1
|
Zhang Y, Wang TW, Tamatani M, Zeng X, Nakamura L, Omori S, Yamaguchi K, Hatakeyama S, Shimizu E, Yamazaki S, Furukawa Y, Imoto S, Johmura Y, Nakanishi M. Signaling networks in cancer stromal senescent cells establish malignant microenvironment. Proc Natl Acad Sci U S A 2025; 122:e2412818122. [PMID: 40168129 PMCID: PMC12002233 DOI: 10.1073/pnas.2412818122] [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: 06/26/2024] [Accepted: 02/27/2025] [Indexed: 04/03/2025] Open
Abstract
The tumor microenvironment (TME) encompasses various cell types, blood and lymphatic vessels, and noncellular constituents like extracellular matrix (ECM) and cytokines. These intricate interactions between cellular and noncellular components contribute to the development of a malignant TME, such as immunosuppressive, desmoplastic, angiogenic conditions, and the formation of a niche for cancer stem cells, but there is limited understanding of the specific subtypes of stromal cells involved in this process. Here, we utilized p16-CreERT2-tdTomato mouse models to investigate the signaling networks established by senescent cancer stromal cells, contributing to the development of a malignant TME. In pancreatic ductal adenocarcinoma (PDAC) allograft models, these senescent cells were found to promote cancer fibrosis, enhance angiogenesis, and suppress cancer immune surveillance. Notably, the selective elimination of senescent cancer stromal cells improves the malignant TME, subsequently reducing tumor progression in PDAC. This highlights the antitumor efficacy of senolytic treatment alone and its synergistic effect when combined with conventional chemotherapy. Taken together, our findings suggest that the signaling crosstalk among senescent cancer stromal cells plays a key role in the progression of PDAC and may be a promising therapeutic target.
Collapse
Affiliation(s)
- Yue Zhang
- Division of Cancer Cell Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo108-8639, Japan
| | - Teh-Wei Wang
- Division of Cancer Cell Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo108-8639, Japan
- Project Division of Generative AI Utilization Aging Cells, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo108-8639, Japan
| | - Maho Tamatani
- Division of Cancer Cell Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo108-8639, Japan
| | - Xinyi Zeng
- Division of Cancer Cell Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo108-8639, Japan
| | - Lindo Nakamura
- Division of Cancer Cell Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo108-8639, Japan
| | - Satotaka Omori
- Division of Cancer Cell Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo108-8639, Japan
| | - Kiyoshi Yamaguchi
- Division of Clinical Genome Research, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo108-8639, Japan
| | - Seira Hatakeyama
- Division of Clinical Genome Research, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo108-8639, Japan
| | - Eigo Shimizu
- Division of Health Medical Intelligence, Human Genome Center, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo108-8639, Japan
| | - Satoshi Yamazaki
- Division of Cell Regulation, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo108-8639, Japan
| | - Yoichi Furukawa
- Division of Clinical Genome Research, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo108-8639, Japan
| | - Seiya Imoto
- Division of Health Medical Intelligence, Human Genome Center, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo108-8639, Japan
| | - Yoshikazu Johmura
- Division of Cancer and Senescence Biology, Cancer Research Institute, Kanazawa University, Kanazawa920-1192, Japan
| | - Makoto Nakanishi
- Division of Cancer Cell Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo108-8639, Japan
| |
Collapse
|
2
|
Karjalainen A, Witalisz-Siepracka A, Prchal-Murphy M, Martin D, Sternberg F, Krunic M, Dolezal M, Fortelny N, Farlik M, Macho-Maschler S, Lassnig C, Meissl K, Amenitsch L, Lederer T, Pohl E, Gotthardt D, Bock C, Decker T, Strobl B, Müller M. Cell-type-specific requirement for TYK2 in murine immune cells under steady state and challenged conditions. Cell Mol Life Sci 2025; 82:98. [PMID: 40025196 PMCID: PMC11872851 DOI: 10.1007/s00018-025-05625-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/31/2025] [Accepted: 02/17/2025] [Indexed: 03/04/2025]
Abstract
Tyrosine kinase 2 (TYK2) deficiency and loss or inhibition of kinase activity in men and mice leads to similar immune compromised phenotypes, predominantly through impairment of interferon (IFN) and interleukin 12 family responses. Here we relate the transcriptome changes to phenotypical changes observed in TYK2-deficient (Tyk2-/-) and TYK2 kinase-inactive (Tyk2K923E) mice in naïve splenic immune cells and upon ex vivo IFN treatment or in vivo tumor transplant infiltration. The TYK2 activities under homeostatic and both challenged conditions are highly cell-type-specific with respect to quantity and quality of transcriptionally dependent genes. The major impact of loss of TYK2 protein or kinase activity in splenic homeostatic macrophages, NK and CD8+ T cells and tumor-derived cytolytic cells is on IFN responses. While reportedly TYK2 deficiency leads to partial impairment of IFN-I responses, we identified cell-type-specific IFN-I-repressed gene sets completely dependent on TYK2 kinase activity. Reported kinase-inactive functions of TYK2 relate to signaling crosstalk, metabolic functions and cell differentiation or maturation. None of these phenotypes relates to respective enriched gene sets in the TYK2 kinase-inactive cell types. Nonetheless, the scaffolding functions of TYK2 are capable to change transcriptional activities at single gene levels and chromatin accessibility at promoter-distal regions upon cytokine treatment most prominently in CD8+ T cells. The cell-type-specific transcriptomic and epigenetic effects of TYK2 shed new light on the biology of this JAK family member and are relevant for current and future treatment of autoimmune and inflammatory diseases with TYK2 inhibitors.
Collapse
Affiliation(s)
- Anzhelika Karjalainen
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Agnieszka Witalisz-Siepracka
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
- Division Pharmacology, Karl Landsteiner University of Health Sciences, Krems an Der Donau, Austria
| | - Michaela Prchal-Murphy
- Pharmacology and Toxicology, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - David Martin
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Felix Sternberg
- Physiology and Biophysics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
- Department of Nutritional Sciences, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Milica Krunic
- Campus Tulln, University of Applied Sciences Wiener Neustadt, Wiener Neustadt, Austria
| | - Marlies Dolezal
- Platform Biostatistics and Bioinformatics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Nikolaus Fortelny
- Department of Biosciences and Medical Biology, Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, Austria
| | - Matthias Farlik
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Sabine Macho-Maschler
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Caroline Lassnig
- Core Facility VetBiomodels, University of Veterinary Medicine, Vienna, Austria
| | - Katrin Meissl
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Lena Amenitsch
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Therese Lederer
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Elena Pohl
- Physiology and Biophysics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Dagmar Gotthardt
- Division Pharmacology, Karl Landsteiner University of Health Sciences, Krems an Der Donau, Austria
| | - Christoph Bock
- Cemm Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Institute of Artificial Intelligence, Center for Medical Data Science, Medical University of Vienna, Vienna, Austria
| | - Thomas Decker
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Center for Molecular Biology, Department of Microbiology, Immunobiology and Genetics, University of Vienna, Vienna, Austria
| | - Birgit Strobl
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Mathias Müller
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria.
| |
Collapse
|
3
|
Senthil Kumar J, Mehboob MZ, Lei X. Exploring CTRP6: a biomarker and therapeutic target in metabolic diseases. Am J Physiol Endocrinol Metab 2025; 328:E139-E147. [PMID: 39701154 DOI: 10.1152/ajpendo.00353.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/11/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024]
Abstract
The rising prevalence of metabolic diseases is a significant global health concern. Beyond lifestyle management, targeting key molecules involved in metabolic regulation is essential. C1q/TNF-related protein 6 (CTRP6) is notably associated with glucose and lipid metabolism, with numerous studies highlighting its regulatory functions in metabolic diseases. This review summarizes the current knowledge on CTRP6, focusing on its gene expression profiles, protein structure, gene regulation, and role in metabolic diseases. CTRP6 is widely expressed across various tissues and features four distinct domains, with the C1q domain predicted to bind to its receptor. Notably, serum levels of CTRP6 are significantly elevated in patients with obesity and type 2 diabetes. In these conditions, adipose tissue serves as a key source of CTRP6 and its involvement in adipose tissue expansion, inflammation, and nutrient sensing has been observed in several studies. CTRP6 is also implicated in type 1 diabetes, gestational diabetes mellitus, and diabetic complications, particularly diabetic nephropathy. Although some studies have suggested that CTRP6 has protective roles in atherosclerotic cell models, myocardial infarction rat models, and ischemia/reperfusion injury mouse models, methodological issues such as unreliable antibodies and unstrict controls make it difficult to draw accurate conclusions from these studies. Patients with polycystic ovary syndrome (PCOS) exhibit elevated serum levels of CTRP6, although its direct impact on PCOS phenotypes remains unclear. In conclusion, CTRP6 emerges as a promising therapeutic target for metabolic diseases. A deeper understanding of CTRP6 will empower the scientific community to develop effective interventions to address the increasing prevalence of these diseases.
Collapse
Affiliation(s)
- Jeevotham Senthil Kumar
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, Oklahoma, United States
| | - Muhammad Zubair Mehboob
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, Oklahoma, United States
| | - Xia Lei
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, Oklahoma, United States
| |
Collapse
|
4
|
Duran P, Zelus EI, Burnett LA, Christman KL, Alperin M. Repeated birth injuries lead to long-term pelvic floor muscle dysfunction in the preclinical rat model. Am J Obstet Gynecol 2025; 232:198.e1-198.e23. [PMID: 39191364 PMCID: PMC11757040 DOI: 10.1016/j.ajog.2024.08.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 08/13/2024] [Accepted: 08/20/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND Vaginal childbirth is a key risk factor for pelvic floor muscle injury and dysfunction, and subsequent pelvic floor disorders. Multiparity further exacerbates these risks. Using the rat model, validated for the studies of the human pelvic floor muscles, we have previously identified that a single simulated birth injury results in pelvic floor muscle atrophy and fibrosis. OBJECTIVE To test the hypothesis that multiple birth injuries would further overwhelm the muscle regenerative capacity, leading to functionally relevant pathological alterations long-term. STUDY DESIGN Sprague-Dawley rats underwent simulated birth injury and were allowed to recover for 8 weeks before undergoing additional birth injury. Animals were sacrificed at acute (3 and 7 days postinjury), subacute (21, 28, and 35 days postinjury), and long-term (8 and 12 weeks postinjury) time points post second injury (N=3-8/time point), and the pubocaudalis portion of the rat levator ani complex was harvested to assess the impact of repeated birth injuries on muscle mechanical and histomorphological properties. The accompanying transcriptional changes were assessed by a customized NanoString panel. Uninjured animals were used as controls. Data with a parametric distribution were analyzed by a 2-way analysis of variance followed by post hoc pairwise comparisons using Tukey's or Sidak's tests; nonparametrically distributed data were compared with Kruskal-Wallis test followed by pairwise comparisons with Dunn's test. Data, analyzed using GraphPad Prism v8.0, San Diego, CA, are presented as mean ± standard error of the mean or median (range). RESULTS Following the first simulated birth injury, active muscle force decreased acutely relative to uninjured controls (12.9±0.9 vs 25.98±2.1 g/mm2, P<.01). At 4 weeks, muscle active force production recovered to baseline and remained unchanged at 8 weeks after birth injury (P>.99). Similarly, precipitous decrease in active force was observed immediately after repeated birth injury (18.07±1.2 vs 25.98±2.1 g/mm2, P<.05). In contrast to the functional recovery after a single birth injury, a long-term decrease in muscle contractile function was observed up to 12 weeks after repeated birth injuries (18.3±1.6 vs 25.98±2.1 g/mm2, P<.05). Fiber size was smaller at the long-term time points after second injury compared to the uninjured group (12 weeks vs uninjured control: 1485 (60.7-5000) vs 1989 (65.6-4702) μm2, P<.0001). The proportion of fibers with centralized nuclei, indicating active myofiber regeneration, returned to baseline at 8 weeks post-first birth injury, (P=.95), but remained elevated as far as 12 weeks post-second injury (12 weeks vs uninjured control: 7.1±1.5 vs 0.84±0.13%, P<0.0001). In contrast to the plateauing intramuscular collagen content after 4 weeks post-first injury, fibrotic degeneration increased progressively over 12 weeks after repeated injury (12 weeks vs uninjured control: 6. 7±1.1 vs 2.03±0.2%, P<.001). Prolonged expression of proinflammatory genes accompanied by a greater immune infiltrate was observed after repeated compared to a single birth injury. CONCLUSION Overall, repeated birth injuries lead to a greater magnitude of pathological alterations compared to a single injury, resulting in more pronounced pelvic floor muscle degeneration and muscle dysfunction in the rat model. The above provides a putative mechanistic link between multiparity and the increased risk of pelvic floor dysfunction in women.
Collapse
Affiliation(s)
- Pamela Duran
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA
| | - Emma I Zelus
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA
| | - Lindsey A Burnett
- Sanford Stem Cell Institute, La Jolla, CA; Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Urogynecology and Reconstructive Pelvic Surgery, University of California San Diego, La Jolla, CA
| | - Karen L Christman
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA; Sanford Consortium for Regenerative Medicine, La Jolla, CA; Sanford Stem Cell Institute, La Jolla, CA.
| | - Marianna Alperin
- Sanford Consortium for Regenerative Medicine, La Jolla, CA; Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Urogynecology and Reconstructive Pelvic Surgery, University of California San Diego, La Jolla, CA.
| |
Collapse
|
5
|
Antonczyk A, Kluzek K, Herbich N, Boroujeni ME, Krist B, Wronka D, Karlik A, Przybyl L, Plewinski A, Wesoly J, Bluyssen HAR. Identification of ALEKSIN as a novel multi-IRF inhibitor of IRF- and STAT-mediated transcription in vascular inflammation and atherosclerosis. Front Pharmacol 2025; 15:1471182. [PMID: 39840103 PMCID: PMC11747033 DOI: 10.3389/fphar.2024.1471182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 12/03/2024] [Indexed: 01/23/2025] Open
Abstract
Cardiovascular diseases (CVDs) include atherosclerosis, which is an inflammatory disease of large and medium vessels that leads to atherosclerotic plaque formation. The key factors contributing to the onset and progression of atherosclerosis include the pro-inflammatory cytokines interferon (IFN)α and IFNγ and the pattern recognition receptor (PRR) Toll-like receptor 4 (TLR4). Together, they trigger the activation of IFN regulatory factors (IRFs) and signal transducer and activator of transcription (STAT)s. Based on their promoting role in atherosclerosis, we hypothesized that the inhibition of pro-inflammatory target gene expression through multi-IRF inhibitors may be a promising strategy to treat CVDs. Using comparative in silico docking of multiple IRF-DNA-binding domain (DBD) models on a multi-million natural compound library, we identified the novel multi-IRF inhibitor, ALEKSIN. This compound targets the DBD of IRF1, IRF2, and IRF8 with the same affinity and simultaneously inhibits the expression of multiple IRF target genes in human microvascular endothelial cells (HMECs) in response to IIFNα and IFNγ. Under the same conditions, ALEKSIN also inhibited the phosphorylation of STATs, potentially through low-affinity STAT-SH2 binding but with lower potency than the known multi-STAT inhibitor STATTIC. This was in line with the common inhibition of ALEKSIN and STATTIC observed on the genome-wide expression of pro-inflammatory IRF/STAT/NF-κB target genes, as well as on the migration of HMECs. Finally, we identified a novel signature of 46 ALEKSIN and STATTIC commonly inhibited pro-atherogenic target genes, which was upregulated in atherosclerotic plaques in the aortas of high-fat diet-fed ApoEKO mice and associated with inflammation, proliferation, adhesion, chemotaxis, and response to lipids. Interestingly, the majority of these genes could be linked to macrophage subtypes present in aortic plaques in HFD-fed LDLR-KO mice. Together, this suggests that ALEKSIN represents a novel class of multi-IRF inhibitors, which inhibits IRF-, STAT-, and NF-κB-mediated transcription and could offer great promise for the treatment of CVDs. Furthermore, the ALEKSIN and STATTIC commonly inhibited pro-inflammatory gene signature could help monitor plaque progression during experimental atherosclerosis.
