1
|
Liang MZ, Huang XF, Zhu JC, Bao JX, Chen CL, Wang XW, Lou YW, Pan YT, Dai YW. A machine learning-based glycolysis and fatty acid metabolism-related prognostic signature is constructed and identified ACSL5 as a novel marker inhibiting the proliferation of breast cancer. Comput Biol Chem 2025; 119:108507. [PMID: 40403353 DOI: 10.1016/j.compbiolchem.2025.108507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 04/27/2025] [Accepted: 05/09/2025] [Indexed: 05/24/2025]
Abstract
INTRODUCTION A new perspective on cancer metabolism suggests that it varies by context and is diverse. Cancer metabolism reprogramming can create a heterogeneous microenvironment that affects immune cell infiltration and function, complicating the selection of treatment methods. However, the specifics of this relationship remain unclear in breast cancer. This research aims to explore how glycolysis and fatty acid metabolism (GF) influence the immune microenvironment and their predictive capabilities for immunotherapy responses and overall survival. METHODS We at first time identified 602 GF-related genes. Utilizing multiple datasets from various centers and employing 10 different machine learning algorithms, we developed a GF-related signature called GFSscore, driven by artificial intelligence. RESULTS The GFSscore served as an independent prognostic indicator and demonstrated greater robustness than other models. Its validity was validated through multiple databases. Our study found that breast cancer patients with a high GFSscore, indicative of a greater tendency towards glycolytic activity, experienced poorer prognosis due to immunosuppression from distinct immune evasion mechanisms. Conversely, those with a low GFSscore, more inclined towards fatty acid metabolism, had better outcomes. Additionally, the GFSscore has the potential to forecast how well a patient might respond to immunotherapy and their susceptibility to chemotherapy medications. Moreover, we found that the overexpressed ACSL5 gene inhibits the proliferation of BRCA through experiments. CONCLUSIONS The GFSscore may offer patients personalized therapy by identifying new therapeutic targets for tumors. By understanding the relationship between cancer metabolism and the immune microenvironment, we can better tailor treatments to individual patients.
Collapse
Affiliation(s)
- Mei-Zhen Liang
- Department of Thyroid and Breast Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Xian-Feng Huang
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Jun-Chang Zhu
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Jing-Xia Bao
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Cheng-Liang Chen
- Department of Thyroid and Breast Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Xiao-Wu Wang
- Department of Burns and Skin Repair Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Yun-Wei Lou
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Ya-Ting Pan
- Yongkang First People's Hospital Medical Group, Jinhua, Zhejiang, China.
| | - Yin-Wei Dai
- Department of Thyroid and Breast Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
2
|
Zhuo Z, Wu H, Xu L, Ji Y, Li J, Liu L, Zhang H, Yang Q, Zheng Z, Lun W. Machine learning-based integration reveals immunological heterogeneity and the clinical potential of T cell receptor (TCR) gene pattern in hepatocellular carcinoma. Apoptosis 2025; 30:955-975. [PMID: 39904860 DOI: 10.1007/s10495-025-02080-6] [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] [Accepted: 01/13/2025] [Indexed: 02/06/2025]
Abstract
The T Cell Receptor (TCR) significantly contributes to tumor immunity, whereas the intricate interplay with the Hepatocellular Carcinoma (HCC) microenvironment and clinical significance remains largely unexplored. Here, we aimed to examine the function of TCR signaling in tumor immunity and its clinical significance in HCC. Our objective was to employ TCR signaling genes and a machine learning-based integrative methodology to construct a prognostic prediction system termed the TCR score. Herein, we revealed that the TCR score serves as an independent risk factor for overall survival in HCC patients, demonstrating stable and robust performance. The accuracy of the TCR score significantly exceeds that of traditional clinical variables and published signatures. Additionally, the immune infiltration was abundant in patients with low TCR scores. Single-cell cohort analysis further demonstrates that patients with low TCR scores possess an immune-active tumor microenvironment (TME), with T/NK cells enhancing interactions with myeloid cells through signaling networks such as MIF, MK, and SPP1. In response to these changes in the TME, patients with high TCR scores exhibit poorer outcomes and shorter survival in immunotherapy cohorts. In vitro experiments demonstrated that the key TCR signaling biomarker SOS1 knockdown significantly suppresses the HCC cells' capability to proliferate, invade, and migrate while enhancing tumor cell apoptosis. The TCR score could function as a robust and potential tool to predict immune activity and improve clinical outcomes for HCC patients.
Collapse
MESH Headings
- Humans
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/mortality
- Liver Neoplasms/genetics
- Liver Neoplasms/immunology
- Liver Neoplasms/pathology
- Liver Neoplasms/mortality
- Machine Learning
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Tumor Microenvironment/immunology
- Tumor Microenvironment/genetics
- Male
- Prognosis
- Female
- Signal Transduction
- Gene Expression Regulation, Neoplastic
- Cell Line, Tumor
- Middle Aged
Collapse
Affiliation(s)
- Zewei Zhuo
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
- Department of Gastroenterology, The Sixth Affiliated Hospital, South China University of Technology, Foshan, 510315, China
- Heyuan People's Hospital, Heyuan, Guangdong, 517001, China
| | - Huihuan Wu
- Department of Gastroenterology, The Sixth Affiliated Hospital, South China University of Technology, Foshan, 510315, China
| | - Lingli Xu
- Dadong Street Community Health Service Center, Guangzhou, 510080, China
| | - Yuran Ji
- Heyuan People's Hospital, Heyuan, Guangdong, 517001, China
| | - Jiezhuang Li
- Heyuan People's Hospital, Heyuan, Guangdong, 517001, China
| | - Liehui Liu
- Heyuan People's Hospital, Heyuan, Guangdong, 517001, China
| | - Hong Zhang
- Department of Lymphoma, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, China.
| | - Qi Yang
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, China.
| | - Zhongwen Zheng
- Heyuan People's Hospital, Heyuan, Guangdong, 517001, China.
