1
|
Qu Y, Liang W, Yu M, Wang C, Luo M, Zhong L, Li Z, Wang F. MYO1F in neutrophils is required for the response to immune checkpoint blockade therapy. J Exp Med 2025; 222:e20241957. [PMID: 40202509 PMCID: PMC11980683 DOI: 10.1084/jem.20241957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/29/2025] [Accepted: 02/21/2025] [Indexed: 04/10/2025] Open
Abstract
Tumor-associated neutrophils (TANs) represent a significant barrier to the effectiveness of immune checkpoint blockade (ICB) therapy. A comprehensive understanding of TANs' regulatory mechanisms is therefore essential for predicting ICB efficacy and improving immunotherapy strategies. Our study reveals that MYO1F is selectively downregulated in neutrophils within both human cancers and murine tumor models, showing a negative correlation with ICB response. Mechanistically, MYO1F normally inhibits neutrophil immunosuppression and proliferation by restraining STAT3 activity. However, during tumorigenesis, tumor-derived TGF-β1 disrupts the binding of SPI1 to intron 8 of Myo1f via DNA methylation, thereby suppressing Myo1f transcription. The resultant decrease in MYO1F reprograms neutrophils into an immunosuppressive state through the STAT3-dependent signaling pathways. This immunosuppressive state further contributes to tumor microenvironment (TME) remodeling by inducing CTL exhaustion. These findings establish MYO1F as a critical regulator within TANs, highlighting its significant role in modulating ICB therapy efficacy.
Collapse
Affiliation(s)
- Yingying Qu
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Center for Microbiota and Immunological Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenhua Liang
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingzhu Yu
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenhui Wang
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Min Luo
- Institute of Pediatrics of Children’s Hospital of Fudan University, The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Lin Zhong
- Department of Liver Surgery and Organ Transplantation Center, Shenzhen Third People’s Hospital, Second Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhigang Li
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Wang
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
2
|
Dong D, Yu X, Liu H, Xu J, Guo J, Guo W, Li X, Wang F, Zhang D, Liu K, Sun Y. Study of immunosenescence in the occurrence and immunotherapy of gastrointestinal malignancies. Semin Cancer Biol 2025; 111:16-35. [PMID: 39929408 DOI: 10.1016/j.semcancer.2025.01.007] [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: 12/15/2024] [Revised: 01/18/2025] [Accepted: 01/26/2025] [Indexed: 02/25/2025]
Abstract
In human beings heterogenous, pervasive and lethal malignancies of different parts of the gastrointestinal (GI) tract viz., tumours of the oesophagus, stomach, small intestine, colon, and rectum, represent gastrointestinal malignancies. Primary treatment modality for gastric cancer includes chemotherapy, surgical interventions, radiotherapy, monoclonal antibodies and inhibitors of angiogenesis. However, there is a need to improve upon the existing treatment modality due to associated adverse events and the development of resistance towards treatment. Additionally, age has been found to contribute to increasing the incidence of tumours due to immunosenescence-associated immunosuppression. Immunosenescence is the natural process of ageing, wherein immune cells as well as organs begin to deteriorate resulting in a dysfunctional or malfunctioning immune system. Accretion of senescent cells in immunosenescence results in the creation of a persistent inflammatory environment or inflammaging, marked with elevated expression of pro-inflammatory and immunosuppressive cytokines and chemokines. Perturbation in the T-cell pools and persistent stimulation by the antigens facilitate premature senility of the immune cells, and senile immune cells exacerbate inflammaging conditions and the inefficiency of the immune system to identify the tumour antigen. Collectively, these conditions contribute positively towards tumour generation, growth and eventually proliferation. Thus, activating the immune cells to distinguish the tumour cells from normal cells and invade them seems to be a logical strategy for the treatment of cancer. Consequently, various approaches to immunotherapy, viz., programmed death ligand-1 (PD-1) inhibitors, Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitors etc are being extensively evaluated for their efficiency in gastric cancer. In fact, PD-1 inhibitors have been sanctioned as late late-line therapy modality for gastric cancer. The present review will focus on deciphering the link between the immune system and gastric cancer, and the alterations in the immune system that incur during the development of gastrointestinal malignancies. Also, the mechanism of evasion by tumour cells and immune checkpoints involved along with different approaches of immunotherapy being evaluated in different clinical trials will be discussed.
Collapse
Affiliation(s)
- Daosong Dong
- Department of Pain, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Xue Yu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Molecular Pathology and Epidemiology of Gastric Cancer in the Universities of Liaoning Province, Shenyang, Liaoning 110001, China
| | - Haoran Liu
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Jingjing Xu
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Jiayan Guo
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Wei Guo
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Xiang Li
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Fei Wang
- Department of Otolaryngology, The First Hospital of China Medical University, Shenyang 110001, China.
| | - Dongyong Zhang
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang 110001, China.
| | - Kaiwei Liu
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Yanbin Sun
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang 110001, China.
| |
Collapse
|
3
|
Fang B, Lu Y, Li X, Wei Y, Ye D, Wei G, Zhu Y. Targeting the tumor microenvironment, a new therapeutic approach for prostate cancer. Prostate Cancer Prostatic Dis 2025; 28:260-269. [PMID: 38565910 DOI: 10.1038/s41391-024-00825-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/17/2024] [Accepted: 03/21/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND A growing number of studies have shown that in addition to adaptive immune cells such as CD8 + T cells and CD4 + T cells, various other cellular components within prostate cancer (PCa) tumor microenvironment (TME), mainly tumor-associated macrophages (TAMs), cancer-associated fibroblasts (CAFs) and myeloid-derived suppressor cells (MDSCs), have been increasingly recognized as important modulators of tumor progression and promising therapeutic targets. OBJECTIVE In this review, we aim to delineate the mechanisms by which TAMs, CAFs and MDSCs interact with PCa cells in the TME, summarize the therapeutic advancements targeting these cells and discuss potential new therapeutic avenues. METHODS We searched PubMed for relevant studies published through December 10 2023 on TAMs, CAFs and MDSCs in PCa. RESULTS TAMs, CAFs and MDSCs play a critical role in the tumorigenesis, progression, and metastasis of PCa. Moreover, they substantially mediate therapeutic resistance against conventional treatments including anti-androgen therapy, chemotherapy, and immunotherapy. Therapeutic interventions targeting these cellular components have demonstrated promising effects in preclinical models and several clinical trials for PCa, when administrated alone, or combined with other anti-cancer therapies. However, the lack of reliable biomarkers for patient selection and incomplete understanding of the mechanisms underlying the interactions between these cellular components and PCa cells hinder their clinical translation and utility. CONCLUSION New therapeutic strategies targeting TAMs, CAFs, and MDSCs in PCa hold promising prospects. Future research endeavors should focus on a more comprehensive exploration of the specific mechanisms by which these cells contribute to PCa, aiming to identify additional drug targets and conduct more clinical trials to validate the safety and efficacy of these treatment strategies.
Collapse
Affiliation(s)
- Bangwei Fang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Ying Lu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xiaomeng Li
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Yu Wei
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Gonghong Wei
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Yao Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China.
| |
Collapse
|
4
|
Jiang X, Wu A, Yan J, Chen J, Wen Y, Wu H, Yan N, Yang Z, Liu F, Li P. Eleutheroside A inhibits PI3K/AKT1/mTOR-mediated glycolysis in MDSCs to alleviate their immunosuppressive function in gastric cancer. Int Immunopharmacol 2025; 159:114907. [PMID: 40409102 DOI: 10.1016/j.intimp.2025.114907] [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: 04/10/2025] [Revised: 05/10/2025] [Accepted: 05/16/2025] [Indexed: 05/25/2025]
Abstract
BACKGROUND An immune-suppressive tumor microenvironment (TME) that encourages tumor growth is a hallmark of gastric cancer (GC), which is implicated in the development, metastasis, and unfavorable prognosis of GC. Acanthopanax senticosus (Rupr.&Maxim.) Harms (AS), also called Siberian Ginseng (Chinese: Ci wu jia), is a commonly used traditional Chinese herbal medicine with immune-enhancing, anti-tumor, anti-fatigue, neuroregulatory, blood circulation-improving, and antioxidant properties. Recently, it has also been demonstrated to improve anti-tumor immunity in GC. Eleutheroside A (EA), one of the primary bioactive saponins of AS, has immunoregulatory functions. Given the immunomodulatory and anti-tumor effects of EA, it is crucial to investigate its regulatory impact on the immune landscape of GC. MATERIALS AND METHODS To determine the effects of EA on immune responses in GC, a subcutaneous GC mouse model was established. Tumor growth, body weight changes, and immune responses in the mice treated with EA were measured. The proportion of CD4+T, CD8+T, B cells, NK cells, TAMs, DCs and MDSCs in the spleens were analyzed using flow cytometry. MDSCs and CD4+/CD8+ T cell infiltration in tumor tissue were analyzed using immunofluorescence. Bulk RNA sequencing (bulk RNA-seq) data from the Cancer Genome Atlas (TCGA) and two single-cell RNA sequencing (scRNA-seq) datasets (accession numbers GSE183904 and GSE150290) were used to examine changes in MDSCs and T cell infiltration within the TME of GC and to identify MDSCs-related targets. Network pharmacology analysis, protein-protein interaction (PPI) network analysis, dynamics simulations, molecular docking and surface plasmon resonance (SPR) were applied to explore the potential mechanisms underlying EA's intervention in MDSCs. Flow cytometry, qPCR, and western blotting and Seahorse assays were applied for analyzing MDSCs isolated from in vivo and in vitro-induced conditions, aiming to delineate the mechanism of EA on MDSCs glycolysis and immunosuppressive functions mediated by the PI3K/AKT1/mTOR signaling pathway. RESULTS In vivo, EA treatment effectively suppressed GC tumor growth and progression in mice, reducing the prevalence of MDSCs and increasing CD4+/CD8+ T cell levels. In vitro, EA not only decreased the frequency of MDSCs but also alleviated their immune-suppressing capabilities on CD4+/CD8+ T cells. Network pharmacology, coupled with scRNA-seq analysis, dynamic simulations, and molecular docking studies, suggested that EA might modulate the PI3K/AKT1/mTOR signaling pathway to influence glycolysis in MDSCs. Surface plasmon resonance (SPR) analysis confirmed that EA directly interacts with AKT1. Further validation experiments revealed that in the GC TME, EA treatment decreased the expression of p-PI3K, p-AKT1, p-mTOR, HIF1α, as well as glycolytic genes and glycolytic activity in MDSCs. Additionally, EA led to the downregulation of p-STAT3 and its downstream immunosuppressive factors within these cells. Restoring AKT1 activation could reverse the inhibitory effects of EA on MDSCs glycolysis and the downregulation of immunosuppressive molecules. Moreover, HIF-1α inhibition abolished EA's inhibitory effects on MDSCs. CONCLUSION EA can attenuate the immune-suppressive capacity of MDSCs in GC by inhibiting the PI3K/AKT1/mTOR pathway and suppressing HIF-1α-mediated glycolysis, thereby offering a novel therapeutic approach to targeting the immune-suppressive microenvironment in GC.
Collapse
Affiliation(s)
- Xiaotao Jiang
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China
| | - Anzhou Wu
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China; The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China
| | - Jiaxing Yan
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China; The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China
| | - Jingming Chen
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China; The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China
| | - Yi Wen
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China
| | - Hui Wu
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China; The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China
| | - Ning Yan
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China
| | - Zehong Yang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China
| | - Fengbin Liu
- Baiyun Hospital of The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510470, Guangdong, China; Lingnan Institute of Spleen and Stomach Diseases, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China.
| | - Peiwu Li
- Department of hepatobiliary diseases, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou 510405, Guangdong, China.
| |
Collapse
|
5
|
Teng G, Zhang M, Pan Y, Karampoor S, Mirzaei R. Modulating the tumor microenvironment: The role of traditional Chinese medicine in improving lung cancer treatment. Open Life Sci 2025; 20:20251100. [PMID: 40417000 PMCID: PMC12103189 DOI: 10.1515/biol-2025-1100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 02/02/2025] [Accepted: 03/17/2025] [Indexed: 05/27/2025] Open
Abstract
The holistic approach of traditional Chinese medicine (TCM) has been increasingly being focused on as a potential adjuvant to conventional lung cancer therapies in an attempt at modulating the tumor microenvironment (TME). Covering a diverse range of herbal medicine, acupuncture, and dietary therapy, TCM brings a unique perspective to influencing the TME. Importantly, the study has found the effects of specific TCM compounds, such as cantharidin, boehmenan, shikonin, and salidroside, on lung cancer in the TME. These compounds interact intricately with key apoptotic regulators, oxidative stress pathways, and inflammation-related mechanisms, suggesting their potential role in enhancing conventional therapies. TCM compounds could modulate a variety of cellular and molecular pathways, potentially inhibiting tumor proliferation, invasion, and metastasis. Besides, the practices of TCM alleviate the side effects of conventional treatments and enhance immune function, hence promoting the quality of life among lung cancer patients. In this regard, this review gives a contemporary account of the state of affairs on the part of TCM within the framework of the treatment of lung cancer with reference to its recent developments, and diverse roles.
Collapse
Affiliation(s)
- Geling Teng
- Department of Respiratory and Critical Care Medicine, Shandong Public Health Clinical Center, Shandong University, Jinan, 250013, China
| | - Min Zhang
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Yuling Pan
- School of Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
6
|
Liu Y, Ren Y, Luo R, Li X, Xie L, Kang H, Li Y, Dong X, He Y. FK506 prolongs corneal allograft survival and prevents dendritic cell infiltration in an MDSC-dependent manner. Am J Transplant 2025:S1600-6135(25)00272-2. [PMID: 40398562 DOI: 10.1016/j.ajt.2025.05.018] [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: 11/24/2024] [Revised: 05/08/2025] [Accepted: 05/11/2025] [Indexed: 05/23/2025]
Abstract
FK506 (a.k.a. Tacrolimus) is one of most widely used immunosuppressive drugs in the postsurgery management of transplantation. To date, the cellular mechanism by which FK506 suppresses immune activation and elongates allograft survival remains largely unclear. Here, we employed a mouse model for corneal penetrating keratoplasty to interrogate this critical question. Administration of FK506 led to increased expansion myeloid-derived suppressor cells (MDSC) in recipient mice and prolonged survival of corneal allografts. In contrast, antibody-mediated depletion of MDSC abolished the FK506-mediated beneficial effects, which is associated with increased dendritic cell (DC) activation and recruitment to the graft bed and allografts. Of note, unlike continuous depletion and temporary early depletion (in the first week), delayed depletion of MDSC that started on day 8 posttransplant failed to disrupt the FK506-induced elongation of corneal allograft survival. Single-cell RNA sequencing analysis and immunofluorescence staining of corneal grafts reveal that FK506 reduced graft infiltration of immune cells including DC and T cells in an MDSC-dependent and temporal fashion. Moreover, depletion of MDSC reverted the FK506's suppression of DC maturation in the draining lymph node on day 7. Taken together, these findings indicate that FK506 prolongs allograft survival through induction of MDSC-mediated suppression of early DC activation.