Collapse
Affiliation(s)
- Aleksandra Antonczyk
- Human Molecular Genetics Research Unit, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznan, Poland
| | - Katarzyna Kluzek
- Human Molecular Genetics Research Unit, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznan, Poland
| | - Natalia Herbich
- Human Molecular Genetics Research Unit, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznan, Poland
| | - Mahdi Eskandarian Boroujeni
- Human Molecular Genetics Research Unit, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznan, Poland
| | - Bart Krist
- Human Molecular Genetics Research Unit, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznan, Poland
| | - Dorota Wronka
- Laboratory of Mammalian Model Organisms, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Anna Karlik
- Laboratory of Mammalian Model Organisms, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Lukasz Przybyl
- Laboratory of Mammalian Model Organisms, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Adam Plewinski
- Animal Facility, Center for Advanced Technologies, Adam Mickiewicz University, Poznan, Poland
| | - Joanna Wesoly
- Laboratory of High Throughput Technologies, Faculty of Biology, Adam Mickiewicz University, Poznan, Poland
| | - Hans A. R. Bluyssen
- Human Molecular Genetics Research Unit, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznan, Poland
| |
Collapse
|
6
|
Qi Y, Li R, Han M. Tumor-associated macrophages induce epithelial-mesenchymal transition and promote lung metastasis in breast cancer by activating the IL-6/STAT3/TGM2 axis. Int Immunopharmacol 2024; 143:113387. [PMID: 39426226 DOI: 10.1016/j.intimp.2024.113387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/28/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024]
Abstract
Breast cancer is one of the most common tumors in the world and metastasis is the major cause of tumor-related death. Tumor-associated macrophages (TAMs) are a major component of the tumor microenvironment (TME) and often associated with cancer metastasis. Nevertheless, the mechanism by which TAMs regulate breast cancer metastasis remain unclear. In this study, we found that transglutaminase 2 (TGM2) could serve as a crucial target in the modulation of TAMs-induced epithelial-mesenchymal transition (EMT) and invasion of breast cancer cells. Further analysis revealed that IL-6 secreted from TAMs, which was capable of inducing TGM2 expression through the activation of the JAK/STAT3 signaling pathway. Subsequent luciferase reporter assays demonstrated that STAT3 binds to the TGM2 promoter region, thereby transcriptionally enhancing TGM2 expression. In conclusion, our current research has identified the IL-6/STAT3/TGM2 axis as a pivotal regulator in breast tumorigenesis caused by TAMs, presenting a novel target for the treatment of breast cancer.
Collapse
Affiliation(s)
- Yana Qi
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong Province, China
| | - Ranran Li
- Cancer Therapy and Research Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| | - Mingyong Han
- Cancer therapy and Research Center, Shandong Provincal Hospital, Cheeloo College of Medicine, Shandong University, 324 Jingwuweiqi Road, Jinan, 250021, Shandong, China.
| |
Collapse
|
7
|
Jin H, Kim W, Yuan M, Li X, Yang H, Li M, Shi M, Turkez H, Uhlen M, Zhang C, Mardinoglu A. Identification of SPP1 + macrophages as an immune suppressor in hepatocellular carcinoma using single-cell and bulk transcriptomics. Front Immunol 2024; 15:1446453. [PMID: 39691723 PMCID: PMC11649653 DOI: 10.3389/fimmu.2024.1446453] [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: 06/09/2024] [Accepted: 11/19/2024] [Indexed: 12/19/2024] Open
Abstract
Introduction Macrophages and T cells play crucial roles in liver physiology, but their functional diversity in hepatocellular carcinoma (HCC) remains largely unknown. Methods Two bulk RNA-sequencing (RNA-seq) cohorts for HCC were analyzed using gene co-expression network analysis. Key gene modules and networks were mapped to single-cell RNA-sequencing (scRNA-seq) data of HCC. Cell type fraction of bulk RNA-seq data was estimated by deconvolution approach using single-cell RNA-sequencing data as a reference. Survival analysis was carried out to estimate the prognosis of different immune cell types in bulk RNA-seq cohorts. Cell-cell interaction analysis was performed to identify potential links between immune cell types in HCC. Results In this study, we analyzed RNA-seq data from two large-scale HCC cohorts, revealing a major and consensus gene co-expression cluster with significant implications for immunosuppression. Notably, these genes exhibited higher enrichment in liver macrophages than T cells, as confirmed by scRNA-seq data from HCC patients. Integrative analysis of bulk and single-cell RNA-seq data pinpointed SPP1 + macrophages as an unfavorable cell type, while VCAN + macrophages, C1QA + macrophages, and CD8 + T cells were associated with a more favorable prognosis for HCC patients. Subsequent scRNA-seq investigations and in vitro experiments elucidated that SPP1, predominantly secreted by SPP1 + macrophages, inhibits CD8 + T cell proliferation. Finally, targeting SPP1 in tumor-associated macrophages through inhibition led to a shift towards a favorable phenotype. Discussion This study underpins the potential of SPP1 as a translational target in immunotherapy for HCC.
Collapse
Affiliation(s)
- Han Jin
- Central Laboratory, Tianjin Medical University General Hospital, Tianjin, China
- Science for Life Laboratory, KTH – Royal Institute of Technology, Stockholm, Sweden
| | - Woonghee Kim
- Science for Life Laboratory, KTH – Royal Institute of Technology, Stockholm, Sweden
| | - Meng Yuan
- Science for Life Laboratory, KTH – Royal Institute of Technology, Stockholm, Sweden
| | - Xiangyu Li
- Science for Life Laboratory, KTH – Royal Institute of Technology, Stockholm, Sweden
| | - Hong Yang
- Science for Life Laboratory, KTH – Royal Institute of Technology, Stockholm, Sweden
| | - Mengzhen Li
- Science for Life Laboratory, KTH – Royal Institute of Technology, Stockholm, Sweden
| | - Mengnan Shi
- Science for Life Laboratory, KTH – Royal Institute of Technology, Stockholm, Sweden
| | - Hasan Turkez
- Department of Medical Biology, Faculty of Medicine, Atatürk University, Erzurum, Türkiye
| | - Mathias Uhlen
- Science for Life Laboratory, KTH – Royal Institute of Technology, Stockholm, Sweden
| | - Cheng Zhang
- Science for Life Laboratory, KTH – Royal Institute of Technology, Stockholm, Sweden
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH – Royal Institute of Technology, Stockholm, Sweden
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, United Kingdom
| |
Collapse
|
8
|
Torres G, Salladay-Perez IA, Dhingra A, Covarrubias AJ. Genetic origins, regulators, and biomarkers of cellular senescence. Trends Genet 2024; 40:1018-1031. [PMID: 39341687 PMCID: PMC11717094 DOI: 10.1016/j.tig.2024.08.007] [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: 05/02/2024] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 10/01/2024]
Abstract
This review comprehensively examines the molecular biology and genetic origins of cellular senescence. We focus on various cellular stressors and pathways leading to senescence, including recent advances in the understanding of the genetic influences driving senescence, such as telomere attrition, chemotherapy-induced DNA damage, pathogens, oncogene activation, and cellular and metabolic stress. This review also highlights the complex interplay of various signaling and metabolic pathways involved in cellular senescence and provides insights into potential therapeutic targets for aging-related diseases. Furthermore, this review outlines future research directions to deepen our understanding of senescence biology and develop effective interventions targeting senescent cells (SnCs).
Collapse
Affiliation(s)
- Grasiela Torres
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Interdepartmental Doctoral Program, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ivan A Salladay-Perez
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Interdepartmental Doctoral Program, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Anika Dhingra
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Anthony J Covarrubias
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
9
|
de Juan A, Tabtim-On D, Coillard A, Becher B, Goudot C, Segura E. The aryl hydrocarbon receptor shapes monocyte transcriptional responses to interleukin-4 by prolonging STAT6 binding to promoters. Sci Signal 2024; 17:eadn6324. [PMID: 39405377 DOI: 10.1126/scisignal.adn6324] [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: 12/19/2023] [Revised: 05/25/2024] [Accepted: 09/04/2024] [Indexed: 10/25/2024]
Abstract
Cytokines induce functional and metabolic adaptations in immune cells, typically through transcriptional responses that can be influenced by other extracellular signals and by intracellular factors. The binding of the cytokine interleukin-4 (IL-4) to its receptor induces the phosphorylation and activation of the transcription factor STAT6. The aryl hydrocarbon receptor (AhR), a transcription factor activated by various endogenous and microbe-derived metabolites, modulates the responses of immune cells to danger signals or inflammatory mediators such as cytokines. Here, we investigated cross-talk between the AhR and signaling stimulated by IL-4 in human and mouse monocytes. AhR activation was required for a subset of IL-4-induced transcriptional responses and inhibited the IL-4-induced metabolic switch to fatty acid β-oxidation. The promoters of the genes that were induced by IL-4 in an AhR-dependent manner lacked canonical AhR binding sites, implying a nongenomic mechanism of AhR action. Mechanistically, AhR activation reduced the activity of SHP-1, a phosphatase that targets and inhibits STAT6, and prolonged STAT6 phosphorylation and binding to specific target loci, thus extending the duration of STAT6 activity. Our results identify AhR as a key player in the molecular control of responses to IL-4 in monocytes and suggest a nongenomic mechanism through which AhR ligands may influence the functional responses of cells to IL-4.
Collapse
Affiliation(s)
- Alba de Juan
- Institut Curie, PSL Research University, INSERM, U932, 26 rue d'Ulm, Paris, France
| | - Darawan Tabtim-On
- Institut Curie, PSL Research University, INSERM, U932, 26 rue d'Ulm, Paris, France
| | - Alice Coillard
- Institut Curie, PSL Research University, INSERM, U932, 26 rue d'Ulm, Paris, France
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Christel Goudot
- Institut Curie, PSL Research University, INSERM, U932, 26 rue d'Ulm, Paris, France
| | - Elodie Segura
- Institut Curie, PSL Research University, INSERM, U932, 26 rue d'Ulm, Paris, France
| |
Collapse
|
10
|
Qing F, Tian H, Wang B, Xie B, Sui L, Xie X, He W, He T, Li Y, He L, Guo Q, Liu Z. Interferon regulatory factor 7 alleviates the experimental colitis through enhancing IL-28A-mediated intestinal epithelial integrity. J Transl Med 2024; 22:905. [PMID: 39370517 PMCID: PMC11457333 DOI: 10.1186/s12967-024-05673-y] [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: 03/02/2024] [Accepted: 09/02/2024] [Indexed: 10/08/2024] Open
Abstract
BACKGROUND The incidence of inflammatory bowel disease (IBD) is on the rise in developing countries, and investigating the underlying mechanisms of IBD is essential for the development of targeted therapeutic interventions. Interferon regulatory factor 7 (IRF7) is known to exert pro-inflammatory effects in various autoimmune diseases, yet its precise role in the development of colitis remains unclear. METHODS We analyzed the clinical significance of IRF7 in ulcerative colitis (UC) by searching RNA-Seq databases and collecting tissue samples from clinical UC patients. And, we performed dextran sodium sulfate (DSS)-induced colitis modeling using WT and Irf7-/- mice to explore the mechanism of IRF7 action on colitis. RESULTS In this study, we found that IRF7 expression is significantly reduced in patients with UC, and also demonstrated that Irf7-/- mice display heightened susceptibility to DSS-induced colitis, accompanied by elevated levels of colonic and serum pro-inflammatory cytokines, suggesting that IRF7 is able to inhibit colitis. This increased susceptibility is linked to compromised intestinal barrier integrity and impaired expression of key molecules, including Muc2, E-cadherin, β-catenin, Occludin, and Interleukin-28A (IL-28A), a member of type III interferon (IFN-III), but independent of the deficiency of classic type I interferon (IFN-I) and type II interferon (IFN-II). The stimulation of intestinal epithelial cells by recombinant IL-28A augments the expression of Muc2, E-cadherin, β-catenin, and Occludin. The recombinant IL-28A protein in mice counteracts the heightened susceptibility of Irf7-/- mice to colitis induced by DSS, while also elevating the expression of Muc2, E-cadherin, β-catenin, and Occludin, thereby promoting the integrity of the intestinal barrier. CONCLUSION These findings underscore the pivotal role of IRF7 in preserving intestinal homeostasis and forestalling the onset of colitis.
Collapse
Affiliation(s)
- Furong Qing
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Hongbo Tian
- Department of Stomatology, Chifeng Maternity Hospital, Chifeng, Inner Mongolia, China
| | - Biyao Wang
- Department of Gastroenterology, The Sixth-Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth-Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Bingyu Xie
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Lina Sui
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xiaoyan Xie
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Wenji He
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Tiansheng He
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Yumei Li
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Liangmei He
- Department of Gastroenterology, The First-Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Qin Guo
- Department of Gastroenterology, The Sixth-Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.
- Biomedical Innovation Center, The Sixth-Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.
| | - Zhiping Liu
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, China.
- Center for Immunology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, China.
| |
Collapse
|
11
|
Thrikawala SU, Anderson MH, Rosowski EE. Glucocorticoids Suppress NF-κB-Mediated Neutrophil Control of Aspergillus fumigatus Hyphal Growth. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:971-987. [PMID: 39178124 PMCID: PMC11408098 DOI: 10.4049/jimmunol.2400021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 07/22/2024] [Indexed: 08/25/2024]
Abstract
Glucocorticoids are a major class of therapeutic anti-inflammatory and immunosuppressive drugs prescribed to patients with inflammatory diseases, to avoid transplant rejection, and as part of cancer chemotherapy. However, exposure to these drugs increases the risk of opportunistic infections such as with the fungus Aspergillus fumigatus, which causes mortality in >50% of infected patients. The mechanisms by which glucocorticoids increase susceptibility to A. fumigatus are poorly understood. In this article, we used a zebrafish larva Aspergillus infection model to identify innate immune mechanisms altered by glucocorticoid treatment. Infected larvae exposed to dexamethasone succumb to infection at a significantly higher rate than control larvae. However, both macrophages and neutrophils are still recruited to the site of infection, and dexamethasone treatment does not significantly affect fungal spore killing. Instead, the primary effect of dexamethasone manifests later in infection with treated larvae exhibiting increased invasive hyphal growth. In line with this, dexamethasone predominantly inhibits neutrophil function rather than macrophage function. Dexamethasone-induced mortality also depends on the glucocorticoid receptor. Dexamethasone partially suppresses NF-κB activation at the infection site by inducing the transcription of IκB via the glucocorticoid receptor. Independent CRISPR/Cas9 targeting of IKKγ to prevent NF-κB activation also increases invasive A. fumigatus growth and larval mortality. However, dexamethasone treatment of IKKγ crispant larvae further increases invasive hyphal growth and host mortality, suggesting that dexamethasone may suppress other pathways in addition to NF-κB to promote host susceptibility. Collectively, we find that dexamethasone acts through the glucocorticoid receptor to suppress NF-κB-mediated neutrophil control of A. fumigatus hyphae in zebrafish larvae.
Collapse
Affiliation(s)
- Savini U. Thrikawala
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
- Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, South Carolina, United States of America
| | - Molly H. Anderson
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
- Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, South Carolina, United States of America
| | - Emily E. Rosowski
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
- Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, South Carolina, United States of America
| |
Collapse
|
12
|
Babadei O, Strobl B, Müller M, Decker T. Transcriptional control of interferon-stimulated genes. J Biol Chem 2024; 300:107771. [PMID: 39276937 PMCID: PMC11489399 DOI: 10.1016/j.jbc.2024.107771] [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: 06/01/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/17/2024] Open
Abstract
Interferon-induced genes are among the best-studied groups of coregulated genes. Nevertheless, intense research into their regulation, supported by new technologies, is continuing to provide insights into their many layers of transcriptional regulation and to reveal how cellular transcriptomes change with pathogen-induced innate and adaptive immunity. This article gives an overview of recent findings on interferon-induced gene regulation, paying attention to contributions beyond the canonical JAK-STAT pathways.
Collapse
Affiliation(s)
- Olga Babadei
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria; University of Vienna, Center for Molecular Biology, Department of Microbiology, Immunobiology and Genetics, Vienna, Austria
| | - Birgit Strobl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Mathias Müller
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Thomas Decker
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria; University of Vienna, Center for Molecular Biology, Department of Microbiology, Immunobiology and Genetics, Vienna, Austria.
| |
Collapse
|
13
|
Kim J, Sullivan O, Lee K, Jao J, Tamayo J, Madany AM, Wong B, Ashwood P, Ciernia AV. Repeated LPS induces training and tolerance of microglial responses across brain regions. J Neuroinflammation 2024; 21:233. [PMID: 39304952 PMCID: PMC11414187 DOI: 10.1186/s12974-024-03198-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 08/08/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Neuroinflammation is involved in the pathogenesis of almost every central nervous system disorder. As the brain's innate immune cells, microglia fine tune their activity to a dynamic brain environment. Previous studies have shown that repeated bouts of peripheral inflammation can trigger long-term changes in microglial gene expression and function, a form of innate immune memory. METHODS AND RESULTS In this study, we used multiple low-dose lipopolysaccharide (LPS) injections in adult mice to study the acute cytokine, transcriptomic, and microglia morphological changes that contribute to the formation of immune memory in the frontal cortex, hippocampus, and striatum, as well as the long-term effects of these changes on behavior. Training and tolerance of gene expression was shared across regions, and we identified 3 unique clusters of DEGs (2xLPS-sensitive, 4xLPS-sensitive, LPS-decreased) enriched for different biological functions. 2xLPS-sensitive DEG promoters were enriched for binding sites for IRF and NFkB family transcription factors, two key regulators of innate immune memory. We quantified shifts in microglia morphological populations and found that while the proportion of ramified and rod-like microglia mostly remained consistent within brain regions and sexes with LPS treatment, there was a shift from ameboid towards hypertrophic morphological states across immune memory states and a dynamic emergence and resolution of events of microglia aligning end-to-end with repeated LPS. CONCLUSIONS Together, findings support the dynamic regulation of microglia during the formation of immune memories in the brain and support future work to exploit this model in brain disease contexts.
Collapse
Affiliation(s)
- Jennifer Kim
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada
| | - Olivia Sullivan
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada
| | - Kristen Lee
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada
| | - Justin Jao
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada
| | - Juan Tamayo
- MIND Institute, University of California Davis, Davis, USA
| | | | - Brandon Wong
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada
| | - Paul Ashwood
- MIND Institute, University of California Davis, Davis, USA
| | - Annie Vogel Ciernia
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada.
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada.
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada.
| |
Collapse
|
14
|
Kulakova K, Lawal TR, Mccarthy E, Floudas A. The Contribution of Macrophage Plasticity to Inflammatory Arthritis and Their Potential as Therapeutic Targets. Cells 2024; 13:1586. [PMID: 39329767 PMCID: PMC11430612 DOI: 10.3390/cells13181586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/06/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024] Open
Abstract
Inflammatory arthritis are common chronic inflammatory autoimmune diseases characterised by progressive, destructive inflammation of the joints leading to a loss of function and significant comorbidities; importantly, there are no cures and only 20% of patients achieve drug-free remission for over 2 years. Macrophages play a vital role in maintaining homeostasis, however, under the wrong environmental cues, become drivers of chronic synovial inflammation. Based on the current "dogma", M1 macrophages secrete pro-inflammatory cytokines and chemokines, promoting tissue degradation and joint and bone erosion which over time lead to accelerated disease progression. On the other hand, M2 macrophages secrete anti-inflammatory mediators associated with wound healing, tissue remodelling and the resolution of inflammation. Currently, four subtypes of M2 macrophages have been identified, namely M2a, M2b, M2c and M2d. However, more subtypes may exist due to macrophage plasticity and the ability for repolarisation. Macrophages are highly plastic, and polarisation exists as a continuum with diverse intermediate phenotypes. This plasticity is achieved by a highly amenable epigenome in response to environmental stimuli and shifts in metabolism. Initiating treatment during the early stages of disease is important for improved prognosis and patient outcomes. Currently, no treatment targeting macrophages specifically is available. Such therapeutics are being investigated in ongoing clinical trials. The repolarisation of pro-inflammatory macrophages towards the anti-inflammatory phenotype has been proposed as an effective approach in targeting the M1/M2 imbalance, and in turn is a potential therapeutic strategy for IA diseases. Therefore, elucidating the mechanisms that govern macrophage plasticity is fundamental for the success of novel macrophage targeting therapeutics.
Collapse
Affiliation(s)
- Karina Kulakova
- School of Biotechnology, Dublin City University, D09 V209 Dublin, Ireland; (K.K.)
- Life Sciences Institute, Dublin City University, D09 V209 Dublin, Ireland
| | - Tope Remilekun Lawal
- School of Biotechnology, Dublin City University, D09 V209 Dublin, Ireland; (K.K.)
| | - Eoghan Mccarthy
- Department of Rheumatology, Beaumont Hospital, D09 V2N0 Dublin, Ireland
- Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
| | - Achilleas Floudas
- School of Biotechnology, Dublin City University, D09 V209 Dublin, Ireland; (K.K.)
- Life Sciences Institute, Dublin City University, D09 V209 Dublin, Ireland
- Medical School, University of Ioannina, 45110 Ioannina, Greece
| |
Collapse
|
15
|
Jhamat N, Guo Y, Han J, Humblot P, Bongcam-Rudloff E, Andersson G, Niazi A. Enrichment of Cis-Acting Regulatory Elements in Differentially Methylated Regions Following Lipopolysaccharide Treatment of Bovine Endometrial Epithelial Cells. Int J Mol Sci 2024; 25:9832. [PMID: 39337320 PMCID: PMC11432661 DOI: 10.3390/ijms25189832] [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: 08/23/2024] [Revised: 09/07/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Endometritis is an inflammatory disease that negatively influences fertility and is common in milk-producing cows. An in vitro model for bovine endometrial inflammation was used to identify enrichment of cis-acting regulatory elements in differentially methylated regions (DMRs) in the genome of in vitro-cultured primary bovine endometrial epithelial cells (bEECs) before and after treatment with lipopolysaccharide (LPS) from E. coli, a key player in the development of endometritis. The enriched regulatory elements contain binding sites for transcription factors with established roles in inflammation and hypoxia including NFKB and Hif-1α. We further showed co-localization of certain enriched cis-acting regulatory motifs including ARNT, Hif-1α, and NRF1. Our results show an intriguing interplay between increased mRNA levels in LPS-treated bEECs of the mRNAs encoding the key transcription factors such as AHR, EGR2, and STAT1, whose binding sites were enriched in the DMRs. Our results demonstrate an extraordinary cis-regulatory complexity in these DMRs having binding sites for both inflammatory and hypoxia-dependent transcription factors. Obtained data using this in vitro model for bacterial-induced endometrial inflammation have provided valuable information regarding key transcription factors relevant for clinical endometritis in both cattle and humans.
Collapse
Affiliation(s)
- Naveed Jhamat
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, P.O. Box 7023, SE-75007 Uppsala, Sweden
| | - Yongzhi Guo
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, P.O. Box 7023, SE-75007 Uppsala, Sweden
| | - Jilong Han
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, P.O. Box 7023, SE-75007 Uppsala, Sweden
| | - Patrice Humblot
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, P.O. Box 7023, SE-75007 Uppsala, Sweden
| | - Erik Bongcam-Rudloff
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, P.O. Box 7023, SE-75007 Uppsala, Sweden
- SLU-Global Bioinformatics Centre, Swedish University of Agricultural Sciences, P.O. Box 7023, SE-75007 Uppsala, Sweden
| | - Göran Andersson
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, P.O. Box 7023, SE-75007 Uppsala, Sweden
| | - Adnan Niazi
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, P.O. Box 7023, SE-75007 Uppsala, Sweden
- SLU-Global Bioinformatics Centre, Swedish University of Agricultural Sciences, P.O. Box 7023, SE-75007 Uppsala, Sweden
| |
Collapse
|
16
|
Vidal CM, Alva-Ornelas JA, Chen NZ, Senapati P, Tomsic J, Robles VM, Resto C, Sanchez N, Sanchez A, Hyslop T, Emwas N, Aljaber D, Bachelder N, Martinez E, Ann D, Jones V, Winn RA, Miele L, Ochoa AC, Dietze EC, Natarajan R, Schones D, Seewaldt VL. Insulin Resistance in Women Correlates with Chromatin Histone Lysine Acetylation, Inflammatory Signaling, and Accelerated Aging. Cancers (Basel) 2024; 16:2735. [PMID: 39123463 PMCID: PMC11311683 DOI: 10.3390/cancers16152735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 07/28/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Epigenetic changes link medical, social, and environmental factors with cardiovascular and kidney disease and, more recently, with cancer. The mechanistic link between metabolic health and epigenetic changes is only starting to be investigated. In our in vitro and in vivo studies, we performed a broad analysis of the link between hyperinsulinemia and chromatin acetylation; our top "hit" was chromatin opening at H3K9ac. METHODS Building on our published preclinical studies, here, we performed a detailed analysis of the link between insulin resistance, chromatin acetylation, and inflammation using an initial test set of 28 women and validation sets of 245, 22, and 53 women. RESULTS ChIP-seq identified chromatin acetylation and opening at the genes coding for TNFα and IL6 in insulin-resistant women. Pathway analysis identified inflammatory response genes, NFκB/TNFα-signaling, reactome cytokine signaling, innate immunity, and senescence. Consistent with this finding, flow cytometry identified increased senescent circulating peripheral T-cells. DNA methylation analysis identified evidence of accelerated aging in insulin-resistant vs. metabolically healthy women. CONCLUSIONS This study shows that insulin-resistant women have increased chromatin acetylation/opening, inflammation, and, perhaps, accelerated aging. Given the role that inflammation plays in cancer initiation and progression, these studies provide a potential mechanistic link between insulin resistance and cancer.
Collapse
Affiliation(s)
- Christina M. Vidal
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (C.M.V.); (J.A.A.-O.); (P.S.); (J.T.); (V.M.R.); (C.R.); (N.S.); (A.S.); (N.E.); (D.A.); (D.A.); (V.J.); (E.C.D.)
| | - Jackelyn A. Alva-Ornelas
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (C.M.V.); (J.A.A.-O.); (P.S.); (J.T.); (V.M.R.); (C.R.); (N.S.); (A.S.); (N.E.); (D.A.); (D.A.); (V.J.); (E.C.D.)
| | - Nancy Zhuo Chen
- Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Duarte, Duarte, CA 91010, USA; (N.Z.C.); (N.B.); (R.N.)
| | - Parijat Senapati
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (C.M.V.); (J.A.A.-O.); (P.S.); (J.T.); (V.M.R.); (C.R.); (N.S.); (A.S.); (N.E.); (D.A.); (D.A.); (V.J.); (E.C.D.)
| | - Jerneja Tomsic
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (C.M.V.); (J.A.A.-O.); (P.S.); (J.T.); (V.M.R.); (C.R.); (N.S.); (A.S.); (N.E.); (D.A.); (D.A.); (V.J.); (E.C.D.)
| | - Vanessa Myriam Robles
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (C.M.V.); (J.A.A.-O.); (P.S.); (J.T.); (V.M.R.); (C.R.); (N.S.); (A.S.); (N.E.); (D.A.); (D.A.); (V.J.); (E.C.D.)
| | - Cristal Resto
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (C.M.V.); (J.A.A.-O.); (P.S.); (J.T.); (V.M.R.); (C.R.); (N.S.); (A.S.); (N.E.); (D.A.); (D.A.); (V.J.); (E.C.D.)
| | - Nancy Sanchez
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (C.M.V.); (J.A.A.-O.); (P.S.); (J.T.); (V.M.R.); (C.R.); (N.S.); (A.S.); (N.E.); (D.A.); (D.A.); (V.J.); (E.C.D.)
| | - Angelica Sanchez
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (C.M.V.); (J.A.A.-O.); (P.S.); (J.T.); (V.M.R.); (C.R.); (N.S.); (A.S.); (N.E.); (D.A.); (D.A.); (V.J.); (E.C.D.)
| | - Terry Hyslop
- Sidney Kimmel Cancer Center, Philadelphia, PA 19107, USA;
| | - Nour Emwas
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (C.M.V.); (J.A.A.-O.); (P.S.); (J.T.); (V.M.R.); (C.R.); (N.S.); (A.S.); (N.E.); (D.A.); (D.A.); (V.J.); (E.C.D.)
| | - Dana Aljaber
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (C.M.V.); (J.A.A.-O.); (P.S.); (J.T.); (V.M.R.); (C.R.); (N.S.); (A.S.); (N.E.); (D.A.); (D.A.); (V.J.); (E.C.D.)
| | - Nick Bachelder
- Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Duarte, Duarte, CA 91010, USA; (N.Z.C.); (N.B.); (R.N.)
| | - Ernest Martinez
- Department of Biochemistry, University of California at Riverside, Riverside, CA 92521, USA;
| | - David Ann
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (C.M.V.); (J.A.A.-O.); (P.S.); (J.T.); (V.M.R.); (C.R.); (N.S.); (A.S.); (N.E.); (D.A.); (D.A.); (V.J.); (E.C.D.)
| | - Veronica Jones
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (C.M.V.); (J.A.A.-O.); (P.S.); (J.T.); (V.M.R.); (C.R.); (N.S.); (A.S.); (N.E.); (D.A.); (D.A.); (V.J.); (E.C.D.)
| | - Robert A. Winn
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Lucio Miele
- School of Medicine, Louisiana State University, New Orleans, LA 70112, USA; (L.M.); (A.C.O.)
| | - Augusto C. Ochoa
- School of Medicine, Louisiana State University, New Orleans, LA 70112, USA; (L.M.); (A.C.O.)
| | - Eric C. Dietze
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (C.M.V.); (J.A.A.-O.); (P.S.); (J.T.); (V.M.R.); (C.R.); (N.S.); (A.S.); (N.E.); (D.A.); (D.A.); (V.J.); (E.C.D.)
| | - Rama Natarajan
- Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Duarte, Duarte, CA 91010, USA; (N.Z.C.); (N.B.); (R.N.)
| | - Dustin Schones
- Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Duarte, Duarte, CA 91010, USA; (N.Z.C.); (N.B.); (R.N.)
| | - Victoria L. Seewaldt
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (C.M.V.); (J.A.A.-O.); (P.S.); (J.T.); (V.M.R.); (C.R.); (N.S.); (A.S.); (N.E.); (D.A.); (D.A.); (V.J.); (E.C.D.)
| |
Collapse
|
17
|
Reddy SU, Sadia FZ, Vancura A, Vancurova I. IFNγ-Induced Bcl3, PD-L1 and IL-8 Signaling in Ovarian Cancer: Mechanisms and Clinical Significance. Cancers (Basel) 2024; 16:2676. [PMID: 39123403 PMCID: PMC11311860 DOI: 10.3390/cancers16152676] [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: 05/19/2024] [Revised: 07/18/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
IFNγ, a pleiotropic cytokine produced not only by activated lymphocytes but also in response to cancer immunotherapies, has both antitumor and tumor-promoting functions. In ovarian cancer (OC) cells, the tumor-promoting functions of IFNγ are mediated by IFNγ-induced expression of Bcl3, PD-L1 and IL-8/CXCL8, which have long been known to have critical cellular functions as a proto-oncogene, an immune checkpoint ligand and a chemoattractant, respectively. However, overwhelming evidence has demonstrated that these three genes have tumor-promoting roles far beyond their originally identified functions. These tumor-promoting mechanisms include increased cancer cell proliferation, invasion, angiogenesis, metastasis, resistance to chemotherapy and immune escape. Recent studies have shown that IFNγ-induced Bcl3, PD-L1 and IL-8 expression is regulated by the same JAK1/STAT1 signaling pathway: IFNγ induces the expression of Bcl3, which then promotes the expression of PD-L1 and IL-8 in OC cells, resulting in their increased proliferation and migration. In this review, we summarize the recent findings on how IFNγ affects the tumor microenvironment and promotes tumor progression, with a special focus on ovarian cancer and on Bcl3, PD-L1 and IL-8/CXCL8 signaling. We also discuss promising novel combinatorial strategies in clinical trials targeting Bcl3, PD-L1 and IL-8 to increase the effectiveness of cancer immunotherapies.
Collapse
Affiliation(s)
| | | | | | - Ivana Vancurova
- Department of Biological Sciences, St. John’s University, New York, NY 11439, USA; (S.U.R.); (F.Z.S.); (A.V.)
| |
Collapse
|
18
|
Mak A, Sung CC, Pisitkun T, Khositseth S, Knepper MA. 'Aquaporin-omics': mechanisms of aquaporin-2 loss in polyuric disorders. J Physiol 2024; 602:3191-3206. [PMID: 37114282 PMCID: PMC10603215 DOI: 10.1113/jp284634] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 04/21/2023] [Indexed: 04/29/2023] Open
Abstract
Animal models of a variety of acquired nephrogenic diabetes insipidus (NDI) disorders have identified a common feature: all such models are associated with the loss of aquaporin-2 (AQP2) from collecting duct principal cells, explaining the associated polyuria. To discover mechanisms of AQP2 loss, previous investigators have carried out either transcriptomics (lithium-induced NDI, unilateral ureteral obstruction, endotoxin-induced NDI) or proteomics (hypokalaemia-associated NDI, hypercalcaemia-associated NDI, bilateral ureteral obstruction), yielding contrasting views. Here, to address whether there may be common mechanisms underlying loss of AQP2 in acquired NDI disorders, we have used bioinformatic data integration techniques to combine information from all transcriptomic and proteomic data sets. The analysis reveals roles for autophagy/apoptosis, oxidative stress and inflammatory signalling as key elements of the mechanism that results in loss of AQP2. These processes can cause AQP2 loss through the combined effects of repression of Aqp2 gene transcription, generalized translational repression, and increased autophagic degradation of proteins including AQP2. Two possible types of stress-sensor proteins, namely death receptors and stress-sensitive protein kinases of the EIF2AK family, are discussed as potential triggers for signalling processes that result in loss of AQP2. KEY POINTS: Prior studies have shown in a variety of animal models of acquired nephrogenic diabetes insipidus (NDI) that loss of the aquaporin-2 (AQP2) protein is a common feature. Investigations of acquired NDI using transcriptomics (RNA-seq) and proteomics (protein mass spectrometry) have led to differing conclusions regarding mechanisms of AQP2 loss. Bioinformatic integration of transcriptomic and proteomic data from these prior studies now reveals that acquired NDI models map to three core processes: oxidative stress, apoptosis/autophagy and inflammatory signalling. These processes cause loss of AQP2 through translational repression, accelerated degradation of proteins, and transcriptional repression.
Collapse
Affiliation(s)
- Angela Mak
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Chih-Chien Sung
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Trairak Pisitkun
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Sookkasem Khositseth
- Department of Pediatrics, Faculty of Medicine, Thammasat University, Bangkok,Thailand
| | - Mark A. Knepper
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
19
|
Lee H, Kang SH, Jeong GH, Lee SS, Chung BY, Kim GJ, Bai HW. Gamma irradiation-engineered macrophage-derived exosomes as potential immunomodulatory therapeutic agents. PLoS One 2024; 19:e0303434. [PMID: 38865377 PMCID: PMC11168684 DOI: 10.1371/journal.pone.0303434] [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: 01/18/2024] [Accepted: 04/23/2024] [Indexed: 06/14/2024] Open
Abstract
The modulation of macrophage polarization is a promising strategy for maintaining homeostasis and improving innate and adaptive immunity. Low-dose ionizing radiation has been implicated in macrophage immunomodulatory responses. However, studies on the relationship between exosomes and regulation of macrophage polarization induced by ionizing radiation are limited. Therefore, this study investigated the alterations in macrophages and exosomes induced by gamma irradiation and elucidated the underlying mechanisms. We used the mouse macrophage cell line RAW 264.7 to generate macrophages and performed western blot, quantitative reverse transcription-PCR, and gene ontology analyses to elucidate the molecular profiles of macrophage-derived exosomes under varying treatment conditions, including 10 Gy gamma irradiation. Exosomes isolated from gamma-irradiated M1 macrophages exhibited an enhanced M1 phenotype. Irradiation induced the activation of NF-κB and NLRP3 signaling in M1 macrophages, thereby promoting the expression of pro-inflammatory cytokines. Cytokine expression was also upregulated in gamma-irradiated M1 macrophage-released exosomes. Therefore, gamma irradiation has a remarkable effect on the immunomodulatory mechanisms and cytokine profiles of gamma-irradiated M1 macrophage-derived exosomes, and represents a potential immunotherapeutic modality.
Collapse
Affiliation(s)
- Hanui Lee
- Radiation Biotechnology Division, Advanced Radiation Technology Institute (ARTI), Korea Atomic Energy Research Institute (KAERI), Jeongeup, Republic of Korea
- Department of Biological Sciences and Research Center of Ecomimetics, College of Natural Science, Chonnam National University, Gwangju, Republic of Korea
| | - Seong Hee Kang
- Radiation Biotechnology Division, Advanced Radiation Technology Institute (ARTI), Korea Atomic Energy Research Institute (KAERI), Jeongeup, Republic of Korea
| | - Gyeong Han Jeong
- Radiation Biotechnology Division, Advanced Radiation Technology Institute (ARTI), Korea Atomic Energy Research Institute (KAERI), Jeongeup, Republic of Korea
| | - Seoung Sik Lee
- Radiation Biotechnology Division, Advanced Radiation Technology Institute (ARTI), Korea Atomic Energy Research Institute (KAERI), Jeongeup, Republic of Korea
- Radiation Biotechnology and Applied Radioisotope Science, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Byung Yeoup Chung
- Radiation Biotechnology Division, Advanced Radiation Technology Institute (ARTI), Korea Atomic Energy Research Institute (KAERI), Jeongeup, Republic of Korea
| | - Geun-Joong Kim
- Department of Biological Sciences and Research Center of Ecomimetics, College of Natural Science, Chonnam National University, Gwangju, Republic of Korea
| | - Hyoung-Woo Bai
- Radiation Biotechnology Division, Advanced Radiation Technology Institute (ARTI), Korea Atomic Energy Research Institute (KAERI), Jeongeup, Republic of Korea
- Radiation Biotechnology and Applied Radioisotope Science, University of Science and Technology (UST), Daejeon, Republic of Korea
| |
Collapse
|
20
|
Omodaka S, Kato Y, Sato Y, Falcone-Juengert J, Zhang H, Kanoke A, Eckalbar WL, Endo H, Hsieh CL, Aran D, Liu J. Defective interferon signaling in the circulating monocytes of type 2 diabetic mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.03.597050. [PMID: 38895236 PMCID: PMC11185546 DOI: 10.1101/2024.06.03.597050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Type 2 diabetes mellitus (T2DM) is associated with poor outcome after stroke. Peripheral monocytes play a critical role in the secondary injury and recovery of damaged brain tissue after stroke, but the underlying mechanisms are largely unclear. To investigate transcriptome changes and molecular networks across monocyte subsets in response to T2DM and stroke, we performed single-cell RNA-sequencing (scRNAseq) from peripheral blood mononuclear cells and bulk RNA-sequencing from blood monocytes from four groups of adult mice, consisting of T2DM model db/db and normoglycemic control db/+ mice with or without ischemic stroke. Via scRNAseq we found that T2DM expands the monocyte population at the expense of lymphocytes, which was validated by flow cytometry. Among the monocytes, T2DM also disproportionally increased the inflammatory subsets with Ly6C+ and negative MHC class II expression (MO.6C+II-). Conversely, monocytes from control mice without stroke are enriched with steady-state classical monocyte subset of MO.6C+II+ but with the least percentage of MO.6C+II- subtype. Apart from enhancing inflammation and coagulation, enrichment analysis from both scRNAseq and bulk RNAseq revealed that T2DM specifically suppressed type-1 and type-2 interferon signaling pathways crucial for antigen presentation and the induction of ischemia tolerance. Preconditioning by lipopolysaccharide conferred neuroprotection against ischemic brain injury in db/+ but not in db/db mice and coincided with a lesser induction of brain Interferon-regulatory-factor-3 in the brains of the latter mice. Our results suggest that the increased diversity and altered transcriptome in the monocytes of T2DM mice underlie the worse stroke outcome by exacerbating secondary injury and potentiating stroke-induced immunosuppression. Significance Statement The mechanisms involved in the detrimental diabetic effect on stroke are largely unclear. We show here, for the first time, that peripheral monocytes have disproportionally altered the subsets and changed transcriptome under diabetes and/or stroke conditions. Moreover, genes in the IFN-related signaling pathways are suppressed in the diabetic monocytes, which underscores the immunosuppression and impaired ischemic tolerance under the T2DM condition. Our data raise a possibility that malfunctioned monocytes may systemically and focally affect the host, leading to the poor outcome of diabetes in the setting of stroke. The results yield important clues to molecular mechanisms involved in the detrimental diabetic effect on stroke outcome.
Collapse
|
21
|
Mildenberger J, Rebours C. Green ( Ulva fenestrata) and Brown ( Saccharina latissima) Macroalgae Similarly Modulate Inflammatory Signaling by Activating NF- κB and Dampening IRF in Human Macrophage-Like Cells. J Immunol Res 2024; 2024:8121284. [PMID: 38799117 PMCID: PMC11126347 DOI: 10.1155/2024/8121284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 02/22/2024] [Accepted: 04/23/2024] [Indexed: 05/29/2024] Open
Abstract
Macroalgae are considered healthy food ingredients due to their content in numerous bioactive compounds, and the traditional use of whole macroalgae in Asian cuisine suggests a contribution to longevity. Although much information is available about the bioactivity of pure algal compounds, such as different polyphenols and polysaccharides, documentation of potential effects of whole macroalgae as part of Western diets is limited. Lifestyle- and age-related diseases, which have a high impact on population health, are closely connected to underlying chronic inflammation. Therefore, we have studied crude extracts of green (Ulva fenestrata) and brown (Saccharina latissima) macroalgae, as two of the most promising food macroalgae in the Nordic countries for their effect on inflammation in vitro. Human macrophage-like reporter THP-1 cells were treated with macroalgae extracts and stimulated with lipopolysaccharide (LPS) to induce inflammatory signalling. Effects of the macroalgae extracts were assessed on transcription factor activity of NF-κB and IRF as well as secretion and/or expression of the cytokines TNF-α and IFN-β and chemokines IL-8 and CXCL10. The crude macroalgae extracts were further separated into polyphenol-enriched and polysaccharide-enriched fractions, which were also tested for their effect on transcription factor activity. Interestingly, we observed a selective activation of NF-κB, when cells were treated with macroalgae extracts. On the other hand, pretreatment with macroalgae extracts selectively repressed IRF activation when inflammatory signaling was subsequently induced by LPS. This effect was consistent for both tested species as well as for polyphenol- and polysaccharide-enriched fractions, of which the latter had more pronounced effects. Overall, this is the first indication of how macroalgae could modulate inflammatory signaling by selective activation and subsequent repression of different pathways. Further in vitro and in vivo studies of this mechanism would be needed to understand how macroalgae consumption could influence the prevention of noncommunicable, lifestyle- and age-related diseases that are highly related to unbalanced inflammatory processes.
Collapse
|
22
|
Ghazanfari D, Courreges MC, Belinski LE, Hogrell MJ, Lloyd J, C Bergmeier S, McCall KD, Goetz DJ. Mechanistic insights into SARS-CoV-2 spike protein induction of the chemokine CXCL10. Sci Rep 2024; 14:11179. [PMID: 38750069 PMCID: PMC11096305 DOI: 10.1038/s41598-024-61906-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/10/2024] [Indexed: 05/18/2024] Open
Abstract
During a SARS-CoV-2 infection, macrophages recognize viral components resulting in cytokine production. While this response fuels virus elimination, overexpression of cytokines can lead to severe COVID-19. Previous studies suggest that the spike protein (S) of SARS-CoV-2 can elicit cytokine production via the transcription factor NF-κB and the toll-like receptors (TLRs). In this study, we found that: (i) S and the S2 subunit induce CXCL10, a chemokine implicated in severe COVID-19, gene expression by human macrophage cells (THP-1); (ii) a glycogen synthase kinase-3 inhibitor attenuates this induction; (iii) S and S2 do not activate NF-κB but do activate the transcription factor IRF; (iv) S and S2 do not require TLR2 to elicit CXCL10 production or activate IRF; and (v) S and S2 elicit CXCL10 production by peripheral blood mononuclear cells (PBMCs). We also discovered that the cellular response, or lack thereof, to S and S2 is a function of the recombinant S and S2 used. While such a finding raises the possibility of confounding LPS contamination, we offer evidence that potential contaminating LPS does not underly induced increases in CXCL10. Combined, these results provide insights into the complex immune response to SARS-CoV-2 and suggest possible therapeutic targets for severe COVID-19.
Collapse
Affiliation(s)
- Davoud Ghazanfari
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, 45701, USA
| | | | - Lydia E Belinski
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, 45701, USA
- Biomedical Engineering Program, Ohio University, Athens, OH, 45701, USA
| | - Michael J Hogrell
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, 45701, USA
- Biomedical Engineering Program, Ohio University, Athens, OH, 45701, USA
| | - Jacob Lloyd
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, 45701, USA
| | - Stephen C Bergmeier
- Biomedical Engineering Program, Ohio University, Athens, OH, 45701, USA
- Department of Chemistry and Biochemistry, Ohio University, Athens, OH, 45701, USA
| | - Kelly D McCall
- Department of Specialty Medicine, Ohio University, Athens, OH, 45701, USA
- Biomedical Engineering Program, Ohio University, Athens, OH, 45701, USA
- The Diabetes Institute, Ohio University, Athens, OH, 45701, USA
- Molecular and Cellular Biology Program, Ohio University College of Arts & Sciences, Athens, OH, 45701, USA
- Department of Biological Sciences, Ohio University College of Arts & Sciences, Athens, OH, 45701, USA
- Department of Biomedical Sciences, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, 45701, USA
| | - Douglas J Goetz
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, 45701, USA.
- Biomedical Engineering Program, Ohio University, Athens, OH, 45701, USA.
| |
Collapse
|
23
|
Nagy G, Bojcsuk D, Tzerpos P, Cseh T, Nagy L. Lineage-determining transcription factor-driven promoters regulate cell type-specific macrophage gene expression. Nucleic Acids Res 2024; 52:4234-4256. [PMID: 38348998 PMCID: PMC11077085 DOI: 10.1093/nar/gkae088] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 01/18/2024] [Accepted: 01/29/2024] [Indexed: 05/09/2024] Open
Abstract
Mammalian promoters consist of multifarious elements, which make them unique and support the selection of the proper transcript variants required under diverse conditions in distinct cell types. However, their direct DNA-transcription factor (TF) interactions are mostly unidentified. Murine bone marrow-derived macrophages (BMDMs) are a widely used model for studying gene expression regulation. Thus, this model serves as a rich source of various next-generation sequencing data sets, including a large number of TF cistromes. By processing and integrating the available cistromic, epigenomic and transcriptomic data from BMDMs, we characterized the macrophage-specific direct DNA-TF interactions, with a particular emphasis on those specific for promoters. Whilst active promoters are enriched for certain types of typically methylatable elements, more than half of them contain non-methylatable and prototypically promoter-distal elements. In addition, circa 14% of promoters-including that of Csf1r-are composed exclusively of 'distal' elements that provide cell type-specific gene regulation by specialized TFs. Similar to CG-rich promoters, these also contain methylatable CG sites that are demethylated in a significant portion and show high polymerase activity. We conclude that this unusual class of promoters regulates cell type-specific gene expression in macrophages, and such a mechanism might exist in other cell types too.
Collapse
Affiliation(s)
- Gergely Nagy
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dóra Bojcsuk
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Petros Tzerpos
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tímea Cseh
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - László Nagy
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA
| |
Collapse
|
24
|
Mishra B, Ivashkiv LB. Interferons and epigenetic mechanisms in training, priming and tolerance of monocytes and hematopoietic progenitors. Immunol Rev 2024; 323:257-275. [PMID: 38567833 PMCID: PMC11102283 DOI: 10.1111/imr.13330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 03/11/2024] [Indexed: 05/18/2024]
Abstract
Training and priming of innate immune cells involve preconditioning by PAMPs, DAMPs, and/or cytokines that elicits stronger induction of inflammatory genes upon secondary challenge. Previous models distinguish training and priming based upon whether immune activation returns to baseline prior to secondary challenge. Tolerance is a protective mechanism whereby potent stimuli induce refractoriness to secondary challenge. Training and priming are important for innate memory responses that protect against infection, efficacy of vaccines, and maintaining innate immune cells in a state of readiness; tolerance prevents toxicity from excessive immune activation. Dysregulation of these processes can contribute to pathogenesis of autoimmune/inflammatory conditions, post-COVID-19 hyperinflammatory states, or sepsis-associated immunoparalysis. Training, priming, and tolerance regulate similar "signature" inflammatory genes such as TNF, IL6, and IL1B and utilize overlapping epigenetic mechanisms. We review how interferons (IFNs), best known for activating JAK-STAT signaling and interferon-stimulated genes, also play a key role in regulating training, priming, and tolerance via chromatin-mediated mechanisms. We present new data on how monocyte-to-macrophage differentiation modulates IFN-γ-mediated priming, affects regulation of AP-1 and CEBP activity, and attenuates superinduction of inflammatory genes. We present a "training-priming continuum" model that integrates IFN-mediated priming into current concepts about training and tolerance and proposes a central role for STAT1 and IRF1.
Collapse
Affiliation(s)
- Bikash Mishra
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, New York, USA
| | - Lionel B Ivashkiv
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, New York, USA
- Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
25
|
Moe A, Rayasam A, Sauber G, Shah RK, Doherty A, Yuan CY, Szabo A, Moore BM, Colonna M, Cui W, Romero J, Zamora AE, Hillard CJ, Drobyski WR. Type 2 cannabinoid receptor expression on microglial cells regulates neuroinflammation during graft-versus-host disease. J Clin Invest 2024; 134:e175205. [PMID: 38662453 PMCID: PMC11142740 DOI: 10.1172/jci175205] [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: 09/07/2023] [Accepted: 04/12/2024] [Indexed: 04/29/2024] Open
Abstract
Neuroinflammation is a recognized complication of immunotherapeutic approaches such as immune checkpoint inhibitor treatment, chimeric antigen receptor therapy, and graft versus host disease (GVHD) occurring after allogeneic hematopoietic stem cell transplantation. While T cells and inflammatory cytokines play a role in this process, the precise interplay between the adaptive and innate arms of the immune system that propagates inflammation in the central nervous system remains incompletely understood. Using a murine model of GVHD, we demonstrate that type 2 cannabinoid receptor (CB2R) signaling plays a critical role in the pathophysiology of neuroinflammation. In these studies, we identify that CB2R expression on microglial cells induces an activated inflammatory phenotype that potentiates the accumulation of donor-derived proinflammatory T cells, regulates chemokine gene regulatory networks, and promotes neuronal cell death. Pharmacological targeting of this receptor with a brain penetrant CB2R inverse agonist/antagonist selectively reduces neuroinflammation without deleteriously affecting systemic GVHD severity. Thus, these findings delineate a therapeutically targetable neuroinflammatory pathway and have implications for the attenuation of neurotoxicity after GVHD and potentially other T cell-based immunotherapeutic approaches.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Aniko Szabo
- Division of Biostatistics, Institute of Health and Equity, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Bob M. Moore
- College of Pharmacy, Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University, Saint Louis, Missouri, USA
| | - Weiguo Cui
- Department of Pathology, Northwestern University, Chicago, Illinois, USA
| | - Julian Romero
- Faculty of Experimental Sciences, Francisco de Vitoria University, Madrid, Spain
| | | | | | | |
Collapse
|
26
|
Thottakara T, Padmanabhan A, Tanriverdi T, Thambidurai T, Diaz-RG JA, Amonkar SR, Olgin JE, Long CS, Roselle Abraham M. Single-nucleus RNA/ATAC-seq in early-stage HCM models predicts SWI/SNF-activation in mutant-myocytes, and allele-specific differences in fibroblasts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.24.589078. [PMID: 38903075 PMCID: PMC11188105 DOI: 10.1101/2024.04.24.589078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Hypertrophic cardiomyopathy (HCM) is associated with phenotypic variability. To gain insights into transcriptional regulation of cardiac phenotype, single-nucleus linked RNA-/ATAC-seq was performed in 5-week-old control mouse-hearts (WT) and two HCM-models (R92W-TnT, R403Q-MyHC) that exhibit differences in heart size/function and fibrosis; mutant data was compared to WT. Analysis of 23,304 nuclei from mutant hearts, and 17,669 nuclei from WT, revealed similar dysregulation of gene expression, activation of AP-1 TFs (FOS, JUN) and the SWI/SNF complex in both mutant ventricular-myocytes. In contrast, marked differences were observed between mutants, for gene expression/TF enrichment, in fibroblasts, macrophages, endothelial cells. Cellchat predicted activation of pro-hypertrophic IGF-signaling in both mutant ventricular-myocytes, and profibrotic TGFβ-signaling only in mutant-TnT fibroblasts. In summary, our bioinformatics analyses suggest that activation of IGF-signaling, AP-1 TFs and the SWI/SNF chromatin remodeler complex promotes myocyte hypertrophy in early-stage HCM. Selective activation of TGFβ-signaling in mutant-TnT fibroblasts contributes to genotype-specific differences in cardiac fibrosis.
Collapse
Affiliation(s)
- Tilo Thottakara
- Department of Medicine, University of California San Francisco, Division of Cardiology, San Francisco
- Department of Cardiology, University Heart and Vascular Center Hamburg, Germany
| | - Arun Padmanabhan
- Department of Medicine, University of California San Francisco, Division of Cardiology, San Francisco
- Gladstone Institutes, San Francisco, CA, USA
| | - Talha Tanriverdi
- Department of Medicine, University of California San Francisco, Division of Cardiology, San Francisco
| | - Tharika Thambidurai
- Department of Medicine, University of California San Francisco, Division of Cardiology, San Francisco
| | - Jose A. Diaz-RG
- Department of Medicine, University of California San Francisco, Division of Cardiology, San Francisco
| | - Sanika R. Amonkar
- Department of Medicine, University of California San Francisco, Division of Cardiology, San Francisco
| | - Jeffrey E. Olgin
- Department of Medicine, University of California San Francisco, Division of Cardiology, San Francisco
| | - Carlin S. Long
- Department of Medicine, University of California San Francisco, Division of Cardiology, San Francisco
| | - M. Roselle Abraham
- Department of Medicine, University of California San Francisco, Division of Cardiology, San Francisco
| |
Collapse
|
27
|
Pandey S, Anang V, Schumacher MM. Mitochondria driven innate immune signaling and inflammation in cancer growth, immune evasion, and therapeutic resistance. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 386:223-247. [PMID: 38782500 DOI: 10.1016/bs.ircmb.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Mitochondria play an important and multifaceted role in cellular function, catering to the cell's energy and biosynthetic requirements. They modulate apoptosis while responding to diverse extracellular and intracellular stresses including reactive oxygen species (ROS), nutrient and oxygen scarcity, endoplasmic reticulum stress, and signaling via surface death receptors. Integral components of mitochondria, such as mitochondrial DNA (mtDNA), mitochondrial RNA (mtRNA), Adenosine triphosphate (ATP), cardiolipin, and formyl peptides serve as major damage-associated molecular patterns (DAMPs). These molecules activate multiple innate immune pathways both in the cytosol [such as Retionoic Acid-Inducible Gene-1 (RIG-1) and Cyclic GMP-AMP Synthase (cGAS)] and on the cell surface [including Toll-like receptors (TLRs)]. This activation cascade leads to the release of various cytokines, chemokines, interferons, and other inflammatory molecules and oxidative species. The innate immune pathways further induce chronic inflammation in the tumor microenvironment which either promotes survival and proliferation or promotes epithelial to mesenchymal transition (EMT), metastasis and therapeutic resistance in the cancer cell's. Chronic activation of innate inflammatory pathways in tumors also drives immunosuppressive checkpoint expression in the cancer cells and boosts the influx of immune-suppressive populations like Myeloid-Derived Suppressor Cells (MDSCs) and Regulatory T cells (Tregs) in cancer. Thus, sensing of cellular stress by the mitochondria may lead to enhanced tumor growth. In addition to that, the tumor microenvironment also becomes a source of immunosuppressive cytokines. These cytokines exert a debilitating effect on the functioning of immune effector cells, and thus foster immune tolerance and facilitate immune evasion. Here we describe how alteration of the mitochondrial homeostasis and cellular stress drives innate inflammatory pathways in the tumor microenvironment.
Collapse
Affiliation(s)
- Sanjay Pandey
- Department of Radiation Oncology, Montefiore Medical Center, Bronx, NY, United States.
| | - Vandana Anang
- International Center for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Michelle M Schumacher
- Department of Radiation Oncology, Montefiore Medical Center, Bronx, NY, United States; Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
28
|
Zheng P, He J, Fu Y, Yang Y, Li S, Duan B, Yang Y, Hu Y, Yang Z, Wang M, Liu Q, Zheng X, Hua L, Li W, Li D, Ding Y, Yang X, Bai H, Long Q, Huang W, Ma Y. Engineered Bacterial Biomimetic Vesicles Reprogram Tumor-Associated Macrophages and Remodel Tumor Microenvironment to Promote Innate and Adaptive Antitumor Immune Responses. ACS NANO 2024; 18:6863-6886. [PMID: 38386537 DOI: 10.1021/acsnano.3c06987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Tumor-associated macrophages (TAMs) are among the most abundant infiltrating leukocytes in the tumor microenvironment (TME). Reprogramming TAMs from protumor M2 to antitumor M1 phenotype is a promising strategy for remodeling the TME and promoting antitumor immunity; however, the development of an efficient strategy remains challenging. Here, a genetically modified bacterial biomimetic vesicle (BBV) with IFN-γ exposed on the surface in a nanoassembling membrane pore structure was constructed. The engineered IFN-γ BBV featured a nanoscale structure of protein and lipid vesicle, the existence of rich pattern-associated molecular patterns (PAMPs), and the costimulation of introduced IFN-γ molecules. In vitro, IFN-γ BBV reprogrammed M2 macrophages to M1, possibly through NF-κB and JAK-STAT signaling pathways, releasing nitric oxide (NO) and inflammatory cytokines IL-1β, IL-6, and TNF-α and increasing the expression of IL-12 and iNOS. In tumor-bearing mice, IFN-γ BBV demonstrated a targeted enrichment in tumors and successfully reprogrammed TAMs into the M1 phenotype; notably, the response of antigen-specific cytotoxic T lymphocyte (CTL) in TME was promoted while the immunosuppressive myeloid-derived suppressor cell (MDSC) was suppressed. The tumor growth was found to be significantly inhibited in both a TC-1 tumor and a CT26 tumor. It was indicated that the antitumor effects of IFN-γ BBV were macrophage-dependent. Further, the modulation of TME by IFN-γ BBV produced synergistic effects against tumor growth and metastasis with an immune checkpoint inhibitor in an orthotopic 4T1 breast cancer model which was insensitive to anti-PD-1 mAb alone. In conclusion, IFN-γ-modified BBV demonstrated a strong capability of efficiently targeting tumor and tuning a cold tumor hot through reprogramming TAMs, providing a potent approach for tumor immunotherapy.
Collapse
Affiliation(s)
- Peng Zheng
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, People's Republic of China
| | - Jinrong He
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, People's Republic of China
| | - Yuting Fu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, People's Republic of China
| | - Ying Yang
- Cell Biology & Molecular Biology Laboratory of Experimental Teaching Center, Faculty of Basic Medical Science, Kunming Medical University, Kunming 650500, People's Republic of China
| | - Shuqin Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- Kunming Medical University, Kunming 650500, People's Republic of China
| | - Biao Duan
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- Kunming Medical University, Kunming 650500, People's Republic of China
| | - Ying Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
| | - Yongmao Hu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- School of Life Sciences, Yunnan University, Kunming 650091, People's Republic of China
| | - Zhongqian Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
| | - Mengzhen Wang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
| | - Qingwen Liu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- Kunming Medical University, Kunming 650500, People's Republic of China
| | - Xiao Zheng
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- School of Life Sciences, Yunnan University, Kunming 650091, People's Republic of China
| | - Liangqun Hua
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- School of Life Sciences, Yunnan University, Kunming 650091, People's Republic of China
| | - Weiran Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
| | - Duo Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- Department of Acute Infectious Diseases Control and Prevention, Yunnan Provincial Centers for Disease Control and Prevention, Kunming 530112, People's Republic of China
| | - Yiting Ding
- School of Life Sciences, Yunnan University, Kunming 650091, People's Republic of China
| | - Xu Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, People's Republic of China
| | - Hongmei Bai
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, People's Republic of China
| | - Qiong Long
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, People's Republic of China
| | - Weiwei Huang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, People's Republic of China
| | - Yanbing Ma
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, People's Republic of China
| |
Collapse
|
29
|
Hu Z, Sui Q, Jin X, Shan G, Huang Y, Yi Y, Zeng D, Zhao M, Zhan C, Wang Q, Lin Z, Lu T, Chen Z. IL6-STAT3-C/EBPβ-IL6 positive feedback loop in tumor-associated macrophages promotes the EMT and metastasis of lung adenocarcinoma. J Exp Clin Cancer Res 2024; 43:63. [PMID: 38424624 PMCID: PMC10903044 DOI: 10.1186/s13046-024-02989-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/19/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Lung cancer is one of the most common tumors in the world, and metastasis is one of the major causes of tumor-related death in lung cancer patients. Tumor-associated macrophages (TAMs) are a major component of the tumor microenvironment (TME) and are frequently associated with tumor metastasis in human cancers. However, the regulatory mechanisms of TAMs in lung cancer metastasis remain unclear. METHODS Single-cell sequencing analysis of lung cancer and normal tissues from public databases and from 14 patients who underwent surgery at Zhongshan Hospital was performed. In vitro co-culture experiments were performed to evaluate the effects of TAMs on lung cancer migration and invasion. Changes in the expression of IL-6, STAT3, C/EBPΒ, and EMT pathway were verified using RT-qPCR, western blotting, and immunofluorescence. Dual luciferase reporter assays and ChIP were used to reveal potential regulatory sites on the transcription factor sets. In addition, the effects of TAMs on lung cancer progression and metastasis were confirmed by in vivo models. RESULTS TAM infiltration is associated with tumor progression and poor prognosis. IL-6 secreted by TAMs can activate the JAK2/STAT3 pathway through autocrine secretion, and STAT3 acts as a transcription factor to activate the expression of C/EBPβ, which further promotes the transcription and expression of IL-6, forming positive feedback loops for IL6-STAT3-C/EBPβ-IL6 in TAMs. IL-6 secreted by TAMs promotes lung cancer progression and metastasis in vivo and in vitro by activating the EMT pathway, which can be attenuated by the use of JAK2/STAT3 pathway inhibitors or IL-6 monoclonal antibodies. CONCLUSIONS Our data suggest that TAMs promote IL-6 expression by forming an IL6-STAT3-C/EBPβ-IL6 positive feedback loop. Released IL-6 can induce the EMT pathway in lung cancer to enhance migration, invasion, and metastasis. The use of IL-6-neutralizing antibody can partially counteract the promotion of LUAD by TAMs. A novel mechanism of macrophage-promoted tumor progression was revealed, and the IL6-STAT3-C/EBPβ-IL6 signaling cascade may be a potential therapeutic target against lung cancer.
Collapse
Affiliation(s)
- Zhengyang Hu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Qihai Sui
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Xing Jin
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Guangyao Shan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Yiwei Huang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Yanjun Yi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Dejun Zeng
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Mengnan Zhao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Cheng Zhan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Qun Wang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Zongwu Lin
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China.
| | - Tao Lu
- Department of Thoracic Surgery, Shanxi Province Cancer Hospital / Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences / Cancer Hospital Affiliated to Shanxi Medical University, No. 3 Gongren Xin Jie, Xinghualing District, Taiyuan, 030013, Shanxi Province, China.
| | - Zhencong Chen
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China.
| |
Collapse
|
30
|
Banerjee R, Ajithkumar P, Keestra N, Smith J, Gimenez G, Rodger EJ, Eccles MR, Antony J, Weeks RJ, Chatterjee A. Targeted DNA Methylation Editing Using an All-in-One System Establishes Paradoxical Activation of EBF3. Cancers (Basel) 2024; 16:898. [PMID: 38473261 DOI: 10.3390/cancers16050898] [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: 01/20/2024] [Revised: 02/17/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Cutaneous melanoma is rapidly on the rise globally, surpassing the growth rate of other cancers, with metastasis being the primary cause of death in melanoma patients. Consequently, understanding the mechanisms behind this metastatic process and exploring innovative treatments is of paramount importance. Recent research has shown promise in unravelling the role of epigenetic factors in melanoma progression to metastasis. While DNA hypermethylation at gene promoters typically suppresses gene expression, we have contributed to establishing the newly understood mechanism of paradoxical activation of genes via DNA methylation, where high methylation coincides with increased gene activity. This mechanism challenges the conventional paradigm that promoter methylation solely silences genes, suggesting that, for specific genes, it might actually activate them. Traditionally, altering DNA methylation in vitro has involved using global demethylating agents, which is insufficient for studying the mechanism and testing the direct consequence of gene methylation changes. To investigate promoter hypermethylation and its association with gene activation, we employed a novel approach utilising a CRISPR-SunTag All-in-one system. Here, we focused on editing the DNA methylation of a specific gene promoter segment (EBF3) in melanoma cells using the All-in-one system. Using bisulfite sequencing and qPCR with RNA-Seq, we successfully demonstrated highly effective methylation and demethylation of the EBF3 promoter, with subsequent gene expression changes, to establish and validate the paradoxical role of DNA methylation. Further, our study provides novel insights into the function of the EBF3 gene, which remains largely unknown. Overall, this study challenges the conventional view of methylation as solely a gene-silencing mechanism and demonstrates a potential function of EBF3 in IFN pathway signalling, potentially uncovering new insights into epigenetic drivers of malignancy and metastasis.
Collapse
Affiliation(s)
- Rakesh Banerjee
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand
| | - Priyadarshana Ajithkumar
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand
| | - Nicholas Keestra
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand
| | - Jim Smith
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand
| | - Gregory Gimenez
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand
| | - Euan J Rodger
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand
| | - Michael R Eccles
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand
| | - Jisha Antony
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand
| | - Robert J Weeks
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand
| | - Aniruddha Chatterjee
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand
- School of Health Sciences and Technology, UPES University, Dehradun 248007, India
| |
Collapse
|
31
|
Cano-Cano F, Martín-Loro F, Gallardo-Orihuela A, González-Montelongo MDC, Ortuño-Miquel S, Hervás-Corpión I, de la Villa P, Ramón-Marco L, Navarro-Calvo J, Gómez-Jaramillo L, Arroba AI, Valor LM. Retinal dysfunction in Huntington's disease mouse models concurs with local gliosis and microglia activation. Sci Rep 2024; 14:4176. [PMID: 38378796 PMCID: PMC10879138 DOI: 10.1038/s41598-024-54347-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 02/12/2024] [Indexed: 02/22/2024] Open
Abstract
Huntington's disease (HD) is caused by an aberrant expansion of CAG repeats in the HTT gene that mainly affects basal ganglia. Although striatal dysfunction has been widely studied in HD mouse models, other brain areas can also be relevant to the pathology. In this sense, we have special interest on the retina as this is the most exposed part of the central nervous system that enable health monitoring of patients using noninvasive techniques. To establish the retina as an appropriate tissue for HD studies, we need to correlate the retinal alterations with those in the inner brain, i.e., striatum. We confirmed the malfunction of the transgenic R6/1 retinas, which underwent a rearrangement of their transcriptome as extensive as in the striatum. Although tissue-enriched genes were downregulated in both areas, a neuroinflammation signature was only clearly induced in the R6/1 retina in which the observed glial activation was reminiscent of the situation in HD patient's brains. The retinal neuroinflammation was confirmed in the slow progressive knock-in zQ175 strain. Overall, these results demonstrated the suitability of the mouse retina as a research model for HD and its associated glial activation.
Collapse
Affiliation(s)
- Fátima Cano-Cano
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Unidad de Investigación, Hospital Universitario Puerta del Mar, Av. Ana de Viya 21, 11009, Cádiz, Spain
| | - Francisco Martín-Loro
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Unidad de Investigación, Hospital Universitario Puerta del Mar, Av. Ana de Viya 21, 11009, Cádiz, Spain
| | - Andrea Gallardo-Orihuela
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Unidad de Investigación, Hospital Universitario Puerta del Mar, Av. Ana de Viya 21, 11009, Cádiz, Spain
| | - María Del Carmen González-Montelongo
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Unidad de Investigación, Hospital Universitario Puerta del Mar, Av. Ana de Viya 21, 11009, Cádiz, Spain
| | - Samanta Ortuño-Miquel
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Unidad de Bioinformática, Hospital General Universitario Dr. Balmis, 03010, Alicante, Spain
| | - Irati Hervás-Corpión
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Unidad de Investigación, Hospital Universitario Puerta del Mar, Av. Ana de Viya 21, 11009, Cádiz, Spain
- Programa de Tumores Sólidos, Centro de Investigación Médica Aplicada (CIMA), Departamento de Pediatría, Clínica Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008, Pamplona, Spain
| | - Pedro de la Villa
- Departamento de Biología de Sistemas, Universidad de Alcalá de Henares, 28871, Alcalá de Henares, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034, Madrid, Spain
| | - Lucía Ramón-Marco
- Laboratorio de Investigación, Diagnostics Building, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Hospital General Universitario Dr. Balmis, Av. Pintor Baeza 12, 03010, Alicante, Spain
| | - Jorge Navarro-Calvo
- Laboratorio de Investigación, Diagnostics Building, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Hospital General Universitario Dr. Balmis, Av. Pintor Baeza 12, 03010, Alicante, Spain
| | - Laura Gómez-Jaramillo
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Unidad de Investigación, Hospital Universitario Puerta del Mar, Av. Ana de Viya 21, 11009, Cádiz, Spain
| | - Ana I Arroba
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Unidad de Investigación, Hospital Universitario Puerta del Mar, Av. Ana de Viya 21, 11009, Cádiz, Spain.
| | - Luis M Valor
- Laboratorio de Investigación, Diagnostics Building, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Hospital General Universitario Dr. Balmis, Av. Pintor Baeza 12, 03010, Alicante, Spain.
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), 03202, Elche, Spain.
| |
Collapse
|
32
|
de Ávila MJR, López-López S, García-Blázquez A, Ruiz-García A, González-Gómez MJ, Nueda ML, Baladrón V, Pérez-Roger I, Poch E, Ballester-Lurbe B, García-Ramírez JJ, Monsalve EM, Díaz-Guerra MJM. RND3 Potentiates Proinflammatory Activation through NOTCH Signaling in Activated Macrophages. J Immunol Res 2024; 2024:2264799. [PMID: 38343633 PMCID: PMC10857877 DOI: 10.1155/2024/2264799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/27/2023] [Accepted: 01/10/2024] [Indexed: 02/15/2024] Open
Abstract
Macrophage activation is a complex process with multiple control elements that ensures an adequate response to the aggressor pathogens and, on the other hand, avoids an excess of inflammatory activity that could cause tissue damage. In this study, we have identified RND3, a small GTP-binding protein, as a new element in the complex signaling process that leads to macrophage activation. We show that RND3 expression is transiently induced in macrophages activated through Toll receptors and potentiated by IFN-γ. We also demonstrate that RND3 increases NOTCH signaling in macrophages by favoring NOTCH1 expression and its nuclear activity; however, Rnd3 expression seems to be inhibited by NOTCH signaling, setting up a negative regulatory feedback loop. Moreover, increased RND3 protein levels seem to potentiate NFκB and STAT1 transcriptional activity resulting in increased expression of proinflammatory genes, such as Tnf-α, Irf-1, or Cxcl-10. Altogether, our results indicate that RND3 seems to be a new regulatory element which could control the activation of macrophages, able to fine tune the inflammatory response through NOTCH.
Collapse
Affiliation(s)
- María José Romero de Ávila
- Medical School, Biomedicine Institute (IB-UCLM)/Biomedicine Unit, University of Castilla-La Mancha/CSIC, C/Almansa 14, 02008, Albacete, Spain
| | - Susana López-López
- Medical School, Biomedicine Institute (IB-UCLM)/Biomedicine Unit, University of Castilla-La Mancha/CSIC, C/Almansa 14, 02008, Albacete, Spain
- Research Unit, University Hospital Complex of Albacete, C/Laurel s/n, 02008, Albacete, Spain
| | - Aarón García-Blázquez
- Medical School, Biomedicine Institute (IB-UCLM)/Biomedicine Unit, University of Castilla-La Mancha/CSIC, C/Almansa 14, 02008, Albacete, Spain
| | - Almudena Ruiz-García
- Biochemistry and Molecular Biology Branch, School of Pharmacy/CRIB/Biomedicine Unit, Department of Inorganic and Organic Chemistry and Biochemistry, University of Castilla-La Mancha/CSIC, Albacete, Spain
| | - María Julia González-Gómez
- Medical School, Biomedicine Institute (IB-UCLM)/Biomedicine Unit, University of Castilla-La Mancha/CSIC, C/Almansa 14, 02008, Albacete, Spain
| | - María Luisa Nueda
- Biochemistry and Molecular Biology Branch, School of Pharmacy/CRIB/Biomedicine Unit, Department of Inorganic and Organic Chemistry and Biochemistry, University of Castilla-La Mancha/CSIC, Albacete, Spain
| | - Victoriano Baladrón
- Medical School, Biomedicine Institute (IB-UCLM)/Biomedicine Unit, University of Castilla-La Mancha/CSIC, C/Almansa 14, 02008, Albacete, Spain
| | - Ignacio Pérez-Roger
- Department of Biomedical Sciences School of Health Sciences, University Cardenal Herrera-CEU, CEU Universities, 46115 Alfara del Patriarca, E-46115 Alfara del Patriarca, Valencia, Spain
| | - Enric Poch
- Department of Biomedical Sciences School of Health Sciences, University Cardenal Herrera-CEU, CEU Universities, 46115 Alfara del Patriarca, E-46115 Alfara del Patriarca, Valencia, Spain
| | - Begoña Ballester-Lurbe
- Department of Biomedical Sciences School of Health Sciences, University Cardenal Herrera-CEU, CEU Universities, 46115 Alfara del Patriarca, E-46115 Alfara del Patriarca, Valencia, Spain
| | - José Javier García-Ramírez
- Medical School, Biomedicine Institute (IB-UCLM)/Biomedicine Unit, University of Castilla-La Mancha/CSIC, C/Almansa 14, 02008, Albacete, Spain
| | - Eva M. Monsalve
- Medical School, Biomedicine Institute (IB-UCLM)/Biomedicine Unit, University of Castilla-La Mancha/CSIC, C/Almansa 14, 02008, Albacete, Spain
| | - María José M. Díaz-Guerra
- Medical School, Biomedicine Institute (IB-UCLM)/Biomedicine Unit, University of Castilla-La Mancha/CSIC, C/Almansa 14, 02008, Albacete, Spain
| |
Collapse
|
33
|
Hwang YS, Lim J, Yoon HR, Park SH, Kim A, Jang JP, Cho HJ, Lee HG. Astragalus Complanatus Ethanol Attenuates Septic Shock by Exerting Anti-Inflammatory Effects on Macrophages. Int J Mol Sci 2023; 25:384. [PMID: 38203555 PMCID: PMC10778658 DOI: 10.3390/ijms25010384] [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: 11/27/2023] [Revised: 12/22/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Sepsis is a systemic inflammatory syndrome that results in multiple-organ failure caused by a dysregulated host immune response to microbial infection. Astragali complanati semen extract (ACSE) exhibits pharmacological activities, including antioxidant, anticancer, antiaging, and anti-diabetes effects. It is widely used in traditional medicine to treat liver and kidney diseases; however, the protective effect of ACSE on sepsis and its mechanisms are unknown. In the present study, we investigated the anti-inflammatory effects and potential mechanisms of the action of ACSE on sepsis. We show that ACSE improved survival rates in mouse models of acute sepsis induced by CLP (cecal ligation and puncture) and LPS stimulation. ACSE administration decreased aspartate aminotransferase (AST) and alanine aminotransferase (ALT) in sepsis-induced mice. Furthermore, ACSE reduced the levels of nitric oxide (NO), tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6) in the serum of septic mice. ACSE treatment inhibited the expression of these proinflammatory genes in LPS-stimulated J774 macrophages. Moreover, ACSE inhibited the phosphorylation of the IκB kinase (IKK) and the nuclear translocation of p65 NF-κB by LPS stimulation in macrophages. These results reveal the mechanism underlying the protective effect of ACSE against sepsis by inhibiting NF-κB activation and suggest that ACSE could be a potential therapeutic candidate to treat acute inflammatory diseases.
Collapse
Affiliation(s)
- Yo Sep Hwang
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon 34141, Republic of Korea; (Y.S.H.); (J.L.); (H.R.Y.)
| | - Jeewon Lim
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon 34141, Republic of Korea; (Y.S.H.); (J.L.); (H.R.Y.)
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Hyang Ran Yoon
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon 34141, Republic of Korea; (Y.S.H.); (J.L.); (H.R.Y.)
| | - Seong-Hoon Park
- Genetic and Epigenetic Toxicology Research Group, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea;
| | - Aeyung Kim
- Korean Medicine Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea;
| | - Jun-Pil Jang
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea;
| | - Hee Jun Cho
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon 34141, Republic of Korea; (Y.S.H.); (J.L.); (H.R.Y.)
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Hee Gu Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon 34141, Republic of Korea; (Y.S.H.); (J.L.); (H.R.Y.)
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Yuseong-gu, Daejeon 34113, Republic of Korea
| |
Collapse
|
34
|
Ji L, Li T, Chen H, Yang Y, Lu E, Liu J, Qiao W, Chen H. The crucial regulatory role of type I interferon in inflammatory diseases. Cell Biosci 2023; 13:230. [PMID: 38124132 PMCID: PMC10734085 DOI: 10.1186/s13578-023-01188-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/16/2023] [Indexed: 12/23/2023] Open
Abstract
Type I interferon (IFN-I) plays crucial roles in the regulation of inflammation and it is associated with various inflammatory diseases including systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), and periodontitis, impacting people's health and quality of life. It is well-established that IFN-Is affect immune responses and inflammatory factors by regulating some signaling. However, currently, there is no comprehensive overview of the crucial regulatory role of IFN-I in distinctive pathways as well as associated inflammatory diseases. This review aims to provide a narrative of the involvement of IFN-I in different signaling pathways, mainly mediating the related key factors with specific targets in the pathways and signaling cascades to influence the progression of inflammatory diseases. As such, we suggested that IFN-Is induce inflammatory regulation through the stimulation of certain factors in signaling pathways, which displays possible efficient treatment methods and provides a reference for the precise control of inflammatory diseases.
Collapse
Affiliation(s)
- Ling Ji
- Faculty of Dentistry, The University of Hong Kong, Prince Philip Dental Hospital, Hong Kong, SAR, People's Republic of China
| | - Tianle Li
- Faculty of Dentistry, The University of Hong Kong, Prince Philip Dental Hospital, Hong Kong, SAR, People's Republic of China
| | - Huimin Chen
- Faculty of Dentistry, The University of Hong Kong, Prince Philip Dental Hospital, Hong Kong, SAR, People's Republic of China
| | - Yanqi Yang
- Faculty of Dentistry, The University of Hong Kong, Prince Philip Dental Hospital, Hong Kong, SAR, People's Republic of China
- Division of Pediatric Dentistry and Orthodontics, Faculty of Dentistry, The University of Hong Kong, Prince Philip Dental Hospital, Hong Kong, SAR, People's Republic of China
| | - Eryi Lu
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, China
| | - Jieying Liu
- Department of Medical Research Center, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Qiao
- Faculty of Dentistry, The University of Hong Kong, Prince Philip Dental Hospital, Hong Kong, SAR, People's Republic of China.
- Applied Oral Sciences & Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Prince Philip Dental Hospital, Level 3, 34 Hospital Road, Sai Ying Pun, Hong Kong, SAR, People's Republic of China.
| | - Hui Chen
- Faculty of Dentistry, The University of Hong Kong, Prince Philip Dental Hospital, Hong Kong, SAR, People's Republic of China.
- Division of Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Prince Philip Dental Hospital, Level 3, 34 Hospital Road, Sai Ying Pun, Hong Kong, SAR, People's Republic of China.
| |
Collapse
|
35
|
Jin M, Fang J, Wang JJ, Shao X, Xu SW, Liu PQ, Ye WC, Liu ZP. Regulation of toll-like receptor (TLR) signaling pathways in atherosclerosis: from mechanisms to targeted therapeutics. Acta Pharmacol Sin 2023; 44:2358-2375. [PMID: 37550526 PMCID: PMC10692204 DOI: 10.1038/s41401-023-01123-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 06/04/2023] [Indexed: 08/09/2023]
Abstract
Atherosclerosis, one of the life-threatening cardiovascular diseases (CVDs), has been demonstrated to be a chronic inflammatory disease, and inflammatory and immune processes are involved in the origin and development of the disease. Toll-like receptors (TLRs), a class of pattern recognition receptors that trigger innate immune responses by identifying pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs), regulate numerous acute and chronic inflammatory diseases. Recent studies reveal that TLRs have a vital role in the occurrence and development of atherosclerosis, including the initiation of endothelial dysfunction, interaction of various immune cells, and activation of a number of other inflammatory pathways. We herein summarize some other inflammatory signaling pathways, protein molecules, and cellular responses associated with TLRs, such as NLRP3, Nrf2, PCSK9, autophagy, pyroptosis and necroptosis, which are also involved in the development of AS. Targeting TLRs and their regulated inflammatory events could be a promising new strategy for the treatment of atherosclerotic CVDs. Novel drugs that exert therapeutic effects on AS through TLRs and their related pathways are increasingly being developed. In this article, we comprehensively review the current knowledge of TLR signaling pathways in atherosclerosis and actively seek potential therapeutic strategies using TLRs as a breakthrough point in the prevention and therapy of atherosclerosis.
Collapse
Affiliation(s)
- Mei Jin
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511436, China
| | - Jian Fang
- Affiliated Huadu Hospital, Southern Medical University (People's Hospital of Huadu District), Guangzhou, 510800, China
| | - Jiao-Jiao Wang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511436, China
| | - Xin Shao
- Department of Food Science and Engineering, Jinan University, Guangzhou, 511436, China
| | - Suo-Wen Xu
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Pei-Qing Liu
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511436, China.
- National-Local Joint Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Wen-Cai Ye
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511436, China.
| | - Zhi-Ping Liu
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511436, China.
| |
Collapse
|
36
|
Boyer K, Li L, Li T, Zhang B, Zhao G. MORA and EnsembleTFpredictor: An ensemble approach to reveal functional transcription factor regulatory networks. PLoS One 2023; 18:e0294724. [PMID: 38032891 PMCID: PMC10688744 DOI: 10.1371/journal.pone.0294724] [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: 05/16/2023] [Accepted: 11/06/2023] [Indexed: 12/02/2023] Open
Abstract
MOTIVATION Our study aimed to identify biologically relevant transcription factors (TFs) that control the expression of a set of co-expressed or co-regulated genes. RESULTS We developed a fully automated pipeline, Motif Over Representation Analysis (MORA), to detect enrichment of known TF binding motifs in any query sequences. MORA performed better than or comparable to five other TF-prediction tools as evaluated using hundreds of differentially expressed gene sets and ChIP-seq datasets derived from known TFs. Additionally, we developed EnsembleTFpredictor to harness the power of multiple TF-prediction tools to provide a list of functional TFs ranked by prediction confidence. When applied to the test datasets, EnsembleTFpredictor not only identified the target TF but also revealed many TFs known to cooperate with the target TF in the corresponding biological systems. MORA and EnsembleTFpredictor have been used in two publications, demonstrating their power in guiding experimental design and in revealing novel biological insights.
Collapse
Affiliation(s)
- Kevin Boyer
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Louis Li
- Brown University, Providence, RI, United States of America
| | - Tiandao Li
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Bo Zhang
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Guoyan Zhao
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, United States of America
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States of America
| |
Collapse
|
37
|
Pouyabahar D, Chung SW, Pezzutti OI, Perciani CT, Wang X, Ma XZ, Jiang C, Camat D, Chung T, Sekhon M, Manuel J, Chen XC, McGilvray ID, MacParland SA, Bader GD. A rat liver cell atlas reveals intrahepatic myeloid heterogeneity. iScience 2023; 26:108213. [PMID: 38026201 PMCID: PMC10651689 DOI: 10.1016/j.isci.2023.108213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 08/20/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
The large size and vascular accessibility of the laboratory rat (Rattus norvegicus) make it an ideal hepatic animal model for diseases that require surgical manipulation. Often, the disease susceptibility and outcomes of inflammatory pathologies vary significantly between strains. This study uses single-cell transcriptomics to better understand the complex cellular network of the rat liver, as well as to unravel the cellular and molecular sources of inter-strain hepatic variation. We generated single-cell and single-nucleus transcriptomic maps of the livers of healthy Dark Agouti and Lewis rat strains and developed a factor analysis-based bioinformatics analysis pipeline to study data covariates, such as strain and batch. Using this approach, we discovered transcriptomic variation within the hepatocyte and myeloid populations that underlie distinct cell states between rat strains. This finding will help provide a reference for future investigations on strain-dependent outcomes of surgical experiment models.
Collapse
Affiliation(s)
- Delaram Pouyabahar
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Sai W. Chung
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Olivia I. Pezzutti
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Catia T. Perciani
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Xinle Wang
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Xue-Zhong Ma
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Chao Jiang
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Damra Camat
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Trevor Chung
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Manmeet Sekhon
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Justin Manuel
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Xu-Chun Chen
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Ian D. McGilvray
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Sonya A. MacParland
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Gary D. Bader
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada
- Princess Margaret Research Institute, University Health Network, Toronto, ON, Canada
- The Multiscale Human Program, Canadian Institute for Advanced Research, Toronto, ON, Canada
| |
Collapse
|
38
|
Tang Y, Kim JY, Ip CKM, Bahmani A, Chen Q, Rosenberger MG, Esser-Kahn AP, Ferguson AL. Data-driven discovery of innate immunomodulators via machine learning-guided high throughput screening. Chem Sci 2023; 14:12747-12766. [PMID: 38020385 PMCID: PMC10646978 DOI: 10.1039/d3sc03613h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
The innate immune response is vital for the success of prophylactic vaccines and immunotherapies. Control of signaling in innate immune pathways can improve prophylactic vaccines by inhibiting unfavorable systemic inflammation and immunotherapies by enhancing immune stimulation. In this work, we developed a machine learning-enabled active learning pipeline to guide in vitro experimental screening and discovery of small molecule immunomodulators that improve immune responses by altering the signaling activity of innate immune responses stimulated by traditional pattern recognition receptor agonists. Molecules were tested by in vitro high throughput screening (HTS) where we measured modulation of the nuclear factor κ-light-chain-enhancer of activated B-cells (NF-κB) and the interferon regulatory factors (IRF) pathways. These data were used to train data-driven predictive models linking molecular structure to modulation of the NF-κB and IRF responses using deep representational learning, Gaussian process regression, and Bayesian optimization. By interleaving successive rounds of model training and in vitro HTS, we performed an active learning-guided traversal of a 139 998 molecule library. After sampling only ∼2% of the library, we discovered viable molecules with unprecedented immunomodulatory capacity, including those capable of suppressing NF-κB activity by up to 15-fold, elevating NF-κB activity by up to 5-fold, and elevating IRF activity by up to 6-fold. We extracted chemical design rules identifying particular chemical fragments as principal drivers of specific immunomodulation behaviors. We validated the immunomodulatory effect of a subset of our top candidates by measuring cytokine release profiles. Of these, one molecule induced a 3-fold enhancement in IFN-β production when delivered with a cyclic di-nucleotide stimulator of interferon genes (STING) agonist. In sum, our machine learning-enabled screening approach presents an efficient immunomodulator discovery pipeline that has furnished a library of novel small molecules with a strong capacity to enhance or suppress innate immune signaling pathways to shape and improve prophylactic vaccination and immunotherapies.
Collapse
Affiliation(s)
- Yifeng Tang
- Pritzker School of Molecular Engineering, University of Chicago Chicago IL 60637 USA
| | - Jeremiah Y Kim
- Pritzker School of Molecular Engineering, University of Chicago Chicago IL 60637 USA
| | - Carman K M Ip
- Cellular Screening Center, University of Chicago Chicago IL 60637 USA
| | - Azadeh Bahmani
- Cellular Screening Center, University of Chicago Chicago IL 60637 USA
| | - Qing Chen
- Pritzker School of Molecular Engineering, University of Chicago Chicago IL 60637 USA
| | - Matthew G Rosenberger
- Pritzker School of Molecular Engineering, University of Chicago Chicago IL 60637 USA
| | - Aaron P Esser-Kahn
- Pritzker School of Molecular Engineering, University of Chicago Chicago IL 60637 USA
| | - Andrew L Ferguson
- Pritzker School of Molecular Engineering, University of Chicago Chicago IL 60637 USA
| |
Collapse
|
39
|
O’Neil JD, Bolimowska OO, Clayton SA, Tang T, Daley KK, Lara-Reyna S, Warner J, Martin CS, Mahida RY, Hardy RS, Arthur JSC, Clark AR. Dexamethasone impairs the expression of antimicrobial mediators in lipopolysaccharide-activated primary macrophages by inhibiting both expression and function of interferon β. Front Immunol 2023; 14:1190261. [PMID: 37942320 PMCID: PMC10628473 DOI: 10.3389/fimmu.2023.1190261] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 10/11/2023] [Indexed: 11/10/2023] Open
Abstract
Glucocorticoids potently inhibit expression of many inflammatory mediators, and have been widely used to treat both acute and chronic inflammatory diseases for more than seventy years. However, they can have several unwanted effects, amongst which immunosuppression is one of the most common. Here we used microarrays and proteomic approaches to characterise the effect of dexamethasone (a synthetic glucocorticoid) on the responses of primary mouse macrophages to a potent pro-inflammatory agonist, lipopolysaccharide (LPS). Gene ontology analysis revealed that dexamethasone strongly impaired the lipopolysaccharide-induced antimicrobial response, which is thought to be driven by an autocrine feedback loop involving the type I interferon IFNβ. Indeed, dexamethasone strongly and dose-dependently inhibited the expression of IFNβ by LPS-activated macrophages. Unbiased proteomic data also revealed an inhibitory effect of dexamethasone on the IFNβ-dependent program of gene expression, with strong down-regulation of several interferon-induced antimicrobial factors. Surprisingly, dexamethasone also inhibited the expression of several antimicrobial genes in response to direct stimulation of macrophages with IFNβ. We tested a number of hypotheses based on previous publications, but found that no single mechanism could account for more than a small fraction of the broad suppressive impact of dexamethasone on macrophage type I interferon signaling, underlining the complexity of this pathway. Preliminary experiments indicated that dexamethasone exerted similar inhibitory effects on primary human monocyte-derived or alveolar macrophages.
Collapse
Affiliation(s)
- John D. O’Neil
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Oliwia O. Bolimowska
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Sally A. Clayton
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Tina Tang
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Kalbinder K. Daley
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Samuel Lara-Reyna
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, United Kingdom
| | - Jordan Warner
- School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Claire S. Martin
- School of Biomedical Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Rahul Y. Mahida
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Rowan S. Hardy
- School of Biomedical Sciences, University of Birmingham, Birmingham, United Kingdom
| | | | - Andrew R. Clark
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
40
|
Hashimoto R, Koide H, Katoh Y. MEK inhibitors increase the mortality rate in mice with LPS-induced inflammation through IL-12-NO signaling. Cell Death Discov 2023; 9:374. [PMID: 37833247 PMCID: PMC10575927 DOI: 10.1038/s41420-023-01674-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 09/11/2023] [Accepted: 10/03/2023] [Indexed: 10/15/2023] Open
Abstract
Lipopolysaccharide (LPS) is an endotoxin that can cause an acute inflammatory response. Nitric oxide (NO) is one of the most important innate immune system components and is synthesized by inducible NOS (iNOS) in macrophages in response to stimulation with LPS. LPS activates the RAS-RAF-mitogen-activated protein kinase/ERK kinase (MEK)-extracellular-signal-regulated kinase (ERK) signaling cascade in macrophages. The purpose of this study was to examine how the combination of LPS and MEK inhibitors, which have been used as anticancer agents in recent years, affects inflammation. We showed that MEK inhibitors enhanced iNOS expression and NO production in LPS-stimulated mouse bone marrow-derived macrophages. A MEK inhibitor increased the mortality rate in mice with LPS-induced inflammation. The expression of the cytokine interleukin-12 (IL-12) in macrophages was enhanced by the MEK inhibitor, as shown by a cytokine array and ELISA. IL-12 enhanced iNOS expression and NO production in response to LPS. We also showed that tumor necrosis factor (TNF-α) was secreted by macrophage after stimulation with LPS and that TNF-α and IL-12 synergistically induced iNOS expression and NO production. An anti-IL-12 neutralizing antibody prevented NO production and mortality in an LPS-induced inflammation mouse model in the presence of a MEK inhibitor. These results suggest that the MEK inhibitor increases the mortality rate in mice with LPS-induced inflammation through IL-12-NO signaling.
Collapse
Affiliation(s)
- Ryota Hashimoto
- Laboratory of Cell Biology, Biomedical Research Core Facilities, Juntendo University Graduate School of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan.
- Department of Physiology, Juntendo University Faculty of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan.
| | - Hiroshi Koide
- Laboratory of Molecular and Biochemical Research, Biomedical Research Core Facilities, Juntendo University Graduate School of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Youichi Katoh
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan.
- Juntendo University Faculty of International Liberal Arts, Hongo 2-1-1, Bunkyo-ku, Tokyo, 112-8421, Japan.
| |
Collapse
|
41
|
López-López S, Romero de Ávila MJ, González-Gómez MJ, Nueda ML, Baladrón V, Monsalve EM, García-Ramírez JJ, Díaz-Guerra MJM. NOTCH4 potentiates the IL-13 induced genetic program in M2 alternative macrophages through the AP1 and IRF4-JMJD3 axis. Int Immunol 2023; 35:497-509. [PMID: 37478314 DOI: 10.1093/intimm/dxad028] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/19/2023] [Indexed: 07/23/2023] Open
Abstract
IL-13 signaling polarizes macrophages to an M2 alternatively activated phenotype, which regulates tissue repair and anti-inflammatory responses. However, an excessive activation of this pathway leads to severe pathologies, such as allergic airway inflammation and asthma. In this work, we identified NOTCH4 receptor as an important modulator of M2 macrophage activation. We show that the expression of NOTCH4 is induced by IL-13, mediated by Janus kinases and AP1 activity, probably mediated by the IL-13Rα1 and IL-13Rα2 signaling pathway. Furthermore, we demonstrate an important role for NOTCH4 signaling in the IL-13 induced gene expression program in macrophages, including various genes that contribute to pathogenesis of the airways in asthma, such as ARG1, YM1, CCL24, IL-10, or CD-163. We also demonstrate that NOTCH4 signaling modulates IL-13-induced gene expression by increasing IRF4 activity, mediated, at least in part, by the expression of the histone H3K27me3 demethylase JMJD3, and by increasing AP1-dependent transcription. In summary, our results provide evidence for an important role of NOTCH4 signaling in alternative activation of macrophages by IL-13 and suggest that NOTCH4 may contribute to the increased severity of lesions in M2 inflammatory responses, such as allergic asthma, which points to NOTCH4 as a potential new target for the treatment of these pathologies.
Collapse
Affiliation(s)
- Susana López-López
- CRIB/Biomedicine Unit, Medical School, University of Castilla-La Mancha/CSIC, C/Almansa 14, 02008 Albacete, Spain
- Research Unit, Complejo Hospitalario Universitario de Albacete, C/Laurel, s/n, 02008 Albacete, Spain
| | - María José Romero de Ávila
- CRIB/Biomedicine Unit, Medical School, University of Castilla-La Mancha/CSIC, C/Almansa 14, 02008 Albacete, Spain
| | - María Julia González-Gómez
- CRIB/Biomedicine Unit, Medical School, University of Castilla-La Mancha/CSIC, C/Almansa 14, 02008 Albacete, Spain
| | - María Luisa Nueda
- Biochemistry and Molecular Biology Branch, School of Pharmacy/CRIB/Biomedicine Unit, Department of Inorganic and Organic Chemistry and Biochemistry, University of Castilla-La Mancha/CSIC, Albacete, Spain
| | - Victoriano Baladrón
- CRIB/Biomedicine Unit, Medical School, University of Castilla-La Mancha/CSIC, C/Almansa 14, 02008 Albacete, Spain
| | - Eva M Monsalve
- CRIB/Biomedicine Unit, Medical School, University of Castilla-La Mancha/CSIC, C/Almansa 14, 02008 Albacete, Spain
| | - José Javier García-Ramírez
- CRIB/Biomedicine Unit, Medical School, University of Castilla-La Mancha/CSIC, C/Almansa 14, 02008 Albacete, Spain
| | - María José M Díaz-Guerra
- CRIB/Biomedicine Unit, Medical School, University of Castilla-La Mancha/CSIC, C/Almansa 14, 02008 Albacete, Spain
| |
Collapse
|
42
|
Allen-Gondringer A, Gau D, Varghese C, Boone D, Stolz D, Larregina A, Roy P. Vascular endothelial cell-specific disruption of the profilin1 gene leads to severe multiorgan pathology and inflammation causing mortality. PNAS NEXUS 2023; 2:pgad305. [PMID: 37781098 PMCID: PMC10541205 DOI: 10.1093/pnasnexus/pgad305] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 09/11/2023] [Indexed: 10/03/2023]
Abstract
Actin-binding protein Profilin1 is an important regulator of actin cytoskeletal dynamics in cells and critical for embryonic development in higher eukaryotes. The objective of the present study was to examine the consequence of loss-of-function of Pfn1 in vascular endothelial cells (ECs) in vivo. We utilized a mouse model engineered for tamoxifen-inducible biallelic inactivation of the Pfn1 gene selectively in EC (Pfn1EC-KO). Widespread deletion of EC Pfn1 in adult mice leads to severe health complications presenting overt pathologies (endothelial cell death, infarct, and fibrosis) in major organ systems and evidence for inflammatory infiltrates, ultimately compromising the survival of animals within 3 weeks of gene ablation. Mice deficient in endothelial Pfn1 exhibit selective bias toward the proinflammatory myeloid-derived population of immune cells, a finding further supported by systemic elevation of proinflammatory cytokines. We further show that triggering Pfn1 depletion not only directly upregulates proinflammatory cytokine/chemokine gene expression in EC but also potentiates the paracrine effect of EC on proinflammatory gene expression in macrophages. Consistent with these findings, we provide further evidence for increased activation of Interferon Regulatory Factor 7 (IRF7) and STAT1 in EC when depleted of Pfn1. Collectively, these findings for the first time demonstrate a prominent immunological consequence of loss of endothelial Pfn1 and an indispensable role of endothelial Pfn1 in mammalian survival unlike tolerable phenotypes of Pfn1 loss in other differentiated cell types.
Collapse
Affiliation(s)
| | - David Gau
- Bioengineering, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | | | - David Boone
- Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15206, USA
| | - Donna Stolz
- Cell Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Adriana Larregina
- Dermatology, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Partha Roy
- Bioengineering, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
43
|
Naigles B, Narla AV, Soroczynski J, Tsimring LS, Hao N. Quantifying dynamic pro-inflammatory gene expression and heterogeneity in single macrophage cells. J Biol Chem 2023; 299:105230. [PMID: 37689116 PMCID: PMC10579967 DOI: 10.1016/j.jbc.2023.105230] [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: 05/12/2023] [Revised: 09/02/2023] [Accepted: 09/03/2023] [Indexed: 09/11/2023] Open
Abstract
Macrophages must respond appropriately to pathogens and other pro-inflammatory stimuli in order to perform their roles in fighting infection. One way in which inflammatory stimuli can vary is in their dynamics-that is, the amplitude and duration of stimulus experienced by the cell. In this study, we performed long-term live cell imaging in a microfluidic device to investigate how the pro-inflammatory genes IRF1, CXCL10, and CXCL9 respond to dynamic interferon-gamma (IFNγ) stimulation. We found that IRF1 responds to low concentration or short duration IFNγ stimulation, whereas CXCL10 and CXCL9 require longer or higherconcentration stimulation to be expressed. We also investigated the heterogeneity in the expression of each gene and found that CXCL10 and CXCL9 have substantial cell-to-cell variability. In particular, the expression of CXCL10 appears to be largely stochastic with a subpopulation of nonresponding cells across all the stimulation conditions tested. We developed both deterministic and stochastic models for the expression of each gene. Our modeling analysis revealed that the heterogeneity in CXCL10 can be attributed to a slow chromatin-opening step that is on a similar timescale to that of adaptation of the upstream signal. In this way, CXCL10 expression in individual cells can remain stochastic in response to each pulse of repeated stimulation, which we also validated by experiments. Together, we conclude that pro-inflammatory genes in the same signaling pathway can respond to dynamic IFNγ stimulus with very different response features and that upstream signal adaptation can contribute to shaping heterogeneous gene expression.
Collapse
Affiliation(s)
- Beverly Naigles
- Department of Molecular Biology, University of California San Diego, La Jolla, California, USA
| | - Avaneesh V Narla
- Department of Physics, University of California San Diego, La Jolla, California, USA
| | - Jan Soroczynski
- Laboratory of Genome Architecture and Dynamics, The Rockefeller University, New York, New York, USA
| | - Lev S Tsimring
- Synthetic Biology Institute, University of California San Diego, La Jolla, California, USA
| | - Nan Hao
- Department of Molecular Biology, University of California San Diego, La Jolla, California, USA; Synthetic Biology Institute, University of California San Diego, La Jolla, California, USA; Department of Bioengineering, University of California San Diego, La Jolla, California, USA.
| |
Collapse
|
44
|
Khiter F, Kherrouche Z, Dubois V, Slupek S, Petit E, Debrie AS, Cauchi S, Barois N, Rouanet C, Mielcarek N. Combined regulation of pro-inflammatory cytokines production by STAT3 and STAT5 in a model of B. pertussis infection of alveolar macrophages. Front Immunol 2023; 14:1254276. [PMID: 37841236 PMCID: PMC10569487 DOI: 10.3389/fimmu.2023.1254276] [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: 07/06/2023] [Accepted: 09/14/2023] [Indexed: 10/17/2023] Open
Abstract
Bordetella pertussis is a highly contagious respiratory pathogen responsible for whooping-cough or pertussis. Despite high vaccination coverage worldwide, this gram-negative bacterium continues to spread among the population. B. pertussis is transmitted by aerosol droplets from an infected individual to a new host and will colonize its upper respiratory tract. Alveolar macrophages (AMs) are effector cells of the innate immune system that phagocytose B. pertussis and secrete both pro-inflammatory and antimicrobial mediators in the lungs. However, understanding their role in B. pertussis pathogenesis at the molecular level is hampered by the limited number of primary AMs that can be collected in vivo. In order to decipher the regulation of innate response induced by B. pertussis infection, we used for the first time self-renewing, non-transformed cells, called Max Planck Institute (MPI) cells, which are phenotypically and functionally very close to pulmonary AMs. Using optimized infection conditions, we characterized the entry and the clearance of B. pertussis within MPI macrophages. We showed that under these conditions, MPI cells exhibit a pro-inflammatory phenotype with the production of TNF, IL-1β, IL-6 and MIP-2α, similarly to primary AMs purified from broncho-alveolar fluids of mice. In addition, we explored the yet uncharacterized role of the signal transduction activator of transcription (STAT) proteins family in the innate immune response to B. pertussis infection and showed for the first time the parallel regulation of pro-inflammatory cytokines by STAT3 and STAT5 in MPI macrophages infected by B. pertussis. Altogether, this work highlights the interest of using MPI cells for experiments optimization and preliminary data acquisition to understand B. pertussis interaction with AMs, and thus significantly reduce the number of animals to be sacrificed.
Collapse
Affiliation(s)
- Fethi Khiter
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, Lille, France
| | - Zoulika Kherrouche
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille, France
| | - Violaine Dubois
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, Lille, France
| | - Stéphanie Slupek
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, Lille, France
| | - Emmanuelle Petit
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, Lille, France
| | - Anne-Sophie Debrie
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, Lille, France
| | - Stéphane Cauchi
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, Lille, France
| | - Nicolas Barois
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, Lille, France
| | - Carine Rouanet
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, Lille, France
| | - Nathalie Mielcarek
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, Lille, France
| |
Collapse
|
45
|
Gao Y, Na M, Yao X, Li C, Li L, Yang G, Li Y, Hu Y. Integrative single-cell transcriptomic investigation unveils long non-coding RNAs associated with localized cellular inflammation in psoriasis. Front Immunol 2023; 14:1265517. [PMID: 37822943 PMCID: PMC10562854 DOI: 10.3389/fimmu.2023.1265517] [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: 07/23/2023] [Accepted: 09/11/2023] [Indexed: 10/13/2023] Open
Abstract
Psoriasis is a complex, chronic autoimmune disorder predominantly affecting the skin. Accumulating evidence underscores the critical role of localized cellular inflammation in the development and persistence of psoriatic skin lesions, involving cell types such as keratinocytes, mesenchymal cells, and Schwann cells. However, the underlying mechanisms remain largely unexplored. Long non-coding RNAs (lncRNAs), known to regulate gene expression across various cellular processes, have been particularly implicated in immune regulation. We utilized our neural-network learning pipeline to integrate 106,675 cells from healthy human skin and 79,887 cells from psoriatic human skin. This formed the most extensive cell transcriptomic atlas of human psoriatic skin to date. The robustness of our reclassified cell-types, representing full-layer zonation in human skin, was affirmed through neural-network learning-based cross-validation. We then developed a publicly available website to present this integrated dataset. We carried out analysis for differentially expressed lncRNAs, co-regulated gene patterns, and GO-bioprocess enrichment, enabling us to pinpoint lncRNAs that modulate localized cellular inflammation in psoriasis at the single-cell level. Subsequent experimental validation with skin cell lines and primary cells from psoriatic skin confirmed these lncRNAs' functional role in localized cellular inflammation. Our study provides a comprehensive cell transcriptomic atlas of full-layer human skin in both healthy and psoriatic conditions, unveiling a new regulatory mechanism that governs localized cellular inflammation in psoriasis and highlights the therapeutic potential of lncRNAs in this disease's management.
Collapse
Affiliation(s)
- Yuge Gao
- Department of Dermatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Mengxue Na
- Department of Dermatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinyu Yao
- Department of Dermatology, Peking University First Hospital, Beijing, China
| | - Chao Li
- Department of Dermatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Li Li
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guangyu Yang
- Department of Dermatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuzhen Li
- Department of Dermatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yizhou Hu
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
46
|
Gomes I, Gallego-Paez LM, Jiménez M, Santamaria PG, Mansinho A, Sousa R, Abreu C, Suárez EG, Costa L, Casimiro S. Co-targeting RANK pathway treats and prevents acquired resistance to CDK4/6 inhibitors in luminal breast cancer. Cell Rep Med 2023; 4:101120. [PMID: 37451269 PMCID: PMC10439176 DOI: 10.1016/j.xcrm.2023.101120] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 05/11/2023] [Accepted: 06/21/2023] [Indexed: 07/18/2023]
Abstract
The combination of endocrine therapy (ET) and cyclin-dependent kinase 4/6 (CDK4/6) inhibitors (CDK4/6i) was a hallmark in metastatic luminal breast cancer (BC). However, intrinsic and acquired resistance affects long-term efficacy. Here, we study the role of the receptor activator of nuclear factor-κB (RANK) pathway in CDK4/6i resistance. We find that RANK overexpression in luminal BC is associated with intrinsic resistance to CDK4/6i, both in vitro and in mouse xenografts, and decreased proliferation rate and chronic interferon (IFN) γ response are highlighted as resistance drivers. Gene expression data from the NeoPalAna CDK4/6i clinical trial, and studies with palbociclib-resistant cell lines, show that RANK is upregulated after treatment with CDK4/6i, supporting a role in acquired resistance. Our study shows that RANK ligand (RANKL) inhibitors can restore sensitivity to CDK4/6i and prevent acquired resistance. On the basis of these findings, we conclude that pharmacological inhibition of the RANK pathway through RANKL blocking could represent an add-on to ET + CDK4/6i, warranting further clinical studies.
Collapse
Affiliation(s)
- Inês Gomes
- Luis Costa Laboratory, Institute of Molecular Medicine (iMM), Lisbon Medical School, 1649-028 Lisbon, Portugal
| | - Lina M Gallego-Paez
- Luis Costa Laboratory, Institute of Molecular Medicine (iMM), Lisbon Medical School, 1649-028 Lisbon, Portugal
| | - Maria Jiménez
- Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | | | - André Mansinho
- Oncology Division, Hospital de Santa Maria-CHULN, 1649-028 Lisbon, Portugal
| | - Rita Sousa
- Oncology Division, Hospital de Santa Maria-CHULN, 1649-028 Lisbon, Portugal
| | - Catarina Abreu
- Oncology Division, Hospital de Santa Maria-CHULN, 1649-028 Lisbon, Portugal
| | - Eva González Suárez
- Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain; Oncobell, Bellvitge Biomedical Research Institute, IDIBELL, 08908 Barcelona, Spain
| | - Luis Costa
- Luis Costa Laboratory, Institute of Molecular Medicine (iMM), Lisbon Medical School, 1649-028 Lisbon, Portugal; Oncology Division, Hospital de Santa Maria-CHULN, 1649-028 Lisbon, Portugal.
| | - Sandra Casimiro
- Luis Costa Laboratory, Institute of Molecular Medicine (iMM), Lisbon Medical School, 1649-028 Lisbon, Portugal.
| |
Collapse
|
47
|
Cui Y, Zhang H, Wang Z, Gong B, Al-Ward H, Deng Y, Fan O, Wang J, Zhu W, Sun YE. Exploring the shared molecular mechanisms between systemic lupus erythematosus and primary Sjögren's syndrome based on integrated bioinformatics and single-cell RNA-seq analysis. Front Immunol 2023; 14:1212330. [PMID: 37614232 PMCID: PMC10442653 DOI: 10.3389/fimmu.2023.1212330] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 07/19/2023] [Indexed: 08/25/2023] Open
Abstract
Background Systemic lupus erythematosus (SLE) and primary Sjögren's syndrome (pSS) are common systemic autoimmune diseases that share a wide range of clinical manifestations and serological features. This study investigates genes, signaling pathways, and transcription factors (TFs) shared between SLE and pSS. Methods Gene expression profiles of SLE and pSS were obtained from the Gene Expression Omnibus (GEO). Weighted gene co-expression network analysis (WGCNA) and differentially expressed gene (DEG) analysis were conducted to identify shared genes related to SLE and pSS. Overlapping genes were then subject to Gene Ontology (GO) and protein-protein interaction (PPI) network analyses. Cytoscape plugins cytoHubba and iRegulon were subsequently used to screen shared hub genes and predict TFs. In addition, gene set variation analysis (GSVA) and CIBERSORTx were used to calculate the correlations between hub genes and immune cells as well as related pathways. To confirm these results, hub genes and TFs were verified in microarray and single-cell RNA sequencing (scRNA-seq) datasets. Results Following WGCNA and limma analysis, 152 shared genes were identified. These genes were involved in interferon (IFN) response and cytokine-mediated signaling pathway. Moreover, we screened six shared genes, namely IFI44L, ISG15, IFIT1, USP18, RSAD2 and ITGB2, out of which three genes, namely IFI44L, ISG15 and ITGB2 were found to be highly expressed in both microarray and scRNA-seq datasets. IFN response and ITGB2 signaling pathway were identified as potentially relevant pathways. In addition, STAT1 and IRF7 were identified as common TFs in both diseases. Conclusion This study revealed IFI44L, ISG15 and ITGB2 as the shared genes and identified STAT1 and IRF7 as the common TFs of SLE and pSS. Notably, the IFN response and ITGB2 signaling pathway played vital roles in both diseases. Our study revealed common pathogenetic characteristics of SLE and pSS. The particular roles of these pivotal genes and mutually overlapping pathways may provide a basis for further mechanistic research.
Collapse
Affiliation(s)
- Yanling Cui
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Huina Zhang
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhen Wang
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bangdong Gong
- Division of Rheumatology, Tongji Hospital of Tongji University School of Medicine, Shanghai, China
| | - Hisham Al-Ward
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yaxuan Deng
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Orion Fan
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Junbang Wang
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wenmin Zhu
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yi Eve Sun
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
48
|
Czimmerer Z, Nagy L. Epigenomic regulation of macrophage polarization: Where do the nuclear receptors belong? Immunol Rev 2023; 317:152-165. [PMID: 37074820 PMCID: PMC10524119 DOI: 10.1111/imr.13209] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 04/20/2023]
Abstract
Our laboratory has a long-standing research interest in understanding how lipid-activated transcription factors, nuclear hormone receptors, contribute to dendritic cell and macrophage gene expression regulation, subtype specification, and responses to a changing extra and intracellular milieu. This journey in the last more than two decades took us from identifying target genes for various RXR heterodimers to systematically mapping nuclear receptor-mediated pathways in dendritic cells to identifying hierarchies of transcription factors in alternative polarization in macrophages to broaden the role of nuclear receptors beyond strictly ligand-regulated gene expression. We detail here the milestones of the road traveled and draw conclusions regarding the unexpectedly broad role of nuclear hormone receptors as epigenomic components of dendritic cell and macrophage gene regulation as we are getting ready for the next challenges.
Collapse
Affiliation(s)
- Zsolt Czimmerer
- Institute of Genetics, Biological Research Centre, Eotvos Lorand Research Network, Szeged, Hungary
| | - Laszlo Nagy
- Departments Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, and Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St Petersburg, FL, United States
| |
Collapse
|
49
|
Gau D, Daoud A, Allen A, Joy M, Sagan A, Lee S, Lucas PC, Duensing S, Boone D, Osmanbeyoglu HU, Roy P. Vascular endothelial profilin-1 drives a protumorigenic tumor microenvironment and tumor progression in renal cancer. J Biol Chem 2023; 299:105044. [PMID: 37451478 PMCID: PMC10432806 DOI: 10.1016/j.jbc.2023.105044] [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: 04/18/2023] [Revised: 07/06/2023] [Accepted: 07/09/2023] [Indexed: 07/18/2023] Open
Abstract
Overexpression of actin-binding protein profilin-1 (Pfn1) correlates with advanced disease features and adverse clinical outcome of patients with clear cell renal carcinoma, the most prevalent form of renal cancer. We previously reported that Pfn1 is predominantly overexpressed in tumor-associated vascular endothelial cells in human clear cell renal carcinoma. In this study, we combined in vivo strategies involving endothelial cell-specific depletion and overexpression of Pfn1 to demonstrate a role of vascular endothelial Pfn1 in promoting tumorigenicity and enabling progressive growth and metastasis of renal carcinoma cells in a syngeneic orthotopic mouse model of kidney cancer. We established an important role of endothelial Pfn1 in tumor angiogenesis and further identified endothelial Pfn1-dependent regulation of several pro- (VEGF, SERPINE1, CCL2) and anti-angiogenic factors (platelet factor 4) in vivo. Endothelial Pfn1 overexpression increases tumor infiltration by macrophages and concomitantly diminishes tumor infiltration by T cells including CD8+ T cells in vivo, correlating with the pattern of endothelial Pfn1-dependent changes in tumor abundance of several prominent immunomodulatory cytokines. These data were also corroborated by multiplexed quantitative immunohistochemistry and immune deconvolution analyses of RNA-seq data of clinical samples. Guided by Upstream Regulator Analysis of tumor transcriptome data, we further established endothelial Pfn1-induced Hif1α elevation and suppression of STAT1 activation. In conclusion, this study demonstrates for the first time a direct causal relationship between vascular endothelial Pfn1 dysregulation, immunosuppressive tumor microenvironment, and disease progression with mechanistic insights in kidney cancer. Our study also provides a conceptual basis for targeting Pfn1 for therapeutic benefit in kidney cancer.
Collapse
Affiliation(s)
- David Gau
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
| | - Andrew Daoud
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Abigail Allen
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Marion Joy
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - April Sagan
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sanghoon Lee
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Peter C Lucas
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Stefan Duensing
- Department of Urology, University of Heidelberg School of Medicine, Heidelberg, Germany
| | - David Boone
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Hatice U Osmanbeyoglu
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Partha Roy
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
50
|
Samuel CE. Interferon at the crossroads of SARS-CoV-2 infection and COVID-19 disease. J Biol Chem 2023; 299:104960. [PMID: 37364688 PMCID: PMC10290182 DOI: 10.1016/j.jbc.2023.104960] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/17/2023] [Accepted: 06/20/2023] [Indexed: 06/28/2023] Open
Abstract
A novel coronavirus now known as SARS-CoV-2 emerged in late 2019, possibly following a zoonotic crossover from a coronavirus present in bats. This virus was identified as the pathogen responsible for the severe respiratory disease, coronavirus disease-19 (COVID-19), which as of May 2023, has killed an estimated 6.9 million people globally according to the World Health Organization. The interferon (IFN) response, a cornerstone of antiviral innate immunity, plays a key role in determining the outcome of infection by SARS-CoV-2. This review considers evidence that SARS-CoV-2 infection leads to IFN production; that virus replication is sensitive to IFN antiviral action; molecular mechanisms by which the SARS-CoV-2 virus antagonizes IFN action; and how genetic variability of SARS-CoV-2 and the human host affects the IFN response at the level of IFN production or action or both. Taken together, the current understanding suggests that deficiency of an effective IFN response is an important determinant underlying some cases of critical COVID-19 disease and that IFNλ and IFNα/β have potential as therapeutics for the treatment of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Charles E Samuel
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, California, USA.
| |
Collapse
|