1
|
Yan W, Ni T, Zhang Q, Sun X, Xu Z, Li X, Yi M, Wang Y, Zhang H, Shi J, Zhu Z. MCC950 promotes diabetic wound healing through modulating macrophage polarization in an MDSC-dependent manner. Int Immunopharmacol 2024; 142:112983. [PMID: 39217887 DOI: 10.1016/j.intimp.2024.112983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 08/13/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
Diabetic foot ulcers (DFUs) are serious skin injuries whereby the wound healing process is frequently stalled in the inflammatory phase. Currently, there is a lack of effective therapeutic strategies. MCC950, a highly selective nod-like receptor family pyrin domain containing 3 (NLRP3) inhibitor, has been reported to show strong anti-inflammation effects in many diseases. In this study, we unveiled the role of MCC950 in DFU mice model and its underlying molecular mechanisms. MCC950 could significantly accelerate diabetic wound healing, as shown by shortened healing time and better healing quality. Moreover, increased M2 phenotype macrophages and decreased pro-inflammatory genes were observed in MCC950-treated DFU mice. Additionally, myeloid-derived suppressor cells (MDSCs) were significantly increased in blood, spleen and wound tissues at different time courses. Specifically, MCC950 could recruit more MDSCs in an early phase in DFU mice, exerting an anti-inflammation effect. We identified the cell crosstalk between macrophages and MDSCs with MCC950 treatment process. Depleting MDSCs in vivo could eliminate the therapeutic effect of MCC950 on diabetic wound healing through inhibiting M2 macrophage polarization. Besides, MDSCs isolated from the wounds of MCC950 or saline treated mice were cocultured with bone marrow derived macrophage (BMDM) in a transwell system. Results confirmed that MDSCs sorted from MCC950 treated mice caused a significant increased percentage of M2 macrophages. Collectively, our findings suggest that the administration of MCC950 has the potential to accelerate diabetic wound healing by promoting M2 macrophage polarization in an MDSC-dependent manner. This study provides valuable insights into the utilization of pharmacological agents for DFU treatment.
Collapse
Affiliation(s)
- Wei Yan
- Department of Burn and Plastic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, PR China
| | - Tianyi Ni
- Department of Burn and Plastic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, PR China
| | - Qian Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, PR China
| | - Xiaowei Sun
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, PR China
| | - Zibo Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, PR China
| | - Xiangyu Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, PR China
| | - Min Yi
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, PR China
| | - Yingying Wang
- Department of Burn and Plastic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, PR China
| | - Hao Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, PR China.
| | - Jingping Shi
- Department of Burn and Plastic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, PR China.
| | - Zhechen Zhu
- Department of Burn and Plastic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, PR China.
| |
Collapse
|
2
|
Qi Y, Zhang L, Liu Y, Li Y, Liu Y, Zhang Z. Targeted modulation of myeloid-derived suppressor cells in the tumor microenvironment: Implications for cancer therapy. Biomed Pharmacother 2024; 180:117590. [PMID: 39423752 DOI: 10.1016/j.biopha.2024.117590] [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: 08/05/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) represent a heterogeneous population of immature myeloid cells originating from the bone marrow, known for their potent immunosuppressive functions that contribute to tumor immune evasion and progression. This paper provides a comprehensive analysis of the multifaceted interactions between MDSCs and tumors, exploring their distinct phenotypes and immunosuppressive mechanisms. Key roles of MDSCs in tumor biology are discussed, including their involvement in the formation of the pre-metastatic niche, facilitation of angiogenesis, enhancement of vascular permeability, suppression of tumor cell apoptosis, and promotion of resistance to cancer therapies. Additionally, the review highlights recent advances in the development of MDSC-targeting therapies, with a focus on their potential to enhance anti-tumor immunity. The therapeutic potential of Traditional Chinese Medicine (TCM) in modulating MDSC quantity and function is also explored, suggesting a novel approach to cancer treatment by integrating traditional and modern therapeutic strategies.
Collapse
Affiliation(s)
- Yafeng Qi
- Clinical School of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu 730000, China.
| | - Liying Zhang
- School of Integrative Chinese and Western Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu 730000, China.
| | - Yeyuan Liu
- Clinical School of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu 730000, China.
| | - Yangyang Li
- Clinical School of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu 730000, China.
| | - Yongqi Liu
- School of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu 730000, China.
| | - Zhiming Zhang
- Department of Oncology, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, Gansu 730000, China.
| |
Collapse
|
3
|
Pazhouhesh Far N, Hajiheidari Varnousafaderani M, Faghihkhorasani F, Etemad S, Abdulwahid AHRR, Bakhtiarinia N, Mousaei A, Dortaj E, Karimi S, Ebrahimi N, Aref AR. Breaking the barriers: Overcoming cancer resistance by targeting the NLRP3 inflammasome. Br J Pharmacol 2024. [PMID: 39394867 DOI: 10.1111/bph.17352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 08/06/2024] [Accepted: 08/14/2024] [Indexed: 10/14/2024] Open
Abstract
Inflammation has a pivotal role in the initiation and progression of various cancers, contributing to crucial processes such as metastasis, angiogenesis, cell proliferation and invasion. Moreover, the release of cytokines mediated by inflammation within the tumour microenvironment (TME) has a crucial role in orchestrating these events. The activation of inflammatory caspases, facilitated by the recruitment of caspase-1, is initiated by the activation of pattern recognition receptors on the immune cell membrane. This activation results in the production of proinflammatory cytokines, including IL-1β and IL-18, and participates in diverse biological processes with significant implications. The NOD-Like Receptor Protein 3 (NLRP3) inflammasome holds a central role in innate immunity and regulates inflammation through releasing IL-1β and IL-18. Moreover, it interacts with various cellular compartments. Recently, the mechanisms underlying NLRP3 inflammasome activation have garnered considerable attention. Disruption in NLRP3 inflammasome activation has been associated with a spectrum of inflammatory diseases, encompassing diabetes, enteritis, neurodegenerative diseases, obesity and tumours. The NLRP3 impact on tumorigenesis varies across different cancer types, with contrasting roles observed. For example, colorectal cancer associated with colitis can be suppressed by NLRP3, whereas gastric and skin cancers may be promoted by its activity. This review provides comprehensive insights into the structure, biological characteristics and mechanisms of the NLRP3 inflammasome, with a specific focus on the relationship between NLRP3 and tumour-related immune responses, and TME. Furthermore, the review explores potential strategies for targeting cancers via NLRP3 inflammasome modulation. This encompasses innovative approaches, including NLRP3-based nanoparticles, gene-targeted therapy and immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Nazanin Pazhouhesh Far
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Science, Islamic Azad University, Tehran, Iran
| | | | | | - Sareh Etemad
- Department of Pathology, Faculty of Anatomical Pathology, Ghaem Hospital, University of Medicine, Mashhad, Iran
| | | | | | - Afsaneh Mousaei
- Department of Biology, College of Science, Qaemshahr Branch, Islamic Azad University, Qaem Shahr, Iran
| | - Elahe Dortaj
- Department of Ergonomics, School of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soroush Karimi
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Nasim Ebrahimi
- Genetics Division, Department of Cell and Molecular Biology and Microbiology, Faculty of Science and Technology, University of Isfahan, Isfahan, Iran
| | - Amir Reza Aref
- Mass General Cancer Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
4
|
Li LR, Chen L, Sun ZJ. Igniting hope: Harnessing NLRP3 inflammasome-GSDMD-mediated pyroptosis for cancer immunotherapy. Life Sci 2024; 354:122951. [PMID: 39127315 DOI: 10.1016/j.lfs.2024.122951] [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: 05/10/2024] [Revised: 07/19/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
In the contemporary landscape of oncology, immunotherapy, represented by immune checkpoint blockade (ICB) therapy, stands out as a beacon of innovation in cancer treatment. Despite its promise, the therapy's progression is hindered by suboptimal clinical response rates. Addressing this challenge, the modulation of the NLRP3 inflammasome-GSDMD-mediated pyroptosis pathway holds promise as a means to augment the efficacy of immunotherapy. In the pathway, the NLRP3 inflammasome serves as a pivotal molecular sensor that responds to inflammatory stimuli within the organism. Its activation leads to the release of cytokines interleukin 1β and interleukin 18 through the cleavage of GSDMD, thereby forming membrane pores and potentially resulting in pyroptosis. This cascade of processes exerts a profound impact on tumor development and progression, with its function and expression exhibiting variability across different tumor types and developmental stages. Consequently, understanding the specific roles of the NLRP3 inflammasome and GSDMD-mediated pyroptosis in diverse tumors is imperative for comprehending tumorigenesis and crafting precise therapeutic strategies. This review aims to elucidate the structure and activation mechanisms of the NLRP3 inflammasome, as well as the induction mechanisms of GSDMD-mediated pyroptosis. Additionally, we provide a comprehensive overview of the involvement of this pathway in various cancer types and its applications in tumor immunotherapy, nanotherapy, and other fields. Emphasis is placed on the feasibility of leveraging this approach to enhance ICB therapy within the field of immunotherapy. Furthermore, we discuss the potential applications of this pathway in other immunotherapy methods, such as chimeric antigen receptor T-cell (CAR-T) therapy and tumor vaccines.
Collapse
Affiliation(s)
- Ling-Rui Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Lei Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
| | - Zhi-Jun Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
| |
Collapse
|
5
|
Zhou Q, Guo Y, Tian Z, Qiu Y, Liu Y, Liu Q, Liu Y, Yang Y, Shi L, Li X, Gao G, Fan S, Zeng Z, Xiong W, Tan M, Li G, Zhang W. PLUNC inhibits invasion and metastasis in nasopharyngeal carcinoma by inhibiting NLRP3 inflammasome activation. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167352. [PMID: 39004379 DOI: 10.1016/j.bbadis.2024.167352] [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: 02/21/2024] [Revised: 06/26/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024]
Abstract
Nasopharyngeal carcinoma (NPC) is a malignant tumor that occurs in the nasopharynx. Palate, lung, and nasal epithelium clone (PLUNC) has been identified as an early secreted protein that is specifically expressed in the nasopharynx. The aim of this study was to determine the role and mechanism of PLUNC in NPC. We used mRNA sequencing (seq) combined with ribosome-nascent chain complex (RNC)-seq to determine the biological role of PLUNC. The expression of epithelial-to-mesenchymal transition (EMT)-related molecules was detected by western blotting. Then, cell migration and invasion were detected by wound healing and Transwell chamber assays. NPC cells were injected into the tail vein of nude mice to explore the biological role of PLUNC in vivo. The sequencing results showed that PLUNC inhibited the progression of NPC and its expression was correlated with that of NOD-like receptors. Experiments confirmed that PLUNC inhibited the invasion and metastasis of NPC cells by promoting the ubiquitination degradation of NLRP3. PLUNC overexpression in combination with the treatment by MCC950, an inhibitor of NLRP3 inflammasome activation, was most effective in inhibiting NPC invasion and metastasis. In vivo experiments also confirmed that the combination of PLUNC overexpression and MCC950 treatment effectively inhibited the lung metastasis of NPC cells. In summary, our research suggested that PLUNC inhibited the invasion and metastasis of NPC by inhibiting NLRP3 inflammasome activation, and targeting the PLUNC-NLRP3 inflammasome axis could provide a new strategy for the diagnosis and treatment of NPC patients.
Collapse
Affiliation(s)
- Qing Zhou
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Clinical Laboratory, First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China; Department of Medical Laboratory Science, Xiangya Medical College, Central South University, Changsha, Hunan, China
| | - Yilin Guo
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Medical Laboratory Science, Xiangya Medical College, Central South University, Changsha, Hunan, China
| | - Ziying Tian
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Medical Laboratory Science, Xiangya Medical College, Central South University, Changsha, Hunan, China
| | - Yanbing Qiu
- Clinical Laboratory, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Ying Liu
- Department of Clinical Laboratory, Zhengzhou Orthopaedics Hospital, Zhengzhou, Henan, China
| | - Qingluan Liu
- Changsha Hospital for Maternal and Child Health Care, Changsha, Hunan, China
| | - Yijun Liu
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuqin Yang
- Shenzhen Maternity & Child Healthcare Hospital Clinical Laboratory, Shenzhen, Guangdong, China
| | - Lei Shi
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiayu Li
- Hunan Provincial People's Hospital, Changsha, Hunan, China
| | - Ge Gao
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Medical Laboratory Science, Xiangya Medical College, Central South University, Changsha, Hunan, China
| | - Songqing Fan
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
| | - Ming Tan
- Graduate Institute of Biomedical Sciences, China Medical University, Taiwan; Research Center for Cancer Biology, China Medical University, Taiwan
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
| | - Wenling Zhang
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Medical Laboratory Science, Xiangya Medical College, Central South University, Changsha, Hunan, China.
| |
Collapse
|
6
|
Liu Z, Zheng Y, Yuan M, Zhang G, Yang G. Association of CTACK, IL-2, and IL-13 with increased risk of lung cancer: A Mendelian randomization study. Cytokine 2024; 181:156680. [PMID: 38885591 DOI: 10.1016/j.cyto.2024.156680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/03/2024] [Accepted: 06/13/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND In recent years, relevant studies have reported that inflammatory cytokines are related to the occurrence of cancer. However, the correlation with lung cancer is not clear. This study used the Mendelian random grouping method to investigate the correlation between inflammatory factors and lung cancer in different populations. METHODS We obtained the single nucleotide polymorphisms (SNPs) of inflammatory cytokines through the open database and the SNPs of lung cancer (European and East Asian) through the IEU OpenGWAS project. Inverse variance-weighted (IVW) MR analyses were used to determine the causalities of exposures and outcomes. Supplementary analyses were also performed using weighted median and MR-Egger regressions. Afterward, sensitivity analyses were performed to test the robustness. Search the ChEMBL database for target drugs and indications for CTACK, IL-2, and IL-13. RESULTS By IVW method, we found that CTACK, IL-2, and IL-13 were associated with an increased risk of lung cancer in the European population (CTACK, OR = 1.098, 95 % CI 1.001-1.204, P = 0.047; IL-2, OR = 1.112, 95 % CI 1.009-1.225, P = 0.032; IL-13, OR = 1.068, 95 % CI 1.007-1.132, P = 0.029), while only IL-13 was associated with an increased risk of lung cancer in the East Asian population (IL-13, OR = 1.110, 95 % CI 1.010-1.220, P = 0.030). The weighted median and MR-Egger regression methods were in the same direction as the IVW effect sizes. Furthermore, no evidence of multidirectionality was detected using the MR-Egger intercept as a sensitivity analysis. Currently, there are no approved or phase III studied indications for CTACK, IL-2, and IL-13 targets in lung cancer. CONCLUSION The study outcomes supported that the inflammatory cytokines CTACK, IL-2, and IL-13 increase the risk of lung cancer. There is a lack of indications for drugs in these three targets. We explored the causal relationship between inflammatory cytokines and lung cancer, providing a basis for future cancer prediction models and targets for anti-tumor therapy.