Collapse
Affiliation(s)
- Yingyi Liu
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Key Laboratory of Ophthalmology & Visual Sciences, Beijing 100730, China
| | - Yuerong Ren
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan 410011, China
| | - Runxi Luo
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Xiujuan Li
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Limin Xie
- National Clinical Research Center for Metabolic Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Huanmin Kang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yang Li
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Key Laboratory of Ophthalmology & Visual Sciences, Beijing 100730, China
| | - Xiaonan Dong
- State Key Laboratory of Respiratory Disease, Department of Organ transplantation, Advanced Interdisciplinary Studies Center, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, China; Institute of Infection and Health, Fudan University, Shanghai 200438, China; Guangzhou National Laboratory, Guangzhou, Guangdong 510005, China.
| | - Yan He
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Key Laboratory of Ophthalmology & Visual Sciences, Beijing 100730, China.
| |
Collapse
|
7
|
Faa G, Ziranu P, Pretta A, Cau F, Castagnola M, Spanu D, Saba G, D'Agata AP, Tiwari E, Suri JS, Scartozzi M, Saba L. Cancer-associated fibroblasts (CAFs) and plaque-associated fibroblasts (PAFs): Unraveling the cellular crossroads of atherosclerosis and cancer. Biomed Pharmacother 2025; 188:118145. [PMID: 40373629 DOI: 10.1016/j.biopha.2025.118145] [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: 11/11/2024] [Revised: 05/04/2025] [Accepted: 05/05/2025] [Indexed: 05/17/2025] Open
Abstract
Atherosclerosis is a complex process involving various cells and molecules within the atherosclerotic plaque. Recent evidence suggests that plaque-associated fibroblasts (PAFs), also known as atherosclerosis-associated fibroblasts (AAFs), might play a significant role in the development and progression of the disease. The microenvironment of the atherosclerotic plaque, resembling the tumor microenvironment (TME), includes various cellular populations like plaque-associated macrophages (PAMs), plaque-associated neutrophils (PANs), vascular smooth muscle cells (VSMCs), myeloid-derived suppressor cells (MDSCs), and PAFs. Similar to cancer-associated fibroblasts (CAFs) in tumors, PAFs exhibits a wide range of characteristics and functions. Their interactions with endothelial cells, smooth muscle cells, and other stromal cells, including adventitial fibroblast precursors, significantly influence atherosclerosis progression. Moreover, the ability of PAFs to express various markers such as alpha-SMA, Desmin, VEGF, and GFAP, highlights their diverse origins from different precursor cells, including vascular smooth muscle cells, endothelial cells, glial cells of the enteric nervous system, adventitial fibroblast precursors, as well as resident and circulating fibrocytes. This article explores the molecular interactions between PAFs, cells associated with atherosclerosis, and other stromal cells. It further examines the role of PAFs in the development and progression of atherosclerosis, and compares their features with those of CAFs. The research suggests that studying tumor-associated fibroblasts can help understand fibroblast subpopulations in atherosclerotic plaque. Identifying specific subpopulations could provide new insight into atherosclerosis complexity and lead to the development of innovative drugs for medical intervention.
Collapse
Affiliation(s)
- Gavino Faa
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy; Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, USA
| | - Pina Ziranu
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari 09042, Italy.
| | - Andrea Pretta
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari 09042, Italy
| | - Flaviana Cau
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Massimo Castagnola
- Laboratory of Proteomics, Centro Europeo di Ricerca sul Cervello, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Dario Spanu
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari 09042, Italy
| | - Giorgio Saba
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari 09042, Italy
| | - Alessandra Pia D'Agata
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari 09042, Italy
| | - Ekta Tiwari
- Department of Innovation. Global Biomedical Technologies, Inc., Roseville, CA 95661, USA
| | - Jasjit S Suri
- Department of ECE, Idaho State University, Pocatello, ID, 83209, USA; Department of CE, Graphics Era Deemed to be University, Dehradun 248002, India; University Center for Research & Development, Chandigarh University, Mohali, India; Symbiosis Institute of Technology, Nagpur Campus, Symbiosis International (Deemed University), Pune, INDIA; Stroke Diagnostic and Monitoring Division, AtheroPoint, Roseville, CA 95661, USA
| | - Mario Scartozzi
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari 09042, Italy
| | - Luca Saba
- Department of Medical Sciences and Public Health, Unit of Radiology, University fo Cagliari, Cagliari, Italy
| |
Collapse
|
8
|
Lei W, Zhou K, Lei Y, Li Q, Zhu H. Pathogenesis and Systemic Treatment of Hepatocellular Carcinoma: Current Status and Prospects. Mol Cancer Ther 2025; 24:692-708. [PMID: 39417575 DOI: 10.1158/1535-7163.mct-24-0403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/14/2024] [Accepted: 10/08/2024] [Indexed: 10/19/2024]
Abstract
Hepatocellular carcinoma (HCC) remains one of the major threats to human health worldwide. The emergence of systemic therapeutic options has greatly improved the prognosis of patients with HCC, particularly those with advanced stages of the disease. In this review, we discussed the pathogenesis of HCC, genetic alterations associated with the development of HCC, and alterations in the tumor immune microenvironment. Then, important indicators and emerging technologies related to the diagnosis of HCC are summarized. Also, we reviewed the major advances in treatments for HCC, offering insights into future prospects for next-generation managements.
Collapse
Affiliation(s)
- Wanting Lei
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Kexun Zhou
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ye Lei
- College of Liberal Arts, Neijiang Normal University, Neijiang, China
| | - Qiu Li
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Yang Y, Wang H, Xue Q, Peng W, Zhou Q. New advances of natural products in non-small cell lung cancer: From mechanisms to therapies. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119636. [PMID: 40120701 DOI: 10.1016/j.jep.2025.119636] [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: 11/21/2024] [Revised: 03/13/2025] [Accepted: 03/13/2025] [Indexed: 03/25/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE With the rise of immunotherapy, the treatment approach for non-small cell lung cancer (NSCLC) has undergone revolutionary changes. However, the prognosis for NSCLC patients has not been significantly improved due to the development of acquired drug resistance. Therefore, there is an urgent need to develop new and more effective drugs for treating NSCLC or improving tumor treatment resistance. Traditional Chinese medicine (TCM) has been gradually incorporated into the combined treatment of NSCLC. Its active components (also known as natural products) exhibit novel structures, multi-target effects, diverse pathways, minimal toxicity, and varied biological activities, which play a therapeutic role in various diseases. Thus, natural products hold great potential for future clinical applications. AIM OF THE STUDY Screening main traditional plants widely used in NSCLC and their derived natural products, as well as exploring the mechanisms by which these natural products act on NSCLC-particularly focusing on their applications-can provide valuable insights for the development of therapeutic drugs targeting NSCLC. METHODS A comprehensive, computerized literature search was conducted in PubMed, Embase, Web of Science, Cochrane Library, CNKI Scholar, the American Chemical Abstracts, and Wanfang Database up to June 2024, using the following keywords: "traditional Chinese medicine", "herbal medicine", "medicinal plants", and "herbal", paired with terms such as "non-small cell lung cancer", "therapy", "natural products", and "active ingredient". RESULTS Summarizing current research findings, we discovered eleven medicinal plants containing a total of fourteen natural products. Natural products have a significant impact on tumor progression in NSCLC, including apotosis, autophagy, pyrotosis, cell-cycle arrest and metasis. Moreover, natural products can modulate the activities of various immune cells and reshape the immune microenvironment. Combined with conventional cancer treatments, natural products demonstrate promising therapeutic effects and effectively reverse drug resistance. Furthermore,the use of nano-drug delivery systems to address limitations associated with natural products. CONCLUSIONS This review summarizes eleven medicinal plants containing a total of fourteen natural products that can enhance NSCLC treatment and indicates their action mechanisms. Furthermore, we also discuss limitations of natural products and explore the use of nano-drug delivery systems to address limitations associated with natural products.
Collapse
Affiliation(s)
- Yuening Yang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Haolei Wang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Qianqian Xue
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Wenbei Peng
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Qiong Zhou
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China.
| |
Collapse
|
10
|
Marciniak M, Stachowicz-Suhs M, Wagner M. The role of innate immune cells in modulating vascular dynamics in skin malignancies. Biochim Biophys Acta Rev Cancer 2025; 1880:189331. [PMID: 40280501 DOI: 10.1016/j.bbcan.2025.189331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
A developing tumor relies heavily on blood vessels to supply oxygen and nutrients. As a result, angiogenesis, the formation of new blood vessels, supports tumor growth and progression. Similarly, lymphangiogenesis, the formation of new lymphatic vessels, plays a critical role in metastatic dissemination by providing pathways for malignant cells to spread. The tumor microenvironment is crucial for establishing and maintaining these vascular networks, with innate immune cells playing a key regulatory role. Notably, immune cells are specifically enriched in barrier tissues, such as the skin, emphasizing their importance in skin malignancies. Therefore, understanding their role in regulating angiogenesis and lymphangiogenesis is essential for developing novel therapeutic strategies. This review article explores how innate immune cells influence tumor vasculature and highlights the therapeutic potential that may arise from this knowledge.
Collapse
Affiliation(s)
- Mateusz Marciniak
- Innate Immunity Research Group, Life Sciences and Biotechnology Center, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wrocław, Poland; Department of Biochemistry and Immunochemistry, Wrocław Medical University, Wrocław, Poland
| | - Martyna Stachowicz-Suhs
- Innate Immunity Research Group, Life Sciences and Biotechnology Center, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wrocław, Poland
| | - Marek Wagner
- Innate Immunity Research Group, Life Sciences and Biotechnology Center, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wrocław, Poland.
| |
Collapse
|
11
|
Sun Q, Wang N, Poelchen J, Peter M, Novak D, Özbay Kurt FG, Bitsch R, Wu H, Wang Y, Pardo S, Han R, Liu S, Gong L, Zhang Y, Wistuba-Hamprecht K, Umansky V, Utikal JS. Neural crest-associated gene FOXD1 induces an immunosuppressive microenvironment by regulating myeloid-derived suppressor cells in melanoma. J Immunother Cancer 2025; 13:e010352. [PMID: 40210238 PMCID: PMC11987097 DOI: 10.1136/jitc-2024-010352] [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: 08/14/2024] [Accepted: 03/26/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND Neural crest-associated genes play pivotal roles in tumor initiation, progression, and the intricate dynamics of the tumor microenvironment (TME). Myeloid-derived suppressor cells (MDSC) within the TME are important in dampening T cell activity and contributing to resistance against immunotherapeutic interventions. The neural crest-associated gene Forkhead Box D1 (FOXD1) has been identified as an oncogenic factor that induces melanoma dedifferentiation and progression. However, the underlying mechanisms and the impact of FOXD1 on the antitumor immune response remain unclear. METHODS To investigate the impacts of FOXD1 on the melanoma microenvironment, we analyzed publicly available datasets from multiple platforms, including TNMplot, TIMER2.0, etc. In addition, FOXD1 was overexpressed (OE) or knocked down in melanoma cells to identify its biological functions in vitro and in vivo. Flow cytometry and arginase activity assay were used to analyze the phenotype and function of MDSC. Western blot, reverse transcription-PCR, or ELISA assays were employed to analyze the expression of FOXD1 and its downstream effectors. In vivo experiments were conducted to investigate the role of FOXD1 in melanoma progression and the influence on MDSC accumulation within the TME. RESULTS We demonstrate that increased FOXD1 levels inversely correlated with melanoma responsiveness to immunotherapy. Ex-vivo analyses unveiled that monocytes, exposed to conditioned medium from FOXD1-OE melanoma cells, effectively suppressed T cell proliferation and upregulated the expression of programmed death-ligand 1 (PD-L1) and other immunosuppressive factors. FOXD1 was identified as a direct regulator of interleukin 6 (IL6) expression, which is pivotal for MDSC induction. Blocking IL6 reversed MDSC-associated immunosuppression. Additionally, miR-581, a potential negative regulator of FOXD1, attenuated the impact of FOXD1 on IL6 expression and MDSC differentiation. In vivo experiments demonstrated that tumors derived from FOXD1 OE melanoma cells contained a significantly higher frequency of PD-L1+ MDSC compared with controls, while FOXD1 knockdown resulted in reduced tumor growth and diminished MDSC accumulation. CONCLUSION Our study elucidated a novel function of FOXD1 in melanoma pathogenesis, highlighting its role in orchestrating the immunosuppressive TME by promoting the generation of MDSC via IL6 upregulation.
Collapse
Affiliation(s)
- Qian Sun
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and DKFZ Hector Cancer Institute, Heidelberg, Baden-Württemberg, Germany
- Department of Dermatology Venereology and Allergology, Heidelberg University, Mannheim, Baden-Württemberg, Germany
| | - Nina Wang
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and DKFZ Hector Cancer Institute, Heidelberg, Baden-Württemberg, Germany
- Department of Dermatology Venereology and Allergology, Heidelberg University, Mannheim, Baden-Württemberg, Germany
| | - Juliane Poelchen
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and DKFZ Hector Cancer Institute, Heidelberg, Baden-Württemberg, Germany
- Department of Dermatology Venereology and Allergology, Heidelberg University, Mannheim, Baden-Württemberg, Germany
| | - Mareike Peter
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and DKFZ Hector Cancer Institute, Heidelberg, Baden-Württemberg, Germany
- Department of Dermatology Venereology and Allergology, Heidelberg University, Mannheim, Baden-Württemberg, Germany
| | - Daniel Novak
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and DKFZ Hector Cancer Institute, Heidelberg, Baden-Württemberg, Germany
- Department of Dermatology Venereology and Allergology, Heidelberg University, Mannheim, Baden-Württemberg, Germany
| | - Feyza Gül Özbay Kurt
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and DKFZ Hector Cancer Institute, Heidelberg, Baden-Württemberg, Germany
- Department of Dermatology Venereology and Allergology, Heidelberg University, Mannheim, Baden-Württemberg, Germany
| | - Rebekka Bitsch
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and DKFZ Hector Cancer Institute, Heidelberg, Baden-Württemberg, Germany
- Department of Dermatology Venereology and Allergology, Heidelberg University, Mannheim, Baden-Württemberg, Germany
| | - Huizi Wu
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and DKFZ Hector Cancer Institute, Heidelberg, Baden-Württemberg, Germany
- Department of Dermatology Venereology and Allergology, Heidelberg University, Mannheim, Baden-Württemberg, Germany
| | - Yiman Wang
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and DKFZ Hector Cancer Institute, Heidelberg, Baden-Württemberg, Germany
- Department of Dermatology Venereology and Allergology, Heidelberg University, Mannheim, Baden-Württemberg, Germany
| | - Sandra Pardo
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and DKFZ Hector Cancer Institute, Heidelberg, Baden-Württemberg, Germany
- Department of Dermatology Venereology and Allergology, Heidelberg University, Mannheim, Baden-Württemberg, Germany
| | - Rui Han
- Department of Otorhinolaryngology, Head and Neck Surgery, Heidelberg University, Heidelberg, Baden-Württemberg, Germany
| | - Shibo Liu
- State Key Laboratory of Molecular Oncology, Tsinghua university, School of Basic Medical Sciences, Beijing, China
| | - Lidong Gong
- Peking University Institute of Systems Biomedicine, Beijing, China
| | - Yuxin Zhang
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and DKFZ Hector Cancer Institute, Heidelberg, Baden-Württemberg, Germany
- Department of Dermatology Venereology and Allergology, Heidelberg University, Mannheim, Baden-Württemberg, Germany
| | - Kilian Wistuba-Hamprecht
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and DKFZ Hector Cancer Institute, Heidelberg, Baden-Württemberg, Germany
- Department of Dermatology Venereology and Allergology, Heidelberg University, Mannheim, Baden-Württemberg, Germany
| | - Viktor Umansky
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and DKFZ Hector Cancer Institute, Heidelberg, Baden-Württemberg, Germany
- Department of Dermatology Venereology and Allergology, Heidelberg University, Mannheim, Baden-Württemberg, Germany
| | - Jochen Sven Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and DKFZ Hector Cancer Institute, Heidelberg, Baden-Württemberg, Germany
- Department of Dermatology Venereology and Allergology, Heidelberg University, Mannheim, Baden-Württemberg, Germany
| |
Collapse
|
12
|
Zhu Y, Cao S. Unraveling the Complexities of Myeloid-Derived Suppressor Cells in Inflammatory Bowel Disease. Int J Mol Sci 2025; 26:3291. [PMID: 40244120 PMCID: PMC11989781 DOI: 10.3390/ijms26073291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 03/25/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) regulate immune responses in many pathological conditions, one of which is inflammatory bowel disease (IBD), an incurable chronic disorder of the digestive tract and beyond. The pathophysiology of IBD remains unclear, likely involving aberrant innate and adaptive immunity. Studies have reported altered population of MDSCs in patients with IBD. However, their distribution varies among patients and different preclinical models of IBD. The expansion and activation of MDSCs are likely driven by various stimuli during intestinal inflammation, but the in-depth mechanisms remain poorly understood. The role of MDSCs in the pathogenesis of IBD appears to be paradoxical. In addition to intestinal inflammation, suppressive MDSCs may promote colitis-to-colon cancer transition. In this Review, we summarize recent progresses on the features, activation, and roles of MDSCs in the development of IBD and IBD-associated colon cancer.
Collapse
Affiliation(s)
| | - Siyan Cao
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA;
| |
Collapse
|
13
|
Ibrahim A, Mohamady Farouk Abdalsalam N, Liang Z, Kashaf Tariq H, Li R, O Afolabi L, Rabiu L, Chen X, Xu S, Xu Z, Wan X, Yan D. MDSC checkpoint blockade therapy: a new breakthrough point overcoming immunosuppression in cancer immunotherapy. Cancer Gene Ther 2025; 32:371-392. [PMID: 40140724 PMCID: PMC11976280 DOI: 10.1038/s41417-025-00886-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/25/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025]
Abstract
Despite the success of cancer immunotherapy in treating hematologic malignancies, their efficacy in solid tumors remains limited due to the immunosuppressive tumor microenvironment (TME), which is mainly formed by myeloid-derived suppressor cells (MDSCs). MDSCs not only exert potent immunosuppressive effects that hinder the success of immune checkpoint inhibitors (ICIs) and adaptive cellular therapies, but they also promote tumor advancement through non-immunological pathways, including promoting angiogenesis, driving epithelial-mesenchymal transition (EMT), and contributing to the establishment of pre-metastatic environments. While targeting MDSCs alone or in combination with conventional therapies has shown limited success, emerging evidence suggests that MDSC checkpoint blockade in combination with other immunotherapies holds great promise in overcoming both immunological and non-immunological barriers. In this review, we discussed the dual roles of MDSCs, with a particular emphasis on their underexplored checkpoints blockade strategies. We discussed the rationale behind combination strategies, their potential advantages in overcoming MDSC-mediated immunosuppression, and the challenges associated with their development. Additionally, we highlight future research directions aimed at optimizing combination immunotherapies to enhance cancer therapeutic effectiveness, particularly in solid tumor therapies where MDSCs are highly prevalent.
Collapse
Affiliation(s)
- Abdulrahman Ibrahim
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
- University of Chinese Academy of Sciences, 100864, Beijing, China
| | - Nada Mohamady Farouk Abdalsalam
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
- University of Chinese Academy of Sciences, 100864, Beijing, China
| | - Zihao Liang
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
| | - Hafiza Kashaf Tariq
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
- University of Chinese Academy of Sciences, 100864, Beijing, China
| | - Rong Li
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
| | - Lukman O Afolabi
- Department of Pediatrics, Indiana University School of Medicine, 1234 Notre Dame Ave, South Bend, IN, 46617, USA
| | - Lawan Rabiu
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
- University of Chinese Academy of Sciences, 100864, Beijing, China
| | - Xuechen Chen
- College of Pharmacy, Jinan University, 511436, Guangzhou, China.
| | - Shu Xu
- Cancer Center, Shenzhen Guangming District People's Hospital, 518106, Shenzhen, China
| | - Zhiming Xu
- Cancer Center, Shenzhen Guangming District People's Hospital, 518106, Shenzhen, China.
| | - Xiaochun Wan
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China.
- University of Chinese Academy of Sciences, 100864, Beijing, China.
| | - Dehong Yan
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China.
- University of Chinese Academy of Sciences, 100864, Beijing, China.
| |
Collapse
|
14
|
Yan C, Wang XF. Tumor immune evasion: Systemic immunosuppressive networks beyond the local microenvironment. Proc Natl Acad Sci U S A 2025; 122:e2502597122. [PMID: 40112118 PMCID: PMC11962438 DOI: 10.1073/pnas.2502597122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025] Open
Affiliation(s)
- Chengsong Yan
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC27710
| | - Xiao-Fan Wang
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC27710
| |
Collapse
|
15
|
Hao Z, Guan W, Wei W, Li M, Xiao Z, Sun Q, Pan Y, Xin W. Unlocking the therapeutic potential of tumor-derived EVs in ischemia-reperfusion: a breakthrough perspective from glioma and stroke. J Neuroinflammation 2025; 22:84. [PMID: 40089793 PMCID: PMC11909855 DOI: 10.1186/s12974-025-03405-7] [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: 01/12/2025] [Accepted: 03/04/2025] [Indexed: 03/17/2025] Open
Abstract
Clinical studies have revealed a bidirectional relationship between glioma and ischemic stroke, with evidence of spatial overlap between the two conditions. This connection arises from significant similarities in their pathological processes, including the regulation of cellular metabolism, inflammation, coagulation, hypoxia, angiogenesis, and neural repair, all of which involve common biological factors. A significant shared feature of both diseases is the crucial role of extracellular vesicles (EVs) in mediating intercellular communication. Extracellular vesicles, with their characteristic bilayer structure, encapsulate proteins, lipids, and nucleic acids, shielding them from enzymatic degradation by ribonucleases, deoxyribonucleases, and proteases. This structural protection facilitates long-distance intercellular communication in multicellular organisms. In gliomas, EVs are pivotal in intracranial signaling and shaping the tumor microenvironment. Importantly, the cargos carried by glioma-derived EVs closely align with the biological factors involved in ischemic stroke, underscoring the substantial impact of glioma on stroke pathology, particularly through the crucial roles of EVs as key mediators in this interaction. This review explores the pathological interplay between glioma and ischemic stroke, addressing clinical manifestations and pathophysiological processes across the stages of hypoxia, stroke onset, progression, and recovery, with a particular focus on the crucial role of EVs and their cargos in these interactions.
Collapse
Affiliation(s)
- Zhongnan Hao
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P. R. China
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Department of Neurology, The Affiliated Hospital of Qingdao University, Medical School of Qingdao University, Qingdao, 266100, Shandong Province, China
| | - Wenxin Guan
- Queen Mary School, Nanchang University, Xuefu Avenue, Nanchang, Jiangxi, China
| | - Wei Wei
- Department of Neurology, the Affiliated Hospital of Southwest Jiaotong University & The Third People's Hospital of Chengdu, Chengdu, 610031, Sichuan, PR China
| | - Meihua Li
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zhipeng Xiao
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Qinjian Sun
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P. R. China
| | - Yongli Pan
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P. R. China.
| | - Wenqiang Xin
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
16
|
Natusch L, Heiduk M, Klimova A, Beer C, Willms T, Digomann D, Reiche C, Aust DE, Hempel S, Oehme F, Distler M, Weitz J, Seifert AM, Seifert L. The Value of Blood T Cell Frequencies for Risk Prediction of Postoperative Complications in Pancreatic Cancer Surgery. ANNALS OF SURGERY OPEN 2025; 6:e545. [PMID: 40134486 PMCID: PMC11932582 DOI: 10.1097/as9.0000000000000545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 01/01/2025] [Indexed: 03/27/2025] Open
Abstract
Objective To quantify and characterize preoperative blood T cells in patients with pancreatic ductal adenocarcinoma (PDAC), who underwent tumor resection, and to determine their relevance as biomarkers for postoperative pancreas-specific complications. Background Pancreas-specific complications after pancreatic surgery are associated with a high morbidity and mortality, which both deprive patients of adjuvant chemotherapy. Noninvasive biomarkers for risk prediction of postoperative complications are missing, and the role of blood T cells for preoperative risk stratification is unknown. Methods The preoperative frequency of blood T cell subsets was analyzed for 73 patients with PDAC, who underwent proximal pancreatectomy. Patients were screened for postoperative complications such as pancreatic fistula, postpancreatectomy hemorrhage, and postpancreatectomy acute pancreatitis. The frequency of CD8+, conventional CD4+, and regulatory T cells, as well as the differentiation state of each T cell subset in the peripheral blood of patients with PDAC, was analyzed. Results Of 73 patients with PDAC, 19.2% developed pancreas-specific complications. The occurrence of postoperative complications was independent of the type of resection performed (Whipple procedure vs pylorus-preserving pancreaticoduodenectomy). Neither the frequency of CD8+, conventional CD4+, and regulatory T cells nor the state of T cell differentiation in the peripheral blood was associated with postoperative pancreas-specific complications. Notably, a significantly lower preoperative bilirubin serum level was observed in patients, who developed postpancreatectomy hemorrhage after proximal pancreatectomy (P =0.001). Conclusions A low preoperative bilirubin serum level was associated with a higher risk for postpancreatectomy hemorrhage after proximal pancreatectomy. However, the preoperative blood T cell frequency does not predict postoperative pancreas-specific complications.
Collapse
Affiliation(s)
- Loreen Natusch
- From the Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Max Heiduk
- From the Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Anna Klimova
- National Center for Tumor Diseases (NCT), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Carolin Beer
- From the Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Tido Willms
- From the Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - David Digomann
- From the Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Charlotte Reiche
- From the Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Daniela E. Aust
- Faculty of Medicine Carl Gustav Carus, Institute of Pathology, Technische Universität Dresden, Dresden, Germany
- NCT, Biobank Dresden, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Sebastian Hempel
- From the Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Florian Oehme
- From the Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Marius Distler
- From the Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Jürgen Weitz
- From the Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Adrian M. Seifert
- From the Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lena Seifert
- From the Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Else Kröner Clinician Scientist Professorship for Translational Tumor Immunological Research, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
17
|
KA HYEIN, MUN SEHWAN, HAN SORA, YANG YOUNG. Targeting myeloid-derived suppressor cells in the tumor microenvironment: potential therapeutic approaches for osteosarcoma. Oncol Res 2025; 33:519-531. [PMID: 40109854 PMCID: PMC11915044 DOI: 10.32604/or.2024.056860] [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: 08/01/2024] [Accepted: 10/14/2024] [Indexed: 03/22/2025] Open
Abstract
Osteosarcoma is a bone malignancy characterized by strong invasiveness and rapid disease progression. The tumor microenvironment of osteosarcoma contains various types of immune cells, including myeloid-derived suppressor cells, macrophages, T cells, and B cells. Imbalances of these immune cells can promote the proliferation and metastasis of osteosarcoma. Recent studies have indicated a substantial increase in the levels of myeloid-derived suppressor cells, an immune cell associated with immunosuppressive and pro-cancer effects, in the peripheral blood of patients with osteosarcoma. Moreover, the levels of the pro-inflammatory cytokine interleukin 18 are positively correlated with those of myeloid-derived suppressor cells in the peripheral blood of animal models of osteosarcoma. In this review, we explore the function of myeloid-derived suppressor cells in osteosarcoma based on the clinical diagnoses of patients with osteosarcoma and discuss future therapeutic approaches for targeting osteosarcoma. Targeting myeloid-derived suppressor cells represents a promising approach to improving the prognosis and survival rates of patients with osteosarcoma.
Collapse
Affiliation(s)
| | | | | | - YOUNG YANG
- Research Institute of Women’s Health, Sookmyung Women’s University, Seoul, 04312, Republic of Korea
| |
Collapse
|
18
|
Li C, Xue Y, Yinwang E, Ye Z. The Recruitment and Immune Suppression Mechanisms of Myeloid-Derived Suppressor Cells and Their Impact on Bone Metastatic Cancer. Cancer Rep (Hoboken) 2025; 8:e70044. [PMID: 39947253 PMCID: PMC11825175 DOI: 10.1002/cnr2.70044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 09/16/2024] [Accepted: 10/04/2024] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND MDSCs are immature neutrophils and monocytes with immunosuppressive potentials, involving mononuclear MDSCs (M-MDSCs) and polymorphonuclear MDSCs (PMN-MDSCs). RECENT FINDINGS They are significant components of the tumor microenvironment (TME). Besides, recent studies also verified that MDSCs also facilitated the progression of bone metastasis by regulating the network of cytokines and the function of immune cells. CONCLUSION It is necessary to summarize the mechanisms of MDSC recruitment and immunosuppression, and their impact on bone metastasis.
Collapse
Affiliation(s)
- Chengyuan Li
- Department of Orthopedic Surgery, the Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Yucheng Xue
- Department of Orthopedic Surgery, the Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Eloy Yinwang
- Department of Orthopedic Surgery, the Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Zhaoming Ye
- Department of Orthopedic Surgery, the Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Orthopedics Research Institute of Zhejiang UniversityHangzhouChina
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceHangzhouChina
| |
Collapse
|
19
|
He N, Yuan D, Luo M, Xu Q, Wen Z, Wang Z, Zhao J, Liu Y. Ferroptosis contributes to immunosuppression. Front Med 2025; 19:1-22. [PMID: 39560919 DOI: 10.1007/s11684-024-1080-8] [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: 12/26/2023] [Accepted: 04/18/2024] [Indexed: 11/20/2024]
Abstract
As a novel form of cell death, ferroptosis is mainly regulated by the accumulation of soluble iron ions in the cytoplasm and the production of lipid peroxides and is closely associated with several diseases, including acute kidney injury, ischemic reperfusion injury, neurodegenerative diseases, and cancer. The term "immunosuppression" refers to various factors that can directly harm immune cells' structure and function and affect the synthesis, release, and biological activity of immune molecules, leading to the insufficient response of the immune system to antigen production, failure to successfully resist the invasion of foreign pathogens, and even organ damage and metabolic disorders. An immunosuppressive phase commonly occurs in the progression of many ferroptosis-related diseases, and ferroptosis can directly inhibit immune cell function. However, the relationship between ferroptosis and immunosuppression has not yet been published due to their complicated interactions in various diseases. Therefore, this review deeply discusses the contribution of ferroptosis to immunosuppression in specific cases. In addition to offering new therapeutic targets for ferroptosis-related diseases, the findings will help clarify the issues on how ferroptosis contributes to immunosuppression.
Collapse
Affiliation(s)
- Nina He
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, China
- National Medicine Functional Experimental Teaching Center, Changsha, 410008, China
| | - Dun Yuan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Minjie Luo
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, China
- National Medicine Functional Experimental Teaching Center, Changsha, 410008, China
| | - Qing Xu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, China
- National Medicine Functional Experimental Teaching Center, Changsha, 410008, China
| | - Zhongchi Wen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, China
- National Medicine Functional Experimental Teaching Center, Changsha, 410008, China
| | - Ziqin Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, China
- National Medicine Functional Experimental Teaching Center, Changsha, 410008, China
| | - Jie Zhao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, China.
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, China.
- National Medicine Functional Experimental Teaching Center, Changsha, 410008, China.
| | - Ying Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, China.
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, China.
- National Medicine Functional Experimental Teaching Center, Changsha, 410008, China.
| |
Collapse
|
20
|
Shi J, Pei X, Peng J, Wu C, Lv Y, Wang X, Zhou Y, Yuan X, Dong X, Zhou S, Xu D, Zhao J, Liu J, Huang J, Du B, Yao C, Zeng X, Li M, Chen H, Wang Q. Monocyte-macrophage dynamics as key in disparate lung and peripheral immune responses in severe anti-melanoma differentiation-associated gene 5-positive dermatomyositis-related interstitial lung disease. Clin Transl Med 2025; 15:e70226. [PMID: 39902678 PMCID: PMC11791760 DOI: 10.1002/ctm2.70226] [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: 10/22/2024] [Revised: 01/18/2025] [Accepted: 01/28/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Anti-melanoma differentiation-associated gene 5-positive dermatomyositis (anti-MDA5+ DM) is a rare inflammatory autoimmune disorder often complicated by life-threatening rapidly progressive interstitial lung disease (RP-ILD). The underlying mechanisms driving immune dysfunction and lung injury, however, remain poorly understood. The study aims to gain insights into the disrupted immune landscape in peripheral and pulmonary compartments of severe anti-MDA5+ DM and explore potential therapeutic targets. METHODS We employed single-cell RNA sequencing to examine cellular constituents within five patients' bronchoalveolar lavage fluid and paired peripheral blood mononuclear cells. Luminex assay and flow cytometry were further applied to validate the results. RESULTS Our analysis revealed starkly contrasting immune landscapes between the periphery and lungs, with peripheral immune suppression juxtaposed against pulmonary immune hyperactivation. Central to this dysregulation was the monocyte-macrophage lineage. Circulating monocytes exhibited an immunosuppressive phenotype, characterised by diminished cytokine production, reduced MHC II expression, and features resembling myeloid-derived suppressor cells. These monocytes were recruited to the lungs, where they differentiated into monocyte-derived alveolar macrophages (Mo-AMs) with robust proinflammatory and profibrotic activities. Mo-AMs drove cytokine storms and produced chemokines that amplified inflammatory cell recruitment and lung tissue remodelling. Additionally, peripheral T and NK cells exhibited increased cell death and active migration into the lungs, which may be the cause of lymphopenia. CONCLUSIONS Our study underscores the pivotal role of monocyte-macrophage dynamics in the immunopathogenesis of anti-MDA5+-associated RP-ILD, offering critical insights into compartment-specific immune dysregulation. These findings suggest potential therapeutic strategies targeting monocyte recruitment and macrophage activation to mitigate disease progression. KEY POINTS Peripheral immune suppression and pulmonary immune hyperactivation characterise the distinct immune landscapes in anti-MDA5+DM with RP-ILD. Circulating monocytes transition from an immunosuppressive phenotype in the periphery to proinflammatory and profibrotic Mo-AMs in the lungs. Chemokines produced by Mo-AMs drive monocyte and other immune cell recruitment to the lungs, amplifying pulmonary inflammation.
Collapse
Affiliation(s)
- Jia Shi
- Department of Rheumatology and Clinical ImmunologyPeking Union Medical College HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeNational Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC‐DID), Ministry of Science & TechnologyState Key Laboratory of Complex Severe and Rare DiseasesKey Laboratory of Rheumatology and Clinical ImmunologyMinistry of EducationBeijingChina
| | - Xiaoya Pei
- Department of Biochemistry and Molecular BiologyState Key Laboratory of Medical Molecular BiologyInstitute of Basic Medical SciencesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jinmin Peng
- Medical Intensive Care UnitState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical SciencesBeijingChina
| | - Chanyuan Wu
- Department of Rheumatology and Clinical ImmunologyPeking Union Medical College HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeNational Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC‐DID), Ministry of Science & TechnologyState Key Laboratory of Complex Severe and Rare DiseasesKey Laboratory of Rheumatology and Clinical ImmunologyMinistry of EducationBeijingChina
| | - Yulin Lv
- Department of Rheumatology and Clinical ImmunologyPeking Union Medical College HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeNational Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC‐DID), Ministry of Science & TechnologyState Key Laboratory of Complex Severe and Rare DiseasesKey Laboratory of Rheumatology and Clinical ImmunologyMinistry of EducationBeijingChina
| | - Xiaoman Wang
- Department of Biochemistry and Molecular BiologyState Key Laboratory of Medical Molecular BiologyInstitute of Basic Medical SciencesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yangzhong Zhou
- Department of Rheumatology and Clinical ImmunologyPeking Union Medical College HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeNational Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC‐DID), Ministry of Science & TechnologyState Key Laboratory of Complex Severe and Rare DiseasesKey Laboratory of Rheumatology and Clinical ImmunologyMinistry of EducationBeijingChina
| | - Xueting Yuan
- Department of Rheumatology and Clinical ImmunologyPeking Union Medical College HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeNational Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC‐DID), Ministry of Science & TechnologyState Key Laboratory of Complex Severe and Rare DiseasesKey Laboratory of Rheumatology and Clinical ImmunologyMinistry of EducationBeijingChina
| | - Xingbei Dong
- Department of Rheumatology and Clinical ImmunologyPeking Union Medical College HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeNational Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC‐DID), Ministry of Science & TechnologyState Key Laboratory of Complex Severe and Rare DiseasesKey Laboratory of Rheumatology and Clinical ImmunologyMinistry of EducationBeijingChina
| | - Shuang Zhou
- Department of Rheumatology and Clinical ImmunologyPeking Union Medical College HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeNational Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC‐DID), Ministry of Science & TechnologyState Key Laboratory of Complex Severe and Rare DiseasesKey Laboratory of Rheumatology and Clinical ImmunologyMinistry of EducationBeijingChina
| | - Dong Xu
- Department of Rheumatology and Clinical ImmunologyPeking Union Medical College HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeNational Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC‐DID), Ministry of Science & TechnologyState Key Laboratory of Complex Severe and Rare DiseasesKey Laboratory of Rheumatology and Clinical ImmunologyMinistry of EducationBeijingChina
| | - Jiuliang Zhao
- Department of Rheumatology and Clinical ImmunologyPeking Union Medical College HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeNational Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC‐DID), Ministry of Science & TechnologyState Key Laboratory of Complex Severe and Rare DiseasesKey Laboratory of Rheumatology and Clinical ImmunologyMinistry of EducationBeijingChina
| | - Jun Liu
- State Key Laboratory of Protein and Plant Gene ResearchSchool of Life Sciences, Peking‐Tsinghua Center for Life SciencesPeking UniversityBeijingChina
| | - Jiao Huang
- Department of RheumatologyAffiliated Hangzhou First People's HospitalWestlake University School of MedicineHangzhouChina
| | - Bin Du
- Medical Intensive Care UnitState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical SciencesBeijingChina
| | - Chen Yao
- College of Pulmonary and Critical Care MedicineChinese PLA General HospitalBeijingChina
| | - Xiaofeng Zeng
- Department of Rheumatology and Clinical ImmunologyPeking Union Medical College HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeNational Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC‐DID)Ministry of Science & TechnologyState Key Laboratory of Common Mechanism Research for Major DiseasesKey Laboratory of Rheumatology and Clinical ImmunologyMinistry of EducationBeijingChina
| | - Mengtao Li
- Department of Rheumatology and Clinical ImmunologyPeking Union Medical College HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeNational Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC‐DID), Ministry of Science & TechnologyState Key Laboratory of Complex Severe and Rare DiseasesKey Laboratory of Rheumatology and Clinical ImmunologyMinistry of EducationBeijingChina
| | - Houzao Chen
- Department of Biochemistry and Molecular BiologyState Key Laboratory of Medical Molecular BiologyInstitute of Basic Medical SciencesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Qian Wang
- Department of Rheumatology and Clinical ImmunologyPeking Union Medical College HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeNational Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC‐DID)Ministry of Science & TechnologyState Key Laboratory of Common Mechanism Research for Major DiseasesKey Laboratory of Rheumatology and Clinical ImmunologyMinistry of EducationBeijingChina
| |
Collapse
|
21
|
Bouzeineddine NZ, Philippi A, Gee K, Basta S. Granulocyte macrophage colony stimulating factor in virus-host interactions and its implication for immunotherapy. Cytokine Growth Factor Rev 2025; 81:54-63. [PMID: 39755463 DOI: 10.1016/j.cytogfr.2024.12.002] [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: 10/03/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 01/06/2025]
Abstract
Viruses have evolved to strategically exploit cellular signaling pathways to evade host immune defenses. GM-CSF signaling plays a pivotal role in regulating inflammation, activating myeloid cells, and enhancing the immune response to infections. Due to its central role in the immune system, viruses may target this pathway to further establish infection. This review focuses on key studies elucidating virus interactions with GM-CSF signaling proteins and summarizes findings on the impact of viral infections on GM-CSF production. Additionally, therapeutic strategies centered around GM-CSF are investigated, such as the potential benefits of administering GM-CSF versus inhibiting GM-CSF signaling to mitigate viral-induced aberrant inflammation. Understanding these virus-host interactions provides valuable insights that help further our understanding to develop future therapeutic approaches in modulating the immune response during viral infections.
Collapse
Affiliation(s)
- Nasry Zane Bouzeineddine
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Alecco Philippi
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Katrina Gee
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada.
| | - Sam Basta
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada.
| |
Collapse
|
22
|
Das L, Das S. A comprehensive insights of cancer immunotherapy resistance. Med Oncol 2025; 42:57. [PMID: 39883235 DOI: 10.1007/s12032-025-02605-8] [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: 12/07/2024] [Accepted: 01/09/2025] [Indexed: 01/31/2025]
Abstract
Cancer is a major global health issue that is usually treated with multiple therapies, such as chemotherapy and targeted therapies like immunotherapy. Immunotherapy is a new and alternative approach to treating various types of cancer that are difficult to treat with other methods. Although immune checkpoint inhibitors have shown promise for long-term efficacy, they have limited effectiveness in common cancer types such as breast, prostate, and lung. Some patients do not respond to immunotherapy, while others develop resistance to the treatment over time, which is classified as primary or acquired resistance. Cancer immunotherapy, specifically immune checkpoint inhibitor-based resistance involves multiple factors such as genes, metabolism, inflammation, and angiogenesis. However, cutting-edge research has identified the mechanisms of immunotherapy resistance and possible solutions. Current research may improve biomarker identification and modify treatment strategies, which will lead to better clinical outcomes. This review provides a comprehensive discussion of the current mechanisms of immunotherapy resistance, related biomarker modulation, and strategies to overcome resistance.
Collapse
Affiliation(s)
- Laavanya Das
- Department of Food and Nutrition, Brainware University, 398, Ramkrishnapur Rd, Barasat, Kolkata, West Bengal, 700125, India
| | - Subhadip Das
- Department of In Vivo Pharmacology, TCG Lifesciences Pvt. Ltd, BN 7, Sector V, Salt Lake City, Kolkata, West Bengal, 700091, India.
| |
Collapse
|
23
|
Cheng Z, Lu J, Chen Y, Cao W, Shao Q. The role of CD101 and Tim3 in the immune microenvironment of gastric cancer and their potential as prognostic biomarkers. Int Immunopharmacol 2025; 146:113835. [PMID: 39700955 DOI: 10.1016/j.intimp.2024.113835] [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: 10/30/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND Gastric cancer (GC) is a prevalent malignancy. Current treatment modalities, including surgery, chemotherapy, radiotherapy, and targeted therapy, have limitations in early detection and personalized treatment, necessitating the discovery of novel biomarkers and therapeutic strategies. This study aims to elucidate the molecular mechanisms underlying GC, focusing on the differentially expressed genes (DEGs) of CD101- Tim3+ CD8+ T cells (CCT precursors) and CD101+ Tim3+ CD8+ T cells (CCT). METHODS Utilizing a multi-omics approach, including transcriptomic sequencing, single-cell RNA sequencing, cell communication analysis, and enrichment analysis. RESULTS We identified 140 genes significantly associated with overall survival in GC patients, including LYAR, ASCL2, and EMP2. A risk score model based on 14 prognostic genes was constructed, demonstrating a significant inverse correlation with survival time (p < 0.05). Immune response analysis indicated decreased infiltration of Activated B cells, CD56bright natural killer cells, and Monocytes in the high-risk group, while CD56dim natural killer cells and Gamma delta T cells were significantly increased, suggesting alterations in the immune microenvironment that influence patient prognosis. Furthermore, drug sensitivity analysis revealed potential responsiveness of high-risk patients to BI-2536, supporting personalized treatment approaches. Cell communication analysis indicated reduced intercellular interactions in PD-1 immunotherapy groups, highlighting the impact of immunotherapy on the tumor microenvironment. GSEA (Gene Set Enrichment Analysis) and GSVA (Gene Set Variation Analysis) revealed enrichment in DNA replication and proteasome pathways in high-risk groups, providing insights into the molecular mechanisms of GC. CONCLUSIONS This study established a foundation for future exploration of targeted therapies and personalized treatment strategies in GC.
Collapse
Affiliation(s)
- Zhouyang Cheng
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Junfen Lu
- Department of Chemotherapy, Afliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Yaping Chen
- Department of Chemotherapy, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Wei Cao
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China.
| | - Qi Shao
- Department of Chemotherapy, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
24
|
Mestiri S, Sami A, Sah N, El-Ella DMA, Khatoon S, Shafique K, Raza A, Mathkor DM, Haque S. Cellular plasticity and non-small cell lung cancer: role of T and NK cell immune evasion and acquisition of resistance to immunotherapies. Cancer Metastasis Rev 2025; 44:27. [PMID: 39856479 DOI: 10.1007/s10555-025-10244-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 01/08/2025] [Indexed: 01/27/2025]
Abstract
Lung cancer is a leading global cause of mortality, with non-small cell lung cancer (NSCLC) accounting for a significant portion of cases. Immune checkpoint inhibitors (ICIs) have transformed NSCLC treatment; however, many patients remain unresponsive. ICI resistance in NSCLC and its association with cellular plasticity, epithelial-mesenchymal transition (EMT), enhanced adaptability, invasiveness, and resistance is largely influenced by epigenetic changes, signaling pathways, tumor microenvironment, and associated immune cells, fibroblasts, and cytokines. Immunosuppressive cells, including M2 tumor-associated macrophages, myeloid-derived suppressor cells, and regulatory T cells, contribute to resistance by suppressing the immune response. This cellular plasticity is influenced when B cells, natural killer cells, and T cells are exhausted or inhibited by components of the tumor microenvironment. Conversely, diverse T cell, NK cell, and B cell subsets hold potential as predictive response markers particularly cytotoxic CD8+ T cells, effector memory T cells, activated T cells, tumor infiltrated NK cells, tertiary lymphoid structures, etc. influence treatment response. Identifying specific gene expressions and immunophenotypes within T cells may offer insights into early clinical responses to immunotherapy. ICI resistance in NSCLC is a multifaceted process shaped by tumor plasticity, the complex tumor microenvironment, and dynamic immune cell changes. Comprehensive analysis of these factors may lead to the identification of novel biomarkers and combination therapies to enhance ICI efficacy in NSCLC treatment.
Collapse
Affiliation(s)
- Sarra Mestiri
- Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Ana Sami
- Queen Mary University of London, London, UK
| | - Naresh Sah
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, USA
| | - Dina Moustafa Abo El-Ella
- Translational Cancer Research Facility, National Center for Cancer Care and Research/Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Sabiha Khatoon
- Department of Physiology and Biochemistry, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Khadija Shafique
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, UAE
| | - Afsheen Raza
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi, UAE.
| | - Darin Mansor Mathkor
- Research and Scientific Studies Unit, College of Nursing and Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Health Sciences, Jazan University, Jazan, Saudi Arabia.
- Universidad Espiritu Santo, Samborondon, Ecuador.
| |
Collapse
|
25
|
Kishta MS, Khamis A, Am H, Elshaar AH, Gül D. Exploring the tumor-suppressive role of miRNA-200c in head and neck squamous cell carcinoma: Potential and mechanisms of exosome-mediated delivery for therapeutic applications. Transl Oncol 2025; 51:102216. [PMID: 39615277 DOI: 10.1016/j.tranon.2024.102216] [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: 09/05/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 12/11/2024] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) remains a challenging malignancy due to its high rates of recurrence, metastasis, and resistance to conventional therapies. microRNA-200c (miRNA-200c) has emerged as a critical tumor suppressor in HNSCC, with the potential to inhibit epithelial-mesenchymal transition (EMT), which is considered as a key process in cancer metastasis and progression. Interestingly, there are also controversial findings in HNSCC characterizing miRNA-200c as oncogenic factor. This review article provides a comprehensive overview of the current understanding of miRNA-200c's general role in cancer, and particularly in HNSCC, highlighting its mechanisms of action, including the regulation of EMT and other oncogenic pathways. Additionally, the review explores the innovative approach of exosome-mediated delivery of miRNA-200c as a therapeutic strategy. Exosomes, as natural nanocarriers, offer a promising vehicle for the targeted delivery of miRNA-200c to tumor cells, potentially overcoming the limitations of traditional delivery methods and enhancing therapeutic efficacy. The review also discusses the challenges and future directions in the clinical application of miRNA-200c, particularly focusing on its potential to improve outcomes for HNSCC patients. This article seeks to provide valuable insights for researchers and clinicians working towards innovative treatments for this aggressive cancer type.
Collapse
Affiliation(s)
- Mohamed S Kishta
- Hormones Department, Medical Research and Clinical Studies Institute, Stem Cell Lab., Center of Excellence for Advanced Sciences, National Research Centre, 33 El Bohouth St., Dokki, 12622 Cairo, Egypt.
| | - Aya Khamis
- Maxillofacial and Oral Surgery, University Medical Center, 55131 Mainz, Germany; Oral Pathology Department, Faculty of Dentistry, Alexandria University, 5372066 Alexandria, Egypt
| | - Hafez Am
- Medical Biochemistry Department Faculty of medicine KafrElSheikh University, Kafr El-Sheikh, Egypt
| | | | - Désirée Gül
- Department of Otorhinolaryngology Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany.
| |
Collapse
|
26
|
Wicher G, Roy A, Vaccaro A, Vemuri K, Ramachandran M, Olofsson T, Imbria RN, Belting M, Nilsson G, Dimberg A, Forsberg-Nilsson K. Lack of ST2 aggravates glioma invasiveness, vascular abnormality, and immune suppression. Neurooncol Adv 2025; 7:vdaf010. [PMID: 39931535 PMCID: PMC11808570 DOI: 10.1093/noajnl/vdaf010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Background Glioblastoma (GBM) is the most common primary malignant brain tumor in adults, characterized by aggressive growth and a dismal prognosis. Interleukin-33 (IL-33) and its receptor ST2 have emerged as regulators of glioma growth, but their exact function in tumorigenesis has not been deciphered. Indeed, previous studies on IL-33 in cancer have yielded somewhat opposing results as to whether it is pro- or anti-tumorigenic. Methods IL-33 expression was assessed in a GBM tissue microarray and public databases. As in vivo models we used orthotopic xenografts of patient-derived GBM cells, and syngenic models with grafted mouse glioma cells. Results We analyzed the role of IL-33 and its receptor ST2 in nonmalignant cells of the glioma microenvironment and found that IL-33 levels are increased in cells surrounding the tumor. Protein complexes of IL-33 and ST2 are mainly found outside of the tumor core. The IL-33-producing cells consist primarily of oligodendrocytes. To determine the function of IL-33 in the tumor microenvironment, we used mice lacking the ST2 receptor. When glioma cells were grafted to ST2-deficient mouse brains, the resulting tumors exhibited a more invasive growth pattern, and are associated with poorer survival, compared to wild-type mice. Tumors in ST2-deficient hosts are more invasive, with increased expression of extracellular matrix remodeling enzymes and enhanced tumor angiogenesis. Furthermore, the absence of ST2 leads to a more immunosuppressive environment. Conclusions Our findings reveal that glia-derived IL-33 and its receptor ST2 participate in modulating tumor invasiveness, tumor vasculature, and immunosuppression in glioma.
Collapse
Affiliation(s)
- Grzegorz Wicher
- Science for Life Laboratory, Uppsala University, Sweden
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Ananya Roy
- Science for Life Laboratory, Uppsala University, Sweden
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Alessandra Vaccaro
- Science for Life Laboratory, Uppsala University, Sweden
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Kalyani Vemuri
- Science for Life Laboratory, Uppsala University, Sweden
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Mohanraj Ramachandran
- Science for Life Laboratory, Uppsala University, Sweden
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Tommie Olofsson
- Academic Laboratory, Uppsala University Hospital, Uppsala, Sweden
| | - Rebeca-Noemi Imbria
- Science for Life Laboratory, Uppsala University, Sweden
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Mattias Belting
- Department of Hematology, Oncology and Radiophysics, Skåne University Hospital, Lund, Sweden
- Department of Clinical Sciences, Section of Oncology, Lund University, Lund, Sweden
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Gunnar Nilsson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, and Centre for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Anna Dimberg
- Science for Life Laboratory, Uppsala University, Sweden
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Karin Forsberg-Nilsson
- Division of Cancer and Stem Cells, University of Nottingham Biodiscovery Institute, Nottingham, UK
- Science for Life Laboratory, Uppsala University, Sweden
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
27
|
DU N, Wan H, Guo H, Zhang X, Wu X. [Myeloid-derived suppressor cells as important factors and potential targets for breast cancer progression]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024; 53:785-795. [PMID: 39686697 PMCID: PMC11736353 DOI: 10.3724/zdxbyxb-2024-0353] [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: 07/31/2024] [Accepted: 10/28/2024] [Indexed: 12/18/2024]
Abstract
Recurrence and metastasis remain the leading cause of death in breast cancer patients due to the lack of effective treatment. A microenvironment suitable for cancer cell growth, referred to as pre-metastatic niche (PMN), is formed in distant organs before metastasis occurs. Myeloid-derived suppressor cells (MDSCs) are a heterogenous population of immature myeloid cells with immunosuppressive effects. They can expand in large numbers in breast cancer patients and participate in the formation of PMN. MDSCs can remodel the extracellular matrix of pulmonary vascular endothelial cells and recruit cancer stem cells to promote the lung metastasis of breast cancer. Furthermore, MDSCs facilitate immune evasion of breast cancer cells to impact the efficacy of immunotherapy. It is proposed that MDSCs represent a potential therapeutic target for the inhibition of recurrence and metastasis in breast cancer. Therapeutic strategies targeting MDSCs have shown promising efficacy in preclinical studies and clinical trials. This review presents a summary of the principal factors involved in the recruitment and activation of MDSCs during the formation of PMN, and outlines MDSCs functions such as immunosuppression and the current targeted therapies against MDSCs, aiming to provide new ideas for the treatment of distant metastases in breast cancer.
Collapse
Affiliation(s)
- Nannan DU
- Breast Department, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China.
| | - Hua Wan
- Breast Department, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - Hailing Guo
- Department of Orthopaedics and Traumatology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - Xukuan Zhang
- Breast Department, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - Xueqing Wu
- Breast Department, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China.
| |
Collapse
|
28
|
Reslane I, Watson GF, Handke LD, Fey PD. Regulatory dynamics of arginine metabolism in Staphylococcus aureus. Biochem Soc Trans 2024; 52:2513-2523. [PMID: 39656074 PMCID: PMC11668279 DOI: 10.1042/bst20240710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 12/20/2024]
Abstract
Staphylococcus aureus is a highly significant pathogen with several well studied and defined virulence factors. However, the metabolic pathways that are required to facilitate infection are not well described. Previous data have documented that S. aureus requires glucose catabolism during initial stages of infection. Therefore, certain nutrients whose biosynthetic pathway is under carbon catabolite repression and CcpA, including arginine, must be acquired from the host. However, even though S. aureus encodes pathways to synthesize arginine, biosynthesis of arginine is repressed even in the absence of glucose. Why is S. aureus a functional arginine auxotroph? This review discusses recently described regulatory mechanisms that are linked to repression of arginine biosynthesis using either proline or glutamate as substrates. In addition, recent studies are discussed that shed insight into the ultimate mechanisms linking arginine auxotrophy and infection persistence.
Collapse
Affiliation(s)
- Itidal Reslane
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, U.S.A
| | - Gabrielle F. Watson
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, U.S.A
| | - Luke D. Handke
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, U.S.A
| | - Paul D. Fey
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, U.S.A
| |
Collapse
|
29
|
Wang X, Wang X, Wang D, Zhou C, Lv K, Ma Y, Chang W, Wang B, Hu J, Ji Y, Dai Z, Ma Y. Interleukin-10 overexpression in 4T1 cells: A gateway to suppressing mammary carcinoma growth. Int Immunopharmacol 2024; 142:113089. [PMID: 39244897 DOI: 10.1016/j.intimp.2024.113089] [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: 04/09/2024] [Revised: 08/04/2024] [Accepted: 09/02/2024] [Indexed: 09/10/2024]
Abstract
Interleukin-10 (IL-10) exerts complex effects on tumor growth, exhibiting both pro- and anti-tumor properties. Recent focus on the anti-inflammatory properties of IL-10 has highlighted its potential anti-tumor properties, particularly through the enhancement of CD8+ T cell activity. However, further research is needed to fully elucidate its other anti-tumor mechanisms. Our study investigates novel anti-tumor mechanisms of IL-10 in a murine mammary carcinoma model (4T1). We found that IL-10 overexpression in mouse 4T1 cells suppressed tumor growth in vivo. This suppression was accompanied by an increase in IFN-γ-secreting CD8+ T cells and a decrease in myeloid-derived suppressor cells (MDSCs) in tumor tissue. In vitro experiments showed that IL-10-rich tumor cell-derived supernatants inhibited myeloid cell differentiation into monocytic and granulocytic MDSCs while reducing MDSCs migration. In addition, IL-10 overexpression downregulated CXCL5 expression in 4T1 cells, resulting in decreased CXCR2+ MDSCs infiltration. Using RAG1-deficient mice and CXCL5 knockdown tumor models, we demonstrated that the anti-tumor effects of IL-10 depend on both CD8+ T cells and reduced MDSC infiltration. IL-10 attenuated the immunosuppressive tumor microenvironment by enhancing CD8+ T cell activity and inhibiting MDSCs infiltration. In human breast cancer, we observed a positive correlation between CXCL5 expression and MDSC infiltration. Our findings reveal a dual mechanism of IL-10-mediated tumor suppression: (1) direct enhancement of CD8+ T cell activity and (2) indirect reduction of immunosuppressive MDSCs through CXCL5 downregulation and inhibition of myeloid cell differentiation. This study provides new insights into the role of IL-10 in anti-tumor immunity and suggests potential strategies for breast cancer immunotherapy by modulating the IL-10-CXCL5-MDSCs axis.
Collapse
Affiliation(s)
- Xiaoqin Wang
- The Clinical Laboratory, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiaoqian Wang
- The Clinical Laboratory, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Dan Wang
- The Clinical Laboratory, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Can Zhou
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Kaige Lv
- Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yanfen Ma
- The Clinical Laboratory, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Wenjing Chang
- The Clinical Laboratory, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Baofeng Wang
- Department of Radiotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jian Hu
- The Clinical Laboratory, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Yanhong Ji
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Zhijun Dai
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China.
| | - Yunfeng Ma
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China.
| |
Collapse
|
30
|
Rajkumari S, Singh J, Agrawal U, Agrawal S. Myeloid-derived suppressor cells in cancer: Current knowledge and future perspectives. Int Immunopharmacol 2024; 142:112949. [PMID: 39236460 DOI: 10.1016/j.intimp.2024.112949] [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: 05/30/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024]
Abstract
MDSCs (myeloid-derived suppressor cells) are crucial for immune system evasion in cancer. They accumulate in peripheral blood and tumor microenvironment, suppressing immune cells like T-cells, natural killer cells and dendritic cells. They promote tumor angiogenesis and metastasis by secreting cytokines and growth factors and contribute to a tumor-promoting environment. The accumulation of MDSCs in cancer patients has been linked to poor prognosis and resistance to various cancer therapies. Targeting MDSCs and their immunosuppressive mechanisms may improve treatment outcomes and enhance immune surveillance by developing drugs that inhibit MDSC function, by preventing their accumulation and by disrupting the tumor-promoting environment. This review presents a detailed overview of the MDSC research in cancer with regulation of their development and function. The relevance of MDSC as a prognostic and predictive biomarker in different types of cancers, along with recent advancements on the therapeutic approaches to target MDSCs are discussed in detail.
Collapse
Affiliation(s)
- Sunanda Rajkumari
- ICMR National Institute of Medical Statistics, Ansari Nagar, New Delhi 110029, India
| | - Jaspreet Singh
- ICMR National Institute of Pathology, Safdarjung Hospital Campus, Ansari Nagar, New Delhi 110029, India
| | - Usha Agrawal
- Asian Institute of Public Health University (AIPH) University, 1001 Haridamada, Jatani, Near IIT Bhubaneswar, Bhubaneswar 751002, India
| | - Sandeep Agrawal
- Discovery Research Division, Indian Council of Medical Research, Ansari Nagar, New Delhi 110029, India.
| |
Collapse
|
31
|
Yu K, Wang Y, Yu C, Han L, Li K, Miao K, Ni L, Wen Z, Chen C, Rao X, Wang DW, Zhou L, Zhao C. Regulatory effect of rapamycin on recruitment and function of myeloid-derived suppressor cells in heart failure. Int Immunopharmacol 2024; 141:112965. [PMID: 39186836 DOI: 10.1016/j.intimp.2024.112965] [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: 04/28/2024] [Revised: 07/30/2024] [Accepted: 08/15/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND Immune response and inflammation play important roles in the physiological and pathophysiological processes of heart failure (HF). In our previous study, myeloid-derived suppressor cells (MDSCs), a heterogeneous group of immature myeloid cells with anti-inflammatory and immunosuppressive functions, were shown to exert cardioprotective effects in HF. The pharmacological targeting of MDSCs using rapamycin may emerge as a promising strategy for the prevention and treatment of HF. However, the specific mechanisms underlying rapamycin-induced MDSC accumulation remain unclear. Our study aimed to clarify the effects of rapamycin on the recruitment and function of MDSCs in HF, exploring new therapeutic options for the prevention and treatment of HF. METHODS We used transverse aortic constriction surgery and isoproterenol injection to establish HF models. Flow cytometry, reverse transcription polymerase chain reaction, transcriptomics and western blot were used to explore the regulation of rapamycin on recruitment and function of MDSCs in HF. Furthermore, rapamycin and granulocyte-macrophage colony-stimulating factor (GM-CSF) were combined to induce exogenous MDSCs from bone marrow cells. RESULTS Rapamycin promotes the recruitment of MDSCs by inhibiting their maturation and differentiation via suppression of the Wnt signaling in HF mice and enhanced the immunosuppressive function of MDSCs via the NF-κB signaling. Furthermore, exogenous MDSCs induced by rapamycin and GM-CSF can significantly alleviate transverse aortic constriction-induced cardiac dysfunction. CONCLUSIONS The pharmacological targeting of MDSCs using rapamycin is a promising strategy for the prevention and treatment of HF.
Collapse
Affiliation(s)
- Kun Yu
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yinhui Wang
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chengxin Yu
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Liang Han
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, TongJi Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ke Li
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Kun Miao
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Li Ni
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zheng Wen
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chen Chen
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaoquan Rao
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dao Wen Wang
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ling Zhou
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Chunxia Zhao
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
32
|
Leporati M, Di Genaro MS, Eliçabe RJ. Nitric oxide-producing monocyte-myeloid suppressor cells expand and accumulate in the spleen and mesenteric lymph nodes of Yersinia enterocolitica-infected mice. Front Cell Infect Microbiol 2024; 14:1440514. [PMID: 39529636 PMCID: PMC11551114 DOI: 10.3389/fcimb.2024.1440514] [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: 05/29/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction Yersinia enterocolitica (Ye) is a Gram-negative bacterium that causes gastrointestinal infections. The myeloid-derived suppressor cells (MDSCs) constitute a cellular population with the capacity of inducing the specific suppression of T cells. Although there is evidence supporting the role of MDSCs in controlling the immune responses in several bacterial infections, its role during Ye infection has not yet been reported. Therefore, the purpose of the present work was to analyze MDSCs after oral Ye infection. Methods C57BL/6 wild-type mice were infected with Ye WAP-314 serotype O:8. The proliferation of splenocytes and mesenteric lymph nodes (MLN) cells was measured as well as the levels of cytokines and nitric oxide (NO) in culture supernatants. The frequency and subsets of MDSCs were analyzed in the intestinal mucosa and spleen by flow cytometry. Furthermore, monocytic-MDSCs (Mo-MDSCs) and polymorphonuclear-MDSCs (PMN-MDSCs) were purified from the spleen of infected mice and their suppressor activity was evaluated in co-cultures with purified T cells. Results we observed a marked expansion of CD11b+Gr-1+ cells, a phenotype consistent with MDSCs, in the spleen and intestinal mucosa of Ye-infected mice. Interestingly, a robust proliferation of splenocytes and MLN cells was observed only when the MDSCs were depleted or the NO production was blocked. In addition, we determined that only Mo-MDSCs had the ability to suppress T-cell proliferation. Conclusion Our results highlight a mechanism by which Ye may induce suppression of the immune responses. We suggest that NO-producing Mo-MDSCs expand and accumulate in MLN and spleen of Ye-infected mice. These cells can then suppress the T-cell function without interfering with the anti-bacterial effector response. Instead, these immature myeloid cells may perform an important function in regulating the inflammatory response and protecting affected tissues.
Collapse
Affiliation(s)
- Marianela Leporati
- División de Inmunología, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina
- Laboratorio de Inmunopatología, Instituto Multidisciplinario de Investigaciones Biológicas San Luis (IMIBIO-SL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Luis, Argentina
| | - María Silvia Di Genaro
- División de Inmunología, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina
- Laboratorio de Inmunopatología, Instituto Multidisciplinario de Investigaciones Biológicas San Luis (IMIBIO-SL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Luis, Argentina
| | - Ricardo Javier Eliçabe
- División de Inmunología, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina
- Laboratorio de Inmunopatología, Instituto Multidisciplinario de Investigaciones Biológicas San Luis (IMIBIO-SL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Luis, Argentina
| |
Collapse
|
33
|
Ni X, Wei Y, Li X, Pan J, Fang B, Zhang T, Lu Y, Ye D, Zhu Y. From biology to the clinic - exploring liver metastasis in prostate cancer. Nat Rev Urol 2024; 21:593-614. [PMID: 38671281 DOI: 10.1038/s41585-024-00875-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2024] [Indexed: 04/28/2024]
Abstract
Liver metastases from prostate cancer are associated with an aggressive disease course and poor prognosis. Results from autopsy studies indicate a liver metastasis prevalence of up to 25% in patients with advanced prostate cancer. Population data estimate that ~3-10% of patients with metastatic castration-resistant prostate cancer harbour liver metastases at the baseline, rising to 20-30% in post-treatment cohorts, suggesting that selective pressure imposed by novel therapies might promote metastatic spread to the liver. Liver metastases are associated with more aggressive tumour biology than lung metastases. Molecular profiling of liver lesions showed an enrichment of low androgen receptor, neuroendocrine phenotypes and high genomic instability. Despite advancements in molecular imaging modalities such as prostate-specific membrane antigen PET-CT, and liquid biopsy markers such as circulating tumour DNA, early detection of liver metastases from prostate cancer remains challenging, as both approaches are hampered by false positive and false negative results, impeding the accurate identification of early liver lesions. Current therapeutic strategies showed limited efficacy in this patient population. Emerging targeted radionuclide therapies, metastasis-directed therapy, and novel systemic agents have shown preliminary activity against liver metastases, but require further validation. Treatment with various novel prostate cancer therapies might lead to an increase in the prevalence of liver metastasis, underscoring the urgent need for coordinated efforts across preclinical and clinical researchers to improve characterization, monitoring, and management of liver metastases from prostate cancer. Elucidating molecular drivers of liver tropism and interactions with the liver microenvironment might ultimately help to identify actionable targets to enhance survival in this high-risk patient group.
Collapse
Affiliation(s)
- Xudong Ni
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Yu Wei
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Xiaomeng Li
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Jian Pan
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Bangwei Fang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Tingwei Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Ying Lu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Yao Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Genitourinary Cancer Institute, Shanghai, China.
| |
Collapse
|
34
|
Nair JJ, van Staden J. Anti-inflammatory Principles of the Plant Family Amaryllidaceae. PLANTA MEDICA 2024; 90:900-937. [PMID: 39029914 DOI: 10.1055/a-2369-8104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Abstract
There is considerable interest in the utilisation of plants against inflammation. Over 50 species of the plant family Amaryllidaceae are known for such usage in traditional medicine. This review was undertaken to identify the chemical principles responsible for these anti-inflammatory effects. It describes the findings from in vitro, in vivo and in silico studies, as well as the probes made on the mechanisms of action. The literature search returned over 600 hits, of which around 130 were chosen for their relevance to the text. Over 140 compounds have thus far been screened for anti-inflammatory effects. These were mostly isoquinoline alkaloids but also included other classes of secondary metabolites such as chromones, flavonoids and triterpenoids. In vitro studies were carried out in mononuclear cells such as lymphocytes, monocytes, neutrophils and macrophages, against which no serious side effects were observed. The constituents were also effective against inflammation induced by physical and chemical stimuli in a variety of murine test subjects. Chief among the compounds were the isoquinoline alkaloids lycorine and narciclasine, which displayed potent effects against pain, swelling, asthma and arthritis, amongst others. From a mechanistic perspective, several of the compounds were shown to mediate in inflammatory pathways, notably via the modulation of both pro-inflammatory (such as NF-κB, TNF-α and IL-1) and anti-inflammatory (such as IL-10 and TGF-β) factors. Useful insights also emerged from active-site docking studies of some of the compounds. The Amaryllidaceae affords a rich and diverse platform for the discovery of potential anti-inflammatory drugs.
Collapse
Affiliation(s)
- Jerald J Nair
- Research Centre for Plant Growth and Development, School of Life Sciences, University of KwaZulu-Natal, Pietermaritzburg, South Africa
| | - Johannes van Staden
- Research Centre for Plant Growth and Development, School of Life Sciences, University of KwaZulu-Natal, Pietermaritzburg, South Africa
| |
Collapse
|
35
|
Angell CD, Lapurga G, Sun SH, Johnson C, Savardekar H, Rampersaud IV, Fletcher C, Albertson D, Ren C, Suarez-Kelly LP, Rampersaud AA, Carson WE. Targeting Myeloid-Derived Suppressor Cells via Dual-Antibody Fluorescent Nanodiamond Conjugate. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1509. [PMID: 39330666 PMCID: PMC11434946 DOI: 10.3390/nano14181509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/10/2024] [Accepted: 09/12/2024] [Indexed: 09/28/2024]
Abstract
Fluorescent nanodiamonds (FNDs) are carbon-based nanomaterials that emit bright, photostable fluorescence and exhibit a modifiable surface chemistry. Myeloid-derived suppressor cells (MDSCs) are an immunosuppressive cell population known to expand in cancer patients and contribute to worse patient outcomes. To target MDSC, glycidol-coated FND were conjugated with antibodies against the murine MDSC markers, CD11b and GR1 (dual-Ab FND). In vitro, dual-Ab FND uptake by murine MDSC was significantly higher than IgG-coated FND (94.7% vs. 69.0%, p < 0.05). In vivo, intra-tumorally injected dual-Ab FND primarily localized to the tumor 2 and 24 h post-injection, as measured by in vivo fluorescence imaging and flow cytometry analysis of the spleen and tumor. Dual-Ab FND were preferentially taken up by intra-tumoral MDSC, representing 87.1% and 83.0% of FND+ cells in the tumor 2 and 24 h post-injection, respectively. Treatment of mice with anti-PD-L1 immunotherapy prior to intra-tumoral injection of dual-Ab FND did not significantly alter the uptake of FND by MDSC. These results demonstrate the ability of our novel dual-antibody conjugated FND to target MDSC and reveal a potential strategy for targeted delivery to other specific immune cell populations in future cancer research.
Collapse
Affiliation(s)
- Colin D Angell
- The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Gabriella Lapurga
- The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Steven H Sun
- The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
- Department of Surgery, The Ohio State University, Columbus, OH 43210, USA
| | - Courtney Johnson
- The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Himanshu Savardekar
- The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | | | - Charles Fletcher
- Columbus NanoWorks, Inc., 1507 Chambers Road, Columbus, OH 43212, USA
| | - David Albertson
- Columbus NanoWorks, Inc., 1507 Chambers Road, Columbus, OH 43212, USA
| | - Casey Ren
- The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | | | | | - William E Carson
- Department of Surgery, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
36
|
Karampitsakos T, Tourki B, Jia M, Perrot CY, Visinescu B, Zhao A, Unterman A, Tzouvelekis A, Bandyopadhyay D, Juan-Guardela BM, Prasse A, Noth I, Liggett S, Kaminski N, Benos PV, Herazo-Maya JD. The transcriptome of CD14 + CD163 - HLA-DR low monocytes predicts mortality in Idiopathic Pulmonary Fibrosis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.07.24311386. [PMID: 39211854 PMCID: PMC11361223 DOI: 10.1101/2024.08.07.24311386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Rationale The association between immune-cell-specific transcriptomic profiles and Idiopathic Pulmonary Fibrosis (IPF) mortality is unknown. Objectives To determine immune-cell-specific transcriptomic profiles associated with IPF mortality. Methods We profiled peripheral blood mononuclear cells (PBMC) in 18 participants [University of South Florida: IPF, COVID-19, post-COVID-19 Interstitial Lung Disease (Post-COVID-19 ILD), controls] by single-cell RNA sequencing (scRNA-seq) and identified 16 immune-cell-specific transcriptomic profiles. The Scoring Algorithm of Molecular Subphenotypes (SAMS) was used to calculate Up-scores based on these 16 gene profiles. Their association with outcomes was investigated in peripheral blood, Bronchoalveolar Lavage (BAL) and lung tissue of N=416 IPF patients from six cohorts. Findings were validated in an independent IPF, PBMC scRNA-seq dataset (N=38). Measurements and main results Cox-regression models demonstrated that 230 genes from CD14 + CD163 - HLA-DR low circulating monocytes predicted IPF mortality [Pittsburgh (p=0.02), Chicago (p=0.003)]. PBMC proportions of CD14 + CD163 - HLA-DR low monocytes were higher in progressive versus stable IPF (Yale, 0.13±0.05 versus 0.09±0.05, p=0.034). Receiving operating characteristic identified a 230 gene, Up-score >41.84 (Pittsburgh) predictive of mortality in Chicago (HR: 6.58, 95%CI: 2.15-20.13, p=0.001) and in pooled analysis of BAL cohorts (HR: 2.20, 95%CI: 1.44-3.37, p=0.0003). High-risk patients had decreased expression of the T-cell co-stimulatory genes CD28 , ICOS , ITK and LCK (Pittsburgh and Chicago, p<0.01). 230 gene-up-scores negatively correlated with Forced Vital Capacity (FVC) in IPF lung tissues (LGRC, rho=-0.2, p=0.02). Results were replicated using a subset of 13 genes from the 230-gene signature (pooled PBMC cohorts - HR: 5.34, 95%CI: 2.83-10.06, p<0.0001). Conclusions The transcriptome of CD14 + CD163 - HLA-DR low monocytes is associated with increased IPF mortality.
Collapse
|
37
|
Wang R, Li B, Huang B, Li Y, Liu Q, Lyu Z, Chen R, Qian Q, Liang X, Pu X, Wu Y, Chen Y, Miao Q, Wang Q, Lian M, Xiao X, Leung PSC, Gershwin ME, You Z, Ma X, Tang R. Gut Microbiota-Derived Butyrate Induces Epigenetic and Metabolic Reprogramming in Myeloid-Derived Suppressor Cells to Alleviate Primary Biliary Cholangitis. Gastroenterology 2024; 167:733-749.e3. [PMID: 38810839 DOI: 10.1053/j.gastro.2024.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 03/29/2024] [Accepted: 05/08/2024] [Indexed: 05/31/2024]
Abstract
BACKGROUND & AIMS Gut dysbiosis and myeloid-derived suppressor cells (MDSCs) are implicated in primary biliary cholangitis (PBC) pathogenesis. However, it remains unknown whether gut microbiota or their metabolites can modulate MDSCs homeostasis to rectify immune dysregulation in PBC. METHODS We measured fecal short-chain fatty acids levels using targeted gas chromatography-mass spectrometry and analyzed circulating MDSCs using flow cytometry in 2 independent PBC cohorts. Human and murine MDSCs were differentiated in vitro in the presence of butyrate, followed by transcriptomic, epigenetic (CUT&Tag-seq and chromatin immunoprecipitation-quantitative polymerase chain reaction), and metabolic (untargeted liquid chromatography-mass spectrometry, mitochondrial stress test, and isotope tracing) analyses. The in vivo role of butyrate-MDSCs was evaluated in a 2-octynoic acid-bovine serum albumin-induced cholangitis murine model. RESULTS Decreased butyrate levels and defective MDSC function were found in patients with incomplete response to ursodeoxycholic acid, compared with those with adequate response. Butyrate induced expansion and suppressive activity of MDSCs in a manner dependent on PPARD-driven fatty acid β-oxidation (FAO). Pharmaceutical inhibition or genetic knockdown of the FAO rate-limiting gene CPT1A abolished the effect of butyrate. Furthermore, butyrate inhibited HDAC3 function, leading to enhanced acetylation of lysine 27 on histone H3 at promoter regions of PPARD and FAO genes in MDSCs. Therapeutically, butyrate administration alleviated immune-mediated cholangitis in mice via MDSCs, and adoptive transfer of butyrate-treated MDSCs also displayed protective efficacy. Importantly, reduced expression of FAO genes and impaired mitochondrial physiology were detected in MDSCs from ursodeoxycholic acid nonresponders, and their impaired suppressive function was restored by butyrate. CONCLUSIONS We identify a critical role for butyrate in modulation of MDSC homeostasis by orchestrating epigenetic and metabolic crosstalk, proposing a novel therapeutic strategy for treating PBC.
Collapse
Affiliation(s)
- Rui Wang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Digestive Disease, Shanghai, China
| | - Bo Li
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Digestive Disease, Shanghai, China
| | - Bingyuan Huang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Digestive Disease, Shanghai, China
| | - Yikang Li
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Digestive Disease, Shanghai, China
| | - Qiaoyan Liu
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Digestive Disease, Shanghai, China
| | - Zhuwan Lyu
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Digestive Disease, Shanghai, China
| | - Ruiling Chen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Digestive Disease, Shanghai, China
| | - Qiwei Qian
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Digestive Disease, Shanghai, China
| | - Xueying Liang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Digestive Disease, Shanghai, China
| | - Xiting Pu
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Digestive Disease, Shanghai, China
| | - Yi Wu
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Digestive Disease, Shanghai, China
| | - Yu Chen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Digestive Disease, Shanghai, China
| | - Qi Miao
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Digestive Disease, Shanghai, China
| | - Qixia Wang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Digestive Disease, Shanghai, China; Division of Infectious Diseases, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Lian
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Digestive Disease, Shanghai, China
| | - Xiao Xiao
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Digestive Disease, Shanghai, China
| | - Patrick S C Leung
- Division of Rheumatology, Department of Medicine, Allergy and Clinical Immunology, University of California at Davis, Davis, California
| | - M Eric Gershwin
- Division of Rheumatology, Department of Medicine, Allergy and Clinical Immunology, University of California at Davis, Davis, California
| | - Zhengrui You
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Digestive Disease, Shanghai, China.
| | - Xiong Ma
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Digestive Disease, Shanghai, China; Institute of Aging & Tissue Regeneration, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Ruqi Tang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Digestive Disease, Shanghai, China.
| |
Collapse
|
38
|
Che J, Song J, Long Y, Wang C, Zheng C, Zhou R, Liu Z. Association Between the Neutrophil-Lymphocyte Ratio and Prognosis of Patients Admitted to the Intensive Care Unit With Chronic Heart Failure: A Retrospective Cohort Study. Angiology 2024; 75:786-795. [PMID: 37586709 DOI: 10.1177/00033197231196174] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
The present study aimed to explore the association between the neutrophil-to-lymphocyte ratio (NLR) and prognosis of critically ill chronic heart failure patients. The records of 5298 patients who met the inclusion criteria were extracted from the Medical Information Mart for Intensive Care IV database. The primary outcome was 30-days all-cause mortality and the secondary outcome was 90-days all-cause mortality. Multivariable logistic regression analysis was performed to examine the relationship between NLR and 30-days mortality. Subgroup analysis was carried out to identify whether the association between NLR and 30-days mortality differed across various subgroups. For 30-days mortality, after adjusting for multiple confounders, the odds ratio (OR) (95% confidence interval [CI]) for the second (NLR 4.0-8.4) and the third (NLR ≥8.4) tertiles were 1.52 (1.13-2.03) and 2.53 (1.92-3.34), respectively, compared with the first tertile (NLR <4.0). As for 90-days mortality, the OR for the second (NLR 4.0-8.4) was 1.34 (1.07-1.67) and 2.23 (1.81-2.76) for the third (NLR ≥8.4) tertiles compared with the reference (NLR<4.0). The interactions between the sepsis subgroup and 30-days mortality were significant. Our study concluded that the NLR was an independent predictor of 30- and 90-days mortality for critically ill patients with chronic heart failure.
Collapse
Affiliation(s)
- Jinhang Che
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiaqi Song
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuxiang Long
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chunping Wang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Caiyin Zheng
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ruoyu Zhou
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zengzhang Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
39
|
Takacs GP, Garcia JS, Hodges CA, Kreiger CJ, Sherman A, Harrison JK. CSF1R Ligands Expressed by Murine Gliomas Promote M-MDSCs to Suppress CD8 + T Cells in a NOS-Dependent Manner. Cancers (Basel) 2024; 16:3055. [PMID: 39272914 PMCID: PMC11394022 DOI: 10.3390/cancers16173055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/27/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Glioblastoma (GBM) is the most common malignant primary brain tumor, resulting in poor survival despite aggressive therapies. GBM is characterized by a highly heterogeneous and immunosuppressive tumor microenvironment (TME) made up predominantly of infiltrating peripheral immune cells. One significant immune cell type that contributes to glioma immune evasion is a population of immunosuppressive cells, termed myeloid-derived suppressor cells (MDSCs). Previous studies suggest that a subset of myeloid cells, expressing monocytic (M)-MDSC markers and dual expression of chemokine receptors CCR2 and CX3CR1, utilize CCR2 to infiltrate the TME. This study evaluated the mechanism of CCR2+/CX3CR1+ M-MDSC differentiation and T cell suppressive function in murine glioma models. We determined that bone marrow-derived CCR2+/CX3CR1+ cells adopt an immune suppressive cell phenotype when cultured with glioma-derived factors. Glioma-secreted CSF1R ligands M-CSF and IL-34 were identified as key drivers of M-MDSC differentiation while adenosine and iNOS pathways were implicated in the M-MDSC suppression of T cells. Mining a human GBM spatial RNAseq database revealed a variety of different pathways that M-MDSCs utilize to exert their suppressive function that is driven by complex niches within the microenvironment. These data provide a more comprehensive understanding of the mechanism of M-MDSCs in glioblastoma.
Collapse
Affiliation(s)
| | | | | | | | | | - Jeffrey K. Harrison
- Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610, USA (J.S.G.); (C.A.H.)
| |
Collapse
|
40
|
Zeng W, Liu H, Mao Y, Jiang S, Yi H, Zhang Z, Wang M, Zong Z. Myeloid‑derived suppressor cells: Key immunosuppressive regulators and therapeutic targets in colorectal cancer (Review). Int J Oncol 2024; 65:85. [PMID: 39054950 PMCID: PMC11299769 DOI: 10.3892/ijo.2024.5673] [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: 02/19/2024] [Accepted: 06/03/2024] [Indexed: 07/27/2024] Open
Abstract
Globally, colorectal cancer (CRC) is the third most common type of cancer. CRC has no apparent symptoms in the early stages of disease, and most patients receive a confirmed diagnosis in the middle or late disease stages. The incidence of CRC continues to increase, and the affected population tends to be younger. Therefore, determining how to achieve an early CRC diagnosis and treatment has become a top priority for prolonging patient survival. Myeloid‑derived suppressor cells (MDSCs) are a group of bone marrow‑derived immuno‑negative regulatory cells that are divided into two subpopulations, polymorphonuclear‑MDSCs and monocytic‑MDSCs, based on their phenotypic similarities to neutrophils and monocytes, respectively. These cells can inhibit the immune response and promote cancer cell metastasis in the tumour microenvironment (TME). A large aggregation of MDSCs in the TME is often a marker of cancer and a poor prognosis in inflammatory diseases of the intestine (such as colonic adenoma and ulcerative colitis). In the present review, the phenotypic classification of MDSCs in the CRC microenvironment are first discussed. Then, the amplification, role and metastatic mechanism of MDSCs in the CRC TME are described, focusing on genes, gene modifications, proteins and the intestinal microenvironment. Finally, the progress in CRC‑targeted therapies that aim to modulate the quantity, function and structure of MDSCs are summarized in the hope of identifying potential screening markers for CRC and improving CRC prognosis and therapeutic options.
Collapse
Affiliation(s)
- Wenjuan Zeng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- HuanKui Academy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Haohan Liu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yuanhao Mao
- Fuzhou Medical College, Nanchang University, Fuzhou, Jiangxi 330006, P.R. China
| | - Shihao Jiang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Hao Yi
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zitong Zhang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- HuanKui Academy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Menghui Wang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- HuanKui Academy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhen Zong
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
41
|
Li X, Chen Y, Liang Y, Shi W. 5-Fluorouracil resistance due to sphingosine kinase 2 overexpression in colorectal cancer is associated with myeloid-derived suppressor cell-mediated immunosuppressive effects. BMC Cancer 2024; 24:983. [PMID: 39118083 PMCID: PMC11313101 DOI: 10.1186/s12885-024-12742-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024] Open
Abstract
PURPOSE Colorectal cancer (CRC) is one of the top five cancer-related causes of mortality globally. Acquired resistance has hindered the effectiveness of 5-fluorouracil (5-FU), the main chemotherapeutic drug used to treat CRC. Sphingosine kinase 2 (SphK2) may be a cancer treatment target and involved in 5-FU resistance. METHODS Cell growth was examined using MTT and clone formation assays for SphK2 expression. To identify immune cells in mice, flow cytometry was performed. West blotting demonstrated alterations in cell division and inflammation-related proteins. SphK2 levels and inflammation-related variables were studied using Elisa. RESULTS Due to SphK2 overexpression, immunosuppression, and 5-FU resistance are caused by the development of myeloid-derived suppressor cells (MDSCs) subsequent to IL-6/STAT3 activation and alterations in the arginase (ARG-1) protein. After therapy, the combination of SphK2 inhibitors and 5-FU can effectively suppress MDSCs while increasing CD4+ and CD8+ T cell infiltration into the tumor microenvironment, lowering tumor burden, and exhibiting a therapeutic impact on CRC. CONCLUSIONS Our findings suggest that 5-FU treatment combined with simultaneous Spkh2 inhibition by ABC294640 has anti-tumor synergistic effects by influencing multiple effects on tumor cells, T cells, and MDSCs, potentially improving the poor prognosis of colorectal cancer patients.
Collapse
Affiliation(s)
- Xiuyun Li
- Maternal and Child Health Development Research Center, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, China
| | - Yungao Chen
- Human Resources Department, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yulin Liang
- School of Nursing, Peking Union Medical College, Beijing, China
| | - Wenna Shi
- Department of Pharmacy and Shandong Provincial key Traditional Chinese Medical Discipline of Clinical Chinese pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
42
|
Luo M, He N, Xu Q, Wen Z, Wang Z, Zhao J, Liu Y. Roles of prostaglandins in immunosuppression. Clin Immunol 2024; 265:110298. [PMID: 38909972 DOI: 10.1016/j.clim.2024.110298] [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: 05/07/2024] [Revised: 06/07/2024] [Accepted: 06/21/2024] [Indexed: 06/25/2024]
Abstract
Prostaglandins (PGs) play a crucial and multifaceted role in various physiological processes such as intercellular signaling, inflammation regulation, neurotransmission, vasodilation, vasoconstriction, and reproductive functions. The diversity and biological significance of these effects are contingent upon the specific types or subtypes of PGs, with each PG playing a crucial role in distinct physiological and pathological processes. Particularly within the immune system, PGs are essential in modulating the function of immune cells and the magnitude and orientation of immune responses. Hence, a comprehensive comprehension of the functions PG signaling pathways in immunosuppressive regulation holds substantial clinical relevance for disease prevention and treatment strategies. The manuscript provides a review of recent developments in PG signaling in immunosuppressive regulation. Furthermore, the potential clinical applications of PGs in immunosuppression are also discussed. While research into the immunosuppressive effects of PGs required further exploration, targeted therapies against their immunosuppressive pathways might open new avenues for disease prevention and treatment.
Collapse
Affiliation(s)
- Minjie Luo
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha 410008, Hunan, China; National Medicine Functional Experimental Teaching Center, Changsha 410008, Hunan, China
| | - Nina He
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha 410008, Hunan, China; National Medicine Functional Experimental Teaching Center, Changsha 410008, Hunan, China
| | - Qing Xu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha 410008, Hunan, China; National Medicine Functional Experimental Teaching Center, Changsha 410008, Hunan, China
| | - Zhongchi Wen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha 410008, Hunan, China; National Medicine Functional Experimental Teaching Center, Changsha 410008, Hunan, China
| | - Ziqin Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha 410008, Hunan, China; National Medicine Functional Experimental Teaching Center, Changsha 410008, Hunan, China
| | - Jie Zhao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha 410008, Hunan, China; National Medicine Functional Experimental Teaching Center, Changsha 410008, Hunan, China.
| | - Ying Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha 410008, Hunan, China; National Medicine Functional Experimental Teaching Center, Changsha 410008, Hunan, China.
| |
Collapse
|
43
|
Zhou Z, Lei J, Fang J, Chen P, Zhou J, Wang H, Sun Z, Chen Y, Yin L. Dihydroartemisinin remodels tumor micro-environment and improves cancer immunotherapy through inhibiting cyclin-dependent kinases. Int Immunopharmacol 2024; 139:112637. [PMID: 39033659 DOI: 10.1016/j.intimp.2024.112637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/13/2024] [Accepted: 07/03/2024] [Indexed: 07/23/2024]
Abstract
Cancer immunotherapies are ineffective in nonresponding patients due to absence of immune responses. Here, we identified that dihydroartemisinin (DHA) induced immunogenic cell death (ICD) in hepatocellular carcinoma (HCC), proved by release or surface expose of damage-associated molecular patterns and in vivo protective vaccine activity. Mechanistically, DHA can inhibit cyclin-dependent kinases (CDKs), leading to a buildup of intracellular reactive oxygen species (ROS), which induces immunogenic cell death. In both Hepa1-6 and H22 tumor bearing mice, DHA exerted anti-tumor activity through increasing tumor-infiltrating CD8+ T cells with expression of activation makers (CD25 and CD69), secretion of intracellular cytokines (IFN-γ and TNF-α) and activated dendritic cells expressing MHCⅡ, CD80 and CD86. In hepa1-6 tumor bearing mice, DHA decreased immunosuppressive myeloid-derived suppressor cells. Furthermore, DHA enhanced the anti-PD-1 antibody and chimeric antigen receptor (CAR) T cell-mediated tumor suppression through recruitment and activation of endogenous CD8+ T cells. Overall, we demonstrated that by inhibiting CDKs, DHA can remodel tumor micro-environment to amplify anti-tumor immune responses in HCC. These findings provide a promising therapy option for HCC patients.
Collapse
Affiliation(s)
- Zihao Zhou
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Jun Lei
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China; Department of Laboratory Medicine, Xixi Hospital of Hangzhou, Hangzhou, Zhejiang 310023, China.
| | - Jialing Fang
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Peng Chen
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Jin Zhou
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Hongjian Wang
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Zaiqiao Sun
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Yongshun Chen
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China.
| | - Lei Yin
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology.
| |
Collapse
|
44
|
Zhu Y, Zhou L, Mo L, Hong C, Pan L, Lin J, Qi Y, Tan S, Qian M, Hu T, Zhao Y, Qiu H, Lin P, Ma X, Yang Q. Plasmodium yoelii Infection Enhances the Expansion of Myeloid-Derived Suppressor Cells via JAK/STAT3 Pathway. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:170-186. [PMID: 38819229 DOI: 10.4049/jimmunol.2300541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 05/07/2024] [Indexed: 06/01/2024]
Abstract
Myeloid-derived suppressor cells (MDSCs), the negative immune regulators, have been demonstrated to be involved in immune responses to a variety of pathological conditions, such as tumors, chronic inflammation, and infectious diseases. However, the roles and mechanisms underlying the expansion of MDSCs in malaria remain unclear. In this study, the phenotypic and functional characteristics of splenic MDSCs during Plasmodium yoelii NSM infection are described. Furthermore, we provide compelling evidence that the sera from P. yoelii-infected C57BL/6 mice containing excess IL-6 and granulocyte-macrophage colony-stimulating factor promote the accumulation of MDSCs by inducing Bcl2 expression. Serum-induced MDSCs exert more potent suppressive effects on T cell responses than control MDSCs within both in vivo P. yoelii infection and in vitro serum-treated bone marrow cells experiments. Serum treatment increases the MDSC inhibitory effect, which is dependent on Arg1 expression. Moreover, mechanistic studies reveal that the serum effects are mediated by JAK/STAT3 signaling. By inhibiting STAT3 phosphorylation with the JAK inhibitor JSI-124, effects of serum on MDSCs are almost eliminated. In vivo depletion of MDSCs with anti-Gr-1 or 5-fluorouracil significantly reduces the parasitemia and promotes Th1 immune response in P. yoelii-infected C57BL/6 mice by upregulating IFN-γ expression. In summary, this study indicates that P. yoelii infection facilitates the accumulation and function of MDSCs by upregulating the expression of Bcl2 and Arg1 via JAK/STAT3 signaling pathway in vivo and in vitro. Manipulating the JAK/STAT3 signaling pathway or depleting MDSCs could be promising therapeutic interventions to treat malaria.
Collapse
Affiliation(s)
- Yiqiang Zhu
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, China
| | - Lu Zhou
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Lengshan Mo
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Cansheng Hong
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Lingxia Pan
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jie Lin
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yanwei Qi
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Simin Tan
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Manhongtian Qian
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Tengfei Hu
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yi Zhao
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Huaina Qiu
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Peibin Lin
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Xiancai Ma
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, China
| | - Quan Yang
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Second Affiliated Hospital, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
45
|
Pourbagheri-Sigaroodi A, Momeny M, Rezaei N, Fallah F, Bashash D. Immune landscape of hepatocellular carcinoma: From dysregulation of the immune responses to the potential immunotherapies. Cell Biochem Funct 2024; 42:e4098. [PMID: 39034646 DOI: 10.1002/cbf.4098] [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: 05/25/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 07/23/2024]
Abstract
Hepatocellular carcinoma (HCC) presents a considerable global health burden due to its late diagnosis and high morbidity. The liver's specific anatomical and physiological features expose it to various antigens, requiring precise immune regulation. To the best of our knowledge, this is the first time that a comprehensive overview of the interactions between the immune system and gut microbiota in the development of HCC, as well as the relevant therapeutic approaches are discussed. Dysregulation of immune compartments within the liver microenvironment drives HCC pathogenesis, characterized by elevated regulatory cells such as regulatory T cells (Tregs), myeloid-derived suppressor cells, and M2 macrophages as well as suppressive molecules, alongside reduced number of effector cells like T cells, natural killer cells, and M1 macrophages. Dysbiosis of gut microbiota also contributes to HCC by disrupting intestinal barrier integrity and triggering overactivated immune responses. Immunotherapy approaches, particularly immune checkpoint inhibitors, have exhibited promise in HCC management, yet adoptive cell therapy and cancer vaccination research are in the early steps with relatively less favorable outcomes. Further understanding of immune dysregulation, gut microbiota involvement, and therapeutic combination strategies are essential for advancing precision immunotherapy in HCC.
Collapse
Affiliation(s)
- Atieh Pourbagheri-Sigaroodi
- Pediatric Infections Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Majid Momeny
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Fallah
- Pediatric Infections Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
46
|
Nepal MR, Shah S, Kang KT. Dual roles of myeloid-derived suppressor cells in various diseases: a review. Arch Pharm Res 2024; 47:597-616. [PMID: 39008186 DOI: 10.1007/s12272-024-01504-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 06/30/2024] [Indexed: 07/16/2024]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of cells that originate from bone marrow stem cells. In pathological conditions, such as autoimmune disorders, allergies, infections, and cancer, normal myelopoiesis is altered to facilitate the formation of MDSCs. MDSCs were first shown to promote cancer initiation and progression by immunosuppression with the assistance of various chemokines and cytokines. Recently, various studies have demonstrated that MDSCs play two distinct roles depending on the physiological and pathological conditions. MDSCs have protective roles in autoimmune disorders (such as uveoretinitis, multiple sclerosis, rheumatoid arthritis, ankylosing spondylitis, type 1 diabetes, autoimmune hepatitis, inflammatory bowel disease, alopecia areata, and systemic lupus erythematosus), allergies, and organ transplantation. However, they play negative roles in infections and various cancers. Several immunosuppressive functions and mechanisms of MDSCs have been determined in different disease conditions. This review comprehensively discusses the associations between MDSCs and various pathological conditions and briefly describes therapeutic approaches.
Collapse
Affiliation(s)
- Mahesh Raj Nepal
- College of Pharmacy, Duksung Women's University, Seoul, South Korea
- Duksung Innovative Drug Center, Duksung Women's University, Seoul, South Korea
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Sajita Shah
- College of Pharmacy, Duksung Women's University, Seoul, South Korea
- Duksung Innovative Drug Center, Duksung Women's University, Seoul, South Korea
- The Comprehensive Cancer Center, Department of Radiation Oncology, Ohio State University, Columbus, OH, USA
| | - Kyu-Tae Kang
- College of Pharmacy, Duksung Women's University, Seoul, South Korea.
- Duksung Innovative Drug Center, Duksung Women's University, Seoul, South Korea.
| |
Collapse
|
47
|
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
|
48
|
Zhang X, Liang Q, Cao Y, Yang T, An M, Liu Z, Yang J, Liu Y. Dual depletion of myeloid-derived suppressor cells and tumor cells with self-assembled gemcitabine-celecoxib nano-twin drug for cancer chemoimmunotherapy. J Nanobiotechnology 2024; 22:319. [PMID: 38849938 PMCID: PMC11161946 DOI: 10.1186/s12951-024-02598-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 05/28/2024] [Indexed: 06/09/2024] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) have played a significant role in facilitating tumor immune escape and inducing an immunosuppressive tumor microenvironment. Eliminating MDSCs and tumor cells remains a major challenge in cancer immunotherapy. A novel approach has been developed using gemcitabine-celecoxib twin drug-based nano-assembled carrier-free nanoparticles (GEM-CXB NPs) for dual depletion of MDSCs and tumor cells in breast cancer chemoimmunotherapy. The GEM-CXB NPs exhibit prolonged blood circulation, leading to the preferential accumulation and co-release of GEM and CXB in tumors. This promotes synergistic chemotherapeutic activity by the proliferation inhibition and apoptosis induction against 4T1 tumor cells. In addition, it enhances tumor immunogenicity by immunogenic cell death induction and MDSC-induced immunosuppression alleviation through the depletion of MDSCs. These mechanisms synergistically activate the antitumor immune function of cytotoxic T cells and natural killer cells, inhibit the proliferation of regulatory T cells, and promote the M2 to M1 phenotype repolarization of tumor-associated macrophages, considerably enhancing the overall antitumor and anti-metastasis efficacy in BALB/c mice bearing 4T1 tumors. The simplified engineering of GEM-CXB NPs, with their dual depletion strategy targeting immunosuppressive cells and tumor cells, represents an advanced concept in cancer chemoimmunotherapy.
Collapse
Affiliation(s)
- Xiaojie Zhang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Qiangwei Liang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China
| | - Yongjin Cao
- Department of Pharmacy, School of Nursing, Wuxi Taihu University, Wuxi, 214064, China
| | - Ting Yang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Min An
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Zihan Liu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Jiayu Yang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Yanhua Liu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
49
|
Takacs GP, Garcia JS, Hodges CA, Kreiger CJ, Sherman A, Harrison JK. Glioma-derived M-CSF and IL-34 license M-MDSCs to suppress CD8 + T cells in a NOS-dependent manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.05.597474. [PMID: 38895268 PMCID: PMC11185662 DOI: 10.1101/2024.06.05.597474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Glioblastoma (GBM) is the most common malignant primary brain tumor, resulting in poor survival despite aggressive therapies. GBM is characterized by a highly heterogeneous and immunosuppressive tumor microenvironment (TME) made up predominantly of infiltrating peripheral immune cells. One significant immune cell type that contributes to glioma immune evasion is a population of immunosuppressive cells, termed myeloid-derived suppressor cells (MDSCs). Previous studies suggest that a subset of myeloid cells, expressing monocytic (M)-MDSC markers and dual expression of chemokine receptors CCR2 and CX3CR1, utilize CCR2 to infiltrate the TME. This study evaluated the mechanism of CCR2+/CX3CR1+ M-MDSC differentiation and T cell suppressive function in murine glioma models. We determined that bone marrow-derived CCR2+/CX3CR1+ cells adopt an immune suppressive cell phenotype when cultured with glioma-derived factors. Glioma secreted CSF1R ligands M-CSF and IL-34 were identified as key drivers of M-MDSC differentiation while adenosine and iNOS pathways were implicated in M-MDSC suppression of T cells. Mining a human GBM spatial RNAseq database revealed a variety of different pathways that M-MDSCs utilize to exert their suppressive function that are driven by complex niches within the microenvironment. These data provide a more comprehensive understanding of the mechanism of M-MDSCs in glioblastoma.
Collapse
Affiliation(s)
- Gregory P. Takacs
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, 32610, United States
| | - Julia S. Garcia
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, 32610, United States
| | - Caitlyn A. Hodges
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, 32610, United States
| | - Christian J. Kreiger
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, 32610, United States
| | - Alexandra Sherman
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, 32610, United States
| | - Jeffrey K. Harrison
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, 32610, United States
| |
Collapse
|
50
|
Yaseen MM, Abuharfeil NM, Darmani H. MDSC expansion during HIV infection: regulators, ART and immune reconstitution. Genes Immun 2024; 25:242-253. [PMID: 38605259 DOI: 10.1038/s41435-024-00272-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/13/2024]
Abstract
Myeloid-derived suppressor cells (MDSCs) become expanded in different pathological conditions including human immunodeficiency virus (HIV) infection and this may worsen the disease status and accelerate disease progression. In HIV infection, MDSCs suppress anti-HIV immune responses and hamper immune reconstitution. Understanding the factors and mechanisms of MDSC expansion during HIV infection is central to understanding the pathophysiology of HIV infection. This may pave the way to developing new therapeutic targets or strategies. In this work we addressed (i) the mechanisms that regulate MDSC expansion, (ii) the impact of antiretroviral therapy (ART) on the frequency of MDSCs during HIV infection; (iii) the impact of MDSCs on immune reconstitution during successful ART; and (iv) the potential of MDSCs as a therapeutic target.
Collapse
Affiliation(s)
- Mahmoud Mohammad Yaseen
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan.
| | - Nizar Mohammad Abuharfeil
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan
| | - Homa Darmani
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan
| |
Collapse
|