| | - Weijian Lun
- Department of Gastroenterology, The Sixth Affiliated Hospital, South China University of Technology, Foshan, 510315, China.
| |
Collapse
|
3
|
Strijker JGM, Pascual-Pasto G, Grothusen GP, Kalmeijer YJ, Kalaitsidou E, Zhao C, McIntyre B, Matlaga S, Visser LL, Barisa M, Himsworth C, Shah R, Muller H, Schild LG, Hains PG, Zhong Q, Reddel RR, Robinson PJ, Catena X, Soengas MS, Margaritis T, Dekker FJ, Anderson J, Molenaar JJ, Bosse KR, Wu W, Wienke J. Blocking MIF secretion enhances CAR T-cell efficacy against neuroblastoma. Eur J Cancer 2025; 218:115263. [PMID: 39908652 PMCID: PMC11884407 DOI: 10.1016/j.ejca.2025.115263] [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: 07/23/2024] [Revised: 12/12/2024] [Accepted: 01/21/2025] [Indexed: 02/07/2025]
Abstract
INTRODUCTION Chimeric antigen receptor (CAR) T-cell therapy is a promising and innovative cancer therapy. However, immunosuppressive tumor microenvironments (TME) limit T cell persistence and durable efficacy. Here, we aimed to identify and target immunosuppressive factors in the TME of neuroblastoma, a pediatric extracranial solid tumor, to improve CAR-T efficacy. METHODS Immunosuppressive factors were identified using a multi-omics approach, including single-cell RNA sequencing (scRNA-seq) of 24 neuroblastoma tumors, published bulk-RNA sequencing datasets, and mass-spectrometry of patient-derived tumoroid models. Candidate targets were validated with functional assays in vitro and in vivo. Protein degradation of the top immunosuppressive target by PROTAC technology was used to evaluate the effect on CAR T-cell activity. RESULTS ScRNA-seq revealed 13 immunosuppressive interactions in the TME of neuroblastoma, two effectors of which, Midkine (MDK) and Macrophage Migration Inhibitory Factor (MIF), were validated as candidate targets across multiple published datasets. Both factors were among the top 6 % of most abundantly secreted factors by patient-derived tumoroid models, substantiating their potential relevance in the TME. In vitro and in vivo functional assays confirmed MIF to be a potent inhibitor of CAR T-cell activation and killing capacity. To translate these findings into a potentially clinically applicable treatment, we explored MIF targeting by PROTAC technology, which significantly enhanced activation of CAR T-cells targeting GPC2 and B7-H3. CONCLUSION By defining the immunosuppressive effects of neuroblastoma's TME on CAR T-cell efficacy, revealing the pivotal role of MIF, we provide an analytic pipeline and therapeutic strategy for improving adoptive cell therapies for this pediatric malignancy and potentially other solid tumors.
Collapse
Affiliation(s)
| | - Guillem Pascual-Pasto
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia; Philadelphia, PA 19104, USA
| | - Grant P Grothusen
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia; Philadelphia, PA 19104, USA
| | | | - Elisavet Kalaitsidou
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A⁎STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore; Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Singapore 117543, Singapore
| | - Chunlong Zhao
- Department of Chemical and Pharmaceutical Biology, Groningen, Research Institute of Pharmacy (GRIP), University of Groningen, Antonius Deusinglaan 1, Groningen 9713 AV, the Netherlands
| | - Brendan McIntyre
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia; Philadelphia, PA 19104, USA
| | - Stephanie Matlaga
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia; Philadelphia, PA 19104, USA
| | - Lindy L Visser
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Marta Barisa
- UCL Great Ormond St Institute of Child Health, London, UK
| | | | - Rivani Shah
- UCL Great Ormond St Institute of Child Health, London, UK
| | - Henrike Muller
- UCL Great Ormond St Institute of Child Health, London, UK
| | - Linda G Schild
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Peter G Hains
- ProCan, Children's Medical Research Institute, The University of Sydney, Westmead, NSW, Australia
| | - Qing Zhong
- ProCan, Children's Medical Research Institute, The University of Sydney, Westmead, NSW, Australia
| | - Roger R Reddel
- ProCan, Children's Medical Research Institute, The University of Sydney, Westmead, NSW, Australia
| | - Phillip J Robinson
- ProCan, Children's Medical Research Institute, The University of Sydney, Westmead, NSW, Australia
| | - Xavier Catena
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - María S Soengas
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | - Frank J Dekker
- Department of Chemical and Pharmaceutical Biology, Groningen, Research Institute of Pharmacy (GRIP), University of Groningen, Antonius Deusinglaan 1, Groningen 9713 AV, the Netherlands
| | - John Anderson
- UCL Great Ormond St Institute of Child Health, London, UK
| | - Jan J Molenaar
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Pharmaceutical Sciences, University Utrecht, Utrecht, the Netherlands
| | - Kristopher R Bosse
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia; Philadelphia, PA 19104, USA; Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wei Wu
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A⁎STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore; Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Singapore 117543, Singapore; Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Judith Wienke
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.
| |
Collapse
|
4
|
James BL, Zaidi SN, Aiswarya RK, Shetty V, Vidya Bhushan R, Dokhe Y, Naveen BS, Pillai V, Dhar SK, Kuriakose MA, Suresh A. Modeling the lymph node stromal cells in oral squamous cell carcinoma: insights into the stromal cues in nodal metastasis. Hum Cell 2025; 38:41. [PMID: 39760828 DOI: 10.1007/s13577-024-01166-8] [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: 07/01/2024] [Accepted: 12/16/2024] [Indexed: 01/07/2025]
Abstract
The study explores the development and characterization of lymph node stromal cell cultures (LNSCs) from patients with oral squamous cell carcinoma (OSCC), highlighting the importance of understanding tumor-node cross-talk for effective prognostic and therapeutic interventions. Herein, we describe the development and characterization of primary lymph node stromal cells (LNSCs, N = 14) from nodes of metastatic and non-metastatic OSCC patients. Primary cultures were established by the explant method from positive (N + ; N = 2), and negative nodes (N0m; N = 4) of the metastatic patients (N = 3) as well as negative (N0nm; N = 8) nodes from non-metastatic (N = 4) patients. STR profiling confirmed the purity and novelty, while characterization by immunocytochemistry/flow cytometry revealed heterogeneous cell populations consisting of fibroblastic reticular cells (CD31-Gp38 +) and double negative cells (CD31-Gp38-). Transcriptomic profiling indicated molecular alterations in the cells based on the non-metastatic, the pre-metastatic or metastatic status of the nodes, pro-inflammatory, matrix remodeling, and immune evasion being the primary pathways. Assessment of the protein levels for five selected markers (MX1, ISG15, CPM, ITGB4 and FOS) in the cell lines revealed that CPM levels were significantly reduced in the N + and N0m nodes whereas ISG15 levels reduced in N0m. Significantly, the profiling also provided insights into possible glycosylation of CPM (N0nm) and ISGylation of ISG15 (N0m). Cytokine profiling indicated release of chemokines/anti-proliferative cytokines from the negative nodes, while angiogenic/pro-metastatic cytokines were released from the nodes of metastatic patients. The lymph node stromal cell models established in the study with distinctive transcriptomic/cytokine characteristics will be invaluable in delineating the processes underlying nodal metastasis.
Collapse
Affiliation(s)
- Bonney Lee James
- Integrated Head and Neck Oncology Program (DSRG-5), Mazumdar Shaw Medical Foundation, Narayana Health, Bangalore, India
- Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Shaesta Naseem Zaidi
- Department of Pathology, Mazumdar Shaw Medical Centre, Narayana Health, Bangalore, India
| | - R K Aiswarya
- Integrated Head and Neck Oncology Program (DSRG-5), Mazumdar Shaw Medical Foundation, Narayana Health, Bangalore, India
| | - Vivek Shetty
- Head and Neck Oncology, Mazumdar Shaw Medical Centre, Narayana Health, Bangalore, India
| | - R Vidya Bhushan
- Head and Neck Oncology, Mazumdar Shaw Medical Centre, Narayana Health, Bangalore, India
| | - Yogesh Dokhe
- Head and Neck Oncology, Mazumdar Shaw Medical Centre, Narayana Health, Bangalore, India
| | - B S Naveen
- Head and Neck Oncology, Mazumdar Shaw Medical Centre, Narayana Health, Bangalore, India
| | - Vijay Pillai
- Head and Neck Oncology, Mazumdar Shaw Medical Centre, Narayana Health, Bangalore, India
| | - Sujan K Dhar
- Computational Biology, Mazumdar Shaw Medical Foundation, Narayana Health, Bangalore, India
| | - Moni Abraham Kuriakose
- Integrated Head and Neck Oncology Program (DSRG-5), Mazumdar Shaw Medical Foundation, Narayana Health, Bangalore, India
- Head and Neck Oncology, Mazumdar Shaw Medical Centre, Narayana Health, Bangalore, India
| | - Amritha Suresh
- Integrated Head and Neck Oncology Program (DSRG-5), Mazumdar Shaw Medical Foundation, Narayana Health, Bangalore, India.
- Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India.
- Head and Neck Oncology, Mazumdar Shaw Medical Centre, Narayana Health, Bangalore, India.
| |
Collapse
|
5
|
Zhao L, Zhao BH, Ruze A, Li QL, Deng AX, Gao XM. Distinct roles of MIF in the pathogenesis of ischemic heart disease. Cytokine Growth Factor Rev 2024; 80:121-137. [PMID: 39438226 DOI: 10.1016/j.cytogfr.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/25/2024]
Abstract
The role of macrophage migration inhibitory factor (MIF) as a multifunctional cytokine in immunomodulation and inflammatory response is increasingly appreciated. Ischemic heart disease (IHD), the leading cause of global mortality, remains a focal point of research owing to its intricate pathophysiology. MIF has been identified as a critical player in IHD, where it exerts distinct roles. On one hand, MIF plays a protective role by enhancing energy metabolism through activation of AMPK, resisting oxidative stress, inhibiting activation of the JNK pathway, and maintaining intracellular calcium ion homeostasis. Additionally, MIF exerts protective effects through mesenchymal stem cells and exosomes. On the other hand, MIF can assume a pro-inflammatory role, which contributes to the exacerbation of IHD's development and progression. Furthermore, MIF levels significantly increase in IHD patients, and its genetic polymorphisms are positively correlated with prevalence and severity. These findings position MIF as a potential biomarker and therapeutic target in the management of IHD. This review summarizes the structure, source, signaling pathways and biological functions of MIF and focuses on its roles and clinical characteristics in IHD. The genetic variants of MIF associated with IHD is also discussed, providing more understandings of its complex interplay in the disease's pathology.
Collapse
Affiliation(s)
- Ling Zhao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Bang-Hao Zhao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Amanguli Ruze
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Qiu-Lin Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - An-Xia Deng
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Xiao-Ming Gao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China; Xinjiang Key Laboratory of Cardiovascular Disease, Urumqi, China.
| |
Collapse
|
6
|
Putha L, Kok LK, Fellner M, Rutledge MT, Gamble AB, Wilbanks SM, Vernall AJ, Tyndall JDA. Covalent Isothiocyanate Inhibitors of Macrophage Migration Inhibitory Factor as Potential Colorectal Cancer Treatments. ChemMedChem 2024; 19:e202400394. [PMID: 38977403 DOI: 10.1002/cmdc.202400394] [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: 05/22/2024] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 07/10/2024]
Abstract
Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine that has roles in innate and adaptive human immune responses, as well as inflammation. MIF exerts its biological activity by binding to the cell surface receptor CD74 as well as intracellular signalling proteins. MIF also possesses keto-enol tautomerase activity. Inhibition of the tautomerase activity has been associated with loss of biological activity of MIF and a potential anticancer target. Isothiocyanates (ITCs) are a class of compounds present in cruciferous vegetables that inhibit the MIF tautomerase activity via covalent modification of the N-terminal proline. A range of substituted ITCs featuring benzyl, phenethyl and phenyl propyl isothiocyanates were designed, synthesised and tested to determine any structure activity relationship for inhibiting MIF. Crystal structures of covalent compounds 8 and 9 in complex with rhMIF revealed key hydrogen bonding and edge-to-face π stacking interactions. Compound 9 and 11 with sub micromolar activity were tested in the NCI60 cancer cell lines panel. Both compounds showed tissue-specific reduced growth in colon and renal cancer cell lines, while one of these showed potent, dose-dependent inhibition of growth against all seven colon cancer cell lines (GI50<2.5 μM) and all eight renal cancer cell lines (GI50<2.2 μM).
Collapse
Affiliation(s)
- Lohitha Putha
- School of Pharmacy, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
| | - Liang K Kok
- School of Pharmacy, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
| | - Matthias Fellner
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
| | - Malcolm T Rutledge
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
| | - Allan B Gamble
- School of Pharmacy, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
| | - Sigurd M Wilbanks
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
| | - Andrea J Vernall
- Department of Chemistry, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
| | - Joel D A Tyndall
- School of Pharmacy, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
| |
Collapse
|
7
|
Xiao SM, Xu R, Yang YX, Zhao R, Xie Y, Lei XD, Wu XT. Gastrointestinal stromal tumors regulate macrophage M2 polarization through the MIF/CXCR4 axis to immune escape. Front Immunol 2024; 15:1431535. [PMID: 39464891 PMCID: PMC11502962 DOI: 10.3389/fimmu.2024.1431535] [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: 05/12/2024] [Accepted: 09/19/2024] [Indexed: 10/29/2024] Open
Abstract
Purpose The infiltration of immune cells and their roles of the infiltrating-immune cells in gastrointestinal stromal tumor (GIST) is still unclear. We aimed to discover the infiltration cell types and the relationship between the infiltrating-immune cells and the progression of GIST. Experimental design Single-cell RNA sequencing were performed to discover types of the infiltrating-immune cells and to analyze CellChat between cells. Immunohistochemistry of 80 GIST samples were used to clarify the relation between macrophages and recurrence risk. In vitro, flow cytometry and Real-time PCR were performed to uncover a potential mechanism of tumor cell regulation of macrophages. Results Tumor cells, macrophages, and T-cells were the predominant cell types. The MIF/CXCR4 axis was the most common ligand-receptor interaction between macrophages and tumor cells. As the risk increased, expression levels of CD68, CD206, MIF, and CXCR4 gradually increased. In vitro, we found that GIST882 was able to secrete MIF and GIST882 cell supernatant upregulated M2 polarization. Real-time PCR showed that expression levels of IL-10 mRNA and Arginase-1 mRNA were also the highest in the GIST882 cell supernatant group. Conclusions These findings identify that macrophages are the most abundant infiltrating cells in GIST. The MIF/CXCR4 axis is the most common ligand-receptor interaction between macrophages and tumor cells. GIST cells can regulate macrophage M2 polarization through the MIF/CXCR4 axis.
Collapse
Affiliation(s)
- Shuo-meng Xiao
- Department of Gastric Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Rui Xu
- Department of Gastric Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ying-xin Yang
- Department of Oncology, The First People’s Hospital of Dali, Dali City, Yunnan, China
| | - Rui Zhao
- Department of Gastrointestinal Surgery, West China School of Medicine and West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuan Xie
- Department of Gastric Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Xu-dan Lei
- Radiation Oncology Key Laboratory of Sichuan Province, Department of Experimental Research, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Xiao-ting Wu
- Department of Gastrointestinal Surgery, West China School of Medicine and West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
8
|
Louault K, De Clerck YA, Janoueix-Lerosey I. The neuroblastoma tumor microenvironment: From an in-depth characterization towards novel therapies. EJC PAEDIATRIC ONCOLOGY 2024; 3:100161. [PMID: 39036648 PMCID: PMC11259008 DOI: 10.1016/j.ejcped.2024.100161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Neuroblastoma is a cancer of the sympathetic nervous system that develops in young children, either as low-risk or high-risk disease. The tumor microenvironment (TME) is now recognized as an important player of the tumor ecosystem that may promote drug resistance and immune escape. Targeting the TME in combination with therapies directly targeting tumor cells therefore represents an interesting strategy to prevent the emergence of resistance in cancer and improve patient's outcome. The development of such strategies however requires an in-depth understanding of the TME landscape, due to its high complexity and intra and inter-tumoral heterogeneity. Various approaches have been used in the last years to characterize the immune and non-immune cell populations present in tumors of neuroblastoma patients, both quantitatively and qualitatively, in particular with the use of single-cell transcriptomics. It is anticipated that in the near future, both genomic and TME information in tumors will contribute to a precise approach to therapy in neuroblastoma. Deciphering the mechanisms of interaction between neuroblastoma cells and stromal or immune cells in the TME is key to identify novel therapeutic combinations. Over the last decade, numerous in vitro studies and in vivo pre-clinical experiments in immune-competent and immune-deficient models have identified therapeutic approaches to circumvent drug resistance and immune escape. Some of these studies have formed the basis for early phase I and II clinical trials in children with recurrent and refractory high-risk neuroblastoma. This review summarizes recently published data on the characterization of the TME landscape in neuroblastoma and novel strategies targeting various TME cellular components, molecules and pathways activated as a result of the tumor-host interactions.
Collapse
Affiliation(s)
- Kevin Louault
- Children’s Hospital Los Angeles, Cancer, and Blood Disease Institute, 4650 Sunset Bld., Los Angeles, CA, USA
| | - Yves A. De Clerck
- Children’s Hospital Los Angeles, Cancer, and Blood Disease Institute, 4650 Sunset Bld., Los Angeles, CA, USA
- Department of Pediatrics and Biochemistry and Molecular Medicine, University of Southern California, CA, USA
| | - Isabelle Janoueix-Lerosey
- Curie Institute, PSL Research University, Inserm U830, Paris, France
- SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Curie Institute, Paris, France
| |
Collapse
|
9
|
Valdez CN, Sánchez-Zuno GA, Bucala R, Tran TT. Macrophage Migration Inhibitory Factor (MIF) and D-Dopachrome Tautomerase (DDT): Pathways to Tumorigenesis and Therapeutic Opportunities. Int J Mol Sci 2024; 25:4849. [PMID: 38732068 PMCID: PMC11084905 DOI: 10.3390/ijms25094849] [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: 03/28/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Discovered as inflammatory cytokines, MIF and DDT exhibit widespread expression and have emerged as critical mediators in the response to infection, inflammation, and more recently, in cancer. In this comprehensive review, we provide details on their structures, binding partners, regulatory mechanisms, and roles in cancer. We also elaborate on their significant impact in driving tumorigenesis across various cancer types, supported by extensive in vitro, in vivo, bioinformatic, and clinical studies. To date, only a limited number of clinical trials have explored MIF as a therapeutic target in cancer patients, and DDT has not been evaluated. The ongoing pursuit of optimal strategies for targeting MIF and DDT highlights their potential as promising antitumor candidates. Dual inhibition of MIF and DDT may allow for the most effective suppression of canonical and non-canonical signaling pathways, warranting further investigations and clinical exploration.
Collapse
Affiliation(s)
- Caroline Naomi Valdez
- School of Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA; (C.N.V.); (R.B.)
| | - Gabriela Athziri Sánchez-Zuno
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA;
| | - Richard Bucala
- School of Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA; (C.N.V.); (R.B.)
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA;
- Yale Cancer Center, Yale University, 333 Cedar St., New Haven, CT 06510, USA
| | - Thuy T. Tran
- School of Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA; (C.N.V.); (R.B.)
- Yale Cancer Center, Yale University, 333 Cedar St., New Haven, CT 06510, USA
- Section of Medical Oncology, Department of Internal Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA
| |
Collapse
|
10
|
Gorostegui M, Muñoz JP, Perez-Jaume S, Simao-Rafael M, Larrosa C, Garraus M, Salvador N, Lavarino C, Krauel L, Mañe S, Castañeda A, Mora J. Management of High-Risk Neuroblastoma with Soft-Tissue-Only Disease in the Era of Anti-GD2 Immunotherapy. Cancers (Basel) 2024; 16:1735. [PMID: 38730688 PMCID: PMC11083939 DOI: 10.3390/cancers16091735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Neuroblastoma presents with two patterns of disease: locoregional or systemic. The poor prognostic risk factors of locoregional neuroblastoma (LR-NB) include age, MYCN or MDM2-CDK4 amplification, 11q, histology, diploidy with ALK or TERT mutations, and ATRX aberrations. Anti-GD2 immunotherapy has significantly improved the outcome of high-risk (HR) NB and is mostly effective against osteomedullary minimal residual disease (MRD), but less so against soft tissue disease. The question is whether adding anti-GD2 monoclonal antibodies (mAbs) benefits patients with HR-NB compounded by only soft tissue. We reviewed 31 patients treated at SJD for HR-NB with no osteomedullary involvement at diagnosis. All tumors had molecular genetic features of HR-NB. The outcome after first-line treatment showed 25 (80.6%) patients achieving CR. Thirteen patients remain in continued CR, median follow-up 3.9 years. We analyzed whether adding anti-GD2 immunotherapy to first-line treatment had any prognostic significance. The EFS analysis using Cox models showed a HR of 0.20, p = 0.0054, and an 80% decrease in the risk of relapse in patients treated with anti-GD2 immunotherapy in the first line. Neither EFS nor OS were significantly different by CR status after first-line treatment. In conclusion, adding treatment with anti-GD2 mAbs at the stage of MRD helps prevent relapse that unequivocally portends poor survival.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Jaume Mora
- Pediatric Cancer Center Barcelona, Hospital Sant Joan de Déu, 08950 Barcelona, Spain; (M.G.); (J.P.M.); (M.S.-R.); (C.L.); (M.G.); (N.S.); (C.L.); (L.K.); (S.M.); (A.C.)
| |
Collapse
|
11
|
Spertini C, Bénéchet AP, Birch F, Bellotti A, Román-Trufero M, Arber C, Auner HW, Mitchell RA, Spertini O, Smirnova T. Macrophage migration inhibitory factor blockade reprograms macrophages and disrupts prosurvival signaling in acute myeloid leukemia. Cell Death Discov 2024; 10:157. [PMID: 38548753 PMCID: PMC10978870 DOI: 10.1038/s41420-024-01924-5] [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: 09/29/2023] [Revised: 03/14/2024] [Accepted: 03/19/2024] [Indexed: 04/01/2024] Open
Abstract
The malignant microenvironment plays a major role in the development of resistance to therapies and the occurrence of relapses in acute myeloid leukemia (AML). We previously showed that interactions of AML blasts with bone marrow macrophages (MΦ) shift their polarization towards a protumoral (M2-like) phenotype, promoting drug resistance; we demonstrated that inhibiting the colony-stimulating factor-1 receptor (CSF1R) repolarizes MΦ towards an antitumoral (M1-like) phenotype and that other factors may be involved. We investigated here macrophage migration inhibitory factor (MIF) as a target in AML blast survival and protumoral interactions with MΦ. We show that pharmacologically inhibiting MIF secreted by AML blasts results in their apoptosis. However, this effect is abrogated when blasts are co-cultured in close contact with M2-like MΦ. We next demonstrate that pharmacological inhibition of MIF secreted by MΦ, in the presence of granulocyte macrophage-colony stimulating factor (GM-CSF), efficiently reprograms MΦ to an M1-like phenotype that triggers apoptosis of interacting blasts. Furthermore, contact with reprogrammed MΦ relieves blast resistance to venetoclax and midostaurin acquired in contact with CD163+ protumoral MΦ. Using intravital imaging in mice, we also show that treatment with MIF inhibitor 4-IPP and GM-CSF profoundly affects the tumor microenvironment in vivo: it strikingly inhibits tumor vasculature, reduces protumoral MΦ, and slows down leukemia progression. Thus, our data demonstrate that MIF plays a crucial role in AML MΦ M2-like protumoral phenotype that can be reversed by inhibiting its activity and suggest the therapeutic targeting of MIF as an avenue towards improved AML treatment outcomes.
Collapse
Affiliation(s)
- Caroline Spertini
- Service and Central Laboratory of Hematology, Lausanne University Hospital (CHUV), 1011, Lausanne, Switzerland
| | - Alexandre P Bénéchet
- In Vivo Imaging Facility (IVIF), Department of Research and Training, Lausanne University Hospital and University of Lausanne, Lausanne, 1011, Switzerland
| | - Flora Birch
- Department of oncology UNIL-CHUV, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), 1011, Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, 1015, Lausanne, Switzerland
| | - Axel Bellotti
- Service and Central Laboratory of Hematology, Lausanne University Hospital (CHUV), 1011, Lausanne, Switzerland
| | - Mónica Román-Trufero
- Service and Central Laboratory of Hematology, Lausanne University Hospital (CHUV), 1011, Lausanne, Switzerland
| | - Caroline Arber
- Service and Central Laboratory of Hematology, Lausanne University Hospital (CHUV), 1011, Lausanne, Switzerland
- Department of oncology UNIL-CHUV, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), 1011, Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, 1015, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, 1011, Lausanne, Switzerland
- Service of Immuno-oncology, Lausanne University Hospital (CHUV), 1011, Lausanne, Switzerland
| | - Holger W Auner
- Service and Central Laboratory of Hematology, Lausanne University Hospital (CHUV), 1011, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, 1011, Lausanne, Switzerland
| | - Robert A Mitchell
- Department of Surgery, Division of Immunotherapy, University of Louisville, Louisville, KY, 40202, USA
| | - Olivier Spertini
- Faculty of Biology and Medicine, University of Lausanne, 1011, Lausanne, Switzerland
| | - Tatiana Smirnova
- Service and Central Laboratory of Hematology, Lausanne University Hospital (CHUV), 1011, Lausanne, Switzerland.
| |
Collapse
|
12
|
Tang W, Lo CWS, Ma W, Chu ATW, Tong AHY, Chung BHY. Revealing the role of SPP1 + macrophages in glioma prognosis and therapeutic targeting by investigating tumor-associated macrophage landscape in grade 2 and 3 gliomas. Cell Biosci 2024; 14:37. [PMID: 38515213 PMCID: PMC10956315 DOI: 10.1186/s13578-024-01218-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 03/13/2024] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND Glioma is a highly heterogeneous brain tumor categorized into World Health Organization (WHO) grades 1-4 based on its malignancy. The suppressive immune microenvironment of glioma contributes significantly to unfavourable patient outcomes. However, the cellular composition and their complex interplays within the glioma environment remain poorly understood, and reliable prognostic markers remain elusive. Therefore, in-depth exploration of the tumor microenvironment (TME) and identification of predictive markers are crucial for improving the clinical management of glioma patients. RESULTS Our analysis of single-cell RNA-sequencing data from glioma samples unveiled the immunosuppressive role of tumor-associated macrophages (TAMs), mediated through intricate interactions with tumor cells and lymphocytes. We also discovered the heterogeneity within TAMs, among which a group of suppressive TAMs named TAM-SPP1 demonstrated a significant association with Epidermal Growth Factor Receptor (EGFR) amplification, impaired T cell response and unfavourable patient survival outcomes. Furthermore, by leveraging genomic and transcriptomic data from The Cancer Genome Atlas (TCGA) dataset, two distinct molecular subtypes with a different constitution of TAMs, EGFR status and clinical outcomes were identified. Exploiting the molecular differences between these two subtypes, we developed a four-gene-based prognostic model. This model displayed strong associations with an elevated level of suppressive TAMs and could be used to predict anti-tumor immune response and prognosis in glioma patients. CONCLUSION Our findings illuminated the molecular and cellular mechanisms that shape the immunosuppressive microenvironment in gliomas, providing novel insights into potential therapeutic targets. Furthermore, the developed prognostic model holds promise for predicting immunotherapy response and assisting in more precise risk stratification for glioma patients.
Collapse
Affiliation(s)
- Wenshu Tang
- Hong Kong Genome Institute, 2/F, Building 20E, Hong Kong Science Park, Hong Kong, China
| | - Cario W S Lo
- Hong Kong Genome Institute, 2/F, Building 20E, Hong Kong Science Park, Hong Kong, China
| | - Wei Ma
- Hong Kong Genome Institute, 2/F, Building 20E, Hong Kong Science Park, Hong Kong, China
| | - Annie T W Chu
- Hong Kong Genome Institute, 2/F, Building 20E, Hong Kong Science Park, Hong Kong, China
| | - Amy H Y Tong
- Hong Kong Genome Institute, 2/F, Building 20E, Hong Kong Science Park, Hong Kong, China
| | - Brian H Y Chung
- Hong Kong Genome Institute, 2/F, Building 20E, Hong Kong Science Park, Hong Kong, China.
- Department of Pediatrics and Adolescent Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
13
|
Stip MC, Teeuwen L, Dierselhuis MP, Leusen JHW, Krijgsman D. Targeting the myeloid microenvironment in neuroblastoma. J Exp Clin Cancer Res 2023; 42:337. [PMID: 38087370 PMCID: PMC10716967 DOI: 10.1186/s13046-023-02913-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
Myeloid cells (granulocytes and monocytes/macrophages) play an important role in neuroblastoma. By inducing a complex immunosuppressive network, myeloid cells pose a challenge for the adaptive immune system to eliminate tumor cells, especially in high-risk neuroblastoma. This review first summarizes the pro- and anti-tumorigenic functions of myeloid cells, including granulocytes, monocytes, macrophages, and myeloid-derived suppressor cells (MDSC) during the development and progression of neuroblastoma. Secondly, we discuss how myeloid cells are engaged in the current treatment regimen and explore novel strategies to target these cells in neuroblastoma. These strategies include: (1) engaging myeloid cells as effector cells, (2) ablating myeloid cells or blocking the recruitment of myeloid cells to the tumor microenvironment and (3) reprogramming myeloid cells. Here we describe that despite their immunosuppressive traits, tumor-associated myeloid cells can still be engaged as effector cells, which is clear in anti-GD2 immunotherapy. However, their full potential is not yet reached, and myeloid cell engagement can be enhanced, for example by targeting the CD47/SIRPα axis. Though depletion of myeloid cells or blocking myeloid cell infiltration has been proven effective, this strategy also depletes possible effector cells for immunotherapy from the tumor microenvironment. Therefore, reprogramming of suppressive myeloid cells might be the optimal strategy, which reverses immunosuppressive traits, preserves myeloid cells as effectors of immunotherapy, and subsequently reactivates tumor-infiltrating T cells.
Collapse
Affiliation(s)
- Marjolein C Stip
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands
| | - Loes Teeuwen
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands
| | | | - Jeanette H W Leusen
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands
| | - Daniëlle Krijgsman
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands.
- Center for Molecular Medicine, University Medical Center Utrecht, 3584 CX, Utrecht, the Netherlands.
| |
Collapse
|
14
|
Spiller L, Manjula R, Leissing F, Basquin J, Bourilhon P, Sinitski D, Brandhofer M, Levecque S, Gerra S, Sabelleck B, Zhang L, Feederle R, Flatley A, Hoffmann A, Panstruga R, Bernhagen J, Lolis E. Plant MDL proteins synergize with the cytokine MIF at CXCR2 and CXCR4 receptors in human cells. Sci Signal 2023; 16:eadg2621. [PMID: 37988455 DOI: 10.1126/scisignal.adg2621] [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: 12/20/2022] [Accepted: 10/27/2023] [Indexed: 11/23/2023]
Abstract
Mammalian macrophage migration inhibitory factor (MIF) and its paralog, D-dopachrome tautomerase, are multifunctional inflammatory cytokines. Plants have orthologous MIF and D-dopachrome tautomerase-like (MDL) proteins that mimic some of the effects of MIF on immune cells in vitro. We explored the structural and functional similarities between the three Arabidopsis thaliana MDLs and MIF. X-ray crystallography of the MDLs revealed high structural similarity between MDL and MIF homotrimers and suggested a potential explanation for the lack of tautomerase activity in the MDLs. MDL1 and MDL2 interacted with each other and with MIF in vitro, in yeast, and in plant leaves and formed hetero-oligomeric complexes with MIF in vitro. The MDLs stimulated signaling through the MIF receptors CXCR2 or CXCR4 and enhanced the responses to MIF in a yeast reporter system, in human neutrophils, and in human lung epithelial cells. Pharmacological inhibitors that disrupted MIF activity or prevented the formation of MIF-MDL hetero-oligomers blocked the observed synergism. These findings demonstrate that MDLs can enhance cellular responses to MIF, which may have functional implications in tissues exposed to MDLs from the diet or environment.
Collapse
Affiliation(s)
- Lukas Spiller
- Department of Pharmacology, School of Medicine, Yale University, New Haven, CT 06510, USA
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Ludwig-Maximilians-Universität (LMU) München, LMU University Hospital, 81377 Munich, Germany
| | - Ramu Manjula
- Department of Pharmacology, School of Medicine, Yale University, New Haven, CT 06510, USA
| | - Franz Leissing
- Unit of Plant Molecular Cell Biology, Institute for Biology I, RWTH Aachen University, 52056 Aachen, Germany
| | - Jerome Basquin
- Department of Structural Cell Biology and Crystallization Facility, Max-Planck-Institute for Biochemistry, 82152 Martinsried, Germany
| | - Priscila Bourilhon
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Ludwig-Maximilians-Universität (LMU) München, LMU University Hospital, 81377 Munich, Germany
| | - Dzmitry Sinitski
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Ludwig-Maximilians-Universität (LMU) München, LMU University Hospital, 81377 Munich, Germany
| | - Markus Brandhofer
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Ludwig-Maximilians-Universität (LMU) München, LMU University Hospital, 81377 Munich, Germany
| | - Sophie Levecque
- Unit of Plant Molecular Cell Biology, Institute for Biology I, RWTH Aachen University, 52056 Aachen, Germany
| | - Simona Gerra
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Ludwig-Maximilians-Universität (LMU) München, LMU University Hospital, 81377 Munich, Germany
| | - Björn Sabelleck
- Unit of Plant Molecular Cell Biology, Institute for Biology I, RWTH Aachen University, 52056 Aachen, Germany
| | - Lin Zhang
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Ludwig-Maximilians-Universität (LMU) München, LMU University Hospital, 81377 Munich, Germany
- Department of Anesthesiology, LMU University Hospital, 81377 Munich, Germany
| | - Regina Feederle
- Monoclonal Antibody Core Facility, Helmholtz Center Munich, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Andrew Flatley
- Monoclonal Antibody Core Facility, Helmholtz Center Munich, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany
| | - Adrian Hoffmann
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Ludwig-Maximilians-Universität (LMU) München, LMU University Hospital, 81377 Munich, Germany
- Department of Anesthesiology, LMU University Hospital, 81377 Munich, Germany
| | - Ralph Panstruga
- Unit of Plant Molecular Cell Biology, Institute for Biology I, RWTH Aachen University, 52056 Aachen, Germany
| | - Jürgen Bernhagen
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Ludwig-Maximilians-Universität (LMU) München, LMU University Hospital, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Elias Lolis
- Department of Pharmacology, School of Medicine, Yale University, New Haven, CT 06510, USA
| |
Collapse
|
15
|
Chong ZX, Yong CY, Ong AHK, Yeap SK, Ho WY. Deciphering the roles of aryl hydrocarbon receptor (AHR) in regulating carcinogenesis. Toxicology 2023; 495:153596. [PMID: 37480978 DOI: 10.1016/j.tox.2023.153596] [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: 06/27/2023] [Revised: 07/13/2023] [Accepted: 07/16/2023] [Indexed: 07/24/2023]
Abstract
Aryl hydrocarbon receptor (AHR) is a ligand-dependent receptor that belongs to the superfamily of basic helix-loop-helix (bHLH) transcription factors. The activation of the canonical AHR signaling pathway is known to induce the expression of cytochrome P450 enzymes, facilitating the detoxification metabolism in the human body. Additionally, AHR could interact with various signaling pathways such as epidermal growth factor receptor (EGFR), signal transducer and activator of transcription 3 (STAT3), hypoxia-inducible factor-1α (HIF-1α), nuclear factor ekappa B (NF-κβ), estrogen receptor (ER), and androgen receptor (AR) signaling pathways. Over the past 30 years, several studies have reported that various chemical, physical, or biological agents, such as tobacco, hydrocarbon compounds, industrial and agricultural chemical wastes, drugs, UV, viruses, and other toxins, could affect AHR expression or activity, promoting cancer development. Thus, it is valuable to overview how these factors regulate AHR-mediated carcinogenesis. Current findings have reported that many compounds could act as AHR ligands to drive the expressions of AHR-target genes, such as CYP1A1, CYP1B1, MMPs, and AXL, and other targets that exert a pro-proliferation or anti-apoptotic effect, like XIAP. Furthermore, some other physical and chemical agents, such as UV and 3-methylcholanthrene, could promote AHR signaling activities, increasing the signaling activities of a few oncogenic pathways, such as the phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) and mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) pathways. Understanding how various factors regulate AHR-mediated carcinogenesis processes helps clinicians and scientists plan personalized therapeutic strategies to improve anti-cancer treatment efficacy. As many studies that have reported the roles of AHR in regulating carcinogenesis are preclinical or observational clinical studies that did not explore the detailed mechanisms of how different chemical, physical, or biological agents promote AHR-mediated carcinogenesis processes, future studies should focus on conducting large-scale and functional studies to unravel the underlying mechanism of how AHR interacts with different factors in regulating carcinogenesis processes.
Collapse
Affiliation(s)
- Zhi Xiong Chong
- Faculty of Science and Engineering, University of Nottingham Malaysia, 43500 Semenyih, Selangor, Malaysia
| | - Chean Yeah Yong
- China-ASEAN College of Marine Sciences, Xiamen University Malaysia, 43900 Sepang, Selangor, Malaysia
| | - Alan Han Kiat Ong
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, 43000 Kajang, Malaysia
| | - Swee Keong Yeap
- China-ASEAN College of Marine Sciences, Xiamen University Malaysia, 43900 Sepang, Selangor, Malaysia.
| | - Wan Yong Ho
- Faculty of Science and Engineering, University of Nottingham Malaysia, 43500 Semenyih, Selangor, Malaysia.
| |
Collapse
|
16
|
Ralchev Ralchev N, Lyubenova Bradyanova S, Valerieva Doneva Y, Mihaylova N, Vikentieva Elefterova-Florova E, Ivanov Tchorbanov A, Munoz-Valle JF, Petralia MC, Checconi P, Nicoletti F, Fagone P. Exploring the Role of CD74 and D-Dopachrome Tautomerase in COVID-19: Insights from Transcriptomic and Serum Analyses. J Clin Med 2023; 12:5037. [PMID: 37568438 PMCID: PMC10419634 DOI: 10.3390/jcm12155037] [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: 05/16/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
The COVID-19 pandemic has posed a significant threat to public health worldwide. While some patients experience only mild symptoms or no symptoms at all, others develop severe illness, which can lead to death. The host immune response is believed to play a crucial role in determining disease severity. In this study, we investigated the involvement of CD74 and D-DT in COVID-19 patients with different disease severities, by employing an in silico analysis of a publicly available transcriptomic dataset and by measuring their serum levels by ELISA. Our results showed a significant increase in MIF levels in PBMCs from COVID-19 patients, as well as a significant increase in the D-DT levels in PBMCs. However, we observed no modulation in the serum levels of D-DT. We also observed a concordant reduction in the serum levels and PBMCs expression levels of CD74. Furthermore, we found a negative correlation between CD74 serum levels and IL-13. In conclusion, our study sheds light on the involvement of CD74 and D-DT in COVID-19, with potential implications for disease severity and treatment. Further studies are needed to fully elucidate the mechanisms underlying these observations and to explore the potential therapeutic value of targeting CD74 and IL-13 in COVID-19.
Collapse
Affiliation(s)
- Nikola Ralchev Ralchev
- Department of Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | | | - Yana Valerieva Doneva
- Department of Clinical Laboratory and Immunology, Military Medical Academy, 1606 Sofia, Bulgaria
| | - Nikolina Mihaylova
- Department of Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | | | - Andrey Ivanov Tchorbanov
- Department of Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | - José Francisco Munoz-Valle
- University Center for Health Science, Department of Molecular Biology and Genomics, University of Guadalajara, Guadalajara 49000, Jalisco, Mexico
| | - Maria Cristina Petralia
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Paola Checconi
- Department of Human Sciences and Quality of Life Promotion, San Raffaele University, 20132 Rome, Italy
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| |
Collapse
|