Collapse
Affiliation(s)
- Zishen Liu
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Yingying Zheng
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Mengqi Yuan
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Ganlin Zhang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
| | - Guowang Yang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
| |
Collapse
|
7
|
Vidana Gamage HE, Shahoei SH, Wang Y, Jacquin E, Weisser E, Bautista RO, Henn MA, Schane CP, Nelczyk AT, Ma L, Das Gupta A, Bendre SV, Nguyen T, Tiwari S, Tjoanda E, Krawczynska N, He S, Albright ST, Farmer R, Smith AJ, Fink EC, Chen H, Sverdlov M, Gann PH, Boidot R, Vegran F, Fanning SW, Hergenrother PJ, Apetoh L, Nelson ER. NR0B2 re-educates myeloid immune cells to reduce regulatory T cell expansion and progression of breast and other solid tumors. Cancer Lett 2024; 597:217042. [PMID: 38908543 DOI: 10.1016/j.canlet.2024.217042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 06/24/2024]
Abstract
Although survival from breast cancer has dramatically increased, many will develop recurrent, metastatic disease. Unfortunately, survival for this stage of disease remains very low. Activating the immune system has incredible promise since it has the potential to be curative. However, immune checkpoint blockade (ICB) which works through T cells has been largely disappointing for metastatic breast cancer. One reason for this is a suppressive myeloid immune compartment that is unaffected by ICB. Cholesterol metabolism and proteins involved in cholesterol homeostasis play important regulatory roles in myeloid cells. Here, we demonstrate that NR0B2, a nuclear receptor involved in negative feedback of cholesterol metabolism, works in several myeloid cell types to impair subsequent expansion of regulatory T cells (Tregs); Tregs being a subset known to be highly immune suppressive and associated with poor therapeutic response. Within myeloid cells, NR0B2 serves to decrease many aspects of the inflammasome, ultimately resulting in decreased IL1β; IL1β driving Treg expansion. Importantly, mice lacking NR0B2 exhibit accelerated tumor growth. Thus, NR0B2 represents an important node in myeloid cells dictating ensuing Treg expansion and tumor growth, thereby representing a novel therapeutic target to re-educate these cells, having impact across different solid tumor types. Indeed, a paper co-published in this issue demonstrates the therapeutic utility of targeting NR0B2.
Collapse
Affiliation(s)
- Hashni Epa Vidana Gamage
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Sayyed Hamed Shahoei
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Yu Wang
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | | | - Erin Weisser
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Rafael O Bautista
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Madeline A Henn
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Claire P Schane
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Adam T Nelczyk
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Liqian Ma
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Anasuya Das Gupta
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Shruti V Bendre
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Tiffany Nguyen
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Srishti Tiwari
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Evelyn Tjoanda
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Natalia Krawczynska
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Sisi He
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Samuel T Albright
- Department of Chemistry, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Rachel Farmer
- Department of Chemistry, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Amanda J Smith
- Department of Chemistry, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Emma C Fink
- Department of Cancer Biology, Loyola University Chicago Health Sciences Campus, Illinois, USA
| | - Hong Chen
- Food Science & Human Nutrition, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Maria Sverdlov
- Research Histology and Tissue Imaging Core, University of Illinois at Chicago, Illinois, USA
| | - Peter H Gann
- Research Histology and Tissue Imaging Core, University of Illinois at Chicago, Illinois, USA; Department of Pathology, University of Illinois at Chicago, Illinois, USA
| | - Romain Boidot
- Unit of Molecular Biology, Department of Biology and Pathology of Tumors, Georges-Francois Leclerc Cancer Center, Dijon, France; ICMUB UMR CNRS 6302, Dijon, France
| | | | - Sean W Fanning
- Department of Cancer Biology, Loyola University Chicago Health Sciences Campus, Illinois, USA
| | - Paul J Hergenrother
- Department of Chemistry, University of Illinois at Urbana-Champaign, Illinois, USA; Carl R. Woese Institute for Genomic Biology- Anticancer Discovery from Pets to People, University of Illinois at Urbana-Champaign, Illinois, USA; Cancer Center at Illinois, University of Illinois Urbana-Champaign, University of Illinois at Urbana-Champaign, Illinois, USA
| | | | - Erik R Nelson
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Illinois, USA; Carl R. Woese Institute for Genomic Biology- Anticancer Discovery from Pets to People, University of Illinois at Urbana-Champaign, Illinois, USA; Cancer Center at Illinois, University of Illinois Urbana-Champaign, University of Illinois at Urbana-Champaign, Illinois, USA; Division of Nutritional Sciences, University of Illinois Urbana-Champaign, University of Illinois at Urbana-Champaign, Illinois, USA.
| |
Collapse
|
8
|
Tengesdal IW, Banks M, Dinarello CA, Marchetti C. Screening NLRP3 drug candidates in clinical development: lessons from existing and emerging technologies. Front Immunol 2024; 15:1422249. [PMID: 39188718 PMCID: PMC11345644 DOI: 10.3389/fimmu.2024.1422249] [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: 04/23/2024] [Accepted: 07/11/2024] [Indexed: 08/28/2024] Open
Abstract
Decades of evidence positioned IL-1β as a master regulatory cytokine in acute and chronic inflammatory diseases. Approved biologics aimed at inhibiting IL-1 signaling have shown efficacy but variable safety. More recently, targeting NLRP3 activation, an upstream mediator of IL-1β, has garnered the most attention. Aberrant NLRP3 activation has been demonstrated to participate in the progression of several pathological conditions from neurogenerative diseases to cardio-metabolic syndromes and cancer. Pharmacological and genetic strategies aimed to limit NLRP3 function have proven effective in many preclinical models of diseases. These evidences have lead to a significant effort in the generation and clinical testing of small orally active molecules that can target NLRP3. In this report, we discuss different properties of these molecules with translational potential and describe the technologies currently available to screen NLRP3 targeting molecules highlighting advantages and limitations of each method.
Collapse
Affiliation(s)
- Isak W. Tengesdal
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
| | - Migachelle Banks
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
| | - Charles A. Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
| | - Carlo Marchetti
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
- Department of Research, Rocky Mountain Regional Veteran Affairs (VA) Medical Center, Aurora, CO, United States
| |
Collapse
|
9
|
Kang Q, He L, Zhang Y, Zhong Z, Tan W. Immune-inflammatory modulation by natural products derived from edible and medicinal herbs used in Chinese classical prescriptions. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155684. [PMID: 38788391 DOI: 10.1016/j.phymed.2024.155684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/29/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND Edible and medicinal herbs1 (EMHs) refer to a class of substances with dual attribution of food and medicine. These substances are traditionally used as food and also listed in many international pharmacopoeias, including the European Pharmacopoeia, the United States Pharmacopoeia, and the Chinese Pharmacopoeia. Some classical formulas that are widely used in traditional Chinese medicine include a series of EMHs, which have been shown to be effective with obvious characteristics and advantages. Notably, these EMHs and Chinese classical prescriptions2 (CCPs) have also attracted attention in international herbal medicine research because of their low toxicity and high efficiency as well as the rich body of experience for their long-term clinical use. PURPOSE Our purpose is to explore the potential therapeutic effect of EMHs with immune-inflammatory modulation for the study of modern cancer drugs. STUDY DESIGN In the present study, we present a detailed account of some EMHs used in CCPs that have shown considerable research potential in studies exploring modern drugs with immune-inflammatory modulation. METHODS Approximately 500 publications in the past 30 years were collected from PubMed, Web of Science and ScienceDirect using the keywords, such as natural products, edible and medicinal herbs, Chinese medicine, classical prescription, immune-inflammatory, tumor microenvironment and some related synonyms. The active ingredients instead of herbal extracts or botanical mixtures were focused on and the research conducted over the past decade were discussed emphatically and analyzed comprehensively. RESULTS More than ten natural products derived from EMHs used in CCPs are discussed and their immune-inflammatory modulation activities, including enhancing antitumor immunity, regulating inflammatory signaling pathways, lowering the proportion of immunosuppressive cells, inhibiting the secretion of proinflammatory cytokines, immunosuppressive factors, and inflammatory mediators, are summarized. CONCLUSION Our findings demonstrate the immune-inflammatory modulating role of those EMHs used in CCPs and provide new ideas for cancer treatment in clinical settings.
Collapse
Affiliation(s)
- Qianming Kang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Luying He
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Yang Zhang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Zhangfeng Zhong
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China.
| | - Wen Tan
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
10
|
Schlenker R, Schwalie PC, Dettling S, Huesser T, Irmisch A, Mariani M, Martínez Gómez JM, Ribeiro A, Limani F, Herter S, Yángüez E, Hoves S, Somandin J, Siebourg-Polster J, Kam-Thong T, de Matos IG, Umana P, Dummer R, Levesque MP, Bacac M. Myeloid-T cell interplay and cell state transitions associated with checkpoint inhibitor response in melanoma. MED 2024; 5:759-779.e7. [PMID: 38593812 DOI: 10.1016/j.medj.2024.03.015] [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: 06/14/2023] [Revised: 11/23/2023] [Accepted: 03/17/2024] [Indexed: 04/11/2024]
Abstract
BACKGROUND The treatment of melanoma, the deadliest form of skin cancer, has greatly benefited from immunotherapy. However, many patients do not show a durable response, which is only partially explained by known resistance mechanisms. METHODS We performed single-cell RNA sequencing of tumor immune infiltrates and matched peripheral blood mononuclear cells of 22 checkpoint inhibitor (CPI)-naive stage III-IV metastatic melanoma patients. After sample collection, the same patients received CPI treatment, and their response was assessed. FINDINGS CPI responders showed high levels of classical monocytes in peripheral blood, which preferentially transitioned toward CXCL9-expressing macrophages in tumors. Trajectories of tumor-infiltrating CD8+ T cells diverged at the level of effector memory/stem-like T cells, with non-responder cells progressing into a state characterized by cellular stress and apoptosis-related gene expression. Consistently, predicted non-responder-enriched myeloid-T/natural killer cell interactions were primarily immunosuppressive, while responder-enriched interactions were supportive of T cell priming and effector function. CONCLUSIONS Our study illustrates that the tumor immune microenvironment prior to CPI treatment can be indicative of response. In perspective, modulating the myeloid and/or effector cell compartment by altering the described cell interactions and transitions could improve immunotherapy response. FUNDING This research was funded by Roche Pharma Research and Early Development.
Collapse
Affiliation(s)
- Ramona Schlenker
- Roche Innovation Center Munich, Roche Pharma Research and Early Development (pRED), Penzberg, Germany.
| | | | - Steffen Dettling
- Roche Innovation Center Munich, Roche Pharma Research and Early Development (pRED), Penzberg, Germany
| | - Tamara Huesser
- Roche Innovation Center Zurich, pRED, Schlieren, Switzerland
| | - Anja Irmisch
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Marisa Mariani
- Roche Innovation Center Zurich, pRED, Schlieren, Switzerland
| | - Julia M Martínez Gómez
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Alison Ribeiro
- Roche Innovation Center Zurich, pRED, Schlieren, Switzerland
| | - Florian Limani
- Roche Innovation Center Zurich, pRED, Schlieren, Switzerland
| | - Sylvia Herter
- Roche Innovation Center Zurich, pRED, Schlieren, Switzerland
| | - Emilio Yángüez
- Roche Innovation Center Zurich, pRED, Schlieren, Switzerland
| | - Sabine Hoves
- Roche Innovation Center Munich, Roche Pharma Research and Early Development (pRED), Penzberg, Germany
| | - Jitka Somandin
- Roche Innovation Center Zurich, pRED, Schlieren, Switzerland
| | | | | | | | - Pablo Umana
- Roche Innovation Center Zurich, pRED, Schlieren, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Mitchell P Levesque
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Marina Bacac
- Roche Innovation Center Zurich, pRED, Schlieren, Switzerland
| |
Collapse
|
11
|
Papa V, Li Pomi F, Borgia F, Vaccaro M, Pioggia G, Gangemi S. Alarmins in cutaneous malignant melanoma: An updated overview of emerging evidence on their pathogenetic, diagnostic, prognostic, and therapeutic role. J Dermatol 2024; 51:927-938. [PMID: 38775220 PMCID: PMC11483971 DOI: 10.1111/1346-8138.17278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/04/2024] [Accepted: 05/07/2024] [Indexed: 07/04/2024]
Abstract
Malignant cutaneous melanoma is the leading cause of death for skin cancer to date, with globally increasing incidence rates. In this epidemiological scenario, international scientific research is exerting efforts to identify new clinical strategies aimed at the prognostic amelioration of the disease. Very promising and groundbreaking in this context is the scientific interest related to alarmins and their pioneering utility in the setting of the pathogenetic understanding, diagnosis, prognosis, and therapy for malignant cutaneous melanoma. However, the scientific investigations on this matter should not overlook their still well-presented dual and contradictory role. The aim of our critical analysis is to provide an up-to-date overview of the emerging evidence concerning the dichotomous role of alarmins in the aforementioned clinical settings. Our literature revision was based on the extensive body of both preclinical and clinical findings published on the PubMed database over the past 5 years. In addition to this, we offer a special focus on potentially revolutionary new therapeutic frontiers, which, on the strength of their earliest successes in other clinical areas, could inaugurate a new era of personalized and precision medicine in the field of dermato-oncology.
Collapse
Affiliation(s)
- Vincenzo Papa
- Department of Clinical and Experimental Medicine, School and Operative Unit of Allergy and Clinical ImmunologyUniversity of MessinaMessinaItaly
| | - Federica Li Pomi
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.)University of PalermoPalermoItaly
| | - Francesco Borgia
- Department of Clinical and Experimental Medicine, Section of DermatologyUniversity of MessinaMessinaItaly
| | - Mario Vaccaro
- Department of Clinical and Experimental Medicine, Section of DermatologyUniversity of MessinaMessinaItaly
| | - Giovanni Pioggia
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR)MessinaItaly
| | - Sebastiano Gangemi
- Department of Clinical and Experimental Medicine, School and Operative Unit of Allergy and Clinical ImmunologyUniversity of MessinaMessinaItaly
| |
Collapse
|
12
|
Zhang T, Wen R, Fan H, Yu Y, Jia H, Peng Z, Zhou L, Yu G, Zhang W. Impact and potential value of immunosenescence on solid gastrointestinal tumors. Front Immunol 2024; 15:1375730. [PMID: 39007138 PMCID: PMC11239362 DOI: 10.3389/fimmu.2024.1375730] [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: 01/24/2024] [Accepted: 06/17/2024] [Indexed: 07/16/2024] Open
Abstract
Solid gastrointestinal tumors often respond poorly to immunotherapy for the complex tumor microenvironment (TME), which is exacerbated by immune system alterations. Immunosenescence is the process of increased diversification of immune genes due to aging and other factors, leading to a decrease in the recognition function of the immune system. This process involves immune organs, immune cells, and the senescence-associated secretory phenotype (SASP). The most fundamental change is DNA damage, resulting in TME remodeling. The main manifestations are worsening inflammation, increased immunosuppressive SASP production, decreased immune cell antitumor activity, and the accumulation of tumor-associated fibroblasts and myeloid-derived suppressor cells, making antitumor therapy less effective. Senotherapy strategies to remove senescent cells and block key senescence processes can have synergistic effects with other treatments. This review focuses on immunoenescence and its impact on the solid TME. We characterize the immunosenescent TME and discuss future directions for antitumor therapies targeting senescence.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Leqi Zhou
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Guanyu Yu
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Wei Zhang
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
13
|
Gu X, Li Z, Su J. Air pollution and skin diseases: A comprehensive evaluation of the associated mechanism. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 278:116429. [PMID: 38718731 DOI: 10.1016/j.ecoenv.2024.116429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/28/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024]
Abstract
Air pollutants deteriorate the survival environment and endanger human health around the world. A large number of studies have confirmed that air pollution jeopardizes multiple organs, such as the cardiovascular, respiratory, and central nervous systems. Skin is the largest organ and the first barrier that protects us from the outside world. Air pollutants such as particulate matter (PM), polycyclic aromatic hydrocarbons (PAHs), volatile organic compounds (VOCs) will affect the structure and function of the skin and bring about the development of inflammatory skin diseases (atopic dermatitis (AD), psoriasis), skin accessory diseases (acne, alopecia), auto-immune skin diseases (cutaneous lupus erythematosus(CLE) scleroderma), and even skin tumors (melanoma, basal cell carcinoma (BCC), squamous-cell carcinoma (SCC)). Oxidative stress, skin barrier damage, microbiome dysbiosis, and skin inflammation are the pathogenesis of air pollution stimulation. In this review, we summarize the current evidence on the effects of air pollution on skin diseases and possible mechanisms to provide strategies for future research.
Collapse
Affiliation(s)
- Xiaoyu Gu
- Department of Dermatology | Hunan Engineering Research Center of Skin Health and Disease | Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha 410008, China; Furong Laboratory, Changsha, Hunan 410008, China
| | - Zhengrui Li
- XiangYa School of Medicine, Central South University, Changsha 410008, China
| | - Juan Su
- Department of Dermatology | Hunan Engineering Research Center of Skin Health and Disease | Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha 410008, China; Furong Laboratory, Changsha, Hunan 410008, China.
| |
Collapse
|
14
|
Fan J, Zhu J, Zhu H, Xu H. Potential therapeutic targets in myeloid cell therapy for overcoming chemoresistance and immune suppression in gastrointestinal tumors. Crit Rev Oncol Hematol 2024; 198:104362. [PMID: 38614267 DOI: 10.1016/j.critrevonc.2024.104362] [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: 11/18/2023] [Revised: 03/26/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024] Open
Abstract
In the tumor microenvironment (TME), myeloid cells play a pivotal role. Myeloid-derived immunosuppressive cells, including tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs), are central components in shaping the immunosuppressive milieu of the tumor. Within the TME, a majority of TAMs assume an M2 phenotype, characterized by their pro-tumoral activity. These cells promote tumor cell growth, angiogenesis, invasion, and migration. In contrast, M1 macrophages, under appropriate activation conditions, exhibit cytotoxic capabilities against cancer cells. However, an excessive M1 response may lead to pro-tumoral inflammation. As a result, myeloid cells have emerged as crucial targets in cancer therapy. This review concentrates on gastrointestinal tumors, detailing methods for targeting macrophages to enhance tumor radiotherapy and immunotherapy sensitivity. We specifically delve into monocytes and tumor-associated macrophages' various functions, establishing an immunosuppressive microenvironment, promoting tumorigenic inflammation, and fostering neovascularization and stromal remodeling. Additionally, we examine combination therapeutic strategies.
Collapse
Affiliation(s)
- Jiawei Fan
- Department of Gastroenterology, The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130021, PR China
| | - Jianshu Zhu
- Department of Spine Surgery, The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130021, PR China
| | - He Zhu
- Department of Gastroenterology, The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130021, PR China
| | - Hong Xu
- Department of Gastroenterology, The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130021, PR China.
| |
Collapse
|
15
|
Zhu Y, Chen P, Hu B, Zhong S, Yan K, Wu Y, Li S, Yang Y, Xu Z, Lu Y, Ouyang Y, Bao H, Gu W, Wen L, Zhang Y. MDSC-targeting gold nanoparticles enhance PD-1 tumor immunotherapy by inhibiting NLRP3 inflammasomes. Biomaterials 2024; 307:122533. [PMID: 38493671 DOI: 10.1016/j.biomaterials.2024.122533] [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: 10/16/2023] [Revised: 02/20/2024] [Accepted: 03/09/2024] [Indexed: 03/19/2024]
Abstract
Myeloid-derived suppressor cells (MDSCs) play a crucial role in the immune escape mechanisms that limit the efficacy of immunotherapeutic strategies. In the tumor microenvironment, NLRP3 inflammasome-driven Interleukin-1β (IL-1β) production serves to dampen antitumor immune responses, promoting tumor growth, progression, and immunosuppression. In this study, we revealed that gold nanoparticles (Au NPs) with a size of 30 nm disrupted NLRP3 inflammasome, but not other inflammasomes, in bone marrow-derived macrophages through abrogating NLRP3-NEK7 interactions mediated by reactive oxygen species (ROS). Density functional theory (DFT) calculations provided insights into the mechanism underlying the exceptional ROS scavenging capabilities of Au NPs. Additionally, when coupled with H6, a small peptide targeting MDSCs, Au NPs demonstrated the capacity to effectively reduce IL-1β levels and diminish the MDSCs population in tumor microenvironment, leading to enhanced T cell activation and increased immunotherapeutic efficacy in mouse tumor models that are sensitive and resistant to PD-1 inhibition. Our findings unraveled a novel approach wherein peptide-modified Au NPs relieved the suppressive impact of the tumor microenvironment by inhibiting MDSCs-mediated IL-1β release, which is the first time reported the employing a nanostrategy at modulating MDSCs to reverse the immunosuppressive microenvironment and may hold promise as a potential therapeutic agent for cancer immunotherapy.
Collapse
Affiliation(s)
- Yangyang Zhu
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Pin Chen
- National Supercomputer Center in Guangzhou, School of Data and Computer Science, Sun Yat-Senedi University, 132 East Circle at University City, Guangzhou, 510006, China
| | - Bochuan Hu
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Suqin Zhong
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Kai Yan
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Yu Wu
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Shanshan Li
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Yinyin Yang
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Zexin Xu
- National Supercomputer Center in Guangzhou, School of Data and Computer Science, Sun Yat-Senedi University, 132 East Circle at University City, Guangzhou, 510006, China
| | - Yutong Lu
- National Supercomputer Center in Guangzhou, School of Data and Computer Science, Sun Yat-Senedi University, 132 East Circle at University City, Guangzhou, 510006, China
| | - Ying Ouyang
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China; China-Singapore International Joint Research Institute, Guangzhou, 510700, China
| | - Hui Bao
- Department of Oncology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Weiguang Gu
- Department of Oncology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510006, China.
| | - Longping Wen
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| | - Yunjiao Zhang
- School of Medicine, South China University of Technology, Guangzhou, 510006, China.
| |
Collapse
|
16
|
Silva RCMC. Mitochondria, Autophagy and Inflammation: Interconnected in Aging. Cell Biochem Biophys 2024; 82:411-426. [PMID: 38381268 DOI: 10.1007/s12013-024-01231-x] [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: 10/18/2023] [Accepted: 02/08/2024] [Indexed: 02/22/2024]
Abstract
In this manuscript, I discuss the direct link between abnormalities in inflammatory responses, mitochondrial metabolism and autophagy during the process of aging. It is focused on the cytosolic receptors nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) and cyclic GMP-AMP synthase (cGAS); myeloid-derived suppressor cells (MDSCs) expansion and their associated immunosuppressive metabolite, methyl-glyoxal, all of them negatively regulated by mitochondrial autophagy, biogenesis, metabolic pathways and its distinct metabolites.
Collapse
Affiliation(s)
- Rafael Cardoso Maciel Costa Silva
- Laboratory of Immunoreceptors and signaling, Instituto de Biofísica Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
17
|
Liu H, Huang W, Pu X, Chen Y, Zheng Y, Lei Y, Jiang T. miR-22 negatively regulating NOD-like receptor protein 3 gene in the proliferation, invasion, and migration of malignant melanoma cells. Postepy Dermatol Alergol 2024; 41:284-291. [PMID: 39027690 PMCID: PMC11253316 DOI: 10.5114/ada.2024.140521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 02/14/2024] [Indexed: 07/20/2024] Open
Abstract
Introduction Malignant melanoma (MM) is a highly aggressive skin tumour. Aim To investigate whether miR-22 is involved in the proliferation, invasion, and migration of melanoma cells (MCs) by negatively regulating NOD-like receptor protein 3 (NLRP3) gene. Material and methods Human MCs (WM239a) and human epidermal melanocytes (HEM) were used as study material. The expression levels of miR-22 and NLRP3 were detected by qRT-PCR. The expression of NLRP3 protein was determined by Western blot (WB) analysis. The effects of miR-22 and NLRP3 on the proliferation, invasion, and migration of MCs were evaluated by cell counting kit-8 (CCK-8), Transwell cell invasion assay, and scratch assay. Results The expression of miR-22 was clearly lower in WM239a than in HEM. Up-regulation of miR-22 expression in WM239a clearly raised the expression of miR-22, Caspase-1, and E-cadherin and the apoptotic rate of WM239a; however, the levels of interleukin-1β (IL-1β) and NLRP3, cell proliferation activity, invasion and migration ability were clearly decreased. The negative regulation of NLRP3 by miR-22 may play a major role in activities of MM. Conclusions Further studies will help to reveal the molecular details of this regulatory mechanism and provide new therapeutic strategies.
Collapse
Affiliation(s)
- Hongyan Liu
- Department of Burn Plastic and Aesthetic Surgery, Nanchong Central Hospital, Second Clinical Medical College of North Sichuan Medical College, Nanchong, Sichuan Province, China
| | - Wenlian Huang
- Department of Critical Care Medicine, Nanchong Central Hospital, Second Clinical Medical College of North Sichuan Medical College, Nanchong, Sichuan Province, China
| | - Xiaoshu Pu
- Department of Burn Plastic and Aesthetic Surgery, Nanchong Central Hospital, Second Clinical Medical College of North Sichuan Medical College, Nanchong, Sichuan Province, China
| | - Yi Chen
- Department of Stomatology, Nanchong Central Hospital, Second Clinical Medical College of North Sichuan Medical College, Nanchong, Sichuan Province, China
| | - Yinbin Zheng
- Department of Thoracic Surgery, Nanchong Central Hospital, Second Clinical Medical College of North Sichuan Medical College, Nanchong, Sichuan Province, China
| | - Ying Lei
- Department of Burn Plastic and Aesthetic Surgery, Nanchong Central Hospital, Second Clinical Medical College of North Sichuan Medical College, Nanchong, Sichuan Province, China
| | - Ting Jiang
- Department of Burn Plastic and Aesthetic Surgery, Nanchong Central Hospital, Second Clinical Medical College of North Sichuan Medical College, Nanchong, Sichuan Province, China
| |
Collapse
|
18
|
Belicard F, Tarte K, Tadié JM. NLRP3 inflammasome in sepsis: don't overlook the small steps-they can make a big difference! J Leukoc Biol 2024; 115:999-1001. [PMID: 38498595 DOI: 10.1093/jleuko/qiae056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 02/16/2024] [Accepted: 02/19/2024] [Indexed: 03/20/2024] Open
Abstract
"In sepsis, persistent activation of NLRP3 is associated with expansion of both monocytic and granulocytic MDSCs, along with high plasma concentration of IL-10."
Collapse
Affiliation(s)
- Felicie Belicard
- Department of Infectious Diseases and Intensive Care Unit, CHU Rennes, 35000 Rennes, France
- French Blood Establishment and SITI Laboratory, UMR U1236, INSERM, University of Rennes, University Hospital Center of Rennes, 35000 Rennes, France
| | - Karin Tarte
- Department of Infectious Diseases and Intensive Care Unit, CHU Rennes, 35000 Rennes, France
- French Blood Establishment and SITI Laboratory, UMR U1236, INSERM, University of Rennes, University Hospital Center of Rennes, 35000 Rennes, France
| | - Jean-Marc Tadié
- Department of Infectious Diseases and Intensive Care Unit, CHU Rennes, 35000 Rennes, France
- French Blood Establishment and SITI Laboratory, UMR U1236, INSERM, University of Rennes, University Hospital Center of Rennes, 35000 Rennes, France
- Inserm, Centre d'investigation clinique de Rennes (CIC1414), CHU Rennes, 35000 Rennes, France
| |
Collapse
|
19
|
Manica D, da Silva GB, Narzetti RA, Dallagnoll P, da Silva AP, Marafon F, Cassol J, de Souza Matias L, Zamoner A, de Oliveira Maciel SFV, Moreno M, Bagatini MD. Curcumin modulates purinergic signaling and inflammatory response in cutaneous metastatic melanoma cells. Purinergic Signal 2024:10.1007/s11302-024-10023-0. [PMID: 38801619 DOI: 10.1007/s11302-024-10023-0] [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: 12/14/2023] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
Cutaneous melanoma (CM) poses a therapeutic challenge due to its aggressive nature and often limited response to conventional treatments. Exploring novel therapeutic targets is essential, and natural compounds have emerged as potential candidates. This study aimed to elucidate the impact of curcumin, a natural compound known for its anti-inflammatory, antioxidant, and anti-tumor properties, on metastatic melanoma cells, focusing on the purinergic system and immune responses. Human melanoma cell line SK-Mel-28 were exposed to different curcumin concentrations for either 6 or 24 h, after which we assessed components related to the purinergic system and the inflammatory cascade. Using RT-qPCR, we assessed the gene expression of CD39 and CD73 ectonucleotidases, as well as adenosine deaminase (ADA). Curcumin effectively downregulated CD39, CD73, and ADA gene expression. Flow cytometry analysis revealed that curcumin significantly reduced CD39 and CD73 protein expression at specific concentrations. Moreover, the A2A receptor's protein expression decreased across all concentrations. Enzymatic activity assays demonstrated that curcumin modulated CD39, CD73, and ADA activities, with effects dependent on concentration and duration of treatment. Extracellular ATP levels increased after 24 h of curcumin treatment, emphasizing its role in modulating hydrolytic activity. Curcumin also displayed anti-inflammatory properties by reducing NLRP3 gene expression and impacting the levels of key inflammatory cytokines. In conclusion, this study unveils the potential of curcumin as a promising adjuvant in CM treatment. Curcumin modulates the expression and activity of crucial components of the purinergic system and exhibits anti-inflammatory effects, indicating its potential therapeutic role in combating CM. These findings underscore curcumin's promise and warrant further investigation in preclinical and clinical settings for melanoma management.
Collapse
Affiliation(s)
- Daiane Manica
- Department of Biochemistry, Biochemistry Graduate Program, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Gilnei Bruno da Silva
- Multicentric Graduate Program in Biochemistry and Molecular Biology, State University of Santa Catarina, Lages, SC, Brazil
| | - Rafael Antônio Narzetti
- Department of Biochemistry, Biochemistry Graduate Program, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Paula Dallagnoll
- Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapeco, SC, Brazil
| | - Alana Patrícia da Silva
- Department of Biochemistry, Biochemistry Graduate Program, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Filomena Marafon
- Department of Biochemistry, Biochemistry Graduate Program, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Joana Cassol
- Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapeco, SC, Brazil
| | - Letícia de Souza Matias
- Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapeco, SC, Brazil
| | - Ariane Zamoner
- Department of Biochemistry, Biochemistry Graduate Program, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | | | - Marcelo Moreno
- Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapeco, SC, Brazil.
| | - Margarete Dulce Bagatini
- Department of Biochemistry, Biochemistry Graduate Program, Federal University of Santa Catarina, Florianopolis, SC, Brazil.
- Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapeco, SC, Brazil.
| |
Collapse
|
20
|
Nie SC, Jing YH, Lu L, Ren SS, Ji G, Xu HC. Mechanisms of myeloid-derived suppressor cell-mediated immunosuppression in colorectal cancer and related therapies. World J Gastrointest Oncol 2024; 16:1690-1704. [PMID: 38764816 PMCID: PMC11099432 DOI: 10.4251/wjgo.v16.i5.1690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/30/2024] [Accepted: 03/11/2024] [Indexed: 05/09/2024] Open
Abstract
Severe immunosuppression is a hallmark of colorectal cancer (CRC). Myeloid-derived suppressor cells (MDSCs), one of the most abundant components of the tumor stroma, play an important role in the invasion, metastasis, and immune escape of CRC. MDSCs create an immunosuppressive microenvironment by inhibiting the proliferation and activation of immunoreactive cells, including T and natural killer cells, as well as by inducing the proliferation of immunosuppressive cells, such as regulatory T cells and tumor-associated macrophages, which, in turn, promote the growth of cancer cells. Thus, MDSCs are key contributors to the emergence of an immunosuppressive microenvironment in CRC and play an important role in the breakdown of antitumor immunity. In this narrative review, we explore the mechanisms through which MDSCs contribute to the immunosuppressive microenvironment, the current therapeutic approaches and technologies targeting MDSCs, and the therapeutic potential of modulating MDSCs in CRC treatment. This study provides ideas and methods to enhance survival rates in patients with CRC.
Collapse
Affiliation(s)
- Shu-Chang Nie
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yan-Hua Jing
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Lu Lu
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- Shanghai Frontiers Science Center of Disease and Syndrome Biology of Inflammatory Cancer Transformation, Shanghai 200032, China
| | - Si-Si Ren
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- Shanghai Frontiers Science Center of Disease and Syndrome Biology of Inflammatory Cancer Transformation, Shanghai 200032, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine), Shanghai 200032, China
| | - Han-Chen Xu
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- Shanghai Frontiers Science Center of Disease and Syndrome Biology of Inflammatory Cancer Transformation, Shanghai 200032, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine), Shanghai 200032, China
| |
Collapse
|
21
|
Shi F, Qiu H, Yan J, Ke C, Li Y. Effect of thymalfasin on myeloid-derived suppressor cells in patients with non-small cell lung cancer. Am J Transl Res 2024; 16:1790-1797. [PMID: 38883367 PMCID: PMC11170616 DOI: 10.62347/qsws7848] [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: 08/25/2023] [Accepted: 04/24/2024] [Indexed: 06/18/2024]
Abstract
OBJECTIVE To observe the effect of thymalfasin on myeloid-derived suppressor cells (MDSCs) subsets in peripheral blood of patients with non-small cell lung cancer (NSCLC). METHODS 50 cases of NSCLC (NSCLC group) diagnosed in Chest Hospital of Jiangxi Province were selected as the research subjects, and 50 healthy subjects who underwent physical examination in our hospital during the same period were selected as the healthy control group. The expression of HLA-DR-CD14-CD33+ MDSCs in peripheral blood mononuclear cells and tumor tissue single cell suspension of NSCLC patients before and after thymalfasin treatment was explored by flow cytometry. RESULTS The proportion of MDSCs in peripheral blood of NSCLC group was 1.70±0.52%, which was significantly higher than that in peripheral blood (0.51±0.15%) of healthy controls (P < 0.05). The proportion of HLA-DR-CD14-CD33+ MDSCs in the tissues of NSCLC group was 1.65±0.43% before treatment and 1.15±0.50% after treatment (P < 0.05). The proportion of MDSCs in peripheral blood of NSCLC patients before treatment was 1.70±0.52%, and that after treatment was 0.59±0.18% (P < 0.05). CONCLUSION Thymalfasin can reduce the number of MDSCs in peripheral blood mononuclear cells. The application of thymalfasin in the treatment of NSCLC patients can help to enhance the anti-tumor effect.
Collapse
Affiliation(s)
- Fang Shi
- Department of Oncology, Chest Hospital of Jiangxi Province Nanchang 330096, Jiangxi, China
| | - Huiping Qiu
- Department of Oncology, Chest Hospital of Jiangxi Province Nanchang 330096, Jiangxi, China
| | - Jinjin Yan
- Department of Oncology, Chest Hospital of Jiangxi Province Nanchang 330096, Jiangxi, China
| | - Changlin Ke
- Department of Oncology, Chest Hospital of Jiangxi Province Nanchang 330096, Jiangxi, China
| | - Yao Li
- Department of Oncology, Chest Hospital of Jiangxi Province Nanchang 330096, Jiangxi, China
| |
Collapse
|
22
|
Lu J, Luo Y, Rao D, Wang T, Lei Z, Chen X, Zhang B, Li Y, Liu B, Xia L, Huang W. Myeloid-derived suppressor cells in cancer: therapeutic targets to overcome tumor immune evasion. Exp Hematol Oncol 2024; 13:39. [PMID: 38609997 PMCID: PMC11010322 DOI: 10.1186/s40164-024-00505-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 03/31/2024] [Indexed: 04/14/2024] Open
Abstract
Paradoxically, tumor development and progression can be inhibited and promoted by the immune system. After three stages of immune editing, namely, elimination, homeostasis and escape, tumor cells are no longer restricted by immune surveillance and thus develop into clinical tumors. The mechanisms of immune escape include abnormalities in antitumor-associated immune cells, selection for immune resistance to tumor cells, impaired transport of T cells, and the formation of an immunosuppressive tumor microenvironment. A population of distinct immature myeloid cells, myeloid-derived suppressor cells (MDSCs), mediate immune escape primarily by exerting immunosuppressive effects and participating in the constitution of an immunosuppressive microtumor environment. Clinical trials have found that the levels of MDSCs in the peripheral blood of cancer patients are strongly correlated with tumor stage, metastasis and prognosis. Moreover, animal experiments have confirmed that elimination of MDSCs inhibits tumor growth and metastasis to some extent. Therefore, MDSCs may become the target of immunotherapy for many cancers, and eliminating MDSCs can help improve the response rate to cancer treatment and patient survival. However, a clear definition of MDSCs and the specific mechanism involved in immune escape are lacking. In this paper, we review the role of the MDSCs population in tumor development and the mechanisms involved in immune escape in different tumor contexts. In addition, we discuss the use of these cells as targets for tumor immunotherapy. This review not only contributes to a systematic and comprehensive understanding of the essential role of MDSCs in immune system reactions against tumors but also provides information to guide the development of cancer therapies targeting MDSCs.
Collapse
Affiliation(s)
- Junli Lu
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, Hubei, China
| | - Yiming Luo
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, Hubei, China
| | - Dean Rao
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, Hubei, China
| | - Tiantian Wang
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, Hubei, China
| | - Zhen Lei
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, Hubei, China
| | - Xiaoping Chen
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, Hubei, China
- Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, 430030, Hubei, China
| | - Bixiang Zhang
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, Hubei, China
- Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, 430030, Hubei, China
| | - Yiwei Li
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Bifeng Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Limin Xia
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Wenjie Huang
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, Hubei, China.
- Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, 430030, Hubei, China.
| |
Collapse
|
23
|
Yang S, Hu C, Chen X, Tang Y, Li J, Yang H, Yang Y, Ying B, Xiao X, Li SZ, Gu L, Zhu Y. Crosstalk between metabolism and cell death in tumorigenesis. Mol Cancer 2024; 23:71. [PMID: 38575922 PMCID: PMC10993426 DOI: 10.1186/s12943-024-01977-1] [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: 06/17/2023] [Accepted: 03/02/2024] [Indexed: 04/06/2024] Open
Abstract
It is generally recognized that tumor cells proliferate more rapidly than normal cells. Due to such an abnormally rapid proliferation rate, cancer cells constantly encounter the limits of insufficient oxygen and nutrient supplies. To satisfy their growth needs and resist adverse environmental events, tumor cells modify the metabolic pathways to produce both extra energies and substances required for rapid growth. Realizing the metabolic characters special for tumor cells will be helpful for eliminating them during therapy. Cell death is a hot topic of long-term study and targeting cell death is one of the most effective ways to repress tumor growth. Many studies have successfully demonstrated that metabolism is inextricably linked to cell death of cancer cells. Here we summarize the recently identified metabolic characters that specifically impact on different types of cell deaths and discuss their roles in tumorigenesis.
Collapse
Affiliation(s)
- Shichao Yang
- School of Medicine, Chongqing University, Chongqing, 400030, P. R. China
| | - Caden Hu
- School of Medicine, Chongqing University, Chongqing, 400030, P. R. China
| | - Xiaomei Chen
- School of Medicine, Chongqing University, Chongqing, 400030, P. R. China
| | - Yi Tang
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, P. R. China
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, Chongqing, P. R. China
| | - Juanjuan Li
- Department of breast and thyroid surgery, Renmin hospital of Wuhan University, Wuhan, 430060, P. R. China
| | - Hanqing Yang
- School of Medicine, Chongqing University, Chongqing, 400030, P. R. China
| | - Yi Yang
- Institute of Pathology and Southwest Cancer Center, The First Affiliated Hospital, Key Laboratory of Tumor Immunopathology, Third Military Medical University (Army Medical University, Ministry of Education of China, Chongqing, 400038, P. R. China
| | - Binwu Ying
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, P. R. China.
| | - Xue Xiao
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, P. R. China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, P. R. China.
| | - Shang-Ze Li
- School of Medicine, Chongqing University, Chongqing, 400030, P. R. China.
| | - Li Gu
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, P. R. China.
| | - Yahui Zhu
- School of Medicine, Chongqing University, Chongqing, 400030, P. R. China.
| |
Collapse
|
24
|
Xiong J, Cheng S, Gao X, Yu SH, Dai YT, Huang XY, Zhong HJ, Wang CF, Yi HM, Zhang H, Cao WG, Li R, Tang W, Zhao Y, Xu PP, Wang L, Zhao WL. Anti-metabolic agent pegaspargase plus PD-1 antibody sintilimab for first-line treatment in advanced natural killer T cell lymphoma. Signal Transduct Target Ther 2024; 9:62. [PMID: 38448403 PMCID: PMC10917752 DOI: 10.1038/s41392-024-01782-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 02/17/2024] [Accepted: 02/25/2024] [Indexed: 03/08/2024] Open
Abstract
Natural killer T cell lymphoma (NKTCL) is highly aggressive, with advanced stage patients poorly responding to intensive chemotherapy. To explore effective and safe treatment for newly diagnosed advanced stage NKTCL, we conducted a phase II study of anti-metabolic agent pegaspargase plus PD-1 antibody sintilimab (NCT04096690). Twenty-two patients with a median age of 51 years (range, 24-74) were enrolled and treated with induction treatment of pegaspargase 2500 IU/m2 intramuscularly on day 1 and sintilimab 200 mg intravenously on day 2 for 6 cycles of 21 days, followed by maintenance treatment of sintilimab 200 mg for 28 cycles of 21 days. The complete response and overall response rate after induction treatment were 59% (95%CI, 43-79%) and 68% (95%CI, 47-84%), respectively. With a median follow-up of 30 months, the 2 year progression-free and overall survival rates were 68% (95%CI, 45-83%) and 86% (95%CI, 63-95%), respectively. The most frequently grade 3/4 adverse events were neutropenia (32%, n = 7) and hypofibrinogenemia (18%, n = 4), which were manageable and led to no discontinuation of treatment. Tumor proportion score of PD-L1, peripheral blood high-density lipoprotein cholesterol, and apolipoprotein A-I correlated with good response, while PD-1 on tumor infiltrating lymphocytes and peripheral Treg cells with poor response to pegaspargase plus sintilimab treatment. In conclusion, the chemo-free regimen pegaspargase plus sintilimab was effective and safe in newly diagnosed, advanced stage NKTCL. Dysregulated lipid profile and immunosuppressive signature contributed to treatment resistance, providing an alternative therapeutic approach dual targeting fatty acid metabolism and CTLA-4 in NKTCL.
Collapse
Affiliation(s)
- Jie Xiong
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shu Cheng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao Gao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shan-He Yu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu-Ting Dai
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin-Yun Huang
- Department of Nuclear Medicine, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui-Juan Zhong
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chao-Fu Wang
- Department of Pathology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong-Mei Yi
- Department of Pathology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Zhang
- Department of Otolaryngology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei-Guo Cao
- Department of Radiation, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rong Li
- Department of Hematology, Navy Medical Center of PLA, Shanghai, China
| | - Wei Tang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Zhao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peng-Peng Xu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai, China
| | - Wei-Li Zhao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai, China.
| |
Collapse
|
25
|
Luo W, Zeng Y, Song Q, Wang Y, Yuan F, Li Q, Liu Y, Li S, Jannatun N, Zhang G, Li Y. Strengthening the Combinational Immunotherapy from Modulating the Tumor Inflammatory Environment via Hypoxia-Responsive Nanogels. Adv Healthc Mater 2024; 13:e2302865. [PMID: 38062634 DOI: 10.1002/adhm.202302865] [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: 08/28/2023] [Revised: 11/17/2023] [Indexed: 12/19/2023]
Abstract
Despite the success of immuno-oncology in clinical settings, the therapeutic efficacy is lower than the expectation due to the immunosuppressive inflammatory tumor microenvironment (TME) and the lack of functional lymphocytes caused by exhaustion. To enhance the efficacy of immuno-oncotherapy, a synergistic strategy should be used that can effectively improve the inflammatory TME and increase the tumor infiltration of cytotoxic T lymphocytes (CTLs). Herein, a TME hypoxia-responsive nanogel (NG) is developed to enhance the delivery and penetration of diacerein and (-)-epigallocatechin gallate (EGCG) in tumors. After systemic administration, diacerein effectively improves the tumor immunosuppressive condition through a reduction of MDSCs and Tregs in TME, and induces tumor cell apoptosis via the inhibition of IL-6/STAT3 signal pathway, realizing a strong antitumor effect. Additionally, EGCG can effectively inhibit the expression of PD-L1, restoring the tumor-killing function of CTLs. The infiltration of CTLs increases at the tumor site with activation of systemic immunity after the combination of TIM3 blockade therapy, ultimately resulting in a strong antitumor immune response. This study provides valuable insights for future research on eliciting effective antitumor immunity by suppressing adverse tumor inflammation. The feasible strategy proposed in this work may solve the urgent clinical concerns of the dissatisfactory checkpoint-based immuno-oncotherapy.
Collapse
Affiliation(s)
- Wenhe Luo
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
- Laboratory of Immunology and Nanomedicine & China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Yanqiao Zeng
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
- Laboratory of Immunology and Nanomedicine & China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Qingle Song
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
- Laboratory of Immunology and Nanomedicine & China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Yu Wang
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
- Laboratory of Immunology and Nanomedicine & China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Feng Yuan
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
- Laboratory of Immunology and Nanomedicine & China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Qi Li
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
- Laboratory of Immunology and Nanomedicine & China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Yingnan Liu
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
- Laboratory of Immunology and Nanomedicine & China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Su Li
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
- Laboratory of Immunology and Nanomedicine & China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Nahar Jannatun
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
- Laboratory of Immunology and Nanomedicine & China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Guofang Zhang
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
- Laboratory of Immunology and Nanomedicine & China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Yang Li
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
- Laboratory of Immunology and Nanomedicine & China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| |
Collapse
|
26
|
Bourne CM, Taabazuing CY. Harnessing Pyroptosis for Cancer Immunotherapy. Cells 2024; 13:346. [PMID: 38391959 PMCID: PMC10886719 DOI: 10.3390/cells13040346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 02/24/2024] Open
Abstract
Cancer immunotherapy is a novel pillar of cancer treatment that harnesses the immune system to fight tumors and generally results in robust antitumor immunity. Although immunotherapy has achieved remarkable clinical success for some patients, many patients do not respond, underscoring the need to develop new strategies to promote antitumor immunity. Pyroptosis is an immunostimulatory type of regulated cell death that activates the innate immune system. A hallmark of pyroptosis is the release of intracellular contents such as cytokines, alarmins, and chemokines that can stimulate adaptive immune activation. Recent studies suggest that pyroptosis promotes antitumor immunity. Here, we review the mechanisms by which pyroptosis can be induced and highlight new strategies to induce pyroptosis in cancer cells for antitumor defense. We discuss how pyroptosis modulates the tumor microenvironment to stimulate adaptive immunity and promote antitumor immunity. We also suggest research areas to focus on for continued development of pyroptosis as an anticancer treatment. Pyroptosis-based anticancer therapies offer a promising new avenue for treating immunologically 'cold' tumors.
Collapse
Affiliation(s)
| | - Cornelius Y. Taabazuing
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| |
Collapse
|
27
|
Teske KA, Corona C, Wilkinson J, Mamott D, Good DA, Zambrano D, Lazar DF, Cali JJ, Robers MB, O'Brien MA. Interrogating direct NLRP3 engagement and functional inflammasome inhibition using cellular assays. Cell Chem Biol 2024; 31:349-360.e6. [PMID: 37858335 DOI: 10.1016/j.chembiol.2023.09.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 07/20/2023] [Accepted: 09/25/2023] [Indexed: 10/21/2023]
Abstract
As a key regulator of the innate immune system, the NLRP3 inflammasome responds to a variety of environmental insults through activation of caspase-1 and release of the proinflammatory cytokines IL-1β and IL-18. Aberrant NLRP3 inflammasome function is implicated in numerous inflammatory diseases, spurring drug discovery efforts at NLRP3 as a therapeutic target. A diverse array of small molecules is undergoing preclinical/clinical evaluation with a reported mode of action involving direct modulation of the NLRP3 pathway. However, for a subset of these ligands the functional link between live-cell target engagement and pathway inhibition has yet to be fully established. Herein we present a cohort of mechanistic assays to both query direct NLRP3 engagement in cells, and functionally interrogate different nodes of NLRP3 pathway activity. This system enabled the stratification of potency for five confirmed NLRP3 inhibitors, and identification of two reported NLRP3 inhibitors that failed to demonstrate direct pathway antagonism.
Collapse
Affiliation(s)
- Kelly A Teske
- Promega Corporation, Research & Development, Madison, WI 53711, USA
| | - Cesear Corona
- Promega Corporation, Research & Development, San Luis Obispo, CA 93401, USA
| | | | - Daniel Mamott
- Promega Corporation, Research & Development, Madison, WI 53711, USA
| | - David A Good
- Promega Corporation, Research & Development, San Luis Obispo, CA 93401, USA
| | - Delia Zambrano
- Promega Corporation, Research & Development, San Luis Obispo, CA 93401, USA
| | - Dan F Lazar
- Promega Corporation, Research & Development, Madison, WI 53711, USA
| | - James J Cali
- Promega Corporation, Research & Development, Madison, WI 53711, USA
| | - Matthew B Robers
- Promega Corporation, Research & Development, Madison, WI 53711, USA.
| | - Martha A O'Brien
- Promega Corporation, Research & Development, Madison, WI 53711, USA.
| |
Collapse
|
28
|
Tsankov BK, Luchak A, Carr C, Philpott DJ. The effects of NOD-like receptors on adaptive immune responses. Biomed J 2024; 47:100637. [PMID: 37541620 PMCID: PMC10796267 DOI: 10.1016/j.bj.2023.100637] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 08/06/2023] Open
Abstract
It has long been appreciated that cues from the innate immune system orchestrate downstream adaptive immune responses. Although previous work has focused on the roles of Toll-like receptors in this regard, relatively little is known about how Nod-like receptors instruct adaptive immunity. Here we review the functions of different members of the Nod-like receptor family in orchestrating effector and anamnestic adaptive immune responses. In particular, we address the ways in which inflammasome and non-inflammasome members of this family affect adaptive immunity under various infectious and environmental contexts. Furthermore, we identify several key mechanistic questions that studies in this field have left unaddressed. Our aim is to provide a framework through which immunologists in the adaptive immune field may view their questions through an innate-immune lens and vice-versa.
Collapse
Affiliation(s)
- Boyan K Tsankov
- Department of Immunology, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada
| | - Alexander Luchak
- Department of Immunology, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada
| | - Charles Carr
- Department of Immunology, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada
| | - Dana J Philpott
- Department of Immunology, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada.
| |
Collapse
|
29
|
Chen W, He Y, Zhou G, Chen X, Ye Y, Zhang G, Liu H. Multiomics characterization of pyroptosis in the tumor microenvironment and therapeutic relevance in metastatic melanoma. BMC Med 2024; 22:24. [PMID: 38229080 PMCID: PMC10792919 DOI: 10.1186/s12916-023-03175-0] [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: 03/06/2023] [Accepted: 11/14/2023] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND Pyroptosis, mediated by gasdermins with the release of multiple inflammatory cytokines, has emerged as playing an important role in targeted therapy and immunotherapy due to its effectiveness at inhibiting tumor growth. Melanoma is one of the most commonly used models for immunotherapy development, though an inadequate immune response can occur. Moreover, the development of pyroptosis-related therapy and combinations with other therapeutic strategies is limited due to insufficient understanding of the role of pyroptosis in the context of different tumor immune microenvironments (TMEs). METHODS Here, we present a computational model (pyroptosis-related gene score, PScore) to assess the pyroptosis status. We applied PScore to 1388 melanoma samples in our in-house cohort and eight other publicly available independent cohorts and then calculated its prognostic power of and potential as a predictive marker of immunotherapy efficacy. Furthermore, we performed association analysis for PScore and the characteristics of the TME by using bulk, single-cell, and spatial transcriptomics and assessed the association of PScore with mutation status, which contributes to targeted therapy. RESULTS Pyroptosis-related genes (PRGs) showed distinct expression patterns and prognostic predictive ability in melanoma. Most PRGs were associated with better survival in metastatic melanoma. Our PScore model based on genes associated with prognosis exhibits robust performance in survival prediction in multiple metastatic melanoma cohorts. We also found PScore to be associated with BRAF mutation and correlate positively with multiple molecular signatures, such as KRAS signaling and the IFN gamma response pathway. Based on our data, melanoma with an immune-enriched TME had a higher PScore than melanoma with an immune-depleted or fibrotic TME. Additionally, monocytes had the highest PScore and malignant cells and fibroblasts the lowest PScore based on single-cell and spatial transcriptome analyses. Finally, a higher PScore was associated with better therapeutic efficacy of immune checkpoint blockade, suggesting the potential of pyroptosis to serve as a marker of immunotherapy response. CONCLUSIONS Collectively, our findings indicate that pyroptosis is a prognostic factor and is associated with the immune response in metastatic melanoma, as based on multiomics data. Our results provide a theoretical basis for drug combination and reveal potential immunotherapy response markers.
Collapse
Affiliation(s)
- Wenqiong Chen
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, China
| | - Yi He
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, China
| | - Guowei Zhou
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, China
| | - Xiang Chen
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China.
- Furong Laboratory, Changsha, Hunan, China.
| | - Youqiong Ye
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Guanxiong Zhang
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China.
- Furong Laboratory, Changsha, Hunan, China.
| | - Hong Liu
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China.
- Furong Laboratory, Changsha, Hunan, China.
- Research Center of Molecular Metabolomics, Xiangya Hospital, Central South University, Changsha, China.
- Big Data Institute, Central South University, Changsha, 410083, China.
| |
Collapse
|
30
|
Jiang D, Liu X, Tan R, Zhu Y, Zhang L. Euphorbia factor L2 suppresses the generation of liver metastatic ascites in breast cancer via inhibiting NLRP3 inflammasome activation. Int J Mol Med 2024; 53:8. [PMID: 38063231 PMCID: PMC10712698 DOI: 10.3892/ijmm.2023.5332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 11/14/2023] [Indexed: 12/18/2023] Open
Abstract
Metastasis is the leading cause of death in patients with breast cancer, in part due to the lack of effective treatments. Euphorbia factor L2 (EFL2) is a diterpenoid extracted from Euphorbia lathyris L. seeds, which has attracted increasing attention in recent years due to its anticancer effect. However, the role and molecular mechanism of EFL2 in breast cancer liver metastasis remain unclear. In the present study, a breast cancer liver metastasis model was constructed and the effect of EFL2 on ascites generation in mice was examined. H&E staining detected inflammatory cells and tumor cells in the liver, small intestine and tumor tissues. Western blotting and reverse transcription‑quantitative PCR were used to detect the protein and mRNA expression of NLR family pyrin domain containing‑3 (NLRP3) and related molecules in tumor tissues. Immunohistochemistry was used to detect the levels of CD4 and CD8 T cells in tumor tissue and immunofluorescence was used to further detect the expression level of NLRP3. Finally, the aforementioned experiments were further verified by overexpressing NLPR3. It was found that EFL2 inhibited generation of ascites in the model in a dose‑dependent manner. Furthermore, EFL2 inhibited tumor cell metastasis and enhanced immune cell infiltration. Meanwhile, EFL2 dose‑dependently downregulated the mRNA and protein expression of NLRP3 and related molecules in the model, and overexpression of NLRP3 abolished these beneficial effects of EFL2. Taken together, the present experimental data suggested that EFL2 has a significant inhibitory effect on ascites of breast cancer liver metastasis in vivo, which may inhibit tumor cell metastasis by downregulating NLRP3 expression, providing an experimental basis for treating breast cancer metastasis.
Collapse
Affiliation(s)
- Dongjing Jiang
- Traditional Chinese Medicine and Research Office, Suzhou Vocational Health College, Suzhou, Jiangsu 215000, P.R. China
| | - Xun Liu
- Traditional Chinese Medicine and Research Office, Suzhou Vocational Health College, Suzhou, Jiangsu 215000, P.R. China
| | - Rulan Tan
- Traditional Chinese Medicine and Research Office, Suzhou Vocational Health College, Suzhou, Jiangsu 215000, P.R. China
| | - Ye Zhu
- Traditional Chinese Medicine and Research Office, Suzhou Vocational Health College, Suzhou, Jiangsu 215000, P.R. China
| | - Li Zhang
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| |
Collapse
|
31
|
Kulshrestha S, Goel A. Protein therapeutics as an emerging strategy to deal with skin cancer: A short review. Exp Dermatol 2024; 33:e14981. [PMID: 37983960 DOI: 10.1111/exd.14981] [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: 07/25/2023] [Revised: 10/21/2023] [Accepted: 11/01/2023] [Indexed: 11/22/2023]
Abstract
Cancer has turned into a global menace with an exponential increase in the rate of death every year. Amongst all forms of cancers, skin cancer is the one becoming more common day by day because of the increased exposure to ultraviolet rays, chemicals, pollutants, etc. Skin cancer is of three types namely basal cell, squamous cell and melanoma which is one of the most aggressive forms of cancer with a low survival rate and easy relapse. Melanoma is also notorious for being multi-drug resistant which accounts for its low survival rates in it. Many kinds of therapeutics are been practiced in the contemporary world, but among them, protein therapeutics is been emerging as a promising field with multiple molecular pathway targets that have revolutionized the science of oncology. Proteins acts as small-molecule targets for cancer cells by binding to the cell surface receptors. Proteins including bromodomain and extra-terminal domain (BET) and some toxin proteins are been tried on for dealing with melanoma targeting the major pathways including MAPK, NF-κB and PI3K/AKT. The protein therapeutics also targets the tumour microenvironment including myofibrils, lymphatic vessels etc., thus inducing tumour cell death. In the review, several kinds of proteins and their function toward cell death will be highlighted in the context of skin cancer. In addition to this, the review will look into the inhibition of the function of other inflammatory pathways by inflammasomes and cytokines, both of which have a role in preventing cancer.
Collapse
Affiliation(s)
| | - Anjana Goel
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| |
Collapse
|
32
|
Jiao Z, Zhang J. Interplay between inflammasomes and PD-1/PD-L1 and their implications in cancer immunotherapy. Carcinogenesis 2023; 44:795-808. [PMID: 37796835 DOI: 10.1093/carcin/bgad072] [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: 04/11/2023] [Revised: 08/22/2023] [Accepted: 10/04/2023] [Indexed: 10/07/2023] Open
Abstract
The inflammasomes play crucial roles in inflammation and cancer development, while the PD-1/PD-L1 pathway is critical for immune suppression in the tumor microenvironment (TME). Recent research indicates a reciprocal regulatory relationship between inflammasomes and PD-1/PD-L1 signaling in cancer development and PD-1 blockade treatment. By activating in diverse cells in tumor tissues, inflammasome upregulates PD-L1 level in the TME. Moreover, the regulation of PD-1/PD-L1 activity by inflammasome activation involves natural killer cells, tumor-associated macrophages and myeloid-derived suppressor cells. Conversely, PD-1 blockade can activate the inflammasome, potentially influencing treatment outcomes. The interplay between inflammasomes and PD-1/PD-L1 has profound and intricate effects on cancer development and treatment. In this review, we discuss the crosstalk between inflammasomes and PD-1/PD-L1 in cancers, exploring their implications for tumorigenesis, metastasis and immune checkpoint inhibitor (ICI) resistance. The combined therapeutic strategies targeting both inflammasomes and checkpoint molecules hold promising potential as treatments for cancer.
Collapse
Affiliation(s)
- Zhongyu Jiao
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology (Peking University), Peking University Health Science Center, Beijing 100191, P.R. China
| | - Jun Zhang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology (Peking University), Peking University Health Science Center, Beijing 100191, P.R. China
| |
Collapse
|
33
|
Nasir I, McGuinness C, Poh AR, Ernst M, Darcy PK, Britt KL. Tumor macrophage functional heterogeneity can inform the development of novel cancer therapies. Trends Immunol 2023; 44:971-985. [PMID: 37995659 DOI: 10.1016/j.it.2023.10.007] [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: 10/04/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 11/25/2023]
Abstract
Macrophages represent a key component of the tumor microenvironment (TME) and are largely associated with poor prognosis. Therapeutic targeting of macrophages has historically focused on inhibiting their recruitment or reprogramming their phenotype from a protumor (M2-like) to an antitumor (M1-like) one. Unfortunately, this approach has not provided clinical breakthroughs that have changed practice. Emerging studies utilizing single-cell RNA-sequencing (scRNA-seq) and spatial transcriptomics have improved our understanding of the ontogeny, phenotype, and functional plasticity of macrophages. Overlaying the wealth of current information regarding macrophage molecular subtypes and functions has also identified novel therapeutic vulnerabilities that might drive better control of tumor-associated macrophages (TAMs). Here, we discuss the functional profiling of macrophages and provide an update of novel macrophage-targeted therapies in development.
Collapse
Affiliation(s)
- Ibraheem Nasir
- Breast Cancer Risk and Prevention Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
| | - Conor McGuinness
- Breast Cancer Risk and Prevention Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Melbourne, VIC 3000, Australia
| | - Ashleigh R Poh
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria 3084, Australia; La Trobe University School of Cancer Medicine, Heidelberg, Victoria 3084, Australia
| | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria 3084, Australia; La Trobe University School of Cancer Medicine, Heidelberg, Victoria 3084, Australia
| | - Phillip K Darcy
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Melbourne, VIC 3000, Australia; Cancer Immunology Research Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
| | - Kara L Britt
- Breast Cancer Risk and Prevention Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Melbourne, VIC 3000, Australia.
| |
Collapse
|
34
|
Deng J, Pan T, Liu Z, McCarthy C, Vicencio JM, Cao L, Alfano G, Suwaidan AA, Yin M, Beatson R, Ng T. The role of TXNIP in cancer: a fine balance between redox, metabolic, and immunological tumor control. Br J Cancer 2023; 129:1877-1892. [PMID: 37794178 PMCID: PMC10703902 DOI: 10.1038/s41416-023-02442-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 09/07/2023] [Accepted: 09/14/2023] [Indexed: 10/06/2023] Open
Abstract
Thioredoxin-interacting protein (TXNIP) is commonly considered a master regulator of cellular oxidation, regulating the expression and function of Thioredoxin (Trx). Recent work has identified that TXNIP has a far wider range of additional roles: from regulating glucose and lipid metabolism, to cell cycle arrest and inflammation. Its expression is increased by stressors commonly found in neoplastic cells and the wider tumor microenvironment (TME), and, as such, TXNIP has been extensively studied in cancers. In this review, we evaluate the current literature regarding the regulation and the function of TXNIP, highlighting its emerging role in modulating signaling between different cell types within the TME. We then assess current and future translational opportunities and the associated challenges in this area. An improved understanding of the functions and mechanisms of TXNIP in cancers may enhance its suitability as a therapeutic target.
Collapse
Affiliation(s)
- Jinhai Deng
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
- Clinical Research Center (CRC), Clinical Pathology Center (CPC), Chongqing University Three Gorges Hospital, Chongqing University, Wanzhou, Chongqing, China
| | - Teng Pan
- Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, 518172, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Caitlin McCarthy
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - Jose M Vicencio
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - Lulu Cao
- Department of Rheumatology and Immunology, Peking University People's Hospital and Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Giovanna Alfano
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - Ali Abdulnabi Suwaidan
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - Mingzhu Yin
- Clinical Research Center (CRC), Clinical Pathology Center (CPC), Chongqing University Three Gorges Hospital, Chongqing University, Wanzhou, Chongqing, China
| | - Richard Beatson
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College London, London, UK.
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Division of Medicine, University College London (UCL), Rayne 9 Building, London, WC1E 6JF, UK.
| | - Tony Ng
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College London, London, UK.
- UCL Cancer Institute, University College London, London, UK.
- Cancer Research UK City of London Centre, London, UK.
| |
Collapse
|
35
|
Liu W, Zhang F, Quan B, Yao F, Chen R, Ren Z, Yin X. NLRP3/IL-1β induced myeloid-derived suppressor cells recruitment and PD-L1 upregulation promotes oxaliplatin resistance of hepatocellular carcinoma. MedComm (Beijing) 2023; 4:e447. [PMID: 38116060 PMCID: PMC10728756 DOI: 10.1002/mco2.447] [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: 04/22/2023] [Revised: 11/10/2023] [Accepted: 11/15/2023] [Indexed: 12/21/2023] Open
Abstract
Oxaliplatin is commonly used as the first-line chemotherapeutic agent for advanced hepatocellular carcinoma (HCC). Unfortunately, the acquired resistance, limits the effectiveness of oxaliplatin and the underlying mechanisms remain unknown. Therefore, we explored the role of NOD-like receptor protein 3 (NLRP3)/IL-1β in mediating oxaliplatin resistance in HCC. We observed that NLRP3/IL-1β expression was much higher in oxaliplatin-resistant HCC cells. To further understand its impact on drug resistance, we knocked down NLRP3 and observed that it sensitized HCC cells to the growth-inhibitory effects of oxaliplatin and induced cell apoptosis. NLRP3/IL-1β overexpressing tumor cells also attracted polymorphonuclear myeloid-derived suppressor cells. Using mouse models, we demonstrated that NLRP3/IL-1β inhibition by short hairpin RNA or MCC950 effectively overcame oxaliplatin resistance. Furthermore, NLRP3/IL-1β inhibition resulted in reduced expression of PD-L1. We also found that PD-L1 antibody combined with NLRP3/IL-1β blockade displayed significant antitumor effect in HCC. Overall, our study provides compelling evidence supporting the essential role of NLRP3/IL-1β in conferring resistance to oxaliplatin and reshaping the immunosuppressive microenvironment in HCC. Targeting NLRP3/IL-1β presents a potential therapeutic target for overcoming oxaliplatin resistance and reshaping microenvironment of HCC.
Collapse
Affiliation(s)
- Wenfeng Liu
- Department of Hepatic Oncology, Liver Cancer Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Department of National Clinical Research Center for Interventional MedicineZhongshan hospital, Fudan universityShanghaiChina
| | - Feng Zhang
- Department of Hepatic Oncology, Liver Cancer Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Department of National Clinical Research Center for Interventional MedicineZhongshan hospital, Fudan universityShanghaiChina
| | - Bing Quan
- Department of Hepatic Oncology, Liver Cancer Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Department of National Clinical Research Center for Interventional MedicineZhongshan hospital, Fudan universityShanghaiChina
| | - Fan Yao
- Department of Hepatic Oncology, Liver Cancer Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Department of National Clinical Research Center for Interventional MedicineZhongshan hospital, Fudan universityShanghaiChina
| | - Rongxin Chen
- Department of Hepatic Oncology, Liver Cancer Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Department of National Clinical Research Center for Interventional MedicineZhongshan hospital, Fudan universityShanghaiChina
| | - Zhenggang Ren
- Department of Hepatic Oncology, Liver Cancer Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Department of National Clinical Research Center for Interventional MedicineZhongshan hospital, Fudan universityShanghaiChina
| | - Xin Yin
- Department of Hepatic Oncology, Liver Cancer Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Department of National Clinical Research Center for Interventional MedicineZhongshan hospital, Fudan universityShanghaiChina
| |
Collapse
|
36
|
Dinarello A, May M, Amo-Aparicio J, Azam T, Gaballa JM, Marchetti C, Tesoriere A, Ghirardo R, Redzic JS, Webber WS, Atif SM, Li S, Eisenmesser EZ, de Graaf DM, Dinarello CA. IL-38 regulates intestinal stem cell homeostasis by inducing WNT signaling and beneficial IL-1β secretion. Proc Natl Acad Sci U S A 2023; 120:e2306476120. [PMID: 37906644 PMCID: PMC10636342 DOI: 10.1073/pnas.2306476120] [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: 05/02/2023] [Accepted: 09/13/2023] [Indexed: 11/02/2023] Open
Abstract
The IL-1 Family member IL-38 has been characterized primarily as an antiinflammatory cytokine in human and mouse models of systemic diseases. Here, we examined the role of IL-38 in the murine small intestine (SI). Immunostaining of SI revealed that IL-38 expression partially confines to intestinal stem cells. Cultures of intestinal organoids reveal IL-38 functions as a growth factor by increasing organoid size via inducing WNT3a. In contrast, organoids from IL-38-deficient mice develop more slowly. This reduction in size is likely due to the downregulation of intestinal stemness markers (i.e., Fzd5, Ephb2, and Olfm4) expression compared with wild-type organoids. The IL-38 binding to IL-1R6 and IL-1R9 is still a matter of debate. Therefore, to analyze the molecular mechanisms of IL-38 signaling, we also examined organoids from IL-1R9-deficient mice. Unexpectedly, these organoids, although significantly smaller than wild type, respond to IL-38, suggesting that IL-1R9 is not involved in IL-38 signaling in the stem cell crypt. Nevertheless, silencing of IL-1R6 disabled the organoid response to the growth property of IL-38, thus suggesting IL-1R6 as the main receptor used by IL-38 in the crypt compartment. In organoids from wild-type mice, IL-38 stimulation induced low concentrations of IL-1β which contribute to organoid growth. However, high concentrations of IL-1β have detrimental effects on the cultures that were prevented by treatment with recombinant IL-38. Overall, our data demonstrate an important regulatory function of IL-38 as a growth factor, and as an antiinflammatory molecule in the SI, maintaining homeostasis.
Collapse
Affiliation(s)
- Alberto Dinarello
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| | - Makenna May
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| | - Jesus Amo-Aparicio
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| | - Tania Azam
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| | - Joseph M. Gaballa
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| | - Carlo Marchetti
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| | | | | | - Jasmina S. Redzic
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Denver, Aurora, CO80045
| | - William S. Webber
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| | - Shaikh M. Atif
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| | - Suzhao Li
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| | - Elan Z. Eisenmesser
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Denver, Aurora, CO80045
| | - Dennis M. de Graaf
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| | - Charles A. Dinarello
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| |
Collapse
|
37
|
Mao D, Zhou Z, Chen H, Liu X, Li D, Chen X, He Y, Liu M, Zhang C. Pleckstrin-2 promotes tumour immune escape from NK cells by activating the MT1-MMP-MICA signalling axis in gastric cancer. Cancer Lett 2023; 572:216351. [PMID: 37591356 DOI: 10.1016/j.canlet.2023.216351] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/08/2023] [Accepted: 08/13/2023] [Indexed: 08/19/2023]
Abstract
Immune escape is a major challenge in tumour immunotherapy. Pleckstrin-2(PLEK2) plays a critical role in tumour progression, but its role in immune escape in gastric cancer (GC) remains uncharacterized. RNA sequencing was used to explore the differentially expressed genes in a GC cell line that was resistant to the antitumor effect of Natural killer (NK) cells. Apoptosis and the expression of IFN-γ and TNF-α were detected by flow cytometry (FCM). PLEK2 expression was examined by Western blotting and immunohistochemistry (IHC). PLEK2 was upregulated in MGC803R cells that were resistant to the antitumor effect of NK cells. PLEK2 knockout increased the sensitivity of GC cells to NK cell killing. PLEK2 expression was negatively correlated with MICA and positively correlated with MT1-MMP expression both in vitro and in vivo. PLEK2 promoted Sp1 phosphorylation through the PI3K-AKT pathway, thereby upregulating MT1-MMP expression, which ultimately led to MICA shedding. In mouse xenograft models, PLEK2 knockout inhibited intraperitoneal metastasis of GC cells and promoted NK cell infiltration. In summary, PLEK2 suppressed NK cell immune surveillance by promoting MICA shedding, which serves as a potential therapeutic target for GC.
Collapse
Affiliation(s)
- Deli Mao
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China
| | - Zhijun Zhou
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China; Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, United States
| | - Hengxing Chen
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China
| | - Xinran Liu
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China
| | - Dongsheng Li
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China
| | - Xiancong Chen
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China
| | - Yulong He
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China; Department of Gastrointestinal Surgery of the First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China
| | - Mingyang Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, China.
| | - Changhua Zhang
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China.
| |
Collapse
|
38
|
Amo-Aparicio J, Dominguez A, Atif SM, Dinarello A, Azam T, Alula KM, Piper M, Lieu CH, Lentz RW, Leal AD, Bagby SM, Messersmith WA, Karam SD, Dinarello CA, Pitts TM, Marchetti C. Pancreatic Ductal Adenocarcinoma Cells Regulate NLRP3 Activation to Generate a Tolerogenic Microenvironment. CANCER RESEARCH COMMUNICATIONS 2023; 3:1899-1911. [PMID: 37772994 PMCID: PMC10510589 DOI: 10.1158/2767-9764.crc-23-0065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 08/01/2023] [Accepted: 08/25/2023] [Indexed: 09/30/2023]
Abstract
Defining feature of pancreatic ductal adenocarcinoma (PDAC) that participates in the high mortality rate and drug resistance is the immune-tolerant microenvironment which enables tumors to progress unabated by adaptive immunity. In this study, we report that PDAC cells release CSF-1 to induce nucleotide-binding domain, leucine-rich containing family, pyrin domain-containing-3 (NLRP3) activation in myeloid cells. Increased NLRP3 expression was found in the pancreas of patients with PDAC when compared with normal pancreas which correlated with the formation of the NLRP3 inflammasome. Using human primary cells and an orthotopic PDAC mouse model, we show that NLRP3 activation is responsible for the maturation and release of the inflammatory cytokine IL1β which selectively drives Th2-type inflammation via COX2/PGE2 induction. As a result of this inflammation, primary tumors were characterized by reduced cytotoxic CD8+ T-cell activation and increased tumor expansion. Genetic deletion and pharmacologic inhibition of NLRP3 enabled the development of Th1 immunity, increased intratumoral levels of IL2, CD8+ T cell–mediated tumor suppression, and ultimately limited tumor growth. In addition, we observed that NLRP3 inhibition in combination with gemcitabine significantly increased the efficacy of the chemotherapy. In conclusion, this study provides a mechanism by which tumor-mediated NLRP3 activation exploits a distinct adaptive immunity response that facilitates tumor escape and progression. Considering the ability to block NLRP3 activity with safe and small orally active molecules, this protein represents a new promising target to improve the limited therapeutic options in PDAC. SIGNIFICANT This study provides novel molecular insights on how PDAC cells exploit NLRP3 activation to suppress CD8 T-cell activation. From a translational perspective, we demonstrate that the combination of gemcitabine with the orally active NLRP3 inhibitor OLT1177 increases the efficacy of monotherapy.
Collapse
Affiliation(s)
- Jesus Amo-Aparicio
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Adrian Dominguez
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Shaikh M. Atif
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Alberto Dinarello
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Tania Azam
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Kibrom M. Alula
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Miles Piper
- Department of Radiation Oncology, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado
| | - Christopher H. Lieu
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Robert W. Lentz
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Alexis D. Leal
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Stacey M. Bagby
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Wells A. Messersmith
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Sana D. Karam
- Department of Radiation Oncology, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado
| | - Charles A. Dinarello
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Todd M. Pitts
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Carlo Marchetti
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
39
|
Shadab A, Mahjoor M, Abbasi-Kolli M, Afkhami H, Moeinian P, Safdarian AR. Divergent functions of NLRP3 inflammasomes in cancer: a review. Cell Commun Signal 2023; 21:232. [PMID: 37715239 PMCID: PMC10503066 DOI: 10.1186/s12964-023-01235-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 07/17/2023] [Indexed: 09/17/2023] Open
Abstract
The cancer is a serious health problem, which is The cancer death rate (cancer mortality) is 158.3 per 100,000 men and women per year (based on 2013-2017 deaths). Both clinical and translational studies have demonstrated that chronic inflammation is associated with Cancer progression. However, the precise mechanisms of inflammasome, and the pathways that mediate this phenomenon are not fully characterized. One of the most recently identified signaling pathways, whose activation seems to affect many metabolic disorders, is the "inflammasome" a multiprotein complex composed of NLRP3 (nucleotide-binding domain and leucine-rich repeat protein 3), ASC (apoptosis associated speck-like protein containing a CARD), and procaspase-1. NLRP3 inflammasome activation leads to the processing and secretion of the proinflammatory cytokines interleukin-1β (IL-1β) and IL-18. The goal of this paper is to review new insights on the effects of the NLRP3 inflammasome activation in the complex mechanisms of crosstalk between different organs, for a better understanding of the role of chronic inflammation in cancer pathogenesis. We will provide here a perspective on the current research on NLRP3 inflammasome, which may represent an innovative therapeutic target to reverse the malignancy condition consequences of the inflammation. Video Abstract.
Collapse
Affiliation(s)
- Alireza Shadab
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Iran University of Medical Sciences, Deputy of Health, Tehran, Iran
| | - Mohamad Mahjoor
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Immunology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Abbasi-Kolli
- Iran University of Medical Sciences, Deputy of Health, Tehran, Iran
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Parisa Moeinian
- Department of Medical Genetics and Molecular Biology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amir-Reza Safdarian
- Immunology Board for Transplantation and Cell-Based Therapeutics (Immuno TACT), Universal Scientific Education and Research Network (USERN) Chicago, Chicago, IL, USA.
- Department of Immunology and Microbiology, Faculty of Veterinary Medicine, Tehran University, Tehran, Iran.
| |
Collapse
|
40
|
Chaudhri A, Bu X, Wang Y, Gomez M, Torchia JA, Hua P, Hung SH, Davies MA, Lizee GA, von Andrian U, Hwu P, Freeman GJ. The CX3CL1-CX3CR1 chemokine axis can contribute to tumor immune evasion and blockade with a novel CX3CR1 monoclonal antibody enhances response to anti-PD-1 immunotherapy. Front Immunol 2023; 14:1237715. [PMID: 37771579 PMCID: PMC10524267 DOI: 10.3389/fimmu.2023.1237715] [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: 06/09/2023] [Accepted: 08/28/2023] [Indexed: 09/30/2023] Open
Abstract
CX3CL1 secreted in the tumor microenvironment serves as a chemoattractant playing a critical role in metastasis of CX3CR1 expressing cancer cells. CX3CR1 can be expressed in both cancer and immune-inhibitory myeloid cells to facilitate their migration. We generated a novel monoclonal antibody against mouse CX3CR1 that binds to CX3CR1 and blocks the CX3CL1-CX3CR1 interaction. We next explored the immune evasion strategies implemented by the CX3CL1-CX3CR1 axis and find that it initiates a resistance program in cancer cells that results in 1) facilitation of tumor cell migration, 2) secretion of soluble mediators to generate a pro-metastatic niche, 3) secretion of soluble mediators to attract myeloid populations, and 4) generation of tumor-inflammasome. The CX3CR1 monoclonal antibody reduces migration of tumor cells and decreases secretion of immune suppressive soluble mediators by tumor cells. In combination with anti-PD-1 immunotherapy, this CX3CR1 monoclonal antibody enhances survival in an immunocompetent mouse colon carcinoma model through a decrease in tumor-promoting myeloid populations. Thus, this axis is involved in the mechanisms of resistance to anti-PD-1 immunotherapy and the combination therapy can overcome a portion of the resistance mechanisms to anti-PD-1.
Collapse
Affiliation(s)
- Apoorvi Chaudhri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Department of Melanoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Xia Bu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Yunfei Wang
- Department of Clinical Science, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Michael Gomez
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - James A. Torchia
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Ping Hua
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Shao-Hsi Hung
- Department of Melanoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| | - Michael A. Davies
- Department of Melanoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Gregory A. Lizee
- Department of Melanoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ulrich von Andrian
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Department of Immunology & HMS Center for Immune Imaging, Harvard Medical School, Boston, MA, United States
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States
| | - Patrick Hwu
- Department of Clinical Science, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Gordon J. Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
41
|
Scheben A, Mendivil Ramos O, Kramer M, Goodwin S, Oppenheim S, Becker DJ, Schatz MC, Simmons NB, Siepel A, McCombie WR. Long-Read Sequencing Reveals Rapid Evolution of Immunity- and Cancer-Related Genes in Bats. Genome Biol Evol 2023; 15:evad148. [PMID: 37728212 PMCID: PMC10510315 DOI: 10.1093/gbe/evad148] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2023] [Indexed: 09/21/2023] Open
Abstract
Bats are exceptional among mammals for their powered flight, extended lifespans, and robust immune systems and therefore have been of particular interest in comparative genomics. Using the Oxford Nanopore Technologies long-read platform, we sequenced the genomes of two bat species with key phylogenetic positions, the Jamaican fruit bat (Artibeus jamaicensis) and the Mesoamerican mustached bat (Pteronotus mesoamericanus), and carried out a comprehensive comparative genomic analysis with a diverse collection of bats and other mammals. The high-quality, long-read genome assemblies revealed a contraction of interferon (IFN)-α at the immunity-related type I IFN locus in bats, resulting in a shift in relative IFN-ω and IFN-α copy numbers. Contradicting previous hypotheses of constitutive expression of IFN-α being a feature of the bat immune system, three bat species lost all IFN-α genes. This shift to IFN-ω could contribute to the increased viral tolerance that has made bats a common reservoir for viruses that can be transmitted to humans. Antiviral genes stimulated by type I IFNs also showed evidence of rapid evolution, including a lineage-specific duplication of IFN-induced transmembrane genes and positive selection in IFIT2. In addition, 33 tumor suppressors and 6 DNA-repair genes showed signs of positive selection, perhaps contributing to increased longevity and reduced cancer rates in bats. The robust immune systems of bats rely on both bat-wide and lineage-specific evolution in the immune gene repertoire, suggesting diverse immune strategies. Our study provides new genomic resources for bats and sheds new light on the extraordinary molecular evolution in this critically important group of mammals.
Collapse
Affiliation(s)
- Armin Scheben
- Simons Center for Quantitative Biology, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
| | | | - Melissa Kramer
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
| | - Sara Goodwin
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
| | - Sara Oppenheim
- American Museum of Natural History, Institute for Comparative Genomics, New York, New York, USA
| | - Daniel J Becker
- School of Biological Sciences, University of Oklahoma, Norman, Oklahoma, USA
| | - Michael C Schatz
- Simons Center for Quantitative Biology, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
- Departments of Computer Science and Biology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Nancy B Simmons
- Department of Mammalogy, Division of Vertebrate Zoology, American Museum of Natural History, New York, New York, USA
| | - Adam Siepel
- Simons Center for Quantitative Biology, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
| | | |
Collapse
|
42
|
Wang K, Zhou L, Mao H, Liu J, Chen Z, Zhang L. Intercellular mitochondrial transfer alleviates pyroptosis in dental pulp damage. Cell Prolif 2023; 56:e13442. [PMID: 37086012 PMCID: PMC10472516 DOI: 10.1111/cpr.13442] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 04/23/2023] Open
Abstract
Mitochondrial transfer is emerging as a promising therapeutic strategy for tissue repair, but whether it protects against pulpitis remains unclear. Here, we show that hyperactivated nucleotide-binding domain and leucine-rich repeat protein3 (NLRP3) inflammasomes with pyroptotic cell death was present in pulpitis tissues, especially in the odontoblast layer, and mitochondrial oxidative stress (OS) was involved in driving this NLRP3 inflammasome-induced pathology. Using bone marrow mesenchymal stem cells (BMSCs) as mitochondrial donor cells, we demonstrated that BMSCs could donate their mitochondria to odontoblasts via tunnelling nanotubes (TNTs) and, thus, reduce mitochondrial OS and the consequent NLRP3 inflammasome-induced pyroptosis in odontoblasts. These protective effects of BMSCs were mostly blocked by inhibitors of the mitochondrial function or TNT formation. In terms of the mechanism of action, TNF-α secreted from pyroptotic odontoblasts activates NF-κB signalling in BMSCs via the paracrine pathway, thereby promoting the TNT formation in BMSCs and enhancing mitochondrial transfer efficiency. Inhibitions of NF-κB signalling and TNF-α secretion in BMSCs suppressed their mitochondrial donation capacity and TNT formation. Collectively, these findings demonstrated that TNT-mediated mitochondrial transfer is a potential protective mechanism of BMSCs under stress conditions, suggesting a new therapeutic strategy of mitochondrial transfer for dental pulp repair.
Collapse
Affiliation(s)
- Konghuai Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of StomatologyWuhan UniversityWuhanChina
| | - Lu Zhou
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of StomatologyWuhan UniversityWuhanChina
| | - Hanqing Mao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of StomatologyWuhan UniversityWuhanChina
| | - Jiayi Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of StomatologyWuhan UniversityWuhanChina
| | - Zhi Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of StomatologyWuhan UniversityWuhanChina
- Department of Endodontics, School and Hospital of StomatologyWuhan UniversityWuhanChina
| | - Lu Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of StomatologyWuhan UniversityWuhanChina
- Department of Endodontics, School and Hospital of StomatologyWuhan UniversityWuhanChina
| |
Collapse
|
43
|
Zhou B, Wang Z, Dou Q, Li W, Li Y, Yan Z, Sun P, Zhao B, Li X, Shen F, Zhang B, Guo M. Long-term outcomes of esophageal and gastric cancer patients with cardiovascular and metabolic diseases: A two-center propensity score-matched cohort study. J Transl Int Med 2023; 11:234-245. [PMID: 37818156 PMCID: PMC10561076 DOI: 10.2478/jtim-2023-0112] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2023] Open
Abstract
Background and Objectives An increased risk of cardiovascular and metabolic diseases (CVMDs) among patients with cancer suggests a potential link between CVMD and cancer. The impact of CVMD on the survival time of patients with esophageal and gastric cancer remains unknown. We aimed to determine the incidence of CVMD and its impact on the longterm outcomes in esophageal and gastric cancer patients. Methods A total of 2074 cancer patients were enrolled from January 1, 2007 to December 31, 2017 in two hospitals, including 1205 cases of esophageal cancer and 869 cases of gastric cancer, who were followed up for a median of 79.8 and 79.3 months, respectively. Survival time was analyzed using the Kaplan-Meier method before and after propensity score matching. Results The incidence of CVMD in patients with esophageal and gastric cancer was 34.1% (411/1205) and 34.3% (298/869), respectively. The effects of hypertension, diabetes, and stroke on the long-term survival of esophageal and gastric cancer patients were not significant (all P > 0.05). The survival time was significantly longer in esophageal cancer patients without ischemic heart disease than in patients with ischemic heart disease, both before matching (36.5 vs. 29.1 months, P = 0.027) and after matching (37.4 vs. 27.9 months, P = 0.011). The survival time in gastric cancer patients without ischemic heart disease was significantly longer than in patients with ischemic heart disease, both before (28.4 vs.17.5 months, P = 0.032) and after matching (29.5 vs.17.5 months, P = 0.02). Conclusion The survival time of esophageal and gastric cancer patients with ischemic heart disease was significantly reduced compared to that of esophageal and gastric cancer patients without ischemic heart disease.
Collapse
Affiliation(s)
- Bo Zhou
- Department of Gastroenterology and Hepatology, Chinese PLA General Hospital, Beijing100853, China
| | - Zhixin Wang
- Department of Gastroenterology and Hepatology, the First Medical Center, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang453003, Henan Province, China
| | - Qifeng Dou
- Department of Urology Surgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang453199, Henan Province, China
| | - Wenbin Li
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital affiliated to Capital Medical University, Beijing100029, China
| | - Yangyang Li
- Department of Cardiovascular Surgery, Henan Provincial Chest Hospital, Zhengzhou450008, Henan Province, China
| | - Zhengqiang Yan
- Department of General Surgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang453199, Henan Province, China
| | - Peisheng Sun
- Department of General Surgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang453199, Henan Province, China
| | - Baosheng Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang453199, Henan Province, China
| | - Xiumin Li
- Department of Gastroenterology and Hepatology, the First Medical Center, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang453003, Henan Province, China
- Henan Key Laboratory of Tumor Molecular Therapy Medicine, Xinxiang Medical University, Xinxiang453003, Henan Province, China
| | - Fangfang Shen
- The Key Laboratory for Tumor Translational Medicine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang453003, Henan Province, China
| | - Bangjie Zhang
- Department of Gastroenterology and Hepatology, the First Medical Center, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang453003, Henan Province, China
| | - Mingzhou Guo
- Department of Gastroenterology and Hepatology, Chinese PLA General Hospital, Beijing100853, China
- Department of Gastroenterology and Hepatology, the First Medical Center, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang453003, Henan Province, China
| |
Collapse
|
44
|
Zeng B, Wang X, Qin Y, Cao L, Zhang C, Meng F, Chen C, Wang J, Ma L. Differences in serum cytokine levels distinguish between clinically noninvasive lung adenocarcinoma and invasive lung adenocarcinoma: A cross-sectional study. Health Sci Rep 2023; 6:e1522. [PMID: 37692791 PMCID: PMC10486205 DOI: 10.1002/hsr2.1522] [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: 04/19/2023] [Revised: 08/07/2023] [Accepted: 08/09/2023] [Indexed: 09/12/2023] Open
Abstract
Background Lung cancer incidence and mortality remain high and are now the leading cause of cancer-related death. Lung adenocarcinoma (LUAD) is one of the main histological subtypes of lung cancer. Previous studies have shown the role of inflammation in the development of lung cancer, but the relationship between cytokines and LUAD is still unclear. To further differentiate and explore the association of cytokines with the risk of non-invasive and invasive LUAD, we studied and assessed serum cytokine levels in patients with two types of LUAD. Methods A cohort study of 90 non-invasive LUAD and 90 invasive LUAD was retrospectively included, and the clinical characteristics were recorded in detail. The differences in the levels of 12 serum cytokines (IFN-α, IFN-γ, IL-10, IL-12P70, IL-17A, IL-1β, IL-2, IL-4, IL-5, IL-6, IL-8, and TNF-α) between the two groups of patients with LUAD were analyzed and evaluated. And we evaluated the clinical value of cytokine differential diagnosis of invasive LUAD based on receiver operating characteristics (ROC) curves. Results The mean age of the patients was 56.6 years, and the proportions of males and females were 38.9% and 61.1%, respectively. IFN-α, IL-1β, IL-2, IL-6, TNF-α, IL-4, and IL-8 were significantly increased in patients with invasive LUAD compared with the non-invasive LUAD group. Further research found that smoking is an important factor, with changes in the four cytokines IL-1β, IL-6, IL-8, and TNF-α being significantly higher in the smoking group of patients with invasive LUAD. It can be seen from the area under the curve that IL-1β and IL-2 have a significant differential diagnosis. Conclusions We observed differences in preoperative serum cytokine levels between patients with invasive and non-invasive LUAD, which may serve as potential serum biomarkers for clinical differential diagnosis and disease progression assessment.
Collapse
Affiliation(s)
- Bingjie Zeng
- Department of Laboratory Medicine, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xianzhao Wang
- Department of Laboratory Medicine, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yueyang Qin
- Department of Laboratory Medicine, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Leiqun Cao
- Shanghai Institute of Thoracic Oncology, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Anhui University of Science and Technology School of MedicineHuainanAnhuiChina
| | - Congcong Zhang
- Shanghai Institute of Thoracic Oncology, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Anhui University of Science and Technology School of MedicineHuainanAnhuiChina
| | - Fanyu Meng
- Shanghai Institute of Thoracic Oncology, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Changqiang Chen
- Department of Laboratory Medicine, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jiayi Wang
- Department of Laboratory Medicine, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Institute of Thoracic Oncology, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Lifang Ma
- Department of Laboratory Medicine, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Institute of Thoracic Oncology, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
45
|
Gamage HEV, Shahoei SH, Albright ST, Wang Y, Smith AJ, Farmer R, Fink EC, Jacquin E, Weisser E, Bautista RO, Henn MA, Schane CP, Nelczyk AT, Ma L, Gupta AD, Bendre SV, Nguyen T, Tiwari S, Krawczynska N, He S, Tjoanda E, Chen H, Sverdlov M, Gann PH, Boidot R, Vegran F, Fanning SW, Apetoh L, Hergenrother PJ, Nelson ER. Re-education of myeloid immune cells to reduce regulatory T cell expansion and impede breast cancer progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.14.553229. [PMID: 37645737 PMCID: PMC10462080 DOI: 10.1101/2023.08.14.553229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Immune checkpoint blockade (ICB) has revolutionized cancer therapy but has had limited utility in several solid tumors such as breast cancer, a major cause of cancer-related mortality in women. Therefore, there is considerable interest in alternate strategies to promote an anti-cancer immune response. We demonstrate that NR0B2, a protein involved in cholesterol homeostasis, functions within myeloid immune cells to modulate the NLRP3 inflammasome and reduce the expansion of immune-suppressive regulatory T cells (Treg). Loss of NR0B2 increased mammary tumor growth and metastasis. Small molecule agonists, including one developed here, reduced Treg expansion, reduced metastatic growth and improved the efficacy of ICB. This work identifies NR0B2 as a target to re-educate myeloid immune cells providing proof-of-principle that this cholesterol-homeostasis axis may have utility in enhancing ICB.
Collapse
Affiliation(s)
- Hashni Epa Vidana Gamage
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Sayyed Hamed Shahoei
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Samuel T. Albright
- Department of Chemistry, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Yu Wang
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Amanda J. Smith
- Department of Chemistry, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Rachel Farmer
- Department of Chemistry, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Emma C. Fink
- Department of Cancer Biology, Loyola University Chicago Health Sciences Campus, Illinois, USA
| | | | - Erin Weisser
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Rafael O. Bautista
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Madeline A. Henn
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Claire P. Schane
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Adam T. Nelczyk
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Liqian Ma
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Anasuya Das Gupta
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Shruti V. Bendre
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Tiffany Nguyen
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Srishti Tiwari
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Natalia Krawczynska
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Sisi He
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Evelyn Tjoanda
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Hong Chen
- Food Science & Human Nutrition, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Maria Sverdlov
- Research Histology and Tissue Imaging Core, University of Illinois at Chicago, Illinois, USA
| | - Peter H. Gann
- Research Histology and Tissue Imaging Core, University of Illinois at Chicago, Illinois, USA
- Department of Pathology, University of Illinois at Chicago, Illinois, USA
| | - Romain Boidot
- Unit of Molecular Biology, Department of Biology and Pathology of Tumors, Georges-Francois Leclerc cancer Center, Dijon, France, and ICMUB UMR CNRS 6302, Dijon, France
| | | | - Sean W. Fanning
- Department of Cancer Biology, Loyola University Chicago Health Sciences Campus, Illinois, USA
| | | | - Paul J. Hergenrother
- Department of Chemistry, University of Illinois at Urbana-Champaign, Illinois, USA
- Carl R. Woese Institute for Genomic Biology- Anticancer Discovery from Pets to People, University of Illinois at Urbana-Champaign, Illinois, USA
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Erik R. Nelson
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Illinois, USA
- Carl R. Woese Institute for Genomic Biology- Anticancer Discovery from Pets to People, University of Illinois at Urbana-Champaign, Illinois, USA
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, University of Illinois at Urbana-Champaign, Illinois, USA
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, University of Illinois at Urbana-Champaign, Illinois, USA
| |
Collapse
|
46
|
Kim SM, Jeon Y, Jang JY, Lee H. NR1D1 deficiency in the tumor microenvironment promotes lung tumor development by activating the NLRP3 inflammasome. Cell Death Discov 2023; 9:278. [PMID: 37524704 PMCID: PMC10390518 DOI: 10.1038/s41420-023-01554-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/09/2023] [Accepted: 07/12/2023] [Indexed: 08/02/2023] Open
Abstract
Nuclear receptor Rev-erbα (NR1D1) is a major negative regulator of the circadian clock. Numerous studies have investigated the role of circadian clock-related factors in the tumorigenesis of multiple cancer types, but little is known about the role of NR1D1 in cancer development. In this study, we identified the role of NR1D1 in lung tumorigenesis using genetically engineered mouse models of Nr1d1. Although NR1D1 overexpression or knockdown had little effect on the proliferation of NSCLC cells in vitro, NR1D1 deficiency in the tumor microenvironment increased lung cancer development compared with the control in the orthotopic model. NR1D1-deficient mice showed increased NACHT, LRR, and PYD domain-containing protein 3 (NLRP3) inflammasome activation, and conditioned medium (CM) from NR1D1-deficient macrophages increased the proliferation and epithelial-mesenchymal transition (EMT) of lung cancer cells. Treatment with MCC950, a specific inhibitor of NLRP3 inflammasome, blocked tumorigenesis in NR1D1-deficient mice in an orthotopic lung cancer model. In addition, MCC950 treatment blocked the increased proliferation and EMT of cancer cells induced by CM from NR1D1-deficient macrophages in vitro. Our results showed that NR1D1 in the tumor microenvironment functions as a tumor suppressor by negatively regulating the NLRP3 inflammasome, suggesting that the NLRP3 inflammasome blockade via NR1D1 activation could be a therapeutic strategy to overcome lung cancer.
Collapse
Affiliation(s)
- Sun Mi Kim
- Graduate School of Cancer Science and Policy, National Cancer Center, Gyeonggi, 10408, Republic of Korea.
- Research Institute, National Cancer Center, Gyeonggi, 10408, Republic of Korea.
| | - Yoon Jeon
- Research Institute, National Cancer Center, Gyeonggi, 10408, Republic of Korea
| | - Ji Yun Jang
- Research Institute, National Cancer Center, Gyeonggi, 10408, Republic of Korea
- College of Pharmacy, Dongguk University, Seoul, 04620, Republic of Korea
| | - Ho Lee
- Graduate School of Cancer Science and Policy, National Cancer Center, Gyeonggi, 10408, Republic of Korea.
- Research Institute, National Cancer Center, Gyeonggi, 10408, Republic of Korea.
| |
Collapse
|
47
|
Ozbay Kurt FG, Lasser S, Arkhypov I, Utikal J, Umansky V. Enhancing immunotherapy response in melanoma: myeloid-derived suppressor cells as a therapeutic target. J Clin Invest 2023; 133:e170762. [PMID: 37395271 DOI: 10.1172/jci170762] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2023] Open
Abstract
Despite the remarkable success of immune checkpoint inhibitors (ICIs) in melanoma treatment, resistance to them remains a substantial clinical challenge. Myeloid-derived suppressor cells (MDSCs) represent a heterogeneous population of myeloid cells that can suppress antitumor immune responses mediated by T and natural killer cells and promote tumor growth. They are major contributors to ICI resistance and play a crucial role in creating an immunosuppressive tumor microenvironment. Therefore, targeting MDSCs is considered a promising strategy to improve the therapeutic efficacy of ICIs. This Review describes the mechanism of MDSC-mediated immune suppression, preclinical and clinical studies on MDSC targeting, and potential strategies for inhibiting MDSC functions to improve melanoma immunotherapy.
Collapse
Affiliation(s)
- Feyza Gul Ozbay Kurt
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | - Samantha Lasser
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | - Ihor Arkhypov
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Viktor Umansky
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| |
Collapse
|
48
|
Chen Y, Ye X, Escames G, Lei W, Zhang X, Li M, Jing T, Yao Y, Qiu Z, Wang Z, Acuña-Castroviejo D, Yang Y. The NLRP3 inflammasome: contributions to inflammation-related diseases. Cell Mol Biol Lett 2023; 28:51. [PMID: 37370025 DOI: 10.1186/s11658-023-00462-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
The NOD-like receptor protein 3 (NLRP3) inflammasome is a protein complex that regulates innate immune responses by activating caspase-1 and the inflammatory cytokines interleukin (IL)-1β and IL-18. Multiple studies have demonstrated the importance of the NLRP3 inflammasome in the development of immune and inflammation-related diseases, including arthritis, Alzheimer's disease, inflammatory bowel disease, and other autoimmune and autoinflammatory diseases. This review first explains the activation and regulatory mechanism of the NLRP3 inflammasome. Secondly, we focus on the role of the NLRP3 inflammasome in various inflammation-related diseases. Finally, we look forward to new methods for targeting the NLRP3 inflammasome to treat inflammation-related diseases, and provide new ideas for clinical treatment.
Collapse
Affiliation(s)
- Ying Chen
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xingyan Ye
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
- Department of Neurology, Xi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, China
| | - Germaine Escames
- Biomedical Research Center, Health Sciences Technology Park, University of Granada, Avda. del Conocimiento s/n, Granada, Spain
- Ibs. Granada and CIBERfes, Granada, Spain
- UGC of Clinical Laboratories, University San Cecilio's Hospital, Granada, Spain
| | - Wangrui Lei
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
- Department of Neurology, Xi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, China
| | - Xin Zhang
- Department of Cardiology, Affiliated Hospital, Yan'an University, Yan'an, China
| | - Meng Li
- Department of Cardiology, Affiliated Hospital, Yan'an University, Yan'an, China
| | - Tong Jing
- Department of Cardiology, Affiliated Hospital, Yan'an University, Yan'an, China
| | - Yu Yao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
- Department of Neurology, Xi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, China
| | - Zhenye Qiu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
- Department of Neurology, Xi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, China
| | - Zheng Wang
- Department of Cardiothoracic Surgery, Central Theater Command General Hospital of Chinese People's Liberation Army, Wuhan, China
| | - Darío Acuña-Castroviejo
- Biomedical Research Center, Health Sciences Technology Park, University of Granada, Avda. del Conocimiento s/n, Granada, Spain.
- Ibs. Granada and CIBERfes, Granada, Spain.
- UGC of Clinical Laboratories, University San Cecilio's Hospital, Granada, Spain.
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China.
- Department of Neurology, Xi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, China.
| |
Collapse
|
49
|
Amo-Aparicio J, Daly J, Højen JF, Dinarello CA. Pharmacologic inhibition of NLRP3 reduces the levels of α-synuclein and protects dopaminergic neurons in a model of Parkinson's disease. J Neuroinflammation 2023; 20:147. [PMID: 37349821 PMCID: PMC10286423 DOI: 10.1186/s12974-023-02830-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 06/11/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND Parkinson's disease (PD) is characterized by a progressive degeneration of dopaminergic neurons, which leads to irreversible loss of peripheral motor functions. Death of dopaminergic neurons induces an inflammatory response in microglial cells, which further exacerbates neuronal loss. Reducing inflammation is expected to ameliorate neuronal loss and arrest motor dysfunctions. Because of the contribution of the NLRP3 inflammasome to the inflammatory response in PD, we targeted NLRP3 using the specific inhibitor OLT1177®. METHODS We evaluated the effectiveness of OLT1177® in reducing the inflammatory response in an MPTP neurotoxic model of PD. Using a combination of in vitro and in vivo studies, we analyzed the effects of NLRP3 inhibition on pro-inflammatory markers in the brain, α-synuclein aggregation, and dopaminergic neuron survival. We also determined the effects of OLT1177® on locomotor deficits associated with MPTP and brain penetrance. RESULTS Treatment with OLT1177® prevented the loss of motor function, reduced the levels of α-synuclein, modulated pro-inflammatory markers in the nigrostriatal areas of the brain, and protected dopaminergic neurons from degeneration in the MPTP model of PD. We also demonstrated that OLT1177® crosses the blood-brain barrier and reaches therapeutic concentrations in the brain. CONCLUSIONS These data suggest that targeting the NLRP3 inflammasome by OLT1177® may be a safe and novel therapeutic approach to arrest neuroinflammation and protect against neurological deficits of Parkinson's disease in humans.
Collapse
Affiliation(s)
- Jesus Amo-Aparicio
- Department of Medicine, University of Colorado, 12700 E 19th Ave, Aurora, CO, 80045, USA.
| | - Jonathan Daly
- Department of Medicine, University of Colorado, 12700 E 19th Ave, Aurora, CO, 80045, USA
| | - Jesper Falkesgaard Højen
- Department of Medicine, University of Colorado, 12700 E 19th Ave, Aurora, CO, 80045, USA
- Department of Clinical Medicine, Aarhus University, 8200, Aarhus, Denmark
| | - Charles A Dinarello
- Department of Medicine, University of Colorado, 12700 E 19th Ave, Aurora, CO, 80045, USA
| |
Collapse
|
50
|
Kharouf N, Flanagan TW, Hassan SY, Shalaby H, Khabaz M, Hassan SL, Megahed M, Haikel Y, Santourlidis S, Hassan M. Tumor Microenvironment as a Therapeutic Target in Melanoma Treatment. Cancers (Basel) 2023; 15:3147. [PMID: 37370757 DOI: 10.3390/cancers15123147] [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: 05/07/2023] [Revised: 06/02/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
The role of the tumor microenvironment in tumor growth and therapy has recently attracted more attention in research and drug development. The ability of the microenvironment to trigger tumor maintenance, progression, and resistance is the main cause for treatment failure and tumor relapse. Accumulated evidence indicates that the maintenance and progression of tumor cells is determined by components of the microenvironment, which include stromal cells (endothelial cells, fibroblasts, mesenchymal stem cells, and immune cells), extracellular matrix (ECM), and soluble molecules (chemokines, cytokines, growth factors, and extracellular vesicles). As a solid tumor, melanoma is not only a tumor mass of monolithic tumor cells, but it also contains supporting stroma, ECM, and soluble molecules. Melanoma cells are continuously in interaction with the components of the microenvironment. In the present review, we focus on the role of the tumor microenvironment components in the modulation of tumor progression and treatment resistance as well as the impact of the tumor microenvironment as a therapeutic target in melanoma.
Collapse
Affiliation(s)
- Naji Kharouf
- Biomaterials and Bioengineering, Institut National de la Santé et de la Recherche Médicale, Université de Strasbourg, Unité Mixte de Recherche 1121, 67000 Strasbourg, France
- Department of Endodontics and Conservative Dentistry, Faculty of Dental Medicine, University of Strasbourg, 67000 Strasbourg, France
| | - Thomas W Flanagan
- Department of Pharmacology and Experimental Therapeutics, LSU Health Sciences Center, New Orleans, LA 70112, USA
| | - Sofie-Yasmin Hassan
- Department of Chemistry, Faculty of Science, Heinrich-Heine University Duesseldorf, 40225 Dusseldorf, Germany
| | - Hosam Shalaby
- Department of Urology, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Marla Khabaz
- Department of Production, Beta Factory for Veterinary Pharmaceutical Industries, Damascus 0100, Syria
| | - Sarah-Lilly Hassan
- Department of Chemistry, Faculty of Science, Heinrich-Heine University Duesseldorf, 40225 Dusseldorf, Germany
| | - Mosaad Megahed
- Clinic of Dermatology, University Hospital of Aachen, 52074 Aachen, Germany
| | - Youssef Haikel
- Biomaterials and Bioengineering, Institut National de la Santé et de la Recherche Médicale, Université de Strasbourg, Unité Mixte de Recherche 1121, 67000 Strasbourg, France
- Department of Endodontics and Conservative Dentistry, Faculty of Dental Medicine, University of Strasbourg, 67000 Strasbourg, France
- Pôle de Médecine et Chirurgie Bucco-Dentaire, Hôpital Civil, Hôpitaux Universitaire de Strasbourg, 67000 Strasbourg, France
| | - Simeon Santourlidis
- Epigenetics Core Laboratory, Institute of Transplantation Diagnostics and Cell Therapeutics, Medical Faculty, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Mohamed Hassan
- Biomaterials and Bioengineering, Institut National de la Santé et de la Recherche Médicale, Université de Strasbourg, Unité Mixte de Recherche 1121, 67000 Strasbourg, France
- Department of Endodontics and Conservative Dentistry, Faculty of Dental Medicine, University of Strasbourg, 67000 Strasbourg, France
- Research Laboratory of Surgery-Oncology, Department of Surgery, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| |
Collapse
|