics, The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Peter A Morawski
- Benaroya Research Institute at Virginia MasonSeattleUnited States
| | - Peng Sun
- Center for Spatial and Functional Genomics, Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Department of Pathology, The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Chun Su
- Center for Spatial and Functional Genomics, Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Division of Human Genetics, The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Daniel Campbell
- Benaroya Research Institute at Virginia MasonSeattleUnited States
- Department of Immunology, University of Washington School of MedicineSeattleUnited States
| | - Struan FA Grant
- Center for Spatial and Functional Genomics, Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Division of Human Genetics, The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Department of Genetics, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Department of Pediatrics, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Division of Endocrinology and Diabetes, The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Andrew D Wells
- Center for Spatial and Functional Genomics, Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Department of Pathology, The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Institute for Immunology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
21
|
Cheng Z, Liu B, Liu X. Circadian gene signatures in the progression of obesity based on machine learning and Mendelian randomization analysis. Front Nutr 2024; 11:1407265. [PMID: 39351493 PMCID: PMC11439728 DOI: 10.3389/fnut.2024.1407265] [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: 03/26/2024] [Accepted: 09/03/2024] [Indexed: 10/04/2024] Open
Abstract
Objective Obesity, a global health concern, is associated with a spectrum of chronic diseases and cancers. Our research sheds light on the regulatory role of circadian genes in obesity progression, providing insight into the immune landscape of obese patients, and introducing new avenues for therapeutic interventions. Methods Expression files of multiple datasets were retrieved from the GEO database. By 80 machine-learning algorithm combinations and Mendelian randomization analysis, we discovered the key circadian genes contributing to and protecting against obesity. Subsequently, an immune infiltration analysis was conducted to examine the alterations in immune cell types and their abundance in the body and to investigate the relationships between circadian genes and immune cells. Furthermore, we delved into the molecular mechanisms of key genes implicated in obesity. Results Our study identified three key circadian genes (BHLHE40, PPP1CB, and CSNK1E) associated with obesity. BHLHE40 was found to promote obesity through various pathways, while PPP1CB and CSNK1E counteracted lipid metabolism disorders, and modulated cytokines, immune receptors, T cells, and monocytes. Conclusion In conclusion, the key circadian genes (BHLHE40, CSNK1E, and PPP1CB) may serve as novel biomarkers for understanding obesity pathogenesis and have significant correlations with infiltrating immune cells, thus providing potential new targets for obese prevention and treatment.
Collapse
Affiliation(s)
- Zhi’ang Cheng
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Binghong Liu
- College of Horticulture, South China Agricultural University, Guangzhou, China
| | - Xiaoyong Liu
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
- Department of Ophthalmology, The Affiliated Shunde Hospital of Jinan University, Foshan, China
| |
Collapse
|
22
|
Ichiyama K, Long J, Kobayashi Y, Horita Y, Kinoshita T, Nakamura Y, Kominami C, Georgopoulos K, Sakaguchi S. Transcription factor Ikzf1 associates with Foxp3 to repress gene expression in Treg cells and limit autoimmunity and anti-tumor immunity. Immunity 2024; 57:2043-2060.e10. [PMID: 39111316 DOI: 10.1016/j.immuni.2024.07.010] [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: 08/10/2023] [Revised: 02/16/2024] [Accepted: 07/15/2024] [Indexed: 09/13/2024]
Abstract
The master transcription factor of regulatory T (Treg) cells, forkhead box protein P3 (Foxp3), controls Treg cell function by targeting certain genes for activation or repression, but the specific mechanisms by which it mediates this activation or repression under different conditions remain unclear. We found that Ikzf1 associates with Foxp3 via its exon 5 (IkE5) and that IkE5-deficient Treg cells highly expressed genes that would otherwise be repressed by Foxp3 upon T cell receptor stimulation, including Ifng. Treg-specific IkE5-deletion caused interferon-γ (IFN-γ) overproduction, which destabilized Foxp3 expression and impaired Treg suppressive function, leading to systemic autoimmune disease and strong anti-tumor immunity. Pomalidomide, which degrades IKZF1 and IKZF3, induced IFN-γ overproduction in human Treg cells. Mechanistically, the Foxp3-Ikzf1-Ikzf3 complex competed with epigenetic co-activators, such as p300, for binding to target gene loci via chromatin remodeling. Therefore, the Ikzf1 association with Foxp3 is essential for the gene-repressive function of Foxp3 and could be exploited to treat autoimmune disease and cancer.
Collapse
Affiliation(s)
- Kenji Ichiyama
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.
| | - Jia Long
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Yusuke Kobayashi
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan; Department of Medical Innovations, Osaka Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., Osaka, Japan
| | - Yuji Horita
- Joint Research Chair of Immune-therapeutic Drug Discovery, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan; Department of Research Management, Otsuka Pharmaceutical Co., Ltd., Tokushima, Japan
| | - Takeshi Kinoshita
- Joint Research Chair of Immune-therapeutic Drug Discovery, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan; Department of Research Management, Otsuka Pharmaceutical Co., Ltd., Tokushima, Japan
| | - Yamami Nakamura
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Chizuko Kominami
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Katia Georgopoulos
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Shimon Sakaguchi
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan; Department of Experimental Pathology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.
| |
Collapse
|
23
|
Singh Rajkumar M, Ibanez-Carrasco F, Avila CA, Mandadi KK. Insights into Bactericera cockerelli and Candidatus Liberibacter solanacearum interaction: a tissue-specific transcriptomic approach. FRONTIERS IN PLANT SCIENCE 2024; 15:1393994. [PMID: 39280947 PMCID: PMC11392735 DOI: 10.3389/fpls.2024.1393994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 08/14/2024] [Indexed: 09/18/2024]
Abstract
The tomato-potato psyllid, Bactericera cockerelli (Šulc), belonging to the Hemiptera order, is an insect pest of solanaceous crops and vectors a fastidious bacterium, Candidatus Liberibacter solanacearum (CLso), the presumptive causal agent of zebra chip and vein greening diseases in potatoes and tomatoes, respectively. The genome of B. cockerelli has been sequenced recently, providing new avenues to elucidate mechanistic insights into pathogenesis in vegetable crops. In this study, we performed RNA-sequencing of the critical psyllid organs (salivary glands and ovaries) involved in CLso pathology and transmission to host plants. Transcriptome analysis revealed differentially expressed genes and organ-specific enrichment of gene ontology (GO) terms related to metabolic processes, response to stress/stimulus, phagocytosis, proteolysis, endocytosis, and provided candidate genes encoding transcription factors (TFs). To examine gene regulatory networks across the psyllid organs under CLso(-) and CLso(+) conditions, we performed weighted gene co-expression network analysis (WGCNA), and unique modules differentiating the psyllid organs were identified. A comparative GO analysis of the unique gene modules revealed functional terms enriched in response to stress, gene regulation, and cell division processes in the ovaries. In contrast, respiration, transport, and neuronal transmission-related GO terms were enriched in the salivary glands. Altogether, this study reveals new insights into tissue-specific expression of the psyllid organs in the absence or presence of CLso bacterium. This knowledge can be leveraged to develop new pest and disease management strategies by delineating the regulatory networks involved in the psyllid-CLso interaction.
Collapse
Affiliation(s)
| | - Freddy Ibanez-Carrasco
- Texas A&M AgriLife Research and Extension Center, Weslaco, TX, United States
- Department of Entomology, Texas A&M University, College Station, TX, United States
| | - Carlos A Avila
- Texas A&M AgriLife Research and Extension Center, Weslaco, TX, United States
- Department of Horticultural Sciences, Texas A&M University, College Station, TX, United States
| | - Kranthi K Mandadi
- Texas A&M AgriLife Research and Extension Center, Weslaco, TX, United States
- Department of Plant Pathology & Microbiology, Texas A&M University, College Station, TX, United States
- Institute for Advancing Health Through Agriculture, Texas A&M AgriLife, College Station, TX, United States
| |
Collapse
|
24
|
Yang Y, Chen X, Pan J, Ning H, Zhang Y, Bo Y, Ren X, Li J, Qin S, Wang D, Chen MM, Zhang Z. Pan-cancer single-cell dissection reveals phenotypically distinct B cell subtypes. Cell 2024; 187:4790-4811.e22. [PMID: 39047727 DOI: 10.1016/j.cell.2024.06.038] [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: 06/09/2023] [Revised: 04/25/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024]
Abstract
Characterizing the compositional and phenotypic characteristics of tumor-infiltrating B cells (TIBs) is important for advancing our understanding of their role in cancer development. Here, we establish a comprehensive resource of human B cells by integrating single-cell RNA sequencing data of B cells from 649 patients across 19 major cancer types. We demonstrate substantial heterogeneity in their total abundance and subtype composition and observe immunoglobulin G (IgG)-skewness of antibody-secreting cell isotypes. Moreover, we identify stress-response memory B cells and tumor-associated atypical B cells (TAABs), two tumor-enriched subpopulations with prognostic potential, shared in a pan-cancer manner. In particular, TAABs, characterized by a high clonal expansion level and proliferative capacity as well as by close interactions with activated CD4 T cells in tumors, are predictive of immunotherapy response. Our integrative resource depicts distinct clinically relevant TIB subsets, laying a foundation for further exploration of functional commonality and diversity of B cells in cancer.
Collapse
Affiliation(s)
- Yu Yang
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Xueyan Chen
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Jieying Pan
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Huiheng Ning
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Yaojun Zhang
- State Key Laboratory of Oncology in South China, Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yufei Bo
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Xianwen Ren
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Jiesheng Li
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Shishang Qin
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Dongfang Wang
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China.
| | - Min-Min Chen
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | - Zemin Zhang
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China.
| |
Collapse
|
25
|
Ji A, Li H, Fu X, Zhang Y, Liu Y. Long non-coding RNA NEAT1 induced by BHLHE40 activates Wnt/β-catenin signaling and potentiates colorectal cancer progression. Cell Div 2024; 19:25. [PMID: 39098910 PMCID: PMC11299305 DOI: 10.1186/s13008-024-00129-7] [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: 01/31/2024] [Accepted: 07/22/2024] [Indexed: 08/06/2024] Open
Abstract
BACKGROUND Nuclear-enriched abundant transcript 1 (NEAT1), a long noncoding RNA (lncRNA), has been implicated in the colorectal cancer (CRC) progression. However, its upstream mechanism has not been well studied. In the present study, the functions and mechanisms of NEAT1 in CRC were investigated. METHODS The NEAT1 expression in CRC tissues and CRC cells was analyzed by RT-qPCR. The genes co-expressed with NEAT1 in CRC were obtained from UALCAN, which were intersected with the transcription factors targeting NEAT1 from hTFtarget. Dual-luciferase assay, RT-qPCR, and ChIP were conducted to analyze the transcriptional regulatory relationship between BHLHE40 and NEAT1. LoVo and HCT-15 cells knocking down BHLHE40 and overexpressing NEAT1 were subjected to MTT, Transwell, Western blot, and flow cytometry to examine the malignant aggressiveness of CRC cells. The effects of knocking down BHLHE40 and overexpressing NEAT1 on tumor and lung metastasis were investigated in mice using HE and immunohistochemical analyses. RESULTS NEAT1 and BHLHE40 were significantly overexpressed in CRC tissues and cells. BHLHE40 has a binding relationship with the NEAT1 promoter. Knockdown of BHLHE40 resulted in a reverted malignant phenotype in vitro and slowed tumor growth and metastasis dissemination in vivo, which were reversed by NEAT1 overexpression. Overexpression of BHLHE40 increased Wnt/β-catenin pathway activity, but knockdown of NEAT1 decreased Wnt/β-catenin pathway activity. CONCLUSIONS BHLHE40 mediates the transcriptional activation of NEAT1, which activates the Wnt/β-catenin pathway and promotes the CRC progression.
Collapse
Affiliation(s)
- Anlong Ji
- Department of General Surgery, The Second Affiliated Hospital of Hainan Medical University, No. 368, Yehai Avenue, Haikou, 570216, Hainan, People's Republic of China
| | - Hui Li
- Department of Geriatrics, The Second Affiliated Hospital of Hainan Medical University, No. 368, Yehai Avenue, Haikou, 570216, Hainan, People's Republic of China
| | - Xiangwei Fu
- Department of General Surgery, The Second Affiliated Hospital of Hainan Medical University, No. 368, Yehai Avenue, Haikou, 570216, Hainan, People's Republic of China
| | - Yourong Zhang
- Department of General Surgery, The Second Affiliated Hospital of Hainan Medical University, No. 368, Yehai Avenue, Haikou, 570216, Hainan, People's Republic of China
| | - Yanhe Liu
- Department of General Surgery, The Second Affiliated Hospital of Hainan Medical University, No. 368, Yehai Avenue, Haikou, 570216, Hainan, People's Republic of China.
| |
Collapse
|
26
|
Acosta-Iborra B, Gil-Acero AI, Sanz-Gómez M, Berrouayel Y, Puente-Santamaría L, Alieva M, del Peso L, Jiménez B. Bhlhe40 Regulates Proliferation and Angiogenesis in Mouse Embryoid Bodies under Hypoxia. Int J Mol Sci 2024; 25:7669. [PMID: 39062912 PMCID: PMC11277088 DOI: 10.3390/ijms25147669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Knowledge of the molecular mechanisms that underlie the regulation of major adaptive responses to an unbalanced oxygen tension is central to understanding tissue homeostasis and disease. Hypoxia-inducible transcription factors (HIFs) coordinate changes in the transcriptome that control these adaptive responses. Here, we focused on the functional role of the transcriptional repressor basic-helix-loop-helix family member e40 (Bhlhe40), which we previously identified in a meta-analysis as one of the most consistently upregulated genes in response to hypoxia across various cell types. We investigated the role of Bhlhe40 in controlling proliferation and angiogenesis using a gene editing strategy in mouse embryonic stem cells (mESCs) that we differentiated in embryoid bodies (EBs). We observed that hypoxia-induced Bhlhe40 expression was compatible with the rapid proliferation of pluripotent mESCs under low oxygen tension. However, in EBs, hypoxia triggered a Bhlhe40-dependent cell cycle arrest in most progenitor cells and endothelial cells within vascular structures. Furthermore, Bhlhe40 knockout increased the basal vascularization of the EBs in normoxia and exacerbated the hypoxia-induced vascularization, supporting a novel role for Bhlhe40 as a negative regulator of blood vessel formation. Our findings implicate Bhlhe40 in mediating key functional adaptive responses to hypoxia, such as proliferation arrest and angiogenesis.
Collapse
Affiliation(s)
- Bárbara Acosta-Iborra
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Arturo Duperier, 4, 28029 Madrid, Spain
| | - Ana Isabel Gil-Acero
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Arturo Duperier, 4, 28029 Madrid, Spain
| | - Marta Sanz-Gómez
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Arturo Duperier, 4, 28029 Madrid, Spain
| | - Yosra Berrouayel
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Arturo Duperier, 4, 28029 Madrid, Spain
| | - Laura Puente-Santamaría
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Arturo Duperier, 4, 28029 Madrid, Spain
- Biocomputing Unit, Instituto Aragonés de Ciencias de la Salud, San Juan Bosco, 50009 Zaragoza, Spain
| | - Maria Alieva
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Arturo Duperier, 4, 28029 Madrid, Spain
| | - Luis del Peso
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Arturo Duperier, 4, 28029 Madrid, Spain
- IdiPaz, Instituto de Investigación Sanitaria del Hospital Universitario La Paz, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Unidad Asociada de Biomedicina CSIC-UCLM, 02006 Albacete, Spain
| | - Benilde Jiménez
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Arturo Duperier, 4, 28029 Madrid, Spain
- IdiPaz, Instituto de Investigación Sanitaria del Hospital Universitario La Paz, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Unidad Asociada de Biomedicina CSIC-UCLM, 02006 Albacete, Spain
| |
Collapse
|
27
|
Nguyen C, Kudek M, Zander R, Niu H, Shen J, Bauer A, Alson D, Khatun A, Chen Y, Sun J, Drobyski W, Edelson BT, Cui W. Bhlhe40 Promotes CD4+ T Helper 1 Cell and Suppresses T Follicular Helper Cell Differentiation during Viral Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1829-1842. [PMID: 38619295 DOI: 10.4049/jimmunol.2300355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 03/18/2024] [Indexed: 04/16/2024]
Abstract
In response to acute infection, naive CD4+ T cells primarily differentiate into T helper 1 (Th1) or T follicular helper (Tfh) cells that play critical roles in orchestrating cellular or humoral arms of immunity, respectively. However, despite the well established role of T-bet and BCL-6 in driving Th1 and Tfh cell lineage commitment, respectively, whether additional transcriptional circuits also underlie the fate bifurcation of Th1 and Tfh cell subsets is not fully understood. In this article, we study how the transcriptional regulator Bhlhe40 dictates the Th1/Tfh differentiation axis in mice. CD4+ T cell-specific deletion of Bhlhe40 abrogates Th1 but augments Tfh differentiation. We also assessed an increase in germinal center B cells and Ab production, suggesting that deletion of Bhlhe40 in CD4+ T cells not only alters Tfh differentiation but also their capacity to provide help to B cells. To identify molecular mechanisms by which Bhlhe40 regulates Th1 versus Tfh lineage choice, we first performed epigenetic profiling in the virus specific Th1 and Tfh cells following LCMV infection, which revealed distinct promoter and enhancer activities between the two helper cell lineages. Furthermore, we identified that Bhlhe40 directly binds to cis-regulatory elements of Th1-related genes such as Tbx21 and Cxcr6 to activate their expression while simultaneously binding to regions of Tfh-related genes such as Bcl6 and Cxcr5 to repress their expression. Collectively, our data suggest that Bhlhe40 functions as a transcription activator to promote Th1 cell differentiation and a transcription repressor to suppress Tfh cell differentiation.
Collapse
Affiliation(s)
- Christine Nguyen
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
- Versiti Blood Research Institute, Versiti Wisconsin, Milwaukee, WI
| | - Matthew Kudek
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
- Versiti Blood Research Institute, Versiti Wisconsin, Milwaukee, WI
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
| | - Ryan Zander
- Versiti Blood Research Institute, Versiti Wisconsin, Milwaukee, WI
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA
| | - Hongshen Niu
- Department of Pathology, Northwestern University, Chicago, IL
| | - Jian Shen
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
- Versiti Blood Research Institute, Versiti Wisconsin, Milwaukee, WI
- Department of Pathology, Northwestern University, Chicago, IL
| | - Ashley Bauer
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
- Versiti Blood Research Institute, Versiti Wisconsin, Milwaukee, WI
- Department of Pathology, Northwestern University, Chicago, IL
| | - Donia Alson
- Versiti Blood Research Institute, Versiti Wisconsin, Milwaukee, WI
| | - Achia Khatun
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
- Versiti Blood Research Institute, Versiti Wisconsin, Milwaukee, WI
| | - Yao Chen
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
- Versiti Blood Research Institute, Versiti Wisconsin, Milwaukee, WI
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Sun
- University of Virginia School of Medicine, Charlottesville, VA
| | - William Drobyski
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Brian T Edelson
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Weiguo Cui
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
- Versiti Blood Research Institute, Versiti Wisconsin, Milwaukee, WI
- Department of Pathology, Northwestern University, Chicago, IL
| |
Collapse
|
28
|
Li XX, Chen ZD, Sun XJ, Yang YQ, Jin H, Liu NF. Empagliflozin ameliorates vascular calcification in diabetic mice through inhibiting Bhlhe40-dependent NLRP3 inflammasome activation. Acta Pharmacol Sin 2024; 45:751-764. [PMID: 38172306 PMCID: PMC10943241 DOI: 10.1038/s41401-023-01217-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/14/2023] [Indexed: 01/05/2024]
Abstract
Type 2 diabetes mellitus (T2DM) patients exhibit greater susceptibility to vascular calcification (VC), which has a higher risk of death and disability. However, there is no specific drug for VC therapy. NLRP3 inflammasome activation as a hallmark event of medial calcification leads to arterial stiffness, causing vasoconstrictive dysfunction in T2DM. Empagliflozin (EMPA), a sodium-glucose co-transporter 2 inhibitor (SGLT2i), restrains hyperglycemia with definite cardiovascular benefits. Given the anti-inflammatory activity of EMPA, herein we investigated whether EMPA protected against VC in the aorta of T2DM mice by inhibiting NLRP3 inflammasome activation. Since db/db mice receiving a normal diet developed VC at the age of about 20 weeks, we administered EMPA (5, 10, 20 mg·kg-1·d-1, i.g) to 8 week-old db/db mice for 12 weeks. We showed that EMPA intervention dose-dependently ameliorated the calcium deposition, accompanied by reduced expression of RUNX2 and BMP2 proteins in the aortas. We found that EMPA (10 mg·kg-1·d-1 for 6 weeks) also protected against VC in vitamin D3-overloaded mice, suggesting the protective effects independent of metabolism. We showed that EMPA (10 mg·kg-1·d-1) inhibited the abnormal activation of NLRP3 inflammasome in aortic smooth muscle layer of db/db mice. Knockout (KO) of NLRP3 significantly alleviated VC in STZ-induced diabetic mice. The protective effects of EMPA were verified in high glucose (HG)-treated mouse aortic smooth muscle cells (MOVASs). In HG-treated NLRP3 KO MOVASs, EMPA (1 μM) did not cause further improvement. Bioinformatics and Western blot analysis revealed that EMPA significantly increased the expression levels of basic helix-loop-helix family transcription factor e40 (Bhlhe40) in HG-treated MOVASs, which served as a negative transcription factor directly binding to the promotor of Nlrp3. We conclude that EMPA ameliorates VC by inhibiting Bhlhe40-dpendent NLRP3 inflammasome activation. These results might provide potential significance for EMPA in VC therapy of T2DM patients.
Collapse
Affiliation(s)
- Xiao-Xue Li
- Department of Cardiology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, 210009, China
| | - Zheng-Dong Chen
- Department of Cardiology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, 210009, China
| | - Xue-Jiao Sun
- Department of Cardiology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, 210009, China
| | - Yi-Qing Yang
- Department of Cardiology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, 210009, China
| | - Hong Jin
- Department of Cardiology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, 210009, China
| | - Nai-Feng Liu
- Department of Cardiology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, 210009, China.
| |
Collapse
|
29
|
Dos Santos JC, Moreno M, Teufel LU, Chilibroste S, Keating ST, Groh L, Domínguez-Andrés J, Williams DL, Ma Z, Lowman DW, Ensley HE, Novakovic B, Ribeiro-Dias F, Netea MG, Chabalgoity JA, Joosten LAB. Leishmania braziliensis enhances monocyte responses to promote anti-tumor activity. Cell Rep 2024; 43:113932. [PMID: 38457336 PMCID: PMC11000460 DOI: 10.1016/j.celrep.2024.113932] [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: 04/16/2023] [Revised: 11/07/2023] [Accepted: 02/21/2024] [Indexed: 03/10/2024] Open
Abstract
Innate immune cells can undergo long-term functional reprogramming after certain infections, a process called trained immunity (TI). Here, we focus on antigens of Leishmania braziliensis, which induced anti-tumor effects via trained immunity in human monocytes. We reveal that monocytes exposed to promastigote antigens of L. braziliensis develop an enhanced response to subsequent exposure to Toll-like receptor (TLR)2 or TLR4 ligands. Mechanistically, the induction of TI in monocytes by L. braziliensis is mediated by multiple pattern recognition receptors, changes in metabolism, and increased deposition of H3K4me3 at the promoter regions of immune genes. The administration of L. braziliensis exerts potent anti-tumor capabilities by delaying tumor growth and prolonging survival of mice with non-Hodgkin lymphoma. Our work reveals mechanisms of TI induced by L. braziliensis in vitro and identifies its potential for cancer immunotherapy.
Collapse
Affiliation(s)
- Jéssica Cristina Dos Santos
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - María Moreno
- Laboratory for Vaccine Research, Departamento de Desarrollo Biotecnológico, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Lisa U Teufel
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Sofía Chilibroste
- Laboratory for Vaccine Research, Departamento de Desarrollo Biotecnológico, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Samuel T Keating
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Laszlo Groh
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jorge Domínguez-Andrés
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - David L Williams
- Department of Surgery, Quillen College of Medicine, Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, TN, USA
| | - Zuchao Ma
- Department of Surgery, Quillen College of Medicine, Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, TN, USA
| | - Douglas W Lowman
- Department of Surgery, Quillen College of Medicine, Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, TN, USA
| | - Harry E Ensley
- Department of Surgery, Quillen College of Medicine, Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, TN, USA
| | - Boris Novakovic
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands; Murdoch Children's Research Institute and Department of Pediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Fátima Ribeiro-Dias
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands; Department for Immunology and Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - José A Chabalgoity
- Laboratory for Vaccine Research, Departamento de Desarrollo Biotecnológico, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands; Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
| |
Collapse
|
30
|
Hu X, Zou M, Zheng W, Zhu M, Hou Q, Gao H, Zhang X, Liu Y, Cheng Z. Bhlhe40 deficiency attenuates LPS-induced acute lung injury through preventing macrophage pyroptosis. Respir Res 2024; 25:100. [PMID: 38402153 PMCID: PMC10894472 DOI: 10.1186/s12931-024-02740-2] [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: 09/26/2023] [Accepted: 02/19/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND Acute lung injury (ALI) and its more severe form, acute respiratory distress syndrome (ARDS) as common life-threatening lung diseases with high mortality rates are mostly associated with acute and severe inflammation in lungs. Recently, increasing evidence supports activated inflammation and gasdermin D (GSDMD)-mediated pyroptosis in macrophage are closely associated with ALI. Basic helix-loop-helix family member e40 (Bhlhe40) is a transcription factor that is comprehensively involved in inflammation. However, there is little experimental evidence connecting Bhlhe40 and GSDMD-driven pyroptosis. The study sought to verify the hypothesis that Bhlhe40 is required for GSDMD-mediated pyroptosis in lipopolysaccharide (LPS)-induced inflammatory injury. METHOD We performed studies using Bhlhe40-knockout (Bhlhe40 -/-) mice, small interfering RNA (siRNA) targeting Bhlhe40 and pyroptosis inhibitor disulfiram to investigate the potential roles of Bhlhe40 on LPS-induced ALI and the underlying mechanisms. RESULTS Bhlhe40 was highly expressed in total lung tissues and macrophages of LPS-induced mice. Bhlhe40-/- mice showed alleviative lung pathological injury and inflammatory response upon LPS stimulation. Meanwhile, we found that Bhlhe40 deficiency significantly suppressed GSDMD-mediated pyroptosis in macrophage in vivo and in vitro. By further mechanistic analysis, we demonstrated that Bhlhe40 deficiency inhibited GSDMD-mediated pyroptosis and subsequent ALI by repressing canonical (caspase-1-mediated) and non-canonical (caspase-11-mediated) signaling pathways in vivo and in vitro. CONCLUSION These results indicate Bhlhe40 is required for LPS-induced ALI. Bhlhe40 deficiency can inhibit GSDMD-mediated pyroptosis and therefore alleviate ALI. Targeting Bhlhe40 may be a potential therapeutic strategy for LPS-induced ALI.
Collapse
Affiliation(s)
- Xingxing Hu
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Menglin Zou
- Fourth Ward of Medical Care Center, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Weishuai Zheng
- Department of Respiratory and Critical Care Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Minghui Zhu
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qinhui Hou
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Han Gao
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xin Zhang
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Yuan Liu
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Zhenshun Cheng
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, Hubei, China.
- Hubei Engineering Center for Infectious Disease Prevention, Control and Treatment, Wuhan, China.
| |
Collapse
|
31
|
Cao Y, Wang X, Liu Y, Liu P, Qin J, Zhu Y, Zhai S, Jiang Y, Liu Y, Han L, Luo J, Zhang R, Shi M, Wang L, Tang X, Xue M, Liu J, Wang W, Wen C, Deng X, Peng C, Chen H, Cheng D, Jiang L, Shen B. BHLHE40 Inhibits Ferroptosis in Pancreatic Cancer Cells via Upregulating SREBF1. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306298. [PMID: 38064101 PMCID: PMC10870036 DOI: 10.1002/advs.202306298] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/14/2023] [Indexed: 02/17/2024]
Abstract
Pancreatic cancer (PCa) is one of the most fatal human malignancies. The enhanced infiltration of stromal tissue into the PCa tumor microenvironment limits the identification of key tumor-specific transcription factors and epigenomic abnormalities in malignant epithelial cells. Integrated transcriptome and epigenetic multiomics analyses of the paired PCa organoids indicate that the basic helix-loop-helix transcription factor 40 (BHLHE40) is significantly upregulated in tumor samples. Increased chromatin accessibility at the promoter region and enhanced mTOR pathway activity contribute to the elevated expression of BHLHE40. Integrated analysis of chromatin immunoprecipitation-seq, RNA-seq, and high-throughput chromosome conformation capture data, together with chromosome conformation capture assays, indicate that BHLHE40 not only regulates sterol regulatory element-binding factor 1 (SREBF1) transcription as a classic transcription factor but also links the enhancer and promoter regions of SREBF1. It is found that the BHLHE40-SREBF1-stearoyl-CoA desaturase axis protects PCa cells from ferroptosis, resulting in the reduced accumulation of lipid peroxidation. Moreover, fatostatin, an SREBF1 inhibitor, significantly suppresses the growth of PCa tumors with high expressions of BHLHE40. This study highlights the important roles of BHLHE40-mediated lipid peroxidation in inducing ferroptosis in PCa cells and provides a novel mechanism underlying SREBF1 overexpression in PCa.
Collapse
|
32
|
Carbo-Meix A, Guijarro F, Wang L, Grau M, Royo R, Frigola G, Playa-Albinyana H, Buhler MM, Clot G, Duran-Ferrer M, Lu J, Granada I, Baptista MJ, Navarro JT, Espinet B, Puiggros A, Tapia G, Bandiera L, De Canal G, Bonoldi E, Climent F, Ribera-Cortada I, Fernandez-Caballero M, De la Banda E, Do Nascimento J, Pineda A, Vela D, Rozman M, Aymerich M, Syrykh C, Brousset P, Perera M, Yanez L, Ortin JX, Tuset E, Zenz T, Cook JR, Swerdlow SH, Martin-Subero JI, Colomer D, Matutes E, Bea S, Costa D, Nadeu F, Campo E. BCL3 rearrangements in B-cell lymphoid neoplasms occur in two breakpoint clusters associated with different diseases. Haematologica 2024; 109:493-508. [PMID: 37560801 PMCID: PMC10828791 DOI: 10.3324/haematol.2023.283209] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 07/31/2023] [Indexed: 08/11/2023] Open
Abstract
The t(14;19)(q32;q13) often juxtaposes BCL3 with immunoglobulin heavy chain (IGH) resulting in overexpression of the gene. In contrast to other oncogenic translocations, BCL3 rearrangement (BCL3-R) has been associated with a broad spectrum of lymphoid neoplasms. Here we report an integrative whole-genome sequence, transcriptomic, and DNA methylation analysis of 13 lymphoid neoplasms with BCL3-R. The resolution of the breakpoints at single base-pair revealed that they occur in two clusters at 5' (n=9) and 3' (n=4) regions of BCL3 associated with two different biological and clinical entities. Both breakpoints were mediated by aberrant class switch recombination of the IGH locus. However, the 5' breakpoints (upstream) juxtaposed BCL3 next to an IGH enhancer leading to overexpression of the gene whereas the 3' breakpoints (downstream) positioned BCL3 outside the influence of the IGH and were not associated with its expression. Upstream BCL3-R tumors had unmutated IGHV, trisomy 12, and mutated genes frequently seen in chronic lymphocytic leukemia (CLL) but had an atypical CLL morphology, immunophenotype, DNA methylome, and expression profile that differ from conventional CLL. In contrast, downstream BCL3-R neoplasms were atypical splenic or nodal marginal zone lymphomas (MZL) with mutated IGHV, complex karyotypes and mutated genes typical of MZL. Two of the latter four tumors transformed to a large B-cell lymphoma. We designed a novel fluorescence in situ hybridization assay that recognizes the two different breakpoints and validated these findings in 17 independent tumors. Overall, upstream or downstream breakpoints of BCL3-R are mainly associated with two subtypes of lymphoid neoplasms with different (epi)genomic, expression, and clinicopathological features resembling atypical CLL and MZL, respectively.
Collapse
Affiliation(s)
- Anna Carbo-Meix
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona
| | - Francesca Guijarro
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Hematopathology Section, laboratory of Pathology, Hospital Clínic de Barcelona, Barcelona
| | - Luojun Wang
- Hematopathology Section, laboratory of Pathology, Hospital Clínic de Barcelona, Barcelona
| | - Marta Grau
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona
| | - Romina Royo
- Barcelona Supercomputing Center (BSC), Barcelona
| | - Gerard Frigola
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Hematopathology Section, laboratory of Pathology, Hospital Clínic de Barcelona, Barcelona
| | - Heribert Playa-Albinyana
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid
| | - Marco M Buhler
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich
| | - Guillem Clot
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona
| | - Marti Duran-Ferrer
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid
| | - Junyan Lu
- European Molecular Biology Laboratory, Heidelberg
| | - Isabel Granada
- Department of Hematology-Laboratory, Institut Català d'Oncologia, Hospital Germans Trias i Pujol, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona
| | - Maria-Joao Baptista
- Department of Hematology-Laboratory, Institut Català d'Oncologia, Hospital Germans Trias i Pujol, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona
| | - Jose-Tomas Navarro
- Department of Hematology-Laboratory, Institut Català d'Oncologia, Hospital Germans Trias i Pujol, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona
| | - Blanca Espinet
- Molecular Cytogenetics Laboratory, Pathology Department, Hospital del Mar, Barcelona, Spain and Translational Research on Hematological Neoplasms Group (GRETNHE) - Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona
| | - Anna Puiggros
- Molecular Cytogenetics Laboratory, Pathology Department, Hospital del Mar, Barcelona, Spain and Translational Research on Hematological Neoplasms Group (GRETNHE) - Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona
| | - Gustavo Tapia
- Department of Pathology, Hospital Germans Trias i Pujol, Badalona
| | - Laura Bandiera
- Anatomia Istologia Patologica e Citogenetica, Dipartimento Ematologia, Oncologia e Medicina Molecolare, Niguarda Cancer Center, Milano
| | - Gabriella De Canal
- Anatomia Istologia Patologica e Citogenetica, Dipartimento Ematologia, Oncologia e Medicina Molecolare, Niguarda Cancer Center, Milano
| | - Emanuela Bonoldi
- Anatomia Istologia Patologica e Citogenetica, Dipartimento Ematologia, Oncologia e Medicina Molecolare, Niguarda Cancer Center, Milano
| | - Fina Climent
- Department o f Pathology, H ospital Universitari d e Bellvitge, I nstitut d'Investigació B iomèdica d e Bellvitge (IDIBELL), L'Hospitalet De Llobregat
| | | | - Mariana Fernandez-Caballero
- Department of Hematology-Laboratory, Institut Català d'Oncologia, Hospital Germans Trias i Pujol, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona
| | - Esmeralda De la Banda
- Laboratory of Hematology, Hospital Universitari Bellvitge, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet De Llobregat
| | | | | | - Dolors Vela
- Hematologia Clínica, Hospital General de Granollers, Granollers
| | - Maria Rozman
- Hematopathology Section, laboratory of Pathology, Hospital Clínic de Barcelona, Barcelona
| | - Marta Aymerich
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Hematopathology Section, laboratory of Pathology, Hospital Clínic de Barcelona, Barcelona
| | - Charlotte Syrykh
- Department of Pathology, Toulouse University Hospital Center, Cancer Institute University of Toulouse-Oncopole, 1 avenue Irène Joliot-Curie, 31059, Toulouse CEDEX 9
| | - Pierre Brousset
- Department of Pathology, Toulouse University Hospital Center, Cancer Institute University of Toulouse-Oncopole, 1 avenue Irène Joliot-Curie, 31059, Toulouse CEDEX 9, France; INSERM UMR1037 Cancer Research Center of Toulouse (CRCT), ERL 5294 National Center for Scientific Research (CNRS), University of Toulouse III Paul-Sabatier, Toulouse, France; Institut Carnot Lymphome CALYM, Laboratoire d'Excellence 'TOUCAN', Toulouse
| | - Miguel Perera
- Hematology Department, Hospital Dr Negrín, Las Palmas de Gran Canaria
| | - Lucrecia Yanez
- Hematology Department, Hospital Universitario Marqués de Valdecilla-Instituto de Investigación Valdecilla (IDIVAL), Santander
| | | | - Esperanza Tuset
- Hematology Department, Institut Català d'Oncologia, Hospital Dr. Josep Trueta, Institut d'Investigació Biomèdica de Girona (IDIBGI), Girona
| | - Thorsten Zenz
- Department of Medical Oncology and Hematology, University Hospital and University of Zürich, Zurich
| | - James R Cook
- Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH
| | - Steven H Swerdlow
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Jose I Martin-Subero
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Universitat de Barcelona, Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona
| | - Dolors Colomer
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Hematopathology Section, laboratory of Pathology, Hospital Clínic de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Universitat de Barcelona, Barcelona
| | - Estella Matutes
- Hematopathology Section, laboratory of Pathology, Hospital Clínic de Barcelona, Barcelona
| | - Silvia Bea
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Hematopathology Section, laboratory of Pathology, Hospital Clínic de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Universitat de Barcelona, Barcelona
| | - Dolors Costa
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Hematopathology Section, laboratory of Pathology, Hospital Clínic de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid
| | - Ferran Nadeu
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid
| | - Elias Campo
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Hematopathology Section, laboratory of Pathology, Hospital Clínic de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Universitat de Barcelona, Barcelona.
| |
Collapse
|
33
|
Teo WY, Lim YYE, Sio YY, Say YH, Reginald K, Chew FT. Atopic dermatitis-associated genetic variants regulate LOC100294145 expression implicating interleukin-27 production and type 1 interferon signaling. World Allergy Organ J 2024; 17:100869. [PMID: 38298829 PMCID: PMC10827559 DOI: 10.1016/j.waojou.2023.100869] [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: 09/29/2023] [Revised: 12/14/2023] [Accepted: 12/27/2023] [Indexed: 02/02/2024] Open
Abstract
Background Atopic dermatitis (AD) is a complex inflammatory disease with a strong genetic component. A singular approach of genome wide association studies (GWAS) can identify AD-associated genetic variants, but is unable to explain their functional relevance in AD. This study aims to characterize AD-associated genetic variants and elucidate the mechanisms leading to AD through a multi-omics approach. Methods GWAS identified an association between genetic variants at 6p21.32 locus and AD. Genotypes of 6p21.32 locus variants were evaluated against LOC100294145 expression in peripheral blood mononuclear cells (PBMCs). Their influence on LOC100294145 promoter activity was measured in vitro via a dual-luciferase assay. The function of LOC100294145 was then elucidated through a combination of co-expression analyses and gene enrichment with g:Profiler. Mendelian randomization was further used to assess the causal regulatory effect of LOC100294145 on its co-expressed genes. Results Minor alleles of rs116160149 and rs115388857 at 6p21.32 locus were associated with increased AD risk (p = 2.175 × 10-8, OR = 1.552; p = 2.805 × 10-9, OR = 1.55) and higher LOC100294145 expression in PBMCs (adjusted p = 0.182; 8.267 × 10-12). LOC100294145 expression was also found to be increased in those with AD (adjusted p = 3.653 × 10-2). The genotype effect of 6p21.32 locus on LOC100294145 promoter activity was further validated in vitro. Co-expression analyses predicted LOC100294145 protein's involvement in interleukin-27 and type 1 interferon signaling, which was further substantiated through mendelian randomization. Conclusion Genetic variants at 6p21.32 locus increase AD susceptibility through raising LOC100294145 expression. A multi-omics approach enabled the deduction of its pathogenesis model comprising dysregulation of hub genes involved in type 1 interferon and interleukin 27 signaling.
Collapse
Affiliation(s)
- Wei Yi Teo
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Yi Ying Eliza Lim
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Yang Yie Sio
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Yee-How Say
- Department of Biological Sciences, National University of Singapore, Singapore
- Department of Biomedical Science, Faculty of Science, Universiti Tunku Abdul Rahman (UTAR) Kampar Campus, Kampar, Perak, Malaysia
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Malaysia
| | - Kavita Reginald
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Malaysia
| | - Fook Tim Chew
- Department of Biological Sciences, National University of Singapore, Singapore
| |
Collapse
|
34
|
Mercado MAB, Li Q, Quick CM, Kim Y, Palmer R, Huang L, Li LX. BHLHE40 drives protective polyfunctional CD4 T cell differentiation in the female reproductive tract against Chlamydia. PLoS Pathog 2024; 20:e1011983. [PMID: 38271477 PMCID: PMC10846703 DOI: 10.1371/journal.ppat.1011983] [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: 11/21/2023] [Revised: 02/06/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
The protein basic helix-loop-helix family member e40 (BHLHE40) is a transcription factor recently emerged as a key regulator of host immunity to infections, autoimmune diseases and cancer. In this study, we investigated the role of Bhlhe40 in protective T cell responses to the intracellular bacterium Chlamydia in the female reproductive tract (FRT). Mice deficient in Bhlhe40 exhibited severe defects in their ability to control Chlamydia muridarum shedding from the FRT. The heightened bacterial burdens in Bhlhe40-/- mice correlated with a marked increase in IL-10-producing T regulatory type 1 (Tr1) cells and decreased polyfunctional CD4 T cells co-producing IFN-γ, IL-17A and GM-CSF. Genetic ablation of IL-10 or functional blockade of IL-10R increased CD4 T cell polyfunctionality and partially rescued the defects in bacterial control in Bhlhe40-/- mice. Using single-cell RNA sequencing coupled with TCR profiling, we detected a significant enrichment of stem-like T cell signatures in Bhlhe40-deficient CD4 T cells, whereas WT CD4 T cells were further down on the differentiation trajectory with distinct effector functions beyond IFN-γ production by Th1 cells. Altogether, we identified Bhlhe40 as a key molecular driver of CD4 T cell differentiation and polyfunctional responses in the FRT against Chlamydia.
Collapse
Affiliation(s)
- Miguel A. B. Mercado
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Qiang Li
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Charles M. Quick
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Yejin Kim
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Rachel Palmer
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Lu Huang
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Lin-Xi Li
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| |
Collapse
|
35
|
Jiao A, Zhang C, Wang X, Sun L, Liu H, Su Y, Lei L, Li W, Ding R, Ding C, Dou M, Tian P, Sun C, Yang X, Zhang L, Zhang B. Single-cell sequencing reveals the evolution of immune molecules across multiple vertebrate species. J Adv Res 2024; 55:73-87. [PMID: 36871615 PMCID: PMC10770119 DOI: 10.1016/j.jare.2023.02.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 02/11/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
INTRODUCTION Both innate and adaptive immune system undergo evolution from low to high vertebrates. Due to the limitation of conventional approaches in identifying broader spectrum of immune cells and molecules from various vertebrates, it remains unclear how immune molecules evolve among vertebrates. OBJECTIVES Here, we utilized carry out comparative transcriptome analysis in various immune cells across seven vertebrate species. METHODS Single-cell RNA sequencing (scRNA-seq). RESULTS We uncovered both conserved and species-specific profiling of gene expression in innate and adaptive immunity. Macrophages exhibited highly-diversified genes and developed sophisticated molecular signaling networks along with evolution, indicating effective and versatile functions in higher species. In contrast, B cells conservatively evolved with less differentially-expressed genes in analyzed species. Interestingly, T cells represented a dominant immune cell populations in all species and unique T cell populations were identified in zebrafish and pig. We also revealed compensatory TCR cascade components utilized by different species. Inter-species comparison of core gene programs demonstrated mouse species has the highest similarity in immune transcriptomes to human. CONCLUSIONS Therefore, our comparative study reveals gene transcription characteristics across multiple vertebrate species during the evolution of immune system, providing insights for species-specific immunity as well as the translation of animal studies to human physiology and disease.
Collapse
Affiliation(s)
- Anjun Jiao
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Cangang Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Xin Wang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Lina Sun
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Haiyan Liu
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Yanhong Su
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Lei Lei
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi 710061, China; Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shaanxi 710061, China
| | - Wenhua Li
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Renyi Ding
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Chenguang Ding
- The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Meng Dou
- The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Puxun Tian
- The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Chenming Sun
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi 710061, China; Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shaanxi 710061, China
| | - Xiaofeng Yang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi 710061, China; Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shaanxi 710061, China.
| | - Lianjun Zhang
- Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China; Suzhou Institute of Systems Medicine, Suzhou 215123, China.
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi 710061, China; Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
36
|
Lee JD, Menasche BL, Mavrikaki M, Uyemura MM, Hong SM, Kozlova N, Wei J, Alfajaro MM, Filler RB, Müller A, Saxena T, Posey RR, Cheung P, Muranen T, Heng YJ, Paulo JA, Wilen CB, Slack FJ. Differences in syncytia formation by SARS-CoV-2 variants modify host chromatin accessibility and cellular senescence via TP53. Cell Rep 2023; 42:113478. [PMID: 37991919 PMCID: PMC10785701 DOI: 10.1016/j.celrep.2023.113478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 09/13/2023] [Accepted: 11/06/2023] [Indexed: 11/24/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) remains a significant public health threat due to the ability of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants to evade the immune system and cause breakthrough infections. Although pathogenic coronaviruses such as SARS-CoV-2 and Middle East respiratory syndrome (MERS)-CoV lead to severe respiratory infections, how these viruses affect the chromatin proteomic composition upon infection remains largely uncharacterized. Here, we use our recently developed integrative DNA And Protein Tagging methodology to identify changes in host chromatin accessibility states and chromatin proteomic composition upon infection with pathogenic coronaviruses. SARS-CoV-2 infection induces TP53 stabilization on chromatin, which contributes to its host cytopathic effect. We mapped this TP53 stabilization to the SARS-CoV-2 spike and its propensity to form syncytia, a consequence of cell-cell fusion. Differences in SARS-CoV-2 spike variant-induced syncytia formation modify chromatin accessibility, cellular senescence, and inflammatory cytokine release via TP53. Our findings suggest that differences in syncytia formation alter senescence-associated inflammation, which varies among SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Jonathan D Lee
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
| | - Bridget L Menasche
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT 06520, USA; Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Maria Mavrikaki
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Madison M Uyemura
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Su Min Hong
- Department of Genetics, Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Nina Kozlova
- Department of Genetics, Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Jin Wei
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT 06520, USA; Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Mia M Alfajaro
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT 06520, USA; Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Renata B Filler
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT 06520, USA; Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Arne Müller
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Tanvi Saxena
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Ryan R Posey
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Priscilla Cheung
- Stem Cell Program, Boston Children's Hospital, Boston, MA 02115, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Taru Muranen
- Department of Genetics, Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Yujing J Heng
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Craig B Wilen
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT 06520, USA; Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Frank J Slack
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; Department of Genetics, Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; Harvard Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
37
|
Pal A, Ojha A, Ju J. Functional and Potential Therapeutic Implication of MicroRNAs in Pancreatic Cancer. Int J Mol Sci 2023; 24:17523. [PMID: 38139352 PMCID: PMC10744132 DOI: 10.3390/ijms242417523] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/01/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
The alarmingly low five-year survival rate for pancreatic cancer presents a global health challenge, contributing to about 7% of all cancer-related deaths. Late-stage diagnosis and high heterogeneity are the biggest hurdles in treating pancreatic cancer. Thus, there is a pressing need to discover novel biomarkers that could help in early detection as well as improve therapeutic strategies. MicroRNAs (miRNAs), a class of short non-coding RNA, have emerged as promising candidates with regard to both diagnostics and therapeutics. Dysregulated miRNAs play pivotal roles in accelerating tumor growth and metastasis, orchestrating tumor microenvironment, and conferring chemoresistance in pancreatic cancer. The differential expression profiles of miRNAs in pancreatic cancer could be utilized to explore novel therapeutic strategies. In this review, we also covered studies on recent advancements in various miRNA-based therapeutics such as restoring miRNAs with a tumor-suppressive function, suppressing miRNA with an oncogenic function, and combination with chemotherapeutic drugs. Despite several challenges in terms of specificity and targeted delivery, miRNA-based therapies hold the potential to revolutionize the treatment of pancreatic cancer by simultaneously targeting multiple signaling pathways.
Collapse
Affiliation(s)
- Amartya Pal
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; (A.P.); (A.O.)
- Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Anushka Ojha
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; (A.P.); (A.O.)
- Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Jingfang Ju
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; (A.P.); (A.O.)
- The Northport Veteran’s Administration Medical Center, Northport, NY 11768, USA
| |
Collapse
|
38
|
O'Neal KA, Zeltner SL, Foscue CL, Stumhofer JS. Bhlhe40 limits early IL-10 production from CD4 + T cells during Plasmodium yoelii 17X infection. Infect Immun 2023; 91:e0036723. [PMID: 37843306 PMCID: PMC10652903 DOI: 10.1128/iai.00367-23] [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: 09/06/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
The cytokine IL-10 suppresses T-cell-mediated immunity, which is required to control infection with Plasmodium yoelii. Consequently, IL-10 can delay the time needed to resolve this infection, leading to a higher parasite burden. While the pathways that lead to IL-10 production by CD4+ T cells are well defined, much less is known about the mediators that suppress the expression of this potent anti-inflammatory cytokine. Here, we show that the transcription factor basic helix-loop-helix family member e40 (Bhlhe40) contributes to controlling parasite burden in response to P. yoelii infection in mice. Loss of Bhlhe40 expression in mice results in higher Il10 expression, higher peak parasitemia, and a delay in parasite clearance. The observed phenotype was not due to defects in T-cell activation and proliferation or the humoral response. Nor was it due to changes in regulatory T-cell numbers. However, blocking IL-10 signaling reversed the outcome in Bhlhe40-/ - mice, suggesting that excess IL-10 production limits their ability to control the infection properly. In addition to suppressing Il10 expression in CD4+ T cells, Bhlhe40 can promote Ifng expression. Indeed, IFN-γ production by CD4+ T cells isolated from the liver was significantly affected by the loss of Bhlhe40. Lastly, Bhlhe40 deletion in T cells resulted in a phenotype similar to that observed in the Bhlhe40-/ - mice, indicating that Bhlhe40 expression in T cells contributes to the ability of mice to control infection with P. yoelii.
Collapse
Affiliation(s)
- Kara A. O'Neal
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Sheldon L. Zeltner
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Camille L. Foscue
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Jason S. Stumhofer
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
39
|
Kim H, Abbasi A, Sharrock J, Santosa EK, Lau CM, Edelson BT, Sun JC. Cutting Edge: STAT4 Promotes Bhlhe40 Induction to Drive Protective IFN-γ from NK Cells during Viral Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1469-1474. [PMID: 37830760 PMCID: PMC10842983 DOI: 10.4049/jimmunol.2300402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 09/12/2023] [Indexed: 10/14/2023]
Abstract
NK cells represent a cellular component of the mammalian innate immune system, and they mount rapid responses against viral infection, including the secretion of the potent antiviral effector cytokine IFN-γ. Following mouse CMV infection, Bhlhe40 was the most highly induced transcription factor in NK cells among the basic helix-loop-helix family. Bhlhe40 upregulation in NK cells depended upon IL-12 and IL-18 signals, with the promoter of Bhlhe40 enriched for STAT4 and the permissive histone H3K4me3, and with STAT4-deficient NK cells showing an impairment of Bhlhe40 induction and diminished H3K4me3. Transcriptomic and protein analysis of Bhlhe40-deficient NK cells revealed a defect in IFN-γ production during mouse CMV infection, resulting in diminished protective immunity following viral challenge. Finally, we provide evidence that Bhlhe40 directly promotes IFN-γ by binding throughout the Ifng loci in activated NK cells. Thus, our study reveals how STAT4-mediated control of Bhlhe40 drives protective IFN-γ secretion by NK cells during viral infection.
Collapse
Affiliation(s)
- Hyunu Kim
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065
| | - Aamna Abbasi
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065
| | - Jessica Sharrock
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065
| | - Endi K. Santosa
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065
- Department of Immunology and Microbial Pathogenesis, Weill Cornell Medical College, New York, NY 10065
| | - Colleen M. Lau
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065
| | - Brian T. Edelson
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Joseph C. Sun
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065
- Department of Immunology and Microbial Pathogenesis, Weill Cornell Medical College, New York, NY 10065
| |
Collapse
|
40
|
Balmas E, Chen J, Hu AK, DeBerg HA, Rosasco MG, Gersuk VH, Serti E, Speake C, Greenbaum CJ, Nepom GT, Linsley PS, Cerosaletti K. Islet-autoreactive CD4+ T cells are linked with response to alefacept in type 1 diabetes. JCI Insight 2023; 8:e167881. [PMID: 37751304 PMCID: PMC10721267 DOI: 10.1172/jci.insight.167881] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 09/21/2023] [Indexed: 09/27/2023] Open
Abstract
Variation in the preservation of β cell function in clinical trials in type 1 diabetes (T1D) has emphasized the need to define biomarkers to predict treatment response. The T1DAL trial targeted T cells with alefacept (LFA-3-Ig) and demonstrated C-peptide preservation in approximately 30% of new-onset T1D individuals. We analyzed islet antigen-reactive (IAR) CD4+ T cells in PBMC samples collected prior to treatment from alefacept- and placebo-treated individuals using flow cytometry and single-cell RNA sequencing. IAR CD4+ T cells at baseline had heterogeneous phenotypes. Transcript profiles formed phenotypic clusters of cells along a trajectory based on increasing maturation and activation, and T cell receptor (TCR) chains showed clonal expansion. Notably, the frequency of IAR CD4+ T cells with a memory phenotype and a unique transcript profile (cluster 3) were inversely correlated with C-peptide preservation in alefacept-treated, but not placebo-treated, individuals. Cluster 3 cells had a proinflammatory phenotype characterized by expression of the transcription factor BHLHE40 and the cytokines GM-CSF and TNF-α, and shared TCR chains with effector memory-like clusters. Our results suggest IAR CD4+ T cells as a potential baseline biomarker of response to therapies targeting the CD2 pathway and warrant investigation for other T cell-related therapies.
Collapse
Affiliation(s)
| | | | - Alex K. Hu
- Center for Systems Immunology, Benaroya Research Institute, Seattle, Washington
| | - Hannah A. DeBerg
- Center for Systems Immunology, Benaroya Research Institute, Seattle, Washington
| | - Mario G. Rosasco
- Center for Systems Immunology, Benaroya Research Institute, Seattle, Washington
| | - Vivian H. Gersuk
- Center for Systems Immunology, Benaroya Research Institute, Seattle, Washington
| | | | - Cate Speake
- Center for Interventional Immunology and Diabetes Clinical Research Program, Benaroya Research Institute, Seattle, Washington, USA
| | - Carla J. Greenbaum
- Center for Interventional Immunology and Diabetes Clinical Research Program, Benaroya Research Institute, Seattle, Washington, USA
| | | | - Peter S. Linsley
- Center for Systems Immunology, Benaroya Research Institute, Seattle, Washington
| | | |
Collapse
|
41
|
Mercado MAB, Li Q, Quick CM, Kim Y, Palmer R, Huang L, Li LX. BHLHE40 drives protective polyfunctional CD4 T cell differentiation in the female reproductive tract against Chlamydia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.02.565369. [PMID: 37961221 PMCID: PMC10635079 DOI: 10.1101/2023.11.02.565369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
The protein basic helix-loop-helix family member e40 (BHLHE40) is a transcription factor recently emerged as a key regulator of host immunity to infections, autoimmune diseases and cancer. In this study, we investigated the role of Bhlhe40 in protective T cell responses to the intracellular bacterium Chlamydia in the female reproductive tract (FRT). Mice deficient in Bhlhe40 exhibited severe defects in their ability to control Chlamydia muridarum shedding from the FRT. The heightened bacterial burdens in Bhlhe40-/- mice correlated with a marked increase in IL-10-producing T regulatory type 1 (Tr1) cells and decreased polyfunctional CD4 T cells co-producing IFN-γ, IL-17A and GM-CSF. Genetic ablation of IL-10 or functional blockade of IL-10R increased CD4 T cell polyfunctionality and partially rescued the defects in bacterial control in Bhlhe40-/- mice. Using single-cell RNA sequencing coupled with TCR profiling, we detected a significant enrichment of stem-like T cell signatures in Bhlhe40-deficient CD4 T cells, whereas WT CD4 T cells were further down on the differentiation trajectory with distinct effector functions beyond IFN-γ production by Th1 cells. Altogether, we identified Bhlhe40 as a key molecular driver of CD4 T cell differentiation and polyfunctional responses in the FRT against Chlamydia.
Collapse
Affiliation(s)
- Miguel A. B. Mercado
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Qiang Li
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Charles M. Quick
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Yejin Kim
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Rachel Palmer
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Lu Huang
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Lin-Xi Li
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| |
Collapse
|
42
|
Cook ME, Shchukina I, Lin CC, Bradstreet TR, Schwarzkopf EA, Jarjour NN, Webber AM, Zaitsev K, Artyomov MN, Edelson BT. BHLHE40 Mediates Cross-Talk between Pathogenic TH17 Cells and Myeloid Cells during Experimental Autoimmune Encephalomyelitis. Immunohorizons 2023; 7:737-746. [PMID: 37934060 PMCID: PMC10695412 DOI: 10.4049/immunohorizons.2300042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 10/22/2023] [Indexed: 11/08/2023] Open
Abstract
TH17 cells are implicated in the pathogenesis of multiple sclerosis and experimental autoimmune encephalomyelitis (EAE). We previously reported that the transcription factor basic helix-loop-helix family member e40 (BHLHE40) marks cytokine-producing pathogenic TH cells during EAE, and that its expression in T cells is required for clinical disease. In this study, using dual reporter mice, we show BHLHE40 expression within TH1/17 and ex-TH17 cells following EAE induction. Il17a-Cre-mediated deletion of BHLHE40 in TH cells led to less severe EAE with reduced TH cell cytokine production. Characterization of the leukocytes in the CNS during EAE by single-cell RNA sequencing identified differences in the infiltrating myeloid cells when BHLHE40 was present or absent in TH17 cells. Our studies highlight the importance of BHLHE40 in promoting TH17 cell encephalitogenicity and instructing myeloid cell responses during active EAE.
Collapse
Affiliation(s)
- Melissa E. Cook
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Irina Shchukina
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Chih-Chung Lin
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Tara R. Bradstreet
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | | | - Nicholas N. Jarjour
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Ashlee M. Webber
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Konstantin Zaitsev
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Maxim N. Artyomov
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Brian T. Edelson
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
43
|
Xu Y, Jiang K, Su F, Deng R, Cheng Z, Wang D, Yu Y, Xiang Y. A transient wave of Bhlhe41 + resident macrophages enables remodeling of the developing infarcted myocardium. Cell Rep 2023; 42:113174. [PMID: 37751357 DOI: 10.1016/j.celrep.2023.113174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/03/2023] [Accepted: 09/06/2023] [Indexed: 09/28/2023] Open
Abstract
The immune system plays a critical role during myocardial injury, contributing to repair and remodeling post myocardial infarction (MI). The myocardial infarct and border zone exhibit high heterogeneity, in turn leading to reconstructing macrophage subsets and specific functions. Here we use a combination of single-cell RNA sequencing, spatial transcriptomes, and reporter mice to characterize temporal-spatial dynamics of cardiac macrophage subtype in response to MI. We identify that transient appearance of monocyte-derived Bhlhe41+ Mφs in the "developing" infarct zone peaked at day 7, while other monocyte-derived macrophages are identified in "old" infarct zone. Functional characterization by co-culture of Bhlhe41+ Mφs with cardiomyocytes and fibroblasts or depletion of Bhlhe41+ Mφs unveils a crucial contribution of Bhlhe41+ Mφs in suppression of myofibroblast activation. This work highlights the importance of Bhlhe41+ Mφ phenotype and plasticity in preventing excessive fibrosis and limiting the expansion of developing infarct area.
Collapse
Affiliation(s)
- Yue Xu
- Shanghai East Hospital, Key Laboratory of Arrhythmias of the Ministry of Education of China, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Kai Jiang
- Shanghai East Hospital, Key Laboratory of Arrhythmias of the Ministry of Education of China, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Fanghua Su
- Shanghai East Hospital, Key Laboratory of Arrhythmias of the Ministry of Education of China, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Ruhua Deng
- Shanghai East Hospital, Key Laboratory of Arrhythmias of the Ministry of Education of China, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Zhiyang Cheng
- Shanghai East Hospital, Key Laboratory of Arrhythmias of the Ministry of Education of China, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Dandan Wang
- Shanghai East Hospital, Key Laboratory of Arrhythmias of the Ministry of Education of China, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yong Yu
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yaozu Xiang
- Shanghai East Hospital, Key Laboratory of Arrhythmias of the Ministry of Education of China, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| |
Collapse
|
44
|
Qi W, Liu Q, Fu W, Shi J, Shi M, Duan S, Li Z, Song S, Wang J, Liu Y. BHLHE40, a potential immune therapy target, regulated by FGD5-AS1/miR-15a-5p in pancreatic cancer. Sci Rep 2023; 13:16400. [PMID: 37773521 PMCID: PMC10541890 DOI: 10.1038/s41598-023-43577-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 09/26/2023] [Indexed: 10/01/2023] Open
Abstract
Pancreatic cancer, as one of the neoplasms with the highest degree of malignancy, has become a main disease of concerns in recent years. BHLHE40, a critical transcription factor for remodeling of the tumor immune microenvironment, has been described to be substantially increased in a variety of tumor-associated immune cells. Nevertheless, the pro-cancer biological functions and underlying molecular mechanisms of BHLHE40 for pancreatic cancer and its unique microenvironment are unclear. Hereby, we investigated the pro-oncogenic role of BHLHE40 in the pancreatic cancer microenvironment by bioinformatics analysis and cell biology experiments and determined that the expression of BHLHE40 was obviously elevated in pancreatic cancer tissues than in adjacent normal tissues. In parallel, Kaplan-Meier survival analysis unveiled that lower expression of BHLHE40 was strongly associated with better prognosis of patients. Receiver operating characteristic (ROC) curve analysis confirmed the accuracy of the BHLHE40-related prediction model. Subsequent, spearman correlation analysis observed that higher expression of BHLHE40 might be involved in immunosuppression of pancreatic cancer. Silencing of BHLHE40 could inhibit proliferation, invasion, and apoptosis of pancreatic cancer in vitro and in vivo, implying that BHLHE40 is expected to be a potential therapeutic target for pancreatic cancer. In addition, we explored and validated the FGD5-AS1/miR-15a-5p axis as a potential upstream regulatory mode for high expression of BHLHE40 in pancreatic cancer. In summary, our data showed that ceRNA involved in the regulation of BHLHE40 contributes to the promotion of immunosuppressive response in pancreatic and is expected to be a diagnostic marker and potential immunotherapeutic target for pancreatic cancer.
Collapse
Affiliation(s)
- Wenxin Qi
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Qian Liu
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Wenjun Fu
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Jiaming Shi
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Minmin Shi
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Songqi Duan
- College of Food Science, Sichuan Agricultural University, Yaan, China
| | - Zhe Li
- School of Life Sciences, Shanghai University, Shanghai, China.
| | - Shaohua Song
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China.
| | - Jiao Wang
- School of Life Sciences, Shanghai University, Shanghai, China.
| | - Yihao Liu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
45
|
Lu C, Li S, Qing P, Zhang Q, Ji X, Tang Z, Chen C, Wu T, Hu Y, Zhao Y, Zhang X, He Q, Fox DA, Tan C, Luo Y, Liu Y. Single-cell transcriptome analysis and protein profiling reveal broad immune system activation in IgG4-related disease. JCI Insight 2023; 8:e167602. [PMID: 37561593 PMCID: PMC10544205 DOI: 10.1172/jci.insight.167602] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 07/27/2023] [Indexed: 08/12/2023] Open
Abstract
IgG4-related disease (IgG4-RD) is a systemic autoimmune disease with unclear pathogenesis. We performed single-cell RNA-seq and surface proteome analyses on 61,379 PBMCs from 9 treatment-naive IgG4-RD patients and 7 age- and sex-matched healthy controls. Integrative analyses were performed for altered gene expression in IgG4-RD, and flow cytometry and immunofluorescence were used for validation. We observed expansion of plasmablasts with enhanced protein processing and activation, which correlated with the number of involved organs in IgG4-RD. Increased proportions of CD4+ cytotoxic T lymphocytes (CTLs), CD8+ CTLs-GNLY (granulysin), and γδT cells with enhanced chemotaxis and cytotoxicity but with suppressed inhibitory receptors characterize IgG4-RD. Prominent infiltration of lymphocytes with distinct compositions were found in different organs of IgG4-RD patients. Transcription factors (TFs), including PRDM1/XBP1 and RUNX3, were upregulated in IgG4-RD, promoting the differentiation of plasmablasts and CTLs, respectively. Monocytes in IgG4-RD have stronger expression of genes related to cell adhesion and chemotaxis, which may give rise to profibrotic macrophages in lesions. The gene activation pattern in peripheral immune cells indicated activation of multiple interaction pathways between cell types, in part through chemokines or growth factors and their receptors. Specific upregulation of TFs and expansion of plasmablasts and CTLs may be involved in the pathogenesis of IgG4-RD, and each of these populations are candidate targets for therapeutic interventions in this disease.
Collapse
Affiliation(s)
- Chenyang Lu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
- Division of Rheumatology, Department of Internal Medicine, and
| | - Shasha Li
- Guangdong Provincial Key Laboratory of Diabetology & Guangzhou Municipal Key Laboratory of Mechanistic and Translational Obesity Research, Medical Center for Comprehensive Weight Control, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Pingying Qing
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Qiuping Zhang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Xing Ji
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Zhigang Tang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Chunyan Chen
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Tong Wu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Yidan Hu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Zhao
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaohui Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qi He
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - David A. Fox
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Chunyu Tan
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Yubin Luo
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
46
|
Fowler JWM, Boutagy NE, Zhang R, Horikami D, Whalen MB, Romanoski CE, Sessa WC. SREBP2 regulates the endothelial response to cytokines via direct transcriptional activation of KLF6. J Lipid Res 2023; 64:100411. [PMID: 37437844 PMCID: PMC10407908 DOI: 10.1016/j.jlr.2023.100411] [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: 03/22/2023] [Revised: 06/14/2023] [Accepted: 06/27/2023] [Indexed: 07/14/2023] Open
Abstract
The transcription factor SREBP2 is the main regulator of cholesterol homeostasis and is central to the mechanism of action of lipid-lowering drugs, such as statins, which are responsible for the largest overall reduction in cardiovascular risk and mortality in humans with atherosclerotic disease. Recently, SREBP2 has been implicated in leukocyte innate and adaptive immune responses by upregulation of cholesterol flux or direct transcriptional activation of pro-inflammatory genes. Here, we investigate the role of SREBP2 in endothelial cells (ECs), since ECs are at the interface of circulating lipids with tissues and crucial to the pathogenesis of cardiovascular disease. Loss of SREBF2 inhibits the production of pro-inflammatory chemokines but amplifies type I interferon response genes in response to inflammatory stimulus. Furthermore, SREBP2 regulates chemokine expression not through enhancement of endogenous cholesterol synthesis or lipoprotein uptake but partially through direct transcriptional activation. Chromatin immunoprecipitation sequencing of endogenous SREBP2 reveals that SREBP2 bound to the promoter regions of two nonclassical sterol responsive genes involved in immune modulation, BHLHE40 and KLF6. SREBP2 upregulation of KLF6 was responsible for the downstream amplification of chemokine expression, highlighting a novel relationship between cholesterol homeostasis and inflammatory phenotypes in ECs.
Collapse
Affiliation(s)
- Joseph Wayne M Fowler
- Department of Pharmacology, Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Nabil E Boutagy
- Department of Pharmacology, Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Rong Zhang
- Department of Pharmacology, Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Daiki Horikami
- Department of Pharmacology, Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Michael B Whalen
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ, USA
| | - Casey E Romanoski
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ, USA
| | - William C Sessa
- Department of Pharmacology, Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
47
|
Pérez-Cabello JA, Silvera-Carrasco L, Franco JM, Capilla-González V, Armaos A, Gómez-Lima M, García-García R, Yap XW, Leal-Lasarte M, Lall D, Baloh RH, Martínez S, Miyata Y, Tartaglia GG, Sawarkar R, García-Domínguez M, Pozo D, Roodveldt C. MAPK/MAK/MRK overlapping kinase (MOK) controls microglial inflammatory/type-I IFN responses via Brd4 and is involved in ALS. Proc Natl Acad Sci U S A 2023; 120:e2302143120. [PMID: 37399380 PMCID: PMC10334760 DOI: 10.1073/pnas.2302143120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 05/26/2023] [Indexed: 07/05/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal and incurable neurodegenerative disease affecting motor neurons and characterized by microglia-mediated neurotoxic inflammation whose underlying mechanisms remain incompletely understood. In this work, we reveal that MAPK/MAK/MRK overlapping kinase (MOK), with an unknown physiological substrate, displays an immune function by controlling inflammatory and type-I interferon (IFN) responses in microglia which are detrimental to primary motor neurons. Moreover, we uncover the epigenetic reader bromodomain-containing protein 4 (Brd4) as an effector protein regulated by MOK, by promoting Ser492-phospho-Brd4 levels. We further demonstrate that MOK regulates Brd4 functions by supporting its binding to cytokine gene promoters, therefore enabling innate immune responses. Remarkably, we show that MOK levels are increased in the ALS spinal cord, particularly in microglial cells, and that administration of a chemical MOK inhibitor to ALS model mice can modulate Ser492-phospho-Brd4 levels, suppress microglial activation, and modify the disease course, indicating a pathophysiological role of MOK kinase in ALS and neuroinflammation.
Collapse
Affiliation(s)
- Jesús A. Pérez-Cabello
- Centro Andaluz de Biología Molecular y Medicina Regenerativa, Universidad de Sevilla-Consejo Superior de Investigaciones Científicas, Seville41092, Spain
- Department of Medical Biochemistry, Molecular Biology and Immunology, Faculty of Medicine, University of Seville, Seville41009, Spain
| | - Lucía Silvera-Carrasco
- Centro Andaluz de Biología Molecular y Medicina Regenerativa, Universidad de Sevilla-Consejo Superior de Investigaciones Científicas, Seville41092, Spain
- Department of Medical Biochemistry, Molecular Biology and Immunology, Faculty of Medicine, University of Seville, Seville41009, Spain
| | - Jaime M. Franco
- Centro Andaluz de Biología Molecular y Medicina Regenerativa, Universidad de Sevilla-Consejo Superior de Investigaciones Científicas, Seville41092, Spain
| | - Vivian Capilla-González
- Centro Andaluz de Biología Molecular y Medicina Regenerativa, Universidad de Sevilla-Consejo Superior de Investigaciones Científicas, Seville41092, Spain
| | - Alexandros Armaos
- Center for Human Technologies, Istituto Italiano di Tecnologia, Genova16152, Italy
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Genova16152, Italy
| | - María Gómez-Lima
- Centro Andaluz de Biología Molecular y Medicina Regenerativa, Universidad de Sevilla-Consejo Superior de Investigaciones Científicas, Seville41092, Spain
| | - Raquel García-García
- Centro Andaluz de Biología Molecular y Medicina Regenerativa, Universidad de Sevilla-Consejo Superior de Investigaciones Científicas, Seville41092, Spain
- Department of Medical Biochemistry, Molecular Biology and Immunology, Faculty of Medicine, University of Seville, Seville41009, Spain
| | - Xin Wen Yap
- The Medical Research Council Toxicology Unit, University of Cambridge, CambridgeCB1 2QR, United Kingdom
| | - Magdalena Leal-Lasarte
- Centro Andaluz de Biología Molecular y Medicina Regenerativa, Universidad de Sevilla-Consejo Superior de Investigaciones Científicas, Seville41092, Spain
| | - Deepti Lall
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA90048
| | - Robert H. Baloh
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA90048
| | - Salvador Martínez
- Instituto de Neurociencias, Universidad Miguel Hernández de Elche-CSIC, Alicante03550, Spain
| | - Yoshihiko Miyata
- Department of Cell and Developmental Biology, Graduate School of Biostudies, Kyoto University, Kyoto606-8501, Japan
| | - Gian G. Tartaglia
- Center for Human Technologies, Istituto Italiano di Tecnologia, Genova16152, Italy
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Genova16152, Italy
- Department of Biology and Biotechnologies, University Sapienza Rome, Rome00185, Italy
| | - Ritwick Sawarkar
- The Medical Research Council Toxicology Unit, University of Cambridge, CambridgeCB1 2QR, United Kingdom
| | - Mario García-Domínguez
- Centro Andaluz de Biología Molecular y Medicina Regenerativa, Universidad de Sevilla-Consejo Superior de Investigaciones Científicas, Seville41092, Spain
| | - David Pozo
- Centro Andaluz de Biología Molecular y Medicina Regenerativa, Universidad de Sevilla-Consejo Superior de Investigaciones Científicas, Seville41092, Spain
- Department of Medical Biochemistry, Molecular Biology and Immunology, Faculty of Medicine, University of Seville, Seville41009, Spain
| | - Cintia Roodveldt
- Centro Andaluz de Biología Molecular y Medicina Regenerativa, Universidad de Sevilla-Consejo Superior de Investigaciones Científicas, Seville41092, Spain
- Department of Medical Biochemistry, Molecular Biology and Immunology, Faculty of Medicine, University of Seville, Seville41009, Spain
| |
Collapse
|
48
|
Lagumdzic E, Pernold CPS, Ertl R, Palmieri N, Stadler M, Sawyer S, Stas MR, Kreutzmann H, Rümenapf T, Ladinig A, Saalmüller A. Gene expression of peripheral blood mononuclear cells and CD8 + T cells from gilts after PRRSV infection. Front Immunol 2023; 14:1159970. [PMID: 37409113 PMCID: PMC10318438 DOI: 10.3389/fimmu.2023.1159970] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 06/05/2023] [Indexed: 07/07/2023] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is a positive-stranded RNA virus, which emerged in Europe and U.S.A. in the late 1980s and has since caused huge economic losses. Infection with PRRSV causes mild to severe respiratory and reproductive clinical symptoms in pigs. Alteration of the host immune response by PRRSV is associated with the increased susceptibility to secondary viral and bacterial infections resulting in more serious and chronic disease. However, the expression profiles underlying innate and adaptive immune responses to PRRSV infection are yet to be further elucidated. In this study, we investigated gene expression profiles of PBMCs and CD8+ T cells after PRRSV AUT15-33 infection. We identified the highest number of differentially expressed genes in PBMCs and CD8+ T cells at 7 dpi and 21 dpi, respectively. The gene expression profile of PBMCs from infected animals was dominated by a strong innate immune response at 7 dpi which persisted through 14 dpi and 21 dpi and was accompanied by involvement of adaptive immunity. The gene expression pattern of CD8+ T cells showed a strong adaptive immune response to PRRSV, leading to the formation of highly differentiated CD8+ T cells starting from 14 dpi. The hallmark of the CD8+ T-cell response was the increased expression of effector and cytolytic genes (PRF1, GZMA, GZMB, GZMK, KLRK1, KLRD1, FASL, NKG7), with the highest levels observed at 21 dpi. Temporal clustering analysis of DEGs of PBMCs and CD8+ T cells from PRRSV-infected animals revealed three and four clusters, respectively, suggesting tight transcriptional regulation of both the innate and the adaptive immune response to PRRSV. The main cluster of PBMCs was related to the innate immune response to PRRSV, while the main clusters of CD8+ T cells represented the initial transformation and differentiation of these cells in response to the PRRSV infection. Together, we provided extensive transcriptomics data explaining gene signatures of the immune response of PBMCs and CD8+ T cells after PRRSV infection. Additionally, our study provides potential biomarker targets useful for vaccine and therapeutics development.
Collapse
Affiliation(s)
- Emil Lagumdzic
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Clara P. S. Pernold
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Reinhard Ertl
- VetCore Facility for Research, University of Veterinary Medicine, Vienna, Austria
| | - Nicola Palmieri
- University Clinic for Poultry and Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Maria Stadler
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Spencer Sawyer
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Melissa R. Stas
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Heinrich Kreutzmann
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Till Rümenapf
- Institute of Virology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Andrea Ladinig
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Armin Saalmüller
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
49
|
Chen Z, Javed N, Moore M, Wu J, Sun G, Vinyard M, Collins A, Pinello L, Najm FJ, Bernstein BE. Integrative dissection of gene regulatory elements at base resolution. CELL GENOMICS 2023; 3:100318. [PMID: 37388913 PMCID: PMC10300548 DOI: 10.1016/j.xgen.2023.100318] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 02/21/2023] [Accepted: 03/31/2023] [Indexed: 07/01/2023]
Abstract
Although vast numbers of putative gene regulatory elements have been cataloged, the sequence motifs and individual bases that underlie their functions remain largely unknown. Here, we combine epigenetic perturbations, base editing, and deep learning to dissect regulatory sequences within the exemplar immune locus encoding CD69. We converge on a ∼170 base interval within a differentially accessible and acetylated enhancer critical for CD69 induction in stimulated Jurkat T cells. Individual C-to-T base edits within the interval markedly reduce element accessibility and acetylation, with corresponding reduction of CD69 expression. The most potent base edits may be explained by their effect on regulatory interactions between the transcriptional activators GATA3 and TAL1 and the repressor BHLHE40. Systematic analysis suggests that the interplay between GATA3 and BHLHE40 plays a general role in rapid T cell transcriptional responses. Our study provides a framework for parsing regulatory elements in their endogenous chromatin contexts and identifying operative artificial variants.
Collapse
Affiliation(s)
- Zeyu Chen
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Gene Regulation Observatory, Broad Institute, Cambridge, MA, USA
- Department of Cell Biology and Pathology, Harvard Medical School, Boston, MA, USA
| | - Nauman Javed
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Gene Regulation Observatory, Broad Institute, Cambridge, MA, USA
- Department of Cell Biology and Pathology, Harvard Medical School, Boston, MA, USA
| | - Molly Moore
- Gene Regulation Observatory, Broad Institute, Cambridge, MA, USA
| | - Jingyi Wu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Gene Regulation Observatory, Broad Institute, Cambridge, MA, USA
- Department of Cell Biology and Pathology, Harvard Medical School, Boston, MA, USA
| | - Gary Sun
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology and Pathology, Harvard Medical School, Boston, MA, USA
| | - Michael Vinyard
- Gene Regulation Observatory, Broad Institute, Cambridge, MA, USA
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | | | - Luca Pinello
- Gene Regulation Observatory, Broad Institute, Cambridge, MA, USA
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Fadi J. Najm
- Gene Regulation Observatory, Broad Institute, Cambridge, MA, USA
| | - Bradley E. Bernstein
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Gene Regulation Observatory, Broad Institute, Cambridge, MA, USA
- Department of Cell Biology and Pathology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
50
|
Liu C, Du J, Zheng J, Zhang R, Zhu J, Xing B, Dong L, Zhou Q, Yao X, Gao S, Wang Y, Ren Y, Zhou X. The role of BHLHE40 in clinical features and prognosis value of PDAC by comprehensive analysis and in vitro validation. Front Oncol 2023; 13:1151321. [PMID: 37377917 PMCID: PMC10291124 DOI: 10.3389/fonc.2023.1151321] [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: 01/28/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the leading cause of cancer-related mortality, primarily due to the abundance of cancer-associated fibroblasts (CAFs), depleted effector T cells, and increased tumor cell stemness; hence, there is an urgent need for efficient biomarkers with prognostic and therapeutic potential. Here, we identified BHLHE40 as a promising target for PDAC through comprehensive analysis and weighted gene coexpression network analysis of RNA sequencing data and public databases, taking into account the unique characteristics of PDAC such as cancer-associated fibroblasts, infiltration of effector T cells, and tumor cell stemness. Additionally, we developed a prognostic risk model based on BHLHE40 and three other candidate genes (ITGA2, ITGA3, and ADAM9) to predict outcomes in PDAC patients. Furthermore, we found that the overexpression of BHLHE40 was significantly associated with T stage, lymph node metastasis, and American Joint Committee on Cancer (AJCC) stage in a cohort of 61 PDAC patients. Moreover, elevated expression levels of BHLHE40 were validated to promote epithelial-mesenchymal transition (EMT) and stemness-related proteins in BXPC3 cell lines. Compared to the parent cells, BXPC3 cells with BHLHE40 overexpression showed resistance to anti-tumor immunity when co-cultured with CD8+ T cells. In summary, these findings suggest that BHLHE40 is a highly effective biomarker for predicting prognosis in PDAC and holds great promise as a target for cancer therapy.
Collapse
Affiliation(s)
- Chao Liu
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer; Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Jiang Du
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer; Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Jianwei Zheng
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer; Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Ruizhe Zhang
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer; Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Jialin Zhu
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Bofan Xing
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer; Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Lin Dong
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer; Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Qianqian Zhou
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer; Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Xiaofeng Yao
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer; Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Song Gao
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Yu Wang
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer; Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Yu Ren
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xuan Zhou
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer; Